51
|
A new method for predicting the prognosis of ischemic stroke based vascular structure features and lesion location features. Clin Imaging 2023; 98:1-7. [PMID: 36934582 DOI: 10.1016/j.clinimag.2023.03.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 02/17/2023] [Accepted: 03/07/2023] [Indexed: 03/17/2023]
Abstract
OBJECTIVE Determining the changes in the prognosis of the cerebral infarction area has an important guiding role in the selection of the treatment plan. The goal of this study is to propose a machine learning-based method that can predict the prognosis of stroke effectively and efficiently. METHODS 97 cases of stroke were analyzed retrospectively. Firstly, we extracted vascular structural features from computed tomography angiography (CTA) images and stroke location features from diffusion-weighted imaging (DWI) images to comprehensively characterize the lesions, respectively. Then, we performed sparse representation-based feature selection and classification to predict the prognosis of stroke based on the extracted features. Finally, we randomly divided the 97 cases into cross-validation set, independent testing set 1 and independent testing set 2 to validate the proposed model. RESULTS 464 vascular structure features and 116 positional features were extracted. After feature selection, 52 features were finally applied to build the classification model. The proposed model achieved promising prediction performance on the two independent testing sets, with the classification accuracies of 85.19% and 81.25%, respectively. CONCLUSION The proposed machine learning approach can effectively mine and accurately quantify the features related to the prognosis, which include the vascular structural features and the stroke location features. In addition, the established prognostic prediction model based on these features has achieved interesting performances, which may provide valuable guidance for the clinical treatment of stroke.
Collapse
|
52
|
Hu X, Geng P, Zhao X, Wang Q, Liu C, Guo C, Dong W, Jin X. The NG2-glia is a potential target to maintain the integrity of neurovascular unit after acute ischemic stroke. Neurobiol Dis 2023; 180:106076. [PMID: 36921779 DOI: 10.1016/j.nbd.2023.106076] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/07/2023] [Accepted: 03/07/2023] [Indexed: 03/18/2023] Open
Abstract
The neurovascular unit (NVU) plays a critical role in health and disease. In the current review, we discuss the critical role of a class of neural/glial antigen 2 (NG2)-expressing glial cells (NG2-glia) in regulating NVU after acute ischemic stroke (AIS). We first introduce the role of NG2-glia in the formation of NVU during development as well as aging-induced damage to NVU and accompanying NG2-glia change. We then discuss the reciprocal interactions between NG2-glia and the other component cells of NVU, emphasizing the factors that could influence NG2-glia. Damage to the NVU integrity is the pathological basis of edema and hemorrhagic transformation, the most dreaded complication after AIS. The role of NG2-glia in AIS-induced NVU damage and the effect of NG2-glia transplantation on AIS-induced NVU damage are summarized. We next discuss the role of NG2-glia and the effect of NG2-glia transplantation in oligodendrogenesis and white matter repair as well as angiogenesis which is associated with the outcome of the patients after AIS. Finally, we review the current strategies to promote NG2-glia proliferation and differentiation and propose to use the dental pulp stem cells (DPSC)-derived exosome as a promising strategy to reduce AIS-induced injury and promote repair through maintaining the integrity of NVU by regulating endogenous NG2-glia proliferation and differentiation.
Collapse
Affiliation(s)
- Xiaoyan Hu
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
| | - Panpan Geng
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
| | - Xiaoyun Zhao
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
| | - Qian Wang
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
| | - Changqing Liu
- Department of Neurosurgery, Beijing Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Chun Guo
- School of Biosciences, University of Sheffield, Firth Court, Western Bank, Sheffield, UK
| | - Wen Dong
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Xinchun Jin
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China; Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China.
| |
Collapse
|
53
|
Jo H, Lee SU, Jeong HG, Kim YD, Kim T, Sunwoo L, Ban SP, Bang JS, Kwon O, Oh CW. Long-term outcomes and quantitative radiologic analysis of extracranial-intracranial bypass for hemodynamically compromised chronic large artery occlusive disease. Sci Rep 2023; 13:3717. [PMID: 36879127 PMCID: PMC9988922 DOI: 10.1038/s41598-023-30874-8] [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: 09/16/2022] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
This study aimed to demonstrate the effectiveness of nonemergent extracranial-to-intracranial bypass (EIB) in symptomatic chronic large artery atherosclerotic stenosis or occlusive disease (LAA) through quantitative analysis of computed tomography perfusion (CTP) parameters using RAPID software. We retrospectively analyzed 86 patients who underwent nonemergent EIB due to symptomatic chronic LAA. CTP data obtained preoperatively, immediately postoperatively (PostOp0), and 6 months postoperatively (PostOp6M) after EIB were quantitatively analyzed through RAPID software, and their association with intraoperative bypass flow (BF) was assessed. The clinical outcomes, including neurologic state, incidence of recurrent infarction and complications, were also analyzed. The time-to-maximum (Tmax) > 8 s, > 6 s and > 4 s volumes decreased significantly at PostOp0 and up through PostOp6M (preoperative, 5, 51, and 223 ml (median), respectively; PostOp0, 0, 20.25, and 143 ml, respectively; PostOp6M, 0, 7.5, and 148.5 ml, respectively; p < 0.001, p < 0.001, and p < 0.001, respectively). The postoperative improvement in the Tmax > 6 s and > 4 s volumes was significantly correlated with the BF at PostOp0 and PostOp6M (PostOp0, r = 0.367 (p = 0.001) and r = 0.275 (p = 0.015), respectively; PostOp6M r = 0.511 (p < 0.001) and r = 0.391 (p = 0.001), respectively). The incidence of recurrent cerebral infarction was 4.7%, and there were no major complications that produced permanent neurological impairment. Nonemergent EIB under strict operation indications can be a feasible treatment for symptomatic, hemodynamically compromised LAA patients.
Collapse
Affiliation(s)
- Hyunjun Jo
- Department of Neurosurgery, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea
| | - Si Un Lee
- Department of Neurosurgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, 82 Gumi-ro 173 beon-gil, Bundang-gu, Seongnam-si, 13620, Gyeonggi-do, Korea.
| | - Han-Gil Jeong
- Department of Neurosurgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, 82 Gumi-ro 173 beon-gil, Bundang-gu, Seongnam-si, 13620, Gyeonggi-do, Korea.,Department of Neurology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam-si, Korea
| | - Young-Deok Kim
- Department of Neurosurgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, 82 Gumi-ro 173 beon-gil, Bundang-gu, Seongnam-si, 13620, Gyeonggi-do, Korea
| | - Tackeun Kim
- Department of Neurosurgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, 82 Gumi-ro 173 beon-gil, Bundang-gu, Seongnam-si, 13620, Gyeonggi-do, Korea
| | - Leonard Sunwoo
- Department of Radiology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam-si, Korea
| | - Seung Pil Ban
- Department of Neurosurgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, 82 Gumi-ro 173 beon-gil, Bundang-gu, Seongnam-si, 13620, Gyeonggi-do, Korea
| | - Jae Seung Bang
- Department of Neurosurgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, 82 Gumi-ro 173 beon-gil, Bundang-gu, Seongnam-si, 13620, Gyeonggi-do, Korea
| | - Oki Kwon
- Department of Neurosurgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, 82 Gumi-ro 173 beon-gil, Bundang-gu, Seongnam-si, 13620, Gyeonggi-do, Korea
| | - Chang Wan Oh
- Department of Neurosurgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, 82 Gumi-ro 173 beon-gil, Bundang-gu, Seongnam-si, 13620, Gyeonggi-do, Korea
| |
Collapse
|
54
|
Jiang G, Li X, Liu M, Li H, Shen H, liao J, You W, Fang Q, Chen G. Remote ischemic postconditioning ameliorates stroke injury via the SDF-1α/CXCR4 signaling axis in rats. Brain Res Bull 2023; 197:31-41. [PMID: 36990325 DOI: 10.1016/j.brainresbull.2023.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/14/2023] [Accepted: 03/26/2023] [Indexed: 03/29/2023]
Abstract
Remote Ischemic Postconditioning (RIPostC) has become a research hotspot due to its protective effect on the brain in clinical studies related to ischemic stroke. The purpose of this study is to investigate the protective effect of RIPostC after ischemic stroke in rats. The middle cerebral artery occlusion/reperfusion (MCAO/R) model was established by the wire embolization method. RIPostC was obtained by inducing temporary ischemia in the hind limbs of rats. First, based on the results of short-term behavioral measures and long-term neurological function experiments, RIPostC was found to have a protective effect on the MCAO/R model and to improve neurological recovery in rats. Compared to the sham group, RIPostC upregulated the expression levels of C-X-C motif chemokine receptor 4(CXCR4) in the brain and stromal cell-derived factor-1(SDF-1α) in peripheral blood. In addition, RIPostC upregulated CXCR4 expression on CD34+ stem cells in peripheral blood in flow cytometric assays. Meanwhile, according to the results of EdU/DCX co-staining and CD31 staining, it was found that the effect of RIPostC on ameliorating brain injury via SDF-1α/CXCR4 signaling axis may be associated with vascular neogenesis. Finally, after inhibiting the SDF-1α/CXCR4 signaling axis using AMD3100(Plerixafor), we found that the neuroprotective effect of RIPostC was diminished. Taken together, RIPostC can improve neurobehavioral damage induced by MCAO/R in rats, and its mechanism may be related to SDF-1α/CXCR4 signaling axis. Therefore, RIPostC can be used as an intervention strategy for stroke. SDF-1α/CXCR4 signaling axis can also be a potential target for intervention.
Collapse
|
55
|
Zhang W, Han L, Wen Y, Su L, Li Y, Luo X. Electroacupuncture reverses endothelial cell death and promotes angiogenesis through the VEGF/Notch signaling pathway after focal cerebral ischemia-reperfusion injury. Brain Behav 2023; 13:e2912. [PMID: 36786352 PMCID: PMC10013937 DOI: 10.1002/brb3.2912] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 12/18/2022] [Accepted: 01/16/2023] [Indexed: 02/15/2023] Open
Abstract
BACKGROUND Angiogenesis is an important mechanism of recovery from ischemic stroke. Recent studies have found that there is a close relationship between the VEGF/Notch pathway and angiogenesis. It is unknown whether EA can exert a brain protection effect and promote angiogenesis by acting on the VEGF/Notch signaling pathway after focal cerebral ischemia-reperfusion injury (CIRI). METHODS The Middle Cerebral Artery occlusion/Reperfusion (MCAo/R) model was established, in which rats were subjected to occlusion with ischemic intervention for 30 min, followed by reperfusion for 8 h, 1 day, 3 days, and 7 days. The first EA treatment was performed 90 min after the animal model was successfully established, and then EA treatments were performed once a day for 7 days. The 2,3,5-triphenyltetrazolium chloride staining and neurological deficit examination were performed to assess the level of CIRI and neuroprotection by EA. Expression levels of VEGFA, Notch1, and Hes1 proteins were measured via western blotting, while the morphological changes of ECs and microvasculature in the cortex were determined using an ultrastructural observation method. RESULTS EA treatment of PC6, GV26, and SP6 can significantly improve the neurological function of MCAO/R rats, reduce the volume of cerebral infarction, and modulate the ultrastructure of ECs and microvessels in pathological states. Western blotting revealed that EA increased VEGFA protein expression at 8 h and 3 days after CIRI, as well as Notch1 protein expression at 1 and 7 days. Subsequently, EA activated the VEGF/Notch pathway, increasing the expression of the downstream target protein Hes1, reversing EC death, and promoting angiogenesis. CONCLUSION Our findings showed that EA plays a role in promoting angiogenesis following focal CIRI, and we hypothesized that this was due to the regulation of ECs by the EA-activated VEGF/Notch signaling pathway.
Collapse
Affiliation(s)
- Wenyu Zhang
- Department of Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.,Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lin Han
- Department of Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yan Wen
- Department of Traditional Chinese Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Lixian Su
- Department of Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.,Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yibing Li
- Department of Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.,Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xudong Luo
- Department of Information Science and Engineering, Yunnan University, Kunming, Yunnan Province, China
| |
Collapse
|
56
|
Li W, Xu P, Kong L, Feng S, Shen N, Huang H, Wang W, Xu X, Wang X, Wang G, Zhang Y, Sun W, Hu W, Liu X. Elabela-APJ axis mediates angiogenesis via YAP/TAZ pathway in cerebral ischemia/reperfusion injury. Transl Res 2023; 257:78-92. [PMID: 36813109 DOI: 10.1016/j.trsl.2023.02.002] [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: 08/02/2022] [Revised: 01/13/2023] [Accepted: 02/02/2023] [Indexed: 02/24/2023]
Abstract
Angiogenesis helps to improve neurological recovery by repairing damaged brain tissue and restoring cerebral blood flow (CBF). The role of the Elabela (ELA)-Apelin receptor (APJ) system in angiogenesis has gained much attention. We aimed to investigate the function of endothelial ELA on postischemic cerebral angiogenesis. Here, we demonstrated that the endothelial ELA expression was upregulated in the ischemic brain and treatment with ELA-32 mitigated brain injury and enhanced the restoration of CBF and newly formed functional vessels following cerebral ischemia/reperfusion (I/R) injury. Furthermore, ELA-32 incubation potentiated proliferation, migration, and tube formation abilities of the mouse brain endothelial cells (bEnd.3 cells) under oxygen-glucose deprivation/reoxygenation (OGD/R) condition. RNA sequencing analysis indicated that ELA-32 incubation had a role in the Hippo signaling pathway, and improved angiogenesis-related gene expression in OGD/R-exposed bEnd.3 cells. Mechanistically, we depicted that ELA could bind to APJ and subsequently activate YAP/TAZ signaling pathway. Silence of APJ or pharmacological blockade of YAP abolished the pro-angiogenesis effects of ELA-32. Together, these findings highlight the ELA-APJ axis as a potential therapeutic strategy for ischemic stroke by showing how activation of this pathway promotes poststroke angiogenesis.
Collapse
Affiliation(s)
- Wenyu Li
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Pengfei Xu
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| | - Lingqi Kong
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Shuo Feng
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Nan Shen
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Hongmei Huang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Wuxuan Wang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Xiang Xu
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Xinyue Wang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Guoping Wang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Yan Zhang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Wen Sun
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Wei Hu
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Xinfeng Liu
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
57
|
Yang L, Du H, Zhang X, Gao B, Zhang D, Qiao Z, Su X, Bao T, Han S. Circ VRK1/microRNA-17/PTEN axis modulates the angiogenesis of human brain microvascular endothelial cells to affect injury induced by oxygen-glucose deprivation/reperfusion. BMC Neurosci 2023; 24:8. [PMID: 36707796 PMCID: PMC9881374 DOI: 10.1186/s12868-023-00774-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 01/04/2023] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Circular RNAs (circRNAs) can act as microRNA (miRNA) sponges, thus regulating gene expression. The role of circRNAs in the process of oxygen-glucose deprivation/reoxygenation (OGD/R) is unclear. Here, we explored the mechanism underlying Circ VRK1 in human brain microvascular endothelial cells (HBMVECs) injury induced by OGD/R. METHODS The OGD/R cell model was established in HBMVECs. The microarray was applied to detect differentially expressed circRNAs, followed by subcellular fractionation assay. Colony formation assay, flow cytometry, ELISA, tube formation, Transwell and western blot assays were performed for loss-of-function assay. HE staining, TTC staining, immunohistochemistry and western blot were performed in an established mouse model. The relationships between Circ VRK1 and miR-17, and between miR-17 and PTEN were detected by bioinformatics and dual-luciferase assays. Rescue experiments were conducted in vitro and in vivo, and PI3K/AKT activity was detected by Western Blot. RESULTS Circ VRK1, predominantly present in the cytoplasm of cells, was upregulated in the HBMVECs exposed to OGD/R. Circ VRK1 downregulation decreased proliferation, migration, tube formation, inflammatory factors and oxidative stress, while increased apoptosis in HBMVECs. Moreover, Circ VRK1 silencing reduced neurological damage, cerebral infarct size, CD34-positive cell counts and VEGF expression in mice. Circ VRK1 mediated PTEN expression and the PI3K/AKT pathway by targeting miR-17. Deletion of miR-17 inhibited the effects of Circ VRK1 siRNA, and silencing of PTEN suppressed the effects of miR-17 inhibitor. CONCLUSION Circ VRK1 was upregulated during OGD/R. Circ VRK1 downregulation regulates PTEN expression by targeting miR-17, thereby promoting PI3K/AKT pathway activity to alleviate OGD/R injury.
Collapse
Affiliation(s)
- Lei Yang
- Department of Neurosurgery, Shijiazhuang People’s Hospital, No.365, Jianhua South Road, Yuhua District, Shijiazhuang, 050000 Hebei People’s Republic of China
| | - Hong Du
- grid.452702.60000 0004 1804 3009Department of Cardiology, Second Hospital of Hebei Medical University, Shijiazhuang, 050000 Hebei People’s Republic of China
| | - Xuejing Zhang
- Center of Medical Research, Shijiazhuang People’s Hospital, Shijiazhuang, 050000 Hebei People’s Republic of China
| | - Bulang Gao
- Center of Medical Research, Shijiazhuang People’s Hospital, Shijiazhuang, 050000 Hebei People’s Republic of China
| | - Dongliang Zhang
- Department of Neurosurgery, Shijiazhuang People’s Hospital, No.365, Jianhua South Road, Yuhua District, Shijiazhuang, 050000 Hebei People’s Republic of China
| | - Zongrong Qiao
- Department of Neurosurgery, Shijiazhuang People’s Hospital, No.365, Jianhua South Road, Yuhua District, Shijiazhuang, 050000 Hebei People’s Republic of China
| | - Xianhui Su
- Department of Neurosurgery, Shijiazhuang People’s Hospital, No.365, Jianhua South Road, Yuhua District, Shijiazhuang, 050000 Hebei People’s Republic of China
| | - Tong Bao
- Department of Neurosurgery, Shijiazhuang People’s Hospital, No.365, Jianhua South Road, Yuhua District, Shijiazhuang, 050000 Hebei People’s Republic of China
| | - Siqin Han
- grid.256883.20000 0004 1760 8442Graduate School, Hebei Medical University, Shijiazhuang, 050017 Hebei People’s Republic of China
| |
Collapse
|
58
|
Zhou Q, Nozdriukhin D, Chen Z, Glandorf L, Hofmann UAT, Reiss M, Tang L, Deán‐Ben XL, Razansky D. Depth-Resolved Localization Microangiography in the NIR-II Window. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 10:e2204782. [PMID: 36403231 PMCID: PMC9811471 DOI: 10.1002/advs.202204782] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/17/2022] [Indexed: 06/16/2023]
Abstract
Detailed characterization of microvascular alterations requires high-resolution 3D imaging methods capable of providing both morphological and functional information. Existing optical microscopy tools are routinely used for microangiography, yet offer suboptimal trade-offs between the achievable field of view and spatial resolution with the intense light scattering in biological tissues further limiting the achievable penetration depth. Herein, a new approach for volumetric deep-tissue microangiography based on stereovision combined with super-resolution localization imaging is introduced that overcomes the spatial resolution limits imposed by light diffusion and optical diffraction in wide-field imaging configurations. The method capitalizes on localization and tracking of flowing fluorescent particles in the second near-infrared window (NIR-II, ≈1000-1700 nm), with the third (depth) dimension added by triangulation and stereo-matching of images acquired with two short-wave infrared cameras operating in a dual-view mode. The 3D imaging capability enabled with the proposed method facilitates a detailed visualization of microvascular networks and an accurate blood flow quantification. Experiments performed in tissue-mimicking phantoms demonstrate that high resolution is preserved up to a depth of 4 mm in a turbid medium. Transcranial microangiography of the entire murine cortex and penetrating vessels is further demonstrated at capillary level resolution.
Collapse
Affiliation(s)
- Quanyu Zhou
- Institute of Pharmacology and Toxicology and Institute for Biomedical EngineeringFaculty of MedicineUniversity of ZurichZurich8057Switzerland
- Institute for Biomedical EngineeringDepartment of Information Technology and Electrical EngineeringETH ZurichZurich8093Switzerland
| | - Daniil Nozdriukhin
- Institute of Pharmacology and Toxicology and Institute for Biomedical EngineeringFaculty of MedicineUniversity of ZurichZurich8057Switzerland
- Institute for Biomedical EngineeringDepartment of Information Technology and Electrical EngineeringETH ZurichZurich8093Switzerland
| | - Zhenyue Chen
- Institute of Pharmacology and Toxicology and Institute for Biomedical EngineeringFaculty of MedicineUniversity of ZurichZurich8057Switzerland
- Institute for Biomedical EngineeringDepartment of Information Technology and Electrical EngineeringETH ZurichZurich8093Switzerland
| | - Lukas Glandorf
- Institute of Pharmacology and Toxicology and Institute for Biomedical EngineeringFaculty of MedicineUniversity of ZurichZurich8057Switzerland
- Institute for Biomedical EngineeringDepartment of Information Technology and Electrical EngineeringETH ZurichZurich8093Switzerland
| | - Urs A. T. Hofmann
- Institute of Pharmacology and Toxicology and Institute for Biomedical EngineeringFaculty of MedicineUniversity of ZurichZurich8057Switzerland
- Institute for Biomedical EngineeringDepartment of Information Technology and Electrical EngineeringETH ZurichZurich8093Switzerland
| | - Michael Reiss
- Institute of Pharmacology and Toxicology and Institute for Biomedical EngineeringFaculty of MedicineUniversity of ZurichZurich8057Switzerland
- Institute for Biomedical EngineeringDepartment of Information Technology and Electrical EngineeringETH ZurichZurich8093Switzerland
| | - Lin Tang
- Institute of Pharmacology and Toxicology and Institute for Biomedical EngineeringFaculty of MedicineUniversity of ZurichZurich8057Switzerland
- Institute for Biomedical EngineeringDepartment of Information Technology and Electrical EngineeringETH ZurichZurich8093Switzerland
| | - Xosé Luís Deán‐Ben
- Institute of Pharmacology and Toxicology and Institute for Biomedical EngineeringFaculty of MedicineUniversity of ZurichZurich8057Switzerland
- Institute for Biomedical EngineeringDepartment of Information Technology and Electrical EngineeringETH ZurichZurich8093Switzerland
| | - Daniel Razansky
- Institute of Pharmacology and Toxicology and Institute for Biomedical EngineeringFaculty of MedicineUniversity of ZurichZurich8057Switzerland
- Institute for Biomedical EngineeringDepartment of Information Technology and Electrical EngineeringETH ZurichZurich8093Switzerland
| |
Collapse
|
59
|
Fan LS, Chen YC, Liao RJ, Zhao YY, Zhang XN, Chen Z, Jiang L, Hu WW. Antagonism of histamine H 3 receptor promotes angiogenesis following focal cerebral ischemia. Acta Pharmacol Sin 2022; 43:2807-2816. [PMID: 35581293 PMCID: PMC9622736 DOI: 10.1038/s41401-022-00916-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 04/25/2022] [Indexed: 11/08/2022]
Abstract
Our previous study showed that H3 receptor antagonists reduced neuronal apoptosis and cerebral infarction in the acute stage after cerebral ischemia, but through an action independent of activation of histaminergic neurons. Because enhanced angiogenesis facilitates neurogenesis and neurological recovery after ischemic stroke, we herein investigated whether antagonism of H3R promoted angiogenesis after brain ischemia. Photothrombotic stroke was induced in mice. We showed that administration of H3R antagonist thioperamide (THIO, 10 mg·kg-1·d-1, i.p., from D1 after cerebral ischemia) significantly improved angiogenesis assessed on D14, and attenuated neurological defects on D28 after cerebral ischemia. Compared with wild-type mice, Hrh3-/- mice displayed more blood vessels in the ischemic boundary zone on D14, and THIO administration did not promote angiogenesis in these knockout mice. THIO-promoted angiogenesis in mice was reversed by i.c.v. injection of H3R agonist immepip, but not by H1 and H2 receptor antagonists, histidine decarboxylase inhibitor α-fluoromethylhistidine, or histidine decarboxylase gene knockout (HDC-/-), suggesting that THIO-promoted angiogenesis was independent of activation of histaminergic neurons. In vascular endothelial cells (bEnd.3), THIO (10-9-10-7 M) dose-dependently facilitated cell migration and tube formation after oxygen glucose deprivation (OGD), and H3R knockdown caused similar effects. We further revealed that H3R antagonism reduced the interaction between H3R and Annexin A2, while knockdown of Annexin A2 abrogated THIO-promoted angiogenesis in bEnd.3 cells after OGD. Annexin A2-overexpressing mice displayed more blood vessels in the ischemic boundary zone, which was reversed by i.c.v. injection of immepip. In conclusion, this study demonstrates that H3R antagonism promotes angiogenesis after cerebral ischemia, which is independent of activation of histaminergic neurons, but related to the H3R on vascular endothelial cells and its interaction with Annexin A2. Thus, H3R antagonists might be promising drug candidates to improve angiogenesis and neurological recovery after ischemic stroke.
Collapse
Affiliation(s)
- Li-Shi Fan
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, Department of Anatomy, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - You-Chao Chen
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, Department of Anatomy, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Ru-Jia Liao
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, Department of Anatomy, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Yan-Yan Zhao
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, Department of Anatomy, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Xiang-Nan Zhang
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, Department of Anatomy, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Zhong Chen
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, Department of Anatomy, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University School of Medicine, Hangzhou, 310000, China
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310000, China
| | - Lei Jiang
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, Department of Anatomy, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University School of Medicine, Hangzhou, 310000, China.
| | - Wei-Wei Hu
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, Department of Anatomy, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University School of Medicine, Hangzhou, 310000, China.
| |
Collapse
|
60
|
Kwon B, Song Y, Choi YH, Suh DC. Physiologic Flow Diversion Coiling Technique for Wide-Necked Aneurysms with an Asymmetric Bidirectional Flow at the Aneurysm Neck. Neurointervention 2022; 17:133-142. [PMID: 36167347 PMCID: PMC9626610 DOI: 10.5469/neuroint.2022.00311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/09/2022] [Accepted: 08/27/2022] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Wide-necked aneurysms in the circle of Willis (CoW) are prone to recur due to reciprocal bidirectional flow. We present a novel concept of coil embolization to prevent recurrence that uses physiologic flow diversion at the CoW. MATERIALS AND METHODS We enrolled 14 patients (15 aneurysms) who underwent aneurysm coiling for wide-necked aneurysms with asymmetric bidirectional inflow into the aneurysm. Four patients had recurrent aneurysms after coiling. The concept of physiologic flow diversion included obliterating antegrade flow into the aneurysm sac as well as opposite CoW flow by performing compact coil packing with intentional protrusion out of the aneurysm neck to the communicating part. RESULTS Fifteen aneurysms, including 4 recurrent aneurysms, in an anterior communicating artery (n=7), posterior communicating artery (n=5), and tip of the basilar artery (n=3) were treated with coil embolization (n=10) and stent-assisted coiling (n=5). All aneurysms had a wide neck, and the mean largest diameter was 9.0 mm. The mean packing density was 45.1%. Twelve aneurysms were completely occluded, and 3 aneurysms had tiny residual neck remnants. There was neither a neurological event nor recurrence during the mean 12.5 months of follow-up. CONCLUSION Wide-necked aneurysms at the CoW tend to recur. As a strategy to prevent a recurrence, physiologic flow diversion can be an option in treating wide-necked aneurysms in the CoW.
Collapse
Affiliation(s)
- Boseong Kwon
- Neurointervention Clinic, Department of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Yunsun Song
- Neurointervention Clinic, Department of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Yun Hyeok Choi
- Neurointervention Clinic, Department of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Dae Chul Suh
- Neurointervention Clinic, Department of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
- Department of Neurointervention, GangNam St. Peter’s Hospital, Seoul, Korea
| |
Collapse
|
61
|
McCrary MR, Jiang MQ, Jesson K, Gu X, Logun MT, Wu A, Gonsalves N, Karumbaiah L, Yu SP, Wei L. Glycosaminoglycan scaffolding and neural progenitor cell transplantation promotes regenerative immunomodulation in the mouse ischemic brain. Exp Neurol 2022; 357:114177. [PMID: 35868359 PMCID: PMC10066865 DOI: 10.1016/j.expneurol.2022.114177] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 07/04/2022] [Accepted: 07/16/2022] [Indexed: 02/08/2023]
Abstract
Ischemic stroke is a leading cause of morbidity and mortality, with limited treatments that can facilitate brain regeneration. Neural progenitor cells (NPCs) hold promise for replacing tissue lost to stroke, and biomaterial approaches may improve their efficacy to overcome hurdles in clinical translation. The immune response and its role in stroke pathogenesis and regeneration may interplay with critical mechanisms of stem cell and biomaterial therapies. Cellular therapy can modulate the immune response to reduce toxic neuroinflammation early after ischemia. However, few studies have attempted to harness the regenerative effects of neuroinflammation to augment recovery. Our previous studies demonstrated that intracerebrally transplanted NPCs encapsulated in a chondroitin sulfate-A hydrogel (CS-A + NPCs) can improve vascular regeneration after stroke. In this paper, we found that CS-A + NPCs affect the microglia/macrophage response to promote a regenerative phenotype following stroke in mice. Following transplantation, PPARγ-expressing microglia/macrophages, and MCP-1 and IL-10 protein levels are enhanced. Secreted immunomodulatory factor expression of other factors was altered compared to NPC transplantation alone. Post-stroke depression-like behavior was reduced following cellular and material transplantation. Furthermore, we showed in cultures that microglia/macrophages encapsulated in CS-A had increased expression of angiogenic and arteriogenic mediators. Neutralization with anti-IL-10 antibody negated these effects in vitro. Cumulatively, this work provides a framework for understanding the mechanisms by which immunomodulatory biomaterials can enhance the regenerative effects of cellular therapy for ischemic stroke and other brain injuries.
Collapse
Affiliation(s)
- Myles R McCrary
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Michael Q Jiang
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA; Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, GA, USA
| | - Kaleena Jesson
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Xiaohuan Gu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA; Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, GA, USA
| | - Meghan T Logun
- Regenerative Bioscience Center, University of Georgia, Athens, GA, USA; Division of Neuroscience, Biomedical and Translational Sciences Institute, University of Georgia, Athens, GA, USA
| | - Anika Wu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Nathan Gonsalves
- Regenerative Bioscience Center, University of Georgia, Athens, GA, USA; Division of Neuroscience, Biomedical and Translational Sciences Institute, University of Georgia, Athens, GA, USA
| | - Lohitash Karumbaiah
- Regenerative Bioscience Center, University of Georgia, Athens, GA, USA; Division of Neuroscience, Biomedical and Translational Sciences Institute, University of Georgia, Athens, GA, USA; Department of Animal and Dairy Science, College of Agriculture and Environmental Sciences, University of Georgia, Athens, GA, USA
| | - Shan Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA; Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, GA, USA
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
62
|
Cheng K, Wang Z, Bai J, Xiong J, Chen J, Ni J. Research advances in the application of vagus nerve electrical stimulation in ischemic stroke. Front Neurosci 2022; 16:1043446. [PMID: 36389255 PMCID: PMC9650138 DOI: 10.3389/fnins.2022.1043446] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 10/10/2022] [Indexed: 12/04/2022] Open
Abstract
Stroke seriously endangers human well-being and brings a severe burden to family and society. Different post-stroke dysfunctions result in an impaired ability to perform activities of daily living. Standard rehabilitative therapies may not meet the requirements for functional improvement after a stroke; thus, alternative approaches need to be proposed. Currently, vagus nerve stimulation (VNS) is clinically applied for the treatment of epilepsy, depression, cluster headache and migraine, while its treatment of various dysfunctions after an ischemic stroke is still in the clinical research stage. Recent studies have confirmed that VNS has neuroprotective effects in animal models of transient and permanent focal cerebral ischemia, and that its combination with rehabilitative training significantly improves upper limb motor dysfunction and dysphagia. In addition, vagus-related anatomical structures and neurotransmitters are closely implicated in memory–cognition enhancement processes, suggesting that VNS is promising as a potential treatment for cognitive dysfunction after an ischemic stroke. In this review, we outline the current status of the application of VNS (invasive and non-invasive) in diverse functional impairments after an ischemic stroke, followed by an in-depth discussion of the underlying mechanisms of its mediated neuroprotective effects. Finally, we summarize the current clinical implementation challenges and adverse events of VNS and put forward some suggestions for its future research direction. Research on VNS for ischemic stroke has reached a critical stage. Determining how to achieve the clinical transformation of this technology safely and effectively is important, and more animal and clinical studies are needed to clarify its therapeutic mechanism.
Collapse
|
63
|
Sayin ES, Sobczyk O, Poublanc J, Mikulis DJ, Fisher JA, Kuo KHM, Duffin J. Assessment of cerebrovascular function in patients with sickle cell disease using transfer function analysis. Physiol Rep 2022; 10:e15472. [PMID: 36200271 PMCID: PMC9535348 DOI: 10.14814/phy2.15472] [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: 07/04/2022] [Revised: 08/29/2022] [Accepted: 09/01/2022] [Indexed: 11/07/2022] Open
Abstract
In patients with sickle cell disease (SCD), the delivery of oxygen to the brain is compromised by anemia, abnormal rheology, and steno-occlusive vascular disease. Successful compensation depends on an increase in oxygen supply such as that provided by an increase in cerebral blood flow (CBF). We used magnetic resonance imaging to provide a high-resolution assessment of the ability of SCD patients to respond to a vasoactive stimulus in middle, anterior, and posterior cerebral artery territories for both white and gray matter. Cerebrovascular reactivity (CVR) was measured as the blood oxygen level dependent signal (a surrogate for CBF) response to an increase in the end tidal partial pressure of CO2 (PET CO2 ). The dynamic aspect of the response was measured as the time constant of the first order response kinetics (tau). To confirm and support these findings we used an alternative examination of the response, transfer function analysis (TFA), to measure the responsiveness (gain), the speed of response (phase), and the consistency of the response over time (coherence). We tested 34 patients with SCD and compared the results to those of 24 healthy controls participants. The results from a three-way ANOVA showed that patients with SCD have reduced CVR (p < 0.001) and lower coherence (p < 0.001) in gray matter and white matter and reduced gain in gray matter only (p < 0.001). In terms of the speed of the response to CO2 , tau (p < 0.001) and TFA phase (p < 0.001) were increased in SCD patients compared to healthy control subjects. These findings show that the cerebrovascular responsiveness to CO2 in patients with SCD is both decreased and slowed compared to healthy controls.
Collapse
Affiliation(s)
- Ece Su Sayin
- Department of PhysiologyUniversity of TorontoTorontoCanada
- Departments of Anaesthesia and Pain ManagementUniversity Health NetworkTorontoCanada
| | - Olivia Sobczyk
- Department of PhysiologyUniversity of TorontoTorontoCanada
- Departments of Anaesthesia and Pain ManagementUniversity Health NetworkTorontoCanada
- Joint Department of Medical Imaging and the Functional Neuroimaging LaboratoryUniversity Health NetworkTorontoCanada
| | - Julien Poublanc
- Joint Department of Medical Imaging and the Functional Neuroimaging LaboratoryUniversity Health NetworkTorontoCanada
| | - David J. Mikulis
- Joint Department of Medical Imaging and the Functional Neuroimaging LaboratoryUniversity Health NetworkTorontoCanada
- Institute of Medical SciencesUniversity of TorontoTorontoCanada
| | - Joseph A. Fisher
- Department of PhysiologyUniversity of TorontoTorontoCanada
- Departments of Anaesthesia and Pain ManagementUniversity Health NetworkTorontoCanada
| | - Kevin H. M. Kuo
- Division of Medical Oncology and Hematology, Department of MedicineUniversity of TorontoTorontoOntarioCanada
| | - James Duffin
- Department of PhysiologyUniversity of TorontoTorontoCanada
- Departments of Anaesthesia and Pain ManagementUniversity Health NetworkTorontoCanada
| |
Collapse
|
64
|
Yang H, Han M, Li J, Ke H, Kong Y, Wang W, Wang L, Ma W, Qiu J, Wang X, Xin T, Liu H. Delivery of miRNAs through Metal-Organic Framework Nanoparticles for Assisting Neural Stem Cell Therapy for Ischemic Stroke. ACS NANO 2022; 16:14503-14516. [PMID: 36065995 DOI: 10.1021/acsnano.2c04886] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Stroke is the most common cause of disability globally. Neural stem cell (NSC) therapy, which can replace lost and damaged neurons, has been proposed as a potential treatment for stroke. The therapeutic efficacy of NSC therapy is hindered by the fact that only a small number of NSCs undergo neuronal differentiation. Neuron-specific miR-124, which promotes the differentiation of NSCs into mature neurons, can be combined with NSC therapy to cure ischemic stroke. However, the instability and poor internalization of miR-124 seriously hamper its broad clinical application. Herein, an innovative strategy involving delivery of miR-124 via a Ca-MOF@miR-124 nanodelivery system, which effectively prevents the degradation of miR-124 by nucleases and promotes the internalization of miR-124 by NSCs, is presented. The effect of accelerated neuronal directed differentiation of NSCs was assessed through in vitro cell experiments, and the clinical application potential of this nanodelivery system for the treatment of ischemic stroke was assessed through in vivo experiments involving the combination of NSC therapy and Ca-MOF@miR-124 nanoparticles. The results indicate that Ca-MOF@miR-124 nanoparticles can promote the differentiation of NSCs into mature neurons with electrophysiological function within 5 days. The differentiation rate of cells treated with Ca-MOF@miR-124 nanoparticles was at least 5 days faster than that of untreated cells. Moreover, Ca-MOF@miR-124 nanoparticles decreased the ischemic area to almost normal levels by day 7. The combination of Ca-MOF@miR-124 nanoparticles and NSC therapy will enhance the treatment of traumatic nerve injury and neurodegenerative diseases.
Collapse
Affiliation(s)
- Hongru Yang
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, Shandong 250100, People's Republic of China
| | - Min Han
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, People's Republic of China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, People's Republic of China
| | - Jian Li
- State Key Laboratory of Separation Membranes and Membrane Processes, Tiangong University, Tianjin 300387, People's Republic of China
| | - Hongfei Ke
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, People's Republic of China
| | - Ying Kong
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, Shandong 250100, People's Republic of China
| | - Wenhan Wang
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, Shandong 250100, People's Republic of China
| | - Liang Wang
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, Shandong 250100, People's Republic of China
| | - Wenjun Ma
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, Shandong 250100, People's Republic of China
| | - Jichuan Qiu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, Shandong 250100, People's Republic of China
| | - Xiwei Wang
- Institute of Novel Semiconductors, Shandong University, Jinan, Shandong 250100, People's Republic of China
| | - Tao Xin
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, People's Republic of China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, People's Republic of China
| | - Hong Liu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, Shandong 250100, People's Republic of China
- Institute for Advanced Interdisciplinary Research, University of Jinan, Jinan, Shandong 250003, People's Republic of China
| |
Collapse
|
65
|
Sato Y, Falcone-Juengert J, Tominaga T, Su H, Liu J. Remodeling of the Neurovascular Unit Following Cerebral Ischemia and Hemorrhage. Cells 2022; 11:2823. [PMID: 36139398 PMCID: PMC9496956 DOI: 10.3390/cells11182823] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/18/2022] [Accepted: 08/30/2022] [Indexed: 11/23/2022] Open
Abstract
Formulated as a group effort of the stroke community, the transforming concept of the neurovascular unit (NVU) depicts the structural and functional relationship between brain cells and the vascular structure. Composed of both neural and vascular elements, the NVU forms the blood-brain barrier that regulates cerebral blood flow to meet the oxygen demand of the brain in normal physiology and maintain brain homeostasis. Conversely, the dysregulation and dysfunction of the NVU is an essential pathological feature that underlies neurological disorders spanning from chronic neurodegeneration to acute cerebrovascular events such as ischemic stroke and cerebral hemorrhage, which were the focus of this review. We also discussed how common vascular risk factors of stroke predispose the NVU to pathological changes. We synthesized existing literature and first provided an overview of the basic structure and function of NVU, followed by knowledge of how these components remodel in response to ischemic stroke and brain hemorrhage. A greater understanding of the NVU dysfunction and remodeling will enable the design of targeted therapies and provide a valuable foundation for relevant research in this area.
Collapse
Affiliation(s)
- Yoshimichi Sato
- Department of Neurological Surgery, UCSF, San Francisco, CA 94158, USA
- Department of Neurological Surgery, SFVAMC, San Francisco, CA 94158, USA
- Department of Neurosurgery, Graduate School of Medicine, Tohoku University, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Jaime Falcone-Juengert
- Department of Neurological Surgery, UCSF, San Francisco, CA 94158, USA
- Department of Neurological Surgery, SFVAMC, San Francisco, CA 94158, USA
| | - Teiji Tominaga
- Department of Neurosurgery, Graduate School of Medicine, Tohoku University, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Hua Su
- Department of Anesthesia, UCSF, San Francisco, CA 94143, USA
- Center for Cerebrovascular Research, UCSF, San Francisco, CA 94143, USA
| | - Jialing Liu
- Department of Neurological Surgery, UCSF, San Francisco, CA 94158, USA
- Department of Neurological Surgery, SFVAMC, San Francisco, CA 94158, USA
| |
Collapse
|
66
|
Deshpande A, Elliott J, Kari N, Jiang B, Michel P, Toosizadeh N, Fahadan PT, Kidwell C, Wintermark M, Laksari K. Novel imaging markers for altered cerebrovascular morphology in aging, stroke, and Alzheimer's disease. J Neuroimaging 2022; 32:956-967. [PMID: 35838658 PMCID: PMC9474631 DOI: 10.1111/jon.13023] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 06/26/2022] [Accepted: 06/29/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Altered brain vasculature is a key phenomenon in several neurologic disorders. This paper presents a quantitative assessment of the anatomical variations in the Circle of Willis (CoW) and vascular morphology in healthy aging, acute ischemic stroke (AIS) and Alzheimer's Disease (AD). METHODS We used our novel automatic method to segment and extract geometric features of the cerebral vasculature from MR angiography scans of 175 healthy subjects, which were used to create a probabilistic atlas of cerebrovasculature and to study normal aging and intersubject variations in CoW anatomy. Subsequently, we quantified and analyzed vascular alterations in 45AIS and 50 AD patients, two prominent cerebrovascular and neurodegenerative disorders. RESULTS In the sampled cohort, we determined that the CoW is fully formed in only 35% of healthy adults and found significantly (p < .05) increased tortuosity and fractality, with increasing age and also with disease in both AIS and AD. We also found significantly lower vessel length, volume, and number of branches in AIS patients, as expected. The AD cerebral vessels exhibited significantly smaller diameter and more complex branching patterns, compared to age-matched healthy adults. These changes were significantly heightened (p < .05) among healthy, early onset mild AD, and moderate/severe dementia groups. CONCLUSION Although our study does not include longitudinal data due to paucity of such datasets, the specific geometric features and quantitative comparisons demonstrate the potential for using vascular morphology as a noninvasive imaging biomarker for neurologic disorders.
Collapse
Affiliation(s)
| | - Jordan Elliott
- Department of Biomedical Engineering, University of Arizona
| | - Nitya Kari
- Department of Biomedical Engineering, University of Arizona
| | - Bin Jiang
- Department of Radiology, Stanford University
| | - Patrik Michel
- Department of Neurology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Nima Toosizadeh
- Department of Biomedical Engineering, University of Arizona
- Arizona Center on Aging, Department of Medicine, University of Arizona
| | - Pouya Tahsili Fahadan
- Neuroscience Intensive Care Unit, Medical Critical Care Service and Department of Medical Education, University of Virginia School of Medicine, Inova Fairfax Medical Campus
- Departments of Neurology, Johns Hopkins University School of Medicine
| | | | | | - Kaveh Laksari
- Department of Biomedical Engineering, University of Arizona
- Department of Aerospace and Mechanical Engineering, University of Arizona
| |
Collapse
|
67
|
Zhang Y, Liu L, Zhao X, Yan S, Zeng F, Zhou D. New insight into ischemic stroke: Circadian rhythm in post-stroke angiogenesis. Front Pharmacol 2022; 13:927506. [PMID: 36016550 PMCID: PMC9395980 DOI: 10.3389/fphar.2022.927506] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 07/07/2022] [Indexed: 11/29/2022] Open
Abstract
The circadian rhythm is an endogenous clock system that coordinates and optimizes various physiological and pathophysiological processes, which accord with the master and the peripheral clock. Increasing evidence indicates that endogenous circadian rhythm disruption is involved in the lesion volume and recovery of ischemic stroke. As a critical recovery mechanism in post-stroke, angiogenesis reestablishes the regional blood supply and enhances cognitive and behavioral abilities, which is mainly composed of the following processes: endothelial cell proliferation, migration, and pericyte recruitment. The available evidence revealed that the circadian governs many aspects of angiogenesis. This study reviews the mechanism by which circadian rhythms regulate the process of angiogenesis and its contribution to functional recovery in post-stroke at the aspects of the molecular level. A comprehensive understanding of the circadian clock regulating angiogenesis in post-stroke is expected to develop new strategies for the treatment of cerebral infarction.
Collapse
Affiliation(s)
- Yuxing Zhang
- The Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha, China
| | - Lijuan Liu
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xin Zhao
- The Medical School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Siyang Yan
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Fukang Zeng
- The Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha, China
| | - Desheng Zhou
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
- *Correspondence: Desheng Zhou,
| |
Collapse
|
68
|
Association between Deep Medullary Veins in the Unaffected Hemisphere and Functional Outcome in Acute Cardioembolic Stroke: An Observational Retrospective Study. Brain Sci 2022; 12:brainsci12080978. [PMID: 35892419 PMCID: PMC9330894 DOI: 10.3390/brainsci12080978] [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: 06/16/2022] [Revised: 07/16/2022] [Accepted: 07/20/2022] [Indexed: 02/05/2023] Open
Abstract
Objective: To explore whether deep medullary veins (DMVs) in the unaffected hemisphere were associated with functional outcome in acute cardioembolic stroke patients. Methods: Acute cardioembolic stroke patients at a single center were retrospectively included. DMVs visibility in the unaffected hemisphere was assessed using a well-established four-grade scoring method based on susceptibility-weighted imaging (SWI): grades 0−3 (grade 0 for no visible DMVs; grade 1 for the numbers of conspicuous DMVs < 5; grade 2 for numbers raging from 5 to 10; grade 3 for more than 10). Patients were further divided into mild-to-moderate (grade 0−2) and severe DMVs (grade 3) groups. Functional outcomes were evaluated using the modified Rankin scale (mRS) score at three months. Poor outcome was defined as mRS ≥ 3. Binary logistic regression analysis was used to explore the association between DMVs grade and functional outcome. Results: A total of 170 patients were finally included. Compared with the mild-to-moderate DMVs group (149 patients), the severe DMVs group (21 patients) had higher baseline National Institutes of Health Stroke Scale (NIHSS) scores (p = 0.002), lower levels of admission systolic blood pressure (BP) (p = 0.031), and elevated rates of large infarction (p = 0.003). At three months, the severe DMVs group had higher mRS (p = 0.002). Patients in the poor outcome group (82/170, 48.2%) had older age, higher baseline NIHSS score, lower admission diastolic BP, higher rates of hemorrhagic transformation and large infarction, and an increased proportion of severe DMVs (all p < 0.05). After adjusting for confounders, multivariable regression analysis showed that the severe DMVs grade (adjusted odds ratio [OR] = 5.830, 95% confidence interval [CI] = 1.266−26.856, p = 0.024) was significantly associated with three-month functional outcomes without interaction with other potential risk factors (p for interaction > 0.05). Conclusions: DMVs grade in the unaffected hemisphere was independently associated with three-month functional outcome in acute cardioembolic stroke patients. Patients with severe DMVs were more likely to have a poor functional outcome at three months.
Collapse
|
69
|
Brainer Clares de Andrade J, Mohr JP, Oliveira Lima F, José de Freitas Carvalho J, Andre Castro de Oliveira R, Coelho Maia Barros L, Sampaio Silva G. Predictors of hemorrhagic transformation differences between patients treated or not with reperfusion therapy. J Clin Neurosci 2022; 101:9-15. [DOI: 10.1016/j.jocn.2022.04.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 02/08/2022] [Accepted: 04/23/2022] [Indexed: 11/25/2022]
|
70
|
Chen J, He J, Luo L. Brain vascular damage-induced lymphatic ingrowth is directed by Cxcl12b/Cxcr4a. Development 2022; 149:275687. [PMID: 35694896 DOI: 10.1242/dev.200729] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 06/06/2022] [Indexed: 12/15/2022]
Abstract
After ischemic stroke, promotion of vascular regeneration without causing uncontrolled vessel growth appears to be the major challenge for pro-angiogenic therapies. The molecular mechanisms underlying how nascent blood vessels (BVs) are correctly guided into the post-ischemic infarction area remain unknown. Here, using a zebrafish cerebrovascular injury model, we show that chemokine signaling provides crucial guidance cues to determine the growing direction of ingrown lymphatic vessels (iLVs) and, in turn, that of nascent BVs. The chemokine receptor Cxcr4a is transcriptionally activated in the iLVs after injury, whereas its ligand Cxcl12b is expressed in the residual central BVs, the destinations of iLV ingrowth. Mutant and mosaic studies indicate that Cxcl12b/Cxcr4a-mediated chemotaxis is necessary and sufficient to determine the growing direction of iLVs and nascent BVs. This study provides a molecular basis for how the vessel directionality of cerebrovascular regeneration is properly determined, suggesting potential application of Cxcl12b/Cxcr4a in the development of post-ischemic pro-angiogenic therapies.
Collapse
Affiliation(s)
- Jingying Chen
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, 400715 Chongqing, China.,University of Chinese Academy of Sciences (Chongqing), Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Beibei, 400714 Chongqing, China
| | - Jianbo He
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, 400715 Chongqing, China
| | - Lingfei Luo
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, 400715 Chongqing, China.,University of Chinese Academy of Sciences (Chongqing), Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Beibei, 400714 Chongqing, China
| |
Collapse
|
71
|
He T, Yang GY, Zhang Z. Crosstalk of Astrocytes and Other Cells during Ischemic Stroke. LIFE (BASEL, SWITZERLAND) 2022; 12:life12060910. [PMID: 35743941 PMCID: PMC9228674 DOI: 10.3390/life12060910] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/04/2022] [Accepted: 06/07/2022] [Indexed: 12/27/2022]
Abstract
Stroke is a leading cause of death and long-term disability worldwide. Astrocytes structurally compose tripartite synapses, blood–brain barrier, and the neurovascular unit and perform multiple functions through cell-to-cell signaling of neurons, glial cells, and vasculature. The crosstalk of astrocytes and other cells is complicated and incompletely understood. Here we review the role of astrocytes in response to ischemic stroke, both beneficial and detrimental, from a cell–cell interaction perspective. Reactive astrocytes provide neuroprotection through antioxidation and antiexcitatory effects and metabolic support; they also contribute to neurorestoration involving neurogenesis, synaptogenesis, angiogenesis, and oligodendrogenesis by crosstalk with stem cells and cell lineage. In the meantime, reactive astrocytes also play a vital role in neuroinflammation and brain edema. Glial scar formation in the chronic phase hinders functional recovery. We further discuss astrocyte enriched microRNAs and exosomes in the regulation of ischemic stroke. In addition, the latest notion of reactive astrocyte subsets and astrocytic activity revealed by optogenetics is mentioned. This review discusses the current understanding of the intimate molecular conversation between astrocytes and other cells and outlines its potential implications after ischemic stroke. “Neurocentric” strategies may not be sufficient for neurological protection and recovery; future therapeutic strategies could target reactive astrocytes.
Collapse
Affiliation(s)
- Tingting He
- Department of Neurology, Shanghai Tenth People’s Hospital, Tongji University, Shanghai 200072, China;
- Neuroscience and Neuroengineering Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Guo-Yuan Yang
- Neuroscience and Neuroengineering Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
- Correspondence: (G.-Y.Y.); (Z.Z.); Tel.: +86-21-62933186 (G.-Y.Y.); Fax: +86-21-62932302 (G.-Y.Y.)
| | - Zhijun Zhang
- Neuroscience and Neuroengineering Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
- Correspondence: (G.-Y.Y.); (Z.Z.); Tel.: +86-21-62933186 (G.-Y.Y.); Fax: +86-21-62932302 (G.-Y.Y.)
| |
Collapse
|
72
|
Chen S, Zhang J, Li M, Zhou J, Zhang Y. Danhong injection combined with tPA protects the BBB through Notch-VEGF signaling pathway on long-term outcomes of thrombolytic therapy. Biomed Pharmacother 2022; 153:113288. [PMID: 35717787 DOI: 10.1016/j.biopha.2022.113288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/08/2022] [Accepted: 06/09/2022] [Indexed: 11/17/2022] Open
Abstract
Current therapy for ischemic stroke primarily relies on tissue plasminogen activator (tPA), but it is limited by narrow treatment time window, bleeding complications and neurotoxicity. The preliminary study of tPA plus Danhong injection (DHI) shows that it can significantly reduce the side effects of tPA and improve its thrombolytic effect, but the mechanism of this action has not been further studied. In this study, the rats were randomly divided into sham group, vehicle group, DHI group (4 mL/kg), tPA group (5 mg/kg) and DHI+tPA group (4 mL/kg+ 2.5 mg/kg), administered intravenously 4.5 h since focal embolic stroke modeling. After 3 days and 7 days of cerebral ischemia, the neurological function of each treatment group was significantly improved compared with the vehicle group. The combination of DHI and tPA significantly reduced Evans blue (EB) penetration as well as the expressions of the proteins MMP-9, PAI-1 and P-selectin, while upregulating the expressions of claudin-5, occludin, and ZO-1 mRNA. Furthermore, the effect of continuous 7-day treatment was more conspicuous than 3-day treatment. Then, it significantly reduced the expressions of the proteins DLL-4 and VEGFR-2, increased the expressions of Notch-1, HIF-1α and HES-1 mRNA, and promoted the expressions of VEGF/HIF-1α-positive cells at 14 days following stroke. Hematoxylin-eosin (HE) staining and transmission electron microscopy (TEM) also showed that it improved pathological changes of ischemic brain tissue and the cerebral cortex micro-structure. These indicate that DHI combined with tPA may significantly ameliorate blood-brain barrier (BBB) disruption by activating Notch-VEGF signaling pathway to promote angiogenesis for long-term outcomes.
Collapse
Affiliation(s)
- Simiao Chen
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, China.
| | - Jinghui Zhang
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, China.
| | - Min Li
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, China.
| | - Jing Zhou
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, China.
| | - Yuyan Zhang
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, China.
| |
Collapse
|
73
|
Kemps H, Dessy C, Dumas L, Sonveaux P, Alders L, Van Broeckhoven J, Font LP, Lambrichts S, Foulquier S, Hendrix S, Brône B, Lemmens R, Bronckaers A. Extremely low frequency electromagnetic stimulation reduces ischemic stroke volume by improving cerebral collateral blood flow. J Cereb Blood Flow Metab 2022; 42:979-996. [PMID: 35209740 PMCID: PMC9125494 DOI: 10.1177/0271678x221084410] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Extremely low frequency electromagnetic stimulation (ELF-EMS) has been considered as a neuroprotective therapy for ischemic stroke based on its capacity to induce nitric oxide (NO) signaling. Here, we examined whether ELF-EMS reduces ischemic stroke volume by stimulating cerebral collateral perfusion. Moreover, the pathway responsible for ELF-EMS-induced NO production was investigated. ELF-EMS diminished infarct growth following experimental stroke in collateral-rich C57BL/6 mice, but not in collateral-scarce BALB/c mice, suggesting that decreased lesion sizes after ELF-EMS results from improved collateral blood flow. In vitro analysis demonstrated that ELF-EMS increased endothelial NO levels by stimulating the Akt-/eNOS pathway. Furthermore, ELF-EMS augmented perfusion in the hind limb of healthy mice, which was mediated by enhanced Akt-/eNOS signaling. In healthy C57BL/6 mouse brains, ELF-EMS treatment increased cerebral blood flow in a NOS-dependent manner, whereas no improvement in cerebrovascular perfusion was observed in collateral-sparse BALB/c mice. In addition, ELF-EMS enhanced cerebral blood flow in both the contra- and ipsilateral hemispheres of C57BL/6 mice subjected to experimental ischemic stroke. In conclusion, we showed that ELF-EMS enhances (cerebro)vascular perfusion by stimulating NO production, indicating that ELF-EMS could be an attractive therapeutic strategy for acute ischemic stroke by improving cerebral collateral blood flow.
Collapse
Affiliation(s)
- Hannelore Kemps
- Biomedical Research Institute (BIOMED), Hasselt University (UHasselt), Diepenbeek, Belgium
| | - Chantal Dessy
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Laurent Dumas
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Pierre Sonveaux
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Lotte Alders
- Biomedical Research Institute (BIOMED), Hasselt University (UHasselt), Diepenbeek, Belgium
| | - Jana Van Broeckhoven
- Biomedical Research Institute (BIOMED), Hasselt University (UHasselt), Diepenbeek, Belgium
| | - Lena Perez Font
- Centro Nacional de Electromagnetismo Aplicado (CNEA), Universidad de Oriente, Santiago de Cuba, Cuba
| | - Sara Lambrichts
- Department of Pharmacology and Toxicology, School for Mental Health and Neuroscience, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Sébastien Foulquier
- Department of Pharmacology and Toxicology, School for Mental Health and Neuroscience, Maastricht University Medical Center+, Maastricht, The Netherlands.,CARIM, School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| | - Sven Hendrix
- Biomedical Research Institute (BIOMED), Hasselt University (UHasselt), Diepenbeek, Belgium.,Medical School Hamburg, Hamburg, Germany
| | - Bert Brône
- Biomedical Research Institute (BIOMED), Hasselt University (UHasselt), Diepenbeek, Belgium
| | - Robin Lemmens
- KU Leuven, - University of Leuven, Department of Neurosciences, Experimental Neurology, Leuven, Belgium.,VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium.,Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Annelies Bronckaers
- Biomedical Research Institute (BIOMED), Hasselt University (UHasselt), Diepenbeek, Belgium
| |
Collapse
|
74
|
Ma W, Zhu K, Yin L, Yang J, Zhang J, Wu H, Liu K, Li C, Liu W, Guo J, Li L. Effects of ischemic postconditioning and long non-coding RNAs in ischemic stroke. Bioengineered 2022; 13:14799-14814. [PMID: 36420646 PMCID: PMC9704383 DOI: 10.1080/21655979.2022.2108266] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Stroke is a main cause of disability and death among adults in China, and acute ischemic stroke accounts for 80% of cases. The key to ischemic stroke treatment is to recanalize the blocked blood vessels. However, more than 90% of patients cannot receive effective treatment within an appropriate time, and delayed recanalization of blood vessels causes reperfusion injury. Recent research has revealed that ischemic postconditioning has a neuroprotective effect on the brain, but the mechanism has not been fully clarified. Long non-coding RNAs (lncRNAs) have previously been associated with ischemic reperfusion injury in ischemic stroke. LncRNAs regulate important cellular and molecular events through a variety of mechanisms, but a comprehensive analysis of potential lncRNAs involved in the brain protection produced by ischemic postconditioning has not been conducted. In this review, we summarize the common mechanisms of cerebral injury in ischemic stroke and the effect of ischemic postconditioning, and we describe the potential mechanisms of some lncRNAs associated with ischemic stroke.
Collapse
Affiliation(s)
- Wei Ma
- Institute of Neuroscience, Faculty of Basic Medical Science, Kunming Medical University, Kunming, China
| | - Kewei Zhu
- Institute of Neuroscience, Faculty of Basic Medical Science, Kunming Medical University, Kunming, China
| | - Luwei Yin
- Institute of Neuroscience, Faculty of Basic Medical Science, Kunming Medical University, Kunming, China
| | - Jinwei Yang
- Second Department of General Surgery, First People’s Hospital of Yunnan Province, Kunming, China
| | - Jinfen Zhang
- Institute of Neuroscience, Faculty of Basic Medical Science, Kunming Medical University, Kunming, China
| | - Hongjie Wu
- Institute of Neuroscience, Faculty of Basic Medical Science, Kunming Medical University, Kunming, China
| | - Kuangpin Liu
- Institute of Neuroscience, Faculty of Basic Medical Science, Kunming Medical University, Kunming, China
| | - Chunyan Li
- Institute of Neuroscience, Faculty of Basic Medical Science, Kunming Medical University, Kunming, China
| | - Wei Liu
- Institute of Neuroscience, Faculty of Basic Medical Science, Kunming Medical University, Kunming, China
| | - Jianhui Guo
- Second Department of General Surgery, First People’s Hospital of Yunnan Province, Kunming, China,Jianhui Guo Second Department of General Surgery, First People’s Hospital of Yunnan Province, Kunming 650034, Yunnan, China
| | - Liyan Li
- Institute of Neuroscience, Faculty of Basic Medical Science, Kunming Medical University, Kunming, China,CONTACT Liyan Li Institute of Neurosicence, Faculty of Basic Medical Science, Kunming Medical University, Kunming 650500, Yunnan, China
| |
Collapse
|
75
|
Circulating miRNA-195-5p and -451a in Patients with Acute Hemorrhagic Stroke in Emergency Department. Life (Basel) 2022; 12:life12050763. [DOI: 10.3390/life12050763] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/12/2022] [Accepted: 05/17/2022] [Indexed: 12/14/2022] Open
Abstract
(1) Background: In our previous study, acute ischemic stroke (AIS) patients showed increased levels of circulating miRNAs (-195-5p and -451a) involved in vascular endothelial growth factor A (VEGF-A) regulation. Here, we evaluated, for the first time, both circulating miRNAs in acute intracerebral hemorrhagic (ICH) patients. (2) Methods: Circulating miRNAs and serum VEGF-A were assessed by real-time PCR and ELISA in 20 acute ICH, 21 AIS patients, and 21 controls. These were evaluated at hospital admission (T0) and after 96 h (T96) from admission. (3) Results: At T0, circulating miRNAs were five-times up-regulated in AIS patients, tending to decrease at T96. By contrast, in the acute ICH group, circulating miRNAs were significantly increased at both T0 and T96. Moreover, a significant decrease was observed in serum VEGF-A levels at T0 in AIS patients, tending to increase at T96. Conversely, in acute ICH patients, the levels of VEGF-A were significantly decreased at both T0 and T96. (4) Conclusions: The absence of a reduction in circulating miRNAs (195-5p and -451a), reported in acute ICH subjects after 96 h from hospital admission, together with the absence of increment of serum VEGF-A, may represent useful biomarkers indicating the severe brain damage status that characterizes acute ICH patients.
Collapse
|
76
|
Hong JM, Choi MH, Park GH, Shin HS, Lee SJ, Lee JS, Lim YC. Transdural Revascularization by Multiple Burrhole After Erythropoietin in Stroke Patients With Cerebral Hypoperfusion: A Randomized Controlled Trial. Stroke 2022; 53:2739-2748. [PMID: 35579016 PMCID: PMC9389942 DOI: 10.1161/strokeaha.122.038650] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In patients with acute symptomatic stroke, reinforcement of transdural angiogenesis using multiple burr hole (MBH) procedures after EPO (erythropoietin) treatment has rarely been addressed. We aimed to investigate the efficacy and safety of cranial MBH procedures under local anesthesia for augmenting transdural revascularization after EPO treatment in patients with stroke with perfusion impairments.
Collapse
Affiliation(s)
- Ji Man Hong
- Department of Neurology, Ajou University School of Medicine, Ajou University Medical Center, Suwon, South Korea. (J.M.H., M.H.C., G.H.P., S.-J.L., J.S.L)
| | - Mun Hee Choi
- Department of Neurology, Ajou University School of Medicine, Ajou University Medical Center, Suwon, South Korea. (J.M.H., M.H.C., G.H.P., S.-J.L., J.S.L)
| | - Geun Hwa Park
- Department of Neurology, Ajou University School of Medicine, Ajou University Medical Center, Suwon, South Korea. (J.M.H., M.H.C., G.H.P., S.-J.L., J.S.L)
| | - Hee Sun Shin
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, South Korea (H.S.S.)
| | - Seong-Joon Lee
- Department of Neurology, Ajou University School of Medicine, Ajou University Medical Center, Suwon, South Korea. (J.M.H., M.H.C., G.H.P., S.-J.L., J.S.L)
| | - Jin Soo Lee
- Department of Neurology, Ajou University School of Medicine, Ajou University Medical Center, Suwon, South Korea. (J.M.H., M.H.C., G.H.P., S.-J.L., J.S.L)
| | - Yong Cheol Lim
- Department of Neurosurgery, Ajou University School of Medicine, Ajou University Medical Center, Suwon, South Korea. (Y.C.L.)
| |
Collapse
|
77
|
Li M, Wang Y, Gao Y, Yao X, Lan W, Tang W. Effects of electroacupuncture on angiogenesis and cortical VEGF and BDNF expression in rats with focal cerebral ischemia. JOURNAL OF ACUPUNCTURE AND TUINA SCIENCE 2022. [DOI: 10.1007/s11726-022-1300-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
78
|
Naoluo Xintong Decoction Ameliorates Cerebral Ischemia-Reperfusion Injury by Promoting Angiogenesis through Activating the HIF-1α/VEGF Signaling Pathway in Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:9341466. [PMID: 35449809 PMCID: PMC9017488 DOI: 10.1155/2022/9341466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 02/12/2022] [Accepted: 02/22/2022] [Indexed: 11/17/2022]
Abstract
Background Naoluo Xintong decoction (NLXTD) is a traditional Chinese medicine (TCM) formula which has been used to improve neuronal functional recovery after cerebral ischemic stroke. However, the molecular mechanism underlying NLXTD's amelioration of ischemic stroke remains unclear. The present study was designed to explore the effect and mechanism of NLXTD on brain angiogenesis in a rat model with cerebral ischemia-reperfusion (I/R) injury targeting the hypoxia-inducible factor-1α (HIF-1α)/vascular endothelial growth factor (VEGF) pathway. Materials and Methods Cerebral I/R model was established by the classical middle cerebral artery occlusion (MCAO) method. Sprague-Dawley (SD) male rats (n = 80) were randomly divided into the sham-operation group, the model group, the HIF-1α inhibitor 2-methoxyestradiol (2ME2) group, the 2ME2 with NLXTD group, and the NLXTD group. Neurological deficit test, TTC staining, H&E staining, TUNEL staining, immunohistochemistry (IH), immunofluorescence (IF), western blot, and quantitative RT-PCR were performed to evaluate the effect of NLXTD after MCAO. Results Administration of NLXTD significantly decreased neuron deficiency scores, reduced brain infarct volume, and lowered damaged and apoptotic cells after brain I/R injury in rats. Meanwhile, NLXTD had a protective effect on angiogenesis by increasing the MVD and the expressions of BrdU and CD34, which enhanced the number of endothelial cells in the ischemic penumbra brain. NLXTD treatment significantly raised the protein and mRNA levels of HIF-1α, VEGF, VEGFR2, and Notch1 compared with the model treatment. In contrast, a specific HIF-1α inhibitor, 2ME2, inhibited the improvement of neurological function and angiogenesis in NLXTD-induced rats with cerebral I/R injury, suggesting that NLXTD played a positive role in ischemic brain injury by activating the HIF-1α/VEGF signaling pathway. Conclusions NLXTD exerts neuroprotection targeting angiogenesis by upregulating the HIF-1α/VEGF signaling pathway on cerebral I/R injury rats.
Collapse
|
79
|
Yang X, Qiang Q, Li N, Feng P, Wei W, Hölscher C. Neuroprotective Mechanisms of Glucagon-Like Peptide-1-Based Therapies in Ischemic Stroke: An Update Based on Preclinical Research. Front Neurol 2022; 13:844697. [PMID: 35370875 PMCID: PMC8964641 DOI: 10.3389/fneur.2022.844697] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 02/16/2022] [Indexed: 12/16/2022] Open
Abstract
The public and social health burdens of ischemic stroke have been increasing worldwide. Hyperglycemia leads to a greater risk of stroke. This increased risk is commonly seen among patients with diabetes and is in connection with worsened clinical conditions and higher mortality in patients with acute ischemic stroke (AIS). Therapy for stroke focuses mainly on restoring cerebral blood flow (CBF) and ameliorating neurological impairment caused by stroke. Although choices of stroke treatment remain limited, much advance have been achieved in assisting patients in recovering from ischemic stroke, along with progress of recanalization therapy through pharmacological and mechanical thrombolysis. However, it is still necessary to develop neuroprotective therapies for AIS to protect the brain against injury before and during reperfusion, prolong the time window for intervention, and consequently improve neurological prognosis. Glucagon-like peptide-1 receptor agonists (GLP-1 RAs) are broadly regarded as effective drugs in the treatment of type 2 diabetes mellitus (T2DM). Preclinical data on GLP-1 and GLP-1 RAs have displayed an impressive neuroprotective efficacy in stroke, Parkinson's disease (PD), Alzheimer's disease (AD), Amyotrophic lateral sclerosis (ALS), and other neurodegenerative diseases. Based on the preclinical studies in the past decade, we review recent progress in the biological roles of GLP-1 and GLP-1 RAs in ischemic stroke. Emphasis will be placed on their neuroprotective effects in experimental models of cerebral ischemia stroke at cellular and molecular levels.
Collapse
Affiliation(s)
- Xiaoyan Yang
- Department of Neurology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Qiang Qiang
- Department of Neurology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Nan Li
- Department of Neurology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Peng Feng
- Department of Neurology, The Second Affiliated Hospital of Shanxi Medical University, Taiyuan, China
| | - Wenshi Wei
- Department of Neurology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Christian Hölscher
- Department of Neurology, The Second Affiliated Hospital of Shanxi Medical University, Taiyuan, China.,Henan University of Chinese Medicine, Academy of Chinese Medical Science, Zhengzhou, China
| |
Collapse
|
80
|
Zhu T, Wang L, Wang LP, Wan Q. Therapeutic targets of neuroprotection and neurorestoration in ischemic stroke: Applications for natural compounds from medicinal herbs. Biomed Pharmacother 2022; 148:112719. [DOI: 10.1016/j.biopha.2022.112719] [Citation(s) in RCA: 136] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 12/13/2022] Open
|
81
|
Jang M, Han S, Cho H. D* from diffusion MRI reveals a correspondence between ventricular cerebrospinal fluid volume and flow in the ischemic rodent model. J Cereb Blood Flow Metab 2022; 42:572-583. [PMID: 34796772 PMCID: PMC9051140 DOI: 10.1177/0271678x211060741] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Quantitative measurement of cerebrospinal fluid (CSF) flow and volume and longitudinal monitoring of CSF dynamics provide insights into the compensatory characteristics of post-stroke CSF. In this study, we compared the MRI pseudo-diffusion index (D*) of live and sacrificed rat brains to confirm the effect of ventricular CSF flow on diffusion signals. We observed the relationship between the CSF peak velocities and D* through Monte Carlo (MC) simulations to further understand the source of D* contrast. We also determined the dominant CSF flow using D* in three directions. Finally, we investigated the dynamic evolutions of ventricular CSF flow and volume in a stroke rat model (n = 8) from preoperative to up to 45 days after surgery and determined the correlation between ventricular CSF volume and flow. MC simulations showed a strong positive correlation between the CSF peak velocity and D* (r = 0.99). The dominant CSF flow variations in the 3D ventricle could be measured using the maximum D* map. A longitudinal positive correlation between ventricular CSF volume and D* was observed in the lateral (r = 0.74) and ventral-third (r = 0.81) ventricles, respectively. The directional D* measurements provide quantitative CSF volume and flow information, which would provide useful insights into ischemic stroke with diffusion MRI.
Collapse
Affiliation(s)
- MinJung Jang
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan, South Korea
| | - SoHyun Han
- Center for Neuroscience Imaging Research, Sungkyunkwan University, Suwon, South Korea
| | - HyungJoon Cho
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan, South Korea
| |
Collapse
|
82
|
Bai S, Lu X, Pan Q, Wang B, Pong U K, Yang Y, Wang H, Lin S, Feng L, Wang Y, Li Y, Lin W, Wang Y, Zhang X, Li Y, Li L, Yang Z, Wang M, Lee WYW, Jiang X, Li G. Cranial Bone Transport Promotes Angiogenesis, Neurogenesis, and Modulates Meningeal Lymphatic Function in Middle Cerebral Artery Occlusion Rats. Stroke 2022; 53:1373-1385. [PMID: 35135326 DOI: 10.1161/strokeaha.121.037912] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 12/23/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND Ischemic stroke is a leading cause of death and disability worldwide. However, the time window for quickly dissolving clots and restoring cerebral blood flow, using tissue-type plasminogen activator treatment is rather limited, resulting in many patients experiencing long-term functional impairments if not death. This study aims to determine the roles of cranial bone transport (CBT), a novel, effective, and simple surgical technique, in the recovery of ischemic stroke using middle cerebral artery occlusion (MCAO) rat model. METHODS CBT was performed by slowly sliding a bone segment in skull with a special frame and a speed of 0.25 mm/12 hours for 10 days following MCAO. Morris water maze, rotarod test, and catwalk gait analysis were used to study the neurological behaviors, and infarct area and cerebral flow were evaluated during CBT process. Immunofluorescence staining of CD31 and Nestin/Sox2 (sex determining region Y box 2) was performed to study the angiogenesis and neurogenesis. OVA-A647 (ovalbumin-Alexa Fluor 647) was intracisterna magna injected to evaluate the meningeal lymphatic drainage function. RESULTS CBT treatment has significantly reduced the ischemic lesions areas and improved the neurological deficits in MCAO rats compared with the rats in the control groups. CBT treatment significantly promoted angiogenesis and neurogenesis in the brain of MCAO rats. The drainage function of meningeal lymphatic vessels in MCAO rats was significantly impaired compared with normal rats. Ablation of meningeal lymphatic drainage led to increased neuroinflammation and aggravated neurological deficits and ischemic injury in MCAO rats. CBT treatment significantly improved the meningeal lymphatic drainage function and alleviated T-cell infiltration in MCAO rats. CONCLUSIONS This study provided evidence for the possible mechanisms on how CBT attenuates ischemic stroke injury and facilitates rapid neuronal function recovery, suggesting that CBT may be an alternative treatment strategy for managing ischemic stroke.
Collapse
Affiliation(s)
- Shanshan Bai
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China (S.B., X.L., B.W., Y.Y., H.W., S.L., L.F., Yan Wang, Yucong Li, W.L., Yujia Wang, X.Z., Yuan Li, L.L., Z.Y., M.W., W.Y.-W.L., G.L.)
| | - Xuan Lu
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China (S.B., X.L., B.W., Y.Y., H.W., S.L., L.F., Yan Wang, Yucong Li, W.L., Yujia Wang, X.Z., Yuan Li, L.L., Z.Y., M.W., W.Y.-W.L., G.L.)
| | - Qi Pan
- Department of Pediatric Orthopaedics, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, PR China (Q.P.)
| | - Bin Wang
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China (S.B., X.L., B.W., Y.Y., H.W., S.L., L.F., Yan Wang, Yucong Li, W.L., Yujia Wang, X.Z., Yuan Li, L.L., Z.Y., M.W., W.Y.-W.L., G.L.)
| | - Kin Pong U
- Key Laboratory for Regenerative Medicine of the Ministry of Education of China, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, SAR, PR China (K.P.U., X.J.)
| | - Yongkang Yang
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China (S.B., X.L., B.W., Y.Y., H.W., S.L., L.F., Yan Wang, Yucong Li, W.L., Yujia Wang, X.Z., Yuan Li, L.L., Z.Y., M.W., W.Y.-W.L., G.L.)
| | - Haixing Wang
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China (S.B., X.L., B.W., Y.Y., H.W., S.L., L.F., Yan Wang, Yucong Li, W.L., Yujia Wang, X.Z., Yuan Li, L.L., Z.Y., M.W., W.Y.-W.L., G.L.)
| | - Sien Lin
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China (S.B., X.L., B.W., Y.Y., H.W., S.L., L.F., Yan Wang, Yucong Li, W.L., Yujia Wang, X.Z., Yuan Li, L.L., Z.Y., M.W., W.Y.-W.L., G.L.)
| | - Lu Feng
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China (S.B., X.L., B.W., Y.Y., H.W., S.L., L.F., Yan Wang, Yucong Li, W.L., Yujia Wang, X.Z., Yuan Li, L.L., Z.Y., M.W., W.Y.-W.L., G.L.)
| | - Yan Wang
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China (S.B., X.L., B.W., Y.Y., H.W., S.L., L.F., Yan Wang, Yucong Li, W.L., Yujia Wang, X.Z., Yuan Li, L.L., Z.Y., M.W., W.Y.-W.L., G.L.)
| | - Yucong Li
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China (S.B., X.L., B.W., Y.Y., H.W., S.L., L.F., Yan Wang, Yucong Li, W.L., Yujia Wang, X.Z., Yuan Li, L.L., Z.Y., M.W., W.Y.-W.L., G.L.)
| | | | | | - Xiaoting Zhang
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China (S.B., X.L., B.W., Y.Y., H.W., S.L., L.F., Yan Wang, Yucong Li, W.L., Yujia Wang, X.Z., Yuan Li, L.L., Z.Y., M.W., W.Y.-W.L., G.L.)
| | - Yuan Li
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China (S.B., X.L., B.W., Y.Y., H.W., S.L., L.F., Yan Wang, Yucong Li, W.L., Yujia Wang, X.Z., Yuan Li, L.L., Z.Y., M.W., W.Y.-W.L., G.L.)
| | - Linlong Li
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China (S.B., X.L., B.W., Y.Y., H.W., S.L., L.F., Yan Wang, Yucong Li, W.L., Yujia Wang, X.Z., Yuan Li, L.L., Z.Y., M.W., W.Y.-W.L., G.L.)
| | - Zhengmeng Yang
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China (S.B., X.L., B.W., Y.Y., H.W., S.L., L.F., Yan Wang, Yucong Li, W.L., Yujia Wang, X.Z., Yuan Li, L.L., Z.Y., M.W., W.Y.-W.L., G.L.)
| | - Ming Wang
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China (S.B., X.L., B.W., Y.Y., H.W., S.L., L.F., Yan Wang, Yucong Li, W.L., Yujia Wang, X.Z., Yuan Li, L.L., Z.Y., M.W., W.Y.-W.L., G.L.)
| | - Wayne Yuk-Wai Lee
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China (S.B., X.L., B.W., Y.Y., H.W., S.L., L.F., Yan Wang, Yucong Li, W.L., Yujia Wang, X.Z., Yuan Li, L.L., Z.Y., M.W., W.Y.-W.L., G.L.)
| | - Xiaohua Jiang
- Key Laboratory for Regenerative Medicine of the Ministry of Education of China, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, SAR, PR China (K.P.U., X.J.)
| | - Gang Li
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China (S.B., X.L., B.W., Y.Y., H.W., S.L., L.F., Yan Wang, Yucong Li, W.L., Yujia Wang, X.Z., Yuan Li, L.L., Z.Y., M.W., W.Y.-W.L., G.L.)
| |
Collapse
|
83
|
Secondary Analysis of Walking Activities During the Acute Stroke Hospital Stay and Cerebrovascular Health. Cardiopulm Phys Ther J 2022; 33:130-137. [DOI: 10.1097/cpt.0000000000000196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
84
|
Yang L, Li M, Zhan Y, Feng X, Lu Y, Li M, Zhuang Y, Lei J, Zhao H. The Impact of Ischemic Stroke on Gray and White Matter Injury Correlated With Motor and Cognitive Impairments in Permanent MCAO Rats: A Multimodal MRI-Based Study. Front Neurol 2022; 13:834329. [PMID: 35309583 PMCID: PMC8924049 DOI: 10.3389/fneur.2022.834329] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 01/19/2022] [Indexed: 01/12/2023] Open
Abstract
Background Identifying the alterations of the cerebral gray and white matter is an important prerequisite for developing potential pharmacological therapy for stroke. This study aimed to assess the changes of gray and white matter after permanent middle cerebral artery occlusion (pMCAO) in rats using magnetic resonance imaging (MRI), and to correlate them with the behavior performance. Methods Rats were subjected to pMCAO or sham surgery and reared for 30 days. Motor and cognitive function of the rats were examined by gait and Morris water maze (MWM) tests, respectively. Multimodal MRI was conducted to examine the functional and structural changes of the gray and white matter followed with luxol fast blue (LFB) staining. Results The gait and MWM tests revealed significant motor and cognitive dysfunction in pMCAO rats, respectively. Magnetic resonance angiography presented abnormal intracranial arteries in pMCAO rats with reduced signal intensity of the anterior cerebral artery, anterior communicating cerebral artery, internal carotid artery, and increased basilar artery vessel signal compared with sham rats. Arterial spin labeling confirmed the decreased cerebral blood flow in the infarcted sensorimotor cortex and striatum. Structural T2-weighted imaging and T2 mapping showed brain atrophy and elevation of T2 value in the gray (sensorimotor cortex, striatum) and white (external capsule, internal capsule) matter of pMCAO rats. The results from diffusion tensor imaging (DTI) corresponded well with LFB staining showing reduced relative FA accompanied with increased relative AD and RD in the gray and white matter of pMCAO rats compared with sham rats. Fiber tracking derived from DTI further observed significantly reduced fiber density and length in the corresponding brain regions of pMCAO rats compared with sham rats. Specially, the DTI parameters (especially FA) in the relevant gray matter and white matter significantly correlated with the behavior performance in the gait and MWM tests. Conclusion Collectively, the gray and white matter damages could be non-invasively monitored in pMCAO rats by multimodal MRI. DTI-derived parameters, particularly the FA, might be a good imaging index to stage gray and white matter damages associated with post-stroke motor and cognitive impairments.
Collapse
Affiliation(s)
- Le Yang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Manzhong Li
- Department of Pharmacy, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Bio-characteristic Profiling for Evaluation of Rational Drug Use, Beijing, China
| | - Yu Zhan
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Xuefeng Feng
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Yun Lu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Mingcong Li
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Yuming Zhuang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Jianfeng Lei
- Medical Imaging Laboratory of Core Facility Center, Capital Medical University, Beijing, China
| | - Hui Zhao
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| |
Collapse
|
85
|
Liu J, Zhang H, Di K, Hou L, Yu S. Circular noncoding RNA circ_0007865, serves as a competing endogenous RNA, targeting the miR-214-3p/FKBP5 axis to regulate oxygen-glucose deprivation-induced injury in brain microvascular endothelial cells. Neuroreport 2022; 33:163-172. [PMID: 35143446 DOI: 10.1097/wnr.0000000000001751] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Ischemic stroke (IS) is a major cause of permanent morbidity and lifelong disability worldwide. Circular RNA (circRNA) circ_0007865 has been reported to be upregulated in acute ischemic stroke (AIS) patients. Also, AIS patients exhibited increased death of human brain microvascular endothelial cells (HBMECs). This study is designed to explore the role and mechanism of circ_0007865 in the oxygen-glucose deprivation (OGD)-induced cell damage in AIS. METHODS Circ_0007865, microRNA-214-3p (miR-214-3p), and FK506-binding protein 5 (FKBP5) levels were detected by real-time quantitative PCR. Cell proliferative angiogenesis, migration, and apoptosis were assessed by Cell Counting Kit-8, 5-ethynyl-2'-deoxyuridine, colony formation, tube formation, wound healing, transwell, and flow cytometry assays. B-cell lymphoma-2 (Bcl-2), Bcl-2-related X protein (Bax), cleaved caspase-3, and FKBP5 protein levels were determined by western blot assay. The binding relationship between miR-214-3p and circ_0007865 or FKBP5 was predicted by StarBase, and verified by a dual-luciferase reporter, RNA pull-down assay. RESULTS Circ_0007865 and FKBP5 were increased, and miR-214-3p was decreased in OGD-treated HBMECs. Furthermore, the silencing of circ_0007865 could promote cell proliferative angiogenesis, migration, and inhibit apoptosis in OGD-triggered HBMECs in vitro. Mechanically, circ_0007865 acted as a sponge of miR-214-3p to regulate FKBP5. CONCLUSION According to these results, circ_0007865 deficiency could attenuate OGD-induced HBMEC damage by modulating the miR-214-3p/FKBP5 axis, hinting at a promising therapeutic target for future acute IS therapy.
Collapse
Affiliation(s)
- Jinghua Liu
- Department of Neurology, The Second People's Hospital of Dongying, Dongying
| | - Hong Zhang
- School of Medicine, Tianjin Tianshi College, Tianjin
| | - Kuiyi Di
- Department of Neurology, The Second People's Hospital of Dongying, Dongying
| | | | - Shanshan Yu
- Department of Pharmacy, The Second People's Hospital of Dongying, Dongying, China
| |
Collapse
|
86
|
Li N, Zhang X, Zhai J, Yin J, Ma K, Wang R, Qin X, Li Y, Dong X, Wang S. Isoflurane and Netrin-1 combination therapy enhances angiogenesis and neurological recovery by improving the expression of HIF-1α-Netrin-1-UNC5B/VEGF cascade to attenuate cerebral ischemia injury. Exp Neurol 2022; 352:114028. [PMID: 35247371 DOI: 10.1016/j.expneurol.2022.114028] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 02/19/2022] [Accepted: 02/26/2022] [Indexed: 12/01/2022]
Abstract
Ischemic stroke (IS) causes many morbidities and deaths worldwide. However, the current monotherapy strategy is not satisfactory. Therefore, it is urgent to explore possible combined treatment methods. Although both isoflurane (ISO) and Netrin-1 (NT-1) have angiogenesis and neuroprotective effects, it is still unclear whether combining ISO with NT-1 will provide a positive effect and the possible mechanism of action. In this study, we used a photochemical (PTI) method to establish a mouse ischemic stroke model. ISO and NT-1 were used to treat the mice for 1 week. The adhesive removal test, Morris water maze test, modified neurological severity scores and triphenyl tetrazolium chloride staining were performed to test the treatment effect. Western blotting was performed to assess protein expression, immunofluorescence staining (IF) and immunohistochemical staining (IHC) was used to evaluate angiogenesis. The results suggested that combining ISO with NT-1 resulted in a better therapeutic effect than ISO or NT-1 treatment after PTI injury (all P < 0.01). The protein expression of VEGFA and CD34 in the ISO + NT-1 group was significantly increased compared with that in the other groups (all P < 0.05). IF and IHC also showed that the ISO + NT-1 group significantly improved angiogenesis (all P < 0.01). YC-1 (an HIF-1α inhibitor) and Unc5B siRNA were used to inhibit the expression of HIF-1α and UNC5B before and after combination ISO and NT-1 treatment. The combined inhibition group not only expressed the least VEGFA and CD34 but also expressed the least HIF-1α, UNC5B, FAK, and β-catenin in all groups (all P < 0.05). Most importantly, angiogenesis and neurological recovery were also significantly decreased by inhibiting HIF-1α and UNC5B (all P < 0.05). In conclusion, our results suggested that ISO combined with NT-1 could promote angiogenesis, recover long-term neurobehavioral function, and attenuate cerebral ischemia injury by activating the HIF-1α-Netrin-1-UNC5B/VEGF cascade.
Collapse
Affiliation(s)
- Nian Li
- Department of Anesthesiology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Xu Zhang
- Department of Anesthesiology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Jingwen Zhai
- Department of Anesthesiology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Jiangwen Yin
- Department of Anesthesiology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Ketao Ma
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, China; Department of Physiology, School of Medicine, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University, Shihezi, China
| | - Ruixue Wang
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, China; Department of Physiology, School of Medicine, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University, Shihezi, China
| | - Xinlei Qin
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, China; Department of Physiology, School of Medicine, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University, Shihezi, China
| | - Yan Li
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, China; Department of Physiology, School of Medicine, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University, Shihezi, China
| | - Xiwei Dong
- Department of Anesthesiology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China.
| | - Sheng Wang
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
87
|
Methylglyoxal Scavengers Attenuate Angiogenesis Dysfunction Induced by Methylglyoxal and Oxygen-Glucose Deprivation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8854457. [PMID: 35035668 PMCID: PMC8754597 DOI: 10.1155/2022/8854457] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/02/2021] [Accepted: 11/06/2021] [Indexed: 12/11/2022]
Abstract
Cerebral endothelial cells play an essential role in brain angiogenesis, and their function has been found to be impaired in diabetes. Methylglyoxal (MG) is a highly reactive dicarbonyl metabolite of glucose formed mainly during glycolysis, and its levels can be elevated in hyperglycemic conditions. MG is a potent precursor of AGEs (advanced glycation end-products). In this study, we investigated if MG can induce angiogenesis dysfunction and whether MG scavengers can ameliorate angiogenesis dysfunction induced by MG. Here, we used cultured human brain microvascular endothelial cells (HBMECs) treated with MG and oxygen-glucose deprivation (OGD) to mimic diabetic stroke in vitro. We also used the MG challenged chicken embryo chorioallantoic membrane (CAM) to study angiogenesis in vivo. Interestingly, administration of MG significantly impaired cell proliferation, cell migration, and tube formation and decreased protein expression of angiogenesis-related factors, which was rescued by three different MG scavengers, glyoxalase 1 (GLO1), aminoguanidine (AG), and N-acetyl cysteine (NAC). In cultured CAM, MG exposure significantly reduced angiogenesis and the angiogenesis-related dysfunction could be attenuated by pretreatment with AG or NAC. Treatment of cultured HBMECs with MG plus OGD increased cellular apoptosis significantly, which could be prevented by exposure to GLO1, AG, or NAC. We also noted that administration of MG increased cellular oxidative stress as measured by reactive oxygen species (ROS) generation, enhanced AGE accumulation, and receptor for advanced glycation end-product (RAGE) expression in the cultured HBMECs, which were partially reversed by GLO1, AG, or NAC. Taken together, our findings demonstrated that GLO1, AG, or NAC administration can ameliorate MG-induced angiogenesis dysfunction, and this can be mainly attributed to attenuated ROS production, reduced cellular apoptosis, and increased levels of angiogenic factors. Overall, this study suggested that GLO1, AG, or NAC may be promising candidate compounds for the treatment of angiogenesis dysfunction caused by hyperglycemia in diabetic ischemic stroke.
Collapse
|
88
|
Tian DS, Qin C, Zhou LQ, Yang S, Chen M, Xiao J, Shang K, Bosco DB, Wu LJ, Wang W. FSAP aggravated endothelial dysfunction and neurological deficits in acute ischemic stroke due to large vessel occlusion. Signal Transduct Target Ther 2022; 7:6. [PMID: 34992208 PMCID: PMC8738761 DOI: 10.1038/s41392-021-00802-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 10/09/2021] [Accepted: 10/19/2021] [Indexed: 11/09/2022] Open
Abstract
Revascularization and angiogenesis, as substrates of sustained collateral circulation, play a crucial role in determining the severity and clinical outcome of acute ischemic stroke (AIS) due to large vessel occlusion (LVO). Developing an adjunct biomarker to help identify and monitor collateral status would aid stroke diagnosis and prognosis. To screen the potential biomarkers, proteomic analysis was performed in this study to identify those distinct plasma protein profiles in AIS due to LVO with different collateral status. Interestingly, we found that levels of Plasma Factor VII Activating Protease (FSAP) significantly increased in those AIS patients with poor collaterals, and were correlated with worse neurological outcome. Furtherly, both in vitro and in vivo models of ischemic stroke were used to explore pathological mechanisms of FSAP in endothelial dysfunction. We demonstrated that the FSAP inhibitor, high-molecular-weight hyaluronan (HMW-HA), enhanced the pro-angiogenic vascular factors, improved the integrity of brain blood barrier, and promoted newly formed cerebral microvessels in the ischemic penumbra, consequently improving neurological function. To elucidate the pathways that might contribute to revascularization during LVO, we applied transcriptomic analysis via unbiased RNA sequencing and showed that Wnt signaling was highly involved in FSAP mediated endothelial dysfunction. Notably, inhibition of Wnt5a largely reversed the protective effects from HMW-HA treatment, implying that FSAP might aggravate endothelial dysfunction and neurological deficits by regulating Wnt5a signaling. Therefore, FSAP may represent a potential biomarker for collateral status after LVO and a promising therapeutic target to be explored in the treatment of stroke.
Collapse
Affiliation(s)
- Dai-Shi Tian
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Chuan Qin
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Luo-Qi Zhou
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Sheng Yang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Man Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Jun Xiao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Ke Shang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Dale B Bosco
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China.
| |
Collapse
|
89
|
Karampatsi D, Zabala A, Wilhelmsson U, Dekens D, Vercalsteren E, Larsson M, Nyström T, Pekny M, Patrone C, Darsalia V. Diet-induced weight loss in obese/diabetic mice normalizes glucose metabolism and promotes functional recovery after stroke. Cardiovasc Diabetol 2021; 20:240. [PMID: 34937562 PMCID: PMC8697500 DOI: 10.1186/s12933-021-01426-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 12/02/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Post-stroke functional recovery is severely impaired by type 2 diabetes (T2D). This is an important clinical problem since T2D is one of the most common diseases. Because weight loss-based strategies have been shown to decrease stroke risk in people with T2D, we aimed to investigate whether diet-induced weight loss can also improve post-stroke functional recovery and identify some of the underlying mechanisms. METHODS T2D/obesity was induced by 6 months of high-fat diet (HFD). Weight loss was achieved by a short- or long-term dietary change, replacing HFD with standard diet for 2 or 4 months, respectively. Stroke was induced by middle cerebral artery occlusion and post-stroke recovery was assessed by sensorimotor tests. Mechanisms involved in neurovascular damage in the post-stroke recovery phase, i.e. neuroinflammation, impaired angiogenesis and cellular atrophy of GABAergic parvalbumin (PV)+ interneurons were assessed by immunohistochemistry/quantitative microscopy. RESULTS Both short- and long-term dietary change led to similar weight loss. However, only the latter enhanced functional recovery after stroke. This effect was associated with pre-stroke normalization of fasting glucose and insulin resistance, and with the reduction of T2D-induced cellular atrophy of PV+ interneurons. Moreover, stroke recovery was associated with decreased T2D-induced neuroinflammation and reduced astrocyte reactivity in the contralateral striatum. CONCLUSION The global diabetes epidemic will dramatically increase the number of people in need of post-stroke treatment and care. Our results suggest that diet-induced weight loss leading to pre-stroke normalization of glucose metabolism has great potential to reduce the sequelae of stroke in the diabetic population.
Collapse
MESH Headings
- Animals
- Behavior, Animal
- Biomarkers/blood
- Blood Glucose/metabolism
- Brain/metabolism
- Brain/pathology
- Brain/physiopathology
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/diet therapy
- Diabetes Mellitus, Type 2/physiopathology
- Diet, High-Fat
- Disease Models, Animal
- Glycemic Control
- Infarction, Middle Cerebral Artery/blood
- Infarction, Middle Cerebral Artery/diet therapy
- Infarction, Middle Cerebral Artery/pathology
- Infarction, Middle Cerebral Artery/physiopathology
- Male
- Mice, Inbred C57BL
- Obesity/blood
- Obesity/diet therapy
- Obesity/physiopathology
- Recovery of Function
- Stroke/blood
- Stroke/diet therapy
- Stroke/pathology
- Stroke/physiopathology
- Time Factors
- Weight Loss
- Mice
Collapse
Affiliation(s)
- Dimitra Karampatsi
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, 118 83, Stockholm, Sweden
| | - Alexander Zabala
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, 118 83, Stockholm, Sweden
| | - Ulrika Wilhelmsson
- Laboratory of Astrocyte Biology and CNS Regeneration, Department of Clinical Neuroscience and Rehabilitation, Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Doortje Dekens
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, 118 83, Stockholm, Sweden
| | - Ellen Vercalsteren
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, 118 83, Stockholm, Sweden
| | - Martin Larsson
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, 118 83, Stockholm, Sweden
| | - Thomas Nyström
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, 118 83, Stockholm, Sweden
| | - Milos Pekny
- Laboratory of Astrocyte Biology and CNS Regeneration, Department of Clinical Neuroscience and Rehabilitation, Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Cesare Patrone
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, 118 83, Stockholm, Sweden.
| | - Vladimer Darsalia
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, 118 83, Stockholm, Sweden.
| |
Collapse
|
90
|
Tang X, Lu J, Chen H, Zhai L, Zhang Y, Lou H, Wang Y, Sun L, Song B. Underlying Mechanism and Active Ingredients of Tianma Gouteng Acting on Cerebral Infarction as Determined via Network Pharmacology Analysis Combined With Experimental Validation. Front Pharmacol 2021; 12:760503. [PMID: 34867377 PMCID: PMC8635202 DOI: 10.3389/fphar.2021.760503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/20/2021] [Indexed: 12/23/2022] Open
Abstract
Cerebral infarction (CI), a common cerebrovascular disease worldwide, is caused by unknown factors common to many diseases, including hypokalemia, respiratory diseases, and lower extremity venous thrombosis. Tianma Gouteng (TMGT), a traditional Chinese Medicine (TCM) prescription, has been used for the clinical treatment of CI. In this study, high-performance liquid chromatography (HPLC) fingerprint analysis was used to detect and identify major chemical constituents of TMGT. TCMSP and BATMAN-TCM databases were used to screen for active TMGT constituent compounds, while the GeneCards database was used to screen for protein targets associated with CI. Next, GO and KEGG enrichment analysis of these core nodes were performed to determine the identities of key associated biological processes and signal pathways. Meanwhile, a total of six possible gene targets of TMGT, including NFKBIA, PPARG, IL6, IL1B, CXCL8, and HIF1A, were selected for further study using two cellular models of CI. For one model, PC12 cells were treated under oxygen and glucose deprivation (OGD) conditions to generate an OGD cellular model of CI, while for the other model, BV2 cells were stimulated with lipopolysaccharide (LPS) to generate a cellular model of CI-associated inflammation. Ultimately TMGT treatment increased PPARγ expression and downregulated the expression of p-P65, p-IκBα, and HIF-1α in both OGD-induced and LPS-induced cell models of CI. In addition, molecular docking analysis showed that one TMGT chemical constituent, quercetin, may be a bioactive TMGT compound with activity that may be associated with the alleviation of neuronal damage and neuroinflammation triggered by CI. Moreover, additional data obtained in this work revealed that TMGT could inhibit neuroinflammation and protect brain cells from OGD-induced and LPS-induced damage by altering HIF-1α/PPARγ/NF-κB pathway functions. Thus, targeting this pathway through TMGT administration to CI patients may be a strategy for alleviating nerve injury and neuroinflammation triggered by CI.
Collapse
Affiliation(s)
- Xiaolei Tang
- Research Center of Traditional Chinese Medicine, the Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Jing Lu
- Research Center of Traditional Chinese Medicine, the Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China.,College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Haoyuan Chen
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Lu Zhai
- Research Center of Traditional Chinese Medicine, the Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Yuxin Zhang
- Research Center of Traditional Chinese Medicine, the Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Huijuan Lou
- College of Acupuncture and Tuina, Changchun University of Chinese Medicine, Changchun, China
| | - Yufeng Wang
- Department of Tuina, the Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Liwei Sun
- Research Center of Traditional Chinese Medicine, the Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Bailin Song
- Northeast Asian Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
91
|
Sciortino VM, Tran A, Sun N, Cao R, Sun T, Sun YY, Yan P, Zhong F, Zhou Y, Kuan CY, Lee JM, Hu S. Longitudinal cortex-wide monitoring of cerebral hemodynamics and oxygen metabolism in awake mice using multi-parametric photoacoustic microscopy. J Cereb Blood Flow Metab 2021; 41:3187-3199. [PMID: 34304622 PMCID: PMC8669277 DOI: 10.1177/0271678x211034096] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Multi-parametric photoacoustic microscopy (PAM) has emerged as a promising new technique for high-resolution quantification of hemodynamics and oxygen metabolism in the mouse brain. In this work, we have extended the scope of multi-parametric PAM to longitudinal, cortex-wide, awake-brain imaging with the use of a long-lifetime (24 weeks), wide-field (5 × 7 mm2), light-weight (2 g), dual-transparency (i.e., light and ultrasound) cranial window. Cerebrovascular responses to the window installation were examined in vivo, showing a complete recovery in 18 days. In the 22-week monitoring after the recovery, no dura thickening, skull regrowth, or changes in cerebrovascular structure and function were observed. The promise of this technique was demonstrated by monitoring vascular and metabolic responses of the awake mouse brain to ischemic stroke throughout the acute, subacute, and chronic stages. Side-by-side comparison of the responses in the ipsilateral (injury) and contralateral (control) cortices shows that despite an early recovery of cerebral blood flow and an increase in microvessel density, a long-lasting deficit in cerebral oxygen metabolism was observed throughout the chronic stage in the injured cortex, part of which proceeded to infarction. This longitudinal, functional-metabolic imaging technique opens new opportunities to study the chronic progression and therapeutic responses of neurovascular diseases.
Collapse
Affiliation(s)
- Vincent M Sciortino
- Department of Biomedical Engineering, 2358University of Virginia, University of Virginia, Charlottesville, VA, USA
| | - Angela Tran
- Department of Biology, 2358University of Virginia, University of Virginia, Charlottesville, VA, USA
| | - Naidi Sun
- Department of Biomedical Engineering, 2358University of Virginia, University of Virginia, Charlottesville, VA, USA.,Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Rui Cao
- Department of Biomedical Engineering, 2358University of Virginia, University of Virginia, Charlottesville, VA, USA
| | - Tao Sun
- Department of Biomedical Engineering, 2358University of Virginia, University of Virginia, Charlottesville, VA, USA.,Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Yu-Yo Sun
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Ping Yan
- Department of Neuroscience, 2358University of Virginia, University of Virginia, Charlottesville, VA, USA
| | - Fenghe Zhong
- Department of Biomedical Engineering, 2358University of Virginia, University of Virginia, Charlottesville, VA, USA.,Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Yifeng Zhou
- Department of Biomedical Engineering, 2358University of Virginia, University of Virginia, Charlottesville, VA, USA.,Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Chia-Yi Kuan
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Jin-Moo Lee
- Department of Neuroscience, 2358University of Virginia, University of Virginia, Charlottesville, VA, USA.,Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Song Hu
- Department of Biomedical Engineering, 2358University of Virginia, University of Virginia, Charlottesville, VA, USA.,Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
92
|
Huang S, Tan Z, Cai J, Wang Z, Tian Y. Myrtenol improves brain damage and promotes angiogenesis in rats with cerebral infarction by activating the ERK1/2 signalling pathway. PHARMACEUTICAL BIOLOGY 2021; 59:584-593. [PMID: 34010584 PMCID: PMC8143630 DOI: 10.1080/13880209.2021.1917626] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
CONTEXT Cerebral ischaemia/reperfusion (I/R) injury has a high disability and fatality worldwide. Myrtenol has protective effects on myocardial I/R injury through antioxidant and anti-apoptotic effects. OBJECTIVE This study investigated the effect of myrtenol on cerebral ischaemia/reperfusion (I/R) injury and the underlying mechanism. MATERIALS AND METHODS Cerebral I/R injury was induced in adult Sprague-Dawley rats by middle cerebral artery occlusion (MCAO) for 90 min. MCAO rats were treated with or without myrtenol (10, 30, or 50 mg/kg/day) or/and U0126 (10 μL) intraperitoneally for 7 days. RESULTS In the present study, myrtenol had no toxicity at concentrations up to 1.3 g/kg. Myrtenol treatment improved neurological function of MCAO rats, with significantly (p < 0.05) improved neurological deficits (4.31 ± 1.29 vs. 0.00) and reduced brain edoema (78.95 ± 2.27% vs. 85.48 ± 1.24%). Myrtenol extenuated brain tissue injury and neuronal apoptosis, with increased Bcl-2 expression (0.48-fold) and decreased Bax expression (2.02-fold) and caspase-3 activity (1.36-fold). Myrtenol promoted angiogenesis in the brain tissues of MCAO rats, which was reflected by increased VEGF (0.86-fold) and FGF2 (0.51-fold). Myrtenol promoted the phosphorylation of MEK1/2 (0.80-fold) and ERK1/2 (0.97-fold) in MCAO rats. U0126, the inhibitor of ERK1/2 pathway, reversed the protective effects of myrtenol on brain tissue damage and angiogenesis in MCAO rats. DISCUSSION AND CONCLUSIONS Myrtenol reduced brain damage and angiogenesis through activating the ERK1/2 signalling pathway, which may provide a novel alternative strategy for preventing cerebral I/R injury. Further in vitro work detailing its mechanism-of-action for improving ischaemic cerebral infarction is needed.
Collapse
Affiliation(s)
- Shengming Huang
- Department of Neurology, Luohe Central Hospital, Luohe City, China
| | - Zhanguo Tan
- Department of Neurosurgery, Luohe Central Hospital, Luohe City, China
| | - Jirui Cai
- Department of Cardiology, Luohe Central Hospital, Luohe City, China
| | - Zhiping Wang
- Institute of Urology, Second Hospital of Lanzhou University, Lanzhou, China
| | - Yuejun Tian
- Department of Neurology, Luohe Central Hospital, Luohe City, China
- Institute of Urology, Second Hospital of Lanzhou University, Lanzhou, China
- CONTACT Yuejun Tian Department of Neurology, Luohe Central Hospital, Luohe City462000, China; Institute of Urology, Second Hospital of Lanzhou University, Lanzhou730030, China
| |
Collapse
|
93
|
Ma W, Li CY, Zhang SJ, Zang CH, Yang JW, Wu Z, Wang GD, Liu J, Liu W, Liu KP, Liang Y, Zhang XK, Li JJ, Guo JH, Li LY. Neuroprotective effects of long noncoding RNAs involved in ischemic postconditioning after ischemic stroke. Neural Regen Res 2021; 17:1299-1309. [PMID: 34782575 PMCID: PMC8643058 DOI: 10.4103/1673-5374.327346] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
During acute reperfusion, the expression profiles of long noncoding RNAs in adult rats with focal cerebral ischemia undergo broad changes. However, whether long noncoding RNAs are involved in neuroprotective effects following focal ischemic stroke in rats remains unclear. In this study, RNA isolation and library preparation was performed for long noncoding RNA sequencing, followed by determining the coding potential of identified long noncoding RNAs and target gene prediction. Differential expression analysis, long noncoding RNA functional enrichment analysis, and co-expression network analysis were performed comparing ischemic rats with and without ischemic postconditioning rats. Rats were subjected to ischemic postconditioning via the brief and repeated occlusion of the middle cerebral artery or femoral artery. Quantitative real-time reverse transcription-polymerase chain reaction was used to detect the expression levels of differentially expressed long noncoding RNAs after ischemic postconditioning in a rat model of ischemic stroke. The results showed that ischemic postconditioning greatly affected the expression profile of long noncoding RNAs and mRNAs in the brains of rats that underwent ischemic stroke. The predicted target genes of some of the identified long noncoding RNAs (cis targets) were related to the cellular response to ischemia and stress, cytokine signal transduction, inflammation, and apoptosis signal transduction pathways. In addition, 15 significantly differentially expressed long noncoding RNAs were identified in the brains of rats subjected to ischemic postconditioning. Nine candidate long noncoding RNAs that may be related to ischemic postconditioning were identified by a long noncoding RNA expression profile and long noncoding RNA-mRNA co-expression network analysis. Expression levels were verified by quantitative real-time reverse transcription-polymerase chain reaction. These results suggested that the identified long noncoding RNAs may be involved in the neuroprotective effects associated with ischemic postconditioning following ischemic stroke. The experimental animal procedures were approved by the Animal Experiment Ethics Committee of Kunming Medical University (approval No. KMMU2018018) in January 2018.
Collapse
Affiliation(s)
- Wei Ma
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan Province, China
| | - Chun-Yan Li
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan Province, China
| | - Si-Jia Zhang
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan Province, China
| | - Cheng-Hao Zang
- Second Department of General Surgery, First People's Hospital of Yunnan Province, Kunming, Yunnan Province, China
| | - Jin-Wei Yang
- Second Department of General Surgery, First People's Hospital of Yunnan Province, Kunming, Yunnan Province, China
| | - Zhen Wu
- Second Department of General Surgery, First People's Hospital of Yunnan Province, Kunming, Yunnan Province, China
| | - Guo-Dong Wang
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan Province, China
| | - Jie Liu
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan Province, China
| | - Wei Liu
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan Province, China
| | - Kuang-Pin Liu
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan Province, China
| | - Yu Liang
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan Province, China
| | - Xing-Kui Zhang
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan Province, China
| | - Jun-Jun Li
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan Province, China
| | - Jian-Hui Guo
- Second Department of General Surgery, First People's Hospital of Yunnan Province, Kunming, Yunnan Province, China
| | - Li-Yan Li
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan Province, China
| |
Collapse
|
94
|
Yan Y, Gu T, Christensen SDK, Su J, Lassen TR, Hjortbak MV, Lo IJ, Venø ST, Tóth AE, Song P, Nielsen MS, Bøtker HE, Blagoev B, Drasbek KR, Kjems J. Cyclic Hypoxia Conditioning Alters the Content of Myoblast-Derived Extracellular Vesicles and Enhances Their Cell-Protective Functions. Biomedicines 2021; 9:biomedicines9091211. [PMID: 34572398 PMCID: PMC8471008 DOI: 10.3390/biomedicines9091211] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/06/2021] [Accepted: 09/08/2021] [Indexed: 11/16/2022] Open
Abstract
Remote ischemic conditioning (RIC) is a procedure that can attenuate ischemic-reperfusion injury by conducting brief cycles of ischemia and reperfusion in the arm or leg. Extracellular vesicles (EVs) circulating in the bloodstream can release their content into recipient cells to confer protective function on ischemia-reperfusion injured (IRI) organs. Skeletal muscle cells are potential candidates to release EVs as a protective signal during RIC. In this study, we used C2C12 cells as a model system and performed cyclic hypoxia-reoxygenation (HR) to mimic RIC. EVs were collected and subjected to small RNA profiling and proteomics. HR induced a distinct shift in the miRNA profile and protein content in EVs. HR EV treatment restored cell viability, dampened inflammation, and enhanced tube formation in in vitro assays. In vivo, HR EVs showed increased accumulation in the ischemic brain compared to EVs secreted from normoxic culture (N EVs) in a mouse undergoing transient middle cerebral artery occlusion (tMCAO). We conclude that HR conditioning changes the miRNA and protein profile in EVs released by C2C12 cells and enhances the protective signal in the EVs to recipient cells in vitro.
Collapse
Affiliation(s)
- Yan Yan
- Interdisciplinary Nanoscience Center, Aarhus University, 8000 Aarhus, Denmark; (Y.Y.); (J.S.); (I.L.); (P.S.)
- Omiics ApS, 8200 Aarhus, Denmark;
| | - Tingting Gu
- Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark; (T.G.); (K.R.D.)
| | - Stine Duelund Kaas Christensen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark; (S.D.K.C.); (B.B.)
| | - Junyi Su
- Interdisciplinary Nanoscience Center, Aarhus University, 8000 Aarhus, Denmark; (Y.Y.); (J.S.); (I.L.); (P.S.)
| | - Thomas Ravn Lassen
- Department of Cardiology, Aarhus University Hospital, Skejby, 8200 Aarhus, Denmark; (T.R.L.); (M.V.H.); (H.E.B.)
| | - Marie Vognstoft Hjortbak
- Department of Cardiology, Aarhus University Hospital, Skejby, 8200 Aarhus, Denmark; (T.R.L.); (M.V.H.); (H.E.B.)
| | - IJu Lo
- Interdisciplinary Nanoscience Center, Aarhus University, 8000 Aarhus, Denmark; (Y.Y.); (J.S.); (I.L.); (P.S.)
| | | | - Andrea Erzsebet Tóth
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark; (A.E.T.); (M.S.N.)
| | - Ping Song
- Interdisciplinary Nanoscience Center, Aarhus University, 8000 Aarhus, Denmark; (Y.Y.); (J.S.); (I.L.); (P.S.)
| | | | - Hans Erik Bøtker
- Department of Cardiology, Aarhus University Hospital, Skejby, 8200 Aarhus, Denmark; (T.R.L.); (M.V.H.); (H.E.B.)
| | - Blagoy Blagoev
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark; (S.D.K.C.); (B.B.)
| | - Kim Ryun Drasbek
- Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark; (T.G.); (K.R.D.)
| | - Jørgen Kjems
- Interdisciplinary Nanoscience Center, Aarhus University, 8000 Aarhus, Denmark; (Y.Y.); (J.S.); (I.L.); (P.S.)
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
- Correspondence: ; Tel.: +45-289-920-86
| |
Collapse
|
95
|
Xu J, Zhang P, Chen Y, Xu Y, Luan P, Zhu Y, Zhang J. Sodium tanshinone IIA sulfonate ameliorates cerebral ischemic injury through regulation of angiogenesis. Exp Ther Med 2021; 22:1122. [PMID: 34504576 PMCID: PMC8383733 DOI: 10.3892/etm.2021.10556] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 05/26/2021] [Indexed: 12/22/2022] Open
Abstract
Vascular remodeling and neuroprotection are two major adaptable methods for treating ischemic stroke. Edaravone is a protective agent for the treatment of stroke and was used as a positive control in the present study. Sodium tanshinone IIA sulfonate (STS) has demonstrated therapeutic clinical effects in cerebral infarction in China, while its mechanisms of action in ischemic stroke have remained elusive. The angiogenesis and neuroprotective effects of STS were evaluated in a rat model induced by middle cerebral artery occlusion and 3 days of reperfusion. When used at the same dose, the magnitude of the therapeutic effect of STS was similar to that of edaravone in terms of decreased blood-brain barrier damage as indicated by reduced Evans blue leakage, improved neurological deficits, alleviated cerebral edema and inhibition of histopathological changes caused by ischemia/reperfusion. The TUNEL assay demonstrated that the ability of STS to inhibit neuronal apoptosis was equivalent to that of edaravone. Immunofluorescence detection of CD31 and α-smooth muscle actin indicated that the vascular density was significantly reduced in the vehicle group compared with that in the sham operation group, STS increased the microvessel density in the ischemic area. Furthermore, in the vehicle group the protein expression of vascular endothelial growth factor (VEGF) and VEGF receptor 2 (VEGFR) as determined by fluorescence microscopy and immunohistochemistry was significantly reduced compared with that in the sham group. However, STS promoted their expression compared to the vehicle group respectively, and increaed the mRNA expression of VEGF, VEGFR, CD31 and angiopoietin-1 as determined by reverse transcription-quantitative PCR, but these changes were not significant or not present for edaravone apart from Ang-1. In conclusion, STS protected against ischemic brain injury by promoting angiogenesis in ischemic areas and inhibiting neuronal apoptosis. These results provide a potential treatment for stroke recovery.
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
| | - Pei Zhang
- Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Yao Chen
- Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Yulan Xu
- Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Pengwei Luan
- Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, 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
| |
Collapse
|
96
|
Wu L, Islam MR, Lee J, Takase H, Guo S, Andrews AM, Buzhdygan TP, Mathew J, Li W, Arai K, Lo EH, Ramirez SH, Lok J. ErbB3 is a critical regulator of cytoskeletal dynamics in brain microvascular endothelial cells: Implications for vascular remodeling and blood brain barrier modulation. J Cereb Blood Flow Metab 2021; 41:2242-2255. [PMID: 33583260 PMCID: PMC8393293 DOI: 10.1177/0271678x20984976] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Neuregulin (NRG)1 - ErbB receptor signaling has been shown to play an important role in the biological function of peripheral microvascular endothelial cells. However, little is known about how NRG1/ErbB signaling impacts brain endothelial function and blood-brain barrier (BBB) properties. NRG1/ErbB pathways are affected by brain injury; when brain trauma was induced in mice in a controlled cortical impact model, endothelial ErbB3 gene expression was reduced to a greater extent than that of other NRG1 receptors. This finding suggests that ErbB3-mediated processes may be significantly compromised after injury, and that an understanding of ErbB3 function would be important in the of study of endothelial biology in the healthy and injured brain. Towards this goal, cultured brain microvascular endothelial cells were transfected with siRNA to ErbB3, resulting in alterations in F-actin organization and microtubule assembly, cell morphology, migration and angiogenic processes. Importantly, a significant increase in barrier permeability was observed when ErbB3 was downregulated, suggesting ErbB3 involvement in BBB regulation. Overall, these results indicate that neuregulin-1/ErbB3 signaling is intricately connected with the cytoskeletal processes of the brain endothelium and contributes to morphological and angiogenic changes as well as to BBB integrity.
Collapse
Affiliation(s)
- Limin Wu
- Neuroprotection Research Laboratory, Massachusetts General Hospital, Harvard Medical School, Charlestown, USA
| | - Mohammad R Islam
- Neuroprotection Research Laboratory, Massachusetts General Hospital, Harvard Medical School, Charlestown, USA
| | - Janice Lee
- Neuroprotection Research Laboratory, Massachusetts General Hospital, Harvard Medical School, Charlestown, USA
| | - Hajime Takase
- Neuroprotection Research Laboratory, Massachusetts General Hospital, Harvard Medical School, Charlestown, USA
| | - Shuzhen Guo
- Neuroprotection Research Laboratory, Massachusetts General Hospital, Harvard Medical School, Charlestown, USA
| | - Allison M Andrews
- Department of Pathology & Laboratory Medicine, Temple University School of Medicine, Philadelphia, USA
| | - Tetyana P Buzhdygan
- Department of Pathology & Laboratory Medicine, Temple University School of Medicine, Philadelphia, USA
| | - Justin Mathew
- Neuroprotection Research Laboratory, Massachusetts General Hospital, Harvard Medical School, Charlestown, USA
| | - Wenlu Li
- Neuroprotection Research Laboratory, Massachusetts General Hospital, Harvard Medical School, Charlestown, USA
| | - Ken Arai
- Neuroprotection Research Laboratory, Massachusetts General Hospital, Harvard Medical School, Charlestown, USA
| | - Eng H Lo
- Neuroprotection Research Laboratory, Massachusetts General Hospital, Harvard Medical School, Charlestown, USA.,Department of Radiology, Massachusetts General Hospital, Boston, USA.,Department of Neurology, Massachusetts General Hospital, Boston, USA
| | - Servio H Ramirez
- Department of Pathology & Laboratory Medicine, Temple University School of Medicine, Philadelphia, USA.,The Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, USA
| | - Josephine Lok
- Neuroprotection Research Laboratory, Massachusetts General Hospital, Harvard Medical School, Charlestown, USA.,Department of Pediatrics, Pediatric Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| |
Collapse
|
97
|
Ejma M, Madetko N, Brzecka A, Alster P, Budrewicz S, Koszewicz M, Misiuk-Hojło M, Tomilova IK, Somasundaram SG, Kirkland CE, Aliev G. The Role of Stem Cells in the Therapy of Stroke. Curr Neuropharmacol 2021; 20:630-647. [PMID: 34365923 PMCID: PMC9608230 DOI: 10.2174/1570159x19666210806163352] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/19/2021] [Accepted: 06/03/2021] [Indexed: 11/22/2022] Open
Abstract
Background: Stroke is a major challenge in neurology due to its multifactorial genesis and irreversible consequences. Processes of endogenous post-stroke neurogenesis, although insufficient, may indicate possible direction of future therapy. Multiple research considers stem-cell-based approaches in order to maximize neuroregeneration and minimize post-stroke deficits. Objective: Aim of this study is to review current literature considering post-stroke stem-cell-based therapy and possibilities of inducing neuroregeneration after brain vascular damage. Methods: Papers included in this article were obtained from PubMed and MEDLINE databases. The following medical subject headings (MeSH) were used: “stem cell therapy”, “post-stroke neurogenesis”, “stem-cells stroke”, “stroke neurogenesis”, “stroke stem cells”, “stroke”, “cell therapy”, “neuroregeneration”, “neurogenesis”, “stem-cell human”, “cell therapy in human”. Ultimate inclusion was made after manual review of the obtained reference list. Results: Attempts of stimulating neuroregeneration after stroke found in current literature include supporting endogenous neurogenesis, different routes of exogenous stem cells supplying and extracellular vesicles used as a method of particle transport. Conclusion: Although further research in this field is required, post stroke brain recovery supported by exogenous stem cells seems to be promising future therapy revolutionizing modern neurology.
Collapse
Affiliation(s)
- Maria Ejma
- Department of Neurology, Wroclaw Medical University, 50-556 Wrocław, Borowska 213. Poland
| | - Natalia Madetko
- Department of Neurology, Medical University of Warsaw, Kondratowicza 8, 03-242 Warszawa. Poland
| | - Anna Brzecka
- Department of Pulmonology and Lung Oncology, Wroclaw Medical University, Grabiszynska 105, 53-439 Wroclaw. Poland
| | - Piotr Alster
- Department of Neurology, Medical University of Warsaw, Kondratowicza 8, 03-242 Warszawa. Poland
| | - Sławomir Budrewicz
- Department of Neurology, Wroclaw Medical University, 50-556 Wrocław, Borowska 213. Poland
| | - Magdalena Koszewicz
- Department of Neurology, Wroclaw Medical University, 50-556 Wrocław, Borowska 213. Poland
| | - Marta Misiuk-Hojło
- Department of Ophthalmology, Wroclaw Medical University, 50-556 Wroclaw, Borowska 213. Poland
| | - Irina K Tomilova
- Department of Biochemistry, Ivanovo State Medical Academy, Avenue Sheremetyevsky 8, Ivanovo, 153012. Russian Federation
| | - Siva G Somasundaram
- Department of Biological Sciences, Salem University, Salem, WV, 26426. United States
| | - Cecil E Kirkland
- Department of Biological Sciences, Salem University, Salem, WV, 26426. United States
| | - Gjumrakch Aliev
- Wroclaw Medical University, Department of Pulmonology and Lung Oncology, Wroclaw. Poland
| |
Collapse
|
98
|
Nasution I, Sjahrir H, Ilyas S, Ichwan M. The Effect of Ophiocephalus striatus sp. Extract on Nitric Oxide in Ischemic Stroke Model. Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.6299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND: One of alternative medicine in stroke therapy is Ophiocephalus striatus sp. extract. The nutrients contained in the O. striatus sp. extract, namely amino acids, fatty acids, cuprum, and zinc, are useful for the process of angiogenesis in poststroke patients through increased endothelial nitric oxide synthase.
AIM: We hypothesized that there was an effect of giving O. striatus sp. extract to cerebral angiogenesis process of Sprague Dawley rats ischemic stroke models through the level of NO.
METHODS: This was evidenced by conducting experimental studies on rats ischemic stroke models which were divided into five groups, (a) K (−) group (no ligation, no treatment), (b) K (+) group (ligation, no treatment), (c) P1 group (ligation, 200 mg extract), (d) P2 group (ligation, 400 mg extract), and (e) P3 group (ligation, 800 mg extract). Then blood sample was taken on day 3 to assess levels of NO.
RESULTS: There was increased level of NO in P1 (p = 0.001), P2 (p < 0.001), and P3 (p < 0.001) groups compared to K (+) group. The level of NO increases along with the increasing dose of O. striatus sp. extract. Histological examination revealed that there was formation of new blood vessel in the P1, P2, and P3 groups compared to K (+) group.
CONCLUSION: Our study showed that O. striatus sp. extract improves cerebral angiogenesis in rat models of ischemic stroke.
Collapse
|
99
|
Chen W, Xie L, Yu F, Li Y, Chen C, Xie W, Huang T, Zhang Y, Zhang S, Li P. Zebrafish as a Model for In-Depth Mechanistic Study for Stroke. Transl Stroke Res 2021; 12:695-710. [PMID: 34050491 DOI: 10.1007/s12975-021-00907-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/05/2021] [Accepted: 03/08/2021] [Indexed: 12/22/2022]
Abstract
Stroke is one of the world's leading causes of death and disability, posing enormous burden to the society. However, the pathogenesis and mechanisms that underlie brain injury and brain repair remain largely unknown. There's an unmet need of in-depth mechanistic research in this field. Zebrafish (Danio rerio) is a powerful tool in brain science research mainly due to its small size and transparent body, high genome synteny with human, and similar nervous system structures. It can be used to establish both hemorrhagic and ischemic stroke models easily and effectively through different ways. After the establishment of stroke model, research methods including behavioral test, in vivo imaging, and drug screening are available to explore mechanisms that underlie the brain injury and brain repair after stroke. This review focuses on the advantages and the feasibility of zebrafish stroke model, and will also introduce the key methods available for stroke studies in zebrafish, which may drive future mechanistic studies in the pursuit of discovering novel therapeutic targets for stroke patients.
Collapse
Affiliation(s)
- Weijie Chen
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Lv Xie
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Fang Yu
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Yan Li
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Chen Chen
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Wanqing Xie
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Tingting Huang
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Yueman Zhang
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Song Zhang
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China.
| | - Peiying Li
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China.
| |
Collapse
|
100
|
Yu R, Kim NS, Li Y, Jeong JY, Park SJ, Zhou B, Oh WJ. Vascular Sema3E-Plexin-D1 Signaling Reactivation Promotes Post-stroke Recovery through VEGF Downregulation in Mice. Transl Stroke Res 2021; 13:142-159. [PMID: 33978913 PMCID: PMC8766426 DOI: 10.1007/s12975-021-00914-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 04/21/2021] [Accepted: 04/26/2021] [Indexed: 12/18/2022]
Abstract
Post-stroke vascular remodeling, including angiogenesis, facilitates functional recovery. Proper vascular repair is important for efficient post-stroke recovery; however, the underlying mechanisms coordinating the diverse signaling pathways involved in vascular remodeling remain largely unknown. Recently, axon guidance molecules were revealed as key players in injured vessel remodeling. One such molecule, Semaphorin 3E (Sema3E), and its receptor, Plexin-D1, control vascular development by regulating vascular endothelial growth factor (VEGF) signaling. In this study, using a mouse model of transient brain infarction, we aimed to investigate whether Sema3E-Plexin-D1 signaling was involved in cerebrovascular remodeling after ischemic injury. We found that ischemic damage rapidly induced Sema3e expression in the neurons of peri-infarct regions, followed by Plexin-D1 upregulation in remodeling vessels. Interestingly, Plexin-D1 reemergence was concurrent with brain vessels entering an active angiogenic process. In line with this, Plxnd1 ablation worsened neurological deficits, infarct volume, neuronal survival rate, and blood flow recovery. Furthermore, reduced and abnormal vascular morphogenesis was caused by aberrantly increased VEGF signaling. In Plxnd1 knockout mice, we observed significant extravasation of intravenously administered tracers in the brain parenchyma, junctional protein downregulation, and mislocalization in regenerating vessels. This suggested that the absence of Sema3E-Plexin-D1 signaling is associated with blood–brain barrier (BBB) impairment. Finally, the abnormal behavioral performance, aberrant vascular phenotype, and BBB breakdown defects in Plxnd1 knockout mice were restored following the inhibition of VEGF signaling during vascular remodeling. These findings demonstrate that Sema3E-Plexin-D1 signaling can promote functional recovery by downregulating VEGF signaling in the injured adult brain.
Collapse
Affiliation(s)
- Ri Yu
- Neurovascular Biology Laboratory, Neurovascular Unit Research Group, Korea Brain Research Institute, Daegu, 41062, Republic of Korea.,College of Veterinary Medicine, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Nam-Suk Kim
- Neurovascular Biology Laboratory, Neurovascular Unit Research Group, Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Yan Li
- Neurovascular Biology Laboratory, Neurovascular Unit Research Group, Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Jin-Young Jeong
- Neurovascular Biology Laboratory, Neurovascular Unit Research Group, Korea Brain Research Institute, Daegu, 41062, Republic of Korea.,Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, 42988, Republic of Korea
| | - Sang-Joon Park
- College of Veterinary Medicine, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Bin Zhou
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Won-Jong Oh
- Neurovascular Biology Laboratory, Neurovascular Unit Research Group, Korea Brain Research Institute, Daegu, 41062, Republic of Korea.
| |
Collapse
|