1
|
He J, Duan Y, Jiang Y, Luo J, Wang T, Liang R, Tang T. Phosphorylated NPY1R regulates phenotypic transition of vascular smooth muscle cells, inflammatory response and macrophage infiltration to promote intracranial aneurysm progression. Neuropeptides 2024; 108:102465. [PMID: 39353356 DOI: 10.1016/j.npep.2024.102465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 09/02/2024] [Accepted: 09/02/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Rupture of intracranial aneurysm (IA) could give rise to spontaneous subarachnoid hemorrhage, leading to a high disability rate and even death. NPY1R expression was upregulated in aneurysm tissues of IA patients. However, the role and underlying mechanism of NPY1R remains unknown. METHODS The IA model of mice was established using inducing systemic hypertension and injecting elastase. The expression of genes and proteins was detected by RT-qPCR and western blot. The number of T cells, macrophages, and neutrophils in IA mice was detected using flow cytometry and IF assay. The levels of inflammatory factors were measured using ELISA. Patho-morphology and inflammatory cells in aneurysm tissues were evaluated by HE staining. The interaction between TK and NPY1R was validated using Co-IP. RESULTS NPY1R expression was greatly elevated in aneurysm tissues in IA patients and mice, which were positively related to macrophage infiltration. Besides, exogenous overexpression of NPY1R resulted in the promotion of contractile phenotype to the synthetic phenotype of vascular smooth muscle cells (VSMCs), inflammatory response and M1 macrophage polarization. In terms of the underlying mechanism, NPY1R protein could be modified by TK-mediated phosphorylation and TKI could decrease IA formation and suppresse contractile phenotype to synthetic phenotype of VSMCs, inflammatory response and M1 macrophage polarization in IA mice. Furthermore, ablating mouse macrophages abolished NPY1R overexpression-mediated promotion of IA formation and rupture in mice. CONCLUSION Phosphorylated NPY1R contributed to IA progression through promoting contractile phenotype to synthetic phenotype of VSMCs, inflammatory response and M1 macrophage polarization in IA.
Collapse
Affiliation(s)
- Jian He
- The Second Affiliated Hospital, Department of Neurosurgery, Hengyang Medical School, University of South China, Hengyang City, Hunan Province, China
| | - Yonghong Duan
- The Second Affiliated Hospital, Department of Neurosurgery, Hengyang Medical School, University of South China, Hengyang City, Hunan Province, China
| | - Yuanding Jiang
- The Second Affiliated Hospital, Department of Neurosurgery, Hengyang Medical School, University of South China, Hengyang City, Hunan Province, China
| | - Jie Luo
- The Second Affiliated Hospital, Department of Neurosurgery, Hengyang Medical School, University of South China, Hengyang City, Hunan Province, China
| | - Tao Wang
- The Second Affiliated Hospital, Department of Neurosurgery, Hengyang Medical School, University of South China, Hengyang City, Hunan Province, China
| | - Richu Liang
- The Second Affiliated Hospital, Department of Neurosurgery, Hengyang Medical School, University of South China, Hengyang City, Hunan Province, China
| | - Ting Tang
- The Second Affiliated Hospital, Department of Teaching and Student Affairs, Hengyang Medical School, University of South China, Hengyang City, Hunan Province, China.
| |
Collapse
|
2
|
Wu J, Mei Y, Li X, Yu WK, Zhou ZH, Yang Y, Niu P, Wang Y, Shi CH, Zhu H, He W, Gao Y, Xu Y, Li Y. PRCP is a promising drug target for intracranial aneurysm rupture supported via multi-omics analysis. Stroke Vasc Neurol 2024:svn-2023-003076. [PMID: 39181687 DOI: 10.1136/svn-2023-003076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 07/30/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND Cerebral aneurysms are life-threatening cerebrovascular disorders. Currently, there are no effective treatments for preventing disease progression. Mendelian randomisation (MR) is widely used to repurify licensed drugs and identify new therapeutic targets. Therefore, this study aims to investigate effective drug targets for preventing the formation and rupture of cerebral aneurysms and analyse their potential mechanisms. METHODS We performed a comprehensive study integrating two-sample MR analysis, colocalisation analysis and summary data-based Mendelian randomisation (SMR) to assess the causal effects of blood and brain druggable cis-expression quantitative trait loci (cis-eQTLs) on intracranial aneurysm (IA), unruptured intracranial aneurysm (UIA) and subarachnoid haemorrhage of IA rupture (SAH). Druggable genes were obtained from the study by Chris Finan et al, cis-eQTLs from the eQTLGen and PsychENCODE consortia. Results were validated using proteomic and transcriptomic data. Single-gene functional analyses probed potential mechanisms, culminating in the construction of a drug-gene regulation network. RESULTS Through the MR analysis, we identified four potential drug targets in the blood, including prolylcarboxypeptidase (PRCP), proteasome 20S subunit alpha 4 (PSMA4), LTBP4 and GPR160 for SAH. Furthermore, two potential drug targets (PSMA4 and SLC22A4) were identified for IA and one potential drug target (KL) for UIA after accounting for multiple testing (P(inverse-variance weighted)<8.28e-6). Strong evidence of colocalisation and SMR analysis confirmed the relevance of PSMA4 and PRCP in outcomes. Elevated PRCP circulating proteins correlated with a lower SAH risk. PRCP gene expression was significantly downregulated in the disease cohort. CONCLUSIONS This study supports that elevated PRCP gene expression in blood is causally associated with the decreased risk of IA rupture. Conversely, increased PSMA4 expression in the blood is causally related to an increased risk of IA rupture and formation.
Collapse
Affiliation(s)
- Jinghao Wu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yunyun Mei
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai, Shanghai, China
| | - XinYu Li
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wen-Kai Yu
- Department of Neurology, Zhengzhou University, Zhengzhou, Henan, China
| | - Zi Han Zhou
- Reproductive Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yinghao Yang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Pengpeng Niu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yunchao Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chang-He Shi
- Department of Neurology, Zhengzhou University, Zhengzhou, Henan, China
| | - Hanghang Zhu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wenjun He
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yuan Gao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- NHC Key Laboratory of Prevention and treatment of Cerebrovascular Disease, zhengzhou, henan, People's Republic of China
- Henan Key Laboratory of Cerebrovascular Diseases, zhengzhou, henan, People's Republic of China
| | - Yusheng Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- NHC Key Laboratory of Prevention and treatment of Cerebrovascular Disease, zhengzhou, henan, People's Republic of China
- Henan Key Laboratory of Cerebrovascular Diseases, zhengzhou, henan, People's Republic of China
| |
Collapse
|
3
|
Xu Y, Guo P, Wang G, Sun X, Wang C, Li H, Cui Z, Zhang P, Feng Y. Integrated analysis of single-cell sequencing and machine learning identifies a signature based on monocyte/macrophage hub genes to analyze the intracranial aneurysm associated immune microenvironment. Front Immunol 2024; 15:1397475. [PMID: 38979407 PMCID: PMC11228246 DOI: 10.3389/fimmu.2024.1397475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 06/04/2024] [Indexed: 07/10/2024] Open
Abstract
Monocytes are pivotal immune cells in eliciting specific immune responses and can exert a significant impact on the progression, prognosis, and immunotherapy of intracranial aneurysms (IAs). The objective of this study was to identify monocyte/macrophage (Mo/MΦ)-associated gene signatures to elucidate their correlation with the pathogenesis and immune microenvironment of IAs, thereby offering potential avenues for targeted therapy against IAs. Single-cell RNA-sequencing (scRNA-seq) data of IAs were acquired from the Gene Expression Synthesis (GEO) database. The significant infiltration of monocyte subsets in the parietal tissue of IAs was identified using single-cell RNA sequencing and high-dimensional weighted gene co-expression network analysis (hdWGCNA). The integration of six machine learning algorithms identified four crucial genes linked to these Mo/MΦ. Subsequently, we developed a multilayer perceptron (MLP) neural model for the diagnosis of IAs (independent external test AUC=1.0, sensitivity =100%, specificity =100%). Furthermore, we employed the CIBERSORT method and MCP counter to establish the correlation between monocyte characteristics and immune cell infiltration as well as patient heterogeneity. Our findings offer valuable insights into the molecular characterization of monocyte infiltration in IAs, which plays a pivotal role in shaping the immune microenvironment of IAs. Recognizing this characterization is crucial for comprehending the limitations associated with targeted therapies for IAs. Ultimately, the results were verified by real-time fluorescence quantitative PCR and Immunohistochemistry.
Collapse
Affiliation(s)
- Yifan Xu
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Pin Guo
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Guipeng Wang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaojuan Sun
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chao Wang
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Huanting Li
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhenwen Cui
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Pining Zhang
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yugong Feng
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
4
|
Wang K, Xiao Y, Zhang W, Yang H, Li C, Wang J, Li G. Elucidating key immunological biomarkers and immune microenvironment dynamics in aging-related intracranial aneurysm through integrated multi-omics analysis. ENVIRONMENTAL TOXICOLOGY 2024; 39:2642-2654. [PMID: 38214030 DOI: 10.1002/tox.24117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/17/2023] [Accepted: 12/25/2023] [Indexed: 01/13/2024]
Abstract
BACKGROUND The exact cause of intracranial aneurysms (IA) is still unclear. However, pro-inflammatory factors are known to contribute to IA progression. The specific changes in the immune microenvironment of IAs remain largely unexplored. METHODS This study analyzed single-cell sequencing data from a male mouse model of brain aneurysm, focusing on samples before and after elastase-induced Willis aneurysms. The data helped identify eight distinct cell subpopulations: fibroblasts, macrophages, NK cells, endothelial cells, B cells, granulocytes, and monocytes. The study also involved bulk RNA sequencing of 97 IA samples, utilizing ssGSEA and CIBERSORT algorithms for analysis. Intercellular communication among these cells was inferred to understand the immune dynamics in IA. RESULTS The study found that fibroblasts and macrophages are predominant in various disease states of IA. Notably, the onset of IA was marked by a significant increase in fibroblasts and a decrease in macrophages. There was a marked increase in cellular interactions, especially involving macrophages, at the onset of the disease. Through enrichment analysis, 12 potential immunogenic biomarkers were identified. Of these, Rgs1 emerged as a critical molecule in IA formation, confirmed through secondary validation in a single-cell sequencing dataset. CONCLUSION This comprehensive analysis of immune cell composition and intercellular communication in IA tissues highlights the significant roles of macrophages and the molecule Rgs1. These findings shed light on the physiological and pathological conditions of IA, offering new insights into its immune microenvironment.
Collapse
Affiliation(s)
- Kai Wang
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yangyang Xiao
- Department of Clinical Laboratory, Binzhou Medical University Hospital, Binzhou, China
| | - Wenjia Zhang
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Haiguang Yang
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chaoqun Li
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jia Wang
- Department of Obstetrics and Gynecology, Shanghai Putuo District Liqun Hospital, Shanghai, China
| | - Guoshu Li
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
5
|
Ushio Y, Kataoka H, Akagawa H, Sato M, Manabe S, Kawachi K, Makabe S, Akihisa T, Seki M, Teraoka A, Iwasa N, Yoshida R, Tsuchiya K, Nitta K, Hoshino J, Mochizuki T. Factors associated with early-onset intracranial aneurysms in patients with autosomal dominant polycystic kidney disease. J Nephrol 2024; 37:983-992. [PMID: 38315279 DOI: 10.1007/s40620-023-01866-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 12/14/2023] [Indexed: 02/07/2024]
Abstract
BACKGROUND Recently, the importance of attribute-based medicine has been emphasized. The effects of early-onset intracranial aneurysms on patients can be significant and long-lasting. Herein, we compared the factors associated with intracranial aneurysms in patients with autosomal dominant polycystic kidney disease (ADPKD) according to age categories (≥ 50 years, < 50 years). METHODS We included 519 ADPKD patients, with a median age of 44 years, estimated glomerular filtration rate of 54.5 mL/min/1.73 m2, and total follow-up duration of 3104 patient-years. Logistic regression analyses were performed to determine factors associated with intracranial aneurysms. RESULTS Regarding the presence of intracranial aneurysm, significant interactions were identified between the age category (age ≥ 50 years), female sex (P = 0.0027 for the interaction) and hypertension (P = 0.0074 for the interaction). Female sex and hypertension were associated with intracranial aneurysm risk factors only in patients aged ≥ 50 years. The presence of intracranial aneurysm was significantly associated with chronic kidney disease (CKD) stages 4-5 (odds ratio [OR] = 3.87, P = 0.0007) and family history of intracranial aneurysm or subarachnoid hemorrhage (OR = 2.30, P = 0.0217) in patients aged < 50 years. For patients aged ≥ 50 years, in addition to the abovementioned factors [OR = 2.38, P = 0.0355 for CKD stages 4-5; OR = 3.49, P = 0.0094 for family history of intracranial aneurysm or subarachnoid hemorrhage], female sex (OR = 4.51, P = 0.0005), and hypertension (OR = 5.89, P = 0.0012) were also associated with intracranial aneurysm. CONCLUSION Kidney dysfunction and family history of intracranial aneurysm or subarachnoid hemorrhage are risk factors for early-onset intracranial aneurysm. Patients aged < 50 years with a family history of intracranial aneurysm or subarachnoid hemorrhage or with CKD stages 4-5 may be at an increased risk of early-onset intracranial aneurysm.
Collapse
Affiliation(s)
- Yusuke Ushio
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Hiroshi Kataoka
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-ku, Tokyo, 162-8666, Japan.
| | - Hiroyuki Akagawa
- Tokyo Women's Medical University Institute for Integrated Medical Sciences (TIIMS), Tokyo, Japan
| | - Masayo Sato
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Shun Manabe
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Keiko Kawachi
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Shiho Makabe
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Taro Akihisa
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Momoko Seki
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Atsuko Teraoka
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Naomi Iwasa
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Rie Yoshida
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Ken Tsuchiya
- Department of Blood Purification, Tokyo Women's Medical University, Tokyo, Japan
| | - Kosaku Nitta
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Junichi Hoshino
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Toshio Mochizuki
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| |
Collapse
|
6
|
Chen Y, Huang JH, Kang YB, Yao ZJ, Song JH. Bioinformatics analysis revealed the potential crosstalk genes and molecular mechanisms between intracranial aneurysms and periodontitis. BMC Med Genomics 2024; 17:114. [PMID: 38685029 PMCID: PMC11059758 DOI: 10.1186/s12920-024-01864-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 04/05/2024] [Indexed: 05/02/2024] Open
Abstract
OBJECTIVES The risk of intracranial aneurysms (IAs) development and rupture is significantly higher in patients with periodontitis (PD), suggesting an association between the two. However, the specific mechanisms of association between these two diseases have not been fully investigated. MATERIALS AND METHODS In this study, we downloaded IAs and PD data from the Gene Expression Omnibus. Differentially expressed genes (DEGs) were identified, and functional enrichment analysis was performed. The protein-protein interaction (PPI) network and weighted gene co-expression network analysis (WGCNA) was performed to identified key modules and key crosstalk genes. In addition, the immune cell landscape was assessed and the correlation of key crosstalk genes with each immune cell was calculated. Finally, transcription factors (TFs) regulating key crosstalk genes were explored. RESULTS 127 overlapping DEGs were identified and functional enrichment analysis highlighted the important role of immune reflection in the pathogenesis of IAs and PD. We identified ITGAX and COL4A2 as key crosstalk genes. In addition, the expression of multiple immune cells was significantly elevated in PDs and IAs compared to controls, and both key crosstalk genes were significantly negatively associated with Macrophages M2. Finally, GATA2 was identified as a potential key transcription factor (TF), which regulates two key crosstalk gene. CONCLUSIONS The present study identifies key crosstalk genes and TF in PD and IAs, providing new insights for further study of the co-pathogenesis of PD and IAs from an immune and inflammatory perspective. Also, this is the first study to report the above findings.
Collapse
Affiliation(s)
- Yao Chen
- Department of Neurosurgery, Affiliated Hospital of Putian University, Putian, Fujian Province, China
| | - Jian-Huang Huang
- Department of Neurosurgery, Affiliated Hospital of Putian University, Putian, Fujian Province, China.
| | - Yuan-Bao Kang
- Department of Neurosurgery, Affiliated Hospital of Putian University, Putian, Fujian Province, China
| | - Zheng-Jian Yao
- Department of Neurosurgery, Affiliated Hospital of Putian University, Putian, Fujian Province, China
| | - Jian-Hua Song
- Department of Neurosurgery, Affiliated Hospital of Putian University, Putian, Fujian Province, China
| |
Collapse
|
7
|
Khan D, Li X, Hashimoto T, Tanikawa R, Niemela M, Lawton M, Muhammad S. Current Mouse Models of Intracranial Aneurysms: Analysis of Pharmacological Agents Used to Induce Aneurysms and Their Impact on Translational Research. J Am Heart Assoc 2024; 13:e031811. [PMID: 38258667 PMCID: PMC11056163 DOI: 10.1161/jaha.123.031811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 12/06/2023] [Indexed: 01/24/2024]
Abstract
Intracranial aneurysms (IAs) are rare vascular lesions that are more frequently found in women. The pathophysiology behind the formation and growth of IAs is complex. Hence, to date, no single pharmacological option exists to treat them. Animal models, especially mouse models, represent a valuable tool to explore such complex scientific questions. Genetic modification in a mouse model of IAs, including deletion or overexpression of a particular gene, provides an excellent means for examining basic mechanisms behind disease pathophysiology and developing novel pharmacological approaches. All existing animal models need some pharmacological treatments, surgical interventions, or both to develop IAs, which is different from the spontaneous and natural development of aneurysms under the influence of the classical risk factors. The benefit of such animal models is the development of IAs in a limited time. However, clinical translation of the results is often challenging because of the artificial course of IA development and growth. Here, we summarize the continuous improvement in mouse models of IAs. Moreover, we discuss the pros and cons of existing mouse models of IAs and highlight the main translational roadblocks and how to improve them to increase the success of translational IA research.
Collapse
Affiliation(s)
- Dilaware Khan
- Department of NeurosurgeryMedical Faculty and University Hospital Düsseldorf, Heinrich‐Heine‐Universität DüsseldorfDüsseldorfGermany
| | - Xuanchen Li
- Department of NeurosurgeryMedical Faculty and University Hospital Düsseldorf, Heinrich‐Heine‐Universität DüsseldorfDüsseldorfGermany
| | - Tomoki Hashimoto
- Department of Neurosurgery and NeurobiologyBarrow Neurological InstitutePhoenixAZUSA
| | - Rokuya Tanikawa
- Department of Neurosurgery, Stroke CenterSapporo Teishinkai HospitalSapporoHokkaidoJapan
| | - Mika Niemela
- Department of NeurosurgeryUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
| | - Michael Lawton
- Department of Neurological SurgeryBarrow Neurological Institute, St. Joseph’s Hospital and Medical CenterPhoenixAZUSA
| | - Sajjad Muhammad
- Department of NeurosurgeryMedical Faculty and University Hospital Düsseldorf, Heinrich‐Heine‐Universität DüsseldorfDüsseldorfGermany
- Department of NeurosurgeryUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
| |
Collapse
|
8
|
Lauzier DC, Srienc AI, Vellimana AK, Dacey Jr RG, Zipfel GJ. Peripheral macrophages in the development and progression of structural cerebrovascular pathologies. J Cereb Blood Flow Metab 2024; 44:169-191. [PMID: 38000039 PMCID: PMC10993883 DOI: 10.1177/0271678x231217001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/10/2023] [Accepted: 09/15/2023] [Indexed: 11/26/2023]
Abstract
The human cerebrovascular system is responsible for maintaining neural function through oxygenation, nutrient supply, filtration of toxins, and additional specialized tasks. While the cerebrovascular system has resilience imparted by elaborate redundant collateral circulation from supportive tertiary structures, it is not infallible, and is susceptible to developing structural vascular abnormalities. The causes of this class of structural cerebrovascular diseases can be broadly categorized as 1) intrinsic developmental diseases resulting from genetic or other underlying aberrations (arteriovenous malformations and cavernous malformations) or 2) extrinsic acquired diseases that cause compensatory mechanisms to drive vascular remodeling (aneurysms and arteriovenous fistulae). Cerebrovascular diseases of both types pose significant risks to patients, in some cases leading to death or disability. The drivers of such diseases are extensive, yet inflammation is intimately tied to all of their progressions. Central to this inflammatory hypothesis is the role of peripheral macrophages; targeting this critical cell type may lead to diagnostic and therapeutic advancement in this area. Here, we comprehensively review the role that peripheral macrophages play in cerebrovascular pathogenesis, provide a schema through which macrophage behavior can be understood in cerebrovascular pathologies, and describe emerging diagnostic and therapeutic avenues in this area.
Collapse
Affiliation(s)
- David C Lauzier
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Anja I Srienc
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Ananth K Vellimana
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Ralph G Dacey Jr
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Gregory J Zipfel
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
9
|
Duan J, Zhao Q, He Z, Tang S, Duan J, Xing W. Current understanding of macrophages in intracranial aneurysm: relevant etiological manifestations, signaling modulation and therapeutic strategies. Front Immunol 2024; 14:1320098. [PMID: 38259443 PMCID: PMC10800944 DOI: 10.3389/fimmu.2023.1320098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
Macrophages activation and inflammatory response play crucial roles in intracranial aneurysm (IA) formation and progression. The outcome of ruptured IA is considerably poor, and the mechanisms that trigger IA progression and rupture remain to be clarified, thereby developing effective therapy to prevent subarachnoid hemorrhage (SAH) become difficult. Recently, climbing evidences have been expanding our understanding of the macrophages relevant IA pathogenesis, such as immune cells population, inflammatory activation, intra-/inter-cellular signaling transductions and drug administration responses. Crosstalk between macrophages disorder, inflammation and cellular signaling transduction aggravates the devastating consequences of IA. Illustrating the pros and cons mechanisms of macrophages in IA progression are expected to achieve more efficient treatment interventions. In this review, we summarized the current advanced knowledge of macrophages activation, infiltration, polarization and inflammatory responses in IA occurrence and development, as well as the most relevant NF-κB, signal transducer and activator of transcription 1 (STAT1) and Toll-Like Receptor 4 (TLR4) regulatory signaling modulation. The understanding of macrophages regulatory mechanisms is important for IA patients' clinical outcomes. Gaining insight into the macrophages regulation potentially contributes to more precise IA interventions and will also greatly facilitate the development of novel medical therapy.
Collapse
Affiliation(s)
- Jian Duan
- Department of Cerebrovascular Disease, Suining Central Hospital, Suining, Sichuan, China
| | - Qijie Zhao
- Department of Cerebrovascular Disease, Suining Central Hospital, Suining, Sichuan, China
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zeyuan He
- Department of Cerebrovascular Disease, Suining Central Hospital, Suining, Sichuan, China
| | - Shuang Tang
- Department of Cerebrovascular Disease, Suining Central Hospital, Suining, Sichuan, China
| | - Jia Duan
- Department of Cerebrovascular Disease, Suining Central Hospital, Suining, Sichuan, China
| | - Wenli Xing
- Department of Cerebrovascular Disease, Suining Central Hospital, Suining, Sichuan, China
| |
Collapse
|
10
|
Li Z, Huang J, Yang L, Li X, Li W. WNTA5-mediated miR-374a-5p regulates vascular smooth muscle cell phenotype transformation and M1 macrophage polarization impacting intracranial aneurysm progression. Sci Rep 2024; 14:559. [PMID: 38177414 PMCID: PMC10766994 DOI: 10.1038/s41598-024-51243-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 01/02/2024] [Indexed: 01/06/2024] Open
Abstract
miR-374a-5p expression and localization in intracranial aneurysm (IA) tissues were detected, and its correlation with vascular smooth muscle cells (VSMCs) and macrophage markers was analyzed. Using platelet-derived growth factor-BB (PDGF-BB) induced VSMC model, elastase-induced IA rat model. Subsequently, miR-374a-5p was knocked down or overexpressed. We investigated the effects of miR-374a-5p on phenotypic conversion, and in vivo experiments were also carried out to verify the findings. The targeted relationship between miR-374a-5p and WNTA5 was analyzed. The effect of WNT5A inhibition on VSMC phenotypic transformation and THP-1-derived macrophage polarization was explored. Clinical studies have shown that miR-374a-5p was upregulated in IA patients. miR-374a-5p was negatively correlated with SM22α, α-SMA, CD206, and positively correlated with CD86. In vitro experiments showed that knocking down miR-374a-5p reversed the promotion of SM22α and α-SMA expression by PDGF-BB, while overexpression of miR-374a-5p had the opposite effect. In addition, knocking down miR-374a-5p also reversed the decrease in Calponin, TIMP3, TIMP4, and IL-10 levels caused by PDGF-BB, and further reduced the levels of MMP1, MMP3, MMP9, IL-1β, IL-6, and TNF-α. These findings were further validated in vivo. In IA rats, there were notable increases in both systolic and diastolic blood pressure, along with an elevated M1/M2 ratio and the occurrence of vascular lesions. However, these symptoms were improved after knocking down miR-374a-5p. Furthermore, miR-374a-5p could target the WNT signals (WNT2B, WNT3, and WNT5A). miR-374a-5p regulated the VSMC phenotypic conversion and M1 macrophage polarization by targeting WNT5A, thereby impacting the progression of IA.
Collapse
Affiliation(s)
- Zengshi Li
- Department of Neurosurgery, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Junqiang Huang
- Department of Neurosurgery, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Lijian Yang
- Department of Neurosurgery, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Xi Li
- Department of Neurosurgery, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Wei Li
- Department of Neurosurgery, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, China.
| |
Collapse
|
11
|
Li W, Wu X, Wang J, Huang T, Zhou L, Zhou Y, Tan Y, Zhong W, Zhou Z. A novel clinical-radscore nomogram for predicting ruptured intracranial aneurysm. Heliyon 2023; 9:e20718. [PMID: 37842571 PMCID: PMC10570585 DOI: 10.1016/j.heliyon.2023.e20718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 10/02/2023] [Accepted: 10/04/2023] [Indexed: 10/17/2023] Open
Abstract
Objectives Our study aims to find the more practical and powerful method to predict intracranial aneurysm (IA) rupture through verification of predictive power of different models. Methods Clinical and imaging data of 576 patients with IAs including 192 ruptured IAs and matched 384 unruptured IAs was retrospectively analyzed. Radiomics features derived from computed tomography angiography (CTA) images were selected by t-test and Elastic-Net regression. A radiomics score (radscore) was developed based on the optimal radiomics features. Inflammatory markers were selected by multivariate regression. And then 4 models including the radscore, inflammatory, clinical and clinical-radscore models (C-R model) were built. The receiver operating characteristic curve (ROC) was performed to evaluate the performance of each model, PHASES and ELAPSS. The nomogram visualizing the C-R model was constructed to predict the risk of IA rupture. Results Five inflammatory features, 2 radiological characteristics and 7 radiomics features were significantly associated with IA rupture. The areas under ROCs of the radscore, inflammatory, clinical and C-R models were 0.814, 0.935, 0.970 and 0.975 in the training cohort and 0.805, 0.927, 0.952 and 0.962 in the validation cohort, respectively. Conclusion The inflammatory model performs particularly well in predicting the risk of IA rupture, and its predictive power is further improved by combining with radiological and radiomics features and the C-R model performs the best. The C-R nomogram is a more stable and effective tool than PHASES and ELAPSS for individually predicting the risk of rupture for patients with IA.
Collapse
Affiliation(s)
| | | | - Jing Wang
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Tianxing Huang
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Lu Zhou
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Yu Zhou
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Yuanxin Tan
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Weijia Zhong
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Zhiming Zhou
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| |
Collapse
|
12
|
Khan D, Cornelius JF, Muhammad S. The Role of NF-κB in Intracranial Aneurysm Pathogenesis: A Systematic Review. Int J Mol Sci 2023; 24:14218. [PMID: 37762520 PMCID: PMC10531594 DOI: 10.3390/ijms241814218] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/12/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Intracranial aneurysms (IAs) are abnormal dilations of the cerebral vessels, which pose a persistent threat of cerebral hemorrhage. Inflammation is known to contribute to IA development. The nuclear factor "kappa-light-chain-enhancer" of activated B-cells (NF-κB) is the major driver of inflammation. It increases the expression of inflammatory markers and matrix metalloproteinases (MMPs), which contribute heavily to the pathogenesis of IAs. NF-κB activation has been linked to IA rupture and resulting subarachnoid hemorrhage. Moreover, NF-κB activation can result in endothelial dysfunction, smooth muscle cell phenotypic switching, and infiltration of inflammatory cells in the arterial wall, which subsequently leads to the initiation and progression of IAs and consequently results in rupture. After a systematic search, abstract screening, and full-text screening, 30 research articles were included in the review. In this systematic review, we summarized the scientific literature reporting findings on NF-κB's role in the pathogenesis of IAs. In conclusion, the activation of the NF-κB pathway was associated with IA formation, progression, and rupture.
Collapse
Affiliation(s)
- Dilaware Khan
- Department of Neurosurgery, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany; (D.K.)
| | - Jan Frederick Cornelius
- Department of Neurosurgery, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany; (D.K.)
| | - Sajjad Muhammad
- Department of Neurosurgery, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany; (D.K.)
- Department of Neurosurgery, University Hospital Helsinki, Topeliuksenkatu 5, 00260 Helsinki, Finland
- Department of Neurosurgery, King Edward Medical University, Lahore 54000, Pakistan
| |
Collapse
|
13
|
Li S, Xiao J, Yu Z, Li J, Shang H, Zhang L. Integrated analysis of C3AR1 and CD163 associated with immune infiltration in intracranial aneurysms pathogenesis. Heliyon 2023; 9:e14470. [PMID: 36942257 PMCID: PMC10024113 DOI: 10.1016/j.heliyon.2023.e14470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/04/2023] [Accepted: 03/07/2023] [Indexed: 03/14/2023] Open
Abstract
Background To identify potential immune-related biomarkers, molecular mechanism, and therapeutic agents of intracranial aneurysms (IAs). Methods We identified the differentially expressed genes (DEGs) between IAs and control samples from GSE75436, GSE26969, GSE6551, and GSE13353 datasets. We used weighted gene co-expression network analysis (WGCNA) and protein-protein interaction (PPI) analysis to identify immune-related hub genes. We evaluated the expression of hub genes by using qRT-PCR analysis. Using miRNet, NetworkAnalyst, and DGIdb databases, we analyzed the regulatory networks and potential therapeutic agents targeting hub genes. Least absolute shrinkage and selection operator (LASSO) logistic regression was performed to identify optimal biomarkers among hub genes. The diagnostic value was validated by external GSE15629 dataset. Results We identified 227 DEGs and 22 differentially infiltrating immune cells between IAs and control samples from GSE75436, GSE26969, GSE6551, and GSE13353 datasets. We further identified 41 differentially expressed immune-related genes (DEIRGs), which were primarily enriched in the chemokine-mediated signaling pathway, myeloid leukocyte migration, endocytic vesicle membrane, chemokine receptor binding, chemokine activity, and viral protein interactions with cytokines and their receptors. Among 41 DEIRGs, 10 hub genes including C3AR1, CD163, CCL4, CXCL8, CCL3, TLR2, TYROBP, C1QB, FCGR3A, and FCGR1A were identified with good diagnostic values (AUC >0.7). Hsa-mir-27a-3p and transcription factors, including YY1 and GATA2, were identified the primary regulators of hub genes. 92 potential therapeutic agents targeting hub genes were predicted. C3AR1 and CD163 were finally identified as the best diagnostic biomarkers using LASSO logistic regression (AUC = 0.994). The diagnostic value of C3AR1 and CD163 was validated by the external GSE15629 dataset (AUC = 0.914). Conclusions This study revealed the importance of C3AR1 and CD163 in immune infiltration in IAs pathogenesis. Our finding provided a valuable reference for subsequent research on the potential targets for molecular mechanisms and intervention of IAs.
Collapse
Affiliation(s)
- Shengjie Li
- Nanchang University, Nanchang, China
- Department of Neurosurgery, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Corresponding author.
| | - Jinting Xiao
- Department of Medical Ultrasound, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Zaiyang Yu
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Junliang Li
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Hao Shang
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Lei Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| |
Collapse
|
14
|
Lu T, He Y, Liu Z, Ma C, Chen S, Jia R, Duan L, Guo C, Liu Y, Guo D, Li T, He Y. A machine learning-derived gene signature for assessing rupture risk and circulatory immunopathologic landscape in patients with intracranial aneurysms. Front Cardiovasc Med 2023; 10:1075584. [PMID: 36844725 PMCID: PMC9950511 DOI: 10.3389/fcvm.2023.1075584] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 01/30/2023] [Indexed: 02/12/2023] Open
Abstract
Background Intracranial aneurysm (IA) is an uncommon but severe subtype of cerebrovascular disease, with high mortality after aneurysm rupture. Current risk assessments are mainly based on clinical and imaging data. This study aimed to develop a molecular assay tool for optimizing the IA risk monitoring system. Methods Peripheral blood gene expression datasets obtained from the Gene Expression Omnibus were integrated into a discovery cohort. Weighted gene co-expression network analysis (WGCNA) and machine learning integrative approaches were utilized to construct a risk signature. QRT-PCR assay was performed to validate the model in an in-house cohort. Immunopathological features were estimated using bioinformatics methods. Results A four-gene machine learning-derived gene signature (MLDGS) was constructed for identifying patients with IA rupture. The AUC of MLDGS was 1.00 and 0.88 in discovery and validation cohorts, respectively. Calibration curve and decision curve analysis also confirmed the good performance of the MLDGS model. MLDGS was remarkably correlated with the circulating immunopathologic landscape. Higher MLDGS scores may represent higher abundance of innate immune cells, lower abundance of adaptive immune cells, and worse vascular stability. Conclusions The MLDGS provides a promising molecular assay panel for identifying patients with adverse immunopathological features and high risk of aneurysm rupture, contributing to advances in IA precision medicine.
Collapse
Affiliation(s)
- Taoyuan Lu
- Department of Cerebrovascular Disease and Neurosurgery, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, Henan, China,Henan International Joint Laboratory of Cerebrovascular Disease, Henan Provincial NeuroInterventional Engineering Research Center, Henan Engineering Research Center of Cerebrovascular Intervention Innovation, Zhengzhou, China
| | - Yanyan He
- Department of Cerebrovascular Disease and Neurosurgery, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, Henan, China,Henan International Joint Laboratory of Cerebrovascular Disease, Henan Provincial NeuroInterventional Engineering Research Center, Henan Engineering Research Center of Cerebrovascular Intervention Innovation, Zhengzhou, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chi Ma
- Department of Cerebrovascular Disease and Neurosurgery, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, Henan, China,Henan International Joint Laboratory of Cerebrovascular Disease, Henan Provincial NeuroInterventional Engineering Research Center, Henan Engineering Research Center of Cerebrovascular Intervention Innovation, Zhengzhou, China
| | - Song Chen
- Translational Research Institute, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
| | - Rufeng Jia
- Department of Cerebrovascular Disease and Neurosurgery, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, Henan, China,Henan International Joint Laboratory of Cerebrovascular Disease, Henan Provincial NeuroInterventional Engineering Research Center, Henan Engineering Research Center of Cerebrovascular Intervention Innovation, Zhengzhou, China
| | - Lin Duan
- Department of Cerebrovascular Disease and Neurosurgery, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, Henan, China,Henan International Joint Laboratory of Cerebrovascular Disease, Henan Provincial NeuroInterventional Engineering Research Center, Henan Engineering Research Center of Cerebrovascular Intervention Innovation, Zhengzhou, China
| | - Chunguang Guo
- Department of Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yiying Liu
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Dehua Guo
- Department of Cerebrovascular Disease and Neurosurgery, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, Henan, China,Henan International Joint Laboratory of Cerebrovascular Disease, Henan Provincial NeuroInterventional Engineering Research Center, Henan Engineering Research Center of Cerebrovascular Intervention Innovation, Zhengzhou, China
| | - Tianxiao Li
- Department of Cerebrovascular Disease and Neurosurgery, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, Henan, China,Henan International Joint Laboratory of Cerebrovascular Disease, Henan Provincial NeuroInterventional Engineering Research Center, Henan Engineering Research Center of Cerebrovascular Intervention Innovation, Zhengzhou, China,Tianxiao Li,
| | - Yingkun He
- Department of Cerebrovascular Disease and Neurosurgery, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, Henan, China,Henan International Joint Laboratory of Cerebrovascular Disease, Henan Provincial NeuroInterventional Engineering Research Center, Henan Engineering Research Center of Cerebrovascular Intervention Innovation, Zhengzhou, China,*Correspondence: Yingkun He,
| |
Collapse
|
15
|
Zhang Q, Liu H, Zhang M, Liu F, Liu T. Identification of co-expressed central genes and transcription factors in atherosclerosis-related intracranial aneurysm. Front Neurol 2023; 14:1055456. [PMID: 36937519 PMCID: PMC10017537 DOI: 10.3389/fneur.2023.1055456] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 02/06/2023] [Indexed: 03/06/2023] Open
Abstract
Background Numerous clinical studies have shown that atherosclerosis is one of the risk factors for intracranial aneurysms. Calcifications in the intracranial aneurysm walls are frequently correlated with atherosclerosis. However, the pathogenesis of atherosclerosis-related intracranial aneurysms remains unclear. This study aims to investigate this mechanism. Methods The Gene Expression Omnibus (GEO) database was used to download the gene expression profiles for atherosclerosis (GSE100927) and intracranial aneurysms (GSE75436). Following the identification of the common differentially expressed genes (DEGs) of atherosclerosis and intracranial aneurysm, the network creation of protein interactions, functional annotation, the identification of hub genes, and co-expression analysis were conducted. Thereafter, we predicted the transcription factors (TF) of hub genes and verified their expressions. Results A total of 270 common (62 downregulated and 208 upregulated) DEGs were identified for subsequent analysis. Functional analyses highlighted the significant role of phagocytosis, cytotoxicity, and T-cell receptor signaling pathways in this disease progression. Eight hub genes were identified and verified, namely, CCR5, FCGR3A, IL10RA, ITGAX, LCP2, PTPRC, TLR2, and TYROBP. Two TFs were also predicted and verified, which were IKZF1 and SPI1. Conclusion Intracranial aneurysms are correlated with atherosclerosis. We identified several hub genes for atherosclerosis-related intracranial aneurysms and explored the underlying pathogenesis. These discoveries may provide new insights for future experiments and clinical practice.
Collapse
Affiliation(s)
- Quan Zhang
- Department of Neurology, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hengfang Liu
- Department of Neurology, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- *Correspondence: Hengfang Liu
| | - Min Zhang
- Department of Neurology, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Fang Liu
- Department of Neurology, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Tiantian Liu
- Department of Neurology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
16
|
Okada A, Koseki H, Ono I, Kayahara T, Kurita H, Miyamoto S, Kataoka H, Aoki T. Identification of The Unique Subtype of Macrophages in Aneurysm Lesions at the Growth Phase. J Stroke Cerebrovasc Dis 2022; 31:106848. [DOI: 10.1016/j.jstrokecerebrovasdis.2022.106848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/05/2022] [Accepted: 10/18/2022] [Indexed: 11/05/2022] Open
|
17
|
Jin J, Duan J, Du L, Xing W, Peng X, Zhao Q. Inflammation and immune cell abnormalities in intracranial aneurysm subarachnoid hemorrhage (SAH): Relevant signaling pathways and therapeutic strategies. Front Immunol 2022; 13:1027756. [PMID: 36505409 PMCID: PMC9727248 DOI: 10.3389/fimmu.2022.1027756] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/31/2022] [Indexed: 11/25/2022] Open
Abstract
Intracranial aneurysm subarachnoid hemorrhage (SAH) is a cerebrovascular disorder associated with high overall mortality. Currently, the underlying mechanisms of pathological reaction after aneurysm rupture are still unclear, especially in the immune microenvironment, inflammation, and relevant signaling pathways. SAH-induced immune cell population alteration, immune inflammatory signaling pathway activation, and active substance generation are associated with pro-inflammatory cytokines, immunosuppression, and brain injury. Crosstalk between immune disorders and hyperactivation of inflammatory signals aggravated the devastating consequences of brain injury and cerebral vasospasm and increased the risk of infection. In this review, we discussed the role of inflammation and immune cell responses in the occurrence and development of aneurysm SAH, as well as the most relevant immune inflammatory signaling pathways [PI3K/Akt, extracellular signal-regulated kinase (ERK), hypoxia-inducible factor-1α (HIF-1α), STAT, SIRT, mammalian target of rapamycin (mTOR), NLRP3, TLR4/nuclear factor-κB (NF-κB), and Keap1/nuclear factor (erythroid-derived 2)-like 2 (Nrf2)/ARE cascades] and biomarkers in aneurysm SAH. In addition, we also summarized potential therapeutic drugs targeting the aneurysm SAH immune inflammatory responses, such as nimodipine, dexmedetomidine (DEX), fingolimod, and genomic variation-related aneurysm prophylactic agent sunitinib. The intervention of immune inflammatory responses and immune microenvironment significantly reduces the secondary brain injury, thereby improving the prognosis of patients admitted to SAH. Future studies should focus on exploring potential immune inflammatory mechanisms and developing additional therapeutic strategies for precise aneurysm SAH immune inflammatory regulation and genomic variants associated with aneurysm formation.
Collapse
Affiliation(s)
- Jing Jin
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, China,Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jian Duan
- Department of Cerebrovascular Disease, Suining Central Hospital, Suining, Sichuan, China
| | - Leiya Du
- 4Department of Oncology, The Second People Hospital of Yibin, Yibin, Sichuan, China
| | - Wenli Xing
- Department of Cerebrovascular Disease, Suining Central Hospital, Suining, Sichuan, China
| | - Xingchen Peng
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China,*Correspondence: Qijie Zhao, ; Xingchen Peng,
| | - Qijie Zhao
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, China,*Correspondence: Qijie Zhao, ; Xingchen Peng,
| |
Collapse
|
18
|
Moschetti G, Vasco C, Clemente F, Galeota E, Carbonara M, Pluderi M, Locatelli M, Stocchetti N, Abrignani S, Zanier ER, Ortolano F, Zoerle T, Geginat J. Deep Phenotyping of T-Cells Derived From the Aneurysm Wall in a Pediatric Case of Subarachnoid Hemorrhage. Front Immunol 2022; 13:866558. [PMID: 35711453 PMCID: PMC9197186 DOI: 10.3389/fimmu.2022.866558] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/28/2022] [Indexed: 11/13/2022] Open
Abstract
Intracranial aneurysms (IAs) are very rare in children, and the characteristics of the T-cells in the IA wall are largely unknown. A comatose 7-years-old child was admitted to our center because of a subarachnoid hemorrhage due to a ruptured giant aneurysm of the right middle cerebral artery. Two days after the aneurysm clipping the patient was fully awake with left hemiparesis. T-cells from the IA wall and from peripheral blood of this patient were analyzed by multi-dimensional flow cytometry. Unbiased analysis, based on the use of FlowSOM clustering and dimensionality reduction technique UMAP, indicated that there was virtually no overlap between circulating and tissue-infiltrating T-cells. Thus, naïve T-cells and canonical memory T-cells were largely restricted to peripheral blood, while CD4-CD8-T-cells were strongly enriched in the IA wall. The unique CD4+, CD8+ and CD4-CD8-T-cell clusters from the IA wall expressed high levels of CCR5, Granzyme B and CD69, displaying thus characteristics of cytotoxic and tissue-resident effector cells. Low Ki67 expression indicated that they were nevertheless in a resting state. Among regulatory T-cell subsets, Eomes+Tr1-like cells were strongly enriched in the IA wall. Finally, analysis of cytokine producing capacities unveiled that the IA wall contained poly-functional T-cells, which expressed predominantly IFN-γ, TNF and IL-2. CD4+T-cells co-expressed also CD40L, and produced some IL-17, GM-CSF and IL-10. This report provides to our knowledge the first detailed characterization of the human T-cell compartment in the IA wall.
Collapse
Affiliation(s)
| | - Chiara Vasco
- National Institute for Molecular Genetics (INGM), Milan, Italy
| | | | - Eugenia Galeota
- National Institute for Molecular Genetics (INGM), Milan, Italy
| | - Marco Carbonara
- Neuroscience Intensive Care Unit, Department of Anesthesia and Critical Care, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Mauro Pluderi
- Department of Neurosurgery, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marco Locatelli
- Department of Neurosurgery, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Nino Stocchetti
- Neuroscience Intensive Care Unit, Department of Anesthesia and Critical Care, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Sergio Abrignani
- National Institute for Molecular Genetics (INGM), Milan, Italy.,Department of Clinical Sciences and Community Health University Milan, Milan, Italy
| | - Elisa R Zanier
- Laboratory of Acute Brain Injury and Therapeutic Strategies, Department of Neuroscience, Mario Negri Institute for Pharmacological Research IRCCS, Milan, Italy
| | - Fabrizio Ortolano
- Neuroscience Intensive Care Unit, Department of Anesthesia and Critical Care, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Tommaso Zoerle
- Neuroscience Intensive Care Unit, Department of Anesthesia and Critical Care, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Jens Geginat
- National Institute for Molecular Genetics (INGM), Milan, Italy.,Department of Clinical Sciences and Community Health University Milan, Milan, Italy
| |
Collapse
|
19
|
Li Y, Zhang Z, Liu D. Intracranial Aneurysms Induced by RUNX1 Through Regulation of NFKB1 in Patients With Hypertension-An Integrated Analysis Based on Multiple Datasets and Algorithms. Front Neurol 2022; 13:877801. [PMID: 35655614 PMCID: PMC9152011 DOI: 10.3389/fneur.2022.877801] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/22/2022] [Indexed: 11/13/2022] Open
Abstract
Objective The purpose of this study was to identify potential therapeutic targets by examining the hub genes contributing to progression of intracranial aneurysm (IA) in patients with hypertension. Methods The bulk RNA sequencing (RNA-seq) datasets of hypertension and IA were obtained from the Gene Expression Omnibus (www.ncbi.nlm.nih.gov/geo) database. These data were then used to calculate disease-related differentially expressed genes (DEGs) at the individual level. An scRNA-seq dataset of patients with abdominal aortic aneurysms (AAA) was used to analyze monocyte/macrophage-related DEGs. On the basis of the DEG data related to monocytes and macrophages, a TF-genes network has been developed. Hub genes and core sub-networks have also been identified. Furthermore, the key genes have been validated in an external cohort. Results From combined monocyte and macrophage-derived DEGs from abdominal aortic aneurysms, five hub DEGs were detected, including IFI30, SERPINE1, HMOX1, IL24, and RUNX1. A total of 57 genes were found in the IA bulk RNA-seq dataset. A support vector machine-recursive feature elimination algorithm (SVM-RFE) was applied to further screen the seven genes (RPS4Y1, DDX3Y, RUNX1, CLEC10A, PLAC8, SLA, and LILRB3). RUNX1 was the hub gene that regulated NFKB1 in the monocyte/macrophage-related network. And RUNX1 is implicated in IA progression by regulating hematopoietic stem cell differentiation and abnormal platelet production, according to gene set enrichment analysis. Conclusion Among patients with hypertension, RUNX1 in monocytes and macrophages was associated with a higher risk of IA through its regulation of NFKB1.
Collapse
Affiliation(s)
- Yang Li
- Department of Neurosurgery, The First People's Hospital of Yinchuan, Yinchuan, China
| | - Zhen Zhang
- Department of Neurosurgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Donghua Liu
- Department of Neurosurgery, The Second People's Hospital of Yinchuan, Yinchuan, China
| |
Collapse
|
20
|
Lu T, Liu Z, Guo D, Ma C, Duan L, He Y, Jia R, Guo C, Xing Z, Liu Y, Li T, He Y. Transcriptome-Based Dissection of Intracranial Aneurysms Unveils an “Immuno-Thermal” Microenvironment and Defines a Pathological Feature-Derived Gene Signature for Risk Estimation. Front Immunol 2022; 13:878195. [PMID: 35711443 PMCID: PMC9194475 DOI: 10.3389/fimmu.2022.878195] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/19/2022] [Indexed: 12/16/2022] Open
Abstract
Immune inflammation plays an essential role in the formation and rupture of intracranial aneurysm (IA). However, the current limited knowledge of alterations in the immune microenvironment of IA has hampered the mastery of pathological mechanisms and technological advances, such as molecular diagnostic and coated stent-based molecular therapy. In this study, seven IA datasets were enrolled from the GEO database to decode the immune microenvironment and relevant biometric alterations. The ssGSEA algorithm was employed for immune infiltration assessment. IAs displayed abundant immune cell infiltration, activated immune-related pathways, and high expression of immune-related genes. Several immunosuppression cells and genes were also coordinately upregulated in IAs. Five immune-related hub genes, including CXCL10, IL6, IL10, STAT1, and VEGFA, were identified from the protein-protein interaction network and further detected at the protein level. CeRNA networks and latent drugs targeting the hub genes were predicted for targeted therapy reference. Two gene modules recognized via WCGNA were functionally associated with contractile smooth muscle loss and extracellular matrix metabolism, respectively. In blood datasets, a pathological feature-derived gene signature (PFDGS) for IA diagnosis and rupture risk prediction was established using machine learning. Patients with high PFDGS scores may possess adverse biological alterations and present with a high risk of morbidity or IA rupture, requiring more vigilance or prompt intervention. Overall, we systematically unveiled an “immuno-thermal” microenvironment characterized by co-enhanced immune activation and immunosuppression in IA, which provides a novel insight into molecular pathology. The PFDGS is a promising signature for optimizing risk surveillance and clinical decision-making in IA patients.
Collapse
Affiliation(s)
- Taoyuan Lu
- Department of Cerebrovascular Disease, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, China
- Henan Provincial NeuroInterventional Engineering Research Center, Henan International Joint Laboratory of Cerebrovascular Disease, and Henan Engineering Research Center of Cerebrovascular Intervention Innovation, Zhengzhou, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dehua Guo
- Department of Cerebrovascular Disease, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, China
- Henan Provincial NeuroInterventional Engineering Research Center, Henan International Joint Laboratory of Cerebrovascular Disease, and Henan Engineering Research Center of Cerebrovascular Intervention Innovation, Zhengzhou, China
| | - Chi Ma
- Department of Cerebrovascular Disease, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, China
- Henan Provincial NeuroInterventional Engineering Research Center, Henan International Joint Laboratory of Cerebrovascular Disease, and Henan Engineering Research Center of Cerebrovascular Intervention Innovation, Zhengzhou, China
| | - Lin Duan
- Henan Provincial NeuroInterventional Engineering Research Center, Henan International Joint Laboratory of Cerebrovascular Disease, and Henan Engineering Research Center of Cerebrovascular Intervention Innovation, Zhengzhou, China
- Department of Cerebrovascular Disease, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Yanyan He
- Department of Cerebrovascular Disease, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, China
- Henan Provincial NeuroInterventional Engineering Research Center, Henan International Joint Laboratory of Cerebrovascular Disease, and Henan Engineering Research Center of Cerebrovascular Intervention Innovation, Zhengzhou, China
- Department of Cerebrovascular Disease, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Rufeng Jia
- Department of Cerebrovascular Disease, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, China
- Henan Provincial NeuroInterventional Engineering Research Center, Henan International Joint Laboratory of Cerebrovascular Disease, and Henan Engineering Research Center of Cerebrovascular Intervention Innovation, Zhengzhou, China
| | - Chunguang Guo
- Department of Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhe Xing
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yiying Liu
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Tianxiao Li
- Department of Cerebrovascular Disease, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, China
- Henan Provincial NeuroInterventional Engineering Research Center, Henan International Joint Laboratory of Cerebrovascular Disease, and Henan Engineering Research Center of Cerebrovascular Intervention Innovation, Zhengzhou, China
- Department of Cerebrovascular Disease, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, China
- *Correspondence: Yingkun He, ; Tianxiao Li,
| | - Yingkun He
- Department of Cerebrovascular Disease, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, China
- Henan Provincial NeuroInterventional Engineering Research Center, Henan International Joint Laboratory of Cerebrovascular Disease, and Henan Engineering Research Center of Cerebrovascular Intervention Innovation, Zhengzhou, China
- Department of Cerebrovascular Disease, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, China
- *Correspondence: Yingkun He, ; Tianxiao Li,
| |
Collapse
|
21
|
lnc-MRGPRF-6:1 Promotes M1 Polarization of Macrophage and Inflammatory Response through the TLR4-MyD88-MAPK Pathway. Mediators Inflamm 2022; 2022:6979117. [PMID: 35125964 PMCID: PMC8816599 DOI: 10.1155/2022/6979117] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 12/02/2021] [Accepted: 12/21/2021] [Indexed: 11/17/2022] Open
Abstract
Background. Macrophage-mediated inflammation plays an essential role in the development of atherosclerosis (AS). Long noncoding RNAs (lncRNAs), as crucial regulators, participate in this process. We identified that lnc-MRGPRF-6:1 was significantly upregulated in the plasma exosomes of coronary atherosclerotic disease (CAD) patients in a preliminary work. In the present study, we aim to assess the role of lnc-MRGPRF-6:1 in macrophage-mediated inflammatory process of AS. Methods. The correlation between lnc-MRGPRF-6:1 and inflammatory factors was estimated firstly in plasma exosomes of CAD patients. Subsequently, we established lnc-MRGPRF-6:1 knockout macrophage model via the CRISPR/Cas9 system. We then investigated the regulatory effects of lnc-MRGPRF-6:1 on macrophage polarization and foam cell formation. Eventually, transcriptome analysis by RNA sequencing was carried out to explore the contribution of differential genes and signaling pathways in this process. Results. lnc-MRGPRF-6:1 was highly expressed in the plasma exosomes of CAD patients and was positively correlated with the expression of inflammatory cytokines in plasma. lnc-MRGPRF-6:1 inhibition significantly reduced the formation of foam cells. The expression of lnc-MRGPRF-6:1 was upregulated in M1 macrophage, and lnc-MRGPRF-6:1 knockout decreased the polarization of M1 macrophage. lnc-MRGPRF-6:1 regulates macrophage polarization via the TLR4-MyD88-MAPK signaling pathway. Conclusions. lnc-MRGPRF-6:1 knockdown can inhibit M1 polarization of macrophage and inflammatory response through the TLR4-MyD88-MAPK signaling pathway. lnc-MRGPRF-6:1 is a vital regulator in macrophage-mediated inflammatory process of AS.
Collapse
|
22
|
Chau SM, Herting SM, Noltensmeyer DA, Ahmed H, Maitland DJ, Raghavan S. Macrophage activation in response to shape memory polymer foam-coated aneurysm occlusion devices. J Biomed Mater Res B Appl Biomater 2022; 110:1535-1544. [PMID: 35090200 DOI: 10.1002/jbm.b.35015] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 12/17/2021] [Accepted: 01/19/2022] [Indexed: 01/30/2023]
Abstract
Brain aneurysms can be treated with embolic coils using minimally invasive approaches. It is advantageous to modulate the biologic response of platinum embolic coils. Our previous studies demonstrated that shape memory polymer (SMP) foam coated embolization coils (FCC) devices demonstrate enhanced healing responses in animal models compared with standard bare platinum coil (BPC) devices. Macrophages are the most prevalent immune cell type that coordinate the greater immune response to implanted materials. Hence, we hypothesized that the highly porous SMP foam coatings on embolic coils activate a pro-regenerative healing phenotype. To test this hypothesis, we analyzed the number and type of infiltrating macrophages in FCC or BPC devices implanted in a rabbit elastase aneurysm model. FCC devices elicited a great number of infiltration macrophages, skewed significantly to a pro-regenerative M2-like phenotype 90 days following implantation. We devised an in vitro assay, where monocyte-derived macrophages were placed in close association with FCC or BPC devices for 6-72 h. Macrophages encountering SMP FCC-devices demonstrated highly mixed activation phenotypes at 6 h, heavily skewing toward an M2-like phenotype by 72 h, compared with macrophages encountering BPC devices. Macrophage activation was evaluated using gene expression analysis, and secreted cytokine evaluation. Together, our results demonstrate that FCC devices promoted a pro-regenerative macrophage activation phenotype, compared with BPC devices. Our in vitro findings corroborate with in vivo observations that SMP-based modification of embolic coils can promote better healing of the aneurysm site, by sustaining a pro-healing macrophage phenotype.
Collapse
Affiliation(s)
- Sarah M Chau
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
- TAMU Master of Biotechnology Program, Texas A&M University, College Station, Texas, USA
| | - Scott M Herting
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Dillon A Noltensmeyer
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Hamzah Ahmed
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Duncan J Maitland
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Shreya Raghavan
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas, USA
| |
Collapse
|
23
|
Muhammad S, Hänggi D. Inflammation and Anti-Inflammatory Targets after Aneurysmal Subarachnoid Hemorrhage. Int J Mol Sci 2021; 22:ijms22147355. [PMID: 34298971 PMCID: PMC8304004 DOI: 10.3390/ijms22147355] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 07/05/2021] [Indexed: 12/26/2022] Open
|
24
|
Giotta Lucifero A, Baldoncini M, Brambilla I, Rutigliano M, Savioli G, Galzio R, Campero A, Lawton MT, Luzzi S. Gene Polymorphisms Increasing the Risk of Intracranial Aneurysms: Interleukin-6 -174G>C and -572G>C (Part II). ACTA BIO-MEDICA : ATENEI PARMENSIS 2021; 92:e2021420. [PMID: 35441611 PMCID: PMC9179066 DOI: 10.23750/abm.v92is4.12669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/27/2022] [Indexed: 11/25/2022]
Abstract
Introduction The interleukin-6 (IL-6), a proinflammatory cytokine, supports the adaptive immune response and regulates inflammatory processes. The -174 G>C and -572 G>C promoter polymorphisms of the IL-6 gene take part in the pathogenesis of intracranial aneurysms (IAs) and influence the clinical presentation of subarachnoid hemorrhage. This meta-analysis purposes to evaluate whether and which IL-6 allelic variations are related to a risk of IAs formation. Methods A PRISMA-based literature search was performed on the PubMed/Medline and Web of Science databases. The keywords used were "interleukin-6," "IL-6," "polymorphism," "interleukin-6 genotype," combined with "intracranial aneurysms" and "subarachnoid hemorrhage." Only human case-control studies, with a study (IAs) and a control group, written in English, and published in the last 15 years were selected. A meta-analysis was performed, estimating odds ratios and 95% confidence intervals in fixed- or random-effects models, as applicable. Statistical analysis was conducted with RevMan 5.0 software. Results 9 studies were eligible. No associations were found between -174 G>C polymorphisms and IAs susceptibility. Notable results were reported by the analysis of -572G>C polymorphisms. -572GG/GC/CC genotypes were strongly related to IAs occurrence with a statistical significance of p=0.03, p=0.0009, and p=0.00001, respectively. Conclusion A higher incidence of -572G>C promoter polymorphisms were demonstrated in the IAs group, highlighting the pivotal role of inflammatory genes in the natural history of brain aneurysms. Additional studies are required considering the racial heterogenicity and the need to widen the population sample.
Collapse
Affiliation(s)
- Alice Giotta Lucifero
- Neurosurgery Unit, Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Matias Baldoncini
- Department of Neurological Surgery, Hospital San Fernando, Buenos Aires, Argentina
| | - Ilaria Brambilla
- Pediatric Clinic, IRCCS Policlinico San Matteo Foundation, University of Pavia, Pavia, Italy
| | - Monica Rutigliano
- Department of Emergency and Organ Transplantation, Urology, Andrology and Kidney Transplantation Unit, University of Bari, Bari, Italy
| | - Gabriele Savioli
- Emergency Medicine and Surgery, IRCCS Fondazione Policlinico San Matteo, Pavia, Italy
| | - Renato Galzio
- Neurosurgery Unit, Maria Cecilia Hospital, Cotignola, Italy
| | - Alvaro Campero
- Servicio de Neurocirugia, Universidad Nacional de Tucuman, Argentina,Department of Neurosurgery, Hospital Padilla, San Miguel de Tucuman, Tucuman, Argentina
| | - Michael T. Lawton
- Department of Neurosurgery, Barrow Neurological Institute (BNI), United States
| | - Sabino Luzzi
- Neurosurgery Unit, Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy,Neurosurgery Unit, Department of Surgical Sciences, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| |
Collapse
|
25
|
Giotta Lucífero A, Baldoncini M, Foiadelli T, Brambilla I, Savioli G, Galzio R, Campero A, Lawton MT, Luzzi S. Gene Polymorphisms Increasing the Risk of Intracranial Aneurysms: Interleukin-1β -511C>T (Part I). ACTA BIO-MEDICA : ATENEI PARMENSIS 2021; 92:e2021419. [PMID: 35441612 PMCID: PMC9179052 DOI: 10.23750/abm.v92is4.12668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/27/2022] [Indexed: 11/22/2022]
Abstract
Introduction Intracranial aneurysms (IAs) are devastating cerebrovascular diseases with multifactorial etiology. The role of inflammation is indisputable, and interleukins are pivotal in supporting local inflammatory pathways and endothelial dysfunction at the aneurysm wall. In the light of insufficient evidence reported in the literature, this meta-analysis was aimed to investigate the genetic linkage between IL-1β (rs16944) -511C>T polymorphisms and IAs susceptibility. Methods A comprehensive online literature review was completed using the PubMed/Medline and Web of Science databases in accordance with the PRISMA guidelines. "Interleukin-1β," "IL-1β," "polymorphism," "intracranial aneurysm," and "subarachnoid hemorrhage" were the main keywords. Only human case-control studies, published from 2005 to 2021, written in English or translated, were screened. In the statistical analysis, we applied the fixed- and random-effect models, according to the level of heterogeneity, to assess the odds ratios (ORs) and 95% confidence intervals (CIs). RevMan 5.0 software was used for the statistics. Results Only 4 studies were eligible, with a total of 2070 patients, 1050 of which were assigned to the study group. Combined results showed a statistically significant association between the risk of IAs and -511CC (OR=0.79, 95% CI [0.65-0.95], p=0.01), and CT (OR=0.69, 95% CI [0.58-0.82], p<0.0001; OR=0.71, 95% CI [0.55-0.93], p=0.01) allele variations, both in the fixed- and random- models. No correlation was identified for the -511TT genotype (p=0.42; p=0.78). All the texts showed a low level of publication bias. Conclusion The present meta-analysis proved a potential role of IL-1β -511CC/CT genotypes in the pathogenesis of IAs. Additional studies are imperative to explain the underlying neuroimmune mechanisms, also allowing tailoring the potential inflammatory-target therapies for IAs.
Collapse
Affiliation(s)
- Alice Giotta Lucífero
- Neurosurgery Unit, Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Matias Baldoncini
- Department of Neurological Surgery, Hospital San Fernando, Buenos Aires, Argentina
| | - Thomas Foiadelli
- Pediatric Clinic, IRCCS Policlinico San Matteo Foundation, University of Pavia, Pavia, Italy
| | - Ilaria Brambilla
- Pediatric Clinic, IRCCS Policlinico San Matteo Foundation, University of Pavia, Pavia, Italy
| | - Gabriele Savioli
- Emergency Medicine and Surgery, IRCCS Fondazione Policlinico San Matteo, Pavia, Italy
| | - Renato Galzio
- Neurosurgery Unit, Maria Cecilia Hospital, Cotignola, Italy
| | - Alvaro Campero
- Servicio de Neurocirugía, Universidad Nacional de Tucumán; Argentina,Department of Neurosurgery, Hospital Padilla, San Miguel de Tucumán, Tucumán, Argentina
| | - Michael T. Lawton
- Department of Neurosurgery, Barrow Neurological Institute (BNI), United States
| | - Sabino Luzzi
- Neurosurgery Unit, Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy,Neurosurgery Unit, Department of Surgical Sciences, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| |
Collapse
|