Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 350
Filter
1.
Biochim Biophys Acta Mol Basis Dis ; 1870(6): 167278, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38834101

ABSTRACT

BACKGROUND: The dysfunction of human vascular smooth cells (hVSMCs) is significantly connected to the development of intracranial aneurysms (IAs). By suppressing the activity of microRNAs (miRNAs), circular RNAs (circRNAs) participate in IA pathogenesis. Nevertheless, the role of hsa_circ_0008571 in IAs remains unclear. METHODS: circRNA sequencing was used to identify circRNAs from human IA tissues. To determine the function of circ_0008571, Transwell, wound healing, and cell proliferation assays were conducted. To identify the target of circ_0008571, the analyses of CircInteractome and TargetScan, as well as the luciferase assay were carried out. Furthermore, circ_0008571 knockdown and over-expression were performed to investigate its functions in IA development and the underlying molecular mechanisms. RESULTS: Both hsa_circ_0008571 and Integrin beta 8 (ITGB8) were downregulated, while miR-145-5p transcription was elevated in the aneurysm wall of IAs patients compared to superficial temporal artery tissues. In vitro, cell migration and growth were dramatically suppressed after hsa_circ_0008571 overexpression. Mechanistically, has_circ_0008571 could suppress miR-145-5p activity by direct sponging. Moreover, we found that ITGB8 expression and the activation of the TGF-ß-mediated signaling pathway were significantly enhanced. CONCLUSION: The hsa_circ_0008571-miR-145-5p-ITGB8 axis plays an essential role in IA progression.


Subject(s)
Cell Proliferation , Intracranial Aneurysm , MicroRNAs , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , RNA, Circular , MicroRNAs/genetics , MicroRNAs/metabolism , Humans , Intracranial Aneurysm/genetics , Intracranial Aneurysm/pathology , Intracranial Aneurysm/metabolism , RNA, Circular/genetics , RNA, Circular/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Cell Proliferation/genetics , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Cell Movement/genetics , Phenotype , Male , Female , Middle Aged , Cells, Cultured , Integrin beta Chains
2.
OMICS ; 28(5): 234-245, 2024 May.
Article in English | MEDLINE | ID: mdl-38717843

ABSTRACT

Cerebral vasospasm (CV) is a significant complication following aneurysmal subarachnoid hemorrhage (aSAH), and lacks a comprehensive molecular understanding. Given the temporal trajectory of intracranial aneurysm (IA) formation, its rupture, and development of CV, altered gene expression might be a molecular substrate that runs through these clinical events, influencing both disease inception and progression. Utilizing RNA-Seq, we analyzed tissue samples from ruptured IAs with and without vasospasm to identify the dysregulated genes. In addition, temporal gene expression analysis was conducted. We identified seven dysregulated genes in patients with ruptured IA with vasospasm when compared with those without vasospasm. We found 192 common genes when the samples of each clinical subset of patients with IA, that is, unruptured aneurysm, ruptured aneurysm without vasospasm, and ruptured aneurysm with vasospasm, were compared with control samples. Among these common genes, TNFSF13B, PLAUR, OSM, and LAMB3 displayed temporal expression (progressive increase) with the pathological progression of disease that is formation of aneurysm, its rupture, and consequently the development of vasospasm. We validated the temporal gene expression pattern of OSM at both the transcript and protein levels and OSM emerges as a crucial gene implicated in the pathological progression of disease. In addition, RSAD2 and ATP1A2 appear to be pivotal genes for CV development. To the best of our knowledge, this is the first study to compare the transcriptome of aneurysmal tissue samples of aSAH patients with and without CV. The findings collectively provide new insights on the molecular basis of IA and CV and new leads for translational research.


Subject(s)
Gene Expression Profiling , Intracranial Aneurysm , Transcriptome , Vasospasm, Intracranial , Humans , Vasospasm, Intracranial/genetics , Vasospasm, Intracranial/metabolism , Intracranial Aneurysm/genetics , Intracranial Aneurysm/metabolism , Intracranial Aneurysm/complications , Transcriptome/genetics , Gene Expression Profiling/methods , Male , Female , Subarachnoid Hemorrhage/genetics , Subarachnoid Hemorrhage/complications , Subarachnoid Hemorrhage/metabolism , Gene Expression Regulation , Middle Aged , Aneurysm, Ruptured/genetics , Aneurysm, Ruptured/complications
3.
Aging (Albany NY) ; 16(9): 8320-8335, 2024 05 08.
Article in English | MEDLINE | ID: mdl-38728254

ABSTRACT

Exosomal long non-coding RNAs (LncRNAs) play a crucial role in the pathogenesis of cerebrovascular diseases. However, the expression profiles and functional significance of exosomal LncRNAs in intracranial aneurysms (IAs) remain poorly understood. Through high-throughput sequencing, we identified 1303 differentially expressed LncRNAs in the plasma exosomes of patients with IAs and healthy controls. Quantitative real-time polymerase chain reaction (qRT-PCR) verification confirmed the differential expression of LncRNAs, the majority of which aligned with the sequencing results. ATP1A1-AS1 showed the most significant upregulation in the disease group. Importantly, subsequent in vitro experiments validated that ATP1A1-AS1 overexpression induced a phenotype switching in vascular smooth muscle cells, along with promoting apoptosis and upregulating MMP-9 expression, potentially contributing to IAs formation. Furthermore, expanded-sample validation affirmed the high diagnostic value of ATP1A1-AS1. These findings suggest that ATP1A1-AS1 is a potential therapeutic target for inhibiting IAs progression and serves as a valuable clinical diagnostic marker.


Subject(s)
Apoptosis , Exosomes , Intracranial Aneurysm , Myocytes, Smooth Muscle , Phenotype , RNA, Long Noncoding , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Humans , Apoptosis/genetics , Intracranial Aneurysm/genetics , Intracranial Aneurysm/metabolism , Intracranial Aneurysm/pathology , Intracranial Aneurysm/blood , Exosomes/metabolism , Exosomes/genetics , Male , Myocytes, Smooth Muscle/metabolism , Middle Aged , Female , Sodium-Potassium-Exchanging ATPase/genetics , Sodium-Potassium-Exchanging ATPase/metabolism , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/genetics , Case-Control Studies
4.
J Biomed Sci ; 31(1): 51, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38741091

ABSTRACT

BACKGROUND: The fusiform aneurysm is a nonsaccular dilatation affecting the entire vessel wall over a short distance. Although PDGFRB somatic variants have been identified in fusiform intracranial aneurysms, the molecular and cellular mechanisms driving fusiform intracranial aneurysms due to PDGFRB somatic variants remain poorly understood. METHODS: In this study, single-cell sequencing and immunofluorescence were employed to investigate the phenotypic changes in smooth muscle cells within fusiform intracranial aneurysms. Whole-exome sequencing revealed the presence of PDGFRB gene mutations in fusiform intracranial aneurysms. Subsequent immunoprecipitation experiments further explored the functional alterations of these mutated PDGFRB proteins. For the common c.1684 mutation site of PDGFRß, we established mutant smooth muscle cell lines and zebrafish models. These models allowed us to simulate the effects of PDGFRB mutations. We explored the major downstream cellular pathways affected by PDGFRBY562D mutations and evaluated the potential therapeutic effects of Ruxolitinib. RESULTS: Single-cell sequencing of two fusiform intracranial aneurysms sample revealed downregulated smooth muscle cell markers and overexpression of inflammation-related markers in vascular smooth muscle cells, which was validated by immunofluorescence staining, indicating smooth muscle cell phenotype modulation is involved in fusiform aneurysm. Whole-exome sequencing was performed on seven intracranial aneurysms (six fusiform and one saccular) and PDGFRB somatic mutations were detected in four fusiform aneurysms. Laser microdissection and Sanger sequencing results indicated that the PDGFRB mutations were present in smooth muscle layer. For the c.1684 (chr5: 149505131) site mutation reported many times, further cell experiments showed that PDGFRBY562D mutations promoted inflammatory-related vascular smooth muscle cell phenotype and JAK-STAT pathway played a crucial role in the process. Notably, transfection of PDGFRBY562D in zebrafish embryos resulted in cerebral vascular anomalies. Ruxolitinib, the JAK inhibitor, could reversed the smooth muscle cells phenotype modulation in vitro and inhibit the vascular anomalies in zebrafish induced by PDGFRB mutation. CONCLUSION: Our findings suggested that PDGFRB somatic variants played a role in regulating smooth muscle cells phenotype modulation in fusiform aneurysms and offered a potential therapeutic option for fusiform aneurysms.


Subject(s)
Intracranial Aneurysm , Myocytes, Smooth Muscle , Receptor, Platelet-Derived Growth Factor beta , Animals , Female , Humans , Male , Intracranial Aneurysm/genetics , Intracranial Aneurysm/metabolism , Mutation , Myocytes, Smooth Muscle/metabolism , Phenotype , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism , Zebrafish/genetics
5.
Cell Mol Biol (Noisy-le-grand) ; 70(4): 152-157, 2024 Apr 28.
Article in English | MEDLINE | ID: mdl-38678612

ABSTRACT

The purpose of this study was to explore the effects of regulatory B-cells (Breg) on intracranial aneurysms by mediating IL-1ß/IL-1R pathways.  The study involved 60 patients undergoing angiography in a hospital from January to June 2022, divided into two groups: 30 with intracranial aneurysms (observation group) and 30 without (control group). Researchers extracted peripheral blood mononuclear cells (PBMC) to analyze the proportion of CD19+CD24hiCD38hiB cells using flow cytometry. These cells, along with T-cells and regulatory T-cells (Treg), were isolated through magnetic bead cell sorting. Following co-culture, the proliferation of T-cells and their related secretory factors were assessed. Additionally, Breg cells, treated with an IL-1R receptor blocker or IL-1R expression adenovirus, were studied to evaluate the levels of IL-10 and TGF-ß. In the study, the observation group showed lower levels of CD19+CD24hiCD38hiB cells, IL-10, and TGF-ß in PBMC than the control group (P<0.05). T-cell proportions were similar in both groups pre and post co-culture (P>0.05). Post co-culture, IFN-γ decreased while IL-4 increased in both groups. The observation group had higher IFN-γ and lower IL-4 than the control group (P<0.05). TNF-α in CD8+T cells, and granzyme B and perforin mRNA levels decreased post co-culture but were higher in the observation group (P<0.05). IL-10 and TGF-ß in Treg cells increased in both groups post co-culture but were lower in the observation group (P<0.05). The observation group also had fewer CD19+IL-1R+IL-10+B cells (P<0.05). After IL-1R blocker addition, IL-10 and TGF-ß in the supernatant decreased in the observation group (P<0.05). Following transfection, IL-1 and TGF-ß levels increased compared to the blank group (P<0.05). The function of peripheral blood CD19+CD24hiCD38hiB cells is impaired in patients with intracranial aneurysms, which may be related to IL-1ß/IL-1R pathways disorder.


Subject(s)
B-Lymphocytes, Regulatory , Interleukin-1beta , Intracranial Aneurysm , Receptors, Interleukin-1 , Female , Humans , Male , B-Lymphocytes, Regulatory/immunology , B-Lymphocytes, Regulatory/metabolism , Cell Proliferation , Coculture Techniques , Interleukin-10/metabolism , Interleukin-1beta/metabolism , Intracranial Aneurysm/immunology , Intracranial Aneurysm/pathology , Intracranial Aneurysm/metabolism , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/immunology , Receptors, Interleukin-1/metabolism , Receptors, Interleukin-1/genetics , Signal Transduction , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta/metabolism
6.
Acta Neuropathol Commun ; 12(1): 43, 2024 Mar 18.
Article in English | MEDLINE | ID: mdl-38500201

ABSTRACT

Intracerebral aneurysms (IAs) are pathological dilatations of cerebral arteries whose rupture leads to subarachnoid hemorrhage, a significant cause of disability and death. Inflammation is recognized as a critical contributor to the formation, growth, and rupture of IAs; however, its precise actors have not yet been fully elucidated. Here, we report CNS-associated macrophages (CAMs), also known as border-associated macrophages, as one of the key players in IA pathogenesis, acting as critical mediators of inflammatory processes related to IA ruptures. Using a new mouse model of middle cerebral artery (MCA) aneurysms we show that CAMs accumulate in the IA walls. This finding was confirmed in a human MCA aneurysm obtained after surgical clipping, together with other pathological characteristics found in the experimental model including morphological changes and inflammatory cell infiltration. In addition, in vivo longitudinal molecular MRI studies revealed vascular inflammation strongly associated with the aneurysm area, i.e., high expression of VCAM-1 and P-selectin adhesion molecules, which precedes and predicts the bleeding extent in the case of IA rupture. Specific CAM depletion by intracerebroventricular injection of clodronate liposomes prior to IA induction reduced IA formation and rupture rate. Moreover, the absence of CAMs ameliorated the outcome severity of IA ruptures resulting in smaller hemorrhages, accompanied by reduced neutrophil infiltration. Our data shed light on the unexplored role of CAMs as main actors orchestrating the progression of IAs towards a rupture-prone state.


Subject(s)
Aneurysm, Ruptured , Intracranial Aneurysm , Mice , Animals , Humans , Intracranial Aneurysm/etiology , Intracranial Aneurysm/metabolism , Intracranial Aneurysm/pathology , Inflammation/pathology , Central Nervous System/metabolism , Risk Factors , Macrophages/metabolism , Aneurysm, Ruptured/complications , Aneurysm, Ruptured/metabolism , Aneurysm, Ruptured/pathology
7.
CNS Neurosci Ther ; 30(2): e14603, 2024 02.
Article in English | MEDLINE | ID: mdl-38332649

ABSTRACT

INTRODUCTION: Genetic factors play a major part in mediating intracranial aneurysm (IA) rupture. However, research on the role of transcription factors (TFs) in IA rupture is rare. AIMS: Bioinformatics analysis was performed to explore the TFs and related functional pathways involved in IA rupture. RESULTS: A total of 63 differentially expressed transcription factors (DETFs) were obtained. Significantly enriched biological processes of these DETFs were related to regulation of myeloid leukocyte differentiation. The top 10 DETFs were screened based on the MCC algorithm from the protein-protein interaction network. After screening and validation, it was finally determined that CEBPB may be the hub gene for aneurysm rupture. The GSEA results of CEBPB were mainly associated with the inflammatory response, which was also verified by the experimental model of cellular inflammation in vitro. CONCLUSION: The inflammatory and immune response may be closely associated with aneurysm rupture. CEBPB may be the hub gene for aneurysm rupture and may have diagnostic value. Therefore, CEBPB may serve as the diagnostic signature for RIAs and a potential target for intervention.


Subject(s)
Aneurysm, Ruptured , Intracranial Aneurysm , Humans , Intracranial Aneurysm/genetics , Intracranial Aneurysm/metabolism , Gene Expression Regulation , Aneurysm, Ruptured/genetics , Aneurysm, Ruptured/metabolism , Immunity , Transcription Factors/genetics , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism
8.
Gene ; 908: 148253, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38341004

ABSTRACT

OBJECTIVE: This study endeavored to explore the relationship between exosome-derived lncRNA Double Homeobox A Pseudogene 8 (DUXAP8) and Chondroitin Polymerizing Factor 2 (CHPF2), and their roles in the pathogenesis of intracranial aneurysm (IA). METHODS: The shared targeted molecules (DUXAP8 and CHPF2) were detected via GSE122897 and GSE75436 datasets. A total of 312 patients with IAs were incorporated into this study. Exosomes were isolated from serum samples, and their identity was confirmed using Western blotting for exosomal markers (CD9, CD63 and ALIX). Inflammatory responses in IA tissues were evaluated using Hematoxylin-Eosin staining. CHPF2 protein concentration and the expression levels of DUXAP8 and CHPF2 mRNA in exosomal samples were assessed using Immunochemistry (IHC), Western Blotting, and qRT-PCR, respectively. Cell-based assays involving Human Umbilical Vein Endothelial Cells (HuvECs), including transfection with exosomal DUXAP8, Western Blotting, qRT-PCR, and Cell Counting Kit-8, were conducted. Receiver Operating Characteristic (ROC) curves were derived using SPSS. RESULTS: DUXAP8 level affects the level of CHPF2. DUXAP8 expression within exosomes was associated with increased CD9, CD63, ALIX and CHPF2 levels during IA development and inflammatory stress. In HuvECs, transfection with exosomes carrying DUXAP8 siRNA resulted in reduced CHPF2 expression, whereas DUXAP8 mimic increased CHPF2 concentrations. The Area Under the ROC Curve (AUC) for exosomal DUXAP8 expression and CHPF2 levels, and aneurysm size was 0.768 (95% CI, 0.613 to 0.924), 0.937 (95% CI, 0.853 to 1.000), and 0.943 (95% CI, 0.860, 1.000), respectively. CONCLUSION: Exosome-derived DUXAP8 promotes IA progression by affecting CHPF2 expression.


Subject(s)
Exosomes , Intracranial Aneurysm , N-Acetylgalactosaminyltransferases , RNA, Long Noncoding , Humans , Exosomes/genetics , Exosomes/metabolism , Genes, Homeobox , Human Umbilical Vein Endothelial Cells/metabolism , Intracranial Aneurysm/genetics , Intracranial Aneurysm/metabolism , MicroRNAs/metabolism , Pseudogenes , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , N-Acetylgalactosaminyltransferases/metabolism
9.
Sci Rep ; 14(1): 3941, 2024 02 16.
Article in English | MEDLINE | ID: mdl-38366068

ABSTRACT

The significant role of increased activation of 20S proteasomes in the development of abdominal aortic aneurysms has been well-established in a mouse model. The available literature lacks similar studies concerning brain aneurysms. The aim of the study was to verify the hypothesis that patients with unruptured intracranial aneurysms (UIA) have increased 20S proteasome ChT-L activity compared to the control group of individuals without vascular lesions in the brain. In the next step, the relationship between the activity of 20S proteasomes ChT-L and precursor proteins from the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) family, namely NF-κB1 (p105), NF-κB2 (p100), NF-κB p65, and the inflammatory chemokine MCP-1, was examined. Patients with UIA had significantly higher 20S ChT-L proteasome activity compared to the control group. Patients with multiple aneurysms had significantly higher 20S proteasome ChT-L activity compared to those with single aneurysms. In patients with UIA, the activity of the 20S proteasome ChT-L negatively correlated with the concentration of NF-κB1 (p105) and NF-κB p65 precursor proteins and positively correlated with the concentration of the cerebrospinal fluid chemokine MCP-1. Our results may suggest that increased 20S proteasome ChT-L activity in UIA patients modulates inflammation in the cerebral arterial vessel via the MCP-1 chemokine as a result of activation of the canonical NF-κB pathway.


Subject(s)
Intracranial Aneurysm , NF-kappa B , Mice , Animals , Humans , NF-kappa B/metabolism , Proteasome Endopeptidase Complex/metabolism , Intracranial Aneurysm/metabolism , Proteolysis , NF-kappa B p52 Subunit/metabolism
10.
Sci Rep ; 14(1): 559, 2024 01 04.
Article in English | MEDLINE | ID: mdl-38177414

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-1ß, 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.


Subject(s)
Intracranial Aneurysm , MicroRNAs , Humans , Rats , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Muscle, Smooth, Vascular/metabolism , Becaplermin/metabolism , Intracranial Aneurysm/genetics , Intracranial Aneurysm/metabolism , Macrophages/metabolism , Myocytes, Smooth Muscle/metabolism , Phenotype , Cell Proliferation/physiology
11.
Microvasc Res ; 152: 104643, 2024 03.
Article in English | MEDLINE | ID: mdl-38081409

ABSTRACT

OBJECTIVE: This research was dedicated to investigating the impact of the SNHG12/microRNA (miR)-15b-5p/MYLK axis on the modulation of vascular smooth muscle cell (VSMC) phenotype and the formation of intracranial aneurysm (IA). METHODS: SNHG12, miR-15b-5p and MYLK expression in IA tissue samples from IA patients were tested by RT-qPCR and western blot. Human aortic vascular smooth muscle cells (VSMCs) were cultivated with H2O2 to mimic IA-like conditions in vitro, and the cell proliferation and apoptosis were measured by MTT assay and Annexin V/PI staining. IA mouse models were established by induction with systemic hypertension combined with elastase injection. The blood pressure in the tail artery of mice in each group was assessed and the pathological changes in arterial tissues were observed by HE staining and TUNEL staining. The expression of TNF-α and IL-1ß, MCP-1, iNOS, caspase-3, and caspase-9 in the arterial tissues were tested by RT-qPCR and ELISA. The relationship among SNHG12, miR-15b-5p and MYLK was verified by bioinformatics, RIP, RNA pull-down, and luciferase reporter assays. RESULTS: The expression levels of MYLK and SNHG12 were down-regulated and that of miR-15b-5p was up-regulated in IA tissues and H2O2-treated human aortic VSMCs. Overexpressed MYLK or SNHG12 mitigated the decrease in proliferation and increase in apoptosis of VSMCs caused by H2O2 induction, and overexpression of miR-15b-5p exacerbated the decrease in proliferation and increase in apoptosis of VSMCs caused by H2O2 induction. Overexpression of miR-15b-5p reversed the H2O2-treated VSMC phenotypic changes caused by SNHG12 up-regulation, and overexpression of MYLK reversed the H2O2-treated VSMC phenotypic changes caused by up-regulation of miR-15b-5p. Overexpression of SNHG12 reduced blood pressure and ameliorated arterial histopathological damage and VSMC apoptosis in IA mice. The mechanical analysis uncovered that SNHG12 acted as an endogenous RNA that competed with miR-15b-5p, thus modulating the suppression of its endogenous target, MYLK. CONCLUSION: Decreased expression of SNHG12 in IA may contribute to the increasing VSMC apoptosis via increasing miR-15b-5p expression and subsequently decreasing MYLK expression. These findings provide potential new strategies for the clinical treatment of IA.


Subject(s)
Intracranial Aneurysm , MicroRNAs , Animals , Humans , Mice , Apoptosis , Calcium-Binding Proteins/genetics , Cell Proliferation , Hydrogen Peroxide/metabolism , Intracranial Aneurysm/genetics , Intracranial Aneurysm/metabolism , Intracranial Aneurysm/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Myosin-Light-Chain Kinase , Phenotype , RNA, Untranslated/genetics
12.
J Proteomics ; 293: 105060, 2024 02 20.
Article in English | MEDLINE | ID: mdl-38154549

ABSTRACT

Currently, there are no effective methods for predicting the rupture of asymptomatic small intracranial aneurysms (IA) (<7 mm). In this study the aim was to identify early warning biomarkers in peripheral plasma for predicting IA rupture. Four experimental groups were included: ruptured intracranial aneurysm (RIA), unruptured intracranial aneurysm (UIA), traumatic subarachnoid hemorrhage control (tSAHC), and healthy control (HC) groups. Plasma proteomics of these four groups were detected using iTRAQ combined LC-MS/MS. Differentially expressed proteins (DEPs) were identified in RIA, UIA, tSAHC compared with HC. Target proteins associated with aneurysm rupture were obtained by comparing the DEPs of the RIA and UIA groups after filtering out the DEPs of the tSAHC group. The plasma concentrations of target proteins were validated using enzyme-linked immunosorbent assay (ELISA). The iTRAQ analysis showed a significant increase in plasma GPC1 concentration in the RIA group compared to the UIA group, which was further validated among the IA patients. Logistic regression analysis identified GPC1 as an independent risk factor for predicting aneurysm rupture. The ROC curve indicated that the GPC1 plasma cut-off value for predicting aneurysms rupture was 4.99 ng/ml. GPC1 may be an early warning biomarker for predicting the rupture of small intracranial aneurysms. SIGNIFICANCE: The current management approach for asymptomatic small intracranial aneurysms (<7 mm) is limited to conservative observation and surgical intervention. However, the decision-making process regarding these options poses a dilemma due to weighing their respective advantages and disadvantages. Currently, there is a lack of effective diagnostic methods to predict the rupture of small aneurysms. Therefore, our aim is to identify early warning biomarkers in peripheral plasma that can serve as quantitative detection markers for predicting intracranial aneurysm rupture. In this study, four experimental populations were established: small ruptured intracranial aneurysm (sRIA) group, small unruptured intracranial aneurysm (sUIA) group, traumatic subarachnoid hemorrhage control (tSAHC) group, and healthy control (HC) group. The tSAH group was the control group of spontaneous subarachnoid hemorrhage caused by ruptured aneurysm. Compared with patients with UIA, aneurysm tissue and plasma GPC1 in patients with RIA is significantly higher, and GPC1 may be an early warning biomarker for predicting the rupture of intracranial small aneurysms.


Subject(s)
Aneurysm, Ruptured , Intracranial Aneurysm , Subarachnoid Hemorrhage, Traumatic , Humans , Aneurysm, Ruptured/diagnosis , Aneurysm, Ruptured/etiology , Biomarkers , Chromatography, Liquid , Glypicans , Intracranial Aneurysm/diagnosis , Intracranial Aneurysm/metabolism , Risk Factors , Subarachnoid Hemorrhage, Traumatic/complications , Tandem Mass Spectrometry
13.
J Neurosurg ; 140(6): 1745-1752, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38157535

ABSTRACT

OBJECTIVE: Aneurysm wall inflammation is associated with lesion instability in unruptured intracranial aneurysms (UIAs). However, most UIAs remain unruptured during lifelong follow-ups because of simultaneous protective remodeling against the inflammatory response. The protective effects of osteoprotegerin (OPG) in intracranial and abdominal aortic aneurysms have been suggested using rodent models; however, the role of this protein in UIAs in humans remains unclear. Herein, the authors examined the relationship between OPG expression and aneurysm wall integrity in intraoperatively resected UIAs by using immunohistochemical and immunofluorescence staining. METHODS: Sixteen UIA wall tissue specimens resected between 2017 and 2022 were analyzed. Aneurysm growth was defined as an enlargement > 1 mm or an obvious morphological change over the course of more than 6 months. Three high-power fields were randomly selected from areas expressing high and low levels of OPG within the same aneurysm. To clarify the role of OPG in the human aneurysm wall, the authors compared averaged values for the following pathological features between the 2 OPG expression groups: aneurysm wall thickness, collagen, macrophages, smooth muscle cells, and transforming growth factor beta 1 (TGF-ß1). Immunohistochemical staining within the entire tissue area was also analyzed to determine the relationships between OPG expression and different aneurysm growth patterns. Pathological findings were compared between high and low OPG expression levels using the Wilcoxon signed-rank test. RESULTS: The heterogeneous expression of OPG was detected in the walls of UIAs. Lesions expressing high OPG levels had thicker aneurysm walls (327 vs 180 µm, p = 0.002) and higher expression levels of TGF-ß1 (8.5% vs 5.4%, p = 0.002) than those expressing low OPG levels. The expression of TGF-ß1 was colocalized with that of OPG mainly in the tunica media. Furthermore, lesions expressing high OPG levels had larger α-SMA+ areas (25% vs 13%, p = 0.002). Aneurysm growth was observed in 6 of 9 UIAs with available data: whole sac expansion in 4 and secondary aneurysm formation in 2. Among the 6 UIAs with aneurysm growth, OPG expression was relatively higher in the UIAs with an internal elastic lamina than in those without (17% vs 6.9%). CONCLUSIONS: Aneurysm wall integrity was associated with OPG expression in the aneurysm wall. Collectively, the study results indicated that OPG is associated with protective remodeling, which may contribute to the retention of aneurysm wall structures.


Subject(s)
Intracranial Aneurysm , Osteoprotegerin , Transforming Growth Factor beta1 , Humans , Intracranial Aneurysm/metabolism , Intracranial Aneurysm/pathology , Intracranial Aneurysm/surgery , Osteoprotegerin/metabolism , Male , Middle Aged , Female , Aged , Transforming Growth Factor beta1/metabolism , Vascular Remodeling , Adult
14.
Redox Biol ; 67: 102887, 2023 11.
Article in English | MEDLINE | ID: mdl-37717465

ABSTRACT

Reactive Oxygen Species (ROS) are widely accepted as a pernicious factor in the progression of intracranial aneurysm (IA), which is eminently related to cell apoptosis and extracellular matrix degradation, but the mechanism remains to be elucidated. Recent evidence has identified that enhancement of Cyclophilin D (CypD) under stress conditions plays a critical role in ROS output, thus accelerating vascular destruction. However, no study has confirmed whether cypD is a detrimental mediator of cell apoptosis and extracellular matrix degradation in the setting of IA development. Our data indicated that endogenous cypD mRNA was significantly upregulated in human IA lesions and mouse IA wall, accompanied by higher level of ROS, MMPs and cell apoptosis. CypD-/- remarkably reversed vascular smooth muscle cells (VSMCs) apoptosis and elastic fiber degradation, and significantly decreased the incidence of aneurysm and ruptured aneurysm, together with the downregulation of ROS, 8-OHdG, NLRP3 and MMP9 in vivo and vitro. Furthermore, we demonstrated that blockade of cypD with CsA inhibited the above processes, thus preventing IA formation and rupture, these effects were highly dependent on ROS output. Mechanistically, we found that cypD directly interacts with ATP5B to promote ROS release in VSMCs, and 8-OHdG directly bind to NLRP3, which interacted with MMP9 to increased MMP9 level and activity in vivo and vitro. Our data expound an unexpected role of cypD in IA pathogenesis and an undescribed 8-OHdG/NLRP3/MMP9 pathway involved in accelerating VSMCs apoptosis and elastic fiber degradation. Repressing ROS output by CypD inhibition may be a promising therapeutic strategy for prevention IA development.


Subject(s)
Intracranial Aneurysm , NLR Family, Pyrin Domain-Containing 3 Protein , Animals , Humans , Mice , Peptidyl-Prolyl Isomerase F , Intracranial Aneurysm/genetics , Intracranial Aneurysm/metabolism , Matrix Metalloproteinase 9/genetics , Reactive Oxygen Species/metabolism
15.
Vascul Pharmacol ; 153: 107236, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37774964

ABSTRACT

Intracranial aneurysms (IAs) are characterized by abnormal dilatation of the cerebral vessels. Vascular smooth muscle cells (VSMCs) are implicated in maintaining vascular homeostasis. Disordered VSMCs are one of the most common causes for occurrence and development of IAs. The bone morphogenetic protein 4 (BMP4) signalling pathway is involved in regulating cell proliferation, apoptosis, and differentiation. This study aimed to investigate the effects of BMP4 on VSMCs and its underlying mechanisms. BMP4 was upregulated in the VSMCs of IAs and caused apoptosis of VSMCs through Smad1/5 phosphorylation. In addition, BMP4 overexpression significantly promoted the proliferation and migration of VSMCs and induced a phenotypic transformation from contractile to inflammatory. Our findings facilitate further understanding of the occurrence and development of IAs and provide a potential therapeutic target.


Subject(s)
Intracranial Aneurysm , Muscle, Smooth, Vascular , Humans , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein 4/pharmacology , Muscle, Smooth, Vascular/metabolism , Intracranial Aneurysm/metabolism , Signal Transduction , Cell Proliferation , Myocytes, Smooth Muscle/metabolism , Cells, Cultured
16.
Microvasc Res ; 149: 104554, 2023 09.
Article in English | MEDLINE | ID: mdl-37236346

ABSTRACT

Intracranial aneurysm (IA) is a severe cerebrovascular disease characterized by abnormal bulging of cerebral vessels that may rupture and cause a stroke. The expansion of the aneurysm accompanies by the remodeling of vascular matrix. It is well-known that vascular remodeling is a process of synthesis and degradation of extracellular matrix (ECM), which is highly dependent on the phenotype of vascular smooth muscle cells (VSMCs). The phenotypic switching of VSMC is considered to be bidirectional, including the physiological contractile phenotype and alternative synthetic phenotype in response to injury. There is increasing evidence indicating that VSMCs have the ability to switch to various phenotypes, including pro-inflammatory, macrophagic, osteogenic, foamy and mesenchymal phenotypes. Although the mechanisms of VSMC phenotype switching are still being explored, it is becoming clear that phenotype switching of VSMCs plays an essential role in IA formation, progression, and rupture. This review summarized the various phenotypes and functions of VSMCs associated with IA pathology. The possible influencing factors and potential molecular mechanisms of the VSMC phenotype switching were further discussed. Understanding how phenotype switching of VSMC contributed to the pathogenesis of unruptured IAs can bring new preventative and therapeutic strategies for IA.


Subject(s)
Intracranial Aneurysm , Muscle, Smooth, Vascular , Humans , Muscle, Smooth, Vascular/metabolism , Intracranial Aneurysm/genetics , Intracranial Aneurysm/metabolism , Intracranial Aneurysm/pathology , Signal Transduction , Myocytes, Smooth Muscle/pathology , Phenotype , Cells, Cultured , Cell Proliferation
17.
Biomed Pharmacother ; 161: 114480, 2023 May.
Article in English | MEDLINE | ID: mdl-37002575

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.


Subject(s)
Intracranial Aneurysm , Animals , Mice , Rats , Intracranial Aneurysm/metabolism , Muscle, Smooth, Vascular/metabolism , NADPH Oxidase 4/genetics , NADPH Oxidase 4/metabolism , NADPH Oxidases/metabolism , NFATC Transcription Factors/metabolism , Reactive Oxygen Species/metabolism , Transcription Factors/metabolism , TRPC Cation Channels/genetics , TRPC Cation Channels/metabolism , TRPC6 Cation Channel/metabolism
18.
BMC Mol Cell Biol ; 24(1): 3, 2023 Jan 30.
Article in English | MEDLINE | ID: mdl-36717793

ABSTRACT

BACKGROUND: Alterations in vascular smooth muscle cells (VSMCs) contribute to the pathogenesis of intracranial aneurysms (IAs). However, molecular mechanisms underlying these changes remain unknown. The present study aimed to characterize the molecular mechanisms underlying VSMC-mediated IAs. METHODS: Expression of the circular RNA circ-ATL1 and microRNA miR-455 was detected in IAs by RT-qPCR. Interactions between circ-ATL1, miR-455 and SIRT5 were examined by luciferase reporter analysis and RT-qPCR. The regulatory roles of circ-ATL1, miR-455 and SIRT5 in VSMC migration, proliferation and phenotypic modulation were also examined by CCK8, Transwell® migration and western blot assays. RESULTS: Biochemical and bioinformatic techniques were used to demonstrate that circ-ATL1 and miR-455 participated in disparate biological processes relevant to aneurysm formation. Clinically, increased expression of circ-ATL1 and downregulated miR-455 expression were observed in IA patients compared with healthy subjects. Silencing of circ-ATL1 led to suppression of VSMC migration, proliferation and phenotypic modulation. Both SIRT5 and miR-455 were found to be downstream targets of circ-ATL1. SIRT5 upregulation or miR-455 inhibition reversed the inhibitory effects induced by circ-ATL1 silencing on VSMC proliferation, migration and phenotypic modulation. We found that VSMC phenotypic modulation by circ-ATL1 upregulation and miR-455 downregulation had a critical role in the development and formation of AIs. Specifically, circ-ATL1 downregulation reversed IA formation. CONCLUSION: Our data provide the theoretical basis for future studies on potential clinical treatment and prevention of IAs.


Subject(s)
Biological Phenomena , Intracranial Aneurysm , MicroRNAs , RNA, Circular , Sirtuins , Humans , Cell Proliferation/genetics , Intracranial Aneurysm/genetics , Intracranial Aneurysm/metabolism , Intracranial Aneurysm/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , Muscle, Smooth, Vascular , Sirtuins/genetics , Sirtuins/metabolism , RNA, Circular/genetics
19.
BMC Pharmacol Toxicol ; 23(1): 81, 2022 10 23.
Article in English | MEDLINE | ID: mdl-36273189

ABSTRACT

Vascular smooth muscle cell (VSMC) phenotypic modulation regulates the initiation and progression of intracranial aneurysm (IA). Dexmedetomidine (DEX) is suggested to play neuroprotective roles in patients with craniocerebral injury. Therefore, we investigated the biological functions of DEX and its mechanisms against IA formation and progression in the current study. The rat primary VSMCs were isolated from Sprague-Dawley rats. IA and superficial temporal artery (STA) tissue samples were obtained from patients with IA. Flow cytometry was conducted to identify the characteristics of isolated VSMCs. Hydrogen peroxide (H2O2) was used to mimic IA-like conditions in vitro. Cell viability was detected using CCK-8 assays. Wound healing and Transwell assays were performed to detect cell motility. ROS production was determined by immunofluorescence using DCFH-DA probes. Western blotting and RT-qPCR were carried out to measure gene expression levels. Inflammation responses were determined by measuring inflammatory cytokines. Immunohistochemistry staining was conducted to measure α2-adrenergic receptor levels in tissue samples. DEX alleviated the H2O2-induced cytotoxicity, attenuated the promoting effects of H2O2 on cell malignancy, and protected VSMCs against H2O2-induced oxidative damage and inflammation response. DEX regulated the GSK-3ß/MKP-1/NRF2 pathway via the α2AR. DEX alleviates the inflammatory responses and oxidative damage of VSMCs by regulating the GSK-3ß/MKP-1/NRF2 pathway via the α2AR in IA.


Subject(s)
Dexmedetomidine , Intracranial Aneurysm , Rats , Animals , Glycogen Synthase Kinase 3 beta/metabolism , Glycogen Synthase Kinase 3 beta/pharmacology , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Muscle, Smooth, Vascular/metabolism , Dexmedetomidine/pharmacology , Dexmedetomidine/therapeutic use , Hydrogen Peroxide , Intracranial Aneurysm/drug therapy , Intracranial Aneurysm/genetics , Intracranial Aneurysm/metabolism , Rats, Sprague-Dawley , Sincalide/metabolism , Sincalide/pharmacology , Reactive Oxygen Species/metabolism , Oxidative Stress , Receptors, Adrenergic, alpha-2 , Inflammation/drug therapy , Inflammation/metabolism , Cytokines/metabolism
20.
Acta Biochim Pol ; 69(3): 613-618, 2022 Sep 13.
Article in English | MEDLINE | ID: mdl-36099587

ABSTRACT

OBJECTIVES: Cerebral aneurysm (CA) is one of the most common cerebrovascular diseases. The study was conducted to investigate the effect of resveratrol (RES) on the CA formation and its possible mechanisms. MATERIALS AND METHODS: Murine model of CA was constructed by induced hypertension and fed without (model group) or with RES (RES group). A Sham group was used as a control. The CA formation and inflammatory response were examined morphologically and histochemically. The expression of nuclear factor-κB (NF-κB), matrix metalloproteinase (MMP)-2, and MMP-9 was analyzed using qRT-PCR and Western blots. RESULTS: CA was induced in mice after the left common carotid artery was ligated and fed with high sodium chloride. Compared with the model, mice fed with RES had significantly fewer CA with smaller size, normal thickness of the arterial wall (P<0.05), and fewer infiltrated macrophages in the aneurysm wall (P<0.05). qRT-PCR and Western blot analyses showed that the expression of MMP-2, MMP-9 and NF-κB was significantly elevated in the model as compared with the control and significantly decreased after RES treatments (P<0.05). CONCLUSIONS: RES can inhibit the CA formation in mice subjected to induced hypertension and this inhibition is likely mediated via downregulating the NF-κB pathway.


Subject(s)
Intracranial Aneurysm , Resveratrol , Animals , Hypertension/chemically induced , Intracranial Aneurysm/drug therapy , Intracranial Aneurysm/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , NF-kappa B/metabolism , Resveratrol/pharmacology , Resveratrol/therapeutic use , Sodium Chloride
SELECTION OF CITATIONS
SEARCH DETAIL
...