1
|
Sun ZH, Liu F, Kong LL, Ji PM, Huang L, Zhou HM, Sun R, Luo J, Li WZ. Interruption of TRPC6-NFATC1 signaling inhibits NADPH oxidase 4 and VSMCs phenotypic switch in intracranial aneurysm. Biomed Pharmacother 2023; 161:114480. [PMID: 37002575 DOI: 10.1016/j.biopha.2023.114480] [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: 01/06/2023] [Revised: 02/23/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
Intracranial aneurysm (IA) is a frequent cerebrovascular disorder with unclear pathogenesis. The vascular smooth muscle cells (VSMCs) phenotypic switch is essential for IA formation. It has been reported that Ca2+ overload and excessive reactive oxygen species (ROS) are involved in VSMCs phenotypic switch. The transient receptor potential canonical 6 (TRPC6) and NADPH oxidase 4 (NOX4) are the main pathway to participate in Ca2+ overload and ROS production in VSMCs. Ca2+ overload can activate calcineurin (CN), leading to nuclear factor of activated T cell (NFAT) dephosphorylation to regulate the target gene's transcription. We hypothesized that activation of TRPC6-NFATC1 signaling may upregulate NOX4 and involve in VSMCs phenotypic switch contributing to the progression of IA. Our results showed that the expressions of NOX4, p22phox, p47phox, TRPC6, CN and NFATC1 were significantly increased, and VSMCs underwent a significant phenotypic switch in IA tissue and cellular specimens. The VIVIT (NFATC1 inhibitor) and BI-749327 (TRPC6 inhibitor) treatment reduced the expressions of NOX4, p22phox and p47phox and the production of ROS, and significantly improved VSMCs phenotypic switch in IA rats and cells. Consistent results were obtained from IA Trpc6 knockout (Trpc6-/-) mice. Furthermore, the results also revealed that NFATC1 could regulate NOX4 transcription by binding to its promoter. Our findings reveal that interrupting the TRPC6-NFATC1 signaling inhibits NOX4 and improves VSMCs phenotypic switch in IA, and regulating Ca2+ homeostasis may be an important therapeutic strategy for IA.
Collapse
Affiliation(s)
- Zheng-Hao Sun
- Department of Pharmacology, Basic Medicine College; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education; Anhui Medical University, Hefei 230032, Anhui, China
| | - Fei Liu
- Department of neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, Anhui, China
| | - Liang-Liang Kong
- Department of Pharmacology, Basic Medicine College; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education; Anhui Medical University, Hefei 230032, Anhui, China
| | - Peng-Min Ji
- Department of Pharmacology, Basic Medicine College; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education; Anhui Medical University, Hefei 230032, Anhui, China
| | - Lei Huang
- Department of Pharmacology, Basic Medicine College; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education; Anhui Medical University, Hefei 230032, Anhui, China
| | - Hui-Min Zhou
- Department of Pharmacology, Basic Medicine College; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education; Anhui Medical University, Hefei 230032, Anhui, China
| | - Ran Sun
- Department of Pharmacology, Basic Medicine College; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education; Anhui Medical University, Hefei 230032, Anhui, China
| | - Jing Luo
- Department of neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, Anhui, China.
| | - Wei-Zu Li
- Department of Pharmacology, Basic Medicine College; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education; Anhui Medical University, Hefei 230032, Anhui, China.
| |
Collapse
|
2
|
Grüter BE, von Faber-Castell F, Marbacher S. Lumen-oriented versus wall-oriented treatment strategies for intracranial aneurysms - A systematic review of suggested therapeutic concepts. J Cereb Blood Flow Metab 2022; 42:1568-1578. [PMID: 34796752 PMCID: PMC9441732 DOI: 10.1177/0271678x211057498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The development of new treatment strategies for intracranial aneurysms (IAs) has been and continues to be a major interest in neurovascular research. Initial treatment concepts were mainly based on a physical-mechanistic disease understanding for IA occlusion (lumen-oriented therapies). However, a growing body of literature indicates the important role of aneurysm wall biology (wall-oriented therapies) for complete IA obliteration. This systematic literature review identified studies that explored endovascular treatment strategies for aneurysm treatment in a preclinical setting. Of 5278 publications screened, 641 studies were included, categorized, and screened for eventual translation in a clinical trial. Lumen-oriented strategies included (1) enhanced intraluminal thrombus organization, (2) enhanced intraluminal packing, (3) bridging of the intraluminal space, and (4) other, alternative concepts. Wall-oriented strategies included (1) stimulation of proliferative response, (2) prevention of aneurysm wall cell injury, (3) inhibition of inflammation and oxidative stress, and (4) inhibition of extracellular matrix degradation. Overall, lumen-oriented strategies numerically still dominate over wall-oriented strategies. Among the plethora of suggested preclinical treatment strategies, only a small minority were translated into clinically applicable concepts (36 of 400 lumen-oriented and 6 of 241 wall-oriented). This systematic review provides a comprehensive overview that may provide a starting point for the development of new treatment strategies.
Collapse
Affiliation(s)
- Basil E Grüter
- Department of Neurosurgery, 30231Kantonsspital Aarau, Aarau, Switzerland.,Cerebrovascular Research Group, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Fabio von Faber-Castell
- Cerebrovascular Research Group, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Department of Neurosurgery, University Hospital Zurich, Zurich, Switzerland
| | - Serge Marbacher
- Department of Neurosurgery, 30231Kantonsspital Aarau, Aarau, Switzerland.,Cerebrovascular Research Group, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Department of Neurosurgery, Kantonsspital Aarau, University of Bern, Switzerland
| |
Collapse
|
3
|
Edaravone Attenuated Angiotensin II-Induced Atherosclerosis and Abdominal Aortic Aneurysms in Apolipoprotein E-Deficient Mice. Biomolecules 2022; 12:biom12081117. [PMID: 36009011 PMCID: PMC9405883 DOI: 10.3390/biom12081117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 08/09/2022] [Accepted: 08/12/2022] [Indexed: 12/17/2022] Open
Abstract
Background: The aim of the study was to define whether edaravone, a free-radical scavenger, influenced angiotensin II (AngII)-induced atherosclerosis and abdominal aortic aneurysms (AAAs) formation. Methods: Male apolipoprotein E-deficient mice (8–12 weeks old) were fed with a normal diet for 5 weeks. Either edaravone (10 mg/kg/day) or vehicle was injected intraperitoneally for 5 weeks. After 1 week of injections, mice were infused subcutaneously with either AngII (1000 ng/kg/min, n = 16–17 per group) or saline (n = 5 per group) by osmotic minipumps for 4 weeks. Results: AngII increased systolic blood pressure equivalently in mice administered with either edaravone or saline. Edaravone had no effect on plasma total cholesterol concentrations and body weights. AngII infusion significantly increased ex vivo maximal diameters of abdominal aortas and en face atherosclerosis but was significantly attenuated by edaravone administration. Edaravone also reduced the incidence of AngII-induced AAAs. In addition, edaravone diminished AngII-induced aortic MMP-2 activation. Quantitative RT-PCR revealed that edaravone ameliorated mRNA abundance of aortic MCP-1 and IL-1β. Immunostaining demonstrated that edaravone attenuated oxidative stress and macrophage accumulation in the aorta. Furthermore, edaravone administration suppressed thioglycolate-induced mice peritoneal macrophages (MPMs) accumulation and mRNA abundance of MCP-1 in MPMs in male apolipoprotein E-deficient mice. In vitro, edaravone reduced LPS-induced mRNA abundance of MCP-1 in MPMs. Conclusions: Edaravone attenuated AngII-induced AAAs and atherosclerosis in male apolipoprotein E-deficient mice via anti-oxidative action and anti-inflammatory effect.
Collapse
|
4
|
Fréneau M, Baron-Menguy C, Vion AC, Loirand G. Why Are Women Predisposed to Intracranial Aneurysm? Front Cardiovasc Med 2022; 9:815668. [PMID: 35224050 PMCID: PMC8866977 DOI: 10.3389/fcvm.2022.815668] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/11/2022] [Indexed: 12/21/2022] Open
Abstract
Intracranial aneurysm (IA) is a frequent and generally asymptomatic cerebrovascular abnormality characterized as a localized dilation and wall thinning of intracranial arteries that preferentially arises at the arterial bifurcations of the circle of Willis. The devastating complication of IA is its rupture, which results in subarachnoid hemorrhage that can lead to severe disability and death. IA affects about 3% of the general population with an average age for detection of rupture around 50 years. IAs, whether ruptured or unruptured, are more common in women than in men by about 60% overall, and more especially after the menopause where the risk is double-compared to men. Although these data support a protective role of estrogen, differences in the location and number of IAs observed in women and men under the age of 50 suggest that other underlying mechanisms participate to the greater IA prevalence in women. The aim of this review is to provide a comprehensive overview of the current data from both clinical and basic research and a synthesis of the proposed mechanisms that may explain why women are more prone to develop IA.
Collapse
|
5
|
Tutino VM, Lu Y, Ishii D, Poppenberg KE, Rajabzadeh-Oghaz H, Siddiqui AH, Hasan DM. Aberrant Whole Blood Gene Expression in the Lumen of Human Intracranial Aneurysms. Diagnostics (Basel) 2021; 11:diagnostics11081442. [PMID: 34441376 PMCID: PMC8392298 DOI: 10.3390/diagnostics11081442] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 07/31/2021] [Accepted: 08/06/2021] [Indexed: 01/19/2023] Open
Abstract
The rupture of an intracranial aneurysm (IA) causes devastating hemorrhagic strokes. Yet, most IAs remain asymptomatic and undetected until they rupture. In the search for circulating biomarkers of unruptured IAs, we previously performed transcriptome profiling on whole blood and identified an IA-associated panel of 18 genes. In this study, we seek to determine if these genes are also differentially expressed within the IA lumen, which could provide a mechanistic link between the disease and the observed circulating gene expression patterns. To this end, we collected blood from the lumen of 37 IAs and their proximal parent vessels in 31 patients. The expression levels of 18 genes in the lumen and proximal vessel were then measured by quantitative polymerase chain reaction. This analysis revealed that the expression of 6/18 genes (CBWD6, MT2A, MZT2B, PIM3, SLC37A3, and TNFRSF4) was significantly higher in intraluminal blood, while the expression of 3/18 genes (ST6GALNAC1, TCN2, and UFSP1) was significantly lower. There was a significant, positive correlation between intraluminal and proximal expression of CXCL10, MT2A, and MZT2B, suggesting local increases of these genes is reflected in the periphery. Expression of ST6GALNAC1 and TIFAB was significantly positively correlated with IA size, while expression of CCDC85B was significantly positively correlated with IA enhancement on post-contrast MRI, a metric of IA instability and risk. In conclusion, intraluminal expression differences in half of the IA-associated genes observed in this study provide evidence for IA tissue-mediated transcriptional changes in whole blood. Additionally, some genes may be informative in assessing IA risk, as their intraluminal expression was correlated to IA size and aneurysmal wall enhancement.
Collapse
Affiliation(s)
- Vincent M. Tutino
- Canon Stroke and Vascular Research Center, University at Buffalo, Buffalo, NY 14260, USA; (V.M.T.); (K.E.P.); (H.R.-O.); (A.H.S.)
- Department of Pathology and Anatomical Sciences, University at Buffalo, Buffalo, NY 14260, USA
- Department of Neurosurgery, University at Buffalo, Buffalo, NY 14260, USA
| | - Yongjun Lu
- Department of Cardiovascular Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA;
| | - Daizo Ishii
- Department of Neurosurgery, University of Iowa Hospitals and Clinics, 1616 JCP, 200 Hawkins Dr, Iowa City, IA 52242, USA;
| | - Kerry E. Poppenberg
- Canon Stroke and Vascular Research Center, University at Buffalo, Buffalo, NY 14260, USA; (V.M.T.); (K.E.P.); (H.R.-O.); (A.H.S.)
- Department of Neurosurgery, University at Buffalo, Buffalo, NY 14260, USA
| | - Hamidreza Rajabzadeh-Oghaz
- Canon Stroke and Vascular Research Center, University at Buffalo, Buffalo, NY 14260, USA; (V.M.T.); (K.E.P.); (H.R.-O.); (A.H.S.)
- Department of Neurosurgery, University at Buffalo, Buffalo, NY 14260, USA
| | - Adnan H. Siddiqui
- Canon Stroke and Vascular Research Center, University at Buffalo, Buffalo, NY 14260, USA; (V.M.T.); (K.E.P.); (H.R.-O.); (A.H.S.)
- Department of Neurosurgery, University at Buffalo, Buffalo, NY 14260, USA
| | - David M. Hasan
- Department of Neurosurgery, University of Iowa Hospitals and Clinics, 1616 JCP, 200 Hawkins Dr, Iowa City, IA 52242, USA;
- Correspondence: ; Tel.: +1-319-384-8669
| |
Collapse
|
6
|
Giotta Lucifero A, Baldoncini M, Bruno N, Galzio R, Hernesniemi J, Luzzi S. Shedding the Light on the Natural History of Intracranial Aneurysms: An Updated Overview. ACTA ACUST UNITED AC 2021; 57:medicina57080742. [PMID: 34440948 PMCID: PMC8400479 DOI: 10.3390/medicina57080742] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 07/19/2021] [Accepted: 07/20/2021] [Indexed: 11/16/2022]
Abstract
The exact molecular pathways underlying the multifactorial natural history of intracranial aneurysms (IAs) are still largely unknown, to the point that their understanding represents an imperative challenge in neurovascular research. Wall shear stress (WSS) promotes the genesis of IAs through an endothelial dysfunction causing an inflammatory cascade, vessel remodeling, phenotypic switching of the smooth muscle cells, and myointimal hyperplasia. Aneurysm growth is supported by endothelial oxidative stress and inflammatory mediators, whereas low and high WSS determine the rupture in sidewall and endwall IAs, respectively. Angioarchitecture, age older than 60 years, female gender, hypertension, cigarette smoking, alcohol abuse, and hypercholesterolemia also contribute to growth and rupture. The improvements of aneurysm wall imaging techniques and the implementation of target therapies targeted against inflammatory cascade may contribute to significantly modify the natural history of IAs. This narrative review strives to summarize the recent advances in the comprehension of the mechanisms underlying the genesis, growth, and rupture of IAs.
Collapse
Affiliation(s)
- Alice Giotta Lucifero
- Neurosurgery Unit, Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy;
| | - Matías Baldoncini
- Department of Neurological Surgery, Hospital San Fernando, Buenos Aires 1646, Argentina;
| | - Nunzio Bruno
- Division of Neurosurgery, Azienda Ospedaliero Universitaria Consorziale Policlinico di Bari, 70124 Bari, Italy;
| | - Renato Galzio
- Neurosurgery Unit, Maria Cecilia Hospital, 48032 Cotignola, Italy;
| | - Juha Hernesniemi
- Juha Hernesniemi International Center for Neurosurgery, Henan Provincial People’s Hospital, Zhengzhou 450000, China;
| | - Sabino Luzzi
- Neurosurgery Unit, Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy;
- Neurosurgery Unit, Department of Surgical Sciences, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
- Correspondence:
| |
Collapse
|
7
|
Liu K, Sun J, Shao L, He H, Liu Q, Li Y, Ge H. Correlation of periodontal diseases with intracranial aneurysm formation: novel predictive indicators. Chin Neurosurg J 2021; 7:31. [PMID: 34092261 PMCID: PMC8182916 DOI: 10.1186/s41016-021-00249-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 05/13/2021] [Indexed: 12/02/2022] Open
Abstract
Background We investigated whether periodontal diseases, specifically, periodontitis and gingivitis, could be risk factors of the incidence of intracranial aneurysms (IAs). Methods We performed a case–control study to compare the differences in the periodontal disease parameters of 281 cases that were divided into the IAs group and non-IAs group. All cases underwent complete radiographic examination for IAs and examination for periodontal health. Results Comparing with those in the non-IAs group, the cases in the IAs group were older (53.95 ± 8.56 vs 47.79 ± 12.33, p < 0.001) and had a higher incidence of hypertension (76 vs 34, p = 0.006). Univariate logistic regression analysis revealed that age (> 50 years) and hypertension were predictive risk factors of aneurysm formation (odds ratio [OR] 1.047, 95% confidence interval [95% CI] 1.022–1.073, p < 0.001 and OR 2.047, 95% CI 1.232–3.401, p = 0.006). In addition, univariate and multivariate logistic regression analyses showed that the parameters of periodontal diseases, including gingival index, plaque index, clinical attachment loss, and alveolar bone loss, were significantly associated with the occurrence of IAs (all p < 0.05). For further statistical investigation, the parameters of periodontal diseases were divided into four layers based on the quartered data. Poorer periodontal health condition (especially gingival index > 1.1 and plaque index > 1.5) had the correlation with IAs formation (p = 0.007 and p < 0.001). Conclusion Severe gingivitis or periodontitis, combining with hypertension, is significantly associated with the incidence of IAs.
Collapse
Affiliation(s)
- Keyun Liu
- Department of Interventional Neuroradiology, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, No. 119, West Road of South Fourth Ring, Fengtai, Beijing, 100070, People's Republic of China
| | - Jia Sun
- Department of Stomatology, Tianjin Stomatological Hospital, Hospital of Stomatology, Nankai University, Tianjin, 300041, People's Republic of China
| | - Lingling Shao
- Department of Tuberculosis, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, People's Republic of China
| | - Hongwei He
- Department of Interventional Neuroradiology, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, No. 119, West Road of South Fourth Ring, Fengtai, Beijing, 100070, People's Republic of China
| | - Qinglin Liu
- Department of Interventional Neuroradiology, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, No. 119, West Road of South Fourth Ring, Fengtai, Beijing, 100070, People's Republic of China
| | - Youxiang Li
- Department of Interventional Neuroradiology, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, No. 119, West Road of South Fourth Ring, Fengtai, Beijing, 100070, People's Republic of China.
| | - Huijian Ge
- Department of Interventional Neuroradiology, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, No. 119, West Road of South Fourth Ring, Fengtai, Beijing, 100070, People's Republic of China.
| |
Collapse
|
8
|
Hayashi K, Kataoka H, Minami M, Ikedo T, Miyata T, Shimizu K, Nagata M, Yang T, Yamamoto Y, Yokode M, Miyamoto S. Association of zinc administration with growth suppression of intracranial aneurysms via induction of A20. J Neurosurg 2021; 134:992-998. [PMID: 32217803 DOI: 10.3171/2020.1.jns192047] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 01/20/2020] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Zinc is an essential micronutrient with multiple biological effects, including antiinflammation. Previously, the authors demonstrated that the pathogenesis of intracranial aneurysms (IAs) is strongly related to chronic inflammation. In this study, the authors investigated whether administration of zinc inhibits the growth of IAs in a rat model. METHODS The authors analyzed surgically induced IAs in Sprague-Dawley male rats, which were subsequently treated with intraperitoneal injections of zinc sulfate heptahydrate (ZnSO4; 3 mg/kg/day) or vehicle for 4 weeks. RESULTS Size and wall thickness ratios of experimentally induced IAs were assessed in both treatment groups after induction and in a control group. The effects of zinc administration in IAs were examined by immunohistochemistry and Western blotting. Zinc administration significantly suppressed aneurysm size and also preserved the internal elastic lumen. Administration of zinc significantly attenuated infiltration of macrophages into IAs. CONCLUSIONS Zinc treatment significantly increased expression of the antiinflammatory signaling protein A20, an inhibitor of the nuclear factor κB (NF-κB) pathway, in rat IAs. Zinc administration may prevent the growth of rat IAs by inducing A20-attributed inactivation of NF-κB signaling.
Collapse
Affiliation(s)
- Kosuke Hayashi
- 1Department of Neurosurgery and
- 2Department of Clinical Innovative Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | - Manabu Minami
- 2Department of Clinical Innovative Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Taichi Ikedo
- 1Department of Neurosurgery and
- 2Department of Clinical Innovative Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takeshi Miyata
- 1Department of Neurosurgery and
- 2Department of Clinical Innovative Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | - Manabu Nagata
- 1Department of Neurosurgery and
- 2Department of Clinical Innovative Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tao Yang
- 1Department of Neurosurgery and
- 2Department of Clinical Innovative Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yu Yamamoto
- 1Department of Neurosurgery and
- 2Department of Clinical Innovative Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masayuki Yokode
- 2Department of Clinical Innovative Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | |
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
|
Mota Telles JP, Rabelo NN, Junior JR, Teixeira MJ, Figueiredo EG. C-Reactive Protein Levels Are Higher in Patients With Fusiform Intracranial Aneurysms: A Case-Control Study. World Neurosurg 2020; 146:e896-e901. [PMID: 33278646 DOI: 10.1016/j.wneu.2020.11.042] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 11/06/2020] [Accepted: 11/07/2020] [Indexed: 01/17/2023]
Abstract
BACKGROUND Comprehending the risk factors that contribute to the formation of fusiform aneurysms (FAs) might provide some insight into treatment and prevention strategies. This case-control study aimed to compare the levels of serum C-reactive protein (CRP), as a biomarker, between patients with fusiform and saccular intracranial aneurysms. METHODS We retrospectively analyzed medical records from 2010 to 2019. Thirty-five patients were identified as having FAs: 13 (37.1%) were ruptured, and 22 were unruptured. An age-matched sample of 70 controls (2:1) with saccular aneurysms was obtained from the same records: 36 (51.4%) ruptured and 34 unruptured. RESULTS Patients with FAs had median CRP values of 0.61 mg/dL (IQR: 1.5), compared with 0.29 mg/dL (IQR: 0.42) in controls (P < 0.01). Within both the ruptured and the unruptured group, median CRP was higher in patients with FAs compared with controls (P < 0.01). Diabetes, smoking status, hypertension, and sex did not significantly influence CRP levels. Age-adjusted analyses showed that fusiform morphology was independently associated with higher CRP levels for unruptured aneurysms (OR 1.2, 95% CI 1.05-1.43), but not for ruptured aneurysms (OR 1.02, 95%CI 0.99-1.05). CONCLUSIONS CRP was higher in patients with FAs than controls, and it constituted an independent predictor of fusiform morphology for patients with unruptured aneurysms. Inflammation might be an especially important factor in FA formation and growth, and further studies could use this finding to design new treatment strategies.
Collapse
Affiliation(s)
- João Paulo Mota Telles
- Division of Neurosurgery, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Nicollas Nunes Rabelo
- Division of Neurosurgery, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Jefferson Rosi Junior
- Division of Neurosurgery, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Manoel Jacobsen Teixeira
- Division of Neurosurgery, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Eberval Gadelha Figueiredo
- Division of Neurosurgery, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.
| |
Collapse
|
11
|
Ma D, Zheng B, Liu HL, Zhao YB, Liu X, Zhang XH, Li Q, Shi WB, Suzuki T, Wen JK. Klf5 down-regulation induces vascular senescence through eIF5a depletion and mitochondrial fission. PLoS Biol 2020; 18:e3000808. [PMID: 32817651 PMCID: PMC7462304 DOI: 10.1371/journal.pbio.3000808] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 09/01/2020] [Accepted: 07/31/2020] [Indexed: 01/10/2023] Open
Abstract
Although dysregulation of mitochondrial dynamics has been linked to cellular senescence, which contributes to advanced age-related disorders, it is unclear how Krüppel-like factor 5 (Klf5), an essential transcriptional factor of cardiovascular remodeling, mediates the link between mitochondrial dynamics and vascular smooth muscle cell (VSMC) senescence. Here, we show that Klf5 down-regulation in VSMCs is correlated with rupture of abdominal aortic aneurysm (AAA), an age-related vascular disease. Mice lacking Klf5 in VSMCs exacerbate vascular senescence and progression of angiotensin II (Ang II)-induced AAA by facilitating reactive oxygen species (ROS) formation. Klf5 knockdown enhances, while Klf5 overexpression suppresses mitochondrial fission. Mechanistically, Klf5 activates eukaryotic translation initiation factor 5a (eIF5a) transcription through binding to the promoter of eIF5a, which in turn preserves mitochondrial integrity by interacting with mitofusin 1 (Mfn1). Accordingly, decreased expression of eIF5a elicited by Klf5 down-regulation leads to mitochondrial fission and excessive ROS production. Inhibition of mitochondrial fission decreases ROS production and VSMC senescence. Our studies provide a potential therapeutic target for age-related vascular disorders.
Collapse
Affiliation(s)
- Dong Ma
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, China
- School of Public Health, North China University of Science and Technology, Tangshan, China
| | - Bin Zheng
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, China
| | - He-liang Liu
- School of Public Health, North China University of Science and Technology, Tangshan, China
| | - Yong-bo Zhao
- Department of Cardiac surgery, the Fourth Hospital of Hebei Medical University, Shi Jiazhuang, China
| | - Xiao Liu
- Department of Cardiac surgery, the Fourth Hospital of Hebei Medical University, Shi Jiazhuang, China
| | - Xin-hua Zhang
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, China
| | - Qiang Li
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, China
| | - Wei-bo Shi
- Department of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang, China
| | - Toru Suzuki
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
| | - Jin-kun Wen
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
12
|
Texakalidis P, Sweid A, Mouchtouris N, Peterson EC, Sioka C, Rangel-Castilla L, Reavey-Cantwell J, Jabbour P. Aneurysm Formation, Growth, and Rupture: The Biology and Physics of Cerebral Aneurysms. World Neurosurg 2019; 130:277-284. [DOI: 10.1016/j.wneu.2019.07.093] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 07/08/2019] [Accepted: 07/09/2019] [Indexed: 12/18/2022]
|
13
|
Ramesh SS, Christopher R, Indira Devi B, Bhat DI. The vascular protective role of oestradiol: a focus on postmenopausal oestradiol deficiency and aneurysmal subarachnoid haemorrhage. Biol Rev Camb Philos Soc 2019; 94:1897-1917. [DOI: 10.1111/brv.12541] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 06/04/2019] [Accepted: 06/10/2019] [Indexed: 12/24/2022]
Affiliation(s)
- Shruthi S. Ramesh
- Department of NeurochemistryNational Institute of Mental Health and Neuro Sciences Bengaluru‐560029 Karnataka India
| | - Rita Christopher
- Department of NeurochemistryNational Institute of Mental Health and Neuro Sciences Bengaluru‐560029 Karnataka India
| | - Bhagavatula Indira Devi
- Department of NeurosurgeryNational Institute of Mental Health and Neuro Sciences Bengaluru‐560029 Karnataka India
| | - Dhananjaya I. Bhat
- Department of NeurosurgeryNational Institute of Mental Health and Neuro Sciences Bengaluru‐560029 Karnataka India
| |
Collapse
|
14
|
Thompson JW, Elwardany O, McCarthy DJ, Sheinberg DL, Alvarez CM, Nada A, Snelling BM, Chen SH, Sur S, Starke RM. In vivo cerebral aneurysm models. Neurosurg Focus 2019; 47:E20. [DOI: 10.3171/2019.4.focus19219] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 04/09/2019] [Indexed: 11/06/2022]
Abstract
Cerebral aneurysm rupture is a devastating event resulting in subarachnoid hemorrhage and is associated with significant morbidity and death. Up to 50% of individuals do not survive aneurysm rupture, with the majority of survivors suffering some degree of neurological deficit. Therefore, prior to aneurysm rupture, a large number of diagnosed patients are treated either microsurgically via clipping or endovascularly to prevent aneurysm filling. With the advancement of endovascular surgical techniques and devices, endovascular treatment of cerebral aneurysms is becoming the first-line therapy at many hospitals. Despite this fact, a large number of endovascularly treated patients will have aneurysm recanalization and progression and will require retreatment. The lack of approved pharmacological interventions for cerebral aneurysms and the need for retreatment have led to a growing interest in understanding the molecular, cellular, and physiological determinants of cerebral aneurysm pathogenesis, maturation, and rupture. To this end, the use of animal cerebral aneurysm models has contributed significantly to our current understanding of cerebral aneurysm biology and to the development of and training in endovascular devices. This review summarizes the small and large animal models of cerebral aneurysm that are being used to explore the pathophysiology of cerebral aneurysms, as well as the development of novel endovascular devices for aneurysm treatment.
Collapse
Affiliation(s)
- John W. Thompson
- Departments of 1Neurological Surgery and
- 3The University of Miami Cerebrovascular Initiative, University of Miami; and
| | - Omar Elwardany
- Departments of 1Neurological Surgery and
- 3The University of Miami Cerebrovascular Initiative, University of Miami; and
| | - David J. McCarthy
- Departments of 1Neurological Surgery and
- 3The University of Miami Cerebrovascular Initiative, University of Miami; and
| | - Dallas L. Sheinberg
- Departments of 1Neurological Surgery and
- 3The University of Miami Cerebrovascular Initiative, University of Miami; and
| | - Carlos M. Alvarez
- Departments of 1Neurological Surgery and
- 3The University of Miami Cerebrovascular Initiative, University of Miami; and
| | - Ahmed Nada
- Departments of 1Neurological Surgery and
- 3The University of Miami Cerebrovascular Initiative, University of Miami; and
| | - Brian M. Snelling
- Departments of 1Neurological Surgery and
- 3The University of Miami Cerebrovascular Initiative, University of Miami; and
- 4Marcus Neuroscience Institute, Boca Raton Regional Hospital, Boca Raton, Florida
| | - Stephanie H. Chen
- Departments of 1Neurological Surgery and
- 3The University of Miami Cerebrovascular Initiative, University of Miami; and
| | - Samir Sur
- Departments of 1Neurological Surgery and
- 3The University of Miami Cerebrovascular Initiative, University of Miami; and
| | - Robert M. Starke
- Departments of 1Neurological Surgery and
- 2Radiology, University of Miami
- 3The University of Miami Cerebrovascular Initiative, University of Miami; and
| |
Collapse
|
15
|
Sheinberg DL, McCarthy DJ, Elwardany O, Bryant JP, Luther E, Chen SH, Thompson JW, Starke RM. Endothelial dysfunction in cerebral aneurysms. Neurosurg Focus 2019; 47:E3. [PMID: 31389675 DOI: 10.3171/2019.4.focus19221] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Endothelial cell (EC) dysfunction is known to contribute to cerebral aneurysm (CA) pathogenesis. Evidence shows that damage or injury to the EC layer is the first event in CA formation. The mechanisms behind EC dysfunction in CA disease are interrelated and include hemodynamic stress, hazardous nitric oxide synthase (NOS) activity, oxidative stress, estrogen imbalance, and endothelial cell-to-cell junction compromise. Abnormal variations in hemodynamic stress incite pathological EC transformation and inflammatory zone formation, ultimately leading to destruction of the vascular wall and aneurysm dilation. Hemodynamic stress activates key molecular pathways that result in the upregulation of chemotactic cytokines and adhesion molecules, leading to inflammatory cell recruitment and infiltration. Concurrently, oxidative stress damages EC-to-EC junction proteins, resulting in interendothelial gap formation. This further promotes leukocyte traffic into the vessel wall and the release of matrix metalloproteinases, which propagates vascular remodeling and breakdown. Abnormal hemodynamic stress and inflammation also trigger adverse changes in NOS activity, altering proper EC mediation of vascular tone and the local inflammatory environment. Additionally, the vasoprotective hormone estrogen modulates gene expression that often suppresses these harmful processes. Crosstalk between these sophisticated pathways contributes to CA initiation, progression, and rupture. This review aims to outline the complex mechanisms of EC dysfunction in CA pathogenesis.
Collapse
|
16
|
Ikedo T, Kataoka H, Minami M, Hayashi K, Miyata T, Nagata M, Fujikawa R, Yokode M, Imai H, Matsuda T, Miyamoto S. Sequential Inward Bending of Arterial Bifurcations is Associated with Intracranial Aneurysm Formation. World Neurosurg 2019; 129:e361-e366. [PMID: 31176059 DOI: 10.1016/j.wneu.2019.05.153] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 05/15/2019] [Accepted: 05/16/2019] [Indexed: 10/26/2022]
Abstract
OBJECTIVE To investigate the association between vascular morphology and the development of intracranial aneurysms (IAs), the morphological changes of intracranial arteries after IA induction were examined using a rodent model. METHODS The vascular morphology of the circle of Willis in rats was visualized at 1 week and at 3 months after IA induction using 7-T magnetic resonance imaging. The following 2 angle parameters were defined: the angle between the parent artery and the daughter arteries (PD angle), and the widening of the daughter arteries (DD angle). The correlations of the angle parameters with IA size and with the number of macrophages infiltrated in the IA wall by immunohistochemistry were examined. RESULTS Magnetic resonance imaging showed bending of the arteries over time around the predilection site for IAs. The PD angle increased significantly 1 week after IA induction (P < 0.05) and correlated with IA size (P < 0.01). The DD angle did not increase after 1 week, but increased 3 months after IA induction (P < 0.01). The PD angle 1 week after surgery also correlated with the number of infiltrated macrophages in aneurysmal walls (P = 0.01). CONCLUSIONS Sequential inward bending of arterial bifurcations occurred after IA induction in the rat model. The degree of arterial bending correlated with IA development and inflammation in the IA wall, suggesting that the vascular morphology may be strongly associated with IA development through a proinflammatory mechanism.
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
| | - Hiroharu Kataoka
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Manabu Minami
- Department of Clinical Innovative Medicine, 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
| | - 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 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
| | - Masayuki Yokode
- Department of Clinical Innovative Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hirohiko Imai
- Department of Systems Science, Graduate School of Informatics, Kyoto University, Kyoto, Japan
| | - Tetsuya Matsuda
- Department of Systems Science, Graduate School of Informatics, Kyoto University, Kyoto, Japan
| | - Susumu Miyamoto
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
17
|
Shimizu K, Kushamae M, Mizutani T, Aoki T. Intracranial Aneurysm as a Macrophage-mediated Inflammatory Disease. Neurol Med Chir (Tokyo) 2019; 59:126-132. [PMID: 30867357 PMCID: PMC6465529 DOI: 10.2176/nmc.st.2018-0326] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Subarachnoid hemorrhage (SAH) is mainly attributable to the rupture of intracranial aneurysms (IAs). Although the outcome of SAH is considerably poor in spite of the recent intensive medical care, mechanisms regulating the progression of IAs or triggering rupture remain to be clarified, making the development of effective preemptive medicine to prevent SAH difficult. However, a series of recent studies have been expanding our understanding of the pathogenesis of IAs. These studies have suggested the crucial role of macrophage-mediated chronic inflammation in the pathogenesis of IAs. In histopathological analyses of IA lesions in humans and induced in animal models, the number of macrophages infiltrating in lesions is positively correlated with enlargement or rupture of IAs. In animal models, a genetic deletion or an inhibition of monocyte chemotactic protein-1, a major chemoattractant for macrophages, or a pharmacological depletion of macrophages consistently suppresses the development and progression of IAs. Furthermore, a macrophage-specific deletion of Ptger2 (gene for prostaglandin E receptor subtype 2) or a macrophage-specific expression of a mutated form of IκBα which inhibits nuclear translocation of nuclear factor κB significantly suppress the development of IAs, supporting the role of macrophages and the inflammatory signaling functioning there in the pathogenesis of IAs. The development of drug therapies suppressing macrophage-mediated inflammatory responses in situ can thus be a potential strategy in the pre-emptive medicine targeting SAH. In this manuscript, we summarize the experimental evidences about the pathogenesis of IAs focused on inflammatory responses and propose the definition of IAs as a macrophage-mediated inflammatory disease.
Collapse
Affiliation(s)
- Kampei Shimizu
- Department of Molecular Pharmacology, Research Institute, National Cerebral and Cardiovascular Center.,Department of Neurosurgery, Kyoto University Graduate School of Medicine
| | - Mika Kushamae
- Department of Molecular Pharmacology, Research Institute, National Cerebral and Cardiovascular Center.,Department of Neurosurgery, Showa University School of Medicine
| | - Tohru Mizutani
- Department of Neurosurgery, Showa University School of Medicine
| | - Tomohiro Aoki
- Department of Molecular Pharmacology, Research Institute, National Cerebral and Cardiovascular Center
| |
Collapse
|
18
|
Hu J, Luo J, Wang H, Wang C, Long R, Li A, Zhou Y, Fang Z, Chen Q. The active participation of p22phox-214T/C in the formation of intracranial aneurysm and the suppressive potential of edaravone. Int J Mol Med 2018; 42:2952-2960. [PMID: 30226557 DOI: 10.3892/ijmm.2018.3846] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 08/23/2018] [Indexed: 11/06/2022] Open
Abstract
Oxidative stress reactions play an important role in the pathogenesis of intracranial aneurysm (IA). p22phox is involved in the oxidative stress reaction, and it is a critical subunit of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase. The present study investigated the association of genetic variants within the gene encoding p22phox‑214T/C with IA. The p22phox‑214T/C gene polymorphisms in 192 cases of IA and 112 controls were analyzed by polymerase chain reaction‑restriction fragment length polymorphism (PCR‑RFLP). The mRNA expression of NADPH oxidase was also analyzed by RT‑PCR. The results of RT‑PCR were validated by ELISA. In a rabbit model of elastase‑induced aneurysm, we used edaravone for anti‑oxidative stress treatment to observe the curative effects. In the clinical cases, a significant difference in p22phox‑214T/C allele frequencies in the IA group was observed compared with the control group (P<0.001). The expression level of NADPH oxidase was differed significantly between the IA group and the control group. In the rabbit model of elastase‑induced aneurysm, the success rate of the aneurysmal model in the edaravone group and the wound ulcer rate were lower than those in the control group. In addition, the diameter of the aneurysm was smaller than in the edaravone group than in the control group (3.26±0.13 mm vs. 3.85±0.07 mm), and the expression of matrix metalloproteinase‑9 (MMP‑9) was significantly lower than that in the control group (P<0.0001). Thus, these data suggest the active participation of p22phox‑214T/C in the formation of IA and the suppressive potential of edaravone against IA formation.
Collapse
Affiliation(s)
- Juntao Hu
- Department of Neurosurgery, Remin Hospital of Wuhan University, Wuhan, Hubei 30060, P.R. China
| | - Jie Luo
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Hui Wang
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Chaojia Wang
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Rongpei Long
- Department of English, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Anrong Li
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Yi Zhou
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Zhicheng Fang
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Qianxue Chen
- Department of Neurosurgery, Remin Hospital of Wuhan University, Wuhan, Hubei 30060, P.R. China
| |
Collapse
|
19
|
Tulamo R, Frösen J, Hernesniemi J, Niemelä M. Inflammatory changes in the aneurysm wall: a review. J Neurointerv Surg 2018; 10:i58-i67. [DOI: 10.1136/jnis.2009.002055.rep] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 12/17/2009] [Accepted: 12/18/2009] [Indexed: 02/06/2023]
Abstract
Rupture of a saccular intracranial artery aneurysm (IA) causes subarachnoid hemorrhage, a significant cause of stroke and death. The current treatment options, endovascular coiling and clipping, are invasive and somewhat risky. Since only some IAs rupture, those IAs at risk for rupture should be identified. However, to improve the imaging of rupture-prone IAs and improve IA treatment, IA wall pathobiology requires more thorough knowledge. Chronic inflammation has become understood as an important phenomenon in IA wall pathobiology, featuring inflammatory cell infiltration as well as proliferative and fibrotic remodulatory responses. We review the literature on what is known about inflammation in the IA wall and also review the probable mechanisms of how inflammation would result in the degenerative changes that ultimately lead to IA wall rupture. We also discuss current options in imaging inflammation and how knowledge of inflammation in IA walls may improve IA treatment.
Collapse
|
20
|
Zhao J, Zhang M, Li Y, Zhang Z, Chen M, Liu T, Zhang J, Shan A. Therapeutic Effect of Hydrogen Injected Subcutaneously on Onion Poisoned Dogs. J Vet Res 2018; 61:527-533. [PMID: 29978119 PMCID: PMC5937354 DOI: 10.1515/jvetres-2017-0068] [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: 07/11/2017] [Accepted: 12/06/2017] [Indexed: 11/15/2022] Open
Abstract
Introduction The purpose of this study was to investigate the therapeutic effect of hydrogen on the therapy of onion poisoned dogs. Material and Methods A total of 16 adult beagle dogs were divided into two groups (control and hydrogen) and all were fed dehydrated onion powder at the dose of 10 g/kg for three days. The dogs of the experimental group were given subcutaneous injection of 0.2 mL/kg of hydrogen for 12 days after making the poisoned model successful. Blood samples were collected before feeding onions, one day before injecting hydrogen, and 2 h after the injection of hydrogen on days 1, 3, 5, 7, 9, and 12. Control dogs were not treated with hydrogen. Results The levels of leukocyte production, anaemia, red blood cell degeneration which was reflected by the values of Heinz body count, haemolytic ratio, and oxidative products in hydrogen treated group were lower than in control dogs on some days. The capacity of medullary haematopoiesis that was based on reticulocyte counts, and the antioxidation in hydrogen group were higher compared with control group. However, the differences in renal function were not obvious in both groups. Conclusion Accordingly, it was concluded that subcutaneous injection of hydrogen could alleviate the symptoms in onion poisoned dogs.
Collapse
Affiliation(s)
- Jinghua Zhao
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Harbin 150030, China
| | - Ming Zhang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Harbin 150030, China
| | - Yue Li
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Harbin 150030, China
| | - Zhiheng Zhang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Harbin 150030, China
| | - Mingzi Chen
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Harbin 150030, China
| | - Tao Liu
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Harbin 150030, China
| | - Jiantao Zhang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Harbin 150030, China
| | - Anshan Shan
- College of Animal Science and Technology, Northeast Agriculture University, Harbin 150030, China
| |
Collapse
|
21
|
Parikh A, Kathawala K, Tan CC, Garg S, Zhou XF. Self-nanomicellizing solid dispersion of edaravone: part I - oral bioavailability improvement. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:2051-2069. [PMID: 30013324 PMCID: PMC6038876 DOI: 10.2147/dddt.s161940] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background Edaravone (EDR) is known for its free radical scavenging, antiapoptotic, antinecrotic, and anticytokine effects in neurological and non-neurological diseases. It is currently available clinically as Radicava® and Radicut®, intravenous medications, recently approved for the treatment of amyotrophic lateral sclerosis and cerebral infarction. However, the oral use of EDR is still restricted by its poor oral bioavailability (BA) due to poor aqueous solubility, stability, rapid metabolism, and low permeability. The present study reports the development of novel EDR formulation (NEF) using self-nanomicellizing solid dispersion (SNMSD) strategy with the aim to enable its oral use. Materials and methods The selection of a suitable carrier for the development of NEF was performed based on the miscibility study. The optimization of EDR-to-carrier ratio was conducted via kinetic solubility study after preparing SNMSDs using solvent evaporation technique. The drug–polymer carrier interaction and self-nanomicellizing properties of NEF were investigated with advanced characterization studies. In vitro permeation, metabolism, and dissolution study was carried out to examine the effect of the presence of a carrier on physico-chemical properties of EDR. Additionally, the dose-dependent pharmacokinetic study of NEF was conducted and compared with the EDR suspension. Results Soluplus® (SOL) as a carrier was selected based on the potential for improving aqueous solubility. The NEF containing EDR and SOL (1:5) resulted in the highest enhancement in aqueous solubility (17.53-fold) due to amorphization, hydrogen bonding interaction, and micellization. Moreover, the NEF demonstrated significant improvement in metabolism, permeability, and dissolution profile of EDR. Furthermore, the oral BA of NEF showed 10.2-, 16.1-, and 14.8-fold enhancement compared to EDR suspension at 46, 138, and 414 µmol/kg doses. Conclusion The results demonstrated that SNMSD strategy could serve as a promising way to enhance EDR oral BA and NEF could be a potential candidate for the treatment of diseases in which oxidative stress plays a key role in their pathogenesis.
Collapse
Affiliation(s)
- Ankit Parikh
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia, ;
| | - Krishna Kathawala
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia, ;
| | - Chun Chuan Tan
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia, ;
| | - Sanjay Garg
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia, ;
| | - Xin-Fu Zhou
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia, ;
| |
Collapse
|
22
|
Signorelli F, Sela S, Gesualdo L, Chevrel S, Tollet F, Pailler-Mattei C, Tacconi L, Turjman F, Vacca A, Schul DB. Hemodynamic Stress, Inflammation, and Intracranial Aneurysm Development and Rupture: A Systematic Review. World Neurosurg 2018; 115:234-244. [DOI: 10.1016/j.wneu.2018.04.143] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 04/18/2018] [Accepted: 04/19/2018] [Indexed: 10/17/2022]
|
23
|
Starke RM, Thompson JW, Ali MS, Pascale CL, Martinez Lege A, Ding D, Chalouhi N, Hasan DM, Jabbour P, Owens GK, Toborek M, Hare JM, Dumont AS. Cigarette Smoke Initiates Oxidative Stress-Induced Cellular Phenotypic Modulation Leading to Cerebral Aneurysm Pathogenesis. Arterioscler Thromb Vasc Biol 2018; 38:610-621. [PMID: 29348119 DOI: 10.1161/atvbaha.117.310478] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 01/04/2018] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Cigarette smoke exposure (CSE) is a risk factor for cerebral aneurysm (CA) formation, but the molecular mechanisms are unclear. Although CSE is known to contribute to excess reactive oxygen species generation, the role of oxidative stress on vascular smooth muscle cell (VSMC) phenotypic modulation and pathogenesis of CAs is unknown. The goal of this study was to investigate whether CSE activates a NOX (NADPH oxidase)-dependent pathway leading to VSMC phenotypic modulation and CA formation and rupture. APPROACH AND RESULTS In cultured cerebral VSMCs, CSE increased expression of NOX1 and reactive oxygen species which preceded upregulation of proinflammatory/matrix remodeling genes (MCP-1, MMPs [matrix metalloproteinase], TNF-α, IL-1β, NF-κB, KLF4 [Kruppel-like factor 4]) and downregulation of contractile genes (SM-α-actin [smooth muscle α actin], SM-22α [smooth muscle 22α], SM-MHC [smooth muscle myosin heavy chain]) and myocardin. Inhibition of reactive oxygen species production and knockdown of NOX1 with siRNA or antisense decreased CSE-induced upregulation of NOX1 and inflammatory genes and downregulation of VSMC contractile genes and myocardin. p47phox-/- NOX knockout mice, or pretreatment with the NOX inhibitor, apocynin, significantly decreased CA formation and rupture compared with controls. NOX1 protein and mRNA expression were similar in p47phox-/- mice and those pretreated with apocynin but were elevated in unruptured and ruptured CAs. CSE increased CA formation and rupture, which was diminished with apocynin pretreatment. Similarly, NOX1 protein and mRNA and reactive oxygen species were elevated by CSE, and in unruptured and ruptured CAs. CONCLUSIONS CSE initiates oxidative stress-induced phenotypic modulation of VSMCs and CA formation and rupture. These molecular changes implicate oxidative stress in the pathogenesis of CAs and may provide a potential target for future therapeutic strategies.
Collapse
Affiliation(s)
- Robert M Starke
- From the Department of Neurological Surgery & Radiology, University of Miami Cerebrovascular Initiative (R.M.S., J.W.T.), Department of Biochemistry and Molecular Biology (M.T.), and Department of Cardiology and Molecular and Cellular Pharmacology (J.M.H.), University of Miami, FL; Department of Neurosurgery, University of Iowa, Iowa City (M.S.A., D.M.H.); Department of Neurological Surgery, Tulane University, New Orleans, LA (C.L.P., A.M.L., A.S.D.); Department of Neurosurgery (D.D.) and Department of Molecular Physiology & Biophysics, Robert M. Berne Cardiovascular Research Center (G.K.O.), University of Virginia, Charlottesville; and Department of Neurosurgery, Thomas Jefferson University, Philadelphia, PA (N.C., P.J.).
| | - John W Thompson
- From the Department of Neurological Surgery & Radiology, University of Miami Cerebrovascular Initiative (R.M.S., J.W.T.), Department of Biochemistry and Molecular Biology (M.T.), and Department of Cardiology and Molecular and Cellular Pharmacology (J.M.H.), University of Miami, FL; Department of Neurosurgery, University of Iowa, Iowa City (M.S.A., D.M.H.); Department of Neurological Surgery, Tulane University, New Orleans, LA (C.L.P., A.M.L., A.S.D.); Department of Neurosurgery (D.D.) and Department of Molecular Physiology & Biophysics, Robert M. Berne Cardiovascular Research Center (G.K.O.), University of Virginia, Charlottesville; and Department of Neurosurgery, Thomas Jefferson University, Philadelphia, PA (N.C., P.J.)
| | - Muhammad S Ali
- From the Department of Neurological Surgery & Radiology, University of Miami Cerebrovascular Initiative (R.M.S., J.W.T.), Department of Biochemistry and Molecular Biology (M.T.), and Department of Cardiology and Molecular and Cellular Pharmacology (J.M.H.), University of Miami, FL; Department of Neurosurgery, University of Iowa, Iowa City (M.S.A., D.M.H.); Department of Neurological Surgery, Tulane University, New Orleans, LA (C.L.P., A.M.L., A.S.D.); Department of Neurosurgery (D.D.) and Department of Molecular Physiology & Biophysics, Robert M. Berne Cardiovascular Research Center (G.K.O.), University of Virginia, Charlottesville; and Department of Neurosurgery, Thomas Jefferson University, Philadelphia, PA (N.C., P.J.)
| | - Crissey L Pascale
- From the Department of Neurological Surgery & Radiology, University of Miami Cerebrovascular Initiative (R.M.S., J.W.T.), Department of Biochemistry and Molecular Biology (M.T.), and Department of Cardiology and Molecular and Cellular Pharmacology (J.M.H.), University of Miami, FL; Department of Neurosurgery, University of Iowa, Iowa City (M.S.A., D.M.H.); Department of Neurological Surgery, Tulane University, New Orleans, LA (C.L.P., A.M.L., A.S.D.); Department of Neurosurgery (D.D.) and Department of Molecular Physiology & Biophysics, Robert M. Berne Cardiovascular Research Center (G.K.O.), University of Virginia, Charlottesville; and Department of Neurosurgery, Thomas Jefferson University, Philadelphia, PA (N.C., P.J.)
| | - Alejandra Martinez Lege
- From the Department of Neurological Surgery & Radiology, University of Miami Cerebrovascular Initiative (R.M.S., J.W.T.), Department of Biochemistry and Molecular Biology (M.T.), and Department of Cardiology and Molecular and Cellular Pharmacology (J.M.H.), University of Miami, FL; Department of Neurosurgery, University of Iowa, Iowa City (M.S.A., D.M.H.); Department of Neurological Surgery, Tulane University, New Orleans, LA (C.L.P., A.M.L., A.S.D.); Department of Neurosurgery (D.D.) and Department of Molecular Physiology & Biophysics, Robert M. Berne Cardiovascular Research Center (G.K.O.), University of Virginia, Charlottesville; and Department of Neurosurgery, Thomas Jefferson University, Philadelphia, PA (N.C., P.J.)
| | - Dale Ding
- From the Department of Neurological Surgery & Radiology, University of Miami Cerebrovascular Initiative (R.M.S., J.W.T.), Department of Biochemistry and Molecular Biology (M.T.), and Department of Cardiology and Molecular and Cellular Pharmacology (J.M.H.), University of Miami, FL; Department of Neurosurgery, University of Iowa, Iowa City (M.S.A., D.M.H.); Department of Neurological Surgery, Tulane University, New Orleans, LA (C.L.P., A.M.L., A.S.D.); Department of Neurosurgery (D.D.) and Department of Molecular Physiology & Biophysics, Robert M. Berne Cardiovascular Research Center (G.K.O.), University of Virginia, Charlottesville; and Department of Neurosurgery, Thomas Jefferson University, Philadelphia, PA (N.C., P.J.)
| | - Nohra Chalouhi
- From the Department of Neurological Surgery & Radiology, University of Miami Cerebrovascular Initiative (R.M.S., J.W.T.), Department of Biochemistry and Molecular Biology (M.T.), and Department of Cardiology and Molecular and Cellular Pharmacology (J.M.H.), University of Miami, FL; Department of Neurosurgery, University of Iowa, Iowa City (M.S.A., D.M.H.); Department of Neurological Surgery, Tulane University, New Orleans, LA (C.L.P., A.M.L., A.S.D.); Department of Neurosurgery (D.D.) and Department of Molecular Physiology & Biophysics, Robert M. Berne Cardiovascular Research Center (G.K.O.), University of Virginia, Charlottesville; and Department of Neurosurgery, Thomas Jefferson University, Philadelphia, PA (N.C., P.J.)
| | - David M Hasan
- From the Department of Neurological Surgery & Radiology, University of Miami Cerebrovascular Initiative (R.M.S., J.W.T.), Department of Biochemistry and Molecular Biology (M.T.), and Department of Cardiology and Molecular and Cellular Pharmacology (J.M.H.), University of Miami, FL; Department of Neurosurgery, University of Iowa, Iowa City (M.S.A., D.M.H.); Department of Neurological Surgery, Tulane University, New Orleans, LA (C.L.P., A.M.L., A.S.D.); Department of Neurosurgery (D.D.) and Department of Molecular Physiology & Biophysics, Robert M. Berne Cardiovascular Research Center (G.K.O.), University of Virginia, Charlottesville; and Department of Neurosurgery, Thomas Jefferson University, Philadelphia, PA (N.C., P.J.)
| | - Pascal Jabbour
- From the Department of Neurological Surgery & Radiology, University of Miami Cerebrovascular Initiative (R.M.S., J.W.T.), Department of Biochemistry and Molecular Biology (M.T.), and Department of Cardiology and Molecular and Cellular Pharmacology (J.M.H.), University of Miami, FL; Department of Neurosurgery, University of Iowa, Iowa City (M.S.A., D.M.H.); Department of Neurological Surgery, Tulane University, New Orleans, LA (C.L.P., A.M.L., A.S.D.); Department of Neurosurgery (D.D.) and Department of Molecular Physiology & Biophysics, Robert M. Berne Cardiovascular Research Center (G.K.O.), University of Virginia, Charlottesville; and Department of Neurosurgery, Thomas Jefferson University, Philadelphia, PA (N.C., P.J.)
| | - Gary K Owens
- From the Department of Neurological Surgery & Radiology, University of Miami Cerebrovascular Initiative (R.M.S., J.W.T.), Department of Biochemistry and Molecular Biology (M.T.), and Department of Cardiology and Molecular and Cellular Pharmacology (J.M.H.), University of Miami, FL; Department of Neurosurgery, University of Iowa, Iowa City (M.S.A., D.M.H.); Department of Neurological Surgery, Tulane University, New Orleans, LA (C.L.P., A.M.L., A.S.D.); Department of Neurosurgery (D.D.) and Department of Molecular Physiology & Biophysics, Robert M. Berne Cardiovascular Research Center (G.K.O.), University of Virginia, Charlottesville; and Department of Neurosurgery, Thomas Jefferson University, Philadelphia, PA (N.C., P.J.)
| | - Michal Toborek
- From the Department of Neurological Surgery & Radiology, University of Miami Cerebrovascular Initiative (R.M.S., J.W.T.), Department of Biochemistry and Molecular Biology (M.T.), and Department of Cardiology and Molecular and Cellular Pharmacology (J.M.H.), University of Miami, FL; Department of Neurosurgery, University of Iowa, Iowa City (M.S.A., D.M.H.); Department of Neurological Surgery, Tulane University, New Orleans, LA (C.L.P., A.M.L., A.S.D.); Department of Neurosurgery (D.D.) and Department of Molecular Physiology & Biophysics, Robert M. Berne Cardiovascular Research Center (G.K.O.), University of Virginia, Charlottesville; and Department of Neurosurgery, Thomas Jefferson University, Philadelphia, PA (N.C., P.J.)
| | - Joshua M Hare
- From the Department of Neurological Surgery & Radiology, University of Miami Cerebrovascular Initiative (R.M.S., J.W.T.), Department of Biochemistry and Molecular Biology (M.T.), and Department of Cardiology and Molecular and Cellular Pharmacology (J.M.H.), University of Miami, FL; Department of Neurosurgery, University of Iowa, Iowa City (M.S.A., D.M.H.); Department of Neurological Surgery, Tulane University, New Orleans, LA (C.L.P., A.M.L., A.S.D.); Department of Neurosurgery (D.D.) and Department of Molecular Physiology & Biophysics, Robert M. Berne Cardiovascular Research Center (G.K.O.), University of Virginia, Charlottesville; and Department of Neurosurgery, Thomas Jefferson University, Philadelphia, PA (N.C., P.J.)
| | - Aaron S Dumont
- From the Department of Neurological Surgery & Radiology, University of Miami Cerebrovascular Initiative (R.M.S., J.W.T.), Department of Biochemistry and Molecular Biology (M.T.), and Department of Cardiology and Molecular and Cellular Pharmacology (J.M.H.), University of Miami, FL; Department of Neurosurgery, University of Iowa, Iowa City (M.S.A., D.M.H.); Department of Neurological Surgery, Tulane University, New Orleans, LA (C.L.P., A.M.L., A.S.D.); Department of Neurosurgery (D.D.) and Department of Molecular Physiology & Biophysics, Robert M. Berne Cardiovascular Research Center (G.K.O.), University of Virginia, Charlottesville; and Department of Neurosurgery, Thomas Jefferson University, Philadelphia, PA (N.C., P.J.)
| |
Collapse
|
24
|
NADPH Oxidase Deficiency: A Multisystem Approach. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:4590127. [PMID: 29430280 PMCID: PMC5753020 DOI: 10.1155/2017/4590127] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 10/11/2017] [Accepted: 11/02/2017] [Indexed: 02/07/2023]
Abstract
The immune system is a complex system able to recognize a wide variety of host agents, through different biological processes. For example, controlled changes in the redox state are able to start different pathways in immune cells and are involved in the killing of microbes. The generation and release of ROS in the form of an “oxidative burst” represent the pivotal mechanism by which phagocytic cells are able to destroy pathogens. On the other hand, impaired oxidative balance is also implicated in the pathogenesis of inflammatory complications, which may affect the function of many body systems. NADPH oxidase (NOX) plays a pivotal role in the production of ROS, and the defect of its different subunits leads to the development of chronic granulomatous disease (CGD). The defect of the different NOX subunits in CGD affects different organs. In this context, this review will be focused on the description of the effect of NOX2 deficiency in different body systems. Moreover, we will also focus our attention on the novel insight in the pathogenesis of immunodeficiency and inflammation-related manifestations and on the protective role of NOX2 deficiency against the development of atherosclerosis.
Collapse
|
25
|
Tanaka LY, Laurindo FRM. Vascular remodeling: A redox-modulated mechanism of vessel caliber regulation. Free Radic Biol Med 2017; 109:11-21. [PMID: 28109889 DOI: 10.1016/j.freeradbiomed.2017.01.025] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 01/13/2017] [Accepted: 01/15/2017] [Indexed: 11/17/2022]
Abstract
Vascular remodeling, i.e. whole-vessel structural reshaping, determines lumen caliber in (patho)physiology. Here we review mechanisms underlying vessel remodeling, with emphasis in redox regulation. First, we discuss confusing terminology and focus on strictu sensu remodeling. Second, we propose a mechanobiological remodeling paradigm based on the concept of tensional homeostasis as a setpoint regulator. We first focus on shear-mediated models as prototypes of remodeling closely dominated by highly redox-sensitive endothelial function. More detailed discussions focus on mechanosensors, integrins, extracellular matrix, cytoskeleton and inflammatory pathways as potential of mechanisms potentially coupling tensional homeostasis to redox regulation. Further discussion of remodeling associated with atherosclerosis and injury repair highlights important aspects of redox vascular responses. While neointima formation has not shown consistent responsiveness to antioxidants, vessel remodeling has been more clearly responsive, indicating that despite the multilevel redox signaling pathways, there is a coordinated response of the whole vessel. Among mechanisms that may orchestrate redox pathways, we discuss roles of superoxide dismutase activity and extracellular protein disulfide isomerase. We then discuss redox modulation of aneurysms, a special case of expansive remodeling. We propose that the redox modulation of vascular remodeling may reflect (1) remodeling pathophysiology is dominated by a particularly redox-sensitive cell type, e.g., endothelial cells (2) redox pathways are temporospatially coordinated at an organ level across distinct cellular and acellular structures or (3) the tensional homeostasis setpoint is closely connected to redox signaling. The mechanobiological/redox model discussed here can be a basis for improved understanding of remodeling and helps clarifying mechanisms underlying prevalent hard-to-treat diseases.
Collapse
Affiliation(s)
- Leonardo Y Tanaka
- Vascular Biology Laboratory, Heart Institute (InCor), University of São Paulo School of Medicine, Av. Enéas Carvalho Aguiar, 44, Annex II, 9th Floor, São Paulo CEP 05403-000, Brazil
| | - Francisco R M Laurindo
- Vascular Biology Laboratory, Heart Institute (InCor), University of São Paulo School of Medicine, Av. Enéas Carvalho Aguiar, 44, Annex II, 9th Floor, São Paulo CEP 05403-000, Brazil.
| |
Collapse
|
26
|
Schröder K, Weissmann N, Brandes RP. Organizers and activators: Cytosolic Nox proteins impacting on vascular function. Free Radic Biol Med 2017; 109:22-32. [PMID: 28336130 DOI: 10.1016/j.freeradbiomed.2017.03.017] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 02/24/2017] [Accepted: 03/14/2017] [Indexed: 01/25/2023]
Abstract
NADPH oxidases of the Nox family are important enzymatic sources of reactive oxygen species (ROS) in the cardiovascular system. Of the 7 members of the Nox family, at least three depend for their activation on specific cytosolic proteins. These are p47phox and its homologue NoxO1 and p67phox and its homologue NoxA1. Also the Rho-GTPase Rac is important but as this protein has many additional functions, it will not be covered here. The Nox1 enzyme is preferentially activated by the combination of NoxO1 with NoxA1, whereas Nox2 gains highest activity with p47phox together with p67phox. As p47phox, different to NoxO1 contains an auto inhibitory region it has to be phosphorylated prior to complex formation. In the cardio-vascular system, all cytosolic Nox proteins are expressed but the evidence for their contribution to ROS production is not well established. Most data have been collected for p47phox, whereas NoxA1 has basically not yet been studied. In this article the specific aspects of cytosolic Nox proteins in the cardiovascular system with respect to Nox activation, their expression and their importance will be reviewed. Finally, it will be discussed whether cytosolic Nox proteins are suitable pharmacological targets to tamper with vascular ROS production.
Collapse
Affiliation(s)
- Katrin Schröder
- Institut für Kardiovaskuläre Physiologie, Goethe-Universität Frankfurt, Frankfurt, Germany.
| | - Norbert Weissmann
- Excellence Cluster Cardiopulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, 35392 Giessen, Germany
| | - Ralf P Brandes
- Institut für Kardiovaskuläre Physiologie, Goethe-Universität Frankfurt, Frankfurt, Germany
| |
Collapse
|
27
|
Abstract
The prevalence of unruptured cerebral aneurysms (UCAs) in elderly patients is increasing in our aging population. UCA management in elderly patients has some difficulties, such as reduced life expectancy, increased comorbidities and treatment risks, and poor prognosis in case of rupture. In this review article, we summarize the most recent findings on the natural history, therapeutic options and treatment results for UCAs exclusively in elderly patients, and describe possible medical treatments for patients with UCAs.
Collapse
Affiliation(s)
- Tomohito Hishikawa
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences
| | - Isao Date
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences
| |
Collapse
|
28
|
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
|
29
|
Abstract
Most of cerebral aneurysms (CAs) are incidentally discovered without any neurological symptoms and the risk of rupture of CAs is relatively higher in Japanese population. The goal of treatments for patients with CAs is complete exclusion of the aneurysmal rupture risk for their lives. Since two currently available major treatments, microsurgical clipping and endovascular coiling, have inherent incompleteness to achieve cure of CAs with some considerable treatment risks, and there is no effective surgical or medical intervention to inhibit the formation of CAs in patients with ruptured and unruptured CAs, new treatment strategies with lower risk and higher efficacy should be developed to prevent the formation, growth, and rupture of CAs. Preemptive medicine for CAs should be designed to prevent or delay the onset of symptoms from CAs found in an asymptomatic state or inhibit the de novo formation of CAs, but we have no definite methods to distinguish rupture-prone aneurysms from rupture-resistant ones. Recent advancements in the research of CAs have provided us with some clues, and one of the new treatment strategies for CAs will be developed based on the findings that several inflammatory pathways may be involved in the formation, growth, and rupture of CAs. Preemptive medicine for CAs will be established with specific biomarkers and imaging modalities which can sensor the development of CAs.
Collapse
Affiliation(s)
- Tomohiro Aoki
- Innovation Center for Immunoregulation Technologies and Drugs, Kyoto University Graduate School of Medicine
| | | |
Collapse
|
30
|
Sharma AK, Salmon MD, Lu G, Su G, Pope NH, Smith JR, Weiss ML, Upchurch GR. Mesenchymal Stem Cells Attenuate NADPH Oxidase-Dependent High Mobility Group Box 1 Production and Inhibit Abdominal Aortic Aneurysms. Arterioscler Thromb Vasc Biol 2016; 36:908-18. [PMID: 26988591 DOI: 10.1161/atvbaha.116.307373] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 02/23/2016] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Abdominal aortic aneurysm (AAA) formation is characterized by inflammation, smooth muscle activation, and matrix degradation. This study tests the hypothesis that macrophage-produced high mobility group box 1 (HMGB1) production is dependent on nicotinamide adenine dinucleotide phosphate oxidase (Nox2), which leads to increase in interleukin (IL)-17 production resulting in AAA formation and that treatment with human mesenchymal stem cells (MSCs) can attenuate this process thereby inhibiting AAA formation. APPROACH AND RESULTS Human aortic tissue demonstrated a significant increase in HMGB1 expression in AAA patients when compared with controls. An elastase-perfusion model of AAA demonstrated a significant increase in HMGB1 production in C57BL/6 (wild-type [WT]) mice, which was attenuated by MSC treatment. Furthermore, anti-HMGB1 antibody treatment of WT mice attenuated AAA formation, IL-17 production, and immune cell infiltration when compared with elastase-perfused WT mice on day 14. Elastase-perfused Nox2(-/y) mice demonstrated a significant attenuation of HMGB1 and IL-17 production, cellular infiltration, matrix metalloproteinase activity, and AAA formation when compared with WT mice on day 14. In vitro studies showed that elastase-treated macrophages from WT mice, but not from Nox2(-/y) mice, produced HMGB1, which was attenuated by MSC treatment. The production of macrophage-dependent HMGB1 involved Nox2 activation and superoxide anion production, which was mitigated by MSC treatment. CONCLUSIONS These results demonstrate that macrophage-produced HMGB1 leads to aortic inflammation and acts as a trigger for CD4(+) T-cell-produced IL-17 during AAA formation. HMGB1 release is dependent on Nox2 activation, which can be inhibited by MSCs leading to attenuation of proinflammatory cytokines, especially IL-17, and protection against AAA formation.
Collapse
Affiliation(s)
- Ashish K Sharma
- From the Department of Surgery, University of Virginia, Charlottesville (A.K.S., M.D.S., G.L., G.S., N.H.P., G.R.U.); and Department of Anatomy and Physiology, Kansas State University, Manhattan (J.R.S., M.L.W.)
| | - Morgan D Salmon
- From the Department of Surgery, University of Virginia, Charlottesville (A.K.S., M.D.S., G.L., G.S., N.H.P., G.R.U.); and Department of Anatomy and Physiology, Kansas State University, Manhattan (J.R.S., M.L.W.)
| | - Guanyi Lu
- From the Department of Surgery, University of Virginia, Charlottesville (A.K.S., M.D.S., G.L., G.S., N.H.P., G.R.U.); and Department of Anatomy and Physiology, Kansas State University, Manhattan (J.R.S., M.L.W.)
| | - Gang Su
- From the Department of Surgery, University of Virginia, Charlottesville (A.K.S., M.D.S., G.L., G.S., N.H.P., G.R.U.); and Department of Anatomy and Physiology, Kansas State University, Manhattan (J.R.S., M.L.W.)
| | - Nicolas H Pope
- From the Department of Surgery, University of Virginia, Charlottesville (A.K.S., M.D.S., G.L., G.S., N.H.P., G.R.U.); and Department of Anatomy and Physiology, Kansas State University, Manhattan (J.R.S., M.L.W.)
| | - Joseph R Smith
- From the Department of Surgery, University of Virginia, Charlottesville (A.K.S., M.D.S., G.L., G.S., N.H.P., G.R.U.); and Department of Anatomy and Physiology, Kansas State University, Manhattan (J.R.S., M.L.W.)
| | - Mark L Weiss
- From the Department of Surgery, University of Virginia, Charlottesville (A.K.S., M.D.S., G.L., G.S., N.H.P., G.R.U.); and Department of Anatomy and Physiology, Kansas State University, Manhattan (J.R.S., M.L.W.)
| | - Gilbert R Upchurch
- From the Department of Surgery, University of Virginia, Charlottesville (A.K.S., M.D.S., G.L., G.S., N.H.P., G.R.U.); and Department of Anatomy and Physiology, Kansas State University, Manhattan (J.R.S., M.L.W.).
| |
Collapse
|
31
|
Wang Y, Emeto TI, Lee J, Marshman L, Moran C, Seto S, Golledge J. Mouse models of intracranial aneurysm. Brain Pathol 2015; 25:237-47. [PMID: 25041057 PMCID: PMC8029187 DOI: 10.1111/bpa.12175] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 07/09/2014] [Indexed: 01/04/2023] Open
Abstract
Subarachnoid hemorrhage secondary to rupture of an intracranial aneurysm is a highly lethal medical condition. Current management strategies for unruptured intracranial aneurysms involve radiological surveillance and neurosurgical or endovascular interventions. There is no pharmacological treatment available to decrease the risk of aneurysm rupture and subsequent subarachnoid hemorrhage. There is growing interest in the pathogenesis of intracranial aneurysm focused on the development of drug therapies to decrease the incidence of aneurysm rupture. The study of rodent models of intracranial aneurysms has the potential to improve our understanding of intracranial aneurysm development and progression. This review summarizes current mouse models of intact and ruptured intracranial aneurysms and discusses the relevance of these models to human intracranial aneurysms. The article also reviews the importance of these models in investigating the molecular mechanisms involved in the disease. Finally, potential pharmaceutical targets for intracranial aneurysm suggested by previous studies are discussed. Examples of potential drug targets include matrix metalloproteinases, stromal cell-derived factor-1, tumor necrosis factor-α, the renin-angiotensin system and the β-estrogen receptor. An agreed clear, precise and reproducible definition of what constitutes an aneurysm in the models would assist in their use to better understand the pathology of intracranial aneurysm and applying findings to patients.
Collapse
Affiliation(s)
- Yutang Wang
- The Vascular Biology UnitQueensland Research Centre for Peripheral Vascular DiseaseSchool of Medicine and DentistryJames Cook UniversityTownsvilleQueenslandAustralia
| | - Theophilus I. Emeto
- The Vascular Biology UnitQueensland Research Centre for Peripheral Vascular DiseaseSchool of Medicine and DentistryJames Cook UniversityTownsvilleQueenslandAustralia
- Discipline of Public Health and Tropical MedicineSchool of Public HealthTropical Medicine and Rehabilitation SciencesJames Cook UniversityTownsvilleQueenslandAustralia
| | - James Lee
- The Vascular Biology UnitQueensland Research Centre for Peripheral Vascular DiseaseSchool of Medicine and DentistryJames Cook UniversityTownsvilleQueenslandAustralia
- Department of NeurosurgeryThe Townsville HospitalTownsvilleQueenslandAustralia
| | - Laurence Marshman
- The Vascular Biology UnitQueensland Research Centre for Peripheral Vascular DiseaseSchool of Medicine and DentistryJames Cook UniversityTownsvilleQueenslandAustralia
- Department of NeurosurgeryThe Townsville HospitalTownsvilleQueenslandAustralia
| | - Corey Moran
- The Vascular Biology UnitQueensland Research Centre for Peripheral Vascular DiseaseSchool of Medicine and DentistryJames Cook UniversityTownsvilleQueenslandAustralia
| | - Sai‐wang Seto
- The Vascular Biology UnitQueensland Research Centre for Peripheral Vascular DiseaseSchool of Medicine and DentistryJames Cook UniversityTownsvilleQueenslandAustralia
| | - Jonathan Golledge
- The Vascular Biology UnitQueensland Research Centre for Peripheral Vascular DiseaseSchool of Medicine and DentistryJames Cook UniversityTownsvilleQueenslandAustralia
- Department of Vascular and Endovascular SurgeryThe Townsville HospitalTownsvilleQueenslandAustralia
| |
Collapse
|
32
|
Hasan DM, Starke RM, Gu H, Wilson K, Chu Y, Chalouhi N, Heistad DD, Faraci FM, Sigmund CD. Smooth Muscle Peroxisome Proliferator-Activated Receptor γ Plays a Critical Role in Formation and Rupture of Cerebral Aneurysms in Mice In Vivo. Hypertension 2015; 66:211-20. [PMID: 25916724 DOI: 10.1161/hypertensionaha.115.05332] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 04/07/2015] [Indexed: 12/17/2022]
Abstract
Vascular inflammation plays a critical role in the pathogenesis of cerebral aneurysms. Peroxisome proliferator-activated receptor γ (PPARγ) protects against vascular inflammation and atherosclerosis, whereas dominant-negative mutations in PPARγ promote atherosclerosis and vascular dysfunction. We tested the role of PPARγ in aneurysm formation and rupture. Aneurysms were induced with a combination of systemic infusion of angiotensin-II and local injection of elastase in (1) mice that received the PPARγ antagonist GW9662 or the PPARγ agonist pioglitazone, (2) mice carrying dominant-negative PPARγ mutations in endothelial or smooth muscle cells, and (3) mice that received the Cullin inhibitor MLN4924. Incidence of aneurysm formation, rupture, and mortality was quantified. Cerebral arteries were analyzed for expression of Cullin3, Kelch-like ECH-associated protein 1, nuclear factor (erythroid-derived 2)-like 2, NAD(P)H dehydrogenase (quinone)1 (NQO1), and inflammatory marker mRNAs. Neither pioglitazone nor GW9662 altered the incidence of aneurysm formation. GW9662 significantly increased the incidence of aneurysm rupture, whereas pioglitazone tended to decrease the incidence of rupture. Dominant-negative endothelial-specific PPARγ did not alter the incidence of aneurysm formation or rupture. In contrast, dominant-negative smooth muscle-specific PPARγ resulted in an increase in aneurysm formation (P<0.05) and rupture (P=0.05). Dominant-negative smooth muscle-specific PPARγ, but not dominant-negative endothelial-specific PPARγ, resulted in significant decreases in expression of genes encoding Cullin3, Kelch-like ECH-associated protein 1, and nuclear factor (erythroid-derived 2)-like 2, along with significant increases in tumor necrosis factor-α, monocyte chemoattractant protein-1, chemokine (C-X-C motif) ligand 1, CD68, matrix metalloproteinase-3, -9, and -13. MLN4924 did not alter incidence of aneurysm formation, but increased the incidence of rupture (P<0.05). In summary, endogenous PPARγ, specifically smooth muscle PPARγ, plays an important role in protecting from formation and rupture of experimental cerebral aneurysms in mice.
Collapse
Affiliation(s)
- David M Hasan
- From the Department of Neurological Surgery (D.M.H., H.G., K.W., Y.C.) and Departments of Pharmacology and Internal Medicine, Carver College of Medicine (Y.C., D.D.H., F.M.F., C.D.S.), University of Department of Internal Medicine, Iowa; Department of Neurological Surgery, University of Virginia, Charlottesville (R.M.S.); Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA (N.C.); and Department of Internal Medicine, Iowa City Veterans Affairs Healthcare System (F.M.F.)
| | - Robert M Starke
- From the Department of Neurological Surgery (D.M.H., H.G., K.W., Y.C.) and Departments of Pharmacology and Internal Medicine, Carver College of Medicine (Y.C., D.D.H., F.M.F., C.D.S.), University of Department of Internal Medicine, Iowa; Department of Neurological Surgery, University of Virginia, Charlottesville (R.M.S.); Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA (N.C.); and Department of Internal Medicine, Iowa City Veterans Affairs Healthcare System (F.M.F.)
| | - He Gu
- From the Department of Neurological Surgery (D.M.H., H.G., K.W., Y.C.) and Departments of Pharmacology and Internal Medicine, Carver College of Medicine (Y.C., D.D.H., F.M.F., C.D.S.), University of Department of Internal Medicine, Iowa; Department of Neurological Surgery, University of Virginia, Charlottesville (R.M.S.); Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA (N.C.); and Department of Internal Medicine, Iowa City Veterans Affairs Healthcare System (F.M.F.)
| | - Katina Wilson
- From the Department of Neurological Surgery (D.M.H., H.G., K.W., Y.C.) and Departments of Pharmacology and Internal Medicine, Carver College of Medicine (Y.C., D.D.H., F.M.F., C.D.S.), University of Department of Internal Medicine, Iowa; Department of Neurological Surgery, University of Virginia, Charlottesville (R.M.S.); Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA (N.C.); and Department of Internal Medicine, Iowa City Veterans Affairs Healthcare System (F.M.F.)
| | - Yi Chu
- From the Department of Neurological Surgery (D.M.H., H.G., K.W., Y.C.) and Departments of Pharmacology and Internal Medicine, Carver College of Medicine (Y.C., D.D.H., F.M.F., C.D.S.), University of Department of Internal Medicine, Iowa; Department of Neurological Surgery, University of Virginia, Charlottesville (R.M.S.); Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA (N.C.); and Department of Internal Medicine, Iowa City Veterans Affairs Healthcare System (F.M.F.)
| | - Nohra Chalouhi
- From the Department of Neurological Surgery (D.M.H., H.G., K.W., Y.C.) and Departments of Pharmacology and Internal Medicine, Carver College of Medicine (Y.C., D.D.H., F.M.F., C.D.S.), University of Department of Internal Medicine, Iowa; Department of Neurological Surgery, University of Virginia, Charlottesville (R.M.S.); Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA (N.C.); and Department of Internal Medicine, Iowa City Veterans Affairs Healthcare System (F.M.F.)
| | - Donald D Heistad
- From the Department of Neurological Surgery (D.M.H., H.G., K.W., Y.C.) and Departments of Pharmacology and Internal Medicine, Carver College of Medicine (Y.C., D.D.H., F.M.F., C.D.S.), University of Department of Internal Medicine, Iowa; Department of Neurological Surgery, University of Virginia, Charlottesville (R.M.S.); Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA (N.C.); and Department of Internal Medicine, Iowa City Veterans Affairs Healthcare System (F.M.F.)
| | - Frank M Faraci
- From the Department of Neurological Surgery (D.M.H., H.G., K.W., Y.C.) and Departments of Pharmacology and Internal Medicine, Carver College of Medicine (Y.C., D.D.H., F.M.F., C.D.S.), University of Department of Internal Medicine, Iowa; Department of Neurological Surgery, University of Virginia, Charlottesville (R.M.S.); Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA (N.C.); and Department of Internal Medicine, Iowa City Veterans Affairs Healthcare System (F.M.F.)
| | - Curt D Sigmund
- From the Department of Neurological Surgery (D.M.H., H.G., K.W., Y.C.) and Departments of Pharmacology and Internal Medicine, Carver College of Medicine (Y.C., D.D.H., F.M.F., C.D.S.), University of Department of Internal Medicine, Iowa; Department of Neurological Surgery, University of Virginia, Charlottesville (R.M.S.); Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA (N.C.); and Department of Internal Medicine, Iowa City Veterans Affairs Healthcare System (F.M.F.).
| |
Collapse
|
33
|
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
|
34
|
Liaw N, Dolan Fox JM, Siddiqui AH, Meng H, Kolega J. Endothelial nitric oxide synthase and superoxide mediate hemodynamic initiation of intracranial aneurysms. PLoS One 2014; 9:e101721. [PMID: 24992254 PMCID: PMC4081806 DOI: 10.1371/journal.pone.0101721] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 06/10/2014] [Indexed: 01/08/2023] Open
Abstract
Background Hemodynamic insults at arterial bifurcations are believed to play a critical role in initiating intracranial aneurysms. Recent studies in a rabbit model indicate that aneurysmal damage initiates under specific wall shear stress conditions when smooth muscle cells (SMCs) become pro-inflammatory and produce matrix metalloproteinases (MMPs). The mechanisms leading to SMC activation and MMP production during hemodynamic aneurysm initiation are unknown. The goal is to determine if nitric oxide and/or superoxide induce SMC changes, MMP production and aneurysmal remodeling following hemodynamic insult. Methods Bilateral common carotid artery ligation was performed on rabbits (n = 19, plus 5 sham operations) to induce aneurysmal damage at the basilar terminus. Ligated animals were treated with the nitric oxide synthase (NOS) inhibitor LNAME (n = 7) or the superoxide scavenger TEMPOL (n = 5) and compared to untreated animals (n = 7). Aneurysm development was assessed histologically 5 days after ligation. Changes in NOS isoforms, peroxynitrite, reactive oxygen species (ROS), MMP-2, MMP-9, and smooth muscle α-actin were analyzed by immunohistochemistry. Results LNAME attenuated ligation-induced IEL loss, media thinning and bulge formation. In untreated animals, immunofluorescence showed increased endothelial NOS (eNOS) after ligation, but no change in inducible or neuronal NOS. Furthermore, during aneurysm initiation ROS increased in the media, but not the intima, and there was no change in peroxynitrite. In LNAME-treated animals, ROS production did not change. Together, this suggests that eNOS is important for aneurysm initiation but not by producing superoxide. TEMPOL treatment reduced aneurysm development, indicating that the increased medial superoxide is also necessary for aneurysm initiation. LNAME and TEMPOL treatment in ligated animals restored α-actin and decreased MMPs, suggesting that eNOS and superoxide both lead to SMC de-differentiation and MMP production. Conclusion Aneurysm-inducing hemodynamics lead to increased eNOS and superoxide, which both affect SMC phenotype, increasing MMP production and aneurysmal damage.
Collapse
Affiliation(s)
- Nicholas Liaw
- Toshiba Stroke and Vascular Research Center and Department of Mechanical and Aerospace Engineering, State University of New York, Buffalo, New York, United States of America
| | - Jennifer M. Dolan Fox
- Toshiba Stroke and Vascular Research Center and Department of Neurosurgery, State University of New York, Buffalo, New York, United States of America
| | - Adnan H. Siddiqui
- Toshiba Stroke and Vascular Research Center and Departments Neurosurgery and Radiology, State University of New York, Buffalo, New York, United States of America
| | - Hui Meng
- Toshiba Stroke and Vascular Research Center and Departments of Mechanical and Aerospace Engineering, Neurosurgery, and Biomedical Engineering, State University of New York, Buffalo, New York, United States of America
| | - John Kolega
- Toshiba Stroke and Vascular Research Center and Department Pathology and Anatomical Sciences, State University of New York, Buffalo, New York, United States of America
- * E-mail:
| |
Collapse
|
35
|
Tewari A, Mahendru V, Sinha A, Bilotta F. Antioxidants: The new frontier for translational research in cerebroprotection. J Anaesthesiol Clin Pharmacol 2014; 30:160-71. [PMID: 24803750 PMCID: PMC4009632 DOI: 10.4103/0970-9185.130001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
It is important for the anesthesiologist to understand the etiology of free radical damage and how free-radical scavengers attenuate this, so that this knowledge can be applied to diverse neuro-pathological conditions. This review will concentrate on the role of reactive species of oxygen in the pathophysiology of organ dysfunction, specifically sub arachnoid hemorrhage (SAH), traumatic brain injury (TBI) as well as global central nervous system (CNS) hypoxic, ischemic and reperfusion states. We enumerate potential therapeutic modalities that are been currently investigated and of interest for future trials. Antioxidants are perhaps the next frontier of translational research, especially in neuro-anesthesiology.
Collapse
Affiliation(s)
- Anurag Tewari
- Department of Anesthesiology, Dayanand Medical College, Ludhiana, Punjab, India
| | - Vidhi Mahendru
- Department of Anesthesiology, All India Institute of Medical Sciences, New Delhi, India
| | - Ashish Sinha
- Department of Anesthesiology and Perioperative Medicine, Drexel University College of Medicine, Philadelphia, USA
| | - Federico Bilotta
- Department of Anesthesiology, Critical Care and Pain Medicine, "Sapienza" University of Rome, Rome, Italy
| |
Collapse
|
36
|
Zhao Y, Zheng YF, Luo QQ, Yan T, Liu XX, Han L, Zou L. Edaravone inhibits hypoxia-induced trophoblast-soluble Fms-like tyrosine kinase 1 expression: a possible therapeutic approach to preeclampsia. Placenta 2014; 35:476-82. [PMID: 24840734 DOI: 10.1016/j.placenta.2014.04.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 03/15/2014] [Accepted: 04/07/2014] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To investigate the effects of edaravone, a potent free radical scavenger used clinically, on hypoxia-induced trophoblast-soluble Fms-like tyrosine kinase 1 (sFlt-1) expression. METHODS A trophoblast cell line (HRT-8/SVneo) impaired by cobalt chloride (CoCl2) was used as the cell model under hypoxic conditions. 3-[4, 5-dimethylthiazol-2-yl]-2, 5-diphenyl tetrazolium bromide (MTT) was used to measure the viability of cells exposed to CoCl2 and edaravone. The levels of intracellular reactive oxygen species (ROS) were analyzed by flow cytometry. mRNA expression of sFlt-1, vascular endothelial growth factor (VEGF), and placental growth factor (PlGF) in trophoblasts was measured by real-time polymerase chain reaction, and the secretion of sFlt-1, VEGF, and PlGF proteins was analyzed by enzyme-linked immunosorbent assays (ELISAs). A human umbilical vein endothelial cell (HUVEC) tube-formation assay was performed to identify the effects of CoCl2 and edaravone on vascular development. RESULTS CoCl2 treatment caused the loss of trophoblast viability, the formation of ROS, and sFlt-1 mRNA and protein expression in a dose-dependent manner. Pretreatment with edaravone significantly inhibited hypoxia-induced oxidative stress formation and sFlt-1 expression in trophoblasts. Neither PlGF nor VEGF mRNA or protein expression was increased by CoCl2. In the in vitro tube formation assay, edaravone showed a protective role in vascular development under hypoxic conditions. CONCLUSION This study demonstrated that hypoxia leading to increased sFlt-1 release in trophoblasts may contribute to the placental vascular formation abnormalities observed in preeclampsia and suggested that the free radical scavenger edaravone could be a candidate for the effective treatment of preeclampsia.
Collapse
Affiliation(s)
- Y Zhao
- Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Street, Wuhan 430022, China.
| | - Y F Zheng
- Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Street, Wuhan 430022, China
| | - Q Q Luo
- Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Street, Wuhan 430022, China
| | - T Yan
- Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Street, Wuhan 430022, China
| | - X X Liu
- Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Street, Wuhan 430022, China
| | - L Han
- Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Street, Wuhan 430022, China
| | - L Zou
- Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Street, Wuhan 430022, China.
| |
Collapse
|
37
|
Inhibitory effects of edaravone in β-amyloid-induced neurotoxicity in rats. BIOMED RESEARCH INTERNATIONAL 2014; 2014:370368. [PMID: 24804216 PMCID: PMC3996961 DOI: 10.1155/2014/370368] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 02/24/2014] [Indexed: 01/02/2023]
Abstract
Amyloid protein can damage nerve cells through a variety of biological mechanisms including oxidative stress, alterations in calcium homeostasis, and proapoptosis. Edaravone, a potent free radical scavenger possessing antioxidant effects, has been proved neuroprotective effect in stroke patients. The current study aimed to investigate the effects of EDA in an Aβ-induced rat model of AD, by studying Aβ1–40-induced voltage-gated calcium channel currents in hippocampal CA1 pyramidal neurons, learning and memory behavioral tests, the number of surviving cholinergic neurons in the basal forebrain, and the acetylcholine level in the hippocampus in this rat model of AD. The results showed that the Aβ1–40-induced increase of ICa can be inhibited by EDA in a dose-dependent manner. Treatment with EDA significantly improved Aβ1–40-induced learning and memory performance. Choline acetyltransferase positive cells in basal forebrain and acetylcholine content in the hippocampus were increased by the administration of EDA as compared with the non-EDA treated Aβ1–40 group. These results demonstrate that EDA can inhibit the neurotoxic effect of Aβ toxicity. Collectively, these findings suggest that EDA may serve as a potential complemental treatment strategy for AD.
Collapse
|
38
|
Affiliation(s)
- Alexis S Turjman
- Department of Materials Science & Engineering (A.S.T.) and Institute for Medical Engineering and Science, (A.S.T., E.R.E.), Massachusetts Institute of Technology, Cambridge, MA; Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (E.R.E.); Department of Interventional Neuroradiology, Hôpital Neurologique Pierre Wertheimer, Bron, France (F.T.); and Institut des Neurosciences Cognitives, CNRS, Bron, France (F.T.)
| | | | | |
Collapse
|
39
|
Raaz U, Toh R, Maegdefessel L, Adam M, Nakagami F, Emrich FC, Spin JM, Tsao PS. Hemodynamic regulation of reactive oxygen species: implications for vascular diseases. Antioxid Redox Signal 2014; 20:914-28. [PMID: 23879326 PMCID: PMC3924901 DOI: 10.1089/ars.2013.5507] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
SIGNIFICANCE Arterial blood vessels functionally and structurally adapt to altering hemodynamic forces in order to accommodate changing needs and to provide stress homeostasis. This ability is achieved at the cellular level by converting mechanical stimulation into biochemical signals (i.e., mechanotransduction). Physiological mechanical stress helps maintain vascular structure and function, whereas pathologic or aberrant stress may impair cellular mechano-signaling, and initiate or augment cellular processes that drive disease. RECENT ADVANCES Reactive oxygen species (ROS) may represent an intriguing class of mechanically regulated second messengers. Chronically enhanced ROS generation may be induced by adverse mechanical stresses, and is associated with a multitude of vascular diseases. Although a causal relationship has clearly been demonstrated in large numbers of animal studies, an effective ROS-modulating therapy still remains to be established by clinical studies. CRITICAL ISSUES AND FUTURE DIRECTIONS This review article focuses on the role of various mechanical forces (in the form of laminar shear stress, oscillatory shear stress, or cyclic stretch) as modulators of ROS-driven signaling, and their subsequent effects on vascular biology and homeostasis, as well as on specific diseases such as arteriosclerosis, hypertension, and abdominal aortic aneurysms. Specifically, it highlights the significance of the various NADPH oxidase (NOX) isoforms as critical ROS generators in the vasculature. Directed targeting of defined components in the complex network of ROS (mechano-)signaling may represent a key for successful translation of experimental findings into clinical practice.
Collapse
Affiliation(s)
- Uwe Raaz
- 1 Division of Cardiovascular Medicine, Stanford University School of Medicine , Stanford, California
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Tada Y, Wada K, Shimada K, Makino H, Liang EI, Murakami S, Kudo M, Kitazato KT, Nagahiro S, Hashimoto T. Roles of hypertension in the rupture of intracranial aneurysms. Stroke 2013; 45:579-86. [PMID: 24370755 DOI: 10.1161/strokeaha.113.003072] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND AND PURPOSE Systemic hypertension has long been considered a risk factor of aneurysmal rupture. However, a causal link between systemic hypertension and the development of aneurysmal rupture has not been established. In this study, using a mouse model of intracranial aneurysm rupture, we examined the roles of systemic hypertension in the development of aneurysmal rupture. METHODS Aneurysms were induced by a combination of deoxycorticosterone acetate (DOCA)-salt and a single injection of elastase into the cerebrospinal fluid in mice. Antihypertensive treatment was started 6 days after aneurysm induction. Aneurysmal rupture was detected by neurological symptoms and confirmed by the presence of intracranial aneurysm with subarachnoid hemorrhage. Hydralazine (direct vasodilator) or discontinuation of DOCA-salt treatment was used to assess the roles of systemic hypertension. Captopril (angiotensin-converting enzyme inhibitor) or losartan (angiotensin II type 1 receptor antagonist) was used to assess the roles of the local renin-angiotensin system in the vascular wall. RESULTS Normalization of blood pressure by hydralazine significantly reduced the incidence of ruptured aneurysms and the rupture rate. There was a dose-dependent relationship between reduction of blood pressure and prevention of aneurysmal rupture. Captopril and losartan were able to reduce rupture rate without affecting systemic hypertension induced by DOCA-salt treatment. CONCLUSIONS Normalization of blood pressure after aneurysm formation prevented aneurysmal rupture in mice. In addition, we found that the inhibition of the local renin-angiotensin system independent from the reduction of blood pressure can prevent aneurysmal rupture.
Collapse
Affiliation(s)
- Yoshiteru Tada
- From the Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA (Y.T., K.W., K.S., H.M., E.I.L., S.M., M.K., T.H.); and Department of Neurosurgery, School of Medicine, University of Tokushima, Tokushima City, Japan (Y.T., K.T.K, S.N.)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
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
|
42
|
Affiliation(s)
- Nohra Chalouhi
- From the Department of Neurosurgery, Thomas Jefferson University, Philadelphia, PA (N.C.); Jefferson Hospital for Neuroscience, Philadelphia, PA (N.C.); Department of Neurosurgery, University of Florida, Gainesville (B.L.H.); and Department of Neurosurgery, Carver College of Medicine, University of Iowa, Iowa City (D.H.)
| | | | | |
Collapse
|
43
|
Wang H, Albadawi H, Siddiquee Z, Stone JM, Panchenko MP, Watkins MT, Stone JR. Altered vascular activation due to deficiency of the NADPH oxidase component p22phox. Cardiovasc Pathol 2013; 23:35-42. [PMID: 24035466 DOI: 10.1016/j.carpath.2013.08.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Revised: 08/04/2013] [Accepted: 08/05/2013] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Reactive oxygen species generated by nicotinamide adenine dinucleotide phosphate (NADPH) oxidase play important roles in vascular activation. The p22(phox) subunit is necessary for the activity of NADPH oxidase complexes utilizing Nox1, Nox2, Nox3, and Nox4 catalytic subunits. METHODS We assessed p22(phox)-deficient mice and human tissue for altered vascular activation. RESULTS Mice deficient in p22(phox) were smaller than their wild-type littermates but showed no alteration in basal blood pressure. The wild-type littermates were relatively resistant to forming intimal hyperplasia following carotid ligation, and the intimal hyperplasia that developed was not altered by p22(phox) deficiency. However, at the site of carotid artery ligation, the p22(phox)-deficient mice showed significantly less vascular elastic fiber loss compared with their wild-type littermates. This preservation of elastic fibers was associated with a reduced matrix metallopeptidase (MMP) 12/tissue inhibitor of metalloproteinase (TIMP) 1 expression ratio. A similar decrease in the relative MMP12/TIMP1 expression ratio occurred in human coronary artery smooth muscle cells upon knockdown of the hydrogen peroxide responsive kinase CK1αLS. In the ligated carotid arteries, the p22(phox)-deficient mice showed reduced expression of heterogeneous nuclear ribonucleoprotein C (hnRNP-C), suggesting reduced activity of CK1αLS. In a lung biopsy from a human patient with p22(phox) deficiency, there was also reduced vascular hnRNP-C expression. CONCLUSIONS These findings indicate that NADPH oxidase complexes modulate aspects of vascular activation including vascular elastic fiber loss, the MMP12/TIMP1 expression ratio, and the expression of hnRNP-C. Furthermore, these findings suggest that the effects of NADPH oxidase on vascular activation are mediated in part by protein kinase CK1αLS.
Collapse
MESH Headings
- Animals
- Carotid Artery Injuries/enzymology
- Carotid Artery Injuries/pathology
- Carotid Artery, Common/enzymology
- Carotid Artery, Common/pathology
- Case-Control Studies
- Casein Kinase Ialpha/genetics
- Casein Kinase Ialpha/metabolism
- Cells, Cultured
- Coronary Vessels/enzymology
- Coronary Vessels/pathology
- Cytochrome b Group/deficiency
- Cytochrome b Group/genetics
- Elastic Tissue/enzymology
- Elastic Tissue/pathology
- Female
- Granulomatous Disease, Chronic/enzymology
- Granulomatous Disease, Chronic/genetics
- Granulomatous Disease, Chronic/pathology
- Heterogeneous-Nuclear Ribonucleoprotein Group C/metabolism
- Humans
- Hyperplasia
- Infant
- Male
- Matrix Metalloproteinase 12/metabolism
- Mice
- Mice, Knockout
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- NADPH Oxidases/deficiency
- NADPH Oxidases/genetics
- Neointima
- RNA Interference
- Reactive Oxygen Species/metabolism
- Tissue Inhibitor of Metalloproteinase-1/metabolism
- Transfection
Collapse
Affiliation(s)
- He Wang
- Center for Systems Biology, Massachusetts General Hospital and Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Starke RM, Chalouhi N, Ali MS, Jabbour PM, Tjoumakaris SI, Gonzalez LF, Rosenwasser RH, Koch WJ, Dumont AS. The role of oxidative stress in cerebral aneurysm formation and rupture. Curr Neurovasc Res 2013; 10:247-55. [PMID: 23713738 PMCID: PMC3845363 DOI: 10.2174/15672026113109990003] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 04/17/2013] [Accepted: 05/08/2013] [Indexed: 02/07/2023]
Abstract
Oxidative stress is known to contribute to the progression of cerebrovascular disease. Additionally, oxidative stress may be increased by, but also augment inflammation, a key contributor to cerebral aneurysm development and rupture. Oxidative stress can induce important processes leading to cerebral aneurysm formation including direct endothelial injury as well as smooth muscle cell phenotypic switching to an inflammatory phenotype and ultimately apoptosis. Oxidative stress leads to recruitment and invasion of inflammatory cells through upregulation of chemotactic cytokines and adhesion molecules. Matrix metalloproteinases can be activated by free radicals leading to vessel wall remodeling and breakdown. Free radicals mediate lipid peroxidation leading to atherosclerosis and contribute to hemodynamic stress and hypertensive pathology, all integral elements of cerebral aneurysm development. Preliminary studies suggest that therapies targeted at oxidative stress may provide a future beneficial treatment for cerebral aneurysms, but further studies are indicated to define the role of free radicals in cerebral aneurysm formation and rupture. The goal of this review is to assess the role of oxidative stress in cerebral aneurysm pathogenesis.
Collapse
Affiliation(s)
- Robert M. Starke
- Joseph and Marie Field Cerebrovascular Research Laboratory, Division of Neurovascular & Endovascular Surgery, Department of Neurological Surgery, Thomas Jefferson University and Jefferson Hospital for Neuroscience, Philadelphia, Pennsylvania
- Department of Neurological Surgery, University of Virginia, Charlottesville, Virginia
| | - Nohra Chalouhi
- Joseph and Marie Field Cerebrovascular Research Laboratory, Division of Neurovascular & Endovascular Surgery, Department of Neurological Surgery, Thomas Jefferson University and Jefferson Hospital for Neuroscience, Philadelphia, Pennsylvania
| | - Muhammad S. Ali
- Joseph and Marie Field Cerebrovascular Research Laboratory, Division of Neurovascular & Endovascular Surgery, Department of Neurological Surgery, Thomas Jefferson University and Jefferson Hospital for Neuroscience, Philadelphia, Pennsylvania
| | - Pascal M. Jabbour
- Joseph and Marie Field Cerebrovascular Research Laboratory, Division of Neurovascular & Endovascular Surgery, Department of Neurological Surgery, Thomas Jefferson University and Jefferson Hospital for Neuroscience, Philadelphia, Pennsylvania
| | - Stavropoula I. Tjoumakaris
- Joseph and Marie Field Cerebrovascular Research Laboratory, Division of Neurovascular & Endovascular Surgery, Department of Neurological Surgery, Thomas Jefferson University and Jefferson Hospital for Neuroscience, Philadelphia, Pennsylvania
| | - L. Fernando Gonzalez
- Joseph and Marie Field Cerebrovascular Research Laboratory, Division of Neurovascular & Endovascular Surgery, Department of Neurological Surgery, Thomas Jefferson University and Jefferson Hospital for Neuroscience, Philadelphia, Pennsylvania
| | - Robert H. Rosenwasser
- Joseph and Marie Field Cerebrovascular Research Laboratory, Division of Neurovascular & Endovascular Surgery, Department of Neurological Surgery, Thomas Jefferson University and Jefferson Hospital for Neuroscience, Philadelphia, Pennsylvania
| | - Walter J. Koch
- Center for Translational Medicine and Department of Pharmacology, Temple University, Philadelphia, Pennsylvania USA
| | - Aaron S. Dumont
- Joseph and Marie Field Cerebrovascular Research Laboratory, Division of Neurovascular & Endovascular Surgery, Department of Neurological Surgery, Thomas Jefferson University and Jefferson Hospital for Neuroscience, Philadelphia, Pennsylvania
| |
Collapse
|
45
|
Hasan DM, Chalouhi N, Jabbour P, Dumont AS, Kung DK, Magnotta VA, Young WL, Hashimoto T, Richard Winn H, Heistad D. Evidence that acetylsalicylic acid attenuates inflammation in the walls of human cerebral aneurysms: preliminary results. J Am Heart Assoc 2013; 2:e000019. [PMID: 23525414 PMCID: PMC3603234 DOI: 10.1161/jaha.112.000019] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Background Inflammatory cells and molecules may play a critical role in formation and rupture of cerebral aneurysms. Recently, an epidemiologic study reported that acetylsalicylic acid (ASA) decreases the risk of aneurysm rupture. The goal of this study was to determine the effects of ASA on inflammatory cells and molecules in the walls of human cerebral aneurysms, using radiographic and histological techniques. Methods and Results Eleven prospectively enrolled patients harboring unruptured intracranial aneurysms were randomized into an ASA‐treated (81 mg daily) group (n=6) and an untreated (control) group (n=5). Aneurysms were imaged at baseline using ferumoxytol‐enhanced MRI to estimate uptake by macrophages. After 3 months, patients were reimaged before undergoing microsurgical clipping. Aneurysm tissues were collected for immunostaining with monoclonal antibodies for cyclooxygenase‐1 (COX‐1), cyclooxygenase‐2 (COX‐2), microsomal prostaglandin E2 synthase‐1 (mPGES‐1), and macrophages. A decrease in signal intensity on ferumoxytol‐enhanced MRI was observed after 3 months of ASA treatment. Expression of COX‐2 (but not COX‐1), mPGES‐1, and macrophages was lower in the ASA group than in the control group. Conclusions This study provides preliminary radiographical and histological evidence that ASA may attenuate the inflammatory process in the walls of human cerebral aneurysms. Clinical Trial Registration URL: http://www.clinicaltrials.gov. Unique identifier: NCT01710072.
Collapse
Affiliation(s)
- David M Hasan
- Department of Neurosurgery, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
|
47
|
Chalouhi N, Ali MS, Starke RM, Jabbour PM, Tjoumakaris SI, Gonzalez LF, Rosenwasser RH, Koch WJ, Dumont AS. Cigarette smoke and inflammation: role in cerebral aneurysm formation and rupture. Mediators Inflamm 2012; 2012:271582. [PMID: 23316103 PMCID: PMC3532877 DOI: 10.1155/2012/271582] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2012] [Accepted: 10/27/2012] [Indexed: 12/17/2022] Open
Abstract
Smoking is an established risk factor for subarachnoid hemorrhage yet the underlying mechanisms are largely unknown. Recent data has implicated a role of inflammation in the development of cerebral aneurysms. Inflammation accompanying cigarette smoke exposure may thus be a critical pathway underlying the development, progression, and rupture of cerebral aneurysms. Various constituents of the inflammatory response appear to be involved including adhesion molecules, cytokines, reactive oxygen species, leukocytes, matrix metalloproteinases, and vascular smooth muscle cells. Characterization of the molecular basis of the inflammatory response accompanying cigarette smoke exposure will provide a rational approach for future targeted therapy. In this paper, we review the current body of knowledge implicating cigarette smoke-induced inflammation in cerebral aneurysm formation/rupture and attempt to highlight important avenues for future investigation.
Collapse
Affiliation(s)
- Nohra Chalouhi
- Joseph and Marie Field Cerebrovascular Research Laboratory, Division of Neurovascular & Endovascular Surgery, Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Muhammad S. Ali
- Joseph and Marie Field Cerebrovascular Research Laboratory, Division of Neurovascular & Endovascular Surgery, Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Robert M. Starke
- Department of Neurological Surgery, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Pascal M. Jabbour
- Joseph and Marie Field Cerebrovascular Research Laboratory, Division of Neurovascular & Endovascular Surgery, Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Stavropoula I. Tjoumakaris
- Joseph and Marie Field Cerebrovascular Research Laboratory, Division of Neurovascular & Endovascular Surgery, Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - L. Fernando Gonzalez
- Joseph and Marie Field Cerebrovascular Research Laboratory, Division of Neurovascular & Endovascular Surgery, Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Robert H. Rosenwasser
- Joseph and Marie Field Cerebrovascular Research Laboratory, Division of Neurovascular & Endovascular Surgery, Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Walter J. Koch
- Center for Translational Medicine and George Zallie and Family Laboratory for Cardiovascular Gene Therapy, Department of Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Aaron S. Dumont
- Joseph and Marie Field Cerebrovascular Research Laboratory, Division of Neurovascular & Endovascular Surgery, Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Division of Neurovascular & Endovascular Surgery, Department of Neurological Surgery, Thomas Jefferson University, 901 Walnut Street, 3rd Floor, Philadelphia, PA 19107, USA
| |
Collapse
|
48
|
Lu Q, Wainwright MS, Harris VA, Aggarwal S, Hou Y, Rau T, Poulsen DJ, Black SM. Increased NADPH oxidase-derived superoxide is involved in the neuronal cell death induced by hypoxia-ischemia in neonatal hippocampal slice cultures. Free Radic Biol Med 2012; 53:1139-51. [PMID: 22728269 PMCID: PMC3527086 DOI: 10.1016/j.freeradbiomed.2012.06.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Revised: 06/07/2012] [Accepted: 06/08/2012] [Indexed: 11/25/2022]
Abstract
Neonatal brain hypoxia-ischemia (HI) results in neuronal cell death. Previous studies indicate that reactive oxygen species, such as superoxide, play a key role in this process. However, the cellular sources have not been established. In this study we examine the role of the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase complex in neonatal HI brain injury and elucidate its mechanism of activation. Rat hippocampal slices were exposed to oxygen glucose deprivation (OGD) to mimic the conditions seen in HI. Initial studies confirmed an important role for NADPH oxidase-derived superoxide in the oxidative stress associated with OGD. Further, the OGD-mediated increase in apoptotic cell death was inhibited by the NADPH oxidase inhibitor apocynin. The activation of NADPH oxidase was found to be dependent on the p38 mitogen-activated protein kinase-mediated phosphorylation and activation of the p47(phox) subunit. Using an adeno-associated virus antisense construct to selectively decrease p47(phox) expression in neurons showed that this led to inhibition of both the increase in superoxide and the neuronal cell death associated with OGD. We also found that NADPH oxidase inhibition in a neonatal rat model of HI or scavenging hydrogen peroxide reduced brain injury. Thus, we conclude that activation of the NADPH oxidase complex contributes to the oxidative stress during HI and that therapies targeted against this complex could provide neuroprotection against the brain injury associated with neonatal HI.
Collapse
Affiliation(s)
- Qing Lu
- Vascular Biology Center, Georgia Health Sciences University, Augusta, GA 30912, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
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
|
50
|
Nagao M, Moriyama Y, Yamagishi K, Iso H, Tamakoshi A. Relation of serum α- and γ-tocopherol levels to cardiovascular disease-related mortality among Japanese men and women. J Epidemiol 2012; 22:402-10. [PMID: 22672959 PMCID: PMC3798634 DOI: 10.2188/jea.je20120002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background There is limited evidence regarding the relationship between serum tocopherol levels and cardiovascular disease. Methods We conducted a nested case-control study as part of the Japan Collaborative Cohort Study for evaluation of cancer risk (JACC Study). Baseline serum samples were collected from 39 242 participants (age range, 40–79 years) between 1988 and 1990. During the 13-year follow-up, there were 530 stroke deaths (302 ischemic strokes and 210 hemorrhagic strokes) and 211 deaths from coronary heart disease. Controls were matched for sex, age, and area of residence. Results Serum α-tocopherol level was not associated with any type of cardiovascular death in men; however, in women, it was inversely associated with total stroke mortality and hemorrhagic stroke mortality. The multivariate odds ratio (95% CI) for the highest versus the lowest quintile of serum α-tocopherol levels among women was 0.35 (0.16–0.77; P for trend = 0.009) for total stroke and 0.26 (0.07–0.97; P for trend = 0.048) for hemorrhagic stroke. Serum γ-tocopherol was inversely associated with ischemic stroke mortality in men but positively associated with hemorrhagic stroke mortality in women. The respective multivariate odds ratios (95% CI) for the highest versus the lowest quintile and for a 1-standard deviation increment in γ-tocopherol level were 0.48 (0.22–1.06; P for trend = 0.07) and 0.77 (0.58–1.02), respectively, for ischemic stroke in men and 3.10 (0.95–10.12; P for trend = 0.052) and 1.49 (1.04–2.13) for hemorrhagic stroke in women. Conclusions Among women, hemorrhagic stroke mortality was inversely associated with serum α-tocopherol and positively associated with serum γ-tocopherol. These findings are due in part to the antioxidative and antithrombotic activities of these tocopherols.
Collapse
Affiliation(s)
- Masanori Nagao
- Public Health, Department of Social and Environmental Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | | | | | | | | | | |
Collapse
|