1
|
Wen Y, Chen J, Long T, Chen F, Wang Z, Chen S, Zhang G, Li M, Zhang S, Kang H, Feng W, Wang G. miR-6760-5p suppresses neoangiogenesis by targeting Yes-associated protein 1 in patients with moyamoya disease undergoing indirect revascularization. Gene 2025; 937:149152. [PMID: 39662645 DOI: 10.1016/j.gene.2024.149152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/22/2024] [Accepted: 12/05/2024] [Indexed: 12/13/2024]
Abstract
OBJECTIVE The aim of this research was to investigate the specific regulatory role of miR-6760-5p in angiogenesis in moyamoya disease. METHODS HUVECs were transfected with miR-6760-5p inhibitor and mimics fragments, then subjected to assays for cell proliferation, migration, and tube formation. Subsequently, downstream target genes of miR-6760-5p were predicted and the protein expression levels of these genes were evaluated. The presence of miR-6760-5p and YAP1 was verified by a dual luciferase reporter gene test, followed by an assessment of the effects of YAP1 and miR-6760-5p on the HUVECs. RESULTS Comparatively to the control group, increased expression of miR-6760-5p decreased cell growth, movement, and tube formation. YAP1 gene was discovered as a target controlled by miR-6760-5p, with subsequent investigation confirming YAP1 as a gene regulated by miR-6760-5p. Additionally, miR-6760-5p was found to counteract the angiogenesis-promoting effect of YAP1. CONCLUSION The results of this research suggest a possible link between the miR-6760-5p gene found in the cerebrospinal fluid of individuals with moyamoya disease and the process of vascularization in this particular condition. The findings indicate that miR-6760-5p may be a new molecular indicator and potential target for the diagnosis of moyamoya disease.
Collapse
Affiliation(s)
- Yunyu Wen
- Neurosurgery Department, Nanfang Hospital, Southern Medical University, Located in Guangzhou, Guangdong, China; The Laboratory for Precision Neurosurgery is affiliated with Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China; The Institute of Brain Disease is part of Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China
| | - Junda Chen
- Neurosurgery Department, Nanfang Hospital, Southern Medical University, Located in Guangzhou, Guangdong, China; The Laboratory for Precision Neurosurgery is affiliated with Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China
| | - Tinghan Long
- Neurosurgery Department, Nanfang Hospital, Southern Medical University, Located in Guangzhou, Guangdong, China; The Laboratory for Precision Neurosurgery is affiliated with Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China
| | - Fangzhou Chen
- Neurosurgery Department, Nanfang Hospital, Southern Medical University, Located in Guangzhou, Guangdong, China; The Laboratory for Precision Neurosurgery is affiliated with Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China
| | - Zhibin Wang
- Neurosurgery Department, Nanfang Hospital, Southern Medical University, Located in Guangzhou, Guangdong, China; The Laboratory for Precision Neurosurgery is affiliated with Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China
| | - Siyuan Chen
- Neurosurgery Department, Nanfang Hospital, Southern Medical University, Located in Guangzhou, Guangdong, China; The Laboratory for Precision Neurosurgery is affiliated with Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China
| | - Guozhong Zhang
- Neurosurgery Department, Nanfang Hospital, Southern Medical University, Located in Guangzhou, Guangdong, China; The Laboratory for Precision Neurosurgery is affiliated with Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China
| | - Mingzhou Li
- Neurosurgery Department, Nanfang Hospital, Southern Medical University, Located in Guangzhou, Guangdong, China; The Laboratory for Precision Neurosurgery is affiliated with Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China
| | - Shichao Zhang
- Neurosurgery Department, Nanfang Hospital, Southern Medical University, Located in Guangzhou, Guangdong, China; The Laboratory for Precision Neurosurgery is affiliated with Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China
| | - Huibin Kang
- Neurosurgery Department, Nanfang Hospital, Southern Medical University, Located in Guangzhou, Guangdong, China; The Laboratory for Precision Neurosurgery is affiliated with Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China
| | - Wenfeng Feng
- Neurosurgery Department, Nanfang Hospital, Southern Medical University, Located in Guangzhou, Guangdong, China; The Laboratory for Precision Neurosurgery is affiliated with Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China; The Institute of Brain Disease is part of Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China.
| | - Gang Wang
- Neurosurgery Department, Nanfang Hospital, Southern Medical University, Located in Guangzhou, Guangdong, China; The Laboratory for Precision Neurosurgery is affiliated with Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China; The Institute of Brain Disease is part of Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China.
| |
Collapse
|
2
|
Bai S, Zhang G, Chen S, Wu X, Li J, Wang J, Chen D, Liu X, Wang J, Li Y, Tang Y, Tang Z. MicroRNA-451 Regulates Angiogenesis in Intracerebral Hemorrhage by Targeting Macrophage Migration Inhibitory Factor. Mol Neurobiol 2024; 61:10481-10499. [PMID: 38743209 PMCID: PMC11584486 DOI: 10.1007/s12035-024-04207-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 04/22/2024] [Indexed: 05/16/2024]
Abstract
Intracerebral hemorrhage (ICH) is a subtype of stroke with the highest fatality and disability rate. Up to now, commonly used first-line therapies have limited value in improving prognosis. Angiogenesis is essential to neurological recovery after ICH. Recent studies have shown that microRNA-451(miR-451) plays an important role in angiogenesis by regulating the function of vascular endothelial cells. We found miR-451 was significantly decreased in the peripheral blood of ICH patients in the acute stage. Based on the clinical findings, we conducted this study to investigate the potential regulatory effect of miR-451 on angiogenesis after ICH. The expression of miR-451 in ICH mouse model and in a hemin toxicity model of human brain microvascular endothelial cells (hBMECs) was decreased the same as in ICH patients. MiR-451 negatively regulated the proliferation, migration, and tube formation of hBMECs in vitro. MiR-451 negatively regulated the microvessel density in the perihematoma tissue and affected neural functional recovery of ICH mouse model. Knockdown of miR-451 could recovered tight junction and protect the integrity of blood-brain barrier after ICH. Based on bioinformatic programs, macrophage migration inhibitory factor (MIF) was predicted to be the target gene and identified to be regulated by miR-451 inhibiting the protein translation. And p-AKT and p-ERK were verified to be downstream of MIF in angiogenesis. These results all suggest that miR-451 will be a potential target for regulating angiogenesis in ICH.
Collapse
Affiliation(s)
- Shuang Bai
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ge Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shiling Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xuan Wu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiarui Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingxuan Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Danyang Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xia Liu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiahui Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuanwei Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingxin Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Zhouping Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
3
|
Zhang J. Non-coding RNAs and angiogenesis in cardiovascular diseases: a comprehensive review. Mol Cell Biochem 2024; 479:2921-2953. [PMID: 38306012 DOI: 10.1007/s11010-023-04919-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 12/18/2023] [Indexed: 02/03/2024]
Abstract
Non-coding RNAs (ncRNAs) have key roles in the etiology of many illnesses, including heart failure, myocardial infarction, stroke, and in physiological processes like angiogenesis. In transcriptional regulatory circuits that control heart growth, signaling, and stress response, as well as remodeling in cardiac disease, ncRNAs have become important players. Studies on ncRNAs and cardiovascular disease have made great progress recently. Here, we go through the functions of non-coding RNAs (ncRNAs) like circular RNAs (circRNAs), and microRNAs (miRNAs) as well as long non-coding RNAs (lncRNAs) in modulating cardiovascular disorders.
Collapse
Affiliation(s)
- Jie Zhang
- Medical School, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
4
|
Melas K, Talevi V, Imtiaz MA, Etteldorf R, Estrada S, Krüger DM, Pena‐Centeno T, Aziz NA, Fischer A, Breteler MMB. Blood-derived microRNAs are related to cognitive domains in the general population. Alzheimers Dement 2024; 20:7138-7159. [PMID: 39210637 PMCID: PMC11485070 DOI: 10.1002/alz.14197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/02/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024]
Abstract
INTRODUCTION Blood-derived microRNAs (miRNAs) are potential candidates for detecting and preventing subclinical cognitive dysfunction. However, replication of previous findings and identification of novel miRNAs associated with cognitive domains, including their relation to brain structure and the pathways they regulate, are still lacking. METHODS We examined blood-derived miRNAs and miRNA co-expression clusters in relation to cognitive domains, structural magnetic resonance imaging measures, target gene expression, and genetic variants in 2869 participants of a population-based cohort. RESULTS Five previously identified and 14 novel miRNAs were associated with cognitive domains. Eleven of these were also associated with cortical thickness and two with hippocampal volume. Multi-omics analysis showed that certain identified miRNAs were genetically influenced and regulated genes in pathways like neurogenesis and synapse assembly. DISCUSSION We identified miRNAs associated with cognitive domains, brain regions, and neuronal processes affected by aging and neurodegeneration, making them promising candidate blood-based biomarkers or therapeutic targets of subclinical cognitive dysfunction. HIGHLIGHTS We investigated the association of blood-derived microRNAs with cognitive domains. Five previously identified and 14 novel microRNAs were associated with cognition. Eleven cognition-related microRNAs were also associated with cortical thickness. Identified microRNAs were linked to genes associated with neuronal functions. Results provide putative biomarkers or therapeutic targets of cognitive aging.
Collapse
Affiliation(s)
- Konstantinos Melas
- Population Health SciencesGerman Centre for Neurodegenerative Diseases (DZNE)BonnGermany
| | - Valentina Talevi
- Population Health SciencesGerman Centre for Neurodegenerative Diseases (DZNE)BonnGermany
| | - Mohammed Aslam Imtiaz
- Population Health SciencesGerman Centre for Neurodegenerative Diseases (DZNE)BonnGermany
| | - Rika Etteldorf
- Population Health SciencesGerman Centre for Neurodegenerative Diseases (DZNE)BonnGermany
| | - Santiago Estrada
- Population Health SciencesGerman Centre for Neurodegenerative Diseases (DZNE)BonnGermany
- AI in Medical ImagingGerman Centre for Neurodegenerative Diseases (DZNE)BonnGermany
| | - Dennis M. Krüger
- Department for Epigenetics and Systems Medicine in Neurodegenerative DiseasesGerman Center for Neurodegenerative Diseases (DZNE)GöttingenGermany
| | - Tonatiuh Pena‐Centeno
- Department for Epigenetics and Systems Medicine in Neurodegenerative DiseasesGerman Center for Neurodegenerative Diseases (DZNE)GöttingenGermany
- Bioinformatics UnitGerman Centre for Neurodegenerative Diseases (DZNE)GöttingenGermany
| | - N. Ahmad Aziz
- Population Health SciencesGerman Centre for Neurodegenerative Diseases (DZNE)BonnGermany
- Department of NeurologyFaculty of MedicineUniversity of BonnBonnGermany
| | - André Fischer
- Department for Epigenetics and Systems Medicine in Neurodegenerative DiseasesGerman Center for Neurodegenerative Diseases (DZNE)GöttingenGermany
- Department for Psychiatry and PsychotherapyUniversity Medical Center GöttingenGöttingenGermany
- Cluster of Excellence MBExCUniversity of Göttingen & University Medical Center GoettingenGöttingenGermany
| | - Monique M. B. Breteler
- Population Health SciencesGerman Centre for Neurodegenerative Diseases (DZNE)BonnGermany
- Institute for Medical BiometryInformatics and Epidemiology (IMBIE)Faculty of MedicineUniversity of BonnBonnGermany
| |
Collapse
|
5
|
Zeng L, Hu P, Zhang Y, Li M, Zhao Y, Li S, Luo A. Macrophage migration inhibitor factor (MIF): Potential role in cognitive impairment disorders. Cytokine Growth Factor Rev 2024; 77:67-75. [PMID: 38548489 DOI: 10.1016/j.cytogfr.2024.03.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 03/11/2024] [Accepted: 03/15/2024] [Indexed: 06/22/2024]
Abstract
Macrophage migration inhibitory factor (MIF) is a cytokine in the immune system, participated in both innate and adaptive immune responses. Except from immune cells, MIF is also secreted by a variety of non-immune cells, including hematopoietic cells, endothelial cells (ECs), and neurons. MIF plays a crucial role in various diseases, such as sepsis, rheumatoid arthritis, acute kidney injury, and neurodegenerative diseases. The role of MIF in the neuropathogenesis of cognitive impairment disorders is emphasized, as it recruits multiple inflammatory mediators, leading to activating microglia or astrocyte-derived neuroinflammation. Furthermore, it contributes to the cell death of neurons and ECs with the binding of apoptosis-inducing factor (AIF) through parthanatos-associated apoptosis-inducing factor nuclease (PAAN) / MIF pathway. This review comprehensively delves into the relationship between MIF and the neuropathogenesis of cognitive impairment disorders, providing a series of emerging MIF-targeted pharmaceuticals as potential treatments for cognitive impairment disorders.
Collapse
Affiliation(s)
- Lian Zeng
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Pengchao Hu
- Hubei Provincial Clinical Research Center for Parkinson's Disease, Central Laboratory, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang 44100, China; Hubei Key Laboratory of Precision Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yu Zhang
- Hubei Provincial Clinical Research Center for Parkinson's Disease, Central Laboratory, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang 44100, China
| | - Mingyue Li
- Hubei Provincial Clinical Research Center for Parkinson's Disease, Central Laboratory, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang 44100, China
| | - Yilin Zhao
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shiyong Li
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Ailin Luo
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
6
|
He W, Cheng Y, Lai Y. CircRNA_102046 Affects the Occurrence and Development of Ischemic Stroke by Regulating the miR-493-5p/ROCK1 Signaling. Cardiovasc Toxicol 2024; 24:280-290. [PMID: 38376771 DOI: 10.1007/s12012-024-09831-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 01/15/2024] [Indexed: 02/21/2024]
Abstract
In our previous studies, the results have revealed that circRNA_102046 is significantly upregulated in plasma of patients with ischemic stroke, which closely related to NIHSS score. Human neural stem cells (hNSCs) were used for characterization and subcellular localization of circRNA_102046, and hNSCs OGD/R model was generated. The proliferation of cells was examined by CCK-8 assay. The expression levels of associated molecules were evaluated using RT-qPCR, immunofluorescence staining or western blotting. The binding and co-localization of associated molecules were also evaluated by RIP and FISH assay. Furthermore, MCAO mouse model was established to examine the effects of circRNA_102046 on the progression of ischemic stroke. Expression of circRNA_102046 was detected in the cytoplasma of hNSCs. Then OGD/R cell model was established, where the levels of circRNA_102046 was significantly up-regulated. Furthermore, knockdown of circRNA_102046 was able to enhance the proliferation and differentiation of OGD/R hNSCs. In further downstream molecular studies, the results indicated that circRNA_102046 could participate in the occurrence and development of ischemic stroke through targeting miR-493-5p. In addition, ROCK1 was identified as the putative target of miR-493-5p, and circRNA_102046 regulates the proliferation and differentiation of hNSCs via the miR-493-5p/ROCK1 signaling. More importantly, the infarct volumes of MCAO mice were remarkably reduced after the treatment with sh-circRNA_102046, which also up- and down-regulate the expression of miR-493-5p and ROCK1, respectively. Elucidating this novel pathway provides a theoretical basis for the development of new diagnostic approach and targeted treatment for ischemic stroke.
Collapse
Affiliation(s)
- Wentao He
- Department of General Medicine, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| | - Yuanyuan Cheng
- Department of General Medicine, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China.
| | - Yujie Lai
- Department of Neurology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| |
Collapse
|
7
|
Alikiaii B, Bagherniya M, Askari G, Rajendram R, Sahebkar A. MicroRNA Profiles in Critically Ill Patients. Curr Med Chem 2024; 31:6801-6825. [PMID: 37496239 DOI: 10.2174/0929867331666230726095222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 05/19/2023] [Accepted: 06/01/2023] [Indexed: 07/28/2023]
Abstract
The use of biomarkers to expedite diagnosis, prognostication, and treatment could significantly improve patient outcomes. The early diagnosis and treatment of critical illnesses can greatly reduce mortality and morbidity. Therefore, there is great interest in the discovery of biomarkers for critical illnesses. Micro-ribonucleic acids (miRNAs) are a highly conserved group of non-coding RNA molecules. They regulate the expression of genes involved in several developmental, physiological, and pathological processes. The characteristics of miRNAs suggest that they could be versatile biomarkers. Assay panels to measure the expression of several miRNAs could facilitate clinical decision-- making for a range of diseases. We have, in this paper, reviewed the current understanding of the role of miRNAs as biomarkers in critically ill patients.
Collapse
Affiliation(s)
- Babak Alikiaii
- Anesthesia and Critical Care Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Bagherniya
- Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Gholamreza Askari
- Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Rajkumar Rajendram
- Department of Medicine, King Abdulaziz Medical City, King Abdulaziz International Medical Research Center, Ministry of National Guard - Health Affairs, Riyadh, Saudi Arabia
- College of Medicine, King Saud bin Abdulaziz University of Health Sciences, Riyadh, Saudi Arabia
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
8
|
Li W, Xie L, Wang L, Lin F. CircRIMS promotes cerebral ischemia-reperfusion injury through increasing apoptosis and targeting the miR-96-5p/JAK/STAT1 axis. Brain Inj 2023; 37:1235-1244. [PMID: 37515578 DOI: 10.1080/02699052.2023.2237890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 04/07/2023] [Accepted: 06/12/2023] [Indexed: 07/31/2023]
Abstract
OBJECTIVE This study aims to explore the function of circRIMS in cerebral ischemia/reperfusion (CIR) and its regulatory mechanism. METHOD The expression of the circRIMS was examined in GEO chip data and validated by qRT-PCR analysis. A middle cerebral artery occlusion/repression (MCAO/R) model was developed using C57BL/6J mice. Starbase and circinteractome were employed to identify the target miRNA and mRNA. The result was confirmed by dual-luciferase reporter assay, and biotinylated RNA-pulldown assay. The cell viability and apoptosis were confirmed through CCK-8 and flow cytometry assay. RESULTS This study revealed that circRIMS expression was upregulated in MCAO mice model and OGD/RX-simulated cell model. Knockdown circRIMS demonstrated the functional of circRIMS in increasing cell viability, reducing apoptosis, LDH activity and inflammatory factors secretion in OGD/RX-simulated CIR injury in vitro. Additionally, miR-96-5p was identified as a target of circRIMS, while the STAT1 gene is a downstream gene of miR-96-5p, and JAK was also considered to be a downstream gene of the JAK-STAT pathway. Furthermore, inhibition of miR-96-5p or overexpression of STAT1 promoted the progression of CIR injury by elevating apoptosis, reducing cell viability, and increasing the secretion of inflammatory cytokines. CONCLUSION CircRIMS contributes to the progression of CIR injury via regulating miR-96-5p/JAK/STAT1 axis.
Collapse
Affiliation(s)
- Wei Li
- Department of Rehabilitation Medicine, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Lin Xie
- Department of Rehabilitation Medicine, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Lisha Wang
- Department of Neurology Intensive Care Unit, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Faliang Lin
- Department of Rehabilitation Medicine, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| |
Collapse
|
9
|
Fang J, Wang Z, Miao CY. Angiogenesis after ischemic stroke. Acta Pharmacol Sin 2023; 44:1305-1321. [PMID: 36829053 PMCID: PMC10310733 DOI: 10.1038/s41401-023-01061-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 02/01/2023] [Indexed: 02/26/2023] Open
Abstract
Owing to its high disability and mortality rates, stroke has been the second leading cause of death worldwide. Since the pathological mechanisms of stroke are not fully understood, there are few clinical treatment strategies available with an exception of tissue plasminogen activator (tPA), the only FDA-approved drug for the treatment of ischemic stroke. Angiogenesis is an important protective mechanism that promotes neural regeneration and functional recovery during the pathophysiological process of stroke. Thus, inducing angiogenesis in the peri-infarct area could effectively improve hemodynamics, and promote vascular remodeling and recovery of neurovascular function after ischemic stroke. In this review, we summarize the cellular and molecular mechanisms affecting angiogenesis after cerebral ischemia registered in PubMed, and provide pro-angiogenic strategies for exploring the treatment of ischemic stroke, including endothelial progenitor cells, mesenchymal stem cells, growth factors, cytokines, non-coding RNAs, etc.
Collapse
Affiliation(s)
- Jie Fang
- Department of Pharmacology, Second Military Medical University / Naval Medical University, Shanghai, 200433, China
| | - Zhi Wang
- Department of Pharmacology, Second Military Medical University / Naval Medical University, Shanghai, 200433, China
| | - Chao-Yu Miao
- Department of Pharmacology, Second Military Medical University / Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
10
|
Kui L, Li Z, Wang G, Li X, Zhao F, Jiao Y. CircPDS5B Reduction Improves Angiogenesis Following Ischemic Stroke by Regulating MicroRNA-223-3p/NOTCH2 Axis. Neurol Genet 2023; 9:e200074. [PMID: 37152444 PMCID: PMC10162703 DOI: 10.1212/nxg.0000000000200074] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 03/06/2023] [Indexed: 05/09/2023]
Abstract
Background and Objectives Ischemic stroke (IS) is responsible for major causes of global death and disability, for which promoting angiogenesis is a promising therapeutic strategy. This study analyzed circular RNA PDS5B (circPDS5B) and its related mechanisms in angiogenesis in IS. Methods In the permanent middle cerebral artery occlusion (pMCAO) mouse model, circPDS5B, microRNA (miR)-223-3p, and NOTCH2 levels were checked. By testing neurologic function, neuronal apoptosis, and expression of angiogenesis-related proteins in pMCAO mice, the protective effects of circPDS5B knockdown were probed. In human brain microvascular endothelial cells (HBMECs) under oxygen-glucose deprivation (OGD) conditions, the effects of circPDS5B, miR-223-3p, and NOTCH2 on angiogenesis were studied by measuring cellular activities. Results The increase of circPDS5B and NOTCH2 expression and the decrease of miR-223-3p expression were examined in pMCAO mice. Reducing circPDS5B expression indicated protection against neurologic dysfunction, apoptosis, and angiogenesis impairment. For circPDS5B-depleted or miR-223-3p-restored HBMECs under OGD treatment, angiogenesis was promoted. MiR-223-3p inhibition-associated reduction of angiogenesis could be counteracted by knocking down NOTCH2. CircPDS5B depletion-induced angiogenesis in OGD-conditioned HBMECs was repressed after overexpressing NOTCH2. Discussion In IS, the expression of circPDS5B was upregulated, and miR-223-3p inhibited HBMECs activity and promoted NOTCH2 expression, thus promoting IS. CircPDS5B reduction improves angiogenesis following ischemic stroke by regulating microRNA-223-3p/NOTCH2 axis.
Collapse
Affiliation(s)
- Ling Kui
- Dehong People's Hospital (Z.L., F.Z.), Mangshi; Shenzhen Qianhai Shekou Free Trade Zone Hospital (L.K., G.W., Y.J.), Shenzhen; State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and State Key Laboratory of Biological Big Data in Yunnan Province (X.L.), Yunnan Agricultural University, Kunming, China
| | - Zongyu Li
- Dehong People's Hospital (Z.L., F.Z.), Mangshi; Shenzhen Qianhai Shekou Free Trade Zone Hospital (L.K., G.W., Y.J.), Shenzhen; State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and State Key Laboratory of Biological Big Data in Yunnan Province (X.L.), Yunnan Agricultural University, Kunming, China
| | - Guoyun Wang
- Dehong People's Hospital (Z.L., F.Z.), Mangshi; Shenzhen Qianhai Shekou Free Trade Zone Hospital (L.K., G.W., Y.J.), Shenzhen; State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and State Key Laboratory of Biological Big Data in Yunnan Province (X.L.), Yunnan Agricultural University, Kunming, China
| | - Xuzhen Li
- Dehong People's Hospital (Z.L., F.Z.), Mangshi; Shenzhen Qianhai Shekou Free Trade Zone Hospital (L.K., G.W., Y.J.), Shenzhen; State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and State Key Laboratory of Biological Big Data in Yunnan Province (X.L.), Yunnan Agricultural University, Kunming, China
| | - Feng Zhao
- Dehong People's Hospital (Z.L., F.Z.), Mangshi; Shenzhen Qianhai Shekou Free Trade Zone Hospital (L.K., G.W., Y.J.), Shenzhen; State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and State Key Laboratory of Biological Big Data in Yunnan Province (X.L.), Yunnan Agricultural University, Kunming, China
| | - Yinming Jiao
- Dehong People's Hospital (Z.L., F.Z.), Mangshi; Shenzhen Qianhai Shekou Free Trade Zone Hospital (L.K., G.W., Y.J.), Shenzhen; State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and State Key Laboratory of Biological Big Data in Yunnan Province (X.L.), Yunnan Agricultural University, Kunming, China
| |
Collapse
|
11
|
Xuan W, Xie W, Li F, Huang D, Zhu Z, Lin Y, Lu B, Yu W, Li Y, Li P. Dualistic roles and mechanistic insights of macrophage migration inhibitory factor in brain injury and neurodegenerative diseases. J Cereb Blood Flow Metab 2023; 43:341-356. [PMID: 36369735 PMCID: PMC9941868 DOI: 10.1177/0271678x221138412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 09/21/2022] [Accepted: 10/03/2022] [Indexed: 11/15/2022]
Abstract
Macrophage migration inhibitory factor (MIF) is involved in various immune-mediated pathologies and regulates both innate and adaptive immune reactions, thus being related to several acute and chronic inflammatory diseases such as rheumatoid arthritis, septic shock, and atherosclerosis. Its role in acute and chronic brain pathologies, such as stroke and neurodegenerative diseases, has attracted increasing attention in recent years. In response to stimuli like hypoxia, inflammation or infection, different cell types can rapidly release MIF, including immune cells, endothelial cells, and neuron cells. Notably, clinical data from past decades also suggested a possible link between serum MIF levels and the severity of stroke and the evolving of neurodegenerative diseases. In this review, we summarize the major and recent findings focusing on the mechanisms of MIF modulating functions in brain injury and neurodegenerative diseases, which may provide important therapeutic targets meriting further investigation.
Collapse
Affiliation(s)
- Wei Xuan
- Department of Anesthesiology, Clinical Research Center, Renji
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China
| | - Wanqing Xie
- Department of Anesthesiology, Clinical Research Center, Renji
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China
| | - Fengshi Li
- Department of Neurosurgery, Renji Hospital, Shanghai Jiao Tong
University School of Medicine, Shanghai, China
| | - Dan Huang
- Department of Anesthesiology, Clinical Research Center, Renji
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China
| | - Ziyu Zhu
- Department of Anesthesiology, Clinical Research Center, Renji
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China
| | - Yuxuan Lin
- Department of Anesthesiology, Clinical Research Center, Renji
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China
| | - Binwei Lu
- Department of Anesthesiology, Clinical Research Center, Renji
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China
| | - Weifeng Yu
- Department of Anesthesiology, Clinical Research Center, Renji
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China
| | - Yan Li
- Department of Anesthesiology, Clinical Research Center, Renji
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China
| | - Peiying Li
- Department of Anesthesiology, Clinical Research Center, Renji
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China
| |
Collapse
|
12
|
Todoran R, Falcione SR, Clarke M, Joy T, Boghozian R, Jickling GC. microRNA as a therapeutic for ischemic stroke. Neurochem Int 2023; 163:105487. [PMID: 36657721 DOI: 10.1016/j.neuint.2023.105487] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 01/11/2023] [Accepted: 01/15/2023] [Indexed: 01/18/2023]
Abstract
microRNA (miRNA) are important regulators of gene expression. miRNA have the potential as a treatment to modulate genes, pathways and cells involved in ischemic stroke. In this review, we specifically present miRNA in stroke as a treatment to decrease thrombosis, reduce blood brain barrier (BBB) disruption and hemorrhagic transformation (HT), modulate inflammation, and modify angiogenesis. miRNA as a treatment for stroke is an emerging area with evidence from animal studies demonstrating its potential. While no miRNA is currently approved for human use, several have shown promise in clinical trials to treat medical conditions, such as miR-122 for hepatitis C. The role of miRNA as a treatment for specific applications in ischemic stroke is presented including a discussion of the benefits and barriers of miRNA as a treatment, and directions for future advancement.
Collapse
Affiliation(s)
- Raluca Todoran
- Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Sarina R Falcione
- Department of Medicine, Division of Neurology, University of Alberta, Edmonton, Canada
| | - Michael Clarke
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Canada
| | - Twinkle Joy
- Department of Medicine, Division of Neurology, University of Alberta, Edmonton, Canada
| | - Roobina Boghozian
- Department of Medicine, Division of Neurology, University of Alberta, Edmonton, Canada
| | - Glen C Jickling
- Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada; Department of Medicine, Division of Neurology, University of Alberta, Edmonton, Canada; Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Canada.
| |
Collapse
|
13
|
Predicting Microenvironment in CXCR4- and FAP-Positive Solid Tumors-A Pan-Cancer Machine Learning Workflow for Theranostic Target Structures. Cancers (Basel) 2023; 15:cancers15020392. [PMID: 36672341 PMCID: PMC9856808 DOI: 10.3390/cancers15020392] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
(1) Background: C-X-C Motif Chemokine Receptor 4 (CXCR4) and Fibroblast Activation Protein Alpha (FAP) are promising theranostic targets. However, it is unclear whether CXCR4 and FAP positivity mark distinct microenvironments, especially in solid tumors. (2) Methods: Using Random Forest (RF) analysis, we searched for entity-independent mRNA and microRNA signatures related to CXCR4 and FAP overexpression in our pan-cancer cohort from The Cancer Genome Atlas (TCGA) database-representing n = 9242 specimens from 29 tumor entities. CXCR4- and FAP-positive samples were assessed via StringDB cluster analysis, EnrichR, Metascape, and Gene Set Enrichment Analysis (GSEA). Findings were validated via correlation analyses in n = 1541 tumor samples. TIMER2.0 analyzed the association of CXCR4 / FAP expression and infiltration levels of immune-related cells. (3) Results: We identified entity-independent CXCR4 and FAP gene signatures representative for the majority of solid cancers. While CXCR4 positivity marked an immune-related microenvironment, FAP overexpression highlighted an angiogenesis-associated niche. TIMER2.0 analysis confirmed characteristic infiltration levels of CD8+ cells for CXCR4-positive tumors and endothelial cells for FAP-positive tumors. (4) Conclusions: CXCR4- and FAP-directed PET imaging could provide a non-invasive decision aid for entity-agnostic treatment of microenvironment in solid malignancies. Moreover, this machine learning workflow can easily be transferred towards other theranostic targets.
Collapse
|
14
|
Situ Y, Lu X, Cui Y, Xu Q, Deng L, Lin H, Shao Z, Chen J. Systematic Analysis of CXC Chemokine-Vascular Endothelial Growth Factor A Network in Colonic Adenocarcinoma from the Perspective of Angiogenesis. BIOMED RESEARCH INTERNATIONAL 2022; 2022:5137301. [PMID: 36246978 PMCID: PMC9553499 DOI: 10.1155/2022/5137301] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 09/09/2022] [Accepted: 09/14/2022] [Indexed: 12/04/2022]
Abstract
Background Tumor angiogenesis plays a vital role in tumorigenesis, proliferation, and metastasis. Recently, vascular endothelial growth factor A (VEGFA) and CXC chemokines have been shown to play vital roles in angiogenesis. Exploring the expression level, gene regulatory network, prognostic value, and target prediction of the CXC chemokine-VEGFA network in colon adenocarcinoma (COAD) is crucial from the perspective of tumor angiogenesis. Methods In this study, we analyzed gene expression and regulation, prognostic value, target prediction, and immune infiltrates related to the CXC chemokine-VEGFA network in patients with COAD using multiple databases (cBioPortal, UALCAN, Human Protein Atlas, GeneMANIA, GEPIA, TIMER (version 2.0), TRRUST (version 2), LinkedOmics, and Metascape). Results Our results showed that CXCL1/2/3/5/6/8/11/16/17 and VEGFA were markedly overexpressed, while CXCL12/13/14 were underexpressed in patients with COAD. Moreover, genetic alterations in the CXC chemokine-VEGFA network found at varying rates in patients with COAD were as follows: CXCL1/2/17 (2.1%), CXCL3/16 (2.6%), CXCL5/14 (2.4%), CXCL6 (3%), CXCL8 (0.8%), CXCL11/13 (1.9%), CXCL12 (0.6%), and VEGFA (1.3%). Promoter methylation of CXCL1/2/3/11/13/17 was considerably lower in patients with COAD, whereas methylation of CXCL5/6/12/14 and VEGFA was considerably higher. Furthermore, CXCL9/10/11 and VEGFA expression was notably correlated with the pathological stages of COAD. In addition, patients with COAD with high CXCL8/11/14 or low VEGFA expression levels survived longer than patients with dissimilar expression levels. CXC chemokines and VEGFA form a complex regulatory network through coexpression, colocalization, and genetic interactions. Moreover, many transcription factor targets of the CXC chemokine-VEGFA network in patients with COAD were identified: RELA, NFKB1, ZFP36, XBP1, HDAC2, SP1, ATF4, EP300, BRCA1, ESR1, HIF1A, EGR1, STAT3, and JUN. We further identified the top three miRNAs involved in regulating each CXC chemokine within the network: miR-518C, miR-369-3P, and miR-448 regulated CXCL1; miR-518C, miR-218, and miR-493 regulated CXCL2; miR-448, miR-369-3P, and miR-221 regulated CXCL3; miR-423 regulated CXCL13; miR-378, miR-381, and miR-210 regulated CXCL14; miR-369-3P, miR-382, and miR-208 regulated CXCL17; miR-486 and miR-199A regulated VEGFA. Furthermore, the CXC chemokine-VEGFA network in patients with COAD was notably associated with immune infiltration. Conclusions This study revealed that the CXC chemokine-VEGFA network might act as a prognostic biomarker for patients with COAD. Moreover, our study provides new therapeutic targets for COAD, serving as a reference for further research in the future.
Collapse
Affiliation(s)
- Yongli Situ
- Department of Parasitology, Guangdong Medical University, Zhanjiang, 524023 Guangdong, China
| | - Xiaoyong Lu
- Department of Parasitology, Guangdong Medical University, Zhanjiang, 524023 Guangdong, China
| | - Yongshi Cui
- Department of Parasitology, Guangdong Medical University, Zhanjiang, 524023 Guangdong, China
| | - Qinying Xu
- Department of Parasitology, Guangdong Medical University, Zhanjiang, 524023 Guangdong, China
| | - Li Deng
- Department of Parasitology, Guangdong Medical University, Zhanjiang, 524023 Guangdong, China
| | - Hao Lin
- Orthopedic Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524023 Guangdong, China
| | - Zheng Shao
- Department of Parasitology, Guangdong Medical University, Zhanjiang, 524023 Guangdong, China
| | - Jv Chen
- Department of Pharmacy, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001 Guangdong, China
| |
Collapse
|
15
|
Hsu CC, Chuang HK, Hsiao YJ, Teng YC, Chiang PH, Wang YJ, Lin TY, Tsai PH, Weng CC, Lin TC, Hwang DK, Hsieh AR. Polygenic Risk Score Improves Cataract Prediction in East Asian Population. Biomedicines 2022; 10:biomedicines10081920. [PMID: 36009466 PMCID: PMC9406175 DOI: 10.3390/biomedicines10081920] [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: 03/13/2022] [Revised: 06/30/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022] Open
Abstract
Cataracts, characterized by crystalline lens opacities in human eyes, is the leading cause of blindness globally. Due to its multifactorial complexity, the molecular mechanisms remain poorly understood. Larger cohorts of genome-wide association studies (GWAS) are needed to investigate cataracts’ genetic basis. In this study, a GWAS was performed on the largest Han population to date, analyzing a total of 7079 patients and 13,256 controls from the Taiwan Biobank (TWB) 2.0 cohort. Two cataract-associated SNPs with an adjustment of p < 1 × 10−7 in the older groups and nine SNPs with an adjustment of p < 1 × 10−6 in the younger group were identified. Except for the reported AGMO in animal models, most variations, including rs74774546 in GJA1 and rs237885 in OXTR, were not identified before this study. Furthermore, a polygenic risk score (PRS) was created for the young and old populations to identify high-risk cataract individuals, with areas under the receiver operating curve (AUROCs) of 0.829 and 0.785, respectively, after covariate adjustments. Younger individuals had 17.45 times the risk while older people had 10.97 times the risk when comparing individuals in the highest and lowest PRS quantiles. Validation analysis on an independent TWB1.0 cohort revealed AUROCs of 0.744 and 0.659.
Collapse
Affiliation(s)
- Chih-Chien Hsu
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 112027, Taiwan
| | - Hao-Kai Chuang
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112027, Taiwan
- Correspondence: (H.-K.C.); (D.-K.H.); (A.-R.H.); Tel.: +886-02-28757325 (D.-K.H.)
| | - Yu-Jer Hsiao
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112027, Taiwan
| | - Yuan-Chi Teng
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112027, Taiwan
| | - Pin-Hsuan Chiang
- Department of Statistics, Tamkang University, New Taipei 251301, Taiwan
| | - Yu-Jun Wang
- Department of Statistics, Tamkang University, New Taipei 251301, Taiwan
| | - Ting-Yi Lin
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112027, Taiwan
| | - Ping-Hsing Tsai
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112027, Taiwan
| | - Chang-Chi Weng
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 112027, Taiwan
| | - Tai-Chi Lin
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 112027, Taiwan
| | - De-Kuang Hwang
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 112027, Taiwan
- Correspondence: (H.-K.C.); (D.-K.H.); (A.-R.H.); Tel.: +886-02-28757325 (D.-K.H.)
| | - Ai-Ru Hsieh
- Department of Statistics, Tamkang University, New Taipei 251301, Taiwan
- Correspondence: (H.-K.C.); (D.-K.H.); (A.-R.H.); Tel.: +886-02-28757325 (D.-K.H.)
| |
Collapse
|
16
|
Neuroprotective Effect of Macrophage Migration Inhibitory Factor (MIF) in a Mouse Model of Ischemic Stroke. Int J Mol Sci 2022; 23:ijms23136975. [PMID: 35805977 PMCID: PMC9267067 DOI: 10.3390/ijms23136975] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/17/2022] [Accepted: 06/20/2022] [Indexed: 12/10/2022] Open
Abstract
The mechanism of the neuroprotective effect of the macrophage migration inhibitory factor (MIF) in vivo is unclear. We investigated whether the MIF promotes neurological recovery in an in vivo mouse model of ischemic stroke. Transient middle cerebral artery occlusion (MCAO) surgery was performed to make ischemic stroke mouse model. Male mice were allocated to a sham vehicle, a sham MIF, a middle cerebral artery occlusion (MCAO) vehicle, and MCAO+MIF groups. Transient MCAO (tMCAO) was performed in the MCAO groups, and the vehicle and the MIF were administered via the intracerebroventricular route. We evaluated the neurological functional scale, the rotarod test, and T2-weighted magnetic resonance imaging. The expression level of the microtubule-associated protein 2 (MAP2), Bcl2, and the brain-derived neurotrophic factor (BDNF) were further measured by Western blot assay. The Garcia test was significantly higher in the MCAO+MIF group than in the MCAO+vehicle group. The MCAO+MIF group exhibited significantly better performance on the rotarod test than the MCAO+vehicle group, which further had a significantly reduced total infarct volume on T2-weighted MRI imaging than the MCAO vehicle group. Expression levels of BDNF, and MAP2 tended to be higher in the MCAO+MIF group than in the MCAO+vehicle group. The MIF exerts a neuroprotective effect in an in vivo ischemic stroke model. The MIF facilitates neurological recovery and protects brain tissue from ischemic injury, indicating a possibility of future novel therapeutic agents for stroke patients.
Collapse
|
17
|
Noncoding RNA as Diagnostic and Prognostic Biomarkers in Cerebrovascular Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8149701. [PMID: 35498129 PMCID: PMC9042605 DOI: 10.1155/2022/8149701] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 03/22/2022] [Indexed: 02/06/2023]
Abstract
Noncoding RNAs (ncRNAs), such as microRNAs, long noncoding RNAs, and circular RNAs, play an important role in the pathophysiology of cerebrovascular diseases (CVDs). They are effectively detectable in body fluids, potentially suggesting new biomarkers for the early detection and prognosis of CVDs. In this review, the physiological functions of circulating ncRNAs and their potential role as diagnostic and prognostic markers in patients with cerebrovascular diseases are discussed, especially in acute ischemic stroke, subarachnoid hemorrhage, and moyamoya disease.
Collapse
|
18
|
Niu M, Li H, Li X, Yan X, Ma A, Pan X, Zhu X. Circulating Exosomal miRNAs as Novel Biomarkers Perform Superior Diagnostic Efficiency Compared With Plasma miRNAs for Large-Artery Atherosclerosis Stroke. Front Pharmacol 2021; 12:791644. [PMID: 34899352 PMCID: PMC8661454 DOI: 10.3389/fphar.2021.791644] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 11/08/2021] [Indexed: 11/13/2022] Open
Abstract
Recently, exosomal miRNAs have been reported to be associated with some diseases, and these miRNAs can be used for diagnosis and treatment. However, diagnostic biomarkers of exosomal miRNAs for ischemic stroke have rarely been studied. In the present study, we aimed to identify exosomal miRNAs that are associated with large-artery atherosclerosis (LAA) stroke, the most common subtype of ischemic stroke; to further verify their diagnostic efficiency; and to obtain promising biomarkers. High-throughput sequencing was performed on samples from 10 subjects. Quantitative real-time polymerase chain reaction (qRT-PCR) was performed on exosomes and plasma in the discovery phase (66 subjects in total) and the validation phase (520 subjects in total). We identified 5 candidate differentially expressed miRNAs (miR-369-3p, miR-493-3p, miR-379-5p, miR-1296-5p, and miR-1277-5p) in the discovery phase according to their biological functions, 4 of which (miR-369-3p, miR-493-3p, miR-379-5p, and miR-1296-5p) were confirmed in the validation phase. These four exosomal miRNAs could be used to distinguish LAA samples from small artery occlusion (SAO) samples, LAA samples from atherosclerosis (AS) samples, and LAA samples from control samples and were superior to plasma miRNAs. In addition, composite biomarkers achieved higher area under the curve (AUC) values than single biomarkers. According to our analysis, the expression levels of exosomal miR-493-3p and miR-1296-5p were negatively correlated with the National Institutes of Health Stroke Scale (NIHSS) score. The four identified exosomal miRNAs are promising biomarkers for the diagnosis of LAA stroke, and their diagnostic efficiency is superior to that of their counterparts in plasma.
Collapse
Affiliation(s)
- Mengying Niu
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hong Li
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xu Li
- Institute of Cerebrovascular Diseases, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaoqian Yan
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Aijun Ma
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xudong Pan
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China.,Institute of Cerebrovascular Diseases, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaoyan Zhu
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
19
|
Sun P, Ma F, Xu Y, Zhou C, Stetler RA, Yin KJ. Genetic deletion of endothelial microRNA-15a/16-1 promotes cerebral angiogenesis and neurological recovery in ischemic stroke through Src signaling pathway. J Cereb Blood Flow Metab 2021; 41:2725-2742. [PMID: 33910400 PMCID: PMC8504951 DOI: 10.1177/0271678x211010351] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cerebral angiogenesis is tightly controlled by specific microRNAs (miRs), including the miR-15a/16-1 cluster. Recently, we reported that endothelium-specific conditional knockout of the miR-15a/16-1 cluster (EC-miR-15a/16-1 cKO) promotes post-stroke angiogenesis and improves long-term neurological recovery by increasing protein levels of VEGFA, FGF2, and their respective receptors VEGFR2 and FGFR1. Herein, we further investigated the underlying signaling mechanism of these pro-angiogenic factors after ischemic stroke using a selective Src family inhibitor AZD0530. EC-miR-15a/16-1 cKO and age- and sex-matched wild-type littermate (WT) mice were subjected to 1 h middle cerebral artery occlusion (MCAO) and 28d reperfusion. AZD0530 was administered daily by oral gavage to both genotypes of mice 3-21d after MCAO. Compared to WT, AZD0530 administration exacerbated spatial cognitive impairments and brain atrophy in EC-miR-15a/16-1 cKO mice following MCAO. AZD0530 also attenuated long-term recovery of blood flow and inhibited the formation of new microvessels, including functional vessels with blood circulation, in the penumbra of stroked cKO mice. Moreover, AZD0530 blocked the Src signaling pathway by downregulating phospho-Src and its downstream mediators (p-Stat3, p-Akt, p-FAK, p-p44/42 MAPK, p-p38 MAPK) in post-ischemic brains. Collectively, our data demonstrated that endothelium-targeted deletion of the miR-15a/16-1 cluster promotes post-stroke angiogenesis and improves long-term neurological recovery via activating Src signaling pathway.
Collapse
Affiliation(s)
- Ping Sun
- Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Feifei Ma
- Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yang Xu
- Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Chao Zhou
- Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - R Anne Stetler
- Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ke-Jie Yin
- Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA
| |
Collapse
|
20
|
MiRNA: Involvement of the MAPK Pathway in Ischemic Stroke. A Promising Therapeutic Target. MEDICINA-LITHUANIA 2021; 57:medicina57101053. [PMID: 34684090 PMCID: PMC8539390 DOI: 10.3390/medicina57101053] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 12/22/2022]
Abstract
Ischemic stroke (IS) is a cerebrovascular disease with a high rate of disability and mortality. It is classified as the second leading cause of death that arises from the sudden occlusion of small vessels in the brain with consequent lack of oxygen and nutrients in the brain tissue. Following an acute ischemic event, the cascade of events promotes the activation of multiple signaling pathways responsible for irreversible neuronal damage. The mitogen-activated protein kinase (MAPK) signaling pathway transmits signals from the cell membrane to the nucleus in response to different stimuli, regulating proliferation, differentiation, inflammation, and apoptosis. Several lines of evidence showed that MAPK is an important regulator of ischemic and hemorrhagic cerebral vascular disease; indeed, it can impair blood–brain barrier (BBB) integrity and exacerbate neuroinflammation through the release of pro-inflammatory mediators implementing neurovascular damage after ischemic stroke. This review aims to illustrate the miRNAs involved in the regulation of MAPK in IS, in order to highlight possible targets for potential neuroprotective treatments. We also discuss some miRNAs (miR), including miR-145, miR-137, miR-493, and miR-126, that are important as they modulate processes such as apoptosis, neuroinflammation, neurogenesis, and angiogenesis through the regulation of the MAPK pathway in cerebral IS. To date, limited drug therapies are available for the treatment of IS; therefore, it is necessary to implement preclinical and clinical studies aimed at discovering novel therapeutic approaches to minimize post-stroke neurological damage.
Collapse
|
21
|
Zhao F, Xing Y, Jiang P, Hu L, Deng S. LncRNA MEG3 inhibits the proliferation of neural stem cells after ischemic stroke via the miR-493-5P/MIF axis. Biochem Biophys Res Commun 2021; 568:186-192. [PMID: 34273844 DOI: 10.1016/j.bbrc.2021.06.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 06/08/2021] [Indexed: 01/15/2023]
Abstract
OBJECTIVE The proliferation of neural stem cells (NSCs1), or lack thereof, can have profound effects on brain tissue remodeling for ischemic stroke (IS2). In this study, we aimed to reveal the influence of the lncRNA MEG3/miR-493-5p/MIF axis on NSC proliferation after IS. METHODS We established an oxygen glucose-deprivation/reoxygenation (OGD/R3) in vitro model of IS in NSCs. We evaluated NSC isolation efficiency and proliferation by NESTIN, SOX2, and PCNA immunofluorescence staining. MEG3 and miR-493-5P levels were assessed by quantitative real-time polymerase chain reaction (qRT-PCR4). Changes in MIF protein expression levels were analyzed using Western blotting. We then evaluated the role of MEG3 and miR-493-5p by transfection of si-MEG3, a miR-493-5p mimic, or miR-493-5p inhibitor. NSC proliferation was quantified using Cell Counting Kit-8 analysis. RESULTS NESTIN and SOX2 were co-expressed in endogenous NSCs. Following OGD/R, MEG3 and miR-493-5P were significantly upregulated in NSCs, while MIF levels decreased and proliferation was inhibited. Knockdown of MEG3 inhibited miR-493-5p and rescued expression of MIF and PCNA, restoring cellular proliferation levels. In NSCs transfected with a miR-493-5p mimic or inhibitor, MIF levels were down- or upregulated, respectively. Consistently, transfection of a miR-493-5p mimic reduced NSC proliferation, while transfection with a miR-493-5p inhibitor or si-MEG3 rescued the inhibitory effect of OGD/R on NSC proliferation. After co-transfection of si-MEG3 and a miR-493-5p mimic of OGD/R-induced NSCs, levels of PCNA, an indicator of cellular proliferation, were significantly reduced. Conclusion MEG3 inhibits NSC proliferation of after IS via positive regulation of miR-493-5p and potential subsequent downregulation of MIF.
Collapse
Affiliation(s)
- Fan Zhao
- Department of Forensic Medicine, Chongqing Medical University, #1 Yixueyuan Road, Chongqing, 400016, China
| | - Yu Xing
- Department of Forensic Medicine, Chongqing Medical University, #1 Yixueyuan Road, Chongqing, 400016, China
| | - Pu Jiang
- Department of Forensic Medicine, Chongqing Medical University, #1 Yixueyuan Road, Chongqing, 400016, China
| | - Lai Hu
- Department of Forensic Medicine, Chongqing Medical University, #1 Yixueyuan Road, Chongqing, 400016, China
| | - Shixiong Deng
- Department of Forensic Medicine, Chongqing Medical University, #1 Yixueyuan Road, Chongqing, 400016, China.
| |
Collapse
|
22
|
Effective silencing of miR-126 after ischemic stroke by means of intravenous α-tocopherol-conjugated heteroduplex oligonucleotide in mice. Sci Rep 2021; 11:14237. [PMID: 34244578 PMCID: PMC8270953 DOI: 10.1038/s41598-021-93666-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 06/11/2021] [Indexed: 12/03/2022] Open
Abstract
Brain endothelial cells (BECs) are involved in the pathogenesis of ischemic stroke. Recently, several microRNAs (miRNAs) in BECs were reported to regulate the endothelial function in ischemic brain. Therefore, modulation of miRNAs in BECs by a therapeutic oligonucleotide to inhibit miRNA (antimiR) could be a useful strategy for treating ischemic stroke. However, few attempts have been made to achieve this strategy via systemic route due to lack of efficient delivery-method toward BECs. Here, we have developed a new technology for delivering an antimiR into BECs and silencing miRNAs in BECs, using a mouse ischemic stroke model. We designed a heteroduplex oligonucleotide, comprising an antimiR against miRNA-126 (miR-126) known as the endothelial-specific miRNA and its complementary RNA, conjugated to α-tocopherol as a delivery ligand (Toc-HDO targeting miR-126). Intravenous administration of Toc-HDO targeting miR-126 remarkably suppressed miR-126 expression in ischemic brain of the model mice. In addition, we showed that Toc-HDO targeting miR-126 was delivered into BECs more efficiently than the parent antimiR in ischemic brain, and that it was delivered more effectively in ischemic brain than non-ischemic brain of this model mice. Our study highlights the potential of this technology as a new clinical therapeutic option for ischemic stroke.
Collapse
|
23
|
Zeng W, Lei Q, Ma J, Gao S, Ju R. Endothelial Progenitor Cell-Derived Microvesicles Promote Angiogenesis in Rat Brain Microvascular Endothelial Cells In vitro. Front Cell Neurosci 2021; 15:638351. [PMID: 33679329 PMCID: PMC7930325 DOI: 10.3389/fncel.2021.638351] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 01/26/2021] [Indexed: 12/14/2022] Open
Abstract
Brain microvascular endothelial cells (BMECs) are a major component of the blood-brain barrier that maintains brain homeostasis. Preserving and restoring the normal biological functions of BMECs can reverse or reduce brain injury. Endothelial progenitor cells (EPCs) may promote brain vascular remodeling and restore normal endothelial function. As a novel vehicle for cell-cell communication, microvesicles (MVs) have varied biological functions. The present study investigated the biological effects of EPC-derived MVs (EPC-MVs) on BMECs in vitro. We isolated MVs from the supernatant of EPCs in a serum-depleted medium. BMECs were cultured alone or in the presence of EPC-MVs. BMEC viability and proliferation were evaluated with the Cell Counting Kit-8 and by flow cytometry, and the proangiogenic effect of EPC-MVs on BMECs was assessed with the transwell migration, wound healing, and tube formation assays. Our results showed that EPC-derived MVs labeled with DiI were internalized by cultured BMECs; this enhanced BMEC viability and promoted their proliferation. EPC-MVs also stimulated migration and tube formation in BMECs. These results demonstrate that EPC-derived MVs exert a proangiogenic effect on BMECs, which has potential applications in cell-free therapy for brain injury.
Collapse
Affiliation(s)
- Wen Zeng
- Department of Neonatology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Qiaoling Lei
- Department of Neonatology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jiao Ma
- Department of Neonatology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Shuqiang Gao
- Department of Neonatology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Rong Ju
- Department of Neonatology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
24
|
Yu Y, Hou K, Ji T, Wang X, Liu Y, Zheng Y, Xu J, Hou Y, Chi G. The role of exosomal microRNAs in central nervous system diseases. Mol Cell Biochem 2021; 476:2111-2124. [PMID: 33528706 DOI: 10.1007/s11010-021-04053-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 01/09/2021] [Indexed: 02/07/2023]
Abstract
MicroRNAs (miRNA), endogenous non-coding RNAs approximately 22 nucleotides long, regulate gene expression by mediating translational inhibition or mRNA degradation. Exosomes are a tool for intercellular transmission of information in which miRNA exchange plays an important role. Under pathophysiological conditions in the central nervous system (CNS), cellular transmission of exosomal miRNAs can regulate signaling pathways. Exosomal miRNAs are involved in the occurrence and development of diverse CNS diseases, such as traumatic brain injury, spinal cord injury, stroke, neurodegenerative diseases, epilepsy, and glioma. The use of exosomes as transport vehicles for certain miRNAs provides a novel therapeutic strategy for CNS diseases. Furthermore, the exosomes in body fluids change with the occurrence of diseases, indicating that subtle changes in physiological and pathological processes in vivo could be recognized by analyzing exosomes. Exosomal analysis is expected to act as a novel tool for diagnosis and prediction of neurological diseases. In this review, we present the current understanding of the implications of miRNAs in CNS diseases and summarize the role and mechanism of action of exosomal miRNA in nervous system disease models.
Collapse
Affiliation(s)
- Yifei Yu
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130000, People's Republic of China
| | - Kun Hou
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, 130000, People's Republic of China
| | - Tong Ji
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130000, People's Republic of China
| | - Xishu Wang
- Clinical Medical College, Jilin University, Changchun, 130000, People's Republic of China
| | - Yining Liu
- Clinical Medical College, Jilin University, Changchun, 130000, People's Republic of China
| | - Yangyang Zheng
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130000, People's Republic of China
| | - Jinying Xu
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130000, People's Republic of China
| | - Yi Hou
- Department of Regeneration Medicine, School of Pharmaceutical Science of Jilin University, Changchun, 130000, People's Republic of China.
| | - Guangfan Chi
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130000, People's Republic of China.
| |
Collapse
|
25
|
Ding MH, Lozoya EG, Rico RN, Chew SA. The Role of Angiogenesis-Inducing microRNAs in Vascular Tissue Engineering. Tissue Eng Part A 2020; 26:1283-1302. [PMID: 32762306 DOI: 10.1089/ten.tea.2020.0170] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Angiogenesis is an important process in tissue repair and regeneration as blood vessels are integral to supply nutrients to a functioning tissue. In this review, the application of microRNAs (miRNAs) or anti-miRNAs that can induce angiogenesis to aid in blood vessel formation for vascular tissue engineering in ischemic diseases such as peripheral arterial disease and stroke, cardiac diseases, and skin and bone tissue engineering is discussed. Endothelial cells (ECs) form the endothelium of the blood vessel and are recognized as the primary cell type that drives angiogenesis and studied in the applications that were reviewed. Besides ECs, mesenchymal stem cells can also play a pivotal role in these applications, specifically, by secreting growth factors or cytokines for paracrine signaling and/or as constituent cells in the new blood vessel formed. In addition to delivering miRNAs or cells transfected/transduced with miRNAs for angiogenesis and vascular tissue engineering, the utilization of extracellular vesicles (EVs), such as exosomes, microvesicles, and EVs collectively, has been more recently explored. Proangiogenic miRNAs and anti-miRNAs contribute to angiogenesis by targeting the 3'-untranslated region of targets to upregulate proangiogenic factors such as vascular endothelial growth factor (VEGF), basic fibroblast growth factor, and hypoxia-inducible factor-1 and increase the transduction of VEGF signaling through the PI3K/AKT and Ras/Raf/MEK/ERK signaling pathways such as phosphatase and tensin homolog or regulating the signaling of other pathways important for angiogenesis such as the Notch signaling pathway and the pathway to produce nitric oxide. In conclusion, angiogenesis-inducing miRNAs and anti-miRNAs are promising tools for vascular tissue engineering for several applications; however, future work should emphasize optimizing the delivery and usage of these therapies as miRNAs can also be associated with the negative implications of cancer.
Collapse
Affiliation(s)
- May-Hui Ding
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, Texas, USA
| | - Eloy G Lozoya
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, Texas, USA
| | - Rene N Rico
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, Texas, USA
| | - Sue Anne Chew
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, Texas, USA
| |
Collapse
|
26
|
Vasudeva K, Munshi A. miRNA dysregulation in ischaemic stroke: Focus on diagnosis, prognosis, therapeutic and protective biomarkers. Eur J Neurosci 2020; 52:3610-3627. [PMID: 32022336 DOI: 10.1111/ejn.14695] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 01/10/2020] [Accepted: 01/31/2020] [Indexed: 01/14/2023]
Abstract
Stroke is one of the leading causes of death and disability in both developing and developed countries. Biomarkers for stroke and its outcome can greatly facilitate early detection and management of the disease. miRNAs have been explored for their potential as biomarkers for diagnosis, prognosis and brain injury in ischaemic stroke. A substantial body of evidence suggests that miRNAs play key roles in numerous cellular changes following ischaemic stroke including mitochondrial dysfunction, energy failure, cytokine-mediated cytotoxicity, oxidative stress, activation of glial cells, increased intracellular calcium levels inflammatory responses and disruption of the blood-brain barrier (BBB). In addition, targeting specific miRNAs, therapeutic modulation of brain injury and apoptosis can also be achieved. Therefore, the current review has been compiled within an aim to give an overview of the developments exploiting miRNAs at different stages of stroke as prognostic, diagnostic, protective and therapeutic biomarkers.
Collapse
Affiliation(s)
- Kanika Vasudeva
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, India
| | - Anjana Munshi
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, India
| |
Collapse
|
27
|
Zhang M, Ge DJ, Su Z, Qi B. miR-137 alleviates focal cerebral ischemic injury in rats by regulating JAK1/STAT1 signaling pathway. Hum Exp Toxicol 2020; 39:816-827. [PMID: 31961204 DOI: 10.1177/0960327119897103] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The repairing effect and potential mechanism of miR-137 on cerebral ischemic injury in rats was investigated. The volume of cerebral infarction and calculated brain water content was detected by triphenyltetrazolium chloride staining. The expression of inflammatory factors was detected by enzyme-linked immunosorbent assay. The pathological damage of brain tissue was analyzed by hematoxylin and eosin and Nissl staining. The apoptosis in ischemic brain tissue was detected by terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling. The levels of STAT1 and JAK1 proteins were analyzed by Western blot. The expression of miR-137 in primary hippocampal neurons was detected by reverse transcription polymerase chain reaction. miR-137 overexpression significantly improved brain damage in rats. miR-137 overexpression can reduce the expression of TNF-α, IL-1β, and IL-6. miR-137 overexpression can reduce the degree of brain tissue damage and inhibit the expression of JAK1 and STAT1 proteins. miR-137 overexpression can reduce oxygen-glucose deprivation (OGD)/R-induced cell damage, improve cell proliferation, and reduce apoptotic rate. JAK1 and STAT1 protein expression was inhibited in hippocampal neurons after OGD/R treatment after transfection with miR-137 mimic. After the addition of the Filgotinib inhibitor, the levels of JAK1 and STAT1 proteins were significantly reduced. The results suggested that miR-137 overexpression can effectively improve ischemic injury after focal cerebral ischemia and protect against by inhibiting JAK1/STAT1 pathway.
Collapse
Affiliation(s)
- M Zhang
- Department of Anesthesiology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| | - D J Ge
- Department of Anesthesiology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| | - Z Su
- Department of Anesthesiology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| | - B Qi
- Department of Anesthesiology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| |
Collapse
|
28
|
Zhou XB, Lai LF, Xie GB, Ding C, Xu X, Wang Y. LncRNA GAS5 sponges miRNA-221 to promote neurons apoptosis by up-regulated PUMA under hypoxia condition. Neurol Res 2019; 42:8-16. [PMID: 31878844 DOI: 10.1080/01616412.2019.1672382] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Objectives: Long noncoding RNAs (lncRNAs) play substantial roles in cerebral ischemia. Growth arrest-specific 5 (GAS5) was reported to be involved in stroke. In the present study, we aimed to investigate the roles of GAS5 in cerebral condition and unveil the underlying mechanism.Method: Transient focal ischemia was induced by intraluminal occlusion of the right Middle cerebral artery occlusion (MCAO) and 2,3,5-triphenyltetrazolium chloride (TTC) staining was used to evaluate the volume of cerebral infarction. RT-qPCR was applied to evaluate the level of GAS5 and miR-221. Fluorescence activated Cell Sorting (FACS) and Terminal deoxynucleotidyl transferased (TUNEL) were used for detection of apoptosis. Western blotting was applied for protein level. Luciferase assay was applied to reveal the underlying relationship between GAS5 and miR-221 or p53-upregulated modulator of apoptosis (PUMA) and miR-221.Results: The results indicated that GAS5 was up-regulated in MCAO rats and in vitro hypoxia cell model while miR-221 expression was decreased in vitro hypoxia cell model. GAS5 promoted cells apoptosis, while miR-221 inhibited cell apoptosis through regulation of PUMA and downstream JNK/H2AX signaling. Moreover, GAS5 and miR-221 have direct interaction and PUMA was the target of miR-221, indicating that GAS5 regulated PUMA through sponging miR-221.Conclusions: the present study revealed that GAS5 aggravated cell apoptosis in hypoxia condition via miR-221/PUMA axis, which may provide potential targets for the treatment of stroke.
Collapse
Affiliation(s)
- Xiao-Bing Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China
| | - Ling-Feng Lai
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China
| | - Guang-Bin Xie
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China
| | - Cong Ding
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China
| | - Xiang Xu
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China
| | - Yang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China
| |
Collapse
|
29
|
Gareev IF, Novikova LB, Beylerli OA. Application of microRNA in the therapy of ischemic stroke. КАРДИОВАСКУЛЯРНАЯ ТЕРАПИЯ И ПРОФИЛАКТИКА 2019. [DOI: 10.15829/1728-8800-2019-5-66-73] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|
30
|
Zheng Y, Wang L, Chen M, Liu L, Pei A, Zhang R, Gan S, Zhu S. Inhibition of T cell immunoglobulin and mucin-1 (TIM-1) protects against cerebral ischemia-reperfusion injury. Cell Commun Signal 2019; 17:103. [PMID: 31438964 PMCID: PMC6704646 DOI: 10.1186/s12964-019-0417-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 08/07/2019] [Indexed: 02/06/2023] Open
Abstract
Background The T cell Ig domain and mucin domain (TIM)-1 protein expressed on the surface of Th2 cells regulates the immune response by modulating cytokine production. The present study aimed to investigate the role and possible mechanism of TIM-1 in cerebral ischemia-reperfusion injury. Methods Western blot was used to detect TIM-1 and apoptosis-related protein expression, whereas TIM-1 mRNA was examined using quantitative real-time reverse transcription PCR. Flow cytometry and a TdT-mediated biotin-16-dUTP nick-end labeling (TUNEL) assay were used to detect the percentage of apoptotic cells and a pathological examination was performed. The migration of neutrophils and macrophages was analyzed by immunohistochemistry. Results Our results suggest that TIM-1 expression was transiently increased 24 h or 48 h following middle cerebral artery occlusion (MCAO)/reperfusion. The infarct size was markedly increased in MCAO, whereas treatment with a TIM-1-blocking mAb could reduce the infarct size. TIM-1 blocking mAb effectively reduced the number of neutrophils, macrophage functionality, cytokine (i.e., IL-6, IL-1β, and TNF-α) and chemokine (i.e., CXCL-1 and CXCL-2) production in the brain tissue. The effect of in vitro T cell damage on neurons was significantly reduced following treatment with a TIM-1 blocking mAb or the knockdown of TIM-1 in co-cultured T cells and neurons. Conclusion Take together, these results indicated that TIM-1 blockade ameliorated cerebral ischemia-reperfusion injury. Thus, TIM-1 disruption may serve as a novel target for therapy following MCAO.
Collapse
Affiliation(s)
- Yueying Zheng
- Department of Anesthesiology, The 1st Affiliated Hospital, School of Medicine, Zhejiang University, 79# Qingchun Road, 310003, Hangzhou, Zhejiang Province, People's Republic of China
| | - Liqing Wang
- Department of Anesthesiology, The 1st Affiliated Hospital, School of Medicine, Zhejiang University, 79# Qingchun Road, 310003, Hangzhou, Zhejiang Province, People's Republic of China
| | - Manli Chen
- Department of Anesthesiology, The 1st Affiliated Hospital, School of Medicine, Zhejiang University, 79# Qingchun Road, 310003, Hangzhou, Zhejiang Province, People's Republic of China
| | - Lu Liu
- Department of Anesthesiology, The 1st Affiliated Hospital, School of Medicine, Zhejiang University, 79# Qingchun Road, 310003, Hangzhou, Zhejiang Province, People's Republic of China
| | - Aijie Pei
- Department of Anesthesiology, The 1st Affiliated Hospital, School of Medicine, Zhejiang University, 79# Qingchun Road, 310003, Hangzhou, Zhejiang Province, People's Republic of China
| | - Rong Zhang
- Department of Anesthesiology, The 1st Affiliated Hospital, School of Medicine, Zhejiang University, 79# Qingchun Road, 310003, Hangzhou, Zhejiang Province, People's Republic of China
| | - Shuyuan Gan
- Department of Anesthesiology, The 1st Affiliated Hospital, School of Medicine, Zhejiang University, 79# Qingchun Road, 310003, Hangzhou, Zhejiang Province, People's Republic of China.
| | - Shengmei Zhu
- Department of Anesthesiology, The 1st Affiliated Hospital, School of Medicine, Zhejiang University, 79# Qingchun Road, 310003, Hangzhou, Zhejiang Province, People's Republic of China.
| |
Collapse
|
31
|
Li Q, Li Y, Zhang D, Gao H, Gao X. Downregulation of microRNA‑451 improves cell migration, invasion and tube formation in hypoxia‑treated HUVECs by targeting MIF. Mol Med Rep 2019; 20:1167-1177. [PMID: 31173234 PMCID: PMC6625462 DOI: 10.3892/mmr.2019.10357] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 01/04/2019] [Indexed: 12/23/2022] Open
Abstract
Angiogenesis is a critical process of recovery from cerebrovascular disease. A growing body of evidence has confirmed that microRNAs (miRNAs/miRs) have an important role in the modulation of angiogenesis under physiological and pathological conditions including cerebral ischemia injury (CII). Therefore, the aim of the present study was to explore the function and mechanism of microRNAs in regulating angiogenesis using a cell model of CII. Firstly, a miRNA microarray was performed to analyze miRNA expression in serum samples from patients with cerebral ischemia and the results revealed that miR-451 was one of the miRNAs that was the most significantly downregulated. Subsequently, human umbilical vein endothelial cells (HUVECs) were used as an in vitro model to further explore the mechanisms governing angiogenesis during hypoxia. The results demonstrated that overexpression of miR-451 had a significantly anti-angiogenic effect by suppressing tube formation, migration and wound healing in vitro. By contrast, reducing the expression of miR-451 promoted HUVEC migration and tubulogenesis under normoxic conditions. The present study further identified that macrophage migration inhibitory factor (MIF), an important angiogenic regulator, was a novel target of miR-451 that could reverse the effects of miR-451 on the regulation of angiogenesis in HUVECs under hypoxic or normoxic conditions. These results revealed that downregulation of miR-451 promotes angiogenesis by targeting MIF in hypoxic HUVECs and indicated that miR-451 is a potential candidate for CII therapeutics.
Collapse
Affiliation(s)
- Qian Li
- Department of Neurology, The Workers' Hospital of Tangshan City, Tangshan, Hebei 063000, P.R. China
| | - Yongqiu Li
- Department of Neurology, The Workers' Hospital of Tangshan City, Tangshan, Hebei 063000, P.R. China
| | - Dongsen Zhang
- Department of Neurology, The Workers' Hospital of Tangshan City, Tangshan, Hebei 063000, P.R. China
| | - Haifeng Gao
- Department of Neurology, The Workers' Hospital of Tangshan City, Tangshan, Hebei 063000, P.R. China
| | - Xuan Gao
- Department of Neurology, The Workers' Hospital of Tangshan City, Tangshan, Hebei 063000, P.R. China
| |
Collapse
|
32
|
Shen J, Zhao Z, Shang W, Liu C, Zhang B, Xu Z, Cai H. Fabrication and evaluation a transferrin receptor targeting nano-drug carrier for cerebral infarction treatment. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:192-200. [PMID: 30663409 DOI: 10.1080/21691401.2018.1548471] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
After cerebral infarction, the regeneration of microvascular played an important role in the recovery. Ginsenoside Rg1 (Rg1) had good effects on promoting angiogenesis and neuro-protection in cerebral infarction treatment. However, the blood-brain barrier (BBB) restricted Rg1 to enter into cerebral tissue. Transferrin receptor (TfR) was over-expressed in the BBB. In this study, we fabricated a TfR targeting nano-carrier (PATRC) to penetrate the BBB for treatment of cerebral infarction. A TfR targeted peptide was conjugated with the nano-carrier wrapped hydrophobic Rg1. The nanoscale size (132 ± 12 nm), polydispersity index (PDI =0.29) and the zeta potential (-38mv) were tested with dynamic light scattering optical system. Surface morphology (ellipse, mean diameter 122 ± 26 nm) was detected by transmission electron microscope (TEM). PATRC implement cell targeting ability on rat brain microvascular endothelial cells RBE4 in vitro detected by immunofluorescence and flow cytometry methods. Comparing with Rg1 threated group, the PATRC exhibited more prominent ability on the tube formation ability (p < .05) in vitro. Comparing with the Rg1 treated group, PATRC penetrated BBB in vivo detected by HPLC, decreased the brain infarction volume tested with TTC staining and promoted regeneration of microvascular in infarction zone detected by CD31 immunofluorescence. PATRC has great potentiality for wide application in clinic.
Collapse
Affiliation(s)
- Junyi Shen
- a Department of Integrated Traditional and Western Medicine, Jinling Hospital, School of Medicine , Nanjing University , Nanjing , China
| | - Zhiming Zhao
- a Department of Integrated Traditional and Western Medicine, Jinling Hospital, School of Medicine , Nanjing University , Nanjing , China
| | - Wei Shang
- a Department of Integrated Traditional and Western Medicine, Jinling Hospital, School of Medicine , Nanjing University , Nanjing , China
| | - Chunli Liu
- a Department of Integrated Traditional and Western Medicine, Jinling Hospital, School of Medicine , Nanjing University , Nanjing , China
| | - Beibei Zhang
- a Department of Integrated Traditional and Western Medicine, Jinling Hospital, School of Medicine , Nanjing University , Nanjing , China
| | - Zihan Xu
- a Department of Integrated Traditional and Western Medicine, Jinling Hospital, School of Medicine , Nanjing University , Nanjing , China
| | - Hui Cai
- a Department of Integrated Traditional and Western Medicine, Jinling Hospital, School of Medicine , Nanjing University , Nanjing , China
| |
Collapse
|
33
|
Ma Q, Zhang L, Pearce WJ. MicroRNAs in brain development and cerebrovascular pathophysiology. Am J Physiol Cell Physiol 2019; 317:C3-C19. [PMID: 30840494 DOI: 10.1152/ajpcell.00022.2019] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
MicroRNAs (miRNAs) are a class of highly conserved non-coding RNAs with 21-25 nucleotides in length and play an important role in regulating gene expression at the posttranscriptional level via base-paring with complementary sequences of the 3'-untranslated region of the target gene mRNA, leading to either transcript degradation or translation inhibition. Brain-enriched miRNAs act as versatile regulators of brain development and function, including neural lineage and subtype determination, neurogenesis, synapse formation and plasticity, neural stem cell proliferation and differentiation, and responses to insults. Herein, we summarize the current knowledge regarding the role of miRNAs in brain development and cerebrovascular pathophysiology. We review recent progress of the miRNA-based mechanisms in neuronal and cerebrovascular development as well as their role in hypoxic-ischemic brain injury. These findings hold great promise, not just for deeper understanding of basic brain biology but also for building new therapeutic strategies for prevention and treatment of pathologies such as cerebral ischemia.
Collapse
Affiliation(s)
- Qingyi Ma
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine , Loma Linda, California
| | - Lubo Zhang
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine , Loma Linda, California
| | - William J Pearce
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine , Loma Linda, California
| |
Collapse
|
34
|
Zheng Y, Pan C, Chen M, Pei A, Xie L, Zhu S. miR‑29a ameliorates ischemic injury of astrocytes in vitro by targeting the water channel protein aquaporin 4. Oncol Rep 2019; 41:1707-1717. [PMID: 30628716 PMCID: PMC6365700 DOI: 10.3892/or.2019.6961] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 12/13/2018] [Indexed: 01/22/2023] Open
Abstract
Ischemic stroke is the main cause of brain injury and results in a high rate of morbidity, disability and mortality. In the present study, we aimed to determine whether miR-29a played a protective role in oxygen glucose deprivation (OGD) injury via regulation of the water channel protein aquaporin 4 (AQP4). Real-time PCR and western blotting were used to assess miR-29a levels and AQP4 protein levels, respectively. Apoptosis was detected by flow cytometry, and lactate dehydrogenase (LDH) was determined by enzyme-linked immunosorbent assay (ELISA). Overexpression of miR-29a was significantly downregulated in OGD-induced primary astrocytes, and transfection with a miR-29a mimic decreased LDH release and apoptosis, and improved cell health in OGD-induced astrocytes. AQP4 was the target of miR-29a, which suppressed AQP4 expression, and knockdown of AQP4 mitigated OGD-induced astrocyte injury. Furthermore, miR-29a regulated AQP4 expression in OGD-induced astrocytes. AQP4 exacerbated astrocyte injury following ischemic stroke, and knockdown of AQP4 protected OGD/RX-induced primary cultured astrocytes against injury. The effect of miR-29a inhibitor on primary astrocytes was lost following AQP4 knockdown. These findings indicated that miR-29a prevented astrocyte injury in vitro by inhibiting AQP4. Thus, miR-29a may protect primary cultured astrocytes after OGD-induced injury by targeting AQP4, and may be a potential therapeutic target for ischemic injury of astrocytes.
Collapse
Affiliation(s)
- Yueying Zheng
- Department of Anesthesiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Caifei Pan
- Department of Anesthesiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Manli Chen
- Department of Anesthesiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Aijie Pei
- Department of Anesthesiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Liwei Xie
- Department of Anesthesiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Shengmei Zhu
- Department of Anesthesiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| |
Collapse
|
35
|
Rust R, Gantner C, Schwab ME. Pro- and antiangiogenic therapies: current status and clinical implications. FASEB J 2018; 33:34-48. [PMID: 30085886 DOI: 10.1096/fj.201800640rr] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Blood vessels nurture every part of the human body. Consequently, abnormalities in the vasculature are closely associated with a variety of diseases, including cerebral stroke, heart disease, retinopathy, and cancer. Pro- or antiangiogenic therapies can influence these diseases by regulating the growth of new blood vessels from a pre-existing vascular network or dampening excessive blood growth. However, clinical translation of these approaches is slow and challenging. In this review, we discuss recent preclinical approaches to regulate angiogenesis and their potential and risks in a clinical setting.-Rust, R., Gantner, C., Schwab, M. E. Pro- and antiangiogenic therapies: current status and clinical implications.
Collapse
Affiliation(s)
- Ruslan Rust
- Brain Research Institute, University of Zurich, Zurich, Switzerland.,Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland; and
| | - Christina Gantner
- Department of Biology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Martin E Schwab
- Brain Research Institute, University of Zurich, Zurich, Switzerland.,Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland; and
| |
Collapse
|
36
|
Abstract
Central nervous system (CNS) injuries, such as stroke, traumatic brain injury (TBI) and spinal cord injury (SCI), are important causes of death and long-term disability worldwide. MicroRNA (miRNA), small non-coding RNA molecules that negatively regulate gene expression, can serve as diagnostic biomarkers and are emerging as novel therapeutic targets for CNS injuries. MiRNA-based therapeutics include miRNA mimics and inhibitors (antagomiRs) to respectively decrease and increase the expression of target genes. In this review, we summarize current miRNA-based therapeutic applications in stroke, TBI and SCI. Administration methods, time windows and dosage for effective delivery of miRNA-based drugs into CNS are discussed. The underlying mechanisms of miRNA-based therapeutics are reviewed including oxidative stress, inflammation, apoptosis, blood-brain barrier protection, angiogenesis and neurogenesis. Pharmacological agents that protect against CNS injuries by targeting specific miRNAs are presented along with the challenges and therapeutic potential of miRNA-based therapies.
Collapse
Affiliation(s)
- Ping Sun
- Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Da Zhi Liu
- Department of Neurology and the M.I.N.D. Institute, University of California at Davis, Sacramento, CA, USA
| | - Glen C Jickling
- Department of Neurology, University of Alberta, Edmonton, Alberta, Canada
| | - Frank R Sharp
- Department of Neurology and the M.I.N.D. Institute, University of California at Davis, Sacramento, CA, USA
| | - Ke-Jie Yin
- Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Ke-Jie Yin, Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery, University of Pittsburgh School of Medicine, 200 Lothrop Street, BST S514, Pittsburgh, PA 15213, USA. Da Zhi Liu, Department of Neurology, University of California at Davis, Sacramento, CA 95817, USA.
| |
Collapse
|
37
|
Zhu WY, Jin X, Ma YC, Liu ZB. MIF protects against oxygen-glucose deprivation-induced ototoxicity in HEI-OC1 cochlear cells by enhancement of Akt-Nrf2-HO-1 pathway. Biochem Biophys Res Commun 2018; 503:665-670. [PMID: 29908183 DOI: 10.1016/j.bbrc.2018.06.058] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 06/12/2018] [Indexed: 12/17/2022]
Abstract
Ischemia and oxidative stress play crucial roles in the pathophysiology of sudden sensorineural hearing loss (SSNHL). Macrophage migration inhibitory factor (MIF) is a pro-inflammatory cytokine and serves an important role in hearing function. The present study was designed to evaluate the effect of MIF on oxygen-glucose deprivation (OGD)-induced ototoxicity and to elucidate its molecular mechanism. In HEI-OC1 auditory cells, OGD reduced cell viability and increased supernatant lactate dehydrogenase (LDH) and MIF in a time-dependent manner. However, the reduced cell viability exerted by OGD was attenuated by antioxidant and MIF. Luciferase reporter assay demonstrated that MIF could activate NF-E2-related factor 2 (Nrf2), and real-time PCR showed increased mRNA expressions of Nrf2 and two Nrf2-responsive genes, including heme oxygenase-1 (HO-1) and NAD(P)H:quinone oxidoreductase 1 (NQO1). MIF also suppressed oxidative stress induced by OGD, as demonstrated by decreased MDA and increased GSH in cellular supernatant. Inhibition of Nrf2 using siRNA suppressed HO-1 protein expression, the protective effect on OGD-induced injury and decrease in oxidative stress by MIF. Moreover, MIF prevented OGD-induced reduction of Akt1 phosphorylation at Ser473. LY294002, an inhibitor of PI3K/Akt signaling, attenuated the enhancement of Nrf2 protein and protective effect of MIF in OGD-treated cochlear cells. We demonstrate that MIF protects cochlear cells against OGD-induced injury through activation of Akt-Nrf2-HO-1 pathway.
Collapse
Affiliation(s)
- Wen-Yan Zhu
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, 223300, Jinagsu Province, China.
| | - Xin Jin
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, 223300, Jinagsu Province, China
| | - Yong-Chi Ma
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, 223300, Jinagsu Province, China
| | - Zhi-Biao Liu
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, 223300, Jinagsu Province, China
| |
Collapse
|
38
|
Exosomes Secreted by Adipose-Derived Stem Cells Contribute to Angiogenesis of Brain Microvascular Endothelial Cells Following Oxygen-Glucose Deprivation In Vitro Through MicroRNA-181b/TRPM7 Axis. J Mol Neurosci 2018; 65:74-83. [PMID: 29705934 DOI: 10.1007/s12031-018-1071-9] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 04/20/2018] [Indexed: 02/08/2023]
Abstract
Adipose-derived stem cells (ADSCs) have been demonstrated to promote cerebral vascular remodeling processes after stroke. However, the exact molecular mechanism by which ADSCs exert protective roles in ischemic stroke is still poorly understood. In this study, we identified the role of exosomal microRNA-181b-5p (181b-Exos) in regulating post-stroke angiogenesis. The results of migration assay and capillary network formation assay showed that exosomes secreted by ADSCs (ADSCs-Exos) promoted the mobility and angiogenesis of brain microvascular endothelial cells (BMECs) after oxygen-glucose deprivation (OGD). Quantitative real-time polymerase chain reaction (qRT-PCR) showed that microRNA-212-5p (miR-212-5p) and miR-181b-5p were upregulated in BMECs subjected to the brain extract of the middle cerebral artery occlusion rats. The migration distance and tube length were increased in BMECs cultured with 181b-Exos. Furthermore, we identified that transient receptor potential melastatin 7 (TRPM7) was a direct target of miR-181b-5p. TRPM7 mRNA and protein levels were declined in BMECs cultured with 181b-Exos, but not in BMECs cultured with 212-Exos. Overexpression of TRPM7 reversed the effects of 181b-Exos on migration and tube formation of BMECs. In addition, 181b-Exos upregulated the protein expression of hypoxia-inducible factor 1α and vascular endothelial cell growth factor, and downregulated the protein expression of tissue inhibitor of metalloproteinase 3. The regulatory effect of 181b-Exos was attenuated by overexpressing TRPM7. Altogether, ADSCs-Exos promote the angiogenesis of BMECs after OGD via miR-181b-5p/TRPM7 axis, suggesting that ADSCs-Exos may represent a novel therapeutic approach for stroke recovery.
Collapse
|
39
|
Yang X, Ji H, Yao Y, Lai X, Jiang Y, Wu D, Cai L, Zhu W, Gu X, Hu R, Li L, Xu L, Jiang M. Downregulation of circ_008018 protects against cerebral ischemia-reperfusion injury by targeting miR-99a. Biochem Biophys Res Commun 2018; 499:758-764. [PMID: 29605297 DOI: 10.1016/j.bbrc.2018.03.218] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 03/29/2018] [Indexed: 11/28/2022]
Abstract
Circular RNAs (circRNAs) are highly expressed in eukaryotic cells and regulate physiological and pathophysiological processes. However, the role of circRNAs in cerebral ischemia-reperfusion (I/R) injury remains largely unknown. In this study, we found that circ_008018 level was higher in the cortical tissue of mice with middle cerebral artery occlusion as compared to those in the sham group 24 h after reperfusion. Knockdown of circ_008018 attenuated cerebral I/R-induced brain tissue damage and neurological deficits in mice by inducing microRNA miR-99a overexpression. The decreased phosphorylation of Akt and glycogen synthase kinase 3β caused by I/R was partly reversed by circ_008018 silencing or miR-99a overexpression. Taken together, these results provide new insight into the mechanisms of apoptosis resulting from cerebral I/R injury and suggest that targeted inhibition of circ_008018 can protect against subsequent neurological damage.
Collapse
Affiliation(s)
- Xuelian Yang
- Department of Neurology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, PR China
| | - Haifeng Ji
- Department of Neurology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, PR China
| | - Yulan Yao
- Department of Critical Care Medicine, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, PR China
| | - Xiaoyin Lai
- Department of Neurology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, PR China
| | - Yufeng Jiang
- Department of Clinical Medicine, Clinical Medical School of Anhui Medical University, Anhui 230000, PR China
| | - Dayu Wu
- Department of Neurology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, PR China
| | - Liying Cai
- Department of Neurology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, PR China
| | - Wei Zhu
- Department of Neurology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, PR China
| | - Xiaju Gu
- Department of Neurology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, PR China
| | - Rongguo Hu
- Department of Neurology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, PR China
| | - Longxuan Li
- Department of Neurology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, PR China
| | - Lijuan Xu
- Department of Clinical Laboratory, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai, 200135 PR China.
| | - Mei Jiang
- Department of Neurology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, PR China.
| |
Collapse
|
40
|
Shi FP, Wang XH, Zhang HX, Shang MM, Liu XX, Sun HM, Song YP. MiR-103 regulates the angiogenesis of ischemic stroke rats by targeting vascular endothelial growth factor (VEGF). IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2018; 21:318-324. [PMID: 29511499 PMCID: PMC5817176 DOI: 10.22038/ijbms.2018.27267.6657] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 09/28/2017] [Indexed: 11/24/2022]
Abstract
OBJECTIVES To investigate the effect of miR-103 on the angiogenesis of ischemic stroke rats via targeting vascular endothelial growth factor (VEGF) at the molecular level. MATERIALS AND METHODS Rat models had received the middle cerebral artery occlusion (MCAO) or sham operation before grouping, and cell models of oxygen-glucose deprivation (OGD) were performed. FITC-dextran, matrigel, and Trans-well assays were used to evaluate the vascular density, tube formation, and cell migration respectively. The expression levels of miR-103 and VEGF were detected by quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting. Dual-luciferase assay was used for analyzing the targeting relationship between miR-103 and VEGF. RESULTS We found the reduced miR-103 in rats after MCAO. Down-regulating miR-103 with the miR-103 inhibitor enhanced VEGF, ameliorated the neurological scores, decreased infarct volume, and increased vascular density in rats after MCAO. Besides, in OGD human umbilical vein endothelial cells (HUVECs), inhibition of miR-103 could promote the increase of tube length and the migration of cells. Additionally, we found that miR-103 could directly target VEGF and thereby lead to the down-expression of VEGF. Meanwhile, si-VEGF could reverse the effect of miR-103 inhibitor on angiogenesis in rats subjected to MCAO. CONCLUSION Inhibition of miR-103 could promote ischemic stroke angiogenesis and reduce infarction volume via enhancing VEGF, which provides a new target for the clinical treatment of ischemic stroke.
Collapse
Affiliation(s)
- Fu-Ping Shi
- Department of Neurology, Affiliated Hospital of Hebei University, Baoding 071000, Hebei Province, China
| | - Xue-Hong Wang
- Department of Neurology, the Traditional Chinese Medicine Hospital of Yixian, Baoding 074200, Hebei Province, China
| | - Hong-Xin Zhang
- Laboratory Animal Center of Hebei University, Hebei University, Baoding 071000, Hebei Province, China
| | - Meng-Meng Shang
- Department of Neurology, Affiliated Hospital of Hebei University, Baoding 071000, Hebei Province, China
| | - Xiao-Xi Liu
- Department of Neurology, Affiliated Hospital of Hebei University, Baoding 071000, Hebei Province, China
| | - Hai-Min Sun
- Department of Neurology, Affiliated Hospital of Hebei University, Baoding 071000, Hebei Province, China
| | - Yue-Ping Song
- Department of Neurology, Affiliated Hospital of Hebei University, Baoding 071000, Hebei Province, China
| |
Collapse
|
41
|
Abstract
Stroke-induced endothelial cell injury leads to destruction of cerebral microvasculature and significant damage to the brain tissue. A subacute phase of cerebral ischemia is associated with regeneration involving the activation of vascular remodeling, neuroplasticity, neurogenesis, and neuroinflammation processes. Effective restoration and improvement of blood supply to the damaged brain tissue offers a potential therapy for stroke. microRNAs (miRNAs) are recently identified small RNA molecules that regulate gene expression and significantly influence the essential cellular processes associated with brain repair following stroke. A number of specific miRNAs are implicated in regulating the development and propagation of the ischemic tissue damage as well as in mediating post-stroke regeneration. In this review, I discuss the functions of the miRNA miR-155 and the effect of its in vivo inhibition on brain recovery following experimental cerebral ischemia. The article introduces new and unexplored approach to cerebral regeneration: regulation of brain tissue repair through a direct modulation of specific miRNA activity.
Collapse
Affiliation(s)
- Tamara Roitbak
- Department of Neurosurgery, Health Sciences Center, University of New Mexico, Albuquerque, NM, United States
| |
Collapse
|
42
|
Wang Y, Wang MD, Xia YP, Gao Y, Zhu YY, Chen SC, Mao L, He QW, Yue ZY, Hu B. MicroRNA-130a regulates cerebral ischemia-induced blood-brain barrier permeability by targeting Homeobox A5. FASEB J 2018; 32:935-944. [PMID: 29070584 DOI: 10.1096/fj.201700139rrr] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Blood-brain barrier (BBB) disruption plays a critical role in brain injury induced by cerebral ischemia, and preserving BBB integrity during ischemia could alleviate cerebral injury. We examined the role of miR-130a in ischemic BBB disruption by using models of rat middle cerebral artery occlusion and cell oxygen-glucose deprivation. We found that ischemia significantly increased microRNA-130a (miR-130a) level and that miR-130a was predominantly from brain microvascular endothelial cells. Antagomir-130a, an antagonist of miR-130a, could attenuate brain edema, lower BBB permeability, reduce infarct volume, and improve neurologic function. MiR-130a overexpression induced by miR-130a mimic increased monolayer permeability, and intercellular inhibition of miR-130a by a miR-130a inhibitor suppressed oxygen-glucose deprivation-induced increase in monolayer permeability. Moreover, dual luciferase reporter system showed that Homeobox A5 was the direct target of miR-130a. MiR-130a, by inhibiting Homeobox A5 expression, could down-regulate occludin, thereby increasing BBB permeability. Our results suggested that miR-130a might be implicated in ischemia-induced BBB dysfunction and serve as a target for the treatment of ischemic stroke.-Wang, Y., Wang, M.-D., Xia, Y.-P., Gao, Y., Zhu, Y.-Y., Chen, S.-C., Mao, L., He, Q.-W., Yue, Z.-Y., Hu, B. MicroRNA-130a regulates cerebral ischemia-induced blood-brain barrier permeability by targeting Homeobox A5.
Collapse
Affiliation(s)
- Yong Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meng-Die Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan-Peng Xia
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Gao
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi-Yi Zhu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sheng-Cai Chen
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Mao
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Quan-Wei He
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhen-Yu Yue
- Department of Neurology, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Bo Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
43
|
Sargento-Freitas J, Aday S, Nunes C, Cordeiro M, Gouveia A, Silva F, Machado C, Rodrigues B, Santo GC, Ferreira C, Amorim A, Sousa S, Gomes AC, Castelo-Branco M, Ferreira L, Cunha L. Endothelial progenitor cells enhance blood-brain barrier permeability in subacute stroke. Neurology 2017; 90:e127-e134. [PMID: 29237797 DOI: 10.1212/wnl.0000000000004801] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 09/29/2017] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To study the association among endothelial progenitor cells (EPCs), subacute blood-brain barrier (BBB) permeability, and clinical outcome after ischemic stroke, determining the micro RNAs of EPCs responsible for good clinical outcome. METHODS We included consecutive patients with nonlacunar acute ischemic strokes in the territory of a middle cerebral artery and ages between 18 and 80 years. Clinical outcome was defined as modified Rankin Scale score at 3 months. Neuroimaging was performed at day 0 and 7 by MRI, including assessment of BBB permeability by dynamic contrast enhancement. EPCs were isolated from peripheral venous blood, quantified, and submitted to in vitro functional tests, including migratory and angiogenic assays. Stroke hemodynamics were evaluated serially by ultrasound. Statistical significance was set at p < 0.05. RESULTS We included 45 patients; mean age was 70.0 ± 10.0 years. The in vitro functional properties of EPCs were associated with BBB permeability, particularly at day 7. The number of each EPC subset at both timepoints was not associated with BBB permeability. Permeability of BBB at day 7 was independently associated with improved clinical outcome (odds ratio 0.897; 95% confidence interval 0.816-0.986; p = 0.025). The EPCs (CD34+ cell subset) of patients with good clinical outcome showed 24 differentially expressed miRNAs, with a common effect on adherens junction pathway. CONCLUSIONS The functional properties of EPCs are associated with enhanced subacute permeability of BBB and improved clinical outcome after acute ischemic stroke.
Collapse
Affiliation(s)
- João Sargento-Freitas
- From the Stroke Unit (J.S.-F., A.G., F.S., C.M., B.R., G.C., L.C.), Centro Hospitalar e Universitário de Coimbra; Faculdade de Medicina da Universidade de Coimbra (J.S.-F., M.C.-B., L.F., L.C.); Centro de Neurociências e Biologia Celular (J.S.-F., S.A., A.C.G., L.F.); Instituto de Ciências Nucleares Aplicadas à Saúde (C.N., M.C., C.F., A.A., M.C.-B.), Coimbra; and Genomics Unit, Biocant-Biotechnology Innovation Center (S.S., A.C.G.), Cantanhede, Portugal
| | - Sezin Aday
- From the Stroke Unit (J.S.-F., A.G., F.S., C.M., B.R., G.C., L.C.), Centro Hospitalar e Universitário de Coimbra; Faculdade de Medicina da Universidade de Coimbra (J.S.-F., M.C.-B., L.F., L.C.); Centro de Neurociências e Biologia Celular (J.S.-F., S.A., A.C.G., L.F.); Instituto de Ciências Nucleares Aplicadas à Saúde (C.N., M.C., C.F., A.A., M.C.-B.), Coimbra; and Genomics Unit, Biocant-Biotechnology Innovation Center (S.S., A.C.G.), Cantanhede, Portugal
| | - César Nunes
- From the Stroke Unit (J.S.-F., A.G., F.S., C.M., B.R., G.C., L.C.), Centro Hospitalar e Universitário de Coimbra; Faculdade de Medicina da Universidade de Coimbra (J.S.-F., M.C.-B., L.F., L.C.); Centro de Neurociências e Biologia Celular (J.S.-F., S.A., A.C.G., L.F.); Instituto de Ciências Nucleares Aplicadas à Saúde (C.N., M.C., C.F., A.A., M.C.-B.), Coimbra; and Genomics Unit, Biocant-Biotechnology Innovation Center (S.S., A.C.G.), Cantanhede, Portugal
| | - Miguel Cordeiro
- From the Stroke Unit (J.S.-F., A.G., F.S., C.M., B.R., G.C., L.C.), Centro Hospitalar e Universitário de Coimbra; Faculdade de Medicina da Universidade de Coimbra (J.S.-F., M.C.-B., L.F., L.C.); Centro de Neurociências e Biologia Celular (J.S.-F., S.A., A.C.G., L.F.); Instituto de Ciências Nucleares Aplicadas à Saúde (C.N., M.C., C.F., A.A., M.C.-B.), Coimbra; and Genomics Unit, Biocant-Biotechnology Innovation Center (S.S., A.C.G.), Cantanhede, Portugal
| | - Ana Gouveia
- From the Stroke Unit (J.S.-F., A.G., F.S., C.M., B.R., G.C., L.C.), Centro Hospitalar e Universitário de Coimbra; Faculdade de Medicina da Universidade de Coimbra (J.S.-F., M.C.-B., L.F., L.C.); Centro de Neurociências e Biologia Celular (J.S.-F., S.A., A.C.G., L.F.); Instituto de Ciências Nucleares Aplicadas à Saúde (C.N., M.C., C.F., A.A., M.C.-B.), Coimbra; and Genomics Unit, Biocant-Biotechnology Innovation Center (S.S., A.C.G.), Cantanhede, Portugal
| | - Fernando Silva
- From the Stroke Unit (J.S.-F., A.G., F.S., C.M., B.R., G.C., L.C.), Centro Hospitalar e Universitário de Coimbra; Faculdade de Medicina da Universidade de Coimbra (J.S.-F., M.C.-B., L.F., L.C.); Centro de Neurociências e Biologia Celular (J.S.-F., S.A., A.C.G., L.F.); Instituto de Ciências Nucleares Aplicadas à Saúde (C.N., M.C., C.F., A.A., M.C.-B.), Coimbra; and Genomics Unit, Biocant-Biotechnology Innovation Center (S.S., A.C.G.), Cantanhede, Portugal
| | - Cristina Machado
- From the Stroke Unit (J.S.-F., A.G., F.S., C.M., B.R., G.C., L.C.), Centro Hospitalar e Universitário de Coimbra; Faculdade de Medicina da Universidade de Coimbra (J.S.-F., M.C.-B., L.F., L.C.); Centro de Neurociências e Biologia Celular (J.S.-F., S.A., A.C.G., L.F.); Instituto de Ciências Nucleares Aplicadas à Saúde (C.N., M.C., C.F., A.A., M.C.-B.), Coimbra; and Genomics Unit, Biocant-Biotechnology Innovation Center (S.S., A.C.G.), Cantanhede, Portugal
| | - Bruno Rodrigues
- From the Stroke Unit (J.S.-F., A.G., F.S., C.M., B.R., G.C., L.C.), Centro Hospitalar e Universitário de Coimbra; Faculdade de Medicina da Universidade de Coimbra (J.S.-F., M.C.-B., L.F., L.C.); Centro de Neurociências e Biologia Celular (J.S.-F., S.A., A.C.G., L.F.); Instituto de Ciências Nucleares Aplicadas à Saúde (C.N., M.C., C.F., A.A., M.C.-B.), Coimbra; and Genomics Unit, Biocant-Biotechnology Innovation Center (S.S., A.C.G.), Cantanhede, Portugal
| | - Gustavo Cordeiro Santo
- From the Stroke Unit (J.S.-F., A.G., F.S., C.M., B.R., G.C., L.C.), Centro Hospitalar e Universitário de Coimbra; Faculdade de Medicina da Universidade de Coimbra (J.S.-F., M.C.-B., L.F., L.C.); Centro de Neurociências e Biologia Celular (J.S.-F., S.A., A.C.G., L.F.); Instituto de Ciências Nucleares Aplicadas à Saúde (C.N., M.C., C.F., A.A., M.C.-B.), Coimbra; and Genomics Unit, Biocant-Biotechnology Innovation Center (S.S., A.C.G.), Cantanhede, Portugal
| | - Carlos Ferreira
- From the Stroke Unit (J.S.-F., A.G., F.S., C.M., B.R., G.C., L.C.), Centro Hospitalar e Universitário de Coimbra; Faculdade de Medicina da Universidade de Coimbra (J.S.-F., M.C.-B., L.F., L.C.); Centro de Neurociências e Biologia Celular (J.S.-F., S.A., A.C.G., L.F.); Instituto de Ciências Nucleares Aplicadas à Saúde (C.N., M.C., C.F., A.A., M.C.-B.), Coimbra; and Genomics Unit, Biocant-Biotechnology Innovation Center (S.S., A.C.G.), Cantanhede, Portugal
| | - André Amorim
- From the Stroke Unit (J.S.-F., A.G., F.S., C.M., B.R., G.C., L.C.), Centro Hospitalar e Universitário de Coimbra; Faculdade de Medicina da Universidade de Coimbra (J.S.-F., M.C.-B., L.F., L.C.); Centro de Neurociências e Biologia Celular (J.S.-F., S.A., A.C.G., L.F.); Instituto de Ciências Nucleares Aplicadas à Saúde (C.N., M.C., C.F., A.A., M.C.-B.), Coimbra; and Genomics Unit, Biocant-Biotechnology Innovation Center (S.S., A.C.G.), Cantanhede, Portugal
| | - Susana Sousa
- From the Stroke Unit (J.S.-F., A.G., F.S., C.M., B.R., G.C., L.C.), Centro Hospitalar e Universitário de Coimbra; Faculdade de Medicina da Universidade de Coimbra (J.S.-F., M.C.-B., L.F., L.C.); Centro de Neurociências e Biologia Celular (J.S.-F., S.A., A.C.G., L.F.); Instituto de Ciências Nucleares Aplicadas à Saúde (C.N., M.C., C.F., A.A., M.C.-B.), Coimbra; and Genomics Unit, Biocant-Biotechnology Innovation Center (S.S., A.C.G.), Cantanhede, Portugal
| | - Ana Catarina Gomes
- From the Stroke Unit (J.S.-F., A.G., F.S., C.M., B.R., G.C., L.C.), Centro Hospitalar e Universitário de Coimbra; Faculdade de Medicina da Universidade de Coimbra (J.S.-F., M.C.-B., L.F., L.C.); Centro de Neurociências e Biologia Celular (J.S.-F., S.A., A.C.G., L.F.); Instituto de Ciências Nucleares Aplicadas à Saúde (C.N., M.C., C.F., A.A., M.C.-B.), Coimbra; and Genomics Unit, Biocant-Biotechnology Innovation Center (S.S., A.C.G.), Cantanhede, Portugal
| | - Miguel Castelo-Branco
- From the Stroke Unit (J.S.-F., A.G., F.S., C.M., B.R., G.C., L.C.), Centro Hospitalar e Universitário de Coimbra; Faculdade de Medicina da Universidade de Coimbra (J.S.-F., M.C.-B., L.F., L.C.); Centro de Neurociências e Biologia Celular (J.S.-F., S.A., A.C.G., L.F.); Instituto de Ciências Nucleares Aplicadas à Saúde (C.N., M.C., C.F., A.A., M.C.-B.), Coimbra; and Genomics Unit, Biocant-Biotechnology Innovation Center (S.S., A.C.G.), Cantanhede, Portugal
| | - Lino Ferreira
- From the Stroke Unit (J.S.-F., A.G., F.S., C.M., B.R., G.C., L.C.), Centro Hospitalar e Universitário de Coimbra; Faculdade de Medicina da Universidade de Coimbra (J.S.-F., M.C.-B., L.F., L.C.); Centro de Neurociências e Biologia Celular (J.S.-F., S.A., A.C.G., L.F.); Instituto de Ciências Nucleares Aplicadas à Saúde (C.N., M.C., C.F., A.A., M.C.-B.), Coimbra; and Genomics Unit, Biocant-Biotechnology Innovation Center (S.S., A.C.G.), Cantanhede, Portugal.
| | - Luís Cunha
- From the Stroke Unit (J.S.-F., A.G., F.S., C.M., B.R., G.C., L.C.), Centro Hospitalar e Universitário de Coimbra; Faculdade de Medicina da Universidade de Coimbra (J.S.-F., M.C.-B., L.F., L.C.); Centro de Neurociências e Biologia Celular (J.S.-F., S.A., A.C.G., L.F.); Instituto de Ciências Nucleares Aplicadas à Saúde (C.N., M.C., C.F., A.A., M.C.-B.), Coimbra; and Genomics Unit, Biocant-Biotechnology Innovation Center (S.S., A.C.G.), Cantanhede, Portugal.
| |
Collapse
|
44
|
Soliman M, Andreeva K, Nasraoui O, Cooper NGF. A causal mediation model of ischemia reperfusion injury in the retina. PLoS One 2017; 12:e0187426. [PMID: 29121052 PMCID: PMC5679526 DOI: 10.1371/journal.pone.0187426] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 10/19/2017] [Indexed: 11/30/2022] Open
Abstract
The goal of this study is to develop a model that explains the relationship between microRNAs, transcription factors, and their co-target genes. This relationship was previously reported in gene regulatory loops associated with 24 hour (24h) and 7 day (7d) time periods following ischemia-reperfusion injury in a rat's retina. Using a model system of retinal ischemia-reperfusion injury, we propose that microRNAs first influence transcription factors, which in turn act as mediators to influence transcription of genes via triadic regulatory loops. Analysis of the relative contributions of direct and indirect regulatory influences on genes revealed that a substantial fraction of the regulatory loops (69% for 24 hours and 77% for 7 days) could be explained by causal mediation. Over 40% of the mediated loops in both time points were regulated by transcription factors only, while about 20% of the loops were regulated entirely by microRNAs. The remaining fractions of the mediated regulatory loops were cooperatively mediated by both microRNAs and transcription factors. The results from these analyses were supported by the patterns of expression of the genes, transcription factors, and microRNAs involved in the mediated loops in both post-ischemic time points. Additionally, network motif detection for the mediated loops showed a handful of time specific motifs related to ischemia-reperfusion injury in a rat's retina. In summary, the effects of microRNAs on genes are mediated, in large part, via transcription factors.
Collapse
Affiliation(s)
- Maha Soliman
- Department of Anatomical Sciences and Neurobiology, University of Louisville, Louisville, KY, United States of America
| | - Kalina Andreeva
- Department of Anatomical Sciences and Neurobiology, University of Louisville, Louisville, KY, United States of America
| | - Olfa Nasraoui
- Department of Computer Engineering and Computer Science, University of Louisville, Louisville, KY, United States of America
| | - Nigel G. F. Cooper
- Department of Anatomical Sciences and Neurobiology, University of Louisville, Louisville, KY, United States of America
| |
Collapse
|
45
|
Vascular Endothelial Growth Factor Isoform-B Stimulates Neurovascular Repair After Ischemic Stroke by Promoting the Function of Pericytes via Vascular Endothelial Growth Factor Receptor-1. Mol Neurobiol 2017; 55:3611-3626. [PMID: 28290152 DOI: 10.1007/s12035-017-0478-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 03/01/2017] [Indexed: 12/14/2022]
Abstract
Ischemic stroke triggers endogenous angiogenic mechanisms, which correlates with longer survival in patients. As such, promoting angiogenesis appears to be a promising approach. Experimental studies investigated mostly the potent angiogenic factor vascular endothelial growth factor isoform-A (VEGF-A). However, VEGF-A increases the risk of destabilizing the brain microvasculature, thus hindering the translation of its usage in clinics. An attractive alternative VEGF isoform-B (VEGF-B) was recently reported to act as a survival factor rather than a potent angiogenic factor. In this study, we investigated the therapeutic potential of VEGF-B in ischemic stroke using different in vivo and in vitro approaches. We showed that the delayed intranasal administration of VEGF-B reduced neuronal damage and inflammation. Unexpectedly, VEGF-B stimulated the formation of stable brain microvasculature within the injured region by promoting the interaction between endothelial cells and pericytes. Our data indicate that the effects of VEGF-B were mediated via its specific receptor VEGF receptor-1 (VEGFR-1) that is predominately expressed in brain pericytes. Importantly, VEGF-B promoted the survival of pericytes, and not brain endothelial cells, by inducing expression of the anti-apoptotic protein B-cell lymphoma 2 (Bcl-2) and the main protein involved in energy homeostasis AMP-activated protein kinase α (AMPKα). Moreover, we showed that VEGF-B stimulated the pericytic release of factors stimulating a "reparative angiogenesis" that does not compromise microvasculature stability. Our study unraveled hitherto unknown role of VEGF-B/VEGFR-1 signaling in regulating the function of pericytes. Furthermore, our findings suggest that brain microvasculature stabilization via VEGF-B constitutes a safe therapeutic approach for ischemic stroke.
Collapse
|
46
|
Chen S, Wang M, Yang H, Mao L, He Q, Jin H, Ye ZM, Luo XY, Xia YP, Hu B. LncRNA TUG1 sponges microRNA-9 to promote neurons apoptosis by up-regulated Bcl2l11 under ischemia. Biochem Biophys Res Commun 2017; 485:167-173. [DOI: 10.1016/j.bbrc.2017.02.043] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 02/06/2017] [Accepted: 02/07/2017] [Indexed: 12/11/2022]
|