Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 327
Filtrar
1.
Tissue Eng Part A ; 30(1-2): 75-83, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37772690

RESUMEN

Abdominal aortic aneurysm (AAA) is a critical, multifactorial cardiovascular disorder marked by localized dilatation of the abdominal aorta. A major challenge to countering the pathophysiology of AAAs lies in the naturally irreversible breakdown of elastic fibers in the aorta wall, which is linked to the poor elastogenicity of adult and diseased vascular smooth muscle cells (SMCs) and their impaired ability to assemble mature elastic fibers in a chronic proteolytic tissue milieu. We have previously shown that these are downstream effects of neutrophil elastase-induced activation of the epidermal growth factor receptor (EGFR) activity in aneurysmal SMCs. The novelty of this study lies in investigating the benefits of an EGFR inhibitor drug, afatinib (used to treat nonsmall cell lung cancer), for proelastogenic and antiproteolytic stimulation of aneurysmal SMCs. In in vitro cell cultures, we have shown that safe doses of 0.5 and 1 nM afatinib inhibit EGFR and p-extracellular signal-regulated kinases 1/2 protein expression by 50-70% and downstream elastolytic matrix metalloprotease 2 (MMP2) versus untreated control cultures. In addition, elastin production on a per cell basis was significantly upregulated by afatinib doses within the 0.1-1 nM dose range, which was further validated through transmission electron microscopy showing significantly increased presence of tropoelastin coacervates and maturing elastic fibers upon afatinib treatment at the above doses. Therefore, our studies for the first time demonstrate the therapeutic benefits of afatinib toward use for elastic matrix repair in small AAAs.


Asunto(s)
Aneurisma de la Aorta , Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Ratas , Animales , Humanos , Afatinib/farmacología , Afatinib/metabolismo , Ratas Sprague-Dawley , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Neoplasias Pulmonares/metabolismo , Aneurisma de la Aorta/metabolismo , Elastina/metabolismo , Receptores ErbB/metabolismo , Receptores ErbB/farmacología , Miocitos del Músculo Liso
2.
Nat Commun ; 14(1): 7994, 2023 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-38042913

RESUMEN

Aortic aneurysms, which may dissect or rupture acutely and be lethal, can be a part of multisystem disorders that have a heritable basis. We report four patients with deficiency of selenocysteine-containing proteins due to selenocysteine Insertion Sequence Binding Protein 2 (SECISBP2) mutations who show early-onset, progressive, aneurysmal dilatation of the ascending aorta due to cystic medial necrosis. Zebrafish and male mice with global or vascular smooth muscle cell (VSMC)-targeted disruption of Secisbp2 respectively show similar aortopathy. Aortas from patients and animal models exhibit raised cellular reactive oxygen species, oxidative DNA damage and VSMC apoptosis. Antioxidant exposure or chelation of iron prevents oxidative damage in patient's cells and aortopathy in the zebrafish model. Our observations suggest a key role for oxidative stress and cell death, including via ferroptosis, in mediating aortic degeneration.


Asunto(s)
Aneurisma de la Aorta , Pez Cebra , Humanos , Masculino , Ratones , Animales , Selenocisteína , Músculo Liso Vascular/metabolismo , Aneurisma de la Aorta/genética , Aneurisma de la Aorta/metabolismo , Selenoproteínas/genética , Miocitos del Músculo Liso/metabolismo
3.
Cell Commun Signal ; 21(1): 113, 2023 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-37189183

RESUMEN

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.


Asunto(s)
Aneurisma de la Aorta , Músculo Liso Vascular , Humanos , Hibridación Fluorescente in Situ , Aneurisma de la Aorta/genética , Aneurisma de la Aorta/metabolismo , Fenotipo , ARN/metabolismo , Análisis de Secuencia de ARN , Miocitos del Músculo Liso/metabolismo
4.
Sci Rep ; 13(1): 1380, 2023 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-36697439

RESUMEN

Aortic aneurysm (AA) is a vascular disorder characterized pathologically by inflammatory cell invasion and extracellular matrix (ECM) degradation. It is known that regulation of the balance between pro-inflammatory M1 macrophages (M1Ms) and anti-inflammatory M2 macrophages (M2Ms) plays a pivotal role in AA stabilization. We investigated the effects of M2M administration in an apolipoprotein E-deficient (apoE-/-) mouse model in which AA was induced by angiotensin II (ATII) infusion. Mice received intraperitoneal administration of 1 million M2Ms 4 weeks after ATII infusion. Compared with a control group that was administered saline, the M2M group exhibited reduced AA expansion; decreased expression levels of interleukin (IL)-1ß, IL-6, tumor necrosis factor-α (TNF-α), and monocyte chemoattractant protein-1 (MCP-1); and a lower M1M/M2M ratio. Moreover, the M2M group exhibited upregulation of anti-inflammatory factors, including IL-4 and IL-10. PKH26-labeled M2Ms accounted for 6.5% of cells in the aneurysmal site and co-expressed CD206. Taken together, intraperitoneal administration of M2Ms inhibited AA expansion by reducing the inflammatory reaction via regulating the M1M/M2M ratio. This study shows that M2M administration might be useful for the treatment of AA.


Asunto(s)
Aneurisma de la Aorta , Macrófagos , Animales , Ratones , Angiotensina II/metabolismo , Antiinflamatorios/metabolismo , Aneurisma de la Aorta/inducido químicamente , Aneurisma de la Aorta/tratamiento farmacológico , Aneurisma de la Aorta/metabolismo , Modelos Animales de Enfermedad , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Factor de Necrosis Tumoral alfa/metabolismo
5.
J Cardiovasc Pharmacol ; 81(4): 280-291, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36652727

RESUMEN

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.


Asunto(s)
Aneurisma de la Aorta , Mononucleótido de Nicotinamida , Animales , Humanos , Mononucleótido de Nicotinamida/farmacología , Mononucleótido de Nicotinamida/metabolismo , Angiotensina II/metabolismo , Nicotinamida Fosforribosiltransferasa/metabolismo , NAD/metabolismo , Aneurisma de la Aorta/metabolismo , Miocitos del Músculo Liso/metabolismo
6.
Acta Pharmacol Sin ; 44(2): 345-355, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35945313

RESUMEN

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.


Asunto(s)
Aneurisma de la Aorta Abdominal , Aneurisma de la Aorta , Aterosclerosis , Humanos , Ratones , Animales , Factor de Necrosis Tumoral alfa/metabolismo , Peróxido de Hidrógeno/metabolismo , Secretoma , Músculo Liso Vascular/metabolismo , Cicloheximida/metabolismo , Proteómica , Tejido Adiposo/metabolismo , Aneurisma de la Aorta/metabolismo , Aneurisma de la Aorta Abdominal/inducido químicamente , Aneurisma de la Aorta Abdominal/metabolismo , Aorta Abdominal/cirugía , Aterosclerosis/metabolismo , Adipocitos Marrones , Ratones Endogámicos C57BL
7.
Cell Death Differ ; 30(3): 618-631, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36151473

RESUMEN

Phenotypic modulation of vascular smooth muscle cells (VSMCs) plays critical roles in the pathogenesis of aortic aneurysm (AA). The function of nuclear receptor corepressor1 (NCOR1) in regulation of VSMC phenotype and AA is unclear. Herein, using smooth muscle NCOR1 knockout mice, we demonstrated that smooth muscle NCOR1 deficiency decreased both mRNA and protein levels of contractile genes, impaired stress fibers formation and RhoA pathway activation, reduced synthesis of elastin and collagens, and induced the expression and activity of MMPs, manifesting a switch from contractile to degradative phenotype of VSMCs. NCOR1 modulated VSMC phenotype through 3 different mechanisms. First, NCOR1 deficiency increased acetylated FOXO3a to inhibit the expression of Myocd, which downregulated contractile genes. Second, deletion of NCOR1 derepressed NFAT5 to induce the expression of Rgs1, thus impeding RhoA activation. Third, NCOR1 deficiency increased the expression of Mmp12 and Mmp13 by derepressing ATF3. Finally, a mouse model combined apoE knockout mice with angiotensin II was used to study the role of smooth muscle NCOR1 in the development of AA. The results showed that smooth muscle NCOR1 deficiency increased the incidence of aortic aneurysms and exacerbated medial degeneration in angiotensin II-induced AA mouse model. Collectively, our data illustrated that NCOR1 interacts with FOXO3a, NFAT5, and ATF3 to maintain contractile phenotype of VSMCs and suppress AA development. Manipulation of smooth muscle NCOR1 may be a potential approach for AA treatment.


Asunto(s)
Aneurisma de la Aorta , Músculo Liso Vascular , Ratones , Animales , Músculo Liso Vascular/metabolismo , Angiotensina II/metabolismo , Aneurisma de la Aorta/metabolismo , Aneurisma de la Aorta/patología , Ratones Noqueados , Fenotipo , Ratones Noqueados para ApoE , Homeostasis , Células Cultivadas , Co-Represor 1 de Receptor Nuclear/metabolismo
8.
Sci Rep ; 12(1): 22504, 2022 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-36577770

RESUMEN

Patients with Marfan syndrome (MFS) develop thoracic aortic aneurysms as the aorta presents excessive elastin breaks, fibrosis, and vascular smooth muscle cell (vSMC) death due to mutations in the FBN1 gene. Despite elaborate vSMC to aortic endothelial cell (EC) signaling, the contribution of ECs to the development of aortic pathology remains largely unresolved. The aim of this study is to investigate the EC properties in Fbn1C1041G/+ MFS mice. Using en face immunofluorescence confocal microscopy, we showed that EC alignment with blood flow was reduced, EC roundness was increased, individual EC surface area was larger, and EC junctional linearity was decreased in aortae of Fbn1C1041G/+ MFS mice. This modified EC phenotype was most prominent in the ascending aorta and occurred before aortic dilatation. To reverse EC morphology, we performed treatment with resveratrol. This restored EC blood flow alignment, junctional linearity, phospho-eNOS expression, and improved the structural integrity of the internal elastic lamina of Fbn1C1041G/+ mice. In conclusion, these experiments identify the involvement of ECs and underlying internal elastic lamina in MFS aortic pathology, which could act as potential target for future MFS pharmacotherapies.


Asunto(s)
Aneurisma de la Aorta , Enfermedades de la Aorta , Síndrome de Marfan , Ratones , Animales , Aneurisma de la Aorta/metabolismo , Resveratrol/farmacología , Resveratrol/metabolismo , Síndrome de Marfan/genética , Aorta/metabolismo , Enfermedades de la Aorta/metabolismo , Fibrilina-1/genética , Fibrilina-1/metabolismo
9.
Cells ; 11(24)2022 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-36552822

RESUMEN

Vascular smooth muscle cells (VSMCs), the major cell type in the arterial vessel wall, have a contractile phenotype that maintains the normal vessel structure and function under physiological conditions. In response to stress or vascular injury, contractile VSMCs can switch to a less differentiated state (synthetic phenotype) to acquire the proliferative, migratory, and synthetic capabilities for tissue reparation. Imbalances in VSMCs phenotypic switching can result in a variety of cardiovascular diseases, including atherosclerosis, in-stent restenosis, aortic aneurysms, and vascular calcification. It is very important to identify the molecular mechanisms regulating VSMCs phenotypic switching to prevent and treat cardiovascular diseases with high morbidity and mortality. However, the key molecular mechanisms and signaling pathways participating in VSMCs phenotypic switching have still not been fully elucidated despite long-term efforts by cardiovascular researchers. In this review, we provide an updated summary of the recent studies and systematic knowledge of VSMCs phenotypic switching in atherosclerosis, in-stent restenosis, aortic aneurysms, and vascular calcification, which may help guide future research and provide novel insights into the prevention and treatment of related diseases.


Asunto(s)
Aneurisma de la Aorta , Aterosclerosis , Enfermedades Cardiovasculares , Reestenosis Coronaria , Calcificación Vascular , Humanos , Enfermedades Cardiovasculares/terapia , Enfermedades Cardiovasculares/metabolismo , Músculo Liso Vascular/metabolismo , Proliferación Celular , Reestenosis Coronaria/metabolismo , Fenotipo , Calcificación Vascular/metabolismo , Aneurisma de la Aorta/metabolismo , Aterosclerosis/metabolismo
10.
Life Sci ; 311(Pt A): 121159, 2022 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-36368416

RESUMEN

Alpha-Lipoic acid (ALA) plays a protective role in a variety of vascular diseases, however, its effect on aortic aneurysm and dissection (AAD) has not been reported. In this study, we found that Alpha-Lipoic Acid treatment significantly improved the AAD and AAA development, which was demonstrated by ameliorated aneurysmal dilation, decreased aortic dissection and aneurysm incidence, improved aortic morphology and inhibited elastin degradation. ALA blunted extra-cellular matrix degradation, vascular smooth muscle cell (VSMC) loss and phenotype transformation. Moreover, the protective effect of ALA on VSMCs may be related to the amelioration of mitochondrial dysfunction. In conclusion, our study revealed that ALA exerts inhibitory effects against progression of AAD, thus suggesting that ALA may be a novel therapeutic molecule for AAD.


Asunto(s)
Aneurisma de la Aorta , Disección Aórtica , Ácido Tióctico , Humanos , Músculo Liso Vascular/metabolismo , Ácido Tióctico/farmacología , Ácido Tióctico/uso terapéutico , Miocitos del Músculo Liso/metabolismo , Disección Aórtica/metabolismo , Aneurisma de la Aorta/metabolismo
11.
Arterioscler Thromb Vasc Biol ; 42(11): 1355-1374, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36172868

RESUMEN

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.


Asunto(s)
Aneurisma de la Aorta Torácica , Aneurisma de la Aorta , Humanos , Estudio de Asociación del Genoma Completo , Músculo Liso Vascular/metabolismo , Actinina/genética , ARN Nuclear/metabolismo , Aorta/patología , Miocitos del Músculo Liso/metabolismo , Aneurisma de la Aorta Torácica/patología , Aneurisma de la Aorta/metabolismo , Análisis de Secuencia de ARN , Factor de Crecimiento Transformador beta/metabolismo
12.
Arterioscler Thromb Vasc Biol ; 42(9): 1154-1168, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35861960

RESUMEN

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.


Asunto(s)
Aneurisma de la Aorta Torácica , Aneurisma de la Aorta , Síndrome de Marfan , Animales , Aneurisma de la Aorta/genética , Aneurisma de la Aorta/metabolismo , Aneurisma de la Aorta Torácica/genética , Cromatina , Humanos , Síndrome de Marfan/complicaciones , Síndrome de Marfan/genética , Síndrome de Marfan/metabolismo , Ratones , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , ARN , Transposasas/genética
13.
Tissue Cell ; 77: 101810, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35653909

RESUMEN

Bone marrow-derived endothelial progenitor cells (EPCs) have been proven to participate in the recovery of aortic aneurysm. CXCL12 promotes EPC recruitment and revascularization. Therefore, this study was dedicated to fathoming out whether EPCs regulated by CXCL12 can participate in the recovery of aneurysm. The morphology and maker protein activity of EPCs derived from bone marrow were separately detected by microscopic examination and immunofluorescence. Reverse transcription quantitative polymerase chain reaction (RT-qPCR) and western blot were performed to detect the transfection efficiency of CXCL12. Lumen formation, viability, and migration were determined by lumen formation, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), and wound-healing assays, respectively. The aneurysm tissues in rat model of artery aneurysm were used for histopathological observation by haematoxylin-eosin (HE) staining, α-SMA was detected by immunohistochemistry, and the expressions of VEGF, IGF1, FGF, PDGF and matrix metalloproteinase-2/9 (MMP-2/9) were measured by RT-qPCR and western blot assays. As a result, some EPCs showed fusiform on day 6, while others showed a cord-like structure. On day 17, cells presented a paving stone-like pattern. The expressions of CD34, CD133 and VEGFR2 were positive. CXCL12 overexpression promoted lumen formation, viability and migration of EPCs, and further facilitated growth of smooth muscle-like cells in the aneurysm cavity and VEGF expression, while inhibiting MMP-2/9 expression in the aneurysm tissues in vivo. The present study suggested that CXCL12 overexpression strengthened the lumen formation, viability, and migration of EPCs, and CXCL12-primed EPCs had better effects on aneurysm recovery than unmodified EPCs did.


Asunto(s)
Aneurisma de la Aorta , Células Progenitoras Endoteliales , Animales , Aneurisma de la Aorta/metabolismo , Médula Ósea/metabolismo , Movimiento Celular , Células Cultivadas , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Quimiocina CXCL12/farmacología , Células Progenitoras Endoteliales/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 2 de la Matriz/farmacología , Ratas , Factor A de Crecimiento Endotelial Vascular/metabolismo
14.
Arterioscler Thromb Vasc Biol ; 42(7): 811-818, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35587695

RESUMEN

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.


Asunto(s)
Aneurisma de la Aorta Abdominal , Aneurisma de la Aorta Torácica , Aneurisma de la Aorta , Aneurisma de la Aorta/metabolismo , Aneurisma de la Aorta Abdominal/metabolismo , Aneurisma de la Aorta Torácica/patología , Células Endoteliales/metabolismo , Humanos , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo
15.
J Clin Invest ; 132(9)2022 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-35324479

RESUMEN

As blood transitions from steady laminar flow (S-flow) in healthy arteries to disturbed flow (D-flow) in aneurysmal arteries, platelets are subjected to external forces. Biomechanical platelet activation is incompletely understood and is a potential mechanism behind antiplatelet medication resistance. Although it has been demonstrated that antiplatelet drugs suppress the growth of abdominal aortic aneurysms (AAA) in patients, we found that a certain degree of platelet reactivity persisted in spite of aspirin therapy, urging us to consider additional antiplatelet therapeutic targets. Transcriptomic profiling of platelets from patients with AAA revealed upregulation of a signal transduction pathway common to olfactory receptors, and this was explored as a mediator of AAA progression. Healthy platelets subjected to D-flow ex vivo, platelets from patients with AAA, and platelets in murine models of AAA demonstrated increased membrane olfactory receptor 2L13 (OR2L13) expression. A drug screen identified a molecule activating platelet OR2L13, which limited both biochemical and biomechanical platelet activation as well as AAA growth. This observation was further supported by selective deletion of the OR2L13 ortholog in a murine model of AAA that accelerated aortic aneurysm growth and rupture. These studies revealed that olfactory receptors regulate platelet activation in AAA and aneurysmal progression through platelet-derived mediators of aortic remodeling.


Asunto(s)
Aneurisma de la Aorta Abdominal , Aneurisma de la Aorta , Receptores Odorantes , Animales , Aneurisma de la Aorta/genética , Aneurisma de la Aorta/metabolismo , Aneurisma de la Aorta Abdominal/genética , Plaquetas/metabolismo , Modelos Animales de Enfermedad , Humanos , Ratones , Activación Plaquetaria , Inhibidores de Agregación Plaquetaria/uso terapéutico , Receptores Odorantes/genética
16.
IUBMB Life ; 74(10): 982-994, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35293116

RESUMEN

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.


Asunto(s)
Aneurisma de la Aorta , Trombospondinas , Aneurisma de la Aorta/genética , Aneurisma de la Aorta/metabolismo , Aneurisma de la Aorta/patología , Biomarcadores , Proteínas de la Matriz Extracelular , Humanos , Trombospondinas/genética
17.
Hypertension ; 79(3): e42-e55, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35138869

RESUMEN

BACKGROUND: CCN2 (cellular communication network factor 2) is a matricellular protein involved in cell communication and microenvironmental signaling responses. CCN2 is known to be overexpressed in several cardiovascular diseases, but its role is not completely understood. METHODS: Here, CCN2 involvement in aortic wall homeostasis and response to vascular injury was investigated in inducible <i>Ccn2</i>-deficient mice, with induction of vascular damage by infusion of Ang II (angiotensin II; 15 days), which is known to upregulate CCN2 expression in the aorta. RESULTS: Ang II infusion in CCN2-silenced mice lead to 60% mortality within 10 days due to rapid development and rupture of aortic aneurysms, as evidenced by magnetic resonance imaging, echography, and histological examination. <i>Ccn2</i> deletion decreased systolic blood pressure and caused aortic structural and functional changes, including elastin layer disruption, smooth muscle cell alterations, augmented distensibility, and increased metalloproteinase activity, which were aggravated by Ang II administration. Gene ontology analysis of RNA sequencing data identified aldosterone biosynthesis as one of the most enriched terms in CCN2-deficient aortas. Consistently, treatment with the mineralocorticoid receptor antagonist spironolactone before and during Ang II infusion reduced aneurysm formation and mortality, underscoring the importance of the aldosterone pathway in Ang II-induced aorta pathology. CONCLUSIONS: CCN2 is critically involved in the functional and structural homeostasis of the aorta and in maintenance of its integrity under Ang II-induced stress, at least, in part, by disruption of the aldosterone pathway. Thus, this study opens new avenues to future studies in disorders associated to vascular pathologies.


Asunto(s)
Aorta/metabolismo , Aneurisma de la Aorta/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Angiotensina II/farmacología , Animales , Aorta/efectos de los fármacos , Aneurisma de la Aorta/genética , Factor de Crecimiento del Tejido Conjuntivo/genética , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
18.
J Immunol Res ; 2022: 7538649, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35211631

RESUMEN

BACKGROUND: The leading cause of mortality in patients with Marfan syndrome (MFS) is thoracic aortic aneurysm and dissection. Notch signaling is essential for vessel morphogenesis and function. However, the role of Notch signaling in aortic pathology and aortic smooth muscle cell (SMC) differentiation in Marfan syndrome (MFS) is not completely understood. METHODS: RNA-sequencing on ascending aortic tissue from a mouse model of MFS, Fbn1mgR/mgR , and wild-type controls was performed. Notch 3 expression and activation in aortic tissue were confirmed with real-time RT-PCR, immunohistochemistry, and Western blot. Fbn1mgR/mgR and wild-type mice were treated with a γ-secretase inhibitor, DAPT, to block Notch activation. Aortic aneurysms and rupture were evaluated with connective tissue staining, ultrasound, and life table analysis. RESULTS: The murine RNA-sequencing data were validated with mouse and human MFS aortic tissue, demonstrating elevated Notch3 activation in MFS. Data further revealed that upregulation and activation of Notch3 were concomitant with increased expression of SMC contractile markers. Inhibiting Notch3 activation with DAPT attenuated aortic enlargement and improved survival of Fbn1mgR/mgR mice. DAPT treatment reduced elastin fiber fragmentation in the aorta and reversed the differentiation of SMCs. CONCLUSIONS: Our data demonstrated that matrix abnormalities in the aorta of MFS are associated with increased Notch3 activation. Enhanced Notch3 activation in MFS contributed to aortic aneurysm formation in MFS. This might be mediated by inducing a contractile phenotypic change of SMC. Our results suggest that inhibiting Notch3 activation may provide a strategy to prevent and treat aortic aneurysms in MFS.


Asunto(s)
Aorta/patología , Aneurisma de la Aorta/metabolismo , Síndrome de Marfan/metabolismo , Miocitos del Músculo Liso/fisiología , Receptor Notch3/metabolismo , Animales , Aneurisma de la Aorta/genética , Diaminas/administración & dosificación , Diaminas/farmacología , Modelos Animales de Enfermedad , Elastina/metabolismo , Fibrilina-1/genética , Fibrilina-1/metabolismo , Humanos , Síndrome de Marfan/genética , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Terapia Molecular Dirigida , Receptor Notch3/antagonistas & inhibidores , Tiazoles/administración & dosificación , Tiazoles/farmacología
19.
Nat Commun ; 13(1): 512, 2022 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-35082286

RESUMEN

Mechanical overload of the vascular wall is a pathological hallmark of life-threatening abdominal aortic aneurysms (AAA). However, how this mechanical stress resonates at the unicellular level of vascular smooth muscle cells (VSMC) is undefined. Here we show defective mechano-phenotype signatures of VSMC in AAA measured with ultrasound tweezers-based micromechanical system and single-cell RNA sequencing technique. Theoretical modelling predicts that cytoskeleton alterations fuel cell membrane tension of VSMC, thereby modulating their mechanoallostatic responses which are validated by live micromechanical measurements. Mechanistically, VSMC gradually adopt a mechanically solid-like state by upregulating cytoskeleton crosslinker, α-actinin2, in the presence of AAA-promoting signal, Netrin-1, thereby directly powering the activity of mechanosensory ion channel Piezo1. Inhibition of Piezo1 prevents mice from developing AAA by alleviating pathological vascular remodeling. Our findings demonstrate that deviations of mechanosensation behaviors of VSMC is detrimental for AAA and identifies Piezo1 as a novel culprit of mechanically fatigued aorta in AAA.


Asunto(s)
Aneurisma de la Aorta/metabolismo , Canales Iónicos/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Aneurisma , Animales , Aorta Abdominal , Aneurisma de la Aorta/patología , Aneurisma de la Aorta Abdominal/metabolismo , Ingeniería Biomédica , Fenómenos Biofísicos , Modelos Animales de Enfermedad , Canales Iónicos/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Netrina-1/metabolismo , Fenotipo , Estrés Mecánico , Remodelación Vascular
20.
Ann Vasc Surg ; 80: 345-357, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34780941

RESUMEN

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.


Asunto(s)
Aneurisma de la Aorta/genética , Enfermedad de la Arteria Coronaria/genética , Predisposición Genética a la Enfermedad , Polimorfismo de Nucleótido Simple , ARN Largo no Codificante/genética , Aneurisma de la Aorta/diagnóstico , Aneurisma de la Aorta/metabolismo , Enfermedad de la Arteria Coronaria/diagnóstico , Enfermedad de la Arteria Coronaria/metabolismo , Predisposición Genética a la Enfermedad/etnología , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...