Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 331
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Circulation ; 149(24): 1903-1920, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38357802

ABSTRACT

BACKGROUND: S-Nitrosylation (SNO), a prototypic redox-based posttranslational modification, is involved in cardiovascular disease. Aortic aneurysm and dissection are high-risk cardiovascular diseases without an effective cure. The aim of this study was to determine the role of SNO of Septin2 in macrophages in aortic aneurysm and dissection. METHODS: Biotin-switch assay combined with liquid chromatography-tandem mass spectrometry was performed to identify the S-nitrosylated proteins in aortic tissue from both patients undergoing surgery for aortic dissection and Apoe-/- mice infused with angiotensin II. Angiotensin II-induced aortic aneurysm model and ß-aminopropionitrile-induced aortic aneurysm and dissection model were used to determine the role of SNO of Septin2 (SNO-Septin2) in aortic aneurysm and dissection development. RNA-sequencing analysis was performed to recapitulate possible changes in the transcriptome profile of SNO-Septin2 in macrophages in aortic aneurysm and dissection. Liquid chromatography-tandem mass spectrometry and coimmunoprecipitation were used to uncover the TIAM1-RAC1 (Ras-related C3 botulinum toxin substrate 1) axis as the downstream target of SNO-Septin2. Both R-Ketorolac and NSC23766 treatments were used to inhibit the TIAM1-RAC1 axis. RESULTS: Septin2 was identified S-nitrosylated at cysteine 111 (Cys111) in both aortic tissue from patients undergoing surgery for aortic dissection and Apoe-/- mice infused with Angiotensin II. SNO-Septin2 was demonstrated driving the development of aortic aneurysm and dissection. By RNA-sequencing, SNO-Septin2 in macrophages was demonstrated to exacerbate vascular inflammation and extracellular matrix degradation in aortic aneurysm. Next, TIAM1 (T lymphoma invasion and metastasis-inducing protein 1) was identified as a SNO-Septin2 target protein. Mechanistically, compared with unmodified Septin2, SNO-Septin2 reduced its interaction with TIAM1 and activated the TIAM1-RAC1 axis and consequent nuclear factor-κB signaling pathway, resulting in stronger inflammation and extracellular matrix degradation mediated by macrophages. Consistently, both R-Ketorolac and NSC23766 treatments protected against aortic aneurysm and dissection by inhibiting the TIAM1-RAC1 axis. CONCLUSIONS: SNO-Septin2 drives aortic aneurysm and dissection through coupling the TIAM1-RAC1 axis in macrophages and activating the nuclear factor-κB signaling pathway-dependent inflammation and extracellular matrix degradation. Pharmacological blockade of RAC1 by R-Ketorolac or NSC23766 may therefore represent a potential treatment against aortic aneurysm and dissection.


Subject(s)
Aortic Aneurysm , Aortic Dissection , Macrophages , Septins , T-Lymphoma Invasion and Metastasis-inducing Protein 1 , rac1 GTP-Binding Protein , Animals , Humans , Male , Mice , Angiotensin II/metabolism , Aortic Aneurysm/metabolism , Aortic Aneurysm/pathology , Aortic Aneurysm/genetics , Aortic Dissection/metabolism , Aortic Dissection/pathology , Aortic Dissection/genetics , Disease Models, Animal , Macrophages/metabolism , Macrophages/pathology , Mice, Inbred C57BL , Neuropeptides , rac1 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/genetics , Septins/metabolism , Septins/genetics , Signal Transduction , T-Lymphoma Invasion and Metastasis-inducing Protein 1/metabolism , T-Lymphoma Invasion and Metastasis-inducing Protein 1/genetics
2.
Int J Mol Sci ; 25(9)2024 May 04.
Article in English | MEDLINE | ID: mdl-38732244

ABSTRACT

Cardiovascular outcome in Marfan syndrome (MFS) patients most prominently depends on aortic aneurysm progression with subsequent aortic dissection. Angiotensin II receptor blockers (ARBs) prevent aneurysm formation in MFS mouse models. In patients, ARBs only slow down aortic dilation. Downstream signalling from the angiotensin II type 1 receptor (AT1R) is mediated by G proteins and ß-arrestin recruitment. AT1R also interacts with the monocyte chemoattractant protein-1 (MCP-1) receptor, resulting in inflammation. In this study, we explore the targeting of ß-arrestin signalling in MFS mice by administering TRV027. Furthermore, because high doses of the ARB losartan, which has been proven beneficial in MFS, cannot be achieved in humans, we investigate a potential additive effect by combining lower concentrations of losartan (25 mg/kg/day and 5 mg/kg/day) with barbadin, a ß-arrestin blocker, and DMX20, a C-C chemokine receptor type 2 (CCR2) blocker. A high dose of losartan (50 mg/kg/day) slowed down aneurysm progression compared to untreated MFS mice (1.73 ± 0.12 vs. 1.96 ± 0.08 mm, p = 0.0033). TRV027, the combination of barbadin with losartan (25 mg/kg/day), and DMX-200 (90 mg/kg/day) with a low dose of losartan (5 mg/kg/day) did not show a significant beneficial effect. Our results confirm that while losartan effectively halts aneurysm formation in Fbn1C1041G/+ MFS mice, neither TRV027 alone nor any of the other compounds combined with lower doses of losartan demonstrate a notable impact on aneurysm advancement. It appears that complete blockade of AT1R function, achieved by administrating a high dosage of losartan, may be necessary for inhibiting aneurysm progression in MFS.


Subject(s)
Angiotensin II Type 1 Receptor Blockers , Disease Models, Animal , Losartan , Marfan Syndrome , Receptor, Angiotensin, Type 1 , Signal Transduction , Animals , Marfan Syndrome/metabolism , Marfan Syndrome/drug therapy , Marfan Syndrome/complications , Mice , Losartan/pharmacology , Receptor, Angiotensin, Type 1/metabolism , Signal Transduction/drug effects , Angiotensin II Type 1 Receptor Blockers/pharmacology , Aortic Aneurysm/metabolism , Aortic Aneurysm/etiology , Aortic Aneurysm/prevention & control , Aortic Aneurysm/drug therapy , Aortic Aneurysm/pathology , Male , beta-Arrestins/metabolism , Receptors, CCR2/metabolism , Receptors, CCR2/antagonists & inhibitors , Mice, Inbred C57BL
3.
Hum Mol Genet ; 30(23): 2286-2299, 2021 11 16.
Article in English | MEDLINE | ID: mdl-34244757

ABSTRACT

Aortic aneurysms (AAs) are pathological dilatations of the aorta. Pathogenic variants in genes encoding for proteins of the contractile machinery of vascular smooth muscle cells (VSMCs), genes encoding proteins of the transforming growth factor beta signaling pathway and extracellular matrix (ECM) homeostasis play a role in the weakening of the aortic wall. These variants affect the functioning of VSMC, the predominant cell type in the aorta. Many variants have unknown clinical significance, with unknown consequences on VSMC function and AA development. Our goal was to develop functional assays that show the effects of pathogenic variants in aneurysm-related genes. We used a previously developed fibroblast transdifferentiation protocol to induce VSMC-like cells, which are used for all assays. We compared transdifferentiated VSMC-like cells of patients with a pathogenic variant in genes encoding for components of VSMC contraction (ACTA2, MYH11), transforming growth factor beta (TGFß) signaling (SMAD3) and a dominant negative (DN) and two haploinsufficient variants in the ECM elastic laminae (FBN1) to those of healthy controls. The transdifferentiation efficiency, structural integrity of the cytoskeleton, TGFß signaling profile, migration velocity and maximum contraction were measured. Transdifferentiation efficiency was strongly reduced in SMAD3 and FBN1 DN patients. ACTA2 and FBN1 DN cells showed a decrease in SMAD2 phosphorylation. Migration velocity was impaired for ACTA2 and MYH11 cells. ACTA2 cells showed reduced contractility. In conclusion, these assays for showing effects of pathogenic variants may be promising tools to help reclassification of variants of unknown clinical significance in AA-related genes.


Subject(s)
Actins/genetics , Aortic Aneurysm/etiology , Fibrillin-1/genetics , Myosin Heavy Chains/genetics , Smad3 Protein/genetics , Aortic Aneurysm/metabolism , Aortic Aneurysm/pathology , Cell Differentiation/genetics , Cell Transdifferentiation/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Models, Biological , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Smad2 Protein/metabolism
4.
Cell Commun Signal ; 21(1): 113, 2023 05 15.
Article in English | MEDLINE | ID: mdl-37189183

ABSTRACT

BACKGROUND AND OBJECTIVES: Phenotypic switching in vascular smooth muscle cells (VSMCs) has been linked to aortic aneurysm, but the phenotypic landscape in aortic aneurysm is poorly understood. The present study aimed to analyse the phenotypic landscape, phenotypic differentiation trajectory, and potential functions of various VSMCs phenotypes in aortic aneurysm. METHODS: Single-cell sequencing data of 12 aortic aneurysm samples and 5 normal aorta samples (obtained from GSE166676 and GSE155468) were integrated by the R package Harmony. VSMCs were identified according to the expression levels of ACTA2 and MYH11. VSMCs clustering was determined by the R package 'Seurat'. Cell annotation was determined by the R package 'singleR' and background knowledge of VSMCs phenotypic switching. The secretion of collagen, proteinases, and chemokines by each VSMCs phenotype was assessed. Cell‒cell junctions and cell-matrix junctions were also scored by examining the expression of adhesion genes. Trajectory analysis was performed by the R package 'Monocle2'. qPCR was used to quantify VSMCs markers. RNA fluorescence in situ hybridization (RNA FISH) was performed to determine the spatial localization of vital VSMCs phenotypes in aortic aneurysms. RESULTS: A total of 7150 VSMCs were categorize into 6 phenotypes: contractile VSMCs, fibroblast-like VSMCs, T-cell-like VSMCs, adipocyte-like VSMCs, macrophage-like VSMCs, and mesenchymal-like VSMCs. The proportions of T-cell-like VSMCs, adipocyte-like VSMCs, macrophage-like VSMCs, and mesenchymal-like VSMCs were significantly increased in aortic aneurysm. Fibroblast-like VSMCs secreted abundant amounts of collagens. T-cell-like VSMCs and macrophage-like VSMCs were characterized by high chemokine levels and proinflammatory effects. Adipocyte-like VSMCs and mesenchymal-like VSMCs were associated with high proteinase levels. RNA FISH validated the presence of T-cell-like VSMCs and macrophage-like VSMCs in the tunica media and the presence of mesenchymal-like VSMCs in the tunica media and tunica adventitia. CONCLUSION: A variety of VSMCs phenotypes are involved in the formation of aortic aneurysm. T-cell-like VSMCs, macrophage-like VSMCs, and mesenchymal-like VSMCs play pivotal roles in this process. Video Abstract.


Subject(s)
Aortic Aneurysm , Muscle, Smooth, Vascular , Humans , In Situ Hybridization, Fluorescence , Aortic Aneurysm/genetics , Aortic Aneurysm/metabolism , Phenotype , RNA/metabolism , Sequence Analysis, RNA , Myocytes, Smooth Muscle/metabolism
5.
Arterioscler Thromb Vasc Biol ; 42(7): 811-818, 2022 07.
Article in English | MEDLINE | ID: mdl-35587695

ABSTRACT

Smooth muscle cells and endothelial cells have a remarkable level of plasticity in vascular pathologies. In thoracic and abdominal aortic aneurysms, smooth muscle cells have been suggested to undergo phenotypic switching and to contribute to degradation of the aortic wall structure in response to, for example, inflammatory mediators, dysregulation of growth factor signaling or oxidative stress. Recently, endothelial-to-mesenchymal transition, and a clonal expansion of degradative smooth muscle cells and immune cells, as well as mesenchymal stem-like cells have been suggested to contribute to the progression of aortic aneurysms. What are the factors driving the aortic cell phenotype changes and how vascular flow, known to affect aortic wall structure and to be altered in aortic aneurysms, could affect aortic cell remodeling? In this review, we summarize the current literature on aortic cell heterogeneity and phenotypic switching in relation to changes in vascular flow and aortic wall structure in aortic aneurysms in clinical samples with special focus on smooth muscle and endothelial cells. The differences between thoracic and abdominal aortic aneurysms are discussed.


Subject(s)
Aortic Aneurysm, Abdominal , Aortic Aneurysm, Thoracic , Aortic Aneurysm , Aortic Aneurysm/metabolism , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Thoracic/pathology , Endothelial Cells/metabolism , Humans , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism
6.
Arterioscler Thromb Vasc Biol ; 42(11): 1355-1374, 2022 11.
Article in English | MEDLINE | ID: mdl-36172868

ABSTRACT

BACKGROUND: Mural cells in ascending aortic aneurysms undergo phenotypic changes that promote extracellular matrix destruction and structural weakening. To explore this biology, we analyzed the transcriptional features of thoracic aortic tissue. METHODS: Single-nuclear RNA sequencing was performed on 13 samples from human donors, 6 with thoracic aortic aneurysm, and 7 without aneurysm. Individual transcriptomes were then clustered based on transcriptional profiles. Clusters were used for between-disease differential gene expression analyses, subcluster analysis, and analyzed for intersection with genetic aortic trait data. RESULTS: We sequenced 71 689 nuclei from human thoracic aortas and identified 14 clusters, aligning with 11 cell types, predominantly vascular smooth muscle cells (VSMCs) consistent with aortic histology. With unbiased methodology, we found 7 vascular smooth muscle cell and 6 fibroblast subclusters. Differentially expressed genes analysis revealed a vascular smooth muscle cell group accounting for the majority of differential gene expression. Fibroblast populations in aneurysm exhibit distinct behavior with almost complete disappearance of quiescent fibroblasts. Differentially expressed genes were used to prioritize genes at aortic diameter and distensibility genome-wide association study loci highlighting the genes JUN, LTBP4 (latent transforming growth factor beta-binding protein 1), and IL34 (interleukin 34) in fibroblasts, ENTPD1, PDLIM5 (PDZ and LIM domain 5), ACTN4 (alpha-actinin-4), and GLRX in vascular smooth muscle cells, as well as LRP1 in macrophage populations. CONCLUSIONS: Using nuclear RNA sequencing, we describe the cellular diversity of healthy and aneurysmal human ascending aorta. Sporadic aortic aneurysm is characterized by differential gene expression within known cellular classes rather than by the appearance of novel cellular forms. Single-nuclear RNA sequencing of aortic tissue can be used to prioritize genes at aortic trait loci.


Subject(s)
Aortic Aneurysm, Thoracic , Aortic Aneurysm , Humans , Genome-Wide Association Study , Muscle, Smooth, Vascular/metabolism , Actinin/genetics , RNA, Nuclear/metabolism , Aorta/pathology , Myocytes, Smooth Muscle/metabolism , Aortic Aneurysm, Thoracic/pathology , Aortic Aneurysm/metabolism , Sequence Analysis, RNA , Transforming Growth Factor beta/metabolism
7.
Arterioscler Thromb Vasc Biol ; 42(9): 1154-1168, 2022 09.
Article in English | MEDLINE | ID: mdl-35861960

ABSTRACT

BACKGROUND: Aortic root smooth muscle cells (SMC) develop from both the second heart field (SHF) and neural crest. Disparate responses to disease-causing Fbn1 variants by these lineages are proposed to promote focal aortic root aneurysm formation in Marfan syndrome (MFS), but lineage-stratified SMC analysis in vivo is lacking. METHODS: We generated SHF lineage-traced MFS mice and performed integrated multiomic (single-cell RNA and assay for transposase-accessible chromatin sequencing) analysis stratified by embryological origin. SMC subtypes were spatially identified via RNA in situ hybridization. Response to TWIST1 overexpression was determined via lentiviral transduction in human aortic SMCs. RESULTS: Lineage stratification enabled nuanced characterization of aortic root cells. We identified heightened SHF-derived SMC heterogeneity including a subset of Tnnt2 (cardiac troponin T)-expressing cells distinguished by altered proteoglycan expression. MFS aneurysm-associated SMC phenotypic modulation was identified in both SHF-traced and nontraced (neural crest-derived) SMCs; however, transcriptomic responses were distinct between lineages. SHF-derived modulated SMCs overexpressed collagen synthetic genes and small leucine-rich proteoglycans while nontraced SMCs activated chondrogenic genes. These modulated SMCs clustered focally in the aneurysmal aortic root at the region of SHF/neural crest lineage overlap. Integrated RNA-assay for transposase-accessible chromatin analysis identified enriched Twist1 and Smad2/3/4 complex binding motifs in SHF-derived modulated SMCs. TWIST1 overexpression promoted collagen and SLRP gene expression in vitro, suggesting TWIST1 may drive SHF-enriched collagen synthesis in MFS aneurysm. CONCLUSIONS: SMCs derived from both SHF and neural crest lineages undergo phenotypic modulation in MFS aneurysm but are defined by subtly distinct transcriptional responses. Enhanced TWIST1 transcription factor activity may contribute to enriched collagen synthetic pathways SHF-derived SMCs in MFS.


Subject(s)
Aortic Aneurysm, Thoracic , Aortic Aneurysm , Marfan Syndrome , Animals , Aortic Aneurysm/genetics , Aortic Aneurysm/metabolism , Aortic Aneurysm, Thoracic/genetics , Chromatin , Humans , Marfan Syndrome/complications , Marfan Syndrome/genetics , Marfan Syndrome/metabolism , Mice , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , RNA , Transposases/genetics
8.
J Cardiovasc Pharmacol ; 81(4): 280-291, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36652727

ABSTRACT

ABSTRACT: The occurrence and development of aortic aneurysms are accompanied by senescence of human aortic smooth muscle cells (HASMCs). Because the mechanism of HASMC senescence has not been fully elucidated, the efficacy of various antisenescence treatments varies. Decreased nicotinamide adenine dinucleotide (NAD + ) levels are one of the mechanisms of cell senescence, and there is a lack of evidence on whether increasing NAD + levels could alleviate HASMC senescence and further retard the progression of aortic aneurysms.We constructed an HASMC-based organ-on-a-chip microphysiological model. RNA sequencing was performed on cell samples from the vehicle control and angiotensin II groups to explore biological differences. We detected cellular senescence markers and NAD + levels in HASMC-based organ-on-a-chip. Subsequently, we pretreated HASMC using the synthetic precursor of NAD + , nicotinamide mononucleotide, and angiotensin II treatment, and used rhythmic stretching to investigate whether nicotinamide mononucleotide could delay HASMC senescence.The HASMC-based organ-on-a-chip model can simulate the biomechanical microenvironment of HASMCs in vivo, and the use of angiotensin II in the model replicated senescence in HASMCs. The senescence of HASMCs was accompanied by downregulation of the expression level of nicotinamide phosphoribosyltransferase and NAD + . Pretreatment with nicotinamide mononucleotide significantly increased the NAD + level and alleviated the senescence of HASMCs, but did not change the expression level of nicotinamide phosphoribosyltransferase.Our study provides a complementary research platform between traditional cell culture and animal experiments to explore HASMC senescence in aortic aneurysms. Furthermore, it provides evidence for NAD + boosting therapy in the clinical treatment of aortic aneurysms.


Subject(s)
Aortic Aneurysm , Nicotinamide Mononucleotide , Animals , Humans , Nicotinamide Mononucleotide/pharmacology , Nicotinamide Mononucleotide/metabolism , Angiotensin II/metabolism , Nicotinamide Phosphoribosyltransferase/metabolism , NAD/metabolism , Aortic Aneurysm/metabolism , Myocytes, Smooth Muscle/metabolism
9.
Acta Pharmacol Sin ; 44(2): 345-355, 2023 Feb.
Article in English | MEDLINE | ID: mdl-35945313

ABSTRACT

Abdominal aortic aneurysm (AAA) is a dangerous vascular disease without any effective drug therapies so far. Emerging evidence suggests the phenotypic differences in perivascular adipose tissue (PVAT) between regions of the aorta are implicated in the development of atherosclerosis evidenced by the abdominal aorta more vulnerable to atherosclerosis than the thoracic aorta in large animals and humans. The prevalence of thoracic aortic aneurysms (TAA) is much less than that of abdominal aortic aneurysms (AAA). In this study we investigated the effect of thoracic PVAT (T-PVAT) transplantation on aortic aneurysm formation and the impact of T-PVAT on vascular smooth muscle cells. Calcium phosphate-induced mouse AAA model was established. T-PVAT (20 mg) was implanted around the abdominal aorta of recipient mice after removal of endogenous abdominal PVAT (A-PVAT) and calcium phosphate treatment. Mice were sacrificed two weeks after the surgery and the maximum external diameter of infrarenal aorta was measured. We found that T-PVAT displayed a more BAT-like phenotype than A-PVAT; transplantation of T-PVAT significantly attenuated calcium phosphate-induced abdominal aortic dilation and elastic degradation as compared to sham control or A-PVAT transplantation. In addition, T-PVAT transplantation largely preserved smooth muscle cell content in the abdominal aortic wall. Co-culture of T-PVAT with vascular smooth muscle cells (VSMCs) significantly inhibited H2O2- or TNFα plus cycloheximide-induced VSMC apoptosis. RNA sequencing analysis showed that T-PVAT was enriched by browning adipocytes and anti-apoptotic secretory proteins. We further verified that the secretome of mature adipocytes isolated from T-PVAT significantly inhibited H2O2- or TNFα plus cycloheximide-induced VSMC apoptosis. Using proteomic and bioinformatic analyses we identified cartilage oligomeric matrix protein (COMP) as a secreted protein significantly increased in T-PVAT. Recombinant COMP protein significantly inhibited VSMC apoptosis. We conclude that T-PVAT exerts anti-apoptosis effect on VSMCs and attenuates AAA formation, which is possibly attributed to the secretome of browning adipocytes.


Subject(s)
Aortic Aneurysm, Abdominal , Aortic Aneurysm , Atherosclerosis , Humans , Mice , Animals , Tumor Necrosis Factor-alpha/metabolism , Hydrogen Peroxide/metabolism , Secretome , Muscle, Smooth, Vascular/metabolism , Cycloheximide/metabolism , Proteomics , Adipose Tissue/metabolism , Aortic Aneurysm/metabolism , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/metabolism , Aorta, Abdominal/surgery , Atherosclerosis/metabolism , Adipocytes, Brown , Mice, Inbred C57BL
10.
IUBMB Life ; 74(10): 982-994, 2022 10.
Article in English | MEDLINE | ID: mdl-35293116

ABSTRACT

Thrombospondins are a family of matricellular proteins with a multimeric structure that is known to be involved in several biological and pathological processes. Their relationship with vascular disorders has raised special interest recently. Aortic aneurysms are related to the impairment of vascular remodeling, in which extracellular matrix proteins seem to play an important role. Thus, research in thrombospondins, and their potential role in aneurysm development is progressively gaining importance. Nevertheless, studies showing thrombospondin dysregulation in human samples are still scarce. Although studies performed in vitro and in vivo models are essential to understand the molecular mechanisms and pathways underlying the disorder, descriptive studies in human samples are also necessary to ascertain their real value as biomarkers and/or novel therapeutic targets. The present article reviews the latest findings regarding the role of thrombospondins in aortic aneurysm development, paying particular attention to the studies performed in human samples.


Subject(s)
Aortic Aneurysm , Thrombospondins , Aortic Aneurysm/genetics , Aortic Aneurysm/metabolism , Aortic Aneurysm/pathology , Biomarkers , Extracellular Matrix Proteins , Humans , Thrombospondins/genetics
11.
PLoS Comput Biol ; 17(12): e1009683, 2021 12.
Article in English | MEDLINE | ID: mdl-34898595

ABSTRACT

Thoracic aortopathy-aneurysm, dissection, and rupture-is increasingly responsible for significant morbidity and mortality. Advances in medical genetics and imaging have improved diagnosis and thus enabled earlier prophylactic surgical intervention in many cases. There remains a pressing need, however, to understand better the underlying molecular and cellular mechanisms with the hope of finding robust pharmacotherapies. Diverse studies in patients and mouse models of aortopathy have revealed critical changes in multiple smooth muscle cell signaling pathways that associate with disease, yet integrating information across studies and models has remained challenging. We present a new quantitative network model that includes many of the key smooth muscle cell signaling pathways and validate the model using a detailed data set that focuses on hyperactivation of the mechanistic target of rapamycin (mTOR) pathway and its inhibition using rapamycin. We show that the model can be parameterized to capture the primary experimental findings both qualitatively and quantitatively. We further show that simulating a population of cells by varying receptor reaction weights leads to distinct proteomic clusters within the population, and that these clusters emerge due to a bistable switch driven by positive feedback in the PI3K/AKT/mTOR signaling pathway.


Subject(s)
Aortic Aneurysm , Myocytes, Smooth Muscle/metabolism , Signal Transduction/genetics , TOR Serine-Threonine Kinases , Animals , Aortic Aneurysm/genetics , Aortic Aneurysm/metabolism , Humans , Male , Mice , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
12.
Arterioscler Thromb Vasc Biol ; 41(11): 2671-2680, 2021 11.
Article in English | MEDLINE | ID: mdl-34615376

ABSTRACT

The aorta is highly heterogeneous, containing many different types of cells that perform sophisticated functions to maintain aortic homeostasis. Recently, single-cell RNA sequencing studies have provided substantial new insight into the heterogeneity of vascular cell types, the comprehensive molecular features of each cell type, and the phenotypic interrelationship between these cell populations. This new information has significantly improved our understanding of aortic biology and aneurysms at the molecular and cellular level. Here, we summarize these findings, with a focus on what single-cell RNA sequencing analysis has revealed about cellular heterogeneity, cellular transitions, communications among cell populations, and critical transcription factors in the vascular wall. We also review the information learned from single-cell RNA sequencing that has contributed to our understanding of the pathogenesis of vascular disease, such as the identification of cell types in which aneurysm-related genes and genetic variants function. Finally, we discuss the challenges and future directions of single-cell RNA sequencing applications in studies of aortic biology and diseases.


Subject(s)
Aorta/metabolism , Aortic Aneurysm/genetics , Gene Expression Profiling , Single-Cell Analysis , Transcriptome , Animals , Aorta/pathology , Aortic Aneurysm/metabolism , Aortic Aneurysm/pathology , Dilatation, Pathologic , Endothelial Cells/metabolism , Endothelial Cells/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , RNA-Seq
13.
Arterioscler Thromb Vasc Biol ; 41(9): e440-e452, 2021 09.
Article in English | MEDLINE | ID: mdl-34162229

ABSTRACT

Objective: We investigated the effect of a potent TGFß (transforming growth factor ß) inhibitor peptide (P144) from the betaglycan/TGFß receptor III on aortic aneurysm development in a Marfan syndrome mouse model. Approach and Results: We used a chimeric gene encoding the P144 peptide linked to apolipoprotein A-I via a flexible linker expressed by a hepatotropic adeno-associated vector. Two experimental approaches were performed: (1) a preventive treatment where the vector was injected before the onset of the aortic aneurysm (aged 4 weeks) and followed-up for 4 and 20 weeks and (2) a palliative treatment where the vector was injected once the aneurysm was formed (8 weeks old) and followed-up for 16 weeks. We evaluated the aortic root diameter by echocardiography, the aortic wall architecture and TGFß signaling downstream effector expression of pSMAD2 and pERK1/2 by immunohistomorphometry, and Tgfß1 and Tgfß2 mRNA expression levels by real-time polymerase chain reaction. Marfan syndrome mice subjected to the preventive approach showed no aortic dilation in contrast to untreated Marfan syndrome mice, which at the same end point age already presented the aneurysm. In contrast, the palliative treatment with P144 did not halt aneurysm progression. In all cases, P144 improved elastic fiber morphology and normalized pERK1/2-mediated TGFß signaling. Unlike the palliative treatment, the preventive treatment reduced Tgfß1 and Tgfß2 mRNA levels. Conclusions: P144 prevents the onset of aortic aneurysm but not its progression. Results indicate the importance of reducing the excess of active TGFß signaling during the early stages of aortic disease progression.


Subject(s)
Aorta/metabolism , Aortic Aneurysm/prevention & control , Gene Transfer Techniques , Genetic Therapy , Marfan Syndrome/complications , Peptide Fragments/metabolism , Proteoglycans/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/metabolism , Animals , Aorta/pathology , Aortic Aneurysm/genetics , Aortic Aneurysm/metabolism , Aortic Aneurysm/pathology , Dependovirus/genetics , Dilatation, Pathologic , Disease Models, Animal , Female , Fibrillin-1/genetics , Genetic Vectors , Male , Marfan Syndrome/genetics , Mice, Inbred C57BL , Peptide Fragments/genetics , Proteoglycans/genetics , Receptors, Transforming Growth Factor beta/genetics , Signal Transduction , Transforming Growth Factor beta/genetics
15.
Cell Mol Life Sci ; 78(9): 4161-4187, 2021 May.
Article in English | MEDLINE | ID: mdl-33575814

ABSTRACT

A disintegrin and metalloproteases (ADAMs) are key mediators of cell signaling by ectodomain shedding of various growth factors, cytokines, receptors and adhesion molecules at the cellular membrane. ADAMs regulate cell proliferation, cell growth, inflammation, and other regular cellular processes. ADAM17, the most extensively studied ADAM family member, is also known as tumor necrosis factor (TNF)-α converting enzyme (TACE). ADAMs-mediated shedding of cytokines such as TNF-α orchestrates immune system or inflammatory cascades and ADAMs-mediated shedding of growth factors causes cell growth or proliferation by transactivation of the growth factor receptors including epidermal growth factor receptor. Therefore, increased ADAMs-mediated shedding can induce inflammation, tissue remodeling and dysfunction associated with various cardiovascular diseases such as hypertension and atherosclerosis, and ADAMs can be a potential therapeutic target in these diseases. In this review, we focus on the role of ADAMs in cardiovascular pathophysiology and cardiovascular diseases. The main aim of this review is to stimulate new interest in this area by highlighting remarkable evidence.


Subject(s)
ADAM Proteins/metabolism , ADAM17 Protein/metabolism , Cardiovascular Diseases/pathology , Angiotensin II/metabolism , Animals , Aortic Aneurysm/metabolism , Aortic Aneurysm/pathology , Cardiovascular Diseases/metabolism , Cytokines/metabolism , Humans , Hypertension/metabolism , Hypertension/pathology , Signal Transduction
16.
Ann Vasc Surg ; 80: 345-357, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34780941

ABSTRACT

BACKGROUND: Coronary artery disease (CAD) and aortic aneurysms (AA) are 2 cardiovascular diseases that share a multifactorial aetiology. The influence of family history and genetics on the 2 diseases separately and in association is well known, but poorly elucidated. This comprehensive review aims to examine the current literature on the gene ANRIL (antisense non-coding RNA in the INK4 locus) and its associations with CAD and AA. METHODS: A database search on OVID, PubMed and Cochrane to identify articles concerning single nucleotide polymorphisms (SNPs) associated with ANRIL and their respective incidences of, and impact on, CAD and AA across populations. RESULTS: Cohort studies across various ethnicities reveal that various ANRIL SNPs are significantly associated separately with CAD (rs1333040, rs1333049 and rs2383207) and AA (rs564398, rs10757278 and rs1333049), and that these SNPs are present in significant proportions of the population. SNP rs1333049 is significantly associated with both diseases, but is positively correlated with AAA and negatively correlated with CAD. This review further outlines several pathophysiological links via endothelial and adventitial cells, vascular smooth muscle cells and sense gene interaction, which may explain these genetic associations identified. CONCLUSION: Given the associations uncovered between ANRIL polymorphisms and CAD and AA, as well as the molecular mechanisms which may explain the underlying pathophysiology, ANRIL appears to be strongly linked with both diseases. ANRIL may hence have a future application in screening normal patients and risk stratifying patients with both diseases. Its role in linking the 2 diseases is yet unclear, warranting further studies.


Subject(s)
Aortic Aneurysm/genetics , Coronary Artery Disease/genetics , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , RNA, Long Noncoding/genetics , Aortic Aneurysm/diagnosis , Aortic Aneurysm/metabolism , Coronary Artery Disease/diagnosis , Coronary Artery Disease/metabolism , Genetic Predisposition to Disease/ethnology , Humans
17.
Planta Med ; 88(11): 899-912, 2022 Sep.
Article in English | MEDLINE | ID: mdl-34741296

ABSTRACT

An abdominal aortic aneurysm is a life-threatening cardiovascular disorder caused by dissection and rupture. No effective medicine is currently available for the > 90% of patients whose aneurysms are below the surgical threshold. The present study investigated the impact of rosmarinic acid, salvianolic acid C, or salvianolic acid B on experimental abdominal aortic aneurysms. Abdominal aortic aneurysms were induced in apolipoprotein E-deficient mice via infusion of angiotensin II for 4 wks. Rosmarinic acid, salvianolic acid C, salvianolic acid B, or doxycycline as a positive control was provided daily through intraperitoneal injection. Administration of rosmarinic acid was found to decrease the thickness of the aortic wall, as determined by histopathological assay. Rosmarinic acid also exhibited protection against elastin fragmentation in aortic media and down-regulated cell apoptosis and proliferation in the aortic adventitia. Infiltration of macrophages, T lymphocytes, and neutrophils in aortic aneurysms was found, especially at the aortic adventitia. Rosmarinic acid, salvianolic acid C, or salvianolic acid B inhibited the infiltration on macrophages specifically, but these compounds did not influence T lymphocytes and neutrophils. Expression of matrix metalloproteinase 9 and macrophage migration inhibitory factor significantly increased in aortic aneurysms. Rosmarinic acid and salvianolic acid C decreased the expression of matrix metalloproteinase-9 in media, and rosmarinic acid also tended to reduce migration inhibitory factor expression. Further then, partial least squares-discriminate analysis was used to classify metabolic changes among different treatments. Rosmarinic acid affected most of the metabolites in the biosynthesis of the citrate cycle, fatty acid pathway significantly. Our present study on mice demonstrated that rosmarinic acid inhibited multiple pathological processes, which were the key features important in abdominal aortic aneurysm formation. Further study on rosmarinic acid, the novel candidate for aneurysmal therapy, should be undertaken to determine its potential for clinical use.


Subject(s)
Aortic Aneurysm, Abdominal , Aortic Aneurysm , Angiotensin II/metabolism , Angiotensin II/pharmacology , Animals , Aorta, Abdominal/metabolism , Aorta, Abdominal/pathology , Aortic Aneurysm/drug therapy , Aortic Aneurysm/metabolism , Aortic Aneurysm/pathology , Aortic Aneurysm, Abdominal/drug therapy , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/pathology , Cinnamates , Depsides , Disease Models, Animal , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 2/pharmacology , Mice , Mice, Inbred C57BL , Rosmarinic Acid
18.
Proc Natl Acad Sci U S A ; 116(17): 8457-8462, 2019 04 23.
Article in English | MEDLINE | ID: mdl-30948641

ABSTRACT

Prostaglandin E2 (PGE2) plays an important role in vascular homeostasis. Its receptor, E-prostanoid receptor 4 (EP4) is essential for physiological remodeling of the ductus arteriosus (DA). However, the role of EP4 in pathological vascular remodeling remains largely unknown. We found that chronic angiotensin II (AngII) infusion of mice with vascular smooth muscle cell (VSMC)-specific EP4 gene knockout (VSMC-EP4-/-) frequently developed aortic dissection (AD) with severe elastic fiber degradation and VSMC dedifferentiation. AngII-infused VSMC-EP4-/- mice also displayed more profound vascular inflammation with increased monocyte chemoattractant protein-1 (MCP-1) expression, macrophage infiltration, matrix metalloproteinase-2 and -9 (MMP2/9) levels, NADPH oxidase 1 (NOX1) activity, and reactive oxygen species production. In addition, VSMC-EP4-/- mice exhibited higher blood pressure under basal and AngII-infused conditions. Ex vivo and in vitro studies further revealed that VSMC-specific EP4 gene deficiency significantly increased AngII-elicited vasoconstriction of the mesenteric artery, likely by stimulating intracellular calcium release in VSMCs. Furthermore, EP4 gene ablation and EP4 blockade in cultured VSMCs were associated with a significant increase in MCP-1 and NOX1 expression and a marked reduction in α-SM actin (α-SMA), SM22α, and SM differentiation marker genes myosin heavy chain (SMMHC) levels and serum response factor (SRF) transcriptional activity. To summarize, the present study demonstrates that VSMC EP4 is critical for vascular homeostasis, and its dysfunction exacerbates AngII-induced pathological vascular remodeling. EP4 may therefore represent a potential therapeutic target for the treatment of AD.


Subject(s)
Angiotensin II/metabolism , Aortic Dissection/metabolism , Blood Pressure/physiology , Inflammation/metabolism , Receptors, Prostaglandin E, EP4 Subtype , Animals , Aorta/chemistry , Aorta/metabolism , Aortic Aneurysm/metabolism , Dinoprostone/metabolism , Female , Hypertension/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/metabolism , Receptors, Prostaglandin E, EP4 Subtype/genetics , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Vascular Remodeling/genetics
19.
Circ Res ; 124(4): 607-618, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30763207

ABSTRACT

Aortic aneurysms are a common vascular disease in Western populations that can involve virtually any portion of the aorta. Abdominal aortic aneurysms are much more common than thoracic aortic aneurysms and combined they account for >25 000 deaths in the United States annually. Although thoracic and abdominal aortic aneurysms share some common characteristics, including the gross anatomic appearance, alterations in extracellular matrix, and loss of smooth muscle cells, they are distinct diseases. In recent years, advances in genetic analysis, robust molecular tools, and increased availability of animal models have greatly enhanced our knowledge of the pathophysiology of aortic aneurysms. This review examines the various proposed cellular mechanisms responsible for aortic aneurysm formation and identifies opportunities for future studies.


Subject(s)
Aortic Aneurysm/metabolism , Aortic Aneurysm/epidemiology , Aortic Aneurysm/pathology , Cytokines/metabolism , Extracellular Matrix/metabolism , Humans , Oxidative Stress , Renin-Angiotensin System
20.
Arterioscler Thromb Vasc Biol ; 40(4): e78-e86, 2020 04.
Article in English | MEDLINE | ID: mdl-32208998

ABSTRACT

Aortic structure and function are controlled by the coordinated actions of different aortic cells and the extracellular matrix. Several pathways have been identified that control the aortic wall in a cell-type-specific manner and play diverse roles in various phases of aortic injury, repair, and remodeling. This complexity of signaling in the aortic wall poses challenges to the development of therapeutic strategies for treating aortic aneurysms and dissections. Here, in part II of this Recent Highlights series on aortic aneurysms and dissections, we will summarize recent studies published in Arteriosclerosis, Thrombosis, and Vascular Biology that have contributed to our knowledge of the signaling pathway-related mechanisms of aortic aneurysms and dissections.


Subject(s)
Aortic Aneurysm/metabolism , Aortic Dissection/metabolism , Extracellular Matrix/metabolism , Signal Transduction , Aortic Dissection/drug therapy , Aortic Aneurysm/drug therapy , Humans , Mutation , Receptors, Angiotensin/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL