1
|
Kim H, Seo KH, Kim K, Shim J, Lee Y. Application and optimization of the U-Net++ model for cerebral artery segmentation based on computed tomographic angiography images. Eur J Radiol 2025; 188:112137. [PMID: 40367559 DOI: 10.1016/j.ejrad.2025.112137] [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: 01/10/2025] [Revised: 03/20/2025] [Accepted: 04/25/2025] [Indexed: 05/16/2025]
Abstract
Accurate segmentation of cerebral arteries on computed tomography angiography (CTA) images is essential for the diagnosis and management of cerebrovascular diseases, including ischemic stroke. This study implemented a deep learning-based U-Net++ model for cerebral artery segmentation in CTA images, focusing on optimizing pruning levels by analyzing the trade-off between segmentation performance and computational cost. Dual-energy CTA and direct subtraction CTA datasets were utilized to segment the internal carotid and vertebral arteries in close proximity to the bone. We implemented four pruning levels (L1-L4) in the U-Net++ model and evaluated the segmentation performance using accuracy, intersection over union, F1-score, boundary F1-score, and Hausdorff distance. Statistical analyses were conducted to assess the significance of segmentation performance differences across pruning levels. In addition, we measured training and inference times to evaluate the trade-off between segmentation performance and computational efficiency. Applying deep supervision improved segmentation performance across all factors. While the L4 pruning level achieved the highest segmentation performance, L3 significantly reduced training and inference times (by an average of 51.56 % and 22.62 %, respectively), while incurring only a small decrease in segmentation performance (7.08 %) compared to L4. These results suggest that L3 achieves an optimal balance between performance and computational cost. This study demonstrates that pruning levels in U-Net++ models can be optimized to reduce computational cost while maintaining effective segmentation performance. By simplifying deep learning models, this approach can improve the efficiency of cerebrovascular segmentation, contributing to faster and more accurate diagnoses in clinical settings.
Collapse
Affiliation(s)
- Hajin Kim
- Department of Health Science, General Graduate School of Gachon University, 191, Hambakmoero, Yeonsu-gu, Incheon 21936, Republic of Korea
| | - Kang-Hyeon Seo
- Department of Health Science, General Graduate School of Gachon University, 191, Hambakmoero, Yeonsu-gu, Incheon 21936, Republic of Korea
| | - Kyuseok Kim
- Institute of Human Convergence Health Science, Gachon University, 191, Hambakmoero, Yeonsu-gu, Incheon 21936, Republic of Korea
| | - Jina Shim
- Department of Radiotechnology, Wonkwang Health Science University, 514, Iksan-daero, Iksan-si, Jeonbuk-do 54538, Republic of Korea
| | - Youngjin Lee
- Department of Radiological Science, Gachon University, 191, Hambakmoero, Yeonsu-gu, Incheon 21936, Republic of Korea.
| |
Collapse
|
2
|
Li W, George P, Azadian MM, Ning M, Dhand A, Cramer SC, Carmichael ST, Lo EH. Changing genes, cells and networks to reprogram the brain after stroke. Nat Neurosci 2025; 28:1130-1145. [PMID: 40456908 DOI: 10.1038/s41593-025-01981-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 03/04/2025] [Indexed: 06/11/2025]
Abstract
Important advances have been made in reperfusion therapies for acute ischemic stroke. However, a majority of patients are either ineligible for or do not respond to treatments and continue to have considerable functional deficits. Stroke results in a pathological disruption of the neurovascular unit (NVU) that involves blood-brain barrier leakage, glial activation, neuronal damage and chronic inflammation, all of which create a microenvironment that hinders recovery. Therefore, finding ways to promote central nervous system recovery remains the holy grail of stroke research. Here we propose a conceptual framework to synthesize recent progress in the field, which is currently dispersed and disconnected in the literature. We suggest that stroke recovery requires an integrated reprogramming process throughout the brain that occurs at multiple levels, including changes in gene expression, endogenous cellular transdifferentiation within the NVU, and reorganization of larger-scale neural and social networks.
Collapse
Affiliation(s)
- Wenlu Li
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Zhejiang Key Laboratory of Neuropsychopharmacology, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, PR China.
| | - Paul George
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Matine M Azadian
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - MingMing Ning
- Clinical Proteomics Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Amar Dhand
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Steven C Cramer
- Department of Neurology, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA, USA
| | - S Thomas Carmichael
- Department of Neurology, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA, USA
| | - Eng H Lo
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
3
|
Sato Y, Kato Y, Kanoke A, Sun JY, Nishijima Y, Wang RK, Stryker M, Endo H, Liu J. Type 2 diabetes abates retrograde collateral flow and promotes leukocyte adhesion following ischemic stroke. J Cereb Blood Flow Metab 2025:271678X251338203. [PMID: 40439073 PMCID: PMC12122487 DOI: 10.1177/0271678x251338203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 02/25/2025] [Accepted: 04/07/2025] [Indexed: 06/02/2025]
Abstract
Type 2 diabetes mellitus (T2DM) is associated with impaired leptomeningeal collateral compensation and poor stroke outcome. Neutrophils tethering and rolling on endothelium after stroke can also independently reduce flow velocity. However, the chronology and topological changes in collateral circulation in T2DM is not yet defined. Here, we describe the spatial and temporal blood flow dynamics and vessel diameter changes in pial arteries and veins and leukocyte-endothelial adhesion following middle cerebral artery (MCA) stroke using two-photon microscopy in awake control and T2DM mice. Relative to control mice, T2DM mice already exhibited smaller pial vessels with reduced flow velocity prior to stroke. Following stroke, T2DM mice displayed persistently reduced blood flow in pial arteries and veins, resulting in a poor recovery of downstream penetrating arterial flow and a sustained deficit in microvascular flow. There was also persistent increase of leukocyte adhesion to the endothelium of veins, coincided with elevated neutrophils infiltration into brain parenchyma in T2DM mice compared to control mice after stroke. Our data suggest that T2DM-induced increase in inflammation and chronic remodeling of leptomeningeal vessels may contribute to the observed hemodynamics deficiency after stroke and subsequent poor stroke outcome.
Collapse
Affiliation(s)
- Yoshimichi Sato
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- SFVAMC, San Francisco, CA, USA
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Yuya Kato
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- SFVAMC, San Francisco, CA, USA
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Atsushi Kanoke
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- SFVAMC, San Francisco, CA, USA
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Jennifer Y Sun
- Institute of Ophthalmology, University College London, London, UK
- Department of Physiology, University of California San Francisco, San Francisco, CA, USA
| | - Yasuo Nishijima
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- SFVAMC, San Francisco, CA, USA
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Ruikang K Wang
- Department of Bioengineering, College of Engineering and School of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Michael Stryker
- Department of Physiology, University of California San Francisco, San Francisco, CA, USA
| | - Hidenori Endo
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Jialing Liu
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- SFVAMC, San Francisco, CA, USA
| |
Collapse
|
4
|
Wang J, Zhou J, Zhu J, Sheng J, Jiang R, Zhang X. Brain remodeling in stroke patients: A comprehensive review of mechanistic and neuroimaging studies. Behav Brain Res 2025; 486:115548. [PMID: 40122286 DOI: 10.1016/j.bbr.2025.115548] [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: 11/12/2024] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
Stroke-induced brain remodeling involves a complex interplay of neurovascular components, including endothelial cells, microglia, astrocytes, and pericytes, which collectively contribute to the restoration of brain function. These processes are crucial for repairing the blood-brain barrier, regulating inflammation, and promoting neurogenesis. This review examines the mechanisms underlying brain remodeling and the role of advanced neuroimaging techniques-such as functional MRI (fMRI), positron emission tomography (PET), functional near-infrared spectroscopy (fNIRS), and functional ultrasound (fUS)-in assessing these changes. We also discuss various therapeutic approaches aimed at enhancing brain remodeling, including pharmacological agents, stem cell therapy, and rehabilitation strategies that target neurovascular repair and functional recovery. Despite significant progress, challenges remain in translating imaging insights into effective treatments. Future research should focus on integrating multiple imaging modalities to provide a comprehensive view of neurovascular changes and refining therapeutic interventions to optimize recovery and functional outcomes in stroke patients.
Collapse
Affiliation(s)
- Jing Wang
- Department of Radiology, The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, China.
| | - Jian Zhou
- Department of Radiology, No. 945 Hospital of Joint Logistics Support Force of the Chinese People's Liberation Army, Yaan, Sichuan 625000, China.
| | - Jing Zhu
- Department of Radiology, The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, China.
| | - Jinping Sheng
- Department of Radiology, The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, China.
| | - Rui Jiang
- Department of Radiology, The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, China.
| | - Xiao Zhang
- Department of Radiology, The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, China.
| |
Collapse
|
5
|
Deshpande A, Zhang LQ, Balu R, Yahyavi-Firouz-Abadi N, Badjatia N, Laksari K, Tahsili-Fahadan P. Cerebrovascular morphology: Insights into normal variations, aging effects, and disease implications. J Cereb Blood Flow Metab 2025:271678X251328537. [PMID: 40314210 PMCID: PMC12048404 DOI: 10.1177/0271678x251328537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 02/20/2025] [Accepted: 03/04/2025] [Indexed: 05/03/2025]
Abstract
Cerebrovascular morphology plays a critical role in brain health, influencing cerebral blood flow (CBF) and contributing to the pathogenesis of various neurological diseases. This review examines the anatomical structure of the cerebrovascular network and its variations in healthy and diseased populations and highlights age-related changes and their implications in various neurological conditions. Normal variations, including the completeness and anatomical anomalies of the Circle of Willis and collateral circulation, are discussed in relation to their impact on CBF and susceptibility to ischemic events. Age-related changes in the cerebrovascular system, such as alterations in vessel geometry and density, are explored for their contributions to age-related neurological disorders, including Alzheimer's disease and vascular dementia. Advances in medical imaging and computational methods have enabled automatic quantitative assessment of cerebrovascular structures, facilitating the identification of pathological changes in both acute and chronic cerebrovascular disorders. Emerging technologies, including machine learning and computational fluid dynamics, offer new tools for predicting disease risk and patient outcomes based on vascular morphology. This review underscores the importance of understanding cerebrovascular remodeling for early diagnosis and the development of novel therapeutic approaches in brain diseases.
Collapse
Affiliation(s)
- Aditi Deshpande
- Department of Mechanical Engineering, University of California, Riverside, USA
| | - Lucy Q Zhang
- Department of Neurology, Duke University School of Medicine, Durham, NC, USA
| | - Ramani Balu
- Vascular Neurology and Neurocritical Care, Inova Neuroscience and Spine Institute, Inova Fairfax Medical Campus, Falls Church, VA, USA
- Department of Medical Education, University of Virginia, Inova Campus, Falls Church, VA, USA
| | - Noushin Yahyavi-Firouz-Abadi
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Neeraj Badjatia
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kaveh Laksari
- Department of Mechanical Engineering, University of California, Riverside, USA
| | - Pouya Tahsili-Fahadan
- Vascular Neurology and Neurocritical Care, Inova Neuroscience and Spine Institute, Inova Fairfax Medical Campus, Falls Church, VA, USA
- Department of Medical Education, University of Virginia, Inova Campus, Falls Church, VA, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
6
|
Wang BQ, Duan YY, Chen M, Ma YF, Chen R, Huang C, Gao F, Xu R, Duan CM. Endothelial Cell Integrin α6 Regulates Vascular Remodeling Through the PI3K/Akt-eNOS-VEGFA Axis After Stroke. Neurosci Bull 2025:10.1007/s12264-025-01403-6. [PMID: 40316875 DOI: 10.1007/s12264-025-01403-6] [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: 09/10/2024] [Accepted: 12/28/2024] [Indexed: 05/04/2025] Open
Abstract
The angiogenic response is essential for the repair of ischemic brain tissue. Integrin α6 (Itga6) expression has been shown to increase under hypoxic conditions and is expressed exclusively in vascular structures; however, its role in post-ischemic angiogenesis remains poorly understood. In this study, we demonstrate that mice with endothelial cell-specific knockout of Itga6 exhibit reduced neovascularization, reduced pericyte coverage on microvessels, and accelerated breakdown of microvascular integrity in the peri-infarct area. In vitro, endothelial cells with ITGA6 knockdown display reduced proliferation, migration, and tube-formation. Mechanistically, we demonstrated that ITGA6 regulates post-stroke angiogenesis through the PI3K/Akt-eNOS-VEGFA axis. Importantly, the specific overexpression of Itga6 in endothelial cells significantly enhanced neovascularization and enhanced the integrity of microvessels, leading to improved functional recovery. Our results suggest that endothelial cell Itga6 plays a crucial role in key steps of post-stroke angiogenesis, and may represent a promising therapeutic target for promoting recovery after stroke.
Collapse
Affiliation(s)
- Bing-Qiao Wang
- Department of Neurology, Xinqiao Hospital, The Army Medical University, Chongqing, 400037, China
| | - Yang-Ying Duan
- Department of Ultrasound Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Mao Chen
- Department of Neurology, Xinqiao Hospital, The Army Medical University, Chongqing, 400037, China
| | - Yu-Fan Ma
- Department of Neurology, Xinqiao Hospital, The Army Medical University, Chongqing, 400037, China
| | - Ru Chen
- Department of Neurology, Xinqiao Hospital, The Army Medical University, Chongqing, 400037, China
| | - Cheng Huang
- Department of Neurology, Xinqiao Hospital, The Army Medical University, Chongqing, 400037, China
| | - Fei Gao
- Department of Neurology, Xinqiao Hospital, The Army Medical University, Chongqing, 400037, China
| | - Rui Xu
- Department of Neurology, Xinqiao Hospital, The Army Medical University, Chongqing, 400037, China.
| | - Chun-Mei Duan
- Department of Neurology, Xinqiao Hospital, The Army Medical University, Chongqing, 400037, China.
| |
Collapse
|
7
|
Ju H, Minker J, Pavlova I, Cho S, Kim ID. Acute, not delayed, treatment of aflibercept enhances vessel density in post-ischemic brain and promotes long-term stroke recovery in obese mice. J Cereb Blood Flow Metab 2025:271678X251330102. [PMID: 40314397 PMCID: PMC12048399 DOI: 10.1177/0271678x251330102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 03/06/2025] [Accepted: 03/09/2025] [Indexed: 05/03/2025]
Abstract
Vascular comorbidities complicate stroke pathophysiology, worsen outcomes, and delay recovery. Obesity, in particular, significantly increases stroke-induced brain edema, a fatal complication during infarction, which leads to worsened long-term recovery. Treatment of aflibercept, a VEGF-trap, has been shown to reduce stroke-induced brain edema and attenuate acute neurological deficits in obese mice. However, the effect of aflibercept on long-term stroke recovery is unknown. We found that treating obese stroke mice with aflibercept at 3 hours displayed significantly improved long-term motor and cognitive function. Notably, VEGFR2 expression was upregulated at 3- and 7-days post-stroke, indicating sustained VEGF signaling in obese subjects. Unlike acute treatment of aflibercept at 3 hours post-stroke, delayed treatment (3-day) worsened stroke recovery. While the improved long-term stroke recovery in mice treated aflibercept 3 hours is associated with the upregulated Pecam-1 and Angiopoietin-1 mRNAs and vessel densities in peri-infarct area at 3 months post-stroke, the delayed treatment led to a reduction in both angiogenic marker expression and vessel density. These findings highlight the importance of early intervention with VEGF signaling in obese mice to promote subsequent vascular remodeling during the stroke recovery phase and indicate a critical therapeutic window for VEGF inhibition to treat stroke in subjects with vascular comorbidities.
Collapse
Affiliation(s)
- Hyunwoo Ju
- Burke Neurological Institute, White Plains, NY, USA
- Feil Brain Mind Research Institute, Weill Cornell Medicine, NY, USA
| | | | - Ina Pavlova
- Burke Neurological Institute, White Plains, NY, USA
| | - Sunghee Cho
- Burke Neurological Institute, White Plains, NY, USA
- Feil Brain Mind Research Institute, Weill Cornell Medicine, NY, USA
| | - Il-doo Kim
- Burke Neurological Institute, White Plains, NY, USA
- Department of Anatomy, Inha University School of Medicine, Incheon, Republic of Korea
| |
Collapse
|
8
|
Yu D, Liu M, Ding Q, Wu Y, Wang T, Song L, Li X, Qian K, Cheng Z, Gu M, Li Z. Molecular imaging-guided diagnosis and treatment integration for brain diseases. Biomaterials 2025; 316:123021. [PMID: 39705925 DOI: 10.1016/j.biomaterials.2024.123021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 12/03/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
In practical clinical scenarios, improved diagnostic methods have been developed for the precise visualization of molecular targets using molecular imaging in brain diseases. Recently, the introduction of innovative molecular imaging modalities across both macroscopic and mesoscopic dimensions, with remarkable specificity and spatial resolution, has expanded the scope of applications beyond diagnostic testing, with the potential to guide therapeutic interventions, offering real-time feedback in the context of brain therapy. The molecular imaging-guided integration of diagnosis and treatment holds the potential to revolutionize disease management by enabling the real-time monitoring of treatment responses and therapy adjustments. Given the vibrant and ever-evolving nature of this field, this review provides an integrated picture on molecular image-guided diagnosis and treatment integration for brain diseases involving the basic concepts, significant breakthroughs, and recent trends. In addition, based on the current achievements, some critical challenges are also discussed.
Collapse
Affiliation(s)
- Donghu Yu
- Brain Glioma Center & Department of Neurosurgery, International Science and Technology Cooperation Base for Research and Clinical Techniques for Brain Glioma Diagnosis and Treatment, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Menghao Liu
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Qihang Ding
- Department of Chemistry, Korea University, Seoul, 02841, South Korea.
| | - Youxian Wu
- Brain Glioma Center & Department of Neurosurgery, International Science and Technology Cooperation Base for Research and Clinical Techniques for Brain Glioma Diagnosis and Treatment, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Tianqing Wang
- Brain Glioma Center & Department of Neurosurgery, International Science and Technology Cooperation Base for Research and Clinical Techniques for Brain Glioma Diagnosis and Treatment, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Litong Song
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xiaoyu Li
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Kun Qian
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Zhen Cheng
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Meijia Gu
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China.
| | - Zhiqiang Li
- Brain Glioma Center & Department of Neurosurgery, International Science and Technology Cooperation Base for Research and Clinical Techniques for Brain Glioma Diagnosis and Treatment, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
9
|
Wang T, Wang X, Liu S, Li M, Wan K, Zheng J, Liao K, Wang J, Zou K, Wang L, Xu H, Lei W, Chen G, Li W. Transcription Factor-Based Gene Therapy Enables Functional Repair of Rat Following Chronic Ischemic Stroke. CNS Neurosci Ther 2025; 31:e70448. [PMID: 40401537 PMCID: PMC12096174 DOI: 10.1111/cns.70448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/21/2025] [Accepted: 05/07/2025] [Indexed: 05/23/2025] Open
Abstract
OBJECTIVE In vivo transcription factor (TF) -mediated gene therapy through astrocyte-to-neuron (AtN) conversion has shown therapeutic effects on rodent and non-human primate cortical ischemic injury in the subacute phase. However, in the clinic, subcortical regions including striatum as well as white matter are vulnerable regions of stroke, with millions of patients beyond subacute phase. In this study, we investigate whether TF-mediated AtN conversion therapy can be extended to treat chronic-phase ischemic stroke involving subcortical regions (e.g., striatum) and white matter, beyond cortical injuries. METHODS Rat middle cerebral artery occlusion (MCAO)-like models were established to induce broad ischemic injuries including cortical and striatal regions. Then multiple rounds of TF-mediated gene therapy treatments through adeno-associated virus (AAV) system to cover the large-scaled infarct areas were conducted in the chronic phase of the stroke models. Magnetic resonance imaging (MRI), [18F] FDG-PET/CT, behavioral tests, immunohistochemistry and bulk-RNA seq were applied to evaluate the AtN conversion, tissue repair and functional recovery. RESULTS Our results revealed that administrated in the chronic phase of ischemic stroke, TF-mediated gene therapy can efficiently regenerate new neurons in both cortical and striatal regions, and promote tissue repair in both grey and white matter. Compared with single round of AAV administration, multiple rounds of treatment regenerated more neurons and led to a significant functional recovery. CONCLUSIONS Our study demonstrates that TF-mediated gene therapy has a broad therapeutic time window and can be applied multiple rounds to treat severe ischemic stroke, making it an attractive therapeutic intervention in the chronic phase after stroke, when current approaches are largely ineffective.
Collapse
Affiliation(s)
- Tao Wang
- Guangdong‐Hong Kong‐Macau Institute of CNS Regeneration (GHMICR)Jinan UniversityGuangzhouChina
| | - Xu Wang
- Guangdong‐Hong Kong‐Macau Institute of CNS Regeneration (GHMICR)Jinan UniversityGuangzhouChina
| | - Shanggong Liu
- Guangdong‐Hong Kong‐Macau Institute of CNS Regeneration (GHMICR)Jinan UniversityGuangzhouChina
| | - Menglei Li
- Guangdong‐Hong Kong‐Macau Institute of CNS Regeneration (GHMICR)Jinan UniversityGuangzhouChina
| | - Kaiying Wan
- Guangdong‐Hong Kong‐Macau Institute of CNS Regeneration (GHMICR)Jinan UniversityGuangzhouChina
| | - Jiajun Zheng
- Guangdong‐Hong Kong‐Macau Institute of CNS Regeneration (GHMICR)Jinan UniversityGuangzhouChina
| | - Kai Liao
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non‐Human Primate Research, GHM Institute of CNS RegenerationJinan UniversityGuangzhouChina
| | - Jinyu Wang
- Guangdong‐Hong Kong‐Macau Institute of CNS Regeneration (GHMICR)Jinan UniversityGuangzhouChina
| | - Kaiming Zou
- Guangdong‐Hong Kong‐Macau Institute of CNS Regeneration (GHMICR)Jinan UniversityGuangzhouChina
| | - Lu Wang
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non‐Human Primate Research, GHM Institute of CNS RegenerationJinan UniversityGuangzhouChina
| | - Hao Xu
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non‐Human Primate Research, GHM Institute of CNS RegenerationJinan UniversityGuangzhouChina
| | - Wenliang Lei
- Guangdong‐Hong Kong‐Macau Institute of CNS Regeneration (GHMICR)Jinan UniversityGuangzhouChina
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative DrugsJinan UniversityGuangzhouChina
- Department of Nuclear Medicine and PET/CT‐MRI CenterThe First Affiliated Hospital of Jinan University & Institute of Molecular and Functional Imaging, Jinan UniversityGuangzhouChina
| | - Gong Chen
- Guangdong‐Hong Kong‐Macau Institute of CNS Regeneration (GHMICR)Jinan UniversityGuangzhouChina
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative DrugsJinan UniversityGuangzhouChina
- Department of Nuclear Medicine and PET/CT‐MRI CenterThe First Affiliated Hospital of Jinan University & Institute of Molecular and Functional Imaging, Jinan UniversityGuangzhouChina
| | - Wen Li
- Guangdong‐Hong Kong‐Macau Institute of CNS Regeneration (GHMICR)Jinan UniversityGuangzhouChina
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative DrugsJinan UniversityGuangzhouChina
- Department of Nuclear Medicine and PET/CT‐MRI CenterThe First Affiliated Hospital of Jinan University & Institute of Molecular and Functional Imaging, Jinan UniversityGuangzhouChina
| |
Collapse
|
10
|
Yang X, Li B, Wu L, Cui Y. Acupuncture treatment of vascular cognitive impairment through peripheral nerve stimulation pathway: a scoping review. Front Aging Neurosci 2025; 17:1515327. [PMID: 40357233 PMCID: PMC12066784 DOI: 10.3389/fnagi.2025.1515327] [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: 10/22/2024] [Accepted: 04/14/2025] [Indexed: 05/15/2025] Open
Abstract
Objective This study aims to explore the central effects of acupuncture on vascular cognitive impairment (VCI) through peripheral nerve stimulation. Methods This scoping review followed the methodological framework proposed by Arksey and O'Malley and the PRISMA-ScR guidelines. A comprehensive search of databases, including PubMed, Web of Science, MEDLINE, and Embase, was conducted, including 79 studies on acupuncture interventions for VCI. Acupoints and their underlying anatomical structures related to peripheral nerves were summarized, and the potential pathways of acupuncture effects via different peripheral nerves were explored. Results The results showed that acupuncture, by stimulating specific acupoints on the head, face, torso, and limbs, significantly affects peripheral nerve networks, including the cervical, lumbar, and sacral plexuses, thoracic nerves, vagus nerve, trigeminal nerve and its branches. The nerve stimulation effects of acupuncture can enhance the regulation of cerebral blood flow, modulate neuroimmune responses, improve brain function, and promote neuroplasticity through multiple central nervous system pathways, ultimately improving cognitive function and treating VCI. Conclusion Acupuncture is a treatment modality that influences the central nervous system through peripheral nerve stimulation to treat VCI. A deeper understanding of the central effects induced by acupuncture-triggered neural reflexes can contribute to the improvement of existing therapies and help elucidate the scientific principles underlying acupuncture's therapeutic effects.
Collapse
Affiliation(s)
- Xinming Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Bo Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Qinghai Provincial Hospital of Traditional Chinese Medicine, Xining, China
| | - Linna Wu
- The First Clinical Medical School, Yunnan University of Chinese Medicine, Kunming, China
| | - Ying Cui
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
11
|
de Magalhães ACPS, Gurgel GDA, de Barros SMW, Valente MLS, Aquino MDA, Bessa SDS, Baquette RF, Castro AA, Pitta GBB. Gastrointestinal histological injury in pigs subjected to triple stent interposition in the thoracoabdominal aorta. Acta Cir Bras 2025; 40:e402425. [PMID: 40105604 PMCID: PMC11908737 DOI: 10.1590/acb402425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 01/10/2025] [Indexed: 03/20/2025] Open
Abstract
PURPOSE To evaluate gastrointestinal histological injury in pigs subjected to triple stent interposition versus a control group, hypothesizing no significant injury increase with triple stents. METHODS A prospective study with 15 pigs divided into a control group (G0, n = 5) undergoing arteriography only, and a triple stent group (G3, n = 10) undergoing arteriography and three stent implantations in the thoracoabdominal aorta. After an eight-day observation, arteriography, euthanasia, and en bloc gastrointestinal harvesting were performed. Lesions were graded using the Park/Chiu classification, and serum markers were analyzed pre- and post-procedure. RESULTS Arteriography confirmed mesenteric artery patency in all animals. Histological analysis showed ischemic lesions in 88.9% of G3, mainly in the colon (89%), compared to 60% in G0, primarily in the colon (60%) and stomach (40%). Most G3 lesions were grade 1, while G0 had higher-grade lesions. Serum markers showed no significant intergroup differences. CONCLUSION Triple stent interposition did not significantly increase gastrointestinal injury, indicating its safety for maintaining gastrointestinal perfusion in this model.
Collapse
Affiliation(s)
| | - Gutenberg do Amaral Gurgel
- Universidade Federal do Rio Grande do Sul – Faculdade de Medicina – Departamento de Cirurgia – Porto Alegre (RS) – Brazil
| | | | - Miguel Lucas Silva Valente
- Universidade Federal do Acre – Faculdade de Medicina – Departamento de Cirurgia – Rio Branco (AC) – Brazil
| | - Maurício de Amorim Aquino
- Universidade Federal do Rio Grande do Sul – Faculdade de Medicina – Departamento de Cirurgia – Porto Alegre (RS) – Brazil
| | - Sthefanie da Silva Bessa
- Centro Universitário São Lucas – Faculdade de Medicina – Departamento de Cirurgia – Porto Velho (RO) – Brazil
| | | | - Aldemar Araújo Castro
- Universidade Estadual de Ciências da Saúde de Alagoas – Faculdade de Medicina – Departamento de Cirurgia – Maceió (AL) – Brazil
| | | |
Collapse
|
12
|
Alaqel SI, Imran M, Khan A, Nayeem N. Aging, vascular dysfunction, and the blood-brain barrier: unveiling the pathophysiology of stroke in older adults. Biogerontology 2025; 26:67. [PMID: 40044939 DOI: 10.1007/s10522-025-10209-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: 01/23/2025] [Accepted: 02/18/2025] [Indexed: 05/09/2025]
Abstract
The progressive decline of vascular integrity and blood-brain barrier (BBB) function is associated with aging, a major risk factor for stroke. This review describes the cellular and molecular changes in the brain microvasculature of the neurovascular unit (NVU) that contribute to the development of BBB dysfunction in aging, such as endothelial cell senescence, oxidative stress, and degradation of tight junction proteins. Stroke severity and recovery are exacerbated by BBB breakdown, leading to neuroinflammation, neurotoxicity, and cerebral oedema while identifying molecular mechanisms such as the NLRP3 inflammasome, matrix metalloproteinases (MMPs), and non-coding RNAs (e.g., miRNAs and circRNAs) that drive BBB disruption in aging and stroke. Real-time assessment of BBB permeability in stroke pathophysiology is made possible using advanced imaging techniques, such as dynamic contrast-enhanced MRI and positron emission tomography. Furthermore, biomarkers, including claudin-5, PDGFRβ, or albumin concentration, serve as markers of BBB integrity and vascular health. Restoration of BBB function and stroke recovery with emerging therapeutic strategies, including sirtuin modulators (SIRT1 and SIRT3 activators to enhance endothelial function and mitochondrial health), stem cell-derived extracellular vesicles (iPSC-sEVs for BBB repair and neuroprotection), NLRP3 inflammasome inhibitors (MCC950 to attenuate endothelial pyroptosis and inflammation), hydrogen-rich water therapy (to counteract oxidative stress-induced BBB damage), and neuropeptides such as cortistatin (to regulate neuroinflammation and BBB stability), is promising. This review explores the pathophysiological mechanisms of BBB dysfunction in aging and stroke, their relation to potential therapeutic targets, and novel approaches to improve vascular health and neuroprotection.
Collapse
Affiliation(s)
- Saleh I Alaqel
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, 91911, Rafha, Saudi Arabia.
- King Salman Center for Disability Research, 11614, Riyadh, Saudi Arabia.
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, 91911, Rafha, Saudi Arabia
- Center For Health Research, Northern Border University, Arar, Saudi Arabia
| | - Abida Khan
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, 91911, Rafha, Saudi Arabia
- Center For Health Research, Northern Border University, Arar, Saudi Arabia
| | - Naira Nayeem
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, 91911, Rafha, Saudi Arabia
| |
Collapse
|
13
|
Sasegbon A, Cheng I, Hamdy S. The neurorehabilitation of post-stroke dysphagia: Physiology and pathophysiology. J Physiol 2025; 603:617-634. [PMID: 38517302 PMCID: PMC11782911 DOI: 10.1113/jp285564] [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: 12/02/2023] [Accepted: 02/29/2024] [Indexed: 03/23/2024] Open
Abstract
Swallowing is a complex process involving the precise contractions of numerous muscles of the head and neck, which act to process and shepherd ingested material from the oral cavity to its eventual destination, the stomach. Over the past five decades, information from animal and human studies has laid bare the complex network of neurones in the brainstem, cortex and cerebellum that are responsible for orchestrating each normal swallow. Amidst this complexity, problems can and often do occur that result in dysphagia, defined as impaired or disordered swallowing. Dysphagia is common, arising from multiple varied disease processes that can affect any of the neuromuscular structures involved in swallowing. Post-stroke dysphagia (PSD) remains the most prevalent and most commonly studied form of dysphagia and, as such, provides an important disease model to assess dysphagia physiology and pathophysiology. In this review, we explore the complex neuroanatomical processes that occur during normal swallowing and PSD. This includes how strokes cause dysphagia, the mechanisms through which natural neuroplastic recovery occurs, current treatments for patients with persistent dysphagia and emerging neuromodulatory treatments.
Collapse
Affiliation(s)
- Ayodele Sasegbon
- Division of Diabetes, Endocrinology and Gastroenterology, School of Medical Sciences, Centre for Gastrointestinal Sciences, Faculty of Biology, Medicine and HealthSalford Royal Foundation TrustUniversity of ManchesterManchesterUK
| | - Ivy Cheng
- Division of Diabetes, Endocrinology and Gastroenterology, School of Medical Sciences, Centre for Gastrointestinal Sciences, Faculty of Biology, Medicine and HealthSalford Royal Foundation TrustUniversity of ManchesterManchesterUK
- Academic Unit of Human Communication, Learning, and Development, Faculty of EducationThe University of Hong KongHong KongChina
- Institute for Biomagnetism and BiosignalanalysisUniversity of MünsterMünsterGermany
| | - Shaheen Hamdy
- Division of Diabetes, Endocrinology and Gastroenterology, School of Medical Sciences, Centre for Gastrointestinal Sciences, Faculty of Biology, Medicine and HealthSalford Royal Foundation TrustUniversity of ManchesterManchesterUK
| |
Collapse
|
14
|
Ruan J, Wang L, Wang N, Huang P, Chang D, Zhou X, Seto S, Li D, Hou J. Hydroxysafflor Yellow A promotes angiogenesis of brain microvascular endothelial cells from ischemia/reperfusion injury via glycolysis pathway in vitro. J Stroke Cerebrovasc Dis 2025; 34:108107. [PMID: 39515547 DOI: 10.1016/j.jstrokecerebrovasdis.2024.108107] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/31/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Angiogenesis of brain microvascular endothelial cells (BMECs) after cerebral ischemia was conducive to improving the blood supply of ischemia tissues, which was upregulated by glycolysis. Hydroxysafflor Yellow A (HSYA) mends damaged tissues through increasing angiogenesis. METHODS HSYA treated proliferation, migration and angiogenesis of BMECs in vitro in vitro during OGD/R. HSYA regulated the key enzymes of glycolysis, such as hexokinase 2 (HK2) and 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3), glucose uptake and products (pyruvate, ATP and lactate) were detected by western blot and kits, respectively. Scratch wound assay, transwell, tube formation and spheroid sprouting were used to explore the pathway that HSYA recovered migration and angiogenesis of BMECs. We evaluated the potential target of HSYA promoting glycolysis via molecular docking, drug affinity responsive target stability (DARTS) and cellular thermal shift assay (CETSA). RESULTS HSYA promoted the proliferation, migration, tube formation and spheroid sprouting of BMECs during OGD/R, and stimulated the expression of tip phenotype marker protein (CD34), and the receptor (Notch-1) that regulated the differentiation of endothelial cells into tip/stalk phenotype. In glycolysis, PFKFB3 expression was upregulated by HSYA; HSYA also improved ATP and pyruvate levels, as well as lactate release after OGD/R. Finally, upregulating VEGFA and p-VEGFR2 of HSYA was weakened because of suppressing glycolysis; the HSYA's improvement of BMECs migration and angiogenesis was attenuated under the inhibition of glycolysis, which confirmed that HSYA were upregulating angiogenesis and expression of VEGFA/VEGFR2 by glycolysis pathway. The result about molecular docking, DARTS and CETSA suggested that PFKFB3 was the possible target of HSYA. CONCLUSION HSYA promotes angiogenesis of BMECs in vitro through the glycolysis mediated VEGFA/VEGFR2 pathway, and PFKFB3 is the potential target of HSYA to heighten glycolysis.
Collapse
Affiliation(s)
- Juxuan Ruan
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Anhui University of Chinese Medicine, No. 350, Longzihu Road, Xinzhan District, Hefei, Anhui 230012, China; Institute for Pharmacodynamics and Safety Evaluation of Chinese Medicine, Anhui Academy of Chinese Medicine, Hefei 230012, China
| | - Lei Wang
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Anhui University of Chinese Medicine, No. 350, Longzihu Road, Xinzhan District, Hefei, Anhui 230012, China; Institute for Pharmacodynamics and Safety Evaluation of Chinese Medicine, Anhui Academy of Chinese Medicine, Hefei 230012, China
| | - Ning Wang
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Anhui University of Chinese Medicine, No. 350, Longzihu Road, Xinzhan District, Hefei, Anhui 230012, China; Institute for Pharmacodynamics and Safety Evaluation of Chinese Medicine, Anhui Academy of Chinese Medicine, Hefei 230012, China.
| | - Ping Huang
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Anhui University of Chinese Medicine, No. 350, Longzihu Road, Xinzhan District, Hefei, Anhui 230012, China; Institute for Pharmacodynamics and Safety Evaluation of Chinese Medicine, Anhui Academy of Chinese Medicine, Hefei 230012, China
| | - Dennis Chang
- NICM Health Research Institute, Western Sydney University, Westmead, Sydney, NSW 2145, Australia
| | - Xian Zhou
- NICM Health Research Institute, Western Sydney University, Westmead, Sydney, NSW 2145, Australia
| | - Saiwang Seto
- Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
| | - Dan Li
- Shineway Pharmaceutical Group Co. Ltd. Shijiahzuang 51430, China
| | - Jincai Hou
- Shineway Pharmaceutical Group Co. Ltd. Shijiahzuang 51430, China
| |
Collapse
|
15
|
Cai X, Jiang J, Zhou G, Zhang Y. Mechanisms of Vagus Nerve Stimulation in Improving Motor Dysfunction After Stroke. Neuropsychiatr Dis Treat 2024; 20:2593-2601. [PMID: 39723115 PMCID: PMC11669332 DOI: 10.2147/ndt.s492043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024] Open
Abstract
Patients with stroke would have persistent functional deficits despite undergoing physiotherapy and rehabilitation training. Recently, vagus nerve stimulation (VNS), a newly emerging neuroregulatory technique, has been shown to improve motor dysfunction after stroke. Evidence from clinical and preclinical studies has proven the safety, feasibility, and efficacy of invasive and noninvasive VNS. It has been reported that the positive effect may be related to anti-inflammatory effects, mediating neuroplasticity, increasing blood-brain barrier integrity, promoting angiogenesis and reducing spreading depolarization. However, the underlying mechanism remains poorly understood. In this review, we have summarized the potential molecular mechanisms by which VNS promotes stroke prognosis. We believe that VNS combined with upper-extremity rehabilitation can improve impairment and function among moderately to severely impaired stroke survivors. The applications and further exploration are discussed to provide new insights into this novel therapeutic technique.
Collapse
Affiliation(s)
- Xiaohu Cai
- Department of Rehabilitation Medicine, Xishan People’s Hospital of Wuxi City, Wuxi Branch of Zhongda Hospital Southeast University, Wuxi, People’s Republic of China
| | - Jiayao Jiang
- Department of Orthopedics, The 904th Hospital of the Joint Logistics Support Force of the PLA, Wuxi, Jiangsu, People’s Republic of China
| | - Guochao Zhou
- Department of Orthopedics, The Army 947th Hospital, Kashgar, People’s Republic of China
| | - Yelei Zhang
- Department of Neurosurgery, Xishan People’s Hospital of Wuxi City, Wuxi Branch of Zhongda Hospital Southeast University, Wuxi, People’s Republic of China
| |
Collapse
|
16
|
Shen L, Tian Q, Ran Q, Gan Q, Hu Y, Du D, Qin Z, Duan X, Zhu X, Huang W. Z-Ligustilide: A Potential Therapeutic Agent for Atherosclerosis Complicating Cerebrovascular Disease. Biomolecules 2024; 14:1623. [PMID: 39766330 PMCID: PMC11726876 DOI: 10.3390/biom14121623] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Atherosclerosis (AS) is one of the major catalysts of ischemic cerebrovascular disease, and the death and disease burden from AS and its cerebrovascular complications are increasing. Z-ligustilide (Z-LIG) is a key active ingredient in Angelica sinensis (Oliv.) Diels and Ligusticum chuanxiong Hort. In this paper, we first introduced LIG's physicochemical properties and pharmacokinetics. Then, we reviewed Z-LIG's intervention and therapeutic mechanisms on AS and its cerebrovascular complications. The mechanisms of Z-LIG intervention in AS include improving lipid metabolism, antioxidant and anti-inflammatory effects, protecting vascular endothelium, and inhibiting vascular endothelial fibrosis, pathological thickening, and plaque calcification. In ischemic cerebrovascular diseases complicated by AS, Z-LIG exerts practical neuroprotective effects in ischemic stroke (IS), transient ischemic attack (TIA), and vascular dementia (VaD) through anti-neuroinflammatory, anti-oxidation, anti-neuronal apoptosis, protection of the blood-brain barrier, promotion of mitochondrial division and angiogenesis, improvement of cholinergic activity, inhibition of astrocyte proliferation, and endoplasmic reticulum stress. This paper aims to provide a basis for subsequent studies of Z-LIG in the prevention and treatment of AS and its cerebrovascular complications and, thus, to promote the development of interventional drugs for AS.
Collapse
Affiliation(s)
- Longyu Shen
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| | - Qianqian Tian
- Faculty of Social Sciences, The University of Hong Kong, Hong Kong 999077, China
| | - Qiqi Ran
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| | - Qianrong Gan
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| | - Yu Hu
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| | - Donglian Du
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| | - Zehua Qin
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| | - Xinyi Duan
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| | - Xinyun Zhu
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China;
| | - Wei Huang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| |
Collapse
|
17
|
Sinha A, Gupta M, Bhaskar SMM. Evolucollateral dynamics in stroke: Evolutionary pathophysiology, remodelling and emerging therapeutic strategies. Eur J Neurosci 2024; 60:6779-6798. [PMID: 39498733 DOI: 10.1111/ejn.16585] [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: 07/20/2024] [Revised: 10/13/2024] [Accepted: 10/15/2024] [Indexed: 11/07/2024]
Abstract
Leptomeningeal collaterals (LMCs) are crucial in mitigating the impact of acute ischemic stroke (AIS) by providing alternate blood flow routes when primary arteries are obstructed. This article explores the evolutionary pathophysiology of LMCs, highlighting their critical function in stroke and the genetic and molecular mechanisms governing their development and remodelling. We address the translational challenges of applying animal model findings to human clinical scenarios, emphasizing the need for further research to validate emerging therapies-such as pharmacological agents, gene therapy and mechanical interventions-in clinical settings, aimed at enhancing collateral perfusion. Computational modelling emerges as a promising method for integrating experimental data, which requires precise parameterization and empirical validation. We introduce the 'Evolucollateral Dynamics' hypothesis, proposing a novel framework that incorporates evolutionary biology principles into therapeutic strategies, offering new perspectives on enhancing collateral circulation. This hypothesis emphasizes the role of genetic predispositions and environmental influences on collateral circulation, which may impact therapeutic strategies and optimize treatment outcomes. Future research must incorporate human clinical data to create robust treatment protocols, thereby maximizing the therapeutic potential of LMCs and improving outcomes for stroke patients.
Collapse
Affiliation(s)
- Akansha Sinha
- Global Health Neurology Lab, Sydney, NSW, Australia
- UNSW Medicine and Health, University of New South Wales (UNSW), South West Sydney Clinical Campuses, Sydney, NSW, Australia
| | - Muskaan Gupta
- Global Health Neurology Lab, Sydney, NSW, Australia
- UNSW Medicine and Health, University of New South Wales (UNSW), South West Sydney Clinical Campuses, Sydney, NSW, Australia
| | - Sonu M M Bhaskar
- Global Health Neurology Lab, Sydney, NSW, Australia
- UNSW Medicine and Health, University of New South Wales (UNSW), South West Sydney Clinical Campuses, Sydney, NSW, Australia
- NSW Brain Clot Bank, NSW Health Pathology, Sydney, NSW, Australia
- Department of Neurology & Neurophysiology, Liverpool Hospital and South West Sydney Local Health District, Liverpool, NSW, Australia
- Clinical Sciences Stream, Ingham Institute for Applied Medical Research, Liverpool, NSW, Australia
- Department of Neurology, Division of Cerebrovascular Medicine and Neurology, National Cerebral and Cardiovascular Center (NCVC), Suita, Osaka, Japan
| |
Collapse
|
18
|
Howard PG, Zou P, Zhang Y, Huang F, Tesic V, Wu CYC, Lee RHC. Serum/glucocorticoid regulated kinase 1 (SGK1) in neurological disorders: pain or gain. Exp Neurol 2024; 382:114973. [PMID: 39326820 PMCID: PMC11536509 DOI: 10.1016/j.expneurol.2024.114973] [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: 08/26/2024] [Revised: 09/18/2024] [Accepted: 09/21/2024] [Indexed: 09/28/2024]
Abstract
Serum/Glucocorticoid Regulated Kinase 1 (SGK1), a serine/threonine kinase, is ubiquitous across a wide range of tissues, orchestrating numerous signaling pathways and associated with various human diseases. SGK1 has been extensively explored in diverse types of immune and inflammatory diseases, cardiovascular disorders, as well as cancer metastasis. These studies link SGK1 to cellular proliferation, survival, metabolism, membrane transport, and drug resistance. Recently, increasing research has focused on SGK1's role in neurological disorders, including a variety of neurodegenerative diseases (e.g., Alzheimer's disease, Huntington's disease and Parkinson's disease), brain injuries (e.g., cerebral ischemia and traumatic brain injury), psychiatric conditions (e.g., depression and drug addiction). SGK1 is emerging as an increasingly compelling therapeutic target across the spectrum of neurological disorders, supported by the availability of several effective agents. However, the conclusions of many studies observing the prevalence and function of SGK1 in neurological disorders are contradictory, necessitating a review of the SGK1 research within neurological disorders. Herein, we review recent literature on SGK1's primary functions within the nervous system and its impacts within different neurological disorders. We summarize significant findings, identify research gaps, and outline possible future research directions based on the current understanding of SGK1 to help further progress the understanding and treatment of neurological disorders.
Collapse
Affiliation(s)
- Peyton Grace Howard
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA; Department of Neurology, Shreveport, Louisiana State University Health, LA, USA
| | - Peibin Zou
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA; Department of Neurology, Shreveport, Louisiana State University Health, LA, USA
| | - Yulan Zhang
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA; Department of Neurology, Shreveport, Louisiana State University Health, LA, USA
| | - Fang Huang
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA; Department of Neurology, Shreveport, Louisiana State University Health, LA, USA
| | - Vesna Tesic
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA; Department of Neurology, Shreveport, Louisiana State University Health, LA, USA
| | - Celeste Yin-Chieh Wu
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA; Department of Neurology, Shreveport, Louisiana State University Health, LA, USA.
| | - Reggie Hui-Chao Lee
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA; Department of Neurology, Shreveport, Louisiana State University Health, LA, USA; Department of Department of Cell Biology & Anatomy, Louisiana State University Health, Shreveport, LA, USA.
| |
Collapse
|
19
|
Li Y, Lu F, Zhang C, Xu H, Yang S. Dynamic susceptibility contrast-enhanced MRI with USPIO in evaluating angiogenesis of the peri-infarction zones in subacute ischemic stroke in a permanent middle cerebral artery occlusion rat model. Acta Radiol 2024; 65:1529-1539. [PMID: 39449316 DOI: 10.1177/02841851241290646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
BACKGROUND Dynamic susceptibility contrast-enhanced magnetic resonance imaging (DSC-MRI) can reflect the angiogenesis of ischemic stroke. PURPOSE To investigate the value of DSC-MRI with ultrasmall superparamagnetic particles of iron oxides (USPIO) in evaluating angiogenesis in the peri-infarction zones in subacute ischemic stroke in a permanent middle cerebral artery occlusion (pMCAO) rat model. MATERIAL AND METHODS A total of 21 Sprague-Dawley rats were randomly divided into the pMCAO and sham operation groups. Every rat in each group underwent DSC-MRI with USPIO at 3, 5, and 7 days. DSC-MRI parameters of the relative cerebral blood volume (rCBV), relative cerebral blood flow (rCBF), relative mean transit time (rMTT), and relative time to peak (rTTP) were measured, calculated, and compared among the different times. Sequential correlations were analyzed among the histopathological indexes with the microvascular density (MVD) and percentage of vascular area (%VA), the serum factors with vascular endothelial growth factor (VEGF), vascular cell adhesion molecule 1 (VCAM-1), and perfusion parameters, respectively. RESULTS The rCBV and rCBF in the peri-infarction area of pMCAO rats were significantly higher on day 7 than on day 3, whereas no significant changes in rMTT and rTTP were observed at 3, 5, and 7 days. Significantly positive correlations were found between rCBV and MVD, %VA, VEGF, VCAM-1, between rCBF and MVD, %VA, VEGF, and VCAM-1 at 3, 5, and 7 days in the pMCAO group. CONCLUSION The rCBV and rCBF deriving from USPIO-DSC may be potentially useful for evaluating the angiogenesis of the peri-infarction zones in the subacute phase of ischemic stroke.
Collapse
Affiliation(s)
- Yuanchao Li
- Department of Radiology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Fang Lu
- Department of Radiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Cheng Zhang
- Department of Radiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Huihui Xu
- Department of Radiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Shuohui Yang
- Department of Radiology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| |
Collapse
|
20
|
Testa AM, Vignozzi L, Corallo D, Aveic S, Viola A, Allegra M, Angioni R. Hypoxic Human Microglia Promote Angiogenesis Through Extracellular Vesicle Release. Int J Mol Sci 2024; 25:12508. [PMID: 39684220 DOI: 10.3390/ijms252312508] [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: 11/09/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
Microglia, the brain-resident immune cells, orchestrate neuroinflammatory responses and are crucial in the progression of neurological diseases, including ischemic stroke (IS), which accounts for approximately 85% of all strokes worldwide. Initially deemed detrimental, microglial activation has been shown to perform protective functions in the ischemic brain. Besides their effects on neurons, microglia play a role in promoting post-ischemic angiogenesis, a pivotal step for restoring oxygen and nutrient supply. However, the molecular mechanisms underlying microglia-endothelial cell interactions remain largely unresolved, particularly in humans. Using both in vitro and in vivo models, we investigated the angiogenic signature and properties of extracellular vesicles (EVs) released by human microglia upon hypoxia-reperfusion stimulation. EVs were isolated and characterized in terms of their size, concentration, and protein content. Their angiogenic potential was evaluated using endothelial cell assays and a zebrafish xenograft model. The in vivo effects were further assessed in a mouse model of ischemic stroke. Our findings identified key proteins orchestrating the pro-angiogenic functions of human microglial EVs under hypoxic conditions. In vitro assays demonstrated that hypoxic EVs (hypEVs) promoted endothelial cell migration and tube formation. In vivo, hypEVs induced vessel sprouting in zebrafish and increased microvessel density in the perilesional area of mice following ischemic stroke.
Collapse
Affiliation(s)
- Alessandra Maria Testa
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
- Laboratory of Immunity, Inflammation and Angiogenesis, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35127 Padua, Italy
| | - Livia Vignozzi
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
| | - Diana Corallo
- Laboratory of Target Discovery and Biology of Neuroblastoma, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35127 Padua, Italy
| | - Sanja Aveic
- Laboratory of Target Discovery and Biology of Neuroblastoma, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35127 Padua, Italy
| | - Antonella Viola
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
| | - Manuela Allegra
- Laboratory of Neuronal Circuits in Developmental Disorders, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35127 Padua, Italy
- Neuroscience Institute, National Research Council, 35131 Padua, Italy
| | - Roberta Angioni
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
- Laboratory of Immunity, Inflammation and Angiogenesis, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35127 Padua, Italy
| |
Collapse
|
21
|
Ma L, Ge P, Zeng C, Liu C, Yin Z, Ya X, Zhai Y, He Q, Li J, Ye X, Zhang Q, Wang R, Zhang D, Zhang Y, Zhao J. Prognostic value of morphology and hemodynamics in moyamoya disease for long-term outcomes and disease progression. Sci Rep 2024; 14:28182. [PMID: 39548256 PMCID: PMC11568140 DOI: 10.1038/s41598-024-79608-4] [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: 07/09/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024] Open
Abstract
To explore the relationship between morphological and hemodynamic parameters, baseline characteristics, and long-term outcomes in patients with moyamoya disease (MMD) using a computational fluid dynamics model. We retrospectively reviewed 129 patients at Beijing Tiantan hospital between July 2020 and December 2021. Perioperative clinical variables and Suzuki stage were recorded. Logistic regression analysis was employed to identify the risk factors for unfavorable long-term outcomes. The association between morphological, CT perfusion parameters, hemodynamic parameters and the Suzuki stage, clinical variables of MMD was also analyzed. Patients with high relative Wall Shear Stress (rWSS) were older and had more cases with higher Suzuki stage and worse follow-up mRS scores (p < 0.05). High rWSS at the terminal ICA and diabetes mellitus were identified as independent predictors of unfavorable long-term outcomes [OR = 3.039(1.191-7.754), p = 0.020; OR = 3.164(1.141-8.723), p = 0.027, respectively]. ROC analysis demonstrated that predictive models incorporating rWSS improved AUC values, with the highest AUC in Model 2 (AUC = 0.889). High rWSS was significantly associated with future TIA and stroke events (p = 0.032). We speculated that high rWSS and diabetes mellitus were independent risk factors for unfavorable long-term outcomes in patients with MMD. rWSS and morphological parameters are crucial for predicting MMD progression and understanding its pathogenesis.
Collapse
Affiliation(s)
- Long Ma
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Peicong Ge
- China National Clinical Research Center for Neurological Diseases, Beijing, China.
| | - Chaofan Zeng
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Chenglong Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Zihan Yin
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Xiaolong Ya
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yuanren Zhai
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Qiheng He
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Junsheng Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Xun Ye
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Qian Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Rong Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Dong Zhang
- Department of Neurosurgery, Beijing Hospital, Beijing, China
| | - Yan Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Jizong Zhao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.
- China National Clinical Research Center for Neurological Diseases, Beijing, China.
| |
Collapse
|
22
|
Williamson JN, Mulyana B, Peng RHT, Jain S, Hassaneen W, Miranpuri A, Yang Y. How the Somatosensory System Adapts to the Motor Change in Stroke: A Hemispheric Shift? Med Hypotheses 2024; 192:111487. [PMID: 39525858 PMCID: PMC11542668 DOI: 10.1016/j.mehy.2024.111487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Previous studies found that post-stroke motor impairments are associated with damage to the lesioned corticospinal tract and a maladaptive increase in indirect contralesional motor pathways. How the somatosensory system adapts to the change in the use of motor pathways and the role of adaptive sensory feedback to the abnormal movement control of the paretic arm remains largely unknown. We hypothesize that following a unilateral stroke, there is an adaptive hemispheric shift of somatosensory processing toward the contralesional sensorimotor areas to provide sensory feedback support to the contralesional indirect motor pathways. This research could provide new insights related to somatosensory reorganization after stroke, which could enrich future hypothesis-driven therapeutic rehabilitation strategies from a sensory or sensory-motor perspective. Understanding how somatosensory information shifts may provide a target for a novel method to therapeutically prevent and mitigate the emergence and expression of upper limb motor impairments, following a stroke.
Collapse
Affiliation(s)
- Jordan N. Williamson
- University of Illinois Urbana-Champaign, Grainger College of Engineering, Department of Bioengineering, Urbana, IL, USA
| | - Beni Mulyana
- University of Illinois Urbana-Champaign, Grainger College of Engineering, Department of Bioengineering, Urbana, IL, USA
- Carle Foundation Hospital, Stephenson Family Clinical Research Institute, Clinical Imaging Research Center, Urbana, IL, USA
| | - Rita Huan-Ting Peng
- University of Illinois Urbana-Champaign, Grainger College of Engineering, Department of Bioengineering, Urbana, IL, USA
- Carle Foundation Hospital, Stephenson Family Clinical Research Institute, Clinical Imaging Research Center, Urbana, IL, USA
| | - Sanjiv Jain
- Carle Foundation Hospital, Dr. Elizabeth Hosick Rehabilitation Center, Urbana, IL, USA
- Carle Illinois College of Medicine, Urbana, IL, USA
| | - Wael Hassaneen
- Carle Illinois College of Medicine, Urbana, IL, USA
- Carle Foundation Hospital, Neuroscience Institute, Urbana, IL, USA
| | - Amrendra Miranpuri
- Carle Illinois College of Medicine, Urbana, IL, USA
- Carle Foundation Hospital, Neuroscience Institute, Urbana, IL, USA
| | - Yuan Yang
- University of Illinois Urbana-Champaign, Grainger College of Engineering, Department of Bioengineering, Urbana, IL, USA
- Carle Foundation Hospital, Stephenson Family Clinical Research Institute, Clinical Imaging Research Center, Urbana, IL, USA
- University of Illinois Urbana-Champaign, Beckman Institute for Advanced Science and Technology, Urbana, IL, USA
- Northwestern University, Feinberg School of Medicine Department of Physical Therapy and Human Movement Sciences, Chicago, IL, USA
| |
Collapse
|
23
|
Sato Y, Li Y, Kato Y, Kanoke A, Sun JY, Nishijima Y, Wang RK, Stryker M, Endo H, Liu J. Type 2 diabetes remodels collateral circulation and promotes leukocyte adhesion following ischemic stroke. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.23.619748. [PMID: 39484619 PMCID: PMC11526934 DOI: 10.1101/2024.10.23.619748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Type 2 diabetes mellitus (T2DM) is associated with impaired leptomeningeal collateral compensation and poor stroke outcome. Neutrophils tethering and rolling on endothelium after stroke can also independently reduce flow velocity. However, the chronology and topological changes in collateral circulation in T2DM is not yet defined. Here, we describe the spatial and temporal blood flow dynamics and vessel remodeling in pial arteries and veins and leukocyte-endothelial adhesion following middle cerebral artery (MCA) stroke using two-photon microscopy in awake control and T2DM mice. Relative to control mice prior to stroke, T2DM mice already exhibited smaller pial vessels with reduced flow velocity. Following stroke, T2DM mice displayed persistently reduced blood flow in pial arteries and veins, resulting in a poor recovery of downstream penetrating arterial flow and a sustained deficit in microvascular flow. There was also persistent increase of leukocyte adhesion to the endothelium of veins, coincided with elevated neutrophils infiltration into brain parenchyma in T2DM mice compared to control mice after stroke. Our data suggest that T2DM-induced increase in chronic inflammation may contribute to the remodeling of leptomeningeal collateral circulation and the observed hemodynamics deficiency that potentiates poor stroke outcome.
Collapse
Affiliation(s)
- Yoshimichi Sato
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- SFVAMC, San Francisco, CA, USA
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Yuandong Li
- Department of Bioengineering, College of Engineering and School of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Yuya Kato
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- SFVAMC, San Francisco, CA, USA
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Atsushi Kanoke
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- SFVAMC, San Francisco, CA, USA
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Jennifer Y Sun
- Department of Physiology, University of California San Francisco, San Francisco, CA, USA
- University College London, Institute of Ophthalmology, London, UK
| | - Yasuo Nishijima
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- SFVAMC, San Francisco, CA, USA
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Ruikang K. Wang
- Department of Bioengineering, College of Engineering and School of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Michael Stryker
- Department of Physiology, University of California San Francisco, San Francisco, CA, USA
| | - Hidenori Endo
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Jialing Liu
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- SFVAMC, San Francisco, CA, USA
| |
Collapse
|
24
|
Yuan M, Tang Y, Huang T, Ke L, Huang E. In situ direct reprogramming of astrocytes to neurons via polypyrimidine tract-binding protein 1 knockdown in a mouse model of ischemic stroke. Neural Regen Res 2024; 19:2240-2248. [PMID: 38488558 PMCID: PMC11034579 DOI: 10.4103/1673-5374.390957] [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: 05/06/2023] [Revised: 08/09/2023] [Accepted: 10/16/2023] [Indexed: 04/24/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202410000-00025/figure1/v/2024-02-06T055622Z/r/image-tiff In situ direct reprogramming technology can directly convert endogenous glial cells into functional neurons in vivo for central nervous system repair. Polypyrimidine tract-binding protein 1 (PTB) knockdown has been shown to reprogram astrocytes to functional neurons in situ. In this study, we used AAV-PHP.eB-GFAP-shPTB to knockdown PTB in a mouse model of ischemic stroke induced by endothelin-1, and investigated the effects of GFAP-shPTB-mediated direct reprogramming to neurons. Our results showed that in the mouse model of ischemic stroke, PTB knockdown effectively reprogrammed GFAP-positive cells to neurons in ischemic foci, restored neural tissue structure, reduced inflammatory response, and improved behavioral function. These findings validate the effectiveness of in situ transdifferentiation of astrocytes, and suggest that the approach may be a promising strategy for stroke treatment.
Collapse
Affiliation(s)
- Meng Yuan
- Key Laboratory of Brain Aging and Neurodegenerative Diseases of Fujian Province, Fujian Medical University, Fuzhou, Fujian Province, China
- Department of Human Anatomy, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Yao Tang
- Key Laboratory of Brain Aging and Neurodegenerative Diseases of Fujian Province, Fujian Medical University, Fuzhou, Fujian Province, China
- Scientific Research Center, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Tianwen Huang
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Fujian Key Laboratory of Vascular Aging, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Lining Ke
- Key Laboratory of Brain Aging and Neurodegenerative Diseases of Fujian Province, Fujian Medical University, Fuzhou, Fujian Province, China
- Department of Human Anatomy, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China
| | - En Huang
- Key Laboratory of Brain Aging and Neurodegenerative Diseases of Fujian Province, Fujian Medical University, Fuzhou, Fujian Province, China
- Scientific Research Center, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China
| |
Collapse
|
25
|
Zedde M, Pascarella R. The Cerebrovascular Side of Plasticity: Microvascular Architecture across Health and Neurodegenerative and Vascular Diseases. Brain Sci 2024; 14:983. [PMID: 39451997 PMCID: PMC11506257 DOI: 10.3390/brainsci14100983] [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: 09/03/2024] [Revised: 09/24/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
The delivery of nutrients to the brain is provided by a 600 km network of capillaries and microvessels. Indeed, the brain is highly energy demanding and, among a total amount of 100 billion neurons, each neuron is located just 10-20 μm from a capillary. This vascular network also forms part of the blood-brain barrier (BBB), which maintains the brain's stable environment by regulating chemical balance, immune cell transport, and blocking toxins. Typically, brain microvascular endothelial cells (BMECs) have low turnover, indicating a stable cerebrovascular structure. However, this structure can adapt significantly due to development, aging, injury, or disease. Temporary neural activity changes are managed by the expansion or contraction of arterioles and capillaries. Hypoxia leads to significant remodeling of the cerebrovascular architecture and pathological changes have been documented in aging and in vascular and neurodegenerative conditions. These changes often involve BMEC proliferation and the remodeling of capillary segments, often linked with local neuronal changes and cognitive function. Cerebrovascular plasticity, especially in arterioles, capillaries, and venules, varies over different time scales in development, health, aging, and diseases. Rapid changes in cerebral blood flow (CBF) occur within seconds due to increased neural activity. Prolonged changes in vascular structure, influenced by consistent environmental factors, take weeks. Development and aging bring changes over months to years, with aging-associated plasticity often improved by exercise. Injuries cause rapid damage but can be repaired over weeks to months, while neurodegenerative diseases cause slow, varied changes over months to years. In addition, if animal models may provide useful and dynamic in vivo information about vascular plasticity, humans are more complex to investigate and the hypothesis of glymphatic system together with Magnetic Resonance Imaging (MRI) techniques could provide useful clues in the future.
Collapse
Affiliation(s)
- Marialuisa Zedde
- Neurology Unit, Stroke Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Viale Risorgimento 80, 42123 Reggio Emilia, Italy
| | - Rosario Pascarella
- Neuroradiology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Viale Risorgimento 80, 42123 Reggio Emilia, Italy;
| |
Collapse
|
26
|
Wang J, Xiong T, Wu Q, Qin X. Integrated Strategies for Targeting Arteriogenesis and Angiogenesis After Stroke. Transl Stroke Res 2024:10.1007/s12975-024-01291-4. [PMID: 39225878 DOI: 10.1007/s12975-024-01291-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/29/2024] [Accepted: 08/17/2024] [Indexed: 09/04/2024]
Abstract
The interdependence between arteriogenesis and angiogenesis is crucial for enhancing perfusion by synchronously improving leptomeningeal collaterals (LMCs) and microvascular networks after stroke. However, current approaches often focus on promoting arteriogenesis and angiogenesis separately, neglecting the potential synergistic benefits of targeting both processes simultaneously. Therefore, it is imperative to consider both arteriogenesis and angiogenesis as integral and complementary strategies for post-stroke revascularization. To gain a deeper understanding of their relationships after stroke and to facilitate the development of targeted revascularization strategies, we compared them based on their timescale, space, and pathophysiology. The temporal differences in the occurrence of arteriogenesis and angiogenesis allow them to restore blood flow at different stages after stroke. The spatial differences in the effects of arteriogenesis and angiogenesis enable them to specifically target the ischemic penumbra and core infarct region. Additionally, the endothelial cell, as the primary effector cell in their pathophysiological processes, is promising target for enhancing both. Therefore, we provide an overview of key signals that regulate endothelium-mediated arteriogenesis and angiogenesis. Finally, we summarize current therapeutic strategies that involve these signals to promote both processes after stroke, with the aim of inspiring future therapeutic advances in revascularization.
Collapse
Affiliation(s)
- Jing Wang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Taoying Xiong
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Qisi Wu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Xinyue Qin
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
27
|
Preka E, Lastra Romero A, Sun Y, Onetti Vilalta Y, Seitz T, Fragkopoulou A, Betsholtz C, Osman AM, Blomgren K. Rapid and robust isolation of microglia and vascular cells from brain subregions for integrative single-cell analyses. Heliyon 2024; 10:e35838. [PMID: 39211933 PMCID: PMC11357767 DOI: 10.1016/j.heliyon.2024.e35838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 06/20/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Cell isolation protocols from brain tissue include prolonged ex vivo processing durations, rendering them suboptimal for transcriptomic studies. Particularly for microglia and vascular cells, current isolation methods produce lower yields, necessitating addition of an enrichment step, and use of large tissue volumes - in most cases whole brain tissue - to obtain sufficient yields. Here, we developed a simple, rapid, and reproducible cell isolation method for generating single-cell suspensions from micro-dissected brain regions, enriched for microglia and vascular cells, without an enrichment step. Cells isolated using this method are suitable for molecular profiling studies using 10 × Genomics Chromium single-cell RNA sequencing with high reproducibility. Our method is valuable for longitudinal unbiased molecular profiling of microglia and vascular cells within different brain regions, spanning multiple time points across physiological development or disease progression.
Collapse
Affiliation(s)
- Efthalia Preka
- Department of Women's and Children's Health, Biomedicum A4, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Alejandro Lastra Romero
- Department of Women's and Children's Health, Biomedicum A4, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Ying Sun
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden
| | - Yara Onetti Vilalta
- Department of Women's and Children's Health, Biomedicum A4, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Thea Seitz
- Department of Women's and Children's Health, Biomedicum A4, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Adamantia Fragkopoulou
- Department of Women's and Children's Health, Biomedicum A4, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden
- Department of Medicine Huddinge, Campus Flemingsberg, Neo, 141 57, Huddinge, Sweden
| | - Ahmed M. Osman
- Department of Women's and Children's Health, Biomedicum A4, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Klas Blomgren
- Department of Women's and Children's Health, Biomedicum A4, Karolinska Institutet, 171 77, Stockholm, Sweden
- Pediatric Oncology, Karolinska University Hospital, 171 64, Stockholm, Sweden
| |
Collapse
|
28
|
Buizza C, Enström A, Carlsson R, Paul G. The Transcriptional Landscape of Pericytes in Acute Ischemic Stroke. Transl Stroke Res 2024; 15:714-728. [PMID: 37378751 PMCID: PMC11226519 DOI: 10.1007/s12975-023-01169-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/07/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023]
Abstract
The current treatment options for ischemic stroke aim to achieve reperfusion but are time critical. Novel therapeutic approaches that can be given beyond the limited time window of 3-4.5 h are still an unmet need to be addressed to improve stroke outcomes. The lack of oxygen and glucose in the area of ischemic injury initiates a pathological cascade leading to blood-brain barrier (BBB) breakdown, inflammation, and neuronal cell death, a process that may be intercepted to limit stroke progression. Pericytes located at the blood/brain interface are one of the first responders to hypoxia in stroke and therefore a potential target cell for early stroke interventions. Using single-cell RNA sequencing in a mouse model of permanent middle cerebral artery occlusion, we investigated the temporal differences in transcriptomic signatures in pericytes at 1, 12, and 24 h after stroke. Our results reveal a stroke-specific subcluster of pericytes that is present at 12 and 24 h and characterized by the upregulation of genes mainly related to cytokine signaling and immune response. This study identifies temporal transcriptional changes in the acute phase of ischemic stroke that reflect the early response of pericytes to the ischemic insult and its secondary consequences and may constitute potential future therapeutic targets.
Collapse
Affiliation(s)
- Carolina Buizza
- Translational Neurology Group, Department of Clinical Science, Lund University, 22184, Lund, Sweden
| | - Andreas Enström
- Translational Neurology Group, Department of Clinical Science, Lund University, 22184, Lund, Sweden
| | - Robert Carlsson
- Translational Neurology Group, Department of Clinical Science, Lund University, 22184, Lund, Sweden
| | - Gesine Paul
- Translational Neurology Group, Department of Clinical Science, Lund University, 22184, Lund, Sweden.
- Department of Neurology, Scania University Hospital, 22185, Lund, Sweden.
- Wallenberg Centre for Molecular Medicine, Lund University, 22184, Lund, Sweden.
| |
Collapse
|
29
|
Xie Z, Zeinstra N, Kirby MA, Le NM, Murry CE, Zheng Y, Wang RK. Quantifying Microvascular Structure in Healthy and Infarcted Rat Hearts Using Optical Coherence Tomography Angiography. IEEE TRANSACTIONS ON MEDICAL IMAGING 2024; 43:2878-2887. [PMID: 38568757 PMCID: PMC11341234 DOI: 10.1109/tmi.2024.3381934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
Abstract
Myocardial infarction (MI) is a life-threatening medical emergency resulting in coronary microvascular dysregulation and heart muscle damage. One of the primary characteristics of MI is capillary loss, which plays a significant role in the progression of this cardiovascular condition. In this study, we utilized optical coherence tomography angiography (OCTA) to image coronary microcirculation in fixed rat hearts, aiming to analyze coronary microvascular impairment post-infarction. Various angiographic metrics are presented to quantify vascular features, including the vessel area density, vessel complexity index, vessel tortuosity index, and flow impairment. Pathological differences identified from OCTA analysis are corroborated with histological analysis. The quantitative assessments reveal a significant decrease in microvascular density in the capillary-sized vessels and an enlargement for the arteriole/venule-sized vessels. Further, microvascular tortuosity and complexity exhibit an increase after myocardial infarction. The results underscore the feasibility of using OCTA to offer qualitative microvascular details and quantitative metrics, providing insights into coronary vascular network remodeling during disease progression and response to therapy.
Collapse
|
30
|
Hu B, Pei J, Wan C, Liu S, Xu Z, Zou Y, Li Z, Tang Z. Mechanisms of Postischemic Stroke Angiogenesis: A Multifaceted Approach. J Inflamm Res 2024; 17:4625-4646. [PMID: 39045531 PMCID: PMC11264385 DOI: 10.2147/jir.s461427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 06/26/2024] [Indexed: 07/25/2024] Open
Abstract
Ischemic stroke constitutes a significant global health care challenge, and a comprehensive understanding of its recovery mechanisms is imperative for the development of innovative therapeutic strategies. Angiogenesis, a pivotal element of ischemic tissue repair, facilitates the restoration of blood flow to damaged regions, thereby promoting neuronal regeneration and functional recovery. Nevertheless, the mechanisms underlying postischemic stroke angiogenesis remain incompletely elucidated. This review meticulously examines the constituents of the neurovascular unit, ion channels, molecular mediators, and signaling pathways implicated in angiogenesis following stroke. Furthermore, it delves into prospective therapeutic strategies informed by these factors. Our objective is to provide detailed and exhaustive information on the intricate mechanisms governing postischemic stroke angiogenesis, thus providing a robust scientific foundation for the advancement of novel neurorepair therapies.
Collapse
Affiliation(s)
- Bin Hu
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Jingchun Pei
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Cheng Wan
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
- Department of Medical Imaging, The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Shuangshuang Liu
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Zhe Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, People’s Republic of China
- School of Basic Medical Sciences, Qujing Medical College, Qujing, People’s Republic of China
| | - Yongwei Zou
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Zhigao Li
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Zhiwei Tang
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| |
Collapse
|
31
|
Sánchez-Puebla L, López-Cuenca I, Salobrar-García E, González-Jiménez M, Arias-Vázquez A, Matamoros JA, Ramírez AI, Fernández-Albarral JA, Elvira-Hurtado L, Saido TC, Saito T, Nieto-Vaquero C, Cuartero MI, Moro MA, Salazar JJ, de Hoz R, Ramírez JM. Retinal Vascular and Structural Changes in the Murine Alzheimer's APPNL-F/NL-F Model from 6 to 20 Months. Biomolecules 2024; 14:828. [PMID: 39062542 PMCID: PMC11274728 DOI: 10.3390/biom14070828] [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: 05/07/2024] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Alzheimer's disease (AD) may manifest retinal changes preceding brain pathology. A transversal case-control study utilized spectral-domain OCT angiography (SD-OCTA) and Angio-Tool software 0.6a to assess retinal vascular structures and OCT for inner and outer retina thickness in the APPNL-F/NL-F AD model at 6, 9, 12, 15, 17, and 20 months old. Comparisons to age-matched wild type (WT) were performed. The analysis focused on the three vascular plexuses using AngiooTool and on retinal thickness, which was represented with the Early Treatment Diabetic Retinopathy Study (ETDRS) sectors. Compared to WT, the APPNL-F/NL-F group exhibited both vascular and structural changes as early as 6 months persisting and evolving at 15, 17, and 20 months. Significant vascular alterations, principally in the superficial vascular complex (SVC), were observed. There was a significant decrease in the vessel area and the total vessel length in SVC, intermediate, and deep capillary plexus. The inner retina in the APPNL-F/NL-F group predominantly decreased in thickness while the outer retina showed increased thickness in most analyzed time points compared to the control group. There are early vascular and structural retinal changes that precede the cognitive changes, which appear at later stages. Therefore, the natural history of the APPNL-F/NL-F model may be more similar to human AD than other transgenic models.
Collapse
Affiliation(s)
- Lidia Sánchez-Puebla
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (I.L.-C.); (E.S.-G.); (M.G.-J.); (A.A.-V.); (J.A.M.); (A.I.R.); (J.A.F.-A.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Inés López-Cuenca
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (I.L.-C.); (E.S.-G.); (M.G.-J.); (A.A.-V.); (J.A.M.); (A.I.R.); (J.A.F.-A.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - Elena Salobrar-García
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (I.L.-C.); (E.S.-G.); (M.G.-J.); (A.A.-V.); (J.A.M.); (A.I.R.); (J.A.F.-A.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - María González-Jiménez
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (I.L.-C.); (E.S.-G.); (M.G.-J.); (A.A.-V.); (J.A.M.); (A.I.R.); (J.A.F.-A.); (L.E.-H.); (J.J.S.)
| | - Alberto Arias-Vázquez
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (I.L.-C.); (E.S.-G.); (M.G.-J.); (A.A.-V.); (J.A.M.); (A.I.R.); (J.A.F.-A.); (L.E.-H.); (J.J.S.)
| | - José A. Matamoros
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (I.L.-C.); (E.S.-G.); (M.G.-J.); (A.A.-V.); (J.A.M.); (A.I.R.); (J.A.F.-A.); (L.E.-H.); (J.J.S.)
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - Ana I. Ramírez
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (I.L.-C.); (E.S.-G.); (M.G.-J.); (A.A.-V.); (J.A.M.); (A.I.R.); (J.A.F.-A.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - José A. Fernández-Albarral
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (I.L.-C.); (E.S.-G.); (M.G.-J.); (A.A.-V.); (J.A.M.); (A.I.R.); (J.A.F.-A.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - Lorena Elvira-Hurtado
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (I.L.-C.); (E.S.-G.); (M.G.-J.); (A.A.-V.); (J.A.M.); (A.I.R.); (J.A.F.-A.); (L.E.-H.); (J.J.S.)
| | - Takaomi C. Saido
- Laboratory for Proteolytic Neuroscience, Brain Science Institute, RIKEN, Wako 351-0198, Japan;
| | - Takashi Saito
- Institute of Brain Science, Faculty of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan;
| | - Carmen Nieto-Vaquero
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Neurovascular Pathophysiology, Cardiovascular Risk Factor and Brain Function Programme, 28029 Madrid, Spain; (C.N.-V.); (M.A.M.)
- Hospital 12 de Octubre Research Institute (i + 12), 28029 Madrid, Spain;
- University Institute for Research in Neurochemistry, Complutense University of Madrid (UCM), 28040 Madrid, Spain
| | - María I. Cuartero
- Hospital 12 de Octubre Research Institute (i + 12), 28029 Madrid, Spain;
- University Institute for Research in Neurochemistry, Complutense University of Madrid (UCM), 28040 Madrid, Spain
- Department of Pharmacology and Toxicology, Faculty of Medicine, Complutense University of Madrid (UCM), 28040 Madrid, Spain
| | - María A. Moro
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Neurovascular Pathophysiology, Cardiovascular Risk Factor and Brain Function Programme, 28029 Madrid, Spain; (C.N.-V.); (M.A.M.)
| | - Juan J. Salazar
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (I.L.-C.); (E.S.-G.); (M.G.-J.); (A.A.-V.); (J.A.M.); (A.I.R.); (J.A.F.-A.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - Rosa de Hoz
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (I.L.-C.); (E.S.-G.); (M.G.-J.); (A.A.-V.); (J.A.M.); (A.I.R.); (J.A.F.-A.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - José M. Ramírez
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (I.L.-C.); (E.S.-G.); (M.G.-J.); (A.A.-V.); (J.A.M.); (A.I.R.); (J.A.F.-A.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| |
Collapse
|
32
|
Alhadidi QM, Bahader GA, Arvola O, Kitchen P, Shah ZA, Salman MM. Astrocytes in functional recovery following central nervous system injuries. J Physiol 2024; 602:3069-3096. [PMID: 37702572 PMCID: PMC11421637 DOI: 10.1113/jp284197] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 08/07/2023] [Indexed: 09/14/2023] Open
Abstract
Astrocytes are increasingly recognised as partaking in complex homeostatic mechanisms critical for regulating neuronal plasticity following central nervous system (CNS) insults. Ischaemic stroke and traumatic brain injury are associated with high rates of disability and mortality. Depending on the context and type of injury, reactive astrocytes respond with diverse morphological, proliferative and functional changes collectively known as astrogliosis, which results in both pathogenic and protective effects. There is a large body of research on the negative consequences of astrogliosis following brain injuries. There is also growing interest in how astrogliosis might in some contexts be protective and help to limit the spread of the injury. However, little is known about how astrocytes contribute to the chronic functional recovery phase following traumatic and ischaemic brain insults. In this review, we explore the protective functions of astrocytes in various aspects of secondary brain injury such as oedema, inflammation and blood-brain barrier dysfunction. We also discuss the current knowledge on astrocyte contribution to tissue regeneration, including angiogenesis, neurogenesis, synaptogenesis, dendrogenesis and axogenesis. Finally, we discuss diverse astrocyte-related factors that, if selectively targeted, could form the basis of astrocyte-targeted therapeutic strategies to better address currently untreatable CNS disorders.
Collapse
Affiliation(s)
- Qasim M Alhadidi
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, CA, USA
- Department of Pharmacy, Al-Yarmok University College, Diyala, Iraq
| | - Ghaith A Bahader
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
| | - Oiva Arvola
- Division of Anaesthesiology, Jorvi Hospital, Department of Anaesthesiology, Intensive Care and Pain Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Stem Cells and Metabolism Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Philip Kitchen
- College of Health and Life Sciences, Aston University, Birmingham, UK
| | - Zahoor A Shah
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
| | - Mootaz M Salman
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
- Kavli Institute for NanoScience Discovery, University of Oxford, Oxford, UK
| |
Collapse
|
33
|
Amin N, Abbasi IN, Wu F, Shi Z, Sundus J, Badry A, Yuan X, Zhao BX, Pan J, Mi XD, Luo Y, Geng Y, Fang M. The Janus face of HIF-1α in ischemic stroke and the possible associated pathways. Neurochem Int 2024; 177:105747. [PMID: 38657682 DOI: 10.1016/j.neuint.2024.105747] [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: 10/30/2023] [Revised: 03/01/2024] [Accepted: 04/19/2024] [Indexed: 04/26/2024]
Abstract
Stroke is the most devastating disease, causing paralysis and eventually death. Many clinical and experimental trials have been done in search of a new safe and efficient medicine; nevertheless, scientists have yet to discover successful remedies that are also free of adverse effects. This is owing to the variability in intensity, localization, medication routes, and each patient's immune system reaction. HIF-1α represents the modern tool employed to treat stroke diseases due to its functions: downstream genes such as glucose metabolism, angiogenesis, erythropoiesis, and cell survival. Its role can be achieved via two downstream EPO and VEGF strongly related to apoptosis and antioxidant processes. Recently, scientists paid more attention to drugs dealing with the HIF-1 pathway. This review focuses on medicines used for ischemia treatment and their potential HIF-1α pathways. Furthermore, we discussed the interaction between HIF-1α and other biological pathways such as oxidative stress; however, a spotlight has been focused on certain potential signalling contributed to the HIF-1α pathway. HIF-1α is an essential regulator of oxygen balance within cells which affects and controls the expression of thousands of genes related to sustaining homeostasis as oxygen levels fluctuate. HIF-1α's role in ischemic stroke strongly depends on the duration and severity of brain damage after onset. HIF-1α remains difficult to investigate, particularly in ischemic stroke, due to alterations in the acute and chronic phases of the disease, as well as discrepancies between the penumbra and ischemic core. This review emphasizes these contrasts and analyzes the future of this intriguing and demanding field.
Collapse
Affiliation(s)
- Nashwa Amin
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China; Department of Zoology, Faculty of Science, Aswan University, Egypt; Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Irum Naz Abbasi
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Fei Wu
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Zongjie Shi
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Javaria Sundus
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Azhar Badry
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Xia Yuan
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Bing-Xin Zhao
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Jie Pan
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Xiao-Dan Mi
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yuhuan Luo
- Department of Pediatrics, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Geng
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Marong Fang
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China; Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| |
Collapse
|
34
|
Norton CE, Shaw RL, Safa, Dockery B, Domeier TL, Segal SS. Advanced age and female sex protect cerebral arteries from mitochondrial depolarization and apoptosis during acute oxidative stress. Aging Cell 2024; 23:e14110. [PMID: 38380477 PMCID: PMC11113258 DOI: 10.1111/acel.14110] [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: 07/03/2023] [Revised: 01/26/2024] [Accepted: 01/30/2024] [Indexed: 02/22/2024] Open
Abstract
Aging increases reactive oxygen species (ROS) which can impair vascular function and contribute to brain injury. However, aging can also promote resilience to acute oxidative stress. Therefore, we tested the hypothesis that advanced age protects smooth muscle cells (SMCs) and endothelial cells (ECs) of posterior cerebral arteries (PCAs; diameter, ∼80 μm) during exposure to H2O2. PCAs from young (4-6 months) and old (20-26 months) male and female C57BL/6 mice were isolated and pressurized (~70 mm Hg) to evaluate cell death, mitochondrial membrane potential (ΔΨm), ROS production, and [Ca2+]i in response to H2O2 (200 μM, 50 min). SMC death and ΔΨm depolarization were greater in PCAs from males vs. females. Aging increased ROS in PCAs from both sexes but increased SMC resilience to death only in males. Inhibiting TRPV4 channels with HC-067047 (1 μM) or Src kinases with SU6656 (10 μM) reduced Ca2+ entry and SMC death to H2O2 most effectively in PCAs from young males. Activating TRPV4 channels with GSK1016790A (50 nM) evoked greater Ca2+ influx in SMCs and ECs of PCAs from young vs. old mice but did not induce cell death. However, when combined with H2O2, TRPV4 activation exacerbated EC death. Activating Src kinases with spermidine (100 μM) increased Ca2+ influx in PCAs from males vs. females with minimal cell death. We conclude that in males, chronic oxidative stress during aging increases the resilience of cerebral arteries, which contrasts with inherent protection in females. Findings implicate TRP channels and Src kinases as targets to limit vascular damage to acute oxidative injury.
Collapse
Affiliation(s)
- Charles E. Norton
- Department of Medical Pharmacology and PhysiologyUniversity of MissouriColumbiaMissouriUSA
| | - Rebecca L. Shaw
- Department of Medical Pharmacology and PhysiologyUniversity of MissouriColumbiaMissouriUSA
| | - Safa
- Department of Medical Pharmacology and PhysiologyUniversity of MissouriColumbiaMissouriUSA
| | - Beyoncé Dockery
- Department of Medical Pharmacology and PhysiologyUniversity of MissouriColumbiaMissouriUSA
| | - Timothy L. Domeier
- Department of Medical Pharmacology and PhysiologyUniversity of MissouriColumbiaMissouriUSA
| | - Steven S. Segal
- Department of Medical Pharmacology and PhysiologyUniversity of MissouriColumbiaMissouriUSA
- Dalton Cardiovascular Research CenterColumbiaMissouriUSA
- Department of Biomedical SciencesUniversity of MissouriColumbiaMissouriUSA
- Department of Biomedical, Biological and Chemical EngineeringUniversity of MissouriColumbiaMissouriUSA
- Department of Nutrition and Exercise PhysiologyUniversity of MissouriColumbiaMissouriUSA
| |
Collapse
|
35
|
Zhang Q, Huang S, Liu X, Wang W, Zhu Z, Chen L. Innovations in Breaking Barriers: Liposomes as Near-Perfect Drug Carriers in Ischemic Stroke Therapy. Int J Nanomedicine 2024; 19:3715-3735. [PMID: 38681090 PMCID: PMC11046314 DOI: 10.2147/ijn.s462194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 04/13/2024] [Indexed: 05/01/2024] Open
Abstract
Liposomes, noted for their tunable particle size, surface customization, and varied drug delivery capacities, are increasingly acknowledged in therapeutic applications. These vesicles exhibit surface flexibility, enabling the incorporation of targeting moieties or peptides to achieve specific targeting and avoid lysosomal entrapment. Internally, their adaptable architecture permits the inclusion of a broad spectrum of drugs, contingent on their solubility characteristics. This study thoroughly reviews liposome fabrication, surface modifications, and drug release mechanisms post-systemic administration, with a particular emphasis on drugs crossing the blood-brain barrier (BBB) to address lesions. Additionally, the review delves into recent developments in the use of liposomes in ischemic stroke models, offering a comparative evaluation with other nanocarriers like exosomes and nano-micelles, thereby facilitating their clinical advancement.
Collapse
Affiliation(s)
- Qiankun Zhang
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Songze Huang
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Xiaowen Liu
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Wei Wang
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Zhihan Zhu
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Lukui Chen
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| |
Collapse
|
36
|
Tudor T, Spinazzi EF, Alexander JE, Mandigo GK, Lavine SD, Grinband J, Connolly ES. Progressive microvascular failure in acute ischemic stroke: A systematic review, meta-analysis, and time-course analysis. J Cereb Blood Flow Metab 2024; 44:192-208. [PMID: 38016953 PMCID: PMC10993872 DOI: 10.1177/0271678x231216766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 09/01/2023] [Accepted: 10/02/2023] [Indexed: 11/30/2023]
Abstract
This systematic review, meta-analysis, and novel time course analysis examines microvascular failure in the treatment of acute ischemic stroke (AIS) patients undergoing endovascular therapy (EVT) and/or thrombolytic administration for stroke management. A systematic review and meta-analysis following PRIMSA-2020 guidelines was conducted along with a novel curve-of-best fit analysis to elucidate the time-course of microvascular failure. Scopus and PubMed were searched using relevant keywords to identify studies that examine recanalization and reperfusion assessment of AIS patients following large vessel occlusion. Meta-analysis was conducted using a random-effects model. Curve-of-best-fit analysis of microvascular failure rate was performed with a negative exponential model. Twenty-seven studies with 1151 patients were included. Fourteen studies evaluated patients within a standard stroke onset-to-treatment time window (≤6 hours after last known normal) and thirteen studies had an extended time window (>6 hours). Our analysis yields a 22% event rate of microvascular failure following successful recanalization (95% CI: 16-30%). A negative exponential curve modeled a microvascular failure rate asymptote of 28.5% for standard time window studies, with no convergence of the model for extended time window studies. Progressive microvascular failure is a phenomenon that is increasingly identified in clinical studies of AIS patients undergoing revascularization treatment.
Collapse
Affiliation(s)
- Thilan Tudor
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Eleonora F Spinazzi
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Julia E Alexander
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Grace K Mandigo
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Sean D Lavine
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Jack Grinband
- Departments of Psychiatry and Radiology, Columbia University Irving Medical Center, New York, NY, USA
| | - E Sander Connolly
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
37
|
Jeong JW, Lee MH, Luat AF, Xuan Y, Haacke EM, Juhász C. Quantification of enlarged deep medullary vein volumes in Sturge-Weber syndrome. Quant Imaging Med Surg 2024; 14:1916-1929. [PMID: 38415136 PMCID: PMC10895099 DOI: 10.21037/qims-23-1271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 12/05/2023] [Indexed: 02/29/2024]
Abstract
Background Enlarged deep medullary veins (EDMVs) in patients with Sturge-Weber syndrome (SWS) may channel venous blood from the surface to the deep vein system in brain regions affected by the leptomeningeal venous malformation. Thus, the quantification of EDMV volume may provide an objective imaging marker for this vascular compensatory process. The present study proposes a novel analytical method to quantify enlarged EDMV volumes in the affected hemisphere of patients with unilateral SWS. Methods Twenty young subjects, including 10 patients with unilateral SWS and 10 healthy siblings (age 14.5±6.7 and 16.0±7.0 years, respectively) underwent 3T brain MRI scanning using susceptibility-weighted imaging (SWI) and volumetric T1-weighted sequences. The proposed image analytic steps segmented EDMVs in white matter regions, defined on the volumetric T1-weighted images, by statistically associating the likelihood of intensity, location, and tubular shape on SWI. The volumes of the segmented EDMVs, calculated in each hemisphere, were compared between affected and unaffected hemispheres. EDMV volumes were also correlated with visually assessed EDMV scores, hemispheric white matter volumes, and cortical surface areas. Parametric tests including Pearson's correlation, unpaired and paired t-tests, were used. A P value <0.05 was considered statistically significant. Results It was found that EDMVs were identified well in SWS-affected hemispheres while calcified regions were excluded. Mean EDMV volumes in the SWS-affected hemispheres were 10-12-fold greater than in the unaffected or healthy control hemispheres; while white matter volumes and cortical surface areas were lower. EDMV volumes in the SWS-affected hemispheres showed a strong positive correlation with the visual EDMV scores (r=0.88, P=0.001) and an inverse correlation with cortical surface area ratios (r=-0.65, P=0.04) but no correlation with white matter volume ratios. Conclusions EDMVs were detected in the SWS-affected atrophic hemispheres reliably while avoiding calcified regions. The approach can be used to quantify enlarged deep cerebral veins in the human brain, which may provide a potential marker of cerebral venous remodeling.
Collapse
Affiliation(s)
- Jeong-Won Jeong
- Department of Pediatrics, Wayne State University School of Medicine, University Health Center, Detroit, MI, USA
- Department of Neurology, Wayne State University School of Medicine, Detroit, MI, USA
- Translational Imaging Laboratory, University Health Center, Detroit, MI, USA
| | - Min-Hee Lee
- Department of Pediatrics, Wayne State University School of Medicine, University Health Center, Detroit, MI, USA
- Translational Imaging Laboratory, University Health Center, Detroit, MI, USA
| | - Aimee F. Luat
- Department of Pediatrics, Wayne State University School of Medicine, University Health Center, Detroit, MI, USA
- Department of Pediatrics, Central Michigan University, Mount Pleasant, MI, USA
| | - Yang Xuan
- Department of Radiology, Wayne State University School of Medicine, Detroit, MI, USA
| | - E. Mark Haacke
- Department of Neurology, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Radiology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Csaba Juhász
- Department of Pediatrics, Wayne State University School of Medicine, University Health Center, Detroit, MI, USA
- Department of Neurology, Wayne State University School of Medicine, Detroit, MI, USA
- Translational Imaging Laboratory, University Health Center, Detroit, MI, USA
| |
Collapse
|
38
|
Li H, Yu W, Yang Y, Li S, Xu J, Gao C, Zhang W, Shi W, Jin K, Ji X, Ren C. Combination of Atractylenolide I, Atractylenolide III, and Paeoniflorin promotes angiogenesis and improves neurological recovery in a mouse model of ischemic Stroke. Chin Med 2024; 19:3. [PMID: 38178130 PMCID: PMC10768365 DOI: 10.1186/s13020-023-00872-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 12/10/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND Prognosis is critically important in stroke cases, with angiogenesis playing a key role in determining outcomes. This study aimed to investigate the potential protective effects of Atractylenolide I (Atr I), Atractylenolide III (Atr III), and Paeoniflorin (Pae) in promoting angiogenesis following cerebral ischemia. METHODS The bEnd.3 cell line was used to evaluate the effects of these three compounds on vascular endothelial cell proliferation, migration, and tube formation. Male C57BL/6 mice underwent transient middle cerebral artery occlusion (MCAO), followed by daily intragastric administration of the Chinese medicine compounds to assess their impact on brain protection and angiogenesis. In vivo experiments included measuring infarct size and assessing neurological function. Immunofluorescence staining and an angiogenesis antibody array were used to evaluate angiogenesis in ischemic brain tissue. Functional enrichment analysis was performed to further investigate the pathways involved in the protective effects of the compounds. Molecular docking analysis explored the potential binding affinity of the compounds to insulin-like growth factor 2 (IGF-2), and Western blotting was used to measure levels of angiogenesis-related proteins. RESULTS In vitro, the combination of Atr I, Atr III, and Pae enhanced cell proliferation, promoted migration, and stimulated tube formation. In vivo, the combined treatment significantly facilitated neurological function recovery and angiogenesis by day 14. The treatment also increased levels of angiogenesis-related proteins, including IGF-2. Pearson correlation analysis revealed a strong positive association between IGF-2 levels in ischemic brain tissue and angiogenesis, suggesting a good affinity of the compounds for the IGF-2 binding site, as supported by molecular docking analysis. CONCLUSION The administration of Atr I, Atr III, and Pae has shown significant enhancements in long-term stroke recovery in mice, likely due to the promotion of angiogenesis via increased activation of the IGF-2 pathway in ischemic brain tissue.
Collapse
Affiliation(s)
- Haiyan Li
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Center of Stroke, Beijing Institute of Brain Disorder, Capital Medical University, Chang Chun Road 45, Beijing, 100053, China
- School of Chinese Medicine, Beijing University of Chines Medicine, Beijing, 100029, China
| | - Wantong Yu
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Center of Stroke, Beijing Institute of Brain Disorder, Capital Medical University, Chang Chun Road 45, Beijing, 100053, China
| | - Yong Yang
- School of Chinese Medicine, Beijing University of Chines Medicine, Beijing, 100029, China
| | - Sijie Li
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Center of Stroke, Beijing Institute of Brain Disorder, Capital Medical University, Chang Chun Road 45, Beijing, 100053, China
| | - Jun Xu
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Center of Stroke, Beijing Institute of Brain Disorder, Capital Medical University, Chang Chun Road 45, Beijing, 100053, China
| | - Chen Gao
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Center of Stroke, Beijing Institute of Brain Disorder, Capital Medical University, Chang Chun Road 45, Beijing, 100053, China
| | - Wei Zhang
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Center of Stroke, Beijing Institute of Brain Disorder, Capital Medical University, Chang Chun Road 45, Beijing, 100053, China
| | - Wenjie Shi
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Center of Stroke, Beijing Institute of Brain Disorder, Capital Medical University, Chang Chun Road 45, Beijing, 100053, China
| | - Kunlin Jin
- Department of Pharmacology and Neuroscience, Texas Health Science Center, University of North, Fort Worth, TX, 76107, USA
| | - Xunming Ji
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Center of Stroke, Beijing Institute of Brain Disorder, Capital Medical University, Chang Chun Road 45, Beijing, 100053, China
| | - Changhong Ren
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Center of Stroke, Beijing Institute of Brain Disorder, Capital Medical University, Chang Chun Road 45, Beijing, 100053, China.
| |
Collapse
|
39
|
Nie L, He J, Wang J, Wang R, Huang L, Jia L, Kim YT, Bhawal UK, Fan X, Zille M, Jiang C, Chen X, Wang J. Environmental Enrichment for Stroke and Traumatic Brain Injury: Mechanisms and Translational Implications. Compr Physiol 2023; 14:5291-5323. [PMID: 38158368 DOI: 10.1002/cphy.c230007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Acquired brain injuries, such as ischemic stroke, intracerebral hemorrhage (ICH), and traumatic brain injury (TBI), can cause severe neurologic damage and even death. Unfortunately, currently, there are no effective and safe treatments to reduce the high disability and mortality rates associated with these brain injuries. However, environmental enrichment (EE) is an emerging approach to treating and rehabilitating acquired brain injuries by promoting motor, sensory, and social stimulation. Multiple preclinical studies have shown that EE benefits functional recovery, including improved motor and cognitive function and psychological benefits mediated by complex protective signaling pathways. This article provides an overview of the enriched environment protocols used in animal models of ischemic stroke, ICH, and TBI, as well as relevant clinical studies, with a particular focus on ischemic stroke. Additionally, we explored studies of animals with stroke and TBI exposed to EE alone or in combination with multiple drugs and other rehabilitation modalities. Finally, we discuss the potential clinical applications of EE in future brain rehabilitation therapy and the molecular and cellular changes caused by EE in rodents with stroke or TBI. This article aims to advance preclinical and clinical research on EE rehabilitation therapy for acquired brain injury. © 2024 American Physiological Society. Compr Physiol 14:5291-5323, 2024.
Collapse
Affiliation(s)
- Luwei Nie
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinxin He
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
- Key Laboratory for Brain Science Research and Transformation in the Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Junmin Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Ruike Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Leo Huang
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
| | - Lin Jia
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yun Tai Kim
- Division of Functional Food Research, Korea Food Research Institute, Wanju-gun, Jeollabuk-do, Republic of Korea
- Department of Food Biotechnology, Korea University of Science & Technology, Daejeon, Republic of Korea
| | - Ujjal K Bhawal
- Research Institute of Oral Science, Nihon University School of Dentistry at Matsudo, Chiba, Japan
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Xiaochong Fan
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Marietta Zille
- Department of Pharmaceutical Sciences, Division of Pharmacology and Toxicology, University of Vienna, Vienna, Austria
| | - Chao Jiang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Xuemei Chen
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jian Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| |
Collapse
|
40
|
Gao Y, Liu X, Xu B, Zhang X, Wang Y, Ni J, Yang Y. Effect of collateral circulation in patients with multiple craniocervical artery stenoses. Clin Transl Sci 2023; 16:2779-2790. [PMID: 37932924 PMCID: PMC10719488 DOI: 10.1111/cts.13673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 10/06/2023] [Accepted: 10/12/2023] [Indexed: 11/08/2023] Open
Abstract
Based on previous findings, collateral circulation in the brain is vital in mitigating cerebral ischemia's effects and influencing stroke risk. This retrospective study examined collateral circulation, admission ischemic stroke status, and long-term recurrence in patients with multiple craniocervical artery stenoses. Consecutive symptomatic internal carotid artery (ICA) stenosis patients from the First Affiliated Hospital of Soochow University were recruited. Baseline data including medical histories and neurological function at admission were collected. Imaging techniques assessed collateral compensative capacity. Multivariate logistic regression analysis was used to investigate the association between collateral circulation and case status. A total of 559 patients with symptomatic ICA stenosis were included, among whom 153 (27.4%) had concurrent moderate to severe vertebro-basilar artery (VBA) stenosis. Dizziness, weakness/numbness, and slurring of speech were the primary symptoms in all patients. Over 36 months, 71 (12.7%) patients experienced a recurrence of acute ischemic stroke (AIS). In multivariate analysis, collateral circulation was found to be negatively associated with AIS (regional leptomeningeal collateral [rLMC] scores: OR: 0.798, 95% CI: 0.743-0.857, p < 0.001; Tan scores: OR: 0.478, 95% CI: 0.336-0.679, p < 0.001). Meanwhile, the collateral circulation scores were significantly associated with the recurrence of AIS within 3 years (rLMC scores: OR: 0.926, 95% CI: 0.860-0.997, p = 0.042; Tan scores: OR: 0.467, 95% CI: 0.306-0.712, p < 0.001). Most associations remained significant in the subgroup of patients with VBA stenosis. Favorable collateral circulation in multiple craniocervical artery stenosis patients reduced long-term ischemic event recurrence. Stratifying treatment risks is essential for optimizing outcomes.
Collapse
Affiliation(s)
- Ya Gao
- Departments of NeurologyThe First Affiliated Hospital of Soochow UniversitySuzhou, JiangsuChina
- Departments of NeurologySuzhou Guangci Cancer HospitalSuzhou, JiangsuChina
| | - Xuan Liu
- Departments of NeurologyThe First Affiliated Hospital of Soochow UniversitySuzhou, JiangsuChina
| | - Beibei Xu
- Departments of NeurologyThe First Affiliated Hospital of Soochow UniversitySuzhou, JiangsuChina
| | - Ximeng Zhang
- Departments of NeurologyThe First Affiliated Hospital of Soochow UniversitySuzhou, JiangsuChina
| | - Yiqing Wang
- Departments of NeurologyThe First Affiliated Hospital of Soochow UniversitySuzhou, JiangsuChina
| | - Jianqiang Ni
- Departments of NeurologyThe First Affiliated Hospital of Soochow UniversitySuzhou, JiangsuChina
| | - Yi Yang
- Departments of NeurologyThe First Affiliated Hospital of Soochow UniversitySuzhou, JiangsuChina
| |
Collapse
|
41
|
Wang W, Cheng W, Wang X, Li Z, Gao J. CircFKBP3 absence alleviates oxygen glucose deprivation-induced function loss of human brain microvascular endothelial cells in vitro via governing the miR-766-3p/TRAF3 axis. Int J Neurosci 2023:1-12. [PMID: 37982234 DOI: 10.1080/00207454.2023.2279506] [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: 09/21/2023] [Accepted: 10/31/2023] [Indexed: 11/21/2023]
Abstract
BACKGROUND Brain microvascular endothelial cell (BMEC) functions loss is a key event in the development of ischemic stroke, which may be affected by the dysregulation of circular RNAs (circRNAs). We aimed to unveil the role of circRNA FKBP Prolyl Isomerase 3 (circFKBP3) in cell models of ischemic stroke. METHODS Cell models of ischemic stroke were constructed in human BEMCs (HBMECs) with the treatment of oxygen glucose deprivation (OGD). Quantitative real-time PCR (qPCR) and western blotting were conducted for expression analysis of circFKBP3, miR-766-3p and TNF receptor associated factor 3 (TRAF3). CCK-8, transwell, wound healing, flow cytometry, tube formation and ELISA assays were implemented to monitor cell viability, migration, apoptosis, angiogenesis and inflammation production. The putative binding relationship between miR-766-3p and circFKBP3 or TRAF3 was validated by dual-luciferase, RIP and pull-down assays. RESULTS CircFKBP3 expression was elevated in OGD-treated HBMECs. OGD suppressed HBMEC viability, migration, angiogenesis, and provoked cell apoptosis and inflammation production, while knockdown of circFKBP3 attenuated these effects. CircFKBP3 interacted with miR-766-3p, and circFKBP3 absence-repressed HBMEC function loss and inflammation were recovered by miR-766-3p inhibition. CircFKBP3 targeted miR-766-3p to regulate TRAF3 expression. MiR-766-3p enrichment-repressed HBMEC function loss and inflammation were recovered by TRAF3 overexpression. CONCLUSION CircFKBP3 absence alleviated OGD-induced function loss and inflammatory responses of HBMECs via governing the miR-766-3p/TRAF3 axis.
Collapse
Affiliation(s)
- Wenyan Wang
- Department of Neurology, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Wei Cheng
- Department of Neurology, Wuhan Puren Hospital affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Xudong Wang
- Department of Neurology, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Zhixin Li
- Department of Neurology, Wuhan Puren Hospital affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Jinli Gao
- Department of Neurology, Wuhan Puren Hospital affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
42
|
Chen R, Du W, Zhang X, Xu R, Jiang W, Zhang C, Yang Y, Zhang H, Xie X, Song D, Yuan Y, Zhang X. Protective effects of low-intensity pulsed ultrasound (LIPUS) against cerebral ischemic stroke in mice by promoting brain vascular remodeling via the inhibition of ROCK1/p-MLC2 signaling pathway. Cereb Cortex 2023; 33:10984-10996. [PMID: 37771006 DOI: 10.1093/cercor/bhad330] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 09/30/2023] Open
Abstract
Vascular remodeling is essential for patients with cerebral ischemic stroke (CIS). Our previous study proved that low-intensity pulsed ultrasound (LIPUS) could increase cortical hemodynamics. However, the effects and mechanisms of LIPUS on cerebral vascular remodeling after CIS are still unknown. In this study, we applied LIPUS to the mouse brain at 0.5 h after distal middle cerebral artery occlusion (dMCAO) and subsequently daily for a stimulation time of 30 min. Results showed that compared with the dMCAO group, LIPUS markedly increased cerebral blood flow (CBF), reduced brain swelling, and improved functional recovery at day 3 after CIS. LIPUS promoted leptomeningeal vasculature remodeling, enlarged vascular diameter, and increased the average vessel length and density at day 3 after CIS. Proteomic analysis highlighted that LIPUS mainly participated in the regulation of actin cytoskeleton pathway. Rho kinase 1 (ROCK1) was downregulated by LIPUS and participated in regulation of actin cytoskeleton. Subsequently, we verified that ROCK1 was mainly expressed in pericytes. Furthermore, we demonstrated that LIPUS inhibited ROCK1/p-MLC2 signaling pathway after CIS, which had positive effects on vascular remodeling and cerebral blood circulation. In conclusion, our preliminary study revealed the vascular remodeling effects and mechanism of LIPUS in CIS, provided evidence for potential clinical application of LIPUS.
Collapse
Affiliation(s)
- Rong Chen
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei 050000, China
| | - Wei Du
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
- Department of Orthopedics, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, China
| | - Xiao Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei 050000, China
| | - Renhao Xu
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei 050000, China
| | - Wei Jiang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei 050000, China
| | - Cong Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei 050000, China
| | - Yi Yang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei 050000, China
| | - Huiran Zhang
- Department of Biological Pharmacy, Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Xiaoli Xie
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Degang Song
- Department of Neurology, First Hospital of Qinhuangdao, Hebei Medical University, No. 258, Wenhua Road, Qinhuangdao, Hebei 066000, China
| | - Yi Yuan
- School of Electrical Engineering, Key Laboratory of Intelligent Rehabilitation and Neuromodulation of Hebei Province, Yanshan University, Qinhuangdao, Hebei 066004, China
| | - Xiangjian Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei 050000, China
| |
Collapse
|
43
|
Sun S, Xu Y, Yu N, Zhang M, Wang J, Wan D, Tian Z, Zhu H. Catalpol Alleviates Ischemic Stroke Through Promoting Angiogenesis and Facilitating Proliferation and Differentiation of Neural Stem Cells via the VEGF-A/KDR Pathway. Mol Neurobiol 2023; 60:6227-6247. [PMID: 37439957 DOI: 10.1007/s12035-023-03459-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 06/22/2023] [Indexed: 07/14/2023]
Abstract
Stroke is one of the leading causes of disability and death globally with a lack of effective therapeutic strategies. Catalpol is a bioactive compound derived from the traditional Chinese medicine Rehmannia glutinosa and it has been shown to be protective against various neurological diseases. The potential roles of catalpol against ischemic stroke are still not completely clear. In this study, we examined the effect and mechanism of catalpol against ischemic stroke using in vivo rat distal middle cerebral artery occlusion (dMCAO) and in vitro oxygen-glucose deprivation (OGD) models. We demonstrated that catalpol indeed attenuated the neurological deficits caused by dMCAO and improved neurological function. Catalpol remarkably promoted angiogenesis, promoted proliferation and differentiation of neural stem cells (NSCs) in the subventricular zone (SVZ), and prevented neuronal loss and astrocyte activation in the ischemic cortex or hippocampal dentate gyrus (DG) in vivo. The vascular endothelial growth factor receptor 2 (KDR, VEGFR-2) inhibitor SU5416 and VEGF-A shRNA were used to investigate the underlying mechanisms. The results showed that SU5416 administration or VEGF-A-shRNA transfection both attenuated the effects of catalpol. We also found that catalpol promoted the proliferation of cultured brain microvascular endothelial cells (BMECs) and the proliferation and differentiation of NSCs subjected to OGD insult in vitro. Interestingly, the impact of catalpol on cultured cells was also inhibited by SU5416. Moreover, catalpol was shown to protect NSCs against OGD indirectly by promoting BMEC proliferation in the co-cultured system. Taken together, catalpol showed therapeutic potential in cerebral ischemia by promoting angiogenesis and NSC proliferation and differentiation. The protective effects of catalpol were mediated through VEGF-A/KDR pathway activation.
Collapse
Affiliation(s)
- Si Sun
- College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
| | - Yitong Xu
- College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
| | - Ningxi Yu
- College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
| | - Meifeng Zhang
- College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
| | - Jinghui Wang
- College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
| | - Dong Wan
- Department of Emergency and Critical Care Medicine, First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhen Tian
- College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China.
| | - Huifeng Zhu
- College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China.
| |
Collapse
|
44
|
Deshpande A, Elliott J, Jiang B, Tahsili-Fahadan P, Kidwell C, Wintermark M, Laksari K. End to end stroke triage using cerebrovascular morphology and machine learning. Front Neurol 2023; 14:1217796. [PMID: 37941573 PMCID: PMC10628321 DOI: 10.3389/fneur.2023.1217796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 09/20/2023] [Indexed: 11/10/2023] Open
Abstract
Background Rapid and accurate triage of acute ischemic stroke (AIS) is essential for early revascularization and improved patient outcomes. Response to acute reperfusion therapies varies significantly based on patient-specific cerebrovascular anatomy that governs cerebral blood flow. We present an end-to-end machine learning approach for automatic stroke triage. Methods Employing a validated convolutional neural network (CNN) segmentation model for image processing, we extract each patient's cerebrovasculature and its morphological features from baseline non-invasive angiography scans. These features are used to detect occlusion's presence and the site automatically, and for the first time, to estimate collateral circulation without manual intervention. We then use the extracted cerebrovascular features along with commonly used clinical and imaging parameters to predict the 90 days functional outcome for each patient. Results The CNN model achieved a segmentation accuracy of 94% based on the Dice similarity coefficient (DSC). The automatic stroke detection algorithm had a sensitivity and specificity of 92% and 94%, respectively. The models for occlusion site detection and automatic collateral grading reached 96% and 87.2% accuracy, respectively. Incorporating the automatically extracted cerebrovascular features significantly improved the 90 days outcome prediction accuracy from 0.63 to 0.83. Conclusion The fast, automatic, and comprehensive model presented here can improve stroke diagnosis, aid collateral assessment, and enhance prognostication for treatment decisions, using cerebrovascular morphology.
Collapse
Affiliation(s)
- Aditi Deshpande
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ, United States
- Department of Mechanical Engineering, University of California, Riverside, Riverside, CA, United States
| | - Jordan Elliott
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ, United States
| | - Bin Jiang
- Department of Radiology, Stanford University, Stanford, CA, United States
| | - Pouya Tahsili-Fahadan
- Department of Medical Education, University of Virginia, Inova Campus, Falls Church, VA, United States
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Chelsea Kidwell
- Department of Neurology, University of Arizona, Tucson, AZ, United States
| | - Max Wintermark
- Department of Neuroradiology, MD Anderson Center, University of Texas, Houston, TX, United States
| | - Kaveh Laksari
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ, United States
- Department of Mechanical Engineering, University of California, Riverside, Riverside, CA, United States
- Department of Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
45
|
Sperring CP, Savage WM, Argenziano MG, Leifer VP, Alexander J, Echlov N, Spinazzi EF, Connolly ES. No-Reflow Post-Recanalization in Acute Ischemic Stroke: Mechanisms, Measurements, and Molecular Markers. Stroke 2023; 54:2472-2480. [PMID: 37534511 DOI: 10.1161/strokeaha.123.044240] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2023]
Abstract
Acute ischemic stroke remains the primary cause of disability worldwide. For patients with large vessel occlusions, intravenous thrombolysis followed by mechanical thrombectomy remains the standard of care. Revascularization of the large vessel is typically successful. However, despite reopening of the occluded vessel, many patients fail to return to independence. Functional failure, despite macrovascular recanalization, is often referred to as the no-reflow phenomenon. Even with an extensive characterization of reperfusion in animal models, numerous mechanisms may explain no-reflow. Further, uniform measurements of this microvascular dysfunction and prognostic markers associated with no-reflow are lacking. In this review, we highlight a number of mechanisms that may explain no-reflow, characterize current multimodal measurements, and assess its molecular markers.
Collapse
Affiliation(s)
- Colin P Sperring
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital
| | - William M Savage
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital
| | - Michael G Argenziano
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital
| | - Valia P Leifer
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital
| | - Julia Alexander
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital
| | - Nicolas Echlov
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital
| | - Eleonora F Spinazzi
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital
| | - E Sander Connolly
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital
| |
Collapse
|
46
|
Xu J, Shen Y, Luan P, Wang H, Xu Y, Jiang L, Li R, Wang F, Zhu Y, Zhang J. Pro‑angiogenic activity of salvianolate and its potential therapeutic effect against acute cerebral ischemia. Exp Ther Med 2023; 26:409. [PMID: 37522065 PMCID: PMC10375442 DOI: 10.3892/etm.2023.12108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 06/23/2023] [Indexed: 08/01/2023] Open
Abstract
Salvianolate (Sal) is a medicinal composition that is widely used in China for the treatment of coronary heart disease and angina pectoris. The aim of the present study was to investigate the potential macrophage-mediated pro-angiogenic effects of Sal in vitro. In addition, another aim was to explore the effects of Sal in a rat model of transient middle cerebral artery occlusion (tMCAO) along with the potential mechanism by which it promotes angiogenesis. In this study, human umbilical vein endothelial cells (HUVECs) and Raw264.7 macrophages in vitro, and a rat tMCAO model in vivo were used to detect the pro-angiogenic effect and mechanism of Sal. The results of in vitro experiments showed that the viability, migration and tube formation of HUVECs were promoted by the supernatant of Sal-treated Raw264.7 macrophages (s-Sal) but not by Sal alone. s-Sal also increased the levels of phosphorylated (p-)VEGFR-2, p-AKT and p-p38 MAPK in HUVECs while Sal alone did not. In vivo, treatment with Sal significantly reduced the cerebral infarction volume and neurological deficit scores in the rat tMCAO model. Similar to the mechanism observed in the in vitro experiments, Sal treatment upregulated the protein expression of VEGF and VEGFR-2, in addition to the phosphorylation of VEGFR-2, AKT and p38, in the brain tissues of the tMCAO model rats. In summary, the results of the present study suggest that the mechanism of Sal-mediated angiogenesis is associated with stimulation of the VEGF/VEGFR-2 signaling pathway by macrophages. This suggests the potential of Sal as a therapeutic option for the treatment of acute cerebral ischemic injury, which may act via the promotion of angiogenesis.
Collapse
Affiliation(s)
- Jiazhen Xu
- Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, P.R. China
| | - Yue Shen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Pengwei Luan
- Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Haiying Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Yulan Xu
- Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Lixian Jiang
- Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Ruixiang Li
- Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Feiyun Wang
- Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Yuying Zhu
- Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Jiange Zhang
- Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
- Shanghai Frontiers Science Center for Traditional Chinese Medicine Chemical Biology, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| |
Collapse
|
47
|
Guo R, Wang X, Guo Q, Yan Y, Gong W, Zheng W, Zhao G, Wang H, Xu L, Nie S. Cardiac magnetic resonance shows increased adverse ventricular remodeling in younger patients after ST-segment elevation myocardial infarction. Eur Radiol 2023; 33:4637-4647. [PMID: 36700954 PMCID: PMC10289996 DOI: 10.1007/s00330-023-09406-5] [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: 05/20/2022] [Revised: 10/04/2022] [Accepted: 12/23/2022] [Indexed: 01/27/2023]
Abstract
OBJECTIVES Young patients account for about half of ST-segment elevation myocardial infarction (STEMI) patients and display a unique risk profile compared with old patients. Whether these differences are related to disparities in ventricular remodeling remains unknown. This study aimed to evaluate age-related differences in ventricular remodeling after primary percutaneous coronary intervention (PPCI) for STEMI. METHODS In this observational study, consecutive STEMI patients between October 2019 and March 2021 who underwent serial cardiovascular magnetic resonance at index admission (3 to 7 days) and 3 months after PPCI were enrolled. Adverse remodeling was defined as ≥ 10% enlargement in left ventricular end-diastolic volume index (LVEDVi), while reverse remodeling was defined as a decrease in left ventricular end-systolic volume index (LVESVi) of more than 10%. RESULTS A total of 123 patients were included and grouped into young (< 60 years, n = 71) and old (≥ 60 years, n = 52) patients. Despite generally similar baseline structural and infarct characteristics, LVESVi significantly decreased only in old patients during follow-up (p = 0.034). The incidence of adverse remodeling was higher (49.3% vs 30.8%, p = 0.039), while the incidence of reverse remodeling was lower (31.0% vs 53.8%, p = 0.011) in young compared with old patients. Younger age (< 60 years) was associated with a significantly higher risk of adverse remodeling (adjusted OR 3.51, 95% CI 1.41-8.74, p = 0.007) and lower incidence of reverse remodeling (adjusted OR 0.42, 95% CI 0.18-0.97, p = 0.046). CONCLUSIONS In STEMI patients undergoing PPCI, young patients are at a higher risk of adverse remodeling and less probably develop reverse remodeling than old patients. Equal or more attention should be paid to young patients with STEMI compared with their older counterparts. KEY POINTS • In STEMI patients undergoing PPCI, young patients displayed unfavorable remodeling compared with old patients. • Young patients are at a higher risk of adverse remodeling and less probably develop reverse remodeling than old patients. • Equal or more attention should be paid to young patients compared with their older counterparts.
Collapse
Affiliation(s)
- Ruifeng Guo
- Center for Coronary Artery Disease, Division of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Xiao Wang
- Center for Coronary Artery Disease, Division of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Chaoyang District, Beijing, 100029, China.
| | - Qian Guo
- Center for Coronary Artery Disease, Division of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Yan Yan
- Center for Coronary Artery Disease, Division of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Wei Gong
- Center for Coronary Artery Disease, Division of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Wen Zheng
- Center for Coronary Artery Disease, Division of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Guanqi Zhao
- Center for Coronary Artery Disease, Division of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Hui Wang
- Department of Radiology, Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Lei Xu
- Department of Radiology, Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Shaoping Nie
- Center for Coronary Artery Disease, Division of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Chaoyang District, Beijing, 100029, China.
| |
Collapse
|
48
|
Abstract
Owing to its high disability and mortality rates, stroke has been the second leading cause of death worldwide. Since the pathological mechanisms of stroke are not fully understood, there are few clinical treatment strategies available with an exception of tissue plasminogen activator (tPA), the only FDA-approved drug for the treatment of ischemic stroke. Angiogenesis is an important protective mechanism that promotes neural regeneration and functional recovery during the pathophysiological process of stroke. Thus, inducing angiogenesis in the peri-infarct area could effectively improve hemodynamics, and promote vascular remodeling and recovery of neurovascular function after ischemic stroke. In this review, we summarize the cellular and molecular mechanisms affecting angiogenesis after cerebral ischemia registered in PubMed, and provide pro-angiogenic strategies for exploring the treatment of ischemic stroke, including endothelial progenitor cells, mesenchymal stem cells, growth factors, cytokines, non-coding RNAs, etc.
Collapse
Affiliation(s)
- Jie Fang
- Department of Pharmacology, Second Military Medical University / Naval Medical University, Shanghai, 200433, China
| | - Zhi Wang
- Department of Pharmacology, Second Military Medical University / Naval Medical University, Shanghai, 200433, China
| | - Chao-Yu Miao
- Department of Pharmacology, Second Military Medical University / Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
49
|
Li J, Wu J, Zhou X, Lu Y, Ge Y, Zhang X. Targeting neuronal mitophagy in ischemic stroke: an update. BURNS & TRAUMA 2023; 11:tkad018. [PMID: 37274155 PMCID: PMC10232375 DOI: 10.1093/burnst/tkad018] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 01/29/2023] [Accepted: 03/19/2023] [Indexed: 06/06/2023]
Abstract
Cerebral ischemia is a neurological disorder associated with complex pathological mechanisms, including autophagic degradation of neuronal mitochondria, or termed mitophagy, following ischemic events. Despite being well-documented, the cellular and molecular mechanisms underlying the regulation of neuronal mitophagy remain unknown. So far, the evidence suggests neuronal autophagy and mitophagy are separately regulated in ischemic neurons, the latter being more likely activated by reperfusional injury. Specifically, given the polarized morphology of neurons, mitophagy is regulated by different neuronal compartments, with axonal mitochondria being degraded by autophagy in the cell body following ischemia-reperfusion insult. A variety of molecules have been associated with neuronal adaptation to ischemia, including PTEN-induced kinase 1, Parkin, BCL2 and adenovirus E1B 19-kDa-interacting protein 3 (Bnip3), Bnip3-like (Bnip3l) and FUN14 domain-containing 1. Moreover, it is still controversial whether mitophagy protects against or instead aggravates ischemic brain injury. Here, we review recent studies on this topic and provide an updated overview of the role and regulation of mitophagy during ischemic events.
Collapse
Affiliation(s)
- Jun Li
- Department of Clinical Pharmacy, the First Affiliated Hospital, Zhejiang University School of Medicine, Qingchun Road 79, Xiacheng District, Hangzhou, China
| | - Jiaying Wu
- Department of Clinical Pharmacy, the First Affiliated Hospital, Zhejiang University School of Medicine, Qingchun Road 79, Xiacheng District, Hangzhou, China
| | - Xinyu Zhou
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Yuhangtang Road 866, Xihu District, Hangzhou, China
| | - Yangyang Lu
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Yuhangtang Road 866, Xihu District, Hangzhou, China
| | - Yuyang Ge
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Yuhangtang Road 866, Xihu District, Hangzhou, China
| | | |
Collapse
|
50
|
Carlsson R, Enström A, Paul G. Molecular Regulation of the Response of Brain Pericytes to Hypoxia. Int J Mol Sci 2023; 24:5671. [PMID: 36982744 PMCID: PMC10053233 DOI: 10.3390/ijms24065671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/13/2023] [Accepted: 03/14/2023] [Indexed: 03/18/2023] Open
Abstract
The brain needs sufficient oxygen in order to function normally. This is achieved by a large vascular capillary network ensuring that oxygen supply meets the changing demand of the brain tissue, especially in situations of hypoxia. Brain capillaries are formed by endothelial cells and perivascular pericytes, whereby pericytes in the brain have a particularly high 1:1 ratio to endothelial cells. Pericytes not only have a key location at the blood/brain interface, they also have multiple functions, for example, they maintain blood-brain barrier integrity, play an important role in angiogenesis and have large secretory abilities. This review is specifically focused on both the cellular and the molecular responses of brain pericytes to hypoxia. We discuss the immediate early molecular responses in pericytes, highlighting four transcription factors involved in regulating the majority of transcripts that change between hypoxic and normoxic pericytes and their potential functions. Whilst many hypoxic responses are controlled by hypoxia-inducible factors (HIF), we specifically focus on the role and functional implications of the regulator of G-protein signaling 5 (RGS5) in pericytes, a hypoxia-sensing protein that is regulated independently of HIF. Finally, we describe potential molecular targets of RGS5 in pericytes. These molecular events together contribute to the pericyte response to hypoxia, regulating survival, metabolism, inflammation and induction of angiogenesis.
Collapse
Affiliation(s)
- Robert Carlsson
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Centre and Wallenberg Centre for Molecular Medicine, Lund University, 22184 Lund, Sweden
| | - Andreas Enström
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Centre and Wallenberg Centre for Molecular Medicine, Lund University, 22184 Lund, Sweden
| | - Gesine Paul
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Centre and Wallenberg Centre for Molecular Medicine, Lund University, 22184 Lund, Sweden
- Department of Neurology, Scania University Hospital, 22185 Lund, Sweden
| |
Collapse
|