Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 115
Filter
1.
Biomed Pharmacother ; 156: 113765, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36228368

ABSTRACT

BACKGROUND: Severe heart failure refractory to conventional therapy requires alternative treatment modalities. Surgical ventricular reconstruction (SVR) has been used to reverse cardiac remodeling in post-myocardial infarction (MI) patients with large left ventricular (LV) aneurysm, however, residual LV remodeling and dysfunction remain postoperatively. It is unclear whether SVR recovers response to drug treatment and whether the sodium-glucose co-transporter 2 inhibitor dapagliflozin (DAPA) reverses residual LV remodeling after SVR. METHODS: Adult male C57 mice were subjected to MI or sham surgery. Four-week later, MI mice with LV aneurysm underwent modified SVR or second open-chest sham operation and were randomized to DAPA or vehicle for four-week. Cardiac remodeling, LV function, and the underlying mechanisms were evaluated by echocardiography, invasive LV hemodynamic measurements, mRNA sequencing, and bioinformatics analysis. RESULTS: SVR significantly decreased LV volume; increased myocardial strain, LV pressure change rates and end-systolic elastance; and decreased heart-to-body weight ratio and myocardial fibrosis. However, significant residual cardiac remodeling remained. DAPA significantly attenuated residual cardiac remodeling and improved LV function in SVR mice but did not have curative effects in non-SVR mice. Of the 1532 genes differentially expressed in SVR and MI mice, 1037 were associated with cardiac metabolism; Src, Crebbp, Fn1, Grb2, and Mapk14 were the top 5 hub genes. Unlike sham surgery, MI upregulated those 5 genes, and treatment with SVR + DAPA normalized their expression. CONCLUSIONS: SVR restores therapeutic response in the post-MI heart with large LV aneurysm, and DAPA attenuates residual cardiac remodeling after SVR by normalizing some cardiac metabolism-related hub genes.


Subject(s)
Aneurysm , Myocardial Infarction , Sodium-Glucose Transporter 2 Inhibitors , Animals , Male , Mice , Aneurysm/complications , Aneurysm/metabolism , Cardiomegaly/metabolism , Myocardium/metabolism , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Ventricular Remodeling
2.
Cytokine ; 158: 155969, 2022 10.
Article in English | MEDLINE | ID: mdl-35930963

ABSTRACT

BACKGROUND: The study explored the role of platelet TGF-ß1 from the perspective of inhibiting the excessive proliferation, migration and invasion of murine aortic vascular smooth muscle cells (MASMCs). METHOD: The platelets were first extracted from C57BL/6 mice, and the TGF-ß1 protein was obtained after the purification of protein. In vitro, the concentrations of angiotensin Ⅱ (Ang Ⅱ) and TGF-ß1 for intervention were screened by testing the viability of MASMCs, followed by the analysis concerning the effects of platelets, Ang Ⅱ and TGF-ß1 on the proliferation, migration, invasion, and the expressions of pathway-related proteins in MASMCs. In vivo, an Ang Ⅱ-induced mouse model was established. TGF-ß1 was injected into the tail of mice as a therapeutic agent, and its action mechanism was further verified by the treatment of inhibitor SB505124. The results of the cell experiment were validated by evaluating the maximum diameter of abdominal aorta, the proportion of total weight, the changes of both pathology and the expressions of pathway-related proteins in the mice. RESULT: 0.5 ng/mL Ang Ⅱ and 15 ng/mL TGF-ß1 were chosen for treatment. The following results of cell functional experiments and Western blot assay demonstrated that Ang Ⅱ promoted the proliferation, migration and invasion of MASMCs via regulating related pathways, the effects of which were evidently reversed by TGF-ß1 and platelets. Consistent results were also observed in the animal experiments, where TGF-ß1 effectively alleviated Ang Ⅱ-induced abdominal aortic injury in mice. CONCLUSION: TGF-ß1 in platelets inhibits Ang Ⅱ-induced proliferation, migration and invasion of MASMCs.


Subject(s)
Aneurysm , Transforming Growth Factor beta1 , Aneurysm/metabolism , Angiotensin II/metabolism , Angiotensin II/pharmacology , Animals , Aorta, Abdominal/metabolism , Blood Platelets/metabolism , Cell Proliferation , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Transforming Growth Factor beta1/metabolism
3.
Hum Exp Toxicol ; 40(12_suppl): S563-S572, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34796763

ABSTRACT

BACKGROUND: Death-associated protein kinase (DAPK1) is one of the positive regulators of apoptosis, and it is widely involved in apoptosis induced by multiple pathways. We examined that the function of DAPK1 in Clinical treatment of arterial aneurysm and its underlying mechanisms. Arterial aneurysm is a common cerebrovascular disease with high disability and fatality rate. OBJECTIVES: Male C57BL/6 mice or DAPK1-/- mice were injected with 50 mg/kg pentobarbital sodium and then were injected with angiotensin II (AngII) infusion for vivo model. hASMCs (Human artery smooth muscle cell) were treated with murine recombinant IL-6 (20  ng ml-1; Cell Signaling) for vitro model. RESULTS: DAPK1 gene, mRNA expression, and protein expression were induced in mice of arterial aneurysm. DAPK1 mRNA expression was increased and Area Under Curve was 0.9075 in patients with arterial aneurysm. Knockout of DAPK1 decreased inflammation and vascular injury in mice model of arterial aneurysm. Beclin1/NLRP3 (NACHT, LRR, and PYD domains-containing protein 3) signal pathway is a critical downstream effector of DAPK1 by TAP production. The regulation of Beclin1 participated in the effects of DAPK1 on inflammation of arterial aneurysm by ATP-dependent NLRP3 inflammasome. The regulation of NLRP3 participated in the effects of DAPK1 on inflammation of arterial aneurysm. CONCLUSION: Collectively, our data indicated that DAPK1 may be a potential biomarker for arterial aneurysm in clinical treatment and activated inflammation levels in arterial aneurysm through NLRP3 inflammasome by Beclin1. DAPK1 might be a key pathogenic event underlying excess inflammation of arterial aneurysm.


Subject(s)
Aneurysm/metabolism , Beclin-1/metabolism , Death-Associated Protein Kinases/metabolism , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Adult , Animals , Biomarkers/metabolism , Death-Associated Protein Kinases/genetics , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged
4.
J Neuropathol Exp Neurol ; 80(10): 966-974, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34534311

ABSTRACT

Saccular intracranial aneurysm (sIA) rupture leads to a disabling subarachnoid hemorrhage. Chronic inflammation and lipid accumulation in the sIA wall contribute to wall degenerative remodeling that precedes its rupture. A better understanding of the pathobiological process is essential for improved future treatment of patients carrying sIAs. Serum amyloid A (SAA) is an acute-phase protein produced in response to acute and chronic inflammation and tissue damage. Here, we studied the presence and the potential role of SAA in 36 intraoperatively resected sIAs (16 unruptured and 20 ruptured), that had previously been studied by histology and immunohistochemistry. SAA was present in all sIAs, but the extent of immunopositivity varied greatly. SAA immunopositivity correlated with wall degeneration (p = 0.028) and rupture (p = 0.004), with numbers of CD163-positive and CD68-positive macrophages and CD3-positive T lymphocytes (all p < 0.001), and with the expression of myeloperoxidase, matrix metalloproteinase-9, prostaglandin E-2 receptor, and cyclo-oxygenase 2 in the sIA wall. Moreover, SAA positivity correlated with the accumulation of apolipoproteins A-1 and B-100. In conclusion, SAA occurs in the sIA wall and, as an inflammation-related factor, may contribute to the development of a rupture-prone sIA.


Subject(s)
Aneurysm, Ruptured/metabolism , Endothelium, Vascular/metabolism , Inflammation Mediators/metabolism , Intracranial Aneurysm/metabolism , Serum Amyloid A Protein/metabolism , Aneurysm/metabolism , Aneurysm/pathology , Aneurysm, Ruptured/pathology , Endothelium, Vascular/chemistry , Endothelium, Vascular/pathology , Humans , Inflammation Mediators/analysis , Intracranial Aneurysm/pathology , Serum Amyloid A Protein/analysis
5.
Cells ; 10(1)2021 01 15.
Article in English | MEDLINE | ID: mdl-33467682

ABSTRACT

Abdominal aortic aneurysm (AAA) bears a high risk of rupture and sudden death of the patient. The pathogenic mechanisms of AAA remain elusive, and surgical intervention represents the only treatment option. Heme oxygenase-1 (HO-1), a heme degrading enzyme, is induced in AAA, both in mice and humans. HO-1 was reported to mitigate AAA development in an angiotensin II (AngII)-induced model of AAA in hyperlipidemic ApoE-/- mice. Since the role of hyperlipidaemia in the pathogenesis of AAA remains controversial, we aimed to evaluate the significance of HO-1 in the development and progression of AAA in normolipidemic animals. The experiments were performed in HO-1-deficient mice and their wild-type counterparts. We demonstrated in non-hypercholesterolemic mice that the high-dose of AngII leads to the efficient formation of AAA, which is attenuated by HO-1 deficiency. Yet, if formed, they are significantly more prone to rupture upon HO-1 shortage. Differential susceptibility to AAA formation does not rely on enhanced inflammatory response or oxidative stress. AAA-resistant mice are characterized by an increase in regulators of aortic remodeling and angiotensin receptor-2 expression, significant medial thickening, and delayed blood pressure elevation in response to AngII. To conclude, we unveil a dual role of HO-1 deficiency in AAA in normolipidemic mice, where it protects against AAA development, but exacerbates the state of formed AAA.


Subject(s)
Angiotensin II/adverse effects , Aortic Aneurysm, Abdominal/metabolism , Heme Oxygenase-1/metabolism , Membrane Proteins/metabolism , Oxidative Stress , Aneurysm/metabolism , Animals , Cardiovascular Diseases/metabolism , Cell Line , Collagen/metabolism , Genotype , Humans , Hyperlipidemias/metabolism , Male , Mice , Mice, Inbred C57BL , Myocytes, Smooth Muscle/metabolism , Plasminogen Activator Inhibitor 1/biosynthesis , Receptor, Angiotensin, Type 2/metabolism , Serpin E2/metabolism , Skin/metabolism , Swine , Tissue Inhibitor of Metalloproteinase-2/biosynthesis , Tissue Inhibitor of Metalloproteinase-2/metabolism
6.
Hypertension ; 77(2): 557-570, 2021 02.
Article in English | MEDLINE | ID: mdl-33356402

ABSTRACT

No drug therapy has shown to limit abdominal aortic aneurysm (AAA) growth or rupture, and the understanding of the disease biology is incomplete; whereby, one challenge of vascular medicine is the development of good animal models and therapies for this life-threatening condition. The nuclear receptor NOR-1 (neuron-derived orphan receptor 1) controls biological processes involved in AAA; however, whether it plays a role in this pathology is unknown. Through a gain-of-function approach we assessed the impact of NOR-1 expression on the vascular response to Ang II (angiotensin II). We used 2 mouse models that overexpress human NOR-1 in the vasculature, one of them specifically in vascular smooth muscle cells. NOR-1 transgenesis amplifies the response to Ang II enhancing vascular inflammation (production of proinflammatory cytokines, chemokines, and reactive oxygen species), increasing MMP (matrix metalloproteinase) activity and disturbing elastin integrity, thereby broking the resistance of C57BL/6 mice to Ang II-induced AAA. Genes encoding for proteins critically involved in AAA formation (Il [interleukin]-6, Il-1ß, Cxcl2, [C-X-C motif chemokine ligand 2], Mcp-1 [monocyte chemoattractant protein 1], and Mmp2) were upregulated in aneurysmal tissues. Both animal models show a similar incidence and severity of AAA, suggesting that high expression of NOR-1 in vascular smooth muscle cell is a sufficient condition to strengthen the response to Ang II. These alterations, including AAA formation, were prevented by the MMP inhibitor doxycycline. Microarray analysis identified gene sets that could explain the susceptibility of transgenic animals to Ang II-induced aneurysms, including those related with extracellular matrix remodeling, inflammatory/immune response, sympathetic activity, and vascular smooth muscle cell differentiation. These results involve NOR-1 in AAA and validate mice overexpressing this receptor as useful experimental models.


Subject(s)
Aneurysm/metabolism , Angiotensin II/pharmacology , DNA-Binding Proteins/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/metabolism , Aneurysm/genetics , Animals , DNA-Binding Proteins/genetics , Disease Models, Animal , Elastin/metabolism , Inflammation/genetics , Inflammation/metabolism , Matrix Metalloproteinases/metabolism , Mice , Mice, Transgenic , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Nerve Tissue Proteins/genetics , Oxidative Stress/physiology , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/genetics , Signal Transduction/drug effects
7.
Angiology ; 72(6): 539-549, 2021 Jul.
Article in English | MEDLINE | ID: mdl-32851875

ABSTRACT

Rupture of splenic artery aneurysms (SAAs) is associated with a high mortality rate. The aim of this study was to identify the features of SAAs. Tissue sections from SAAs were compared to nonaneurysmal splenic arteries using various stains. The presence of intraluminal thrombus (ILT), vascular smooth muscle cells (VSMCs), cluster of differentiation (CD)-68+ phagocytes, myeloperoxidase+ neutrophils, CD3+, and CD20+ adaptive immune cells were studied using immunofluorescence microscopy. Analysis of SAAs revealed the presence of atherosclerotic lesions, calcifications, and ILT. Splenic artery aneurysms were characterized by a profound vascular remodeling with a dramatic loss of VSMCs, elastin degradation, adventitial fibrosis associated with enhanced apoptosis, and increased matrix metalloproteinase 9 expression. We observed an infiltration of immune cells comprising macrophages, neutrophils, T, and B cells. The T and B cells were found in the adventitial layer of SAAs, but their organization into tertiary lymphoid organs was halted. We failed to detect germinal centers even in the most organized T/B cell follicles and these lymphoid clusters lacked lymphoid stromal cells. This detailed histopathological characterization of the vascular remodeling during SAA showed that lymphoid neogenesis was incomplete, suggesting that critical mediators of their development must be missing.


Subject(s)
Aneurysm/immunology , Aneurysm/pathology , Leukocytes/immunology , Macrophages/immunology , Splenic Artery/immunology , Splenic Artery/pathology , Vascular Remodeling , Adult , Aged , Aged, 80 and over , Aneurysm/metabolism , Aneurysm/surgery , Apoptosis , B-Lymphocytes/immunology , Biomarkers/analysis , Female , Fibrosis , Humans , Macrophages/chemistry , Male , Middle Aged , Neutrophils/immunology , Retrospective Studies , Splenic Artery/chemistry , Splenic Artery/surgery , T-Lymphocytes/immunology
8.
Front Immunol ; 12: 780804, 2021.
Article in English | MEDLINE | ID: mdl-35069552

ABSTRACT

Objectives: Currently, cardiovascular risk associated with COVID-19 has been brought to people's attention, but the mechanism is not clear. The aim of this study is to elucidate the mechanisms based on multiple omics data. Methodology: Weighted gene co-expression network analysis (WGCNA) was used to identify key pathways. Combination analysis with aneurysm and atherosclerosis related pathways, hypoxia induced factor-1 (HIF-1) signaling were identified as key pathways of the increased cardiovascular risk associated with COVID-19. ScMLnet algorithm based on scRNA-seq was used to explore the regulation of HIF-1 pathway by intercellular communication. Proteomic analysis was used to detect the regulatory mechanisms between IL18 and HIF-1 signaling pathway. Pseudo time locus analysis was used to study the regulation of HIF1 signaling pathway in macrophages and vascular smooth muscle cells (VSMC) phenotypic transformation. The Virtual Inference of protein-activity by Enriched Regulon (VIPER) analysis was used to study the activity of regulatory proteins. Epigenetic analysis based on methylation revealed epigenetic changes in PBMC after SARS-CoV-2 infection. Potential therapeutic compounds were explored by using Cmap algorithm. Results: HIF-1 signaling pathway is a common key pathway for aneurysms, atherosclerosis and SARS-CoV-2 infection. Intercellular communication analysis showed that macrophage-derived interleukin-18 (IL-18) activates the HIF-1 signaling pathway through IL18R1. Proteomic analysis showed that IL18/IL18R1 promote NF-κB entry into the nucleus, and activated the HIF-1 signaling pathway. Macrophage-derived IL18 promoted the M1 polarization of macrophages and the syntactic phenotype transformation of VSMCs. MAP2K1 mediates the functional regulation of HIF-1 signaling pathway in various cell types. Epigenetic changes in PBMC after COVID-19 infection are characterized by activation of the type I interferon pathway. MEK inhibitors are the promising compounds for the treatment of HIF-1 overactivation. Conclusions: The IL18/IL18R1/HIF1A axis is expected to be an therapeutic target for cardiovascular protection after SARS-CoV-2 infection. MEK inhibitors may be an choice for cardiovascular protection after SARS-COV-2 infection.


Subject(s)
Aneurysm/etiology , Aneurysm/metabolism , Atherosclerosis/etiology , Atherosclerosis/metabolism , COVID-19/blood , COVID-19/complications , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interleukin-18 Receptor alpha Subunit/metabolism , Interleukin-18/metabolism , SARS-CoV-2 , Signal Transduction , Aneurysm/pathology , Atherosclerosis/pathology , COVID-19/virology , Case-Control Studies , Cells, Cultured , Epigenesis, Genetic , Humans , Interferon Type I/metabolism , Leukocytes, Mononuclear/metabolism , Macrophages/metabolism , Myocytes, Smooth Muscle/metabolism , NF-kappa B/metabolism , Proteomics/methods , RNA-Seq/methods , Risk Factors , Single-Cell Analysis/methods
9.
Cell Cycle ; 19(24): 3608-3621, 2020 12.
Article in English | MEDLINE | ID: mdl-33315517

ABSTRACT

The endothelialization of endothelial progenitor cells (EPCs) was proven to facilitate the vascular repair of aneurysm. MiR-17-5p regulated angiogenesis in various cancers. This research focused on exploring the effect of miR-17-5p on EPCs and the vascular repair of aneurysm. In vivo study: the aneurysm rat model was established and treated with AgomiR-17-5p; the histopathology of aneurysm tissues was examined by hematoxylin-eosin staining; and the level of EPCs in the aneurysm tissues and peripheral blood of rats were evaluated by immunofluorescence and flow cytometry, respectively. In vitro study: EPCs were cultured and identified using flow cytometry; the target of miR-17-5p was proven by dual-luciferase reporter assay; after transfection, the viability, migration, and tube formation of the EPCs were detected by MTT, wound healing, and tube formation assays, respectively; the expressions of VEGFA and factors related to PTEN-mediated PI3K/AKT pathway were detected by ELISA, qPCR, or Western blot as needed. In vivo study: miR-17-5p overexpression promoted the vascular repair in aneurysm rats and increased the level of EPCs in the aneurysm tissues and peripheral blood of the rats. In vitro study: miR-17-5p overexpression promoted the viability, migration, and tube formation of EPCs, up-regulated the expressions of VEGFA, p-PI3K, and p-AKT, and down-regulated the PTEN expression in EPCs; miR-17-5p silencing did the opposite; PTEN was targeted by miR-17-3p and further abrogated the effects of miR-17-5p overexpression on EPCs. MiR-17-5p promoted the endothelialization of EPCs to facilitate the vascular repair of aneurysm by regulating PTEN-mediated PI3K/AKT/VEGFA pathway.


Subject(s)
Aneurysm/metabolism , Endothelial Progenitor Cells/metabolism , MicroRNAs/metabolism , Neovascularization, Physiologic/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics , Vascular Endothelial Growth Factor A/metabolism , Aneurysm/genetics , Animals , Cell Movement/genetics , Cell Proliferation/genetics , Cell Survival/genetics , Cells, Cultured , Disease Models, Animal , Down-Regulation/genetics , Male , MicroRNAs/genetics , Rats , Rats, Sprague-Dawley , Transfection , Up-Regulation/genetics
10.
STAR Protoc ; 1(3): 100108, 2020 12 18.
Article in English | MEDLINE | ID: mdl-33377004

ABSTRACT

Aortic aneurysm rupture is a significant cause of premature mortality worldwide. Although animal models exist, some frequently experience aortic rupture and sudden death. An alternative approach is therefore required that would use human material to aid translation. Accordingly, we present an optimized and validated protocol to isolate human umbilical cord arteries and their subsequent deployment within a bioreactor. Consequently, this reproducible ex vivo human model of aneurysm can be used for pathogenesis studies and accompanying assessment of potential novel therapeutics.


Subject(s)
Aneurysm/physiopathology , Primary Cell Culture/methods , Umbilical Arteries/growth & development , Aneurysm/metabolism , Aortic Aneurysm, Abdominal/complications , Aortic Rupture/complications , Bioreactors , Humans , Models, Biological
11.
World J Pediatr Congenit Heart Surg ; 11(4): NP498-NP500, 2020 07.
Article in English | MEDLINE | ID: mdl-32452246

ABSTRACT

Actin α2 (ACTA2) is a protein crucial for proper functioning of contractile apparatus in smooth muscles. A specific mutation resulting in substitution of arginine at position 179 by histidine (p.R179 H) in ACTA2 has been shown to be associated with multisystemic smooth muscle dysfunction syndrome. Characteristic features include aneurysmal arterial disease. Due to rarity of this disease, we report a nine-year-old girl with this rare genetic variant in whom cardiovascular manifestations were identified in fetal life and who needed neonatal cardiac surgical intervention.


Subject(s)
Actins/genetics , Aneurysm/genetics , DNA/genetics , Ductus Arteriosus, Patent/diagnosis , Ductus Arteriosus/abnormalities , Mutation , Pulmonary Artery/abnormalities , Actins/metabolism , Aneurysm/diagnosis , Aneurysm/metabolism , DNA Mutational Analysis , Ductus Arteriosus/diagnostic imaging , Ductus Arteriosus, Patent/genetics , Echocardiography , Female , Humans , Infant, Newborn , Pregnancy , Pulmonary Artery/diagnostic imaging , Pulmonary Artery/physiopathology , Young Adult
12.
Acta Pharmacol Sin ; 41(6): 745-752, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32024951

ABSTRACT

Circulating or tissue-related biomarkers are of clinical value for risk stratification in patients with abdominal aortic aneurysms. Relaxin-2 (RL2) has been linked to the presence and size of arterial aneurysms, and to the extent of atherosclerosis in human subjects. Here, we assessed the expression levels of RL2 in aneurysmal (AA, n = 16) and atherosclerotic (ATH, n = 22) arteries, and established the correlation between RL2 levels and the presence/size of AA and the clinical severity of atherosclerosis. The expression levels of metalloproteinases (MMPs) and endothelial nitric oxide synthetase (eNOS) were also detected for correlations with different phenotypes of atherosclerosis and AA. Temporal artery biopsy specimens (n = 6) and abdominal aortic tissues harvested from accident victims during autopsy (n = 10) were used as controls. Quantitative tissue biomarker analysis revealed that tissue-specific RL2 was increased in patients with larger or symptomatic AA compared to subjects with atherosclerotic disease and healthy controls. In situ RL2 levels were proportional to the size and the severity of aneurysmatic disease, and were substantially elevated in patients with symptomatic aneurysm of any diameter or asymptomatic aneurysm of a diameter >350% of that of the normal artery. In contrast, tissue RL2 was inversely associated with the clinical severity of atherosclerotic lesions. Correlation between RL2 and MMP2 was different between ATH1 and ATH2, depending on atherosclerosis grade. Overall, tissue RL2 is differentially associated with discrete phenotypes of arterial disease and might exert multipotent biological effects on vascular wall integrity and remodeling in human subjects.


Subject(s)
Aneurysm/metabolism , Atherosclerosis/metabolism , Relaxin/metabolism , Aged , Female , Humans , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Relaxin/genetics , Severity of Illness Index
13.
Nat Commun ; 10(1): 5046, 2019 11 06.
Article in English | MEDLINE | ID: mdl-31695038

ABSTRACT

Abdominal aortic aneurysm (AAA) is a prevalent life-threatening disease, where aortic wall degradation is mediated by accumulated immune cells. Although cytokines regulate inflammation within the aorta, their contribution to AAA via distant alterations, particularly in the control of hematopoietic stem cell (HSC) differentiation, remains poorly defined. Here we report a pathogenic role for the interleukin-27 receptor (IL-27R) in AAA, as genetic ablation of IL-27R protects mice from the disease development. Mitigation of AAA is associated with a blunted accumulation of myeloid cells in the aorta due to the attenuation of Angiotensin II (Ang II)-induced HSC expansion. IL-27R signaling is required to induce transcriptional programming to overcome HSC quiescence and increase differentiation and output of mature myeloid cells in response to stress stimuli to promote their accumulation in the diseased aorta. Overall, our studies illuminate how a prominent vascular disease can be distantly driven by a cytokine-dependent regulation of bone marrow precursors.


Subject(s)
Aortic Aneurysm, Abdominal/metabolism , Interleukin-27/metabolism , Myelopoiesis/physiology , Receptors, Interleukin/metabolism , Aneurysm/metabolism , Angiotensin II/metabolism , Animals , Aorta/pathology , Aortic Aneurysm, Abdominal/pathology , Blood Pressure , Cell Differentiation , Cytokines/metabolism , Disease Models, Animal , Female , Hematopoietic Stem Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Knockout, ApoE , Myeloid Cells/pathology , Receptors, Interleukin/genetics , Signal Transduction
14.
Sci Rep ; 9(1): 15472, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31664168

ABSTRACT

Aortic aneurysms are associated with fatal aortic rupture. Current therapeutic approaches are limited to implantation of aortic prostheses and stent-grafts; no effective drugs are available because the pathogenic mechanisms of aortic aneurysms remain unclear. Here, we aimed to elucidate the molecular mechanisms of the initiation and progression of aortic aneurysm by lipidomics. We performed lipidomics analyses of lipids in the aortic media of normal, border, and aneurysm areas from patients with thoracic atherosclerotic aortic aneurysm (N = 30), thoracic nonatherosclerotic aortic aneurysm (N = 19), and abdominal atherosclerotic aortic aneurysm (N = 11) and from controls (N = 8) using liquid chromatography and mass spectrometry. Significant alterations were observed in the lipid profiles of patients with atherosclerotic aortic aneurysms and to a lesser extent in those with nonatherosclerotic aneurysms. Increased triacylglycerols (TGs) and decreased ether-type phosphatidylethanolamines (ePEs) were observed throughout the normal, border, and aneurysm areas of thoracic and abdominal atherosclerotic aortic aneurysms. Prostaglandin D2 increased, but ePEs and TGs decreased in normal areas of thoracic atherosclerotic aortic aneurysms and thoracic nonatherosclerotic aortic aneurysms compared with the control tissues. These findings expand our knowledge of metabolic changes in aortic aneurysms and provide insights into the pathophysiology of aortic aneurysms.


Subject(s)
Aneurysm/etiology , Aorta/metabolism , Atherosclerosis/complications , Lipidomics , Tunica Media/metabolism , Adult , Aged , Aneurysm/metabolism , Aneurysm/physiopathology , Atherosclerosis/metabolism , Atherosclerosis/physiopathology , Female , Humans , Male , Middle Aged , Risk Factors
15.
J Cell Mol Med ; 23(10): 7000-7009, 2019 10.
Article in English | MEDLINE | ID: mdl-31402541

ABSTRACT

Marfan syndrome (MFS) is a connective tissue disorder that results in aortic root aneurysm formation. Reactive oxygen species (ROS) seem to play a role in aortic wall remodelling in MFS, although the mechanism remains unknown. MFS Fbn1C1039G/+ mouse root/ascending (AS) and descending (DES) aortic samples were examined using DHE staining, lucigenin-enhanced chemiluminescence (LGCL), Verhoeff's elastin-Van Gieson staining (elastin breakdown) and in situ zymography for protease activity. Fbn1C1039G/+ AS- or DES-derived smooth muscle cells (SMC) were treated with anti-TGF-ß antibody, angiotensin II (AngII), anti-TGF-ß antibody + AngII, or isotype control. ROS were detected during early aneurysm formation in the Fbn1C1039G/+ AS aorta, but absent in normal-sized DES aorta. Fbn1C1039G/+ mice treated with the unspecific NADPH oxidase inhibitor, apocynin reduced AS aneurysm formation, with attenuated elastin fragmentation. In situ zymography revealed apocynin treatment decreased protease activity. In vitro SMC studies showed Fbn1C1039G/+ -derived AS SMC had increased NADPH activity compared to DES-derived SMC. AS SMC NADPH activity increased with AngII treatment and appeared TGF-ß dependent. In conclusion, ROS play a role in MFS aneurysm development and correspond anatomically with aneurysmal aortic segments. ROS inhibition via apocynin treatment attenuates MFS aneurysm progression. AngII enhances ROS production in MFS AS SMCs and is likely TGF-ß dependent.


Subject(s)
Aneurysm/complications , Aneurysm/metabolism , Marfan Syndrome/complications , Marfan Syndrome/metabolism , Reactive Oxygen Species/metabolism , Acetophenones/pharmacology , Angiotensin II , Animals , Aorta/metabolism , Aorta/pathology , Disease Models, Animal , Fibrillin-1/deficiency , Fibrillin-1/metabolism , Mice, Inbred C57BL , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , NADPH Oxidases/metabolism
16.
Cytokine ; 122: 154215, 2019 10.
Article in English | MEDLINE | ID: mdl-29198612

ABSTRACT

The interleukin (IL)-1 family is a group of cytokines crucially involved in regulating immune responses to infectious challenges and sterile insults. The family consists of the eponymous pair IL-1α and IL-1ß, IL-18, IL-33, IL-37, IL-38, and several isoforms of IL-36. In addition, two endogenous inhibitors of functional receptor binding, IL-1R antagonist (IL-1Ra) and IL-36Ra complete the family. To gain biological activity IL-1ß and IL-18 require processing by the protease caspase-1 which is associated with the multi-protein complex inflammasome. Numerous clinical association studies and experimental approaches have implicated members of the IL-1 family, their receptors, or component of the processing machinery in underlying processes of cardiovascular diseases (CVDs). Here we summarize the current state of knowledge regarding the pro-inflammatory and disease-modulating role of the IL-1 family in atherosclerosis, myocardial infarction, aneurysm, stroke, and other CVDs. We discuss clinical evidence, experimental approaches and lastly lend a perspective on currently developing therapeutic strategies involving the IL-1 family in CVD.


Subject(s)
Atherosclerosis/metabolism , Inflammasomes/metabolism , Interleukin-1alpha/metabolism , Interleukin-1beta/metabolism , Myocardial Infarction/metabolism , Stroke/metabolism , Aneurysm/metabolism , Aneurysm/therapy , Animals , Atherosclerosis/therapy , Caspase 1/metabolism , Cytokines/metabolism , Diabetes Mellitus/metabolism , Diabetes Mellitus/therapy , Humans , Myocardial Infarction/therapy , Stroke/therapy
18.
ACS Appl Mater Interfaces ; 10(48): 41012-41018, 2018 Dec 05.
Article in English | MEDLINE | ID: mdl-30403126

ABSTRACT

This study explored a new rosuvastatin calcium- and heparin-loaded poly(l-lactide- co-caprolactone) (PLCL) scaffold for covered stents for treating aneurysms. The mechanism of rosuvastatin-induced endothelialization via vascular endothelial growth factor (VEGF)-A elevation was further explored. Rosu50, Rosu75, Rosu100, and phosphate-buffered saline (PBS) nanofibrous scaffolds were fabricated by coaxial electrospinning and observed by electron microscopy. Anticoagulation and pro-endothelialization properties were tested. Sixteen rabbits were selected for an in vivo assay and underwent microsurgery to establish a carotid aneurysm model. The animals were treated with covered stents and followed for 4 months using digital subtraction angiography (DSA), electron microscopy, and histology. Rosuvastatin-treated human umbilical vein endothelial cell (HUVEC) viability, function, and VEGF-A modulation were further studied to elucidate the pro-endothelialization mechanism of rosuvastatin. Our study demonstrates that rosuvastatin and heparin can be incorporated into PLCL nanofibers via electrospinning. Rosu100 nanofiber scaffolds exhibited significant anticoagulation properties. The viability of HUVECs transferred to Rosu100 nanofiber scaffolds was increased significantly. In vivo, DSA revealed that the Rosu100 group had better outcomes than the PBS group. In addition, the Rosu100 stents induced more integrated endothelialization. Further study demonstrated that rosuvastatin promoted HUVEC viability and function in vitro. The effects of rosuvastatin may be attributed to an elevation in VEGF-A. We demonstrated that rosuvastatin- and heparin-loaded PLCL-covered stents show favorable anticoagulation and pro-endothelialization properties in vitro and in vivo in a rabbit aneurysm model. VEGF-A elevation played a crucial role in rosuvastatin-promoted endothelialization. This work provides an additional option for treating cerebral aneurysms with covered stents.


Subject(s)
Aneurysm , Carotid Arteries , Nanofibers/chemistry , Rosuvastatin Calcium , Stents , Vascular Endothelial Growth Factor A , Aneurysm/metabolism , Aneurysm/pathology , Aneurysm/surgery , Animals , Caproates/chemistry , Caproates/pharmacokinetics , Caproates/pharmacology , Carotid Arteries/metabolism , Carotid Arteries/pathology , Carotid Arteries/surgery , Heparin/chemistry , Heparin/pharmacokinetics , Heparin/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Lactones/chemistry , Lactones/pharmacokinetics , Lactones/pharmacology , Polyesters/chemistry , Polyesters/pharmacokinetics , Polyesters/pharmacology , Rabbits , Rosuvastatin Calcium/chemistry , Rosuvastatin Calcium/pharmacokinetics , Rosuvastatin Calcium/pharmacology , Vascular Endothelial Growth Factor A/chemistry , Vascular Endothelial Growth Factor A/pharmacokinetics , Vascular Endothelial Growth Factor A/pharmacology
19.
Can J Cardiol ; 34(4): 362-370, 2018 04.
Article in English | MEDLINE | ID: mdl-29571420

ABSTRACT

Aneurysm formation is a complex multifactorial process with both genetic and environmental influences. Over recent years, there has been increasing recognition of sex-specific differences regarding the prevalence and natural history of cardiovascular diseases in the population. In particular, there is a growing body of evidence showing that aneurysm behaviour differs based on sex. Although most types of aneurysms are more common in men, their growth rates and outcomes are worse in women. This fact raises attention about potential underlying differences in the arteries of men and women that may contribute to differences in aneurysm prevalence and outcomes. There are complex biochemical and mechanical mechanisms at play that contribute to vascular health. Furthermore, many studies have suggested potential differences in the hormonal milieu and underlying arterial anatomy between men and women. Based on the data reviewed in this article, assessment of the underlying pathophysiology of aneurysms in women might prove clinically useful regarding prevention, early detection, and management of aneurysms in women. Sex-specific research, screening, and treatment guidelines for aneurysm disease should be introduced to reflect the differing natural history of these diseases in men and women.


Subject(s)
Aneurysm , Arteries , Disease Management , Aneurysm/epidemiology , Aneurysm/metabolism , Aneurysm/physiopathology , Aneurysm/therapy , Disease Progression , Female , Humans , Prevalence , Risk Factors , Secondary Prevention , Sex Factors
20.
Tissue Eng Part A ; 24(15-16): 1251-1261, 2018 08.
Article in English | MEDLINE | ID: mdl-29431029

ABSTRACT

Most tissue-engineered arterial grafts are complicated by aneurysmal dilation secondary to insufficient neotissue formation after scaffold degradation. The optimal graft would form an organized multilayered structure with a robust extracellular matrix that could withstand arterial pressure. The purpose of the current study was to determine how oversizing a biodegradable arterial scaffold affects long-term neotissue formation. Size-matched (1.0 mm, n = 11) and oversized (1.6 mm, n = 9) electrospun polycaprolactone/chitosan scaffolds were implanted as abdominal aortic interposition grafts in Lewis rats. The mean lumen diameter of the 1.6 mm grafts was initially greater compared with the native vessel, but matched the native aorta by 6 months. In contrast, the 1.0 mm grafts experienced stenosis at 6 and 9 months. Total neotissue area and calponin-positive neotissue area were significantly greater in the 1.6 mm grafts by 6 months and similar to the native aorta. Late-term biomechanical testing was dominated by remaining polymer, but graft oversizing did not adversely affect the biomechanics of the adjacent vessel. Oversizing tissue-engineered arterial grafts may represent a strategy to increase the formation of organized neotissue without thrombosis or adverse remodeling of the adjacent native vessel by harnessing a previously undescribed process of adaptive vascular remodeling.


Subject(s)
Absorbable Implants , Blood Vessel Prosthesis , Chitosan/chemistry , Polyesters/chemistry , Tissue Scaffolds/chemistry , Tunica Intima/metabolism , Aneurysm/metabolism , Aneurysm/pathology , Aneurysm/surgery , Animals , Female , Rats , Rats, Inbred Lew , Tunica Intima/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...