1
|
Chen C, Tang F, Zhu M, Wang C, Zhou H, Zhang C, Feng Y. Role of inflammatory mediators in intracranial aneurysms: A review. Clin Neurol Neurosurg 2024; 242:108329. [PMID: 38781806 DOI: 10.1016/j.clineuro.2024.108329] [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: 01/28/2024] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024]
Abstract
The formation, growth, and rupture of intracranial aneurysms (IAs) involve hemodynamics, blood pressure, external stimuli, and a series of hormonal changes. In addition, inflammatory response causes the release of a series of inflammatory mediators, such as IL, TNF-α, MCP-1, and MMPs, which directly or indirectly promote the development process of IA. However, the specific role of these inflammatory mediators in the pathophysiological process of IA remains unclear. Recently, several anti-inflammatory, lipid-lowering, hormone-regulating drugs have been found to have a potentially protective effect on reducing IA formation and rupture in the population. These therapeutic mechanisms have not been fully elucidated, but we can look for potential therapeutic targets that may interfere with the formation and breakdown of IA by studying the relevant inflammatory response and the mechanism of IA formation and rupture involved in inflammatory mediators.
Collapse
Affiliation(s)
- Cheng Chen
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao city, China
| | - Fengjiao Tang
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao city, China
| | - Meng Zhu
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao city, China
| | - Chao Wang
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao city, China
| | - Han Zhou
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao city, China
| | - Chonghui Zhang
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao city, China
| | - Yugong Feng
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao city, China.
| |
Collapse
|
2
|
Kamińska J, Tylicka M, Sutkowska K, Gacuta KM, Sawicka MM, Kowalewska E, Ćwiklińska-Dworakowska M, Maciejczyk M, Łysoń T, Kornhuber J, Lewczuk P, Matowicka-Karna J, Koper-Lenkiewicz OM. The preliminary study suggests an association between NF-ĸB pathway activation and increased plasma 20S proteasome activity in intracranial aneurysm patients. Sci Rep 2024; 14:3941. [PMID: 38366068 PMCID: PMC10873410 DOI: 10.1038/s41598-024-54692-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 02/15/2024] [Indexed: 02/18/2024] Open
Abstract
The significant role of increased activation of 20S proteasomes in the development of abdominal aortic aneurysms has been well-established in a mouse model. The available literature lacks similar studies concerning brain aneurysms. The aim of the study was to verify the hypothesis that patients with unruptured intracranial aneurysms (UIA) have increased 20S proteasome ChT-L activity compared to the control group of individuals without vascular lesions in the brain. In the next step, the relationship between the activity of 20S proteasomes ChT-L and precursor proteins from the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) family, namely NF-κB1 (p105), NF-κB2 (p100), NF-κB p65, and the inflammatory chemokine MCP-1, was examined. Patients with UIA had significantly higher 20S ChT-L proteasome activity compared to the control group. Patients with multiple aneurysms had significantly higher 20S proteasome ChT-L activity compared to those with single aneurysms. In patients with UIA, the activity of the 20S proteasome ChT-L negatively correlated with the concentration of NF-κB1 (p105) and NF-κB p65 precursor proteins and positively correlated with the concentration of the cerebrospinal fluid chemokine MCP-1. Our results may suggest that increased 20S proteasome ChT-L activity in UIA patients modulates inflammation in the cerebral arterial vessel via the MCP-1 chemokine as a result of activation of the canonical NF-κB pathway.
Collapse
Affiliation(s)
- Joanna Kamińska
- Department of Clinical Laboratory Diagnostics, Medical University of Bialystok, 15A Jerzego Waszyngtona St., 15-269, Białystok, Poland.
| | - Marzena Tylicka
- Department of Biophysics, Medical University of Bialystok, 2A Adama Mickiewicza St., 15-089, Białystok, Poland
| | - Kinga Sutkowska
- Department of Clinical Laboratory Diagnostics, Clinical Hospital of the Medical University of Bialystok, 15A Jerzego Waszyngtona St., 15-269, Białystok, Poland
| | - Karolina Marta Gacuta
- Department of Clinical Laboratory Diagnostics, Clinical Hospital of the Medical University of Bialystok, 15A Jerzego Waszyngtona St., 15-269, Białystok, Poland
| | - Magdalena Maria Sawicka
- Department of Analysis and Bioanalysis of Medicines, Medical University of Bialystok, 2D Mickiewicza St., 15-222, Białystok, Poland
| | - Ewa Kowalewska
- Department of Clinical Laboratory Diagnostics, Clinical Hospital of the Medical University of Bialystok, 15A Jerzego Waszyngtona St., 15-269, Białystok, Poland
| | - Magdalena Ćwiklińska-Dworakowska
- Department of Oncological Surgery and General Surgery, Independent Public Health Care Facility of the Ministry of the Interior and Administration in Bialystok named Marian Zyndram-Kościałkowski, 27 Fabryczna St., 15-471, Białystok, Poland
| | - Mateusz Maciejczyk
- Department of Hygiene, Epidemiology, and Ergonomics, Medical University of Białystok, 2C Mickiewicza St., 15-022, Białystok, Poland
| | - Tomasz Łysoń
- Department of Neurosurgery, Medical University of Bialystok/Clinical Hospital of the Medical University of Bialystok, 24A Marii Skłodowskiej-Curie St., 15-276, Białystok, Poland
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, University Hospital Erlangen, and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Piotr Lewczuk
- Department of Psychiatry and Psychotherapy, University Hospital Erlangen, and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15A Jerzego Waszyngtona St., 15-269, Białystok, Poland
| | - Joanna Matowicka-Karna
- Department of Clinical Laboratory Diagnostics, Medical University of Bialystok, 15A Jerzego Waszyngtona St., 15-269, Białystok, Poland
| | - Olga Martyna Koper-Lenkiewicz
- Department of Clinical Laboratory Diagnostics, Medical University of Bialystok, 15A Jerzego Waszyngtona St., 15-269, Białystok, Poland.
| |
Collapse
|
3
|
Wang H, Wang L, Liu Y, Men W, Hao W, Fang C, Li C, Zhang L. Plasma levels of CD36 and glutathione as biomarkers for ruptured intracranial aneurysm. Open Life Sci 2023; 18:20220757. [PMID: 38196515 PMCID: PMC10775171 DOI: 10.1515/biol-2022-0757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/18/2023] [Accepted: 09/24/2023] [Indexed: 01/11/2024] Open
Abstract
Evidence has proved that intracranial aneurysm (IA) formation and rupture might be closely related to inflammatory response and oxidative stress. Our objective was to evaluate the potential of CD36 and glutathione (GSH) as biomarkers for IA. In this study, the enzyme-linked immunosorbent assay was used to measure the plasma levels of CD36 and GSH in 30 IA patients and 30 healthy controls. Then, correlation analysis, receiver operating characteristic (ROC) curve, and logistic regression analysis were performed. The results showed that the plasma level of CD36 in IA patients was significantly higher than that in the control group (P < 0.0001), and plasma GSH was significantly lower compared with that in the control group (P < 0.0001). ROC analysis showed that CD36 and GSH had high sensitivity (90.0 and 96.6%) and specificity (96.6 and 86.6%) for IA diagnosis. The combined sensitivity and specificity achieved were 100 and 100%, respectively. The plasma levels of CD36 and GSH did not show a significant correlation with age, the Glasgow Coma Scale, Hunter-Hess score, aneurysm size, aneurysm height, aneurysm neck, and aspect ratio. The AUC of the logistic regression model based on CD36 and GSH was 0.505. Our results suggested that the combination of plasma CD36 and GSH could serve as potential biomarkers for IA rupture.
Collapse
Affiliation(s)
- Hanbin Wang
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei Province, China
| | - Luxuan Wang
- Department of Neurological Function Examination, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei Province, China
| | - Yunmei Liu
- Department of Reproductive Medicine, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei Province, China
| | - Weidong Men
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei Province, China
| | - Wanjiao Hao
- Department of Reproductive Medicine, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei Province, China
| | - Chuan Fang
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei Province, China
- Postdoctoral Research Station of Neurosurgery, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei Province, China
- Key Laboratory of Precise Diagnosis and Treatment of Glioma in Hebei Province, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei Province, China
| | - Chunhui Li
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei Province, China
| | - Lijian Zhang
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei Province, China
- Postdoctoral Research Station of Neurosurgery, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei Province, China
- Key Laboratory of Precise Diagnosis and Treatment of Glioma in Hebei Province, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei Province, China
| |
Collapse
|
4
|
Freiholtz D, Bergman O, Pradhananga S, Lång K, Poujade FA, Granath C, Olsson C, Franco-Cereceda A, Sahlén P, Eriksson P, Björck HM. SPP1/osteopontin: a driver of fibrosis and inflammation in degenerative ascending aortic aneurysm? J Mol Med (Berl) 2023; 101:1323-1333. [PMID: 37698712 PMCID: PMC10560177 DOI: 10.1007/s00109-023-02370-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 08/25/2023] [Accepted: 09/04/2023] [Indexed: 09/13/2023]
Abstract
Degenerative ascending aortic aneurysm (AscAA) is a silent and potentially fatal disease characterized by excessive vascular inflammation and fibrosis. We aimed to characterize the cellular and molecular signature for the fibrotic type of endothelial mesenchymal transition (EndMT) that has previously been described in degenerative AscAA. Patients undergoing elective open-heart surgery for AscAA and/or aortic valve repair were recruited. Gene expression in the intima-media of the ascending aorta was measured in 22 patients with non-dilated and 24 with dilated aortas, and candidate genes were identified. Protein expression was assessed using immunohistochemistry. Interacting distal gene enhancer regions were identified using targeted chromosome conformation capture (HiCap) in untreated and LPS-treated THP1 cells, and the associated transcription factors were analyzed. Differential expression analysis identified SPP1 (osteopontin) as a key gene in the signature of fibrotic EndMT in patients with degenerative AscAA. The aortic intima-media expression of SPP1 correlated with the expression of inflammatory markers, the level of macrophage infiltration, and the aortic diameter. HiCap analysis, followed by transcription factor binding analysis, identified ETS1 as a potential regulator of SPP1 expression under inflammatory conditions. In conclusion, the present findings suggest that SPP1 may be involved in the development of the degenerative type of AscAA. KEY MESSAGES: In the original manuscript titled "SPP1/osteopontin, a driver of fibrosis and inflammation in degenerative ascending aortic aneurysm?" by David Freiholtz, Otto Bergman, Saliendra Pradhananga, Karin Lång, Flore-Anne Poujade, Carl Granath, Christian Olsson, Anders Franco-Cereceda, Pelin Sahlén, Per Eriksson, and Hanna M Björck, we present novel findings on regulatory factors on osteopontin (SPP1) expression in immune cells involved in degenerative ascending aortic aneurysms (AscAA). The central findings convey: SPP1 is a potential driver of the fibrotic endothelial-to-mesenchymal transition in AscAA. SPP1/osteopontin expression in AscAA is predominately by immune cells. ETS1 is a regulatory transcription factor of SPP1 expression in AscAA immune cells.
Collapse
Affiliation(s)
- David Freiholtz
- Section of Cardiothoracic Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Division of Cardiovascular Medicine, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Karolinska University Hospital, Solna, Sweden
| | - Otto Bergman
- Division of Cardiovascular Medicine, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Karolinska University Hospital, Solna, Sweden
| | - Sailendra Pradhananga
- KTH Royal Institute of Technology, School of Chemistry, Biotechnology and Health, Science for Life Laboratory, Tomtebodavägen 23A, 171 65, Stockholm, Sweden
| | - Karin Lång
- Division of Cardiovascular Medicine, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Karolinska University Hospital, Solna, Sweden
| | - Flore-Anne Poujade
- Division of Cardiovascular Medicine, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Karolinska University Hospital, Solna, Sweden
| | - Carl Granath
- Section of Cardiothoracic Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Division of Cardiovascular Medicine, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Karolinska University Hospital, Solna, Sweden
| | - Christian Olsson
- Section of Cardiothoracic Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Anders Franco-Cereceda
- Section of Cardiothoracic Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Pelin Sahlén
- KTH Royal Institute of Technology, School of Chemistry, Biotechnology and Health, Science for Life Laboratory, Tomtebodavägen 23A, 171 65, Stockholm, Sweden
| | - Per Eriksson
- Division of Cardiovascular Medicine, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Karolinska University Hospital, Solna, Sweden
| | - Hanna M Björck
- Division of Cardiovascular Medicine, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Karolinska University Hospital, Solna, Sweden.
| |
Collapse
|
5
|
Khan D, Cornelius JF, Muhammad S. The Role of NF-κB in Intracranial Aneurysm Pathogenesis: A Systematic Review. Int J Mol Sci 2023; 24:14218. [PMID: 37762520 PMCID: PMC10531594 DOI: 10.3390/ijms241814218] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/12/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Intracranial aneurysms (IAs) are abnormal dilations of the cerebral vessels, which pose a persistent threat of cerebral hemorrhage. Inflammation is known to contribute to IA development. The nuclear factor "kappa-light-chain-enhancer" of activated B-cells (NF-κB) is the major driver of inflammation. It increases the expression of inflammatory markers and matrix metalloproteinases (MMPs), which contribute heavily to the pathogenesis of IAs. NF-κB activation has been linked to IA rupture and resulting subarachnoid hemorrhage. Moreover, NF-κB activation can result in endothelial dysfunction, smooth muscle cell phenotypic switching, and infiltration of inflammatory cells in the arterial wall, which subsequently leads to the initiation and progression of IAs and consequently results in rupture. After a systematic search, abstract screening, and full-text screening, 30 research articles were included in the review. In this systematic review, we summarized the scientific literature reporting findings on NF-κB's role in the pathogenesis of IAs. In conclusion, the activation of the NF-κB pathway was associated with IA formation, progression, and rupture.
Collapse
Affiliation(s)
- Dilaware Khan
- Department of Neurosurgery, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany; (D.K.)
| | - Jan Frederick Cornelius
- Department of Neurosurgery, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany; (D.K.)
| | - Sajjad Muhammad
- Department of Neurosurgery, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany; (D.K.)
- Department of Neurosurgery, University Hospital Helsinki, Topeliuksenkatu 5, 00260 Helsinki, Finland
- Department of Neurosurgery, King Edward Medical University, Lahore 54000, Pakistan
| |
Collapse
|
6
|
Vu HD, Huynh PT, Ryu J, Kang UR, Youn SW, Kim H, Ahn HJ, Park K, Hwang SK, Chang YC, Lee YJ, Lee HJ, Lee J. Melittin-loaded Iron Oxide Nanoparticles Prevent Intracranial Arterial Dolichoectasia Development through Inhibition of Macrophage-mediated Inflammation. Int J Biol Sci 2021; 17:3818-3836. [PMID: 34671201 PMCID: PMC8495379 DOI: 10.7150/ijbs.60588] [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: 03/08/2021] [Accepted: 08/22/2021] [Indexed: 12/24/2022] Open
Abstract
Rationale: In intracranial arterial dolichoectasia (IADE) development, the feedback loop between inflammatory cytokines and macrophages involves TNF-α and NF-κB signaling pathways and leads to subsequent MMP-9 activation and extracellular matrix (ECM) degeneration. In this proof-of-concept study, melittin-loaded L-arginine-coated iron oxide nanoparticle (MeLioN) was proposed as the protective measure of IADE formation for this macrophage-mediated inflammation and ECM degeneration. Methods: IADE was created in 8-week-old C57BL/6J male mice by inducing hypertension and elastase injection into a basal cistern. Melittin was loaded on the surface of ION as a core-shell structure (hydrodynamic size, 202.4 nm; polydispersity index, 0.158). Treatment of MeLioN (2.5 mg/kg, five doses) started after the IADE induction, and the brain was harvested in the third week. In the healthy control, disease control, and MeLioN-treated group, the morphologic changes of the cerebral arterial wall were measured by diameter, thickness, and ECM composition. The expression level of MMP-9, CD68, MCP-1, TNF-α, and NF-κB was assessed from immunohistochemistry, polymerase chain reaction, and Western blot assay. Results: MeLioN prevented morphologic changes of cerebral arterial wall related to IADE formation by restoring ECM alterations and suppressing MMP-9 expression. MeLioN inhibited MCP-1 expression and reduced CD68-positive macrophage recruitments into cerebral arterial walls. MeLioN blocked TNF-α activation and NF-κB signaling pathway. In the Sylvian cistern, co-localization was found between the CD68-positive macrophage infiltrations and the MeLioN distributions detected on Prussian Blue and T2* gradient-echo MRI, suggesting the role of macrophage harboring MeLioN. Conclusions: The macrophage infiltration into the arterial wall plays a critical role in the MMP-9 secretion. MeLioN, designed for ION-mediated melittin delivery, effectively prevents IADE formation by suppressing macrophage-mediated inflammations and MMP activity. MeLioN can be a promising strategy preventing IADE development in high-risk populations.
Collapse
Affiliation(s)
- Huy Duc Vu
- Department of Radiology, Daegu Catholic University School of Medicine, Daegu, Korea
| | - Phuong Tu Huynh
- Department of Radiology, Daegu Catholic University School of Medicine, Daegu, Korea
| | - Junghwa Ryu
- Department of Radiology, Daegu Catholic University School of Medicine, Daegu, Korea
| | - Ung Rae Kang
- Department of Radiology, Daegu Catholic University School of Medicine, Daegu, Korea
| | - Sung Won Youn
- Department of Radiology, Daegu Catholic University School of Medicine, Daegu, Korea
| | - Hongtae Kim
- Department of Anatomy, Daegu Catholic University School of Medicine, Daegu, Korea
| | - Hyun Jin Ahn
- Department of Pathology, Daegu Catholic University School of Medicine, Daegu, Korea
| | - Kwankyu Park
- Department of Pathology, Daegu Catholic University School of Medicine, Daegu, Korea
| | - Soon-Kyung Hwang
- Department of Molecular Biology, Daegu Catholic University School of Medicine, Daegu, Korea
| | - Young-Chae Chang
- Department of Molecular Biology, Daegu Catholic University School of Medicine, Daegu, Korea
| | - Yong Jig Lee
- Department of Plastic Surgery, Daegu Catholic University School of Medicine, Daegu, Korea
| | - Hui Joong Lee
- Department of Radiology, Kyungpook National University School of Medicine, Daegu, Korea
| | - Jongmin Lee
- Department of Radiology, Kyungpook National University School of Medicine, Daegu, Korea
| |
Collapse
|
7
|
Wang RK, Sun YY, Li GY, Yang HT, Liu XJ, Li KF, Zhu X, Yu GY. MicroRNA-124-5p delays the progression of cerebral aneurysm by regulating FoxO1. Exp Ther Med 2021; 22:1172. [PMID: 34504617 PMCID: PMC8393823 DOI: 10.3892/etm.2021.10606] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 03/17/2021] [Indexed: 12/17/2022] Open
Abstract
Cerebral aneurysm (CA) is a common brain disease, and the development of cerebral aneurysm is driven by inflammation and hemodynamic stress. MicroRNA (miR)-124-5p is reported to be associated with inflammatory response in brain disease such as cerebral ischemia-reperfusion injury. However, the function and molecular mechanism of miR-124-5p in CA are not clear, thus, the effects of miR-124-5p on inflammatory response in CA were explored. Firstly, the expression of miR-124-5p in the peripheral blood of patients with CA and the control group was detected by reverse transcription-quantitative PCR. Then, the human umbilical vein endothelial cells (HUVECs) were used as an in vitro model system and stimulated with interleukin (IL)-1β to simulate the inflammatory environment of CA, and the expression of miR-124-5p was detected. Next, the effect of miR-124-5p on the migration and invasion of HUVECs was detected using Transwell assays. Meanwhile, the function of miR-124-5p on various inflammatory factors was determined by western blotting and enzyme-linked immunosorbent assay (ELISA). Next, the TargetScan website was used to predict FoxO1 as a target gene of miR-124-5p, and this target association was validated by double luciferase reporter assay and western blotting. Finally, the interaction of miR-124-5p with FoxO1 in CA was measured by Transwell western blotting and ELISA assays. The results showed that the expression level of miR-124-5p in the peripheral blood of patients with CA was lower compared with that of control group, and the miR-124-5p in HUVECs stimulated by IL-1β was less compared with that in normal HUVECs. Besides, miR-124-5p could inhibit the migration and invasion abilities of HUVECs and the release of inflammatory factors. Additionally, the overexpression of miR-124-5p was able to inhibit the expression of FoxO1. miR-124-5p-inhibitor promoted the migration and invasion of HUVECs, as well as inflammatory response, which was weakened following the introduction of FoxO1 small interfering RNA. Overall, the present study demonstrated that miR-124-5p could prevent the occurrence and development of cerebral aneurysm by downregulating the expression of FoxO1.
Collapse
Affiliation(s)
- Ru-Ke Wang
- Section 2, Department of Neurosurgery, Handan Central Hospital, Handan, Hebei 056001, P.R. China
| | - Yuan-Yuan Sun
- CT Room, Handan First Hospital, Handan, Hebei 056002, P.R. China
| | - Guang-You Li
- Section 2, Department of Neurosurgery, Handan Central Hospital, Handan, Hebei 056001, P.R. China
| | - Hua-Tang Yang
- Section 2, Department of Neurosurgery, Handan Central Hospital, Handan, Hebei 056001, P.R. China
| | - Xiu-Jie Liu
- Section 2, Department of Neurosurgery, Handan Central Hospital, Handan, Hebei 056001, P.R. China
| | - Ke-Feng Li
- Section 2, Department of Neurosurgery, Handan Central Hospital, Handan, Hebei 056001, P.R. China
| | - Xu Zhu
- Section 2, Department of Neurosurgery, Handan Central Hospital, Handan, Hebei 056001, P.R. China
| | - Guo-Yuan Yu
- Section 2, Department of Neurosurgery, Handan Central Hospital, Handan, Hebei 056001, P.R. China
| |
Collapse
|
8
|
Mahjoubin-Tehran M, Atkin SL, Bezsonov EE, Jamialahmadi T, Sahebkar A. Harnessing the Therapeutic Potential of Decoys in Non-Atherosclerotic Cardiovascular Diseases: State of the Art. J Cardiovasc Dev Dis 2021; 8:jcdd8090103. [PMID: 34564121 PMCID: PMC8467637 DOI: 10.3390/jcdd8090103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/06/2021] [Accepted: 08/20/2021] [Indexed: 01/30/2023] Open
Abstract
Cardiovascular disease (CVD) is the main cause of global death, highlighting the fact that conventional therapeutic approaches for the treatment of CVD patients are insufficient, and there is a need to develop new therapeutic approaches. In recent years, decoy technology, decoy oligodeoxynucleotides (ODN), and decoy peptides show promising results for the future treatment of CVDs. Decoy ODN inhibits transcription by binding to the transcriptional factor, while decoy peptide neutralizes receptors by binding to the ligands. This review focused on studies that have investigated the effects of decoy ODN and decoy peptides on non-atherosclerotic CVD.
Collapse
Affiliation(s)
- Maryam Mahjoubin-Tehran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran;
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Evgeny E. Bezsonov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, 125315 Moscow, Russia;
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Institute of Human Morphology, Moscow, Russia
- Department of Biology and General Genetics, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Tannaz Jamialahmadi
- Department of Food Science and Technology, Quchan Branch, Islamic Azad University, Quchan, Iran;
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - 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
- School of Medicine, The University of Western Australia, Perth, Australia
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
- Correspondence: or
| |
Collapse
|
9
|
Endogenous animal models of intracranial aneurysm development: a review. Neurosurg Rev 2021; 44:2545-2570. [PMID: 33501561 DOI: 10.1007/s10143-021-01481-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/05/2021] [Accepted: 01/18/2021] [Indexed: 12/13/2022]
Abstract
The pathogenesis and natural history of intracranial aneurysm (IA) remains poorly understood. To this end, animal models with induced cerebral vessel lesions mimicking human aneurysms have provided the ability to greatly expand our understanding. In this review, we comprehensively searched the published literature to identify studies that endogenously induced IA formation in animals. Studies that constructed aneurysms (i.e., by surgically creating a sac) were excluded. From the eligible studies, we reported information including the animal species, method for aneurysm induction, aneurysm definitions, evaluation methods, aneurysm characteristics, formation rate, rupture rate, and time course. Between 1960 and 2019, 174 articles reported endogenous animal models of IA. The majority used flow modification, hypertension, and vessel wall weakening (i.e., elastase treatment) to induce IAs, primarily in rats and mice. Most studies utilized subjective or qualitative descriptions to define experimental aneurysms and histology to study them. In general, experimental IAs resembled the pathobiology of the human disease in terms of internal elastic lamina loss, medial layer degradation, and inflammatory cell infiltration. After the early 2000s, many endogenous animal models of IA began to incorporate state-of-the-art technology, such as gene expression profiling and 9.4-T magnetic resonance imaging (MRI) in vivo imaging, to quantitatively analyze the biological mechanisms of IA. Future studies aimed at longitudinally assessing IA pathobiology in models that incorporate aneurysm growth will likely have the largest impact on our understanding of the disease. We believe this will be aided by high-resolution, small animal, survival imaging, in situ live-cell imaging, and next-generation omics technology.
Collapse
|
10
|
Gao Y, Zhao C, Wang J, Li H, Yang B. The potential biomarkers for the formation and development of intracranial aneurysm. J Clin Neurosci 2020; 81:270-278. [PMID: 33222929 DOI: 10.1016/j.jocn.2020.09.072] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 09/28/2020] [Indexed: 01/18/2023]
Abstract
This study is aimed to understand the pathogenesis of intracranial aneurysm (IA), which has a risk of rupture and is the primary cause of subarachnoid hemorrhage. From Gene Expression Omnibus (GEO) database, GSE75436 was extracted (15 IA tissues and 15 superficial temporal artery tissues). The differentially expressed genes (DEGs) was conducted through limma package, which followed by the enrichment analysis. Combining STRING database, protein-protein interaction (PPI) network was constructed. The modules in PPI network were performed utilizing molecular complex detection (MCODE) algorithm. With Cytoscape software, the transcription factor-miRNA-target regulatory network was constructed. Finally, microarray dataset GSE54083 was downloaded (13 IA tissues and 10 superficial temporal artery tissues) for the verification test. A total of 1332 DEGs were screened in IA tissues compared with superficial temporal artery tissues. Besides, the up-regulated TNF, IL10, IL1B, and CTSS, as well as down-regulated IL6 were included in the top 20 nodes in the PPI networks. Furthermore, in the module A of up-regulated PPI network, TNF, IL10, IL1B, and VCAM1 were interact with each other. In the regulatory network, miR-29A/B/C targeted up-regulated genes. Besides, VCAM1 was implicated in the pathway of leukocyte transendothelial migration. In the verification analysis, between GSE75436 and GSE54083, there were 444 up-regulated and 543 down-regulated co-existence DEGs and 11 co-existence genes involved the Leukocyte transendothelial migration pathway. VCAM1, TNF, CTSS, IL10, IL1B, IL6, and miR-29A/B/C might be the potential biomarkers for the formation and development of IA.
Collapse
Affiliation(s)
- Yuyuan Gao
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou 450052, Henan, China; The Academy of Medical Sciences, Zhengzhou University, No. 40 University North Road, Erqi District, Zhengzhou 450052, Henan, China
| | - Chengbin Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou 450052, Henan, China
| | - Jing Wang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou 450052, Henan, China; The Academy of Medical Sciences, Zhengzhou University, No. 40 University North Road, Erqi District, Zhengzhou 450052, Henan, China
| | - Hongwei Li
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou 450052, Henan, China.
| | - Bo Yang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou 450052, Henan, China.
| |
Collapse
|
11
|
Exosomal microRNA-23b-3p from bone marrow mesenchymal stem cells maintains T helper/Treg balance by downregulating the PI3k/Akt/NF-κB signaling pathway in intracranial aneurysm. Brain Res Bull 2020; 165:305-315. [PMID: 32956770 DOI: 10.1016/j.brainresbull.2020.09.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 09/01/2020] [Accepted: 09/06/2020] [Indexed: 01/07/2023]
Abstract
Bone marrow mesenchymal stem cells (BMSCs) are involved in cancer initiation and metastasis, and sometimes mediate cell communication by releasing exosomes and delivering microRNAs (miRNAs). The study aims to investigate the effects of exosomal hsa-miR-23b-3p derived from human BMSCs on intracranial aneurysm (IA). Firstly, human BMSCs-derived exosomes were extracted by ultra-high speed centrifugation. After clinical specimen collection, imbalance of T helper (Th) 17/Treg was found in patients with IA. Then, basilar artery aneurysm models were established and BMSCs-derived exosomes were isolated and identified. The results showed that BMSCs-derived exosomes improved pathological remodeling of IA wall, upregulated the contractile phenotype and inhibited the secretory phenotype of smooth muscle cells and reduced the number of Th17 cells to maintain the balance of Th17/Treg. In addition, human BMSCs-derived exosomes inhibited the activation of the phosphatidylinositol-3 kinase (PI3K)/protein kinase B (Akt)/nuclear factor-kappa B (NF-κB) signaling pathway and maintained Th17/Treg balance, which in turn interfered with aneurysm formation. Finally, the targeting relationship between hsa-miR-23b-3p and KLF5 was confirmed. We further noted that BMSCs-derived exosomal hsa-miR-23b-3p inhibited IA formation by targeting KLF5 through suppression of the PI3k/Akt/NF-κB signaling pathway. All in all, our study concluded that BMSCs-derived exosomal hsa-miR-23b-3p could maintain Th17/Treg balance by targeting KLF5 through suppression of the PI3k/Akt/NF-κB signaling pathway, thus inhibit IA formation.
Collapse
|
12
|
Miyata T, Minami M, Kataoka H, Hayashi K, Ikedo T, Yang T, Yamamoto Y, Yokode M, Miyamoto S. Osteoprotegerin Prevents Intracranial Aneurysm Progression by Promoting Collagen Biosynthesis and Vascular Smooth Muscle Cell Proliferation. J Am Heart Assoc 2020; 9:e015731. [PMID: 32856519 PMCID: PMC7660769 DOI: 10.1161/jaha.119.015731] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background Decreased extracellular matrix formation and few vascular smooth muscle cells (VSMCs) in cerebral vascular walls are the main characteristics of intracranial aneurysm (IA) pathogenesis. Recently, osteoprotegerin was reported to activate collagen biosynthesis and VSMC proliferation via the TGF-β1 (transforming growth factor-β1) signaling. This study aimed to investigate whether osteoprotegerin can prevent IA progression in rats through enhanced collagen expression and VSMC proliferation. Methods and Results IAs were surgically induced in 7-week-old male Sprague-Dawley rats; at 1-week post-operation, recombinant mouse osteoprotegerin or vehicle control was continuously infused for 4 weeks into the lateral ventricle using an osmotic pump. In the osteoprotegerin-treatment group, the aneurysmal size was significantly smaller (37.5 μm versus 60.0 μm; P<0.01) and the media of IA walls was thicker (57.1% versus 36.0%; P<0.01) than in the vehicle-control group. Type-I and type-III collagen, TGF-β1, phosphorylated Smad2/3, and proliferating cell nuclear antigen were significantly upregulated in the IA walls of the osteoprotegerin group than that in the control group. No significant difference was found in the expression of proinflammatory genes between the groups. In mouse VSMC cultures, osteoprotegerin treatment upregulated the expression of collagen and TGF-β1 genes, and activated VSMC proliferation; the inhibition of TGF-β1 signaling nullified this effect. Conclusions Osteoprotegerin suppressed the IA progression by a unique mechanism whereby collagen biosynthesis and VSMC proliferation were activated via TGF-β1 without altering proinflammatory gene expression. Osteoprotegerin may represent a novel therapeutic target for IAs.
Collapse
Affiliation(s)
- Takeshi Miyata
- Department of Neurosurgery Kyoto University Graduate School of Medicine Kyoto Japan.,Department of Clinical Innovative Medicine Kyoto University Graduate School of Medicine Kyoto Japan
| | - Manabu Minami
- Department of Clinical Innovative Medicine Kyoto University Graduate School of Medicine Kyoto Japan
| | - Hiroharu Kataoka
- Department of Neurosurgery Kyoto University Graduate School of Medicine Kyoto Japan
| | - Kosuke Hayashi
- Department of Neurosurgery Kyoto University Graduate School of Medicine Kyoto Japan.,Department of Clinical Innovative Medicine Kyoto University Graduate School of Medicine Kyoto Japan
| | - Taichi Ikedo
- Department of Neurosurgery Kyoto University Graduate School of Medicine Kyoto Japan
| | - Tao Yang
- Department of Neurosurgery Kyoto University Graduate School of Medicine Kyoto Japan.,Department of Clinical Innovative Medicine Kyoto University Graduate School of Medicine Kyoto Japan
| | - Yu Yamamoto
- Department of Neurosurgery Kyoto University Graduate School of Medicine Kyoto Japan.,Department of Clinical Innovative Medicine Kyoto University Graduate School of Medicine Kyoto Japan
| | - Masayuki Yokode
- Department of Clinical Innovative Medicine Kyoto University Graduate School of Medicine Kyoto Japan
| | - Susumu Miyamoto
- Department of Neurosurgery Kyoto University Graduate School of Medicine Kyoto Japan
| |
Collapse
|
13
|
Preclinical Intracranial Aneurysm Models: A Systematic Review. Brain Sci 2020; 10:brainsci10030134. [PMID: 32120907 PMCID: PMC7139747 DOI: 10.3390/brainsci10030134] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/21/2020] [Accepted: 02/23/2020] [Indexed: 12/30/2022] Open
Abstract
Intracranial aneurysms (IA) are characterized by weakened cerebral vessel walls that may lead to rupture and subarachnoid hemorrhage. The mechanisms behind their formation and progression are yet unclear and warrant preclinical studies. This systematic review aims to provide a comprehensive, systematic overview of available animal models for the study of IA pathobiology. We conducted a systematic literature search using the PubMed database to identify preclinical studies employing IA animal models. Suitable articles were selected based on predefined eligibility criteria following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Included studies were reviewed and categorized according to the experimental animal and aneurysm model. Of 4266 returned results, 3930 articles were excluded based on the title and/or abstract and further articles after screening the full text, leaving 123 studies for detailed analysis. A total of 20 different models were found in rats (nine), mice (five), rabbits (four), and dogs (two). Rat models constituted the most frequently employed intracranial experimental aneurysm model (79 studies), followed by mice (31 studies), rabbits (12 studies), and two studies in dogs. The most common techniques to induce cerebral aneurysms were surgical ligation of the common carotid artery with subsequent induction of hypertension by ligation of the renal arteries, followed by elastase-induced creation of IAs in combination with corticosterone- or angiotensin-induced hypertension. This review provides a comprehensive summary of the multitude of available IA models to study various aspects of aneurysm formation, growth, and rupture. It will serve as a useful reference for researchers by facilitating the selection of the most appropriate model and technique to answer their scientific question.
Collapse
|
14
|
Che J. Molecular mechanisms of the intracranial aneurysms and their association with the long noncoding ribonucleic acid ANRIL - A review of literature. Neurol India 2019; 65:718-728. [PMID: 28681739 DOI: 10.4103/neuroindia.ni_1074_15] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Long noncoding ribonucleic acids (RNAs) are important regulators of gene expression. Antisense noncoding RNA in the INK4 locus (ANRIL), which was coded on the Chr9p21.3 loci, participates in the pathogenesis of tumor, coronary artery disease, type 2 diabetes mellitus, and other diseases. A genome-wide association study indicated ANRIL to be a candidate gene that may lead to the development of an intracranial aneurysm (IA) formation. However, the detailed molecular mechanisms are unknown and have not been studied. Through reviewing the molecular mechanisms responsible for the development of IA and the regulation pathway of ANRIL, this paper presents four possible molecular mechanisms that may be responsible for the influence of ANRIL on the development of IAs, that is, cell cycling, Krüppel-like factor 2 (KLF2), caspase recruitment domain family member 8, and retinoid metabolism. ANRIL may become a molecular marker or therapeutic target of IA in the future. To the best of our knowledge, this is the first paper elucidating the molecular linkage between ANRIL and IAs.
Collapse
Affiliation(s)
- Jiang Che
- Department of Neurosurgery, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, Guangdong Province, China
| |
Collapse
|
15
|
WANG Y, JIN J. [Roles of macrophages in formation and progression of intracranial aneurysms]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2019; 48:204-213. [PMID: 31309760 PMCID: PMC8800668 DOI: 10.3785/j.issn.1008-9292.2019.04.13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Studies have shown that chronic inflammatory response plays a key role in intracranial aneurysms (IA) formation and progression, and macrophages regulate the formation and progression of IA through a variety of pathways. Bone marrow monocyte-derived macrophages and resident-tissue macrophages infiltrate the vessel wall, after infiltration macrophages are polarized into various polarization phenotypes dominated by M1-like and M2-like cells. Polarized phenotypes of macrophages can regulate the formation and progression of intracranial aneurysms by releasing cytokines and regulating the inflammatory response of other immune cells, as well as release different cytokines to regulate the process of extracellular matrix remodeling. Some important progresses have been made in the clinical detection and treatment in targeting macrophages. This review provides a summary on the pathogenesis of IA and potential drug targets to prevent the formation and rupture of intracranial aneurysms.
Collapse
Affiliation(s)
| | - Jinghua JIN
- 金静华(1975-), 女, 博士, 副教授, 硕士生导师, 主要从事神经退行性疾病和脑血管疾病的发病机制研究, E-mail:
,
https://orcid.org/0000-0001-6086-3340
| |
Collapse
|
16
|
Lai XL, Deng ZF, Zhu XG, Chen ZH. Apc gene suppresses intracranial aneurysm formation and rupture through inhibiting the NF-κB signaling pathway mediated inflammatory response. Biosci Rep 2019; 39:BSR20181909. [PMID: 30808715 PMCID: PMC6434386 DOI: 10.1042/bsr20181909] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 02/14/2019] [Accepted: 02/19/2019] [Indexed: 01/02/2023] Open
Abstract
Background: Intracranial aneurysm (IA) is a critical acquired cerebrovascular disease that may cause subarachnoid hemorrhage, and nuclear factor-κB (NF-κB)-mediated inflammation is involved in the pathogenesis of IA. Adenomatous polyposis coli (Apc) gene is a tumor suppressor gene associated with both familial and sporadic cancer. Herein, the purpose of our study is to validate effect of Apc gene on IA formation and rupture by regulating the NF-κB signaling pathway mediated inflammatory response. Methods: We collected IA specimens (from incarceration of IA) and normal cerebral arteries (from surgery of traumatic brain injury) to examine expression of Apc and the NF-κB signaling pathway related factors (NF-κB p65 and IκBα). ELISA was used to determine levels of monocyte chemoattractant protein-1 (MCP-1), tumor necrosis factor-α (TNF-α), interleukin (IL)-1β (IL-1β), and IL-6. IA model was established in rats, and Apc-siRNA was treated to verify effect of Apc on IA formation and rupture. Next, regulation of Apc on the NF-κB signaling pathway was investigated. Results: Reduced expression of Apc and IκBα, and increased expression of NF-κB p65 were found in IA tissues. MCP-1, TNF-α, IL-1β, and IL-6 exhibited higher levels in unruptured and ruptured IA, which suggested facilitated inflammatory responses. In addition, the IA rats injected with Apc-siRNA showed further enhanced activation of NF-κB signaling pathway, and up-regulated levels of MCP-1, TNF-α, IL-1β, IL-6, MMP-2, and MMP-9 as well as extent of p65 phosphorylation in IA. Conclusion: Above all, Apc has the potential role to attenuate IA formation and rupture by inhibiting inflammatory response through repressing the activation of the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Xian-Liang Lai
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Zhi-Feng Deng
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Xin-Gen Zhu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Zhi-Hua Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| |
Collapse
|
17
|
Two Diverse Hemodynamic Forces, a Mechanical Stretch and a High Wall Shear Stress, Determine Intracranial Aneurysm Formation. Transl Stroke Res 2019; 11:80-92. [DOI: 10.1007/s12975-019-0690-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 12/20/2018] [Accepted: 01/22/2019] [Indexed: 01/18/2023]
|
18
|
Rodgers JJ, McClure R, Epis MR, Cohen RJ, Leedman PJ, Harvey JM, Thomas MA, Bentel JM. ETS1 induces transforming growth factor β signaling and promotes epithelial-to-mesenchymal transition in prostate cancer cells. J Cell Biochem 2018; 120:848-860. [PMID: 30161276 DOI: 10.1002/jcb.27446] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 07/16/2018] [Indexed: 02/01/2023]
Abstract
Expression of the transcriptional regulator, E26 transformation-specific 1 (ETS1), is elevated in human prostate cancers, and this is associated with more aggressive tumor behavior and a rapid progression to castrate-resistant disease. Multiple ETS1 isoforms with distinct biological activities have been characterized and in 44 matched nonmalignant and malignant human prostate specimens, messenger RNAs for two ETS1 isoforms, ETS1p51 and ETS1p42, were detected, with ETS1p51 levels significantly lower in prostate tumor compared to matched nonmalignant prostate tissues. In contrast, ETS1p51 protein, the only ETS1 isoform detected, was expressed at significantly higher levels in malignant prostate. Analysis of epithelial-to-mesenchymal transition (EMT)-associated genes regulated following overexpression of ETS1p51 in the LNCaP prostate cancer cell line predicted promotion of transforming growth factor β (TGFβ) signaling and of EMT. ETS1p51 overexpression upregulated cellular levels of the EMT transcriptional regulators, ZEB1 and SNAIL1, resulted in reduced expression of the mesenchymal marker vimentin with concomitantly elevated levels of claudin 1, an epithelial tight junction protein, and increased prostate cancer cell migration and invasion. ETS1p51-induced activation of the pro-EMT TGFβ signaling pathway that was predicted in polymerase chain reaction arrays was verified by demonstration of elevated SMAD2 phosphorylation following ETS1p51 overexpression. Attenuation of ETS1p51 effects on prostate cancer cell migration and invasion by inhibition of TGFβ pathway signaling indicated that ETS1p51 effects were in part mediated by induction of TGFβ signaling. Thus, overexpression of ETS1p51, the predominant ETS1 isoform expressed in prostate tumors, promotes an EMT program in prostate cancer cells in part via activation of TGFβ signaling, potentially accounting for the poor prognosis of ETS1-overexpressing prostate tumors.
Collapse
Affiliation(s)
- Jamie J Rodgers
- Anatomical Pathology, PathWest Laboratory Medicine, Royal Perth Hospital, Perth, Western Australia, Australia.,School of Pathology and Laboratory Medicine, University of Western Australia, Crawley, Western Australia, Australia
| | - Robert McClure
- Anatomical Pathology, PathWest Laboratory Medicine, Fiona Stanley Hospital, Murdoch, Western Australia, Australia
| | - Michael R Epis
- Laboratory for Cancer Medicine, Harry Perkins Institute of Medical Research, Nedlands, Western Australia, Australia
| | - Ronald J Cohen
- School of Pathology and Laboratory Medicine, University of Western Australia, Crawley, Western Australia, Australia.,Uropath, West Leederville, Western Australia, Australia
| | - Peter J Leedman
- Laboratory for Cancer Medicine, Harry Perkins Institute of Medical Research, Nedlands, Western Australia, Australia.,Centre for Medical Research and Medical School, University of Western Australia, Crawley, Western Australia, Australia
| | - Jennet M Harvey
- School of Pathology and Laboratory Medicine, University of Western Australia, Crawley, Western Australia, Australia.,Anatomical Pathology, PathWest Laboratory Medicine, QEII Medical Centre, Crawley, Western Australia, Australia
| | -
- Australian Prostate Cancer BioResource (APCB), Brisbane, Queensland, Australia
| | - Marc A Thomas
- Anatomical Pathology, PathWest Laboratory Medicine, Royal Perth Hospital, Perth, Western Australia, Australia.,Anatomical Pathology, PathWest Laboratory Medicine, QEII Medical Centre, Crawley, Western Australia, Australia
| | - Jacqueline M Bentel
- Anatomical Pathology, PathWest Laboratory Medicine, Royal Perth Hospital, Perth, Western Australia, Australia.,Anatomical Pathology, PathWest Laboratory Medicine, Fiona Stanley Hospital, Murdoch, Western Australia, Australia
| |
Collapse
|
19
|
Mu SW, Dang Y, Wang SS, Gu JJ. The role of high mobility group box 1 protein in acute cerebrovascular diseases. Biomed Rep 2018; 9:191-197. [PMID: 30271593 PMCID: PMC6158396 DOI: 10.3892/br.2018.1127] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 06/06/2018] [Indexed: 12/15/2022] Open
Abstract
The occurrence and development of acute cerebrovascular diseases involves an inflammatory response, and high mobility group box protein 1 (HMGB1) is a pro-inflammatory factor that is expressed not only in the early-injury stage of disease, but also during the post-repair process. In the initial stage of disease, HMGB1 is released into the outside of the cell to participate in the cascade amplification reaction of inflammation, causing vasospasm, destruction of the blood-brain barrier and apoptosis of nerve cells. In the recovery stage of disease, HMGB1 can promote tissue repair and remodeling, which can aid in nerve function recovery. This review summarizes the biological characteristics of HMGB1, and the role of HMGB1 in ischemic and hemorrhagic cerebrovascular disease, and cerebral venous thrombosis.
Collapse
Affiliation(s)
- Shu-Wen Mu
- Department of Neurosurgery, Dongfang Affiliated Hospital of Xiamen University, Xiamen University Medical College, Fuzhou, Fujian 350025, P.R. China
| | - Yuan Dang
- Department of Comparative Medicine, Dongfang Affiliated Hospital of Xiamen University, Xiamen University Medical College, Fuzhou, Fujian 350025, P.R. China
| | - Shou-Sen Wang
- Department of Neurosurgery, Dongfang Affiliated Hospital of Xiamen University, Xiamen University Medical College, Fuzhou, Fujian 350025, P.R. China
| | - Jian-Jun Gu
- Department of Neuro-interventional Radiology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450000, P.R. China
| |
Collapse
|
20
|
Cooke DL, Bauer D, Sun Z, Stillson C, Nelson J, Barry D, Hetts SW, Higashida RT, Dowd CF, Halbach VV, Su H, Saeed MM. Endovascular biopsy: Technical feasibility of novel endothelial cell harvesting devices assessed in a rabbit aneurysm model. Interv Neuroradiol 2018; 21:120-8. [PMID: 25934786 DOI: 10.15274/inr-2014-10103] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The lack of safe and reliable methods to sample vascular tissue in situ limits discovery of the underlying genetic and pathophysiological mechanisms of many vascular disorders, including aneurysms. We investigated the feasibility and comparable efficacy of in vivo vascular endothelial cell sampling using a spectrum of endovascular devices. Using the rabbit elastase carotid aneurysm model we evaluated the performance of existing aneurysmal coils, intracranial stents, and stent-like devices to collect vascular endothelial cells. Additionally, we modified a subset of devices to assess the effects of alterations to coil pitch, coil wire contour, and stent surface finishing. Device performance was evaluated by (1) the number of viable endothelial cells harvested, (2) the degree of vascular wall damage analyzed using digital subtraction angiography and histopathological analysis, and (3) the ease of device navigability and retrieval. Isolated cells underwent immunohistochemical analysis to confirm cell type and viability. Coil and stent specifications, technique, and endothelial cell counts were tabulated and statistical analysis performed. Using conventional detachable-type and modified aneurysm coils 11 of 14 (78.6%) harvested endothelial cells with a mean of 7.93 (±8.33) cells/coil, while 15 of 15 (100%) conventional stents, stent-like devices and modified stents harvested endothelial cells with a mean of 831.33 (±887.73) cells/device. Coil stiffness was significantly associated with endothelial cell count in univariate analysis (p = 0.044). For stents and stent-like devices univariate analysis demonstrated stent-to-aorta diameter ratios (p = 0.001), stent length (p = 0.049), and the use of a pulling retrieval technique (p = 0.019) significantly predictive of endothelial cell counts, though a multivariate model using these variables demonstrated only the stent-to-aorta diameter ratio (p = 0.029) predictive of endothelial cell counts. Modified devices did not significantly impact harvesting. The efficacy and safety of existing aneurysm coils, intracranial stents and stent-like devices in collecting viable endothelial cells was confirmed. The technique is reproducible and the quantity and quality of collected endothelial cells is adequate for targeted genetic analysis.
Collapse
Affiliation(s)
- Daniel L Cooke
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Diana Bauer
- Laboratory Animal Resource Center, University of California, San Francisco, CA, USA
| | - Zhengda Sun
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Carol Stillson
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Jeffrey Nelson
- Department of Anesthesiology and Perioperative Care, University of California, San Francisco, CA, USA
| | | | - Steven W Hetts
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Randall T Higashida
- Department of Radiology, Neurology, and Neurological Surgery, University of California, San Francisco, CA, USA
| | - Christopher F Dowd
- Department of Radiology, Neurology, and Neurological Surgery, University of California, San Francisco, CA, USA
| | - Van V Halbach
- Department of Radiology, Neurology, and Neurological Surgery, University of California, San Francisco, CA, USA
| | - Hua Su
- Department of Anesthesiology and Perioperative Care, University of California, San Francisco, CA, USA
| | - Maythem M Saeed
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| |
Collapse
|
21
|
Kim JY, An HJ, Kim WH, Gwon MG, Gu H, Park YY, Park KK. Anti-fibrotic Effects of Synthetic Oligodeoxynucleotide for TGF-β1 and Smad in an Animal Model of Liver Cirrhosis. MOLECULAR THERAPY. NUCLEIC ACIDS 2017; 8:250-263. [PMID: 28918026 PMCID: PMC5511593 DOI: 10.1016/j.omtn.2017.06.022] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 06/29/2017] [Accepted: 06/29/2017] [Indexed: 12/12/2022]
Abstract
Liver fibrosis is characterized by changes in tissue architecture and extracellular matrix composition. Liver fibrosis affects not only hepatocytes but also the non-parenchymal cells such as hepatic stellate cells (HSCs), which are essential for maintaining an intact liver structure and function. Transforming growth factor β1 (TGF-β1) is a multifunctional cytokine that induces liver fibrosis through activation of Smad signaling pathways. To improve a new therapeutic approach, synthetic TGF-β1/Smad oligodeoxynucleotide (ODN) was used to suppress both TGF-β1 expression and Smad transcription factor using a combination of antisense ODN and decoy ODN. The aims of this study are to investigate the anti-fibrotic effects of TGF-β1/Smad ODN on simultaneous suppressions of both Smad transcription factor and TGF-β1 mRNA expression in the hepatic fibrosis model in vitro and in vivo. Synthetic TGF-β1/Smad ODN effectively inhibits Smad binding activity and TGF-β1 expression. TGF-β1/Smad ODN attenuated the epithelial mesenchymal transition (EMT) and activation of HSCs in TGF-β1-induced AML12 and HSC-T6 cells. TGF-β1/Smad ODN prevented the fibrogenesis and deposition of collagen in CCl4-treated mouse model. Synthetic TGF-β1/Smad ODN demonstrates anti-fibrotic effects that are mediated by the suppression of fibrogenic protein and inflammatory cytokines. Therefore, synthetic TGF-β1/Smad ODN has substantial therapeutic feasibility for the treatment of liver fibrotic diseases.
Collapse
Affiliation(s)
- Jung-Yeon Kim
- Department of Pathology, College of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Hyun-Jin An
- Department of Pathology, College of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Woon-Hae Kim
- Department of Pathology, College of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Mi-Gyeong Gwon
- Department of Pathology, College of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Hyemin Gu
- Department of Pathology, College of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Yoon-Yub Park
- Department of Physiology, College of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Kwan-Kyu Park
- Department of Pathology, College of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea.
| |
Collapse
|
22
|
Ikedo T, Minami M, Kataoka H, Hayashi K, Nagata M, Fujikawa R, Higuchi S, Yasui M, Aoki T, Fukuda M, Yokode M, Miyamoto S. Dipeptidyl Peptidase-4 Inhibitor Anagliptin Prevents Intracranial Aneurysm Growth by Suppressing Macrophage Infiltration and Activation. J Am Heart Assoc 2017. [PMID: 28630262 PMCID: PMC5669147 DOI: 10.1161/jaha.116.004777] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background Chronic inflammation plays a key role in the pathogenesis of intracranial aneurysms (IAs). DPP‐4 (dipeptidyl peptidase‐4) inhibitors have anti‐inflammatory effects, including suppressing macrophage infiltration, in various inflammatory models. We examined whether a DPP‐4 inhibitor, anagliptin, could suppress the growth of IAs in a rodent aneurysm model. Methods and Results IAs were surgically induced in 7‐week‐old male Sprague Dawley rats, followed by oral administration of 300 mg/kg anagliptin. We measured the morphologic parameters of aneurysms over time and their local inflammatory responses. To investigate the molecular mechanisms, we used lipopolysaccharide‐treated RAW264.7 macrophages. In the anagliptin‐treated group, aneurysms were significantly smaller 2 to 4 weeks after IA induction. Anagliptin inhibited the accumulation of macrophages in IAs, reduced the expression of MCP‐1 (monocyte chemotactic protein 1), and suppressed the phosphorylation of p65. In lipopolysaccharide‐stimulated RAW264.7 cells, anagliptin treatment significantly reduced the production of tumor necrosis factor α, MCP‐1, and IL‐6 (interleukin 6) independent of GLP‐1 (glucagon‐like peptide 1), the key mediator in the antidiabetic effects of DPP‐4 inhibitors. Notably, anagliptin activated ERK5 (extracellular signal–regulated kinase 5), which mediates the anti‐inflammatory effects of statins, in RAW264.7 macrophages. Preadministration with an ERK5 inhibitor blocked the inhibitory effect of anagliptin on MCP‐1 and IL‐6 expression. Accordingly, the ERK5 inhibitor also counteracted the suppression of p65 phosphorylation in vitro. Conclusions A DPP‐4 inhibitor, anagliptin, prevents the growth of IAs via its anti‐inflammatory effects on macrophages.
Collapse
Affiliation(s)
- Taichi Ikedo
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Department of Clinical Innovative Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Manabu Minami
- Department of Clinical Innovative Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiroharu Kataoka
- Department of Neurosurgery, National Cerebral and Cardiovascular Center, Suita Osaka, Japan
| | - Kosuke Hayashi
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Department of Clinical Innovative Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Manabu Nagata
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Department of Clinical Innovative Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Risako Fujikawa
- Department of Clinical Innovative Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Sei Higuchi
- Department of Clinical Innovative Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Mika Yasui
- Department of Clinical Innovative Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tomohiro Aoki
- Center for Innovation in Immunoregulation Technology and Therapeutics, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Core Research for Evolutional Science and Technology, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Miyuki Fukuda
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Center for Innovation in Immunoregulation Technology and Therapeutics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masayuki Yokode
- Department of Clinical Innovative Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Susumu Miyamoto
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
23
|
Aoki T. [Future prospects for the development of a novel medical therapy for intracranial aneurysm]. Nihon Yakurigaku Zasshi 2016; 148:86-91. [PMID: 27478047 DOI: 10.1254/fpj.148.86] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
|
24
|
Fennell VS, Kalani MYS, Atwal G, Martirosyan NL, Spetzler RF. Biology of Saccular Cerebral Aneurysms: A Review of Current Understanding and Future Directions. Front Surg 2016; 3:43. [PMID: 27504449 PMCID: PMC4958945 DOI: 10.3389/fsurg.2016.00043] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 07/06/2016] [Indexed: 12/24/2022] Open
Abstract
Understanding the biology of intracranial aneurysms is a clinical quandary. How these aneurysms form, progress, and rupture is poorly understood. Evidence indicates that well-established risk factors play a critical role, along with immunologic factors, in their development and clinical outcomes. Much of the expanding knowledge of the inception, progression, and rupture of intracranial aneurysms implicates inflammation as a critical mediator of aneurysm pathogenesis. Thus, therapeutic targets exploiting this arm of aneurysm pathogenesis have been implemented, often with promising outcomes.
Collapse
Affiliation(s)
- Vernard S Fennell
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center , Phoenix, AZ , USA
| | - M Yashar S Kalani
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center , Phoenix, AZ , USA
| | - Gursant Atwal
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center , Phoenix, AZ , USA
| | - Nikolay L Martirosyan
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center , Phoenix, AZ , USA
| | - Robert F Spetzler
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center , Phoenix, AZ , USA
| |
Collapse
|
25
|
Upregulation of HMGB1 in wall of ruptured and unruptured human cerebral aneurysms: preliminary results. Neurol Sci 2015; 37:219-26. [PMID: 26466586 DOI: 10.1007/s10072-015-2391-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 10/01/2015] [Indexed: 02/08/2023]
Abstract
A growing body of evidence suggests that inflammation plays a crucial role in cerebral aneurysm initiation, progression, and rupture. High-mobility group box 1 (HMGB1) is a non-histone nuclear protein that can serve as an alarmin to drive the pathogenesis of inflammatory disease. The purpose of this study was to investigate the expression of HMGB1 in the wall of ruptured and unruptured human cerebral aneurysms. Human cerebral aneurysms (25 ruptured and 16 unruptured) were immunohistochemically stained for HMGB1. As controls, four specimens of the middle cerebral arteries obtained at autopsy were also immunostained. Immunofluorescence double staining was used to determine HMGB1 cellular distribution. HMGB1 was nearly undetectable in the controls. All aneurysm tissues stained positive for HMGB1 monoclonal antibody, and expression of HMGB1 was more abundant in ruptured aneurysm tissue than unruptured aneurysms (p < 0.05). Furthermore, the expression of HMGB1 had no correlation with aneurysm size and time resected after the rupture. HMGB1 nuclear immunoreactivity was co-localized with immunoreactivity of CD3 in T lymphocytes, CD20 in B lymphocytes, CD68 in macrophages, α-SMA in smooth muscle cells, and CD31 in endothelial cells. Cytoplasmic HMGB1 localization was also detected in macrophages and T lymphocytes. Taken together, HMGB1 is expressed in the wall of human cerebral aneurysms and is more abundant in ruptured aneurysms than in unruptured ones. These data indicate a possible role of HMGB1 in the pathophysiology of human cerebral aneurysms.
Collapse
|
26
|
Shimada K, Furukawa H, Wada K, Korai M, Wei Y, Tada Y, Kuwabara A, Shikata F, Kitazato KT, Nagahiro S, Lawton MT, Hashimoto T. Protective Role of Peroxisome Proliferator-Activated Receptor-γ in the Development of Intracranial Aneurysm Rupture. Stroke 2015; 46:1664-72. [PMID: 25931465 DOI: 10.1161/strokeaha.114.007722] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 04/02/2015] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND PURPOSE Inflammation is emerging as a key component of the pathophysiology of intracranial aneurysms. Peroxisome proliferator-activated receptor-γ (PPARγ) is a nuclear hormone receptor of which activation modulates various aspects of inflammation. METHODS Using a mouse model of intracranial aneurysm, we examined the potential roles of PPARγ in the development of rupture of intracranial aneurysm. RESULTS A PPARγ agonist, pioglitazone, significantly reduced the incidence of ruptured aneurysms and the rupture rate without affecting the total incidence aneurysm (unruptured aneurysms and ruptured aneurysms). PPARγ antagonist (GW9662) abolished the protective effect of pioglitazone. The protective effect of pioglitazone was absent in mice lacking macrophage PPARγ. Pioglitazone treatment reduced the mRNA levels of inflammatory cytokines (monocyte chemoattractant factor-1, interleukin-1, and interleukin-6) that are primarily produced by macrophages in the cerebral arteries. Pioglitazone treatment reduced the infiltration of M1 macrophage into the cerebral arteries and the macrophage M1/M2 ratio. Depletion of macrophages significantly reduced the rupture rate. CONCLUSIONS Our data showed that the activation of macrophage PPARγ protects against the development of aneurysmal rupture. PPARγ in inflammatory cells may be a potential therapeutic target for the prevention of aneurysmal rupture.
Collapse
Affiliation(s)
- Kenji Shimada
- From the Departments of Anesthesia and Perioperative Care (K.S., H.F., K.W., M.K., Y.W., A.K., F.S., T.H.) and Neurological Surgery (M.T.L.), University of California, San Francisco; and Department of Neurosurgery (K.S., M.K., Y.T., K.T.K., S.N.), School of Medicine, The University of Tokushima, Tokushima City, Japan
| | - Hajime Furukawa
- From the Departments of Anesthesia and Perioperative Care (K.S., H.F., K.W., M.K., Y.W., A.K., F.S., T.H.) and Neurological Surgery (M.T.L.), University of California, San Francisco; and Department of Neurosurgery (K.S., M.K., Y.T., K.T.K., S.N.), School of Medicine, The University of Tokushima, Tokushima City, Japan
| | - Kosuke Wada
- From the Departments of Anesthesia and Perioperative Care (K.S., H.F., K.W., M.K., Y.W., A.K., F.S., T.H.) and Neurological Surgery (M.T.L.), University of California, San Francisco; and Department of Neurosurgery (K.S., M.K., Y.T., K.T.K., S.N.), School of Medicine, The University of Tokushima, Tokushima City, Japan
| | - Masaaki Korai
- From the Departments of Anesthesia and Perioperative Care (K.S., H.F., K.W., M.K., Y.W., A.K., F.S., T.H.) and Neurological Surgery (M.T.L.), University of California, San Francisco; and Department of Neurosurgery (K.S., M.K., Y.T., K.T.K., S.N.), School of Medicine, The University of Tokushima, Tokushima City, Japan
| | - Yuan Wei
- From the Departments of Anesthesia and Perioperative Care (K.S., H.F., K.W., M.K., Y.W., A.K., F.S., T.H.) and Neurological Surgery (M.T.L.), University of California, San Francisco; and Department of Neurosurgery (K.S., M.K., Y.T., K.T.K., S.N.), School of Medicine, The University of Tokushima, Tokushima City, Japan
| | - Yoshiteru Tada
- From the Departments of Anesthesia and Perioperative Care (K.S., H.F., K.W., M.K., Y.W., A.K., F.S., T.H.) and Neurological Surgery (M.T.L.), University of California, San Francisco; and Department of Neurosurgery (K.S., M.K., Y.T., K.T.K., S.N.), School of Medicine, The University of Tokushima, Tokushima City, Japan
| | - Atsushi Kuwabara
- From the Departments of Anesthesia and Perioperative Care (K.S., H.F., K.W., M.K., Y.W., A.K., F.S., T.H.) and Neurological Surgery (M.T.L.), University of California, San Francisco; and Department of Neurosurgery (K.S., M.K., Y.T., K.T.K., S.N.), School of Medicine, The University of Tokushima, Tokushima City, Japan
| | - Fumiaki Shikata
- From the Departments of Anesthesia and Perioperative Care (K.S., H.F., K.W., M.K., Y.W., A.K., F.S., T.H.) and Neurological Surgery (M.T.L.), University of California, San Francisco; and Department of Neurosurgery (K.S., M.K., Y.T., K.T.K., S.N.), School of Medicine, The University of Tokushima, Tokushima City, Japan
| | - Keiko T Kitazato
- From the Departments of Anesthesia and Perioperative Care (K.S., H.F., K.W., M.K., Y.W., A.K., F.S., T.H.) and Neurological Surgery (M.T.L.), University of California, San Francisco; and Department of Neurosurgery (K.S., M.K., Y.T., K.T.K., S.N.), School of Medicine, The University of Tokushima, Tokushima City, Japan
| | - Shinji Nagahiro
- From the Departments of Anesthesia and Perioperative Care (K.S., H.F., K.W., M.K., Y.W., A.K., F.S., T.H.) and Neurological Surgery (M.T.L.), University of California, San Francisco; and Department of Neurosurgery (K.S., M.K., Y.T., K.T.K., S.N.), School of Medicine, The University of Tokushima, Tokushima City, Japan
| | - Michael T Lawton
- From the Departments of Anesthesia and Perioperative Care (K.S., H.F., K.W., M.K., Y.W., A.K., F.S., T.H.) and Neurological Surgery (M.T.L.), University of California, San Francisco; and Department of Neurosurgery (K.S., M.K., Y.T., K.T.K., S.N.), School of Medicine, The University of Tokushima, Tokushima City, Japan
| | - Tomoki Hashimoto
- From the Departments of Anesthesia and Perioperative Care (K.S., H.F., K.W., M.K., Y.W., A.K., F.S., T.H.) and Neurological Surgery (M.T.L.), University of California, San Francisco; and Department of Neurosurgery (K.S., M.K., Y.T., K.T.K., S.N.), School of Medicine, The University of Tokushima, Tokushima City, Japan.
| |
Collapse
|
27
|
Kataoka H. Molecular mechanisms of the formation and progression of intracranial aneurysms. Neurol Med Chir (Tokyo) 2015; 55:214-29. [PMID: 25761423 PMCID: PMC4533330 DOI: 10.2176/nmc.ra.2014-0337] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Until recently, only a little was understood about molecular mechanisms of the development of an intracranial aneurysm (IA). Recent advancements over the last decade in the field of genetics and molecular biology have provided us a wide variety of evidences supporting the notion that chronic inflammation is closely associated with the pathogenesis of IA development. In the field of genetics, large-scale Genome-wide association studies (GWAS) has identified some IA susceptible loci and genes related to cell cycle and endothelial function. Researches in molecular biology using human samples and animal models have revealed the common pathway of the initiation, progression, and rupture of IAs. IA formation begins with endothelial dysfunction followed by pathological remodeling with degenerative changes of vascular walls. Medical treatments inhibiting inflammatory cascades in IA development are likely to prevent IA progression and rupture. Statins and aspirin are expected to suppress IA progression by their anti-inflammatory effects. Decoy oligodeoxynucleotides (ODNs) inhibiting inflammatory transcription factors such as nuclear factor kappa-B (NF-κB) and Ets-1 are the other promising choice of the prevention of IA development. Further clarification of molecular mechanisms of the formation and progression of IAs will shed light to the pathogenesis of IA development and provide insight into novel diagnostic and therapeutic strategies for IAs.
Collapse
Affiliation(s)
- Hiroharu Kataoka
- Department of Neurosurgery, National Cerebral and Cardiovascular Center
| |
Collapse
|
28
|
Cooke DL, Bauer D, Sun Z, Stillson C, Nelson J, Barry D, Hetts SW, Higashida RT, Dowd CF, Halbach VV, Su H, Saeed MM. Endovascular biopsy: Technical feasibility of novel endothelial cell harvesting devices assessed in a rabbit aneurysm model. Interv Neuroradiol 2015. [DOI: 10.1177/inr-2014-10103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
29
|
Cooke DL, Bauer D, Sun Z, Stillson C, Nelson J, Barry D, Hetts SW, Higashida RT, Dowd CF, Halbach VV, Su H, Saeed MM. Endovascular Biopsy: Technical Feasibility of Novel Endothelial Cell Harvesting Devices Assessed in a Rabbit Aneurysm Model. Interv Neuroradiol 2015. [DOI: 10.15274/inr-2015-10103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
30
|
Metelev VG, Kubareva EA, Oretskaya TS. Regulation of activity of transcription factor NF-κB by synthetic oligonucleotides. BIOCHEMISTRY (MOSCOW) 2014; 78:867-78. [PMID: 24228874 DOI: 10.1134/s0006297913080026] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Eukaryotic dimeric nuclear factor-κB (NF-κB) is one of the main transcription factors that activate expression of genes, products of which play the key role in development of cardiovascular pathologies, carcinogenesis, and inflammatory and viral diseases. In this review, the main attention is given to modulation of the transcription factor NF-κB activity by antisense oligonucleotides and oligonucleotide decoys. Also, current concepts about interactions between NF-κB dimers and DNA and general problems that arise in experimental use of synthetic oligonucleotides in vivo are discussed.
Collapse
Affiliation(s)
- V G Metelev
- Faculty of Chemistry, Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninsky Gory 1, Moscow, 119991, Russia.
| | | | | |
Collapse
|
31
|
Hudson JS, Hoyne DS, Hasan DM. Inflammation and human cerebral aneurysms: current and future treatment prospects. FUTURE NEUROLOGY 2013; 8. [PMID: 24376373 DOI: 10.2217/fnl.13.40] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The formation of cerebral aneurysms and their rupture propensity is of immediate clinical importance. Current management includes observation with expectant management, microsurgical clipping and/or endovascular coiling. The surgical options are invasive and are not without increased risk despite the technological advances. Recent human and animal studies have shown that inflammation plays a critical role in aneurysm formation and progression to rupture. Modulating this inflammatory process may prove to be clinically significant. This review will discuss cerebral aneurysm pathogenesis with a focus on current and future research of potential use of pharmaceutical agents that attenuate inflammation in the aneurysm wall leading to decreased risk of aneurysm rupture.
Collapse
Affiliation(s)
| | - Danielle S Hoyne
- Department of Otolaryngology University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - David M Hasan
- Department of Neurosurgery, University of Iowa Hospitals & Clinics, 200 Hawkins Drive, Iowa City, IA 52240, USA
| |
Collapse
|
32
|
You WC, Wang CX, Pan YX, Zhang X, Zhou XM, Zhang XS, Shi JX, Zhou ML. Activation of nuclear factor-κB in the brain after experimental subarachnoid hemorrhage and its potential role in delayed brain injury. PLoS One 2013; 8:e60290. [PMID: 23536907 PMCID: PMC3607578 DOI: 10.1371/journal.pone.0060290] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Accepted: 02/25/2013] [Indexed: 01/20/2023] Open
Abstract
It has been reported that inflammation is involved in brain injury after subarachnoid hemorrhage (SAH). Nuclear factor-κB (NF-κB) is a key transcriptional regulator of inflammatory genes. Here, we used pyrrolidine dithiocarbamate(PDTC), an inhibitor of NF-κB, through intracisternal injection to study the role of NF-κB in delayed brain injury after SAH. A total of 55 rabbits were randomly divided into five groups: the control group; the SAH groups including Day-3, 5, and 7 SAH groups (the rabbits in these groups were sacrificed at 3, 5, 7 days after SAH, respectively); and the PDTC group (n = 11 for each group). Electrophoretic mobility shift assay (EMSA) was performed to detect NF-κB DNA-binding activity. The mRNA levels of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and intercellular adhesion molecule (ICAM)-1 were evaluated by RT-PCR analysis. Deoxyribonucleic acid fragmentation was detected by TUNEL and p65 immunoactivity was assessed by immunohistochemistry. Our results showed the activation of NF-κB after SAH, especially at day 3 and 5. The activated p65 was detected in neurons. NF-κB DNA-binding activity was suppressed by intracisternal administration of PDTC. Increased levels of the TNF-α, IL-1β, and ICAM-1 mRNA were found in the brain at day 5 after SAH, and which were suppressed in the PDTC group. The number of TUNEL-positive cells also decreased significantly in the PDTC group compared with that in the Day-5 SAH group. These results demonstrated that the activated NF-κB in neurons after SAH plays an important role in regulating the expressions of inflammatory genes in the brain, and ultimately contributes to delayed brain injury.
Collapse
Affiliation(s)
- Wan-Chun You
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Chun-xi Wang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Yun-xi Pan
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Xin Zhang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Xiao-ming Zhou
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Xiang-sheng Zhang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Ji-xin Shi
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Meng-liang Zhou
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
- * E-mail:
| |
Collapse
|
33
|
Chalouhi N, Ali MS, Jabbour PM, Tjoumakaris SI, Gonzalez LF, Rosenwasser RH, Koch WJ, Dumont AS. Biology of intracranial aneurysms: role of inflammation. J Cereb Blood Flow Metab 2012; 32:1659-76. [PMID: 22781330 PMCID: PMC3434628 DOI: 10.1038/jcbfm.2012.84] [Citation(s) in RCA: 361] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Intracranial aneurysms (IAs) linger as a potentially devastating clinical problem. Despite intense investigation, our understanding of the mechanisms leading to aneurysm development, progression and rupture remain incompletely defined. An accumulating body of evidence implicates inflammation as a critical contributor to aneurysm pathogenesis. Intracranial aneurysm formation and progression appear to result from endothelial dysfunction, a mounting inflammatory response, and vascular smooth muscle cell phenotypic modulation producing a pro-inflammatory phenotype. A later final common pathway appears to involve apoptosis of cellular constituents of the vessel wall. These changes result in degradation of the integrity of the vascular wall leading to aneurysmal dilation, progression and eventual rupture in certain aneurysms. Various aspects of the inflammatory response have been investigated as contributors to IA pathogenesis including leukocytes, complement, immunoglobulins, cytokines, and other humoral mediators. Furthermore, gene expression profiling of IA compared with control arteries has prominently featured differential expression of genes involved with immune response/inflammation. Preliminary data suggest that therapies targeting the inflammatory response may have efficacy in the future treatment of IA. Further investigation, however, is necessary to elucidate the precise role of inflammation in IA pathogenesis, which can be exploited to improve the prognosis of patients harboring IA.
Collapse
Affiliation(s)
- Nohra Chalouhi
- Joseph and Marie Field Cerebrovascular Research Laboratory, Division of Neurovascular and Endovascular Surgery, Department of Neurological Surgery, Thomas Jefferson University and Jefferson Hospital for Neuroscience, Philadelphia, Pennsylvania 19107, USA.
| | | | | | | | | | | | | | | |
Collapse
|