Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Angiogenesis ; 2024 May 23.
Article in English | MEDLINE | ID: mdl-38780883

ABSTRACT

The presence of atherosclerotic plaque vessels is a critical factor in plaque destabilization. This may be attributable to the leaky phenotype of these microvessels, although direct proof for this notion is lacking. In this study, we investigated molecular and cellular patterns of stable and hemorrhaged human plaque to identify novel drivers of intraplaque vessel dysfunction. From transcriptome data of a human atherosclerotic lesion cohort, we reconstructed a co-expression network, identifying a gene module strongly and selectively correlated with both plaque microvascular density and inflammation. Spectrin Beta Non-Erythrocytic 1 (sptbn1) was identified as one of the central hubs of this module (along with zeb1 and dock1) and was selected for further study based on its predominant endothelial expression. Silencing of sptbn1 enhanced leukocyte transmigration and vascular permeability in vitro, characterized by an increased number of focal adhesions and reduced junctional VE-cadherin. In vivo, sptbn1 knockdown in zebrafish impaired the development of the caudal vein plexus. Mechanistically, increased substrate stiffness was associated with sptbn1 downregulation in endothelial cells in vitro and in human vessels. Plaque SPTBN1 mRNA and protein expression were found to correlate with an enhanced presence of intraplaque hemorrhage and future cardiovascular disease (CVD) events during follow-up. In conclusion, we identify SPTBN1 as a central hub gene in a gene program correlating with plaque vascularisation. SPTBN1 was regulated by substrate stiffness in vitro while silencing blocked vascular development in vivo, and compromised barrier function in vitro. Together, SPTBN1 is identified as a new potential regulator of the leaky phenotype of atherosclerotic plaque microvessels.

2.
Circ Res ; 127(7): 911-927, 2020 09 11.
Article in English | MEDLINE | ID: mdl-32564697

ABSTRACT

RATIONALE: Vascular calcification, the formation of calcium phosphate crystals in the vessel wall, is mediated by vascular smooth muscle cells (VSMCs). However, the underlying molecular mechanisms remain elusive, precluding mechanism-based therapies. OBJECTIVE: Phenotypic switching denotes a loss of contractile proteins and an increase in migration and proliferation, whereby VSMCs are termed synthetic. We examined how VSMC phenotypic switching influences vascular calcification and the possible role of the uniquely calcium-dependent reactive oxygen species (ROS)-forming Nox5 (NADPH oxidase 5). METHODS AND RESULTS: In vitro cultures of synthetic VSMCs showed decreased expression of contractile markers CNN-1 (calponin 1), α-SMA (α-smooth muscle actin), and SM22-α (smooth muscle protein 22α) and an increase in synthetic marker S100A4 (S100 calcium binding protein A4) compared with contractile VSMCs. This was associated with increased calcification of synthetic cells in response to high extracellular Ca2+. Phenotypic switching was accompanied by increased levels of ROS and Ca2+-dependent Nox5 in synthetic VSMCs. Nox5 itself regulated VSMC phenotype as siRNA knockdown of Nox5 increased contractile marker expression and decreased calcification, while overexpression of Nox5 decreased contractile marker expression. ROS production in synthetic VSMCs was cytosolic Ca2+-dependent, in line with it being mediated by Nox5. Treatment of VSMCs with Ca2+ loaded extracellular vesicles (EVs) lead to an increase in cytosolic Ca2+. Inhibiting EV endocytosis with dynasore blocked the increase in cytosolic Ca2+ and VSMC calcification. Increased ROS production resulted in increased EV release and decreased phagocytosis by VSMCs. CONCLUSIONS: We show here that contractile VSMCs are resistant to calcification and identify Nox5 as a key regulator of VSMC phenotypic switching. Additionally, we describe a new mechanism of Ca2+ uptake via EVs and show that Ca2+ induces ROS production in VSMCs via Nox5. ROS production is required for release of EVs, which promote calcification. Identifying molecular pathways that control Nox5 and VSMC-derived EVs provides potential targets to modulate vascular remodeling and calcification in the context of mineral imbalance. Graphic Abstract: A graphic abstract is available for this article.


Subject(s)
Cell Movement , Cell Proliferation , Extracellular Vesicles/enzymology , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , NADPH Oxidase 5/metabolism , Reactive Oxygen Species/metabolism , Vascular Calcification/enzymology , Aged , Aged, 80 and over , Animals , Cells, Cultured , Extracellular Vesicles/genetics , Extracellular Vesicles/pathology , Female , Humans , Male , Middle Aged , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , NADPH Oxidase 5/genetics , Phagocytosis , Phenotype , Signal Transduction , Sus scrofa , Vascular Calcification/genetics , Vascular Calcification/pathology
3.
PLoS Genet ; 16(1): e1008538, 2020 01.
Article in English | MEDLINE | ID: mdl-31917787

ABSTRACT

Genome-wide association studies have identified multiple novel genomic loci associated with vascular diseases. Many of these loci are common non-coding variants that affect the expression of disease-relevant genes within coronary vascular cells. To identify such genes on a genome-wide level, we performed deep transcriptomic analysis of genotyped primary human coronary artery smooth muscle cells (HCASMCs) and coronary endothelial cells (HCAECs) from the same subjects, including splicing Quantitative Trait Loci (sQTL), allele-specific expression (ASE), and colocalization analyses. We identified sQTLs for TARS2, YAP1, CFDP1, and STAT6 in HCASMCs and HCAECs, and 233 ASE genes, a subset of which are also GTEx eGenes in arterial tissues. Colocalization of GWAS association signals for coronary artery disease (CAD), migraine, stroke and abdominal aortic aneurysm with GTEx eGenes in aorta, coronary artery and tibial artery discovered novel candidate risk genes for these diseases. At the CAD and stroke locus tagged by rs2107595 we demonstrate colocalization with expression of the proximal gene TWIST1. We show that disrupting the rs2107595 locus alters TWIST1 expression and that the risk allele has increased binding of the NOTCH signaling protein RBPJ. Finally, we provide data that TWIST1 expression influences vascular SMC phenotypes, including proliferation and calcification, as a potential mechanism supporting a role for TWIST1 in CAD.


Subject(s)
Coronary Vessels/metabolism , Endothelial Cells/metabolism , Myocytes, Smooth Muscle/metabolism , Nuclear Proteins/genetics , Twist-Related Protein 1/genetics , Vascular Diseases/genetics , Cells, Cultured , Coronary Vessels/cytology , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Nuclear Proteins/metabolism , Polymorphism, Single Nucleotide , Protein Binding , Transcriptome , Twist-Related Protein 1/metabolism
5.
Cardiovasc Res ; 115(1): 230-242, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30107531

ABSTRACT

Aims: Long non-coding RNAs (lncRNAs) have been shown to regulate numerous processes in the human genome, but the function of these transcripts in vascular aging is largely unknown. We aim to characterize the expression of lncRNAs in endothelial aging and analyse the function of the highly conserved lncRNA H19. Methods and results: H19 was downregulated in endothelium of aged mice. In human, atherosclerotic plaques H19 was mainly expressed by endothelial cells and H19 was significantly reduced in comparison to healthy carotid artery biopsies. Loss of H19 led to an upregulation of p16 and p21, reduced proliferation and increased senescence in vitro. Depletion of H19 in aortic rings of young mice inhibited sprouting capacity. We generated endothelial-specific inducible H19 deficient mice (H19iEC-KO), resulting in increased systolic blood pressure compared with control littermates (Ctrl). These H19iEC-KO and Ctrl mice were subjected to hindlimb ischaemia, which showed reduced capillary density in H19iEC-KO mice. Mechanistically, exon array analysis revealed an involvement of H19 in IL-6 signalling. Accordingly, intercellular adhesion molecule 1 and vascular cell adhesion molecule 1 were upregulated upon H19 depletion. A luciferase reporter screen for differential transcription factor activity revealed STAT3 as being induced upon H19 depletion and repressed after H19 overexpression. Furthermore, depletion of H19 increased the phosphorylation of STAT3 at TYR705 and pharmacological inhibition of STAT3 activation abolished the effects of H19 silencing on p21 and vascular cell adhesion molecule 1 expression as well as proliferation. Conclusion: These data reveal a pivotal role for the lncRNA H19 in controlling endothelial cell aging.


Subject(s)
Carotid Artery Diseases/metabolism , Cellular Senescence , Endothelial Cells/metabolism , Ischemia/metabolism , Muscle, Skeletal/blood supply , RNA, Long Noncoding/metabolism , STAT3 Transcription Factor/metabolism , Animals , Carotid Artery Diseases/genetics , Carotid Artery Diseases/pathology , Case-Control Studies , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Disease Models, Animal , Endothelial Cells/pathology , Female , Hindlimb , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Ischemia/genetics , Ischemia/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Physiologic , Phosphorylation , Plaque, Atherosclerotic , RNA, Long Noncoding/genetics , Signal Transduction , Vascular Cell Adhesion Molecule-1/metabolism
6.
Sci Rep ; 8(1): 14668, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30279450

ABSTRACT

The intermediate filament protein nestin is expressed during embryonic development, but considered largely restricted to areas of regeneration in the adult. Here, we perform a body-wide transcriptome and protein-profiling analysis to reveal that nestin is constitutively, and highly-selectively, expressed in adult human endothelial cells (EC), independent of proliferative status. Correspondingly, we demonstrate that it is not a marker for tumour EC in multiple malignancy types. Imaging of EC from different vascular beds reveals nestin subcellular distribution is shear-modulated. siRNA inhibition of nestin increases EC proliferation, and nestin expression is reduced in atherosclerotic plaque neovessels. eQTL analysis reveals an association between SNPs linked to cardiovascular disease and reduced aortic EC nestin mRNA expression. Our study challenges the dogma that nestin is a marker of proliferation, and provides insight into its regulation and function in EC. Furthermore, our systems-based approach can be applied to investigate body-wide expression profiles of any candidate protein.


Subject(s)
Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Nestin/physiology , Systems Biology/methods , Adult , Aorta/cytology , Aorta/pathology , Cardiovascular Diseases/genetics , Cardiovascular Diseases/pathology , Cell Movement , Cell Proliferation , Cells, Cultured , Databases, Genetic/statistics & numerical data , Datasets as Topic , Endothelium, Vascular/cytology , Endothelium, Vascular/pathology , Female , Gene Expression Profiling/methods , Haplotypes , Humans , Male , Neoplasms/pathology , Neovascularization, Pathologic/pathology , Polymorphism, Single Nucleotide , Primary Cell Culture , Proteomics/methods , Quantitative Trait Loci , RNA, Small Interfering/metabolism , Tissue Array Analysis/methods
7.
Circ Genom Precis Med ; 11(9): e002115, 2018 09.
Article in English | MEDLINE | ID: mdl-30354329

ABSTRACT

BACKGROUND: Atherosclerosis is a chronic inflammatory disease in part caused by lipid uptake in the vascular wall, but the exact underlying mechanisms leading to acute myocardial infarction and stroke remain poorly understood. Large consortia identified genetic susceptibility loci that associate with large artery ischemic stroke and coronary artery disease. However, deciphering their underlying mechanisms are challenging. Histological studies identified destabilizing characteristics in human atherosclerotic plaques that associate with clinical outcome. To what extent established susceptibility loci for large artery ischemic stroke and coronary artery disease relate to plaque characteristics is thus far unknown but may point to novel mechanisms. METHODS: We studied the associations of 61 established cardiovascular risk loci with 7 histological plaque characteristics assessed in 1443 carotid plaque specimens from the Athero-Express Biobank Study. We also assessed if the genotyped cardiovascular risk loci impact the tissue-specific gene expression in 2 independent biobanks, Biobank of Karolinska Endarterectomy and Stockholm Atherosclerosis Gene Expression. RESULTS: A total of 21 established risk variants (out of 61) nominally associated to a plaque characteristic. One variant (rs12539895, risk allele A) at 7q22 associated to a reduction of intraplaque fat, P=5.09×10-6 after correction for multiple testing. We further characterized this 7q22 Locus and show tissue-specific effects of rs12539895 on HBP1 expression in plaques and COG5 expression in whole blood and provide data from public resources showing an association with decreased LDL (low-density lipoprotein) and increase HDL (high-density lipoprotein) in the blood. CONCLUSIONS: Our study supports the view that cardiovascular susceptibility loci may exert their effect by influencing the atherosclerotic plaque characteristics.


Subject(s)
Atherosclerosis/genetics , Cardiovascular Diseases/genetics , Genetic Loci/genetics , Genetic Predisposition to Disease/genetics , Polymorphism, Single Nucleotide , Adaptor Proteins, Vesicular Transport/genetics , Aged , Alleles , Carotid Artery Diseases/genetics , Female , Gene Expression Regulation , Genotype , High Mobility Group Proteins/genetics , Humans , Male , Middle Aged , Plaque, Atherosclerotic/genetics , Repressor Proteins/genetics
8.
Am J Physiol Cell Physiol ; 315(6): C873-C884, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30332284

ABSTRACT

The endothelin type B receptor (ETB or EDNRB) is highly plastic and is upregulated in smooth muscle cells (SMCs) by arterial injury and following organ culture in vitro. We hypothesized that this transcriptional plasticity may arise, in part, because EDNRB is controlled by a balance of transcriptional inputs from myocardin-related transcription factors (MRTFs) and ternary complex factors (TCFs). We found significant positive correlations between the TCFs ELK3 and FLI1 versus EDNRB in human arteries. The MRTF MKL2 also correlated with EDNRB. Overexpression of ELK3, FLI1, and MKL2 in human coronary artery SMCs promoted expression of EDNRB, and the effect of MKL2 was antagonized by myocardin (MYOCD), which also correlated negatively with EDNRB at the tissue level. Silencing of MKL2 reduced basal EDNRB expression, but depolymerization of actin using latrunculin B (LatB) or overexpression of constitutively active cofilin, as well as treatment with the Rho-associated kinase (ROCK) inhibitor Y27632, increased EDNRB in a MEK/ERK-dependent fashion. Transcript-specific primers indicated that the second EDNRB transcript (EDNRB_2) was targeted, but this promoter was largely unresponsive to LatB and was inhibited rather than stimulated by MKL2 and FLI1, suggesting distant control elements or an indirect effect. LatB also reduced expression of endothelin-1, but supplementation experiments argued that this was not the cause of EDNRB induction. EDNRB finally changed in parallel with ELK3 and FLI1 in rat and human carotid artery lesions. These studies implicate the actin cytoskeleton and ELK3, FLI1, and MKL2 in the transcriptional control of EDNRB and increase our understanding of the plasticity of this receptor.


Subject(s)
Actin Cytoskeleton/genetics , Carotid Artery Injuries/genetics , Proto-Oncogene Proteins/genetics , Receptor, Endothelin B/genetics , Transcription Factors/genetics , Actin Cytoskeleton/metabolism , Actin Depolymerizing Factors/pharmacology , Amides/pharmacology , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Carotid Artery Injuries/metabolism , Carotid Artery Injuries/pathology , Endothelin-1/genetics , Gene Expression Regulation, Developmental/drug effects , Humans , Myocytes, Smooth Muscle/metabolism , Nuclear Proteins/genetics , Proto-Oncogene Protein c-fli-1/genetics , Proto-Oncogene Proteins c-ets , Pyridines/pharmacology , Rats , Ternary Complex Factors/genetics , Thiazolidines/pharmacology , Trans-Activators/genetics , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/genetics
9.
Sci Rep ; 8(1): 13025, 2018 08 29.
Article in English | MEDLINE | ID: mdl-30158653

ABSTRACT

Nexilin, encoded by the NEXN gene, is expressed in striated muscle and localizes to Z-discs, influencing mechanical stability. We examined Nexilin/NEXN in smooth muscle cells (SMCs), and addressed if Nexilin localizes to dense bodies and dense bands and whether it is regulated by actin-controlled coactivators from the MRTF (MYOCD, MKL1, MKL2) and YAP/TAZ (YAP1 and WWTR1) families. NEXN expression in SMCs was comparable to that in striated muscles. Immunofluorescence and immunoelectron microscopy suggested that Nexilin localizes to dense bodies and dense bands. Correlations at the mRNA level suggested that NEXN expression might be controlled by actin polymerization. Depolymerization of actin using Latrunculin B repressed the NEXN mRNA and protein in bladder and coronary artery SMCs. Overexpression and knockdown supported involvement of both YAP/TAZ and MRTFs in the transcriptional control of NEXN. YAP/TAZ and MRTFs appeared equally important in bladder SMCs, whereas MRTFs dominated in vascular SMCs. Expression of NEXN was moreover reduced in situations of SMC phenotypic modulation in vivo. The proximal promoter of NEXN conferred control by MRTF-A/MKL1 and MYOCD. NEXN silencing reduced actin polymerization and cell migration, as well as SMC marker expression. NEXN targeting by actin-controlled coactivators thus amplifies SMC differentiation through the actin cytoskeleton, probably via dense bodies and dense bands.


Subject(s)
Actins/metabolism , Microfilament Proteins/metabolism , Myocytes, Smooth Muscle/metabolism , Protein Multimerization , Adaptor Proteins, Signal Transducing/metabolism , Cell Differentiation , Cells, Cultured , Gene Expression Profiling , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Microscopy, Fluorescence , Microscopy, Immunoelectron , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , RNA, Messenger/analysis , Trans-Activators/metabolism , Transcription Factors/metabolism , Transcriptional Coactivator with PDZ-Binding Motif Proteins , YAP-Signaling Proteins
10.
Cardiovasc Res ; 113(1): 30-39, 2017 01.
Article in English | MEDLINE | ID: mdl-27864310

ABSTRACT

AIMS: Processes in the development of atherosclerotic lesions can lead to plaque rupture or erosion, which can in turn elicit myocardial infarction or ischaemic stroke. The aims of this study were to determine whether Toll-like receptor 7 (TLR7) gene expression levels influence patient outcome and to explore the mechanisms linked to TLR7 expression in atherosclerosis. METHODS AND RESULTS: Atherosclerotic plaques were removed by carotid endarterectomy (CEA) and subjected to gene array expression analysis (n = 123). Increased levels of TLR7 transcript in the plaques were associated with better outcome in a follow-up study over a maximum of 8 years. Patients with higher TLR7 transcript levels had a lower risk of experiencing major cardiovascular and cerebrovascular events (MACCE) during the follow-up period after CEA (hazard ratio: 2.38, P = 0.012, 95% CI 1.21-4.67). TLR7 was expressed in all plaques by T cells, macrophages and endothelial cells in capillaries, as shown by immunohistochemistry. In short-term tissue cultures, ex vivo treatment of plaques with the TLR7 ligand imiquimod elicited dose-dependent secretion of IL-10, TNF-α, GM-CSF, and IL-12/IL-23p40. This secretion was blocked with a TLR7 inhibitor. Immunofluorescent tissue analysis after TLR7 stimulation showed IL-10 expression in T cells, macrophages and vascular smooth muscle cells. TLR7 mRNA levels in the plaques were correlated with IL-10 receptor (r = 0.4031, P < 0.0001) and GM-CSF receptor A (r = 0.4354, P < 0.0001) transcripts. CONCLUSION: These findings demonstrate that TLR7 is abundantly expressed in human atherosclerotic plaques. TLR7 ligation elicits the secretion of pro-inflammatory and anti-inflammatory cytokines, and high TLR7 expression in plaques is associated with better patient outcome, suggesting that TLR7 is a potential therapeutic target for prevention of complications of atherosclerosis.


Subject(s)
Carotid Arteries/metabolism , Carotid Artery Diseases/metabolism , Cerebrovascular Disorders/metabolism , Heart Diseases/metabolism , Plaque, Atherosclerotic , Toll-Like Receptor 7/metabolism , Aged , Carotid Arteries/drug effects , Carotid Arteries/pathology , Carotid Artery Diseases/genetics , Carotid Artery Diseases/pathology , Carotid Artery Diseases/surgery , Case-Control Studies , Cells, Cultured , Cerebrovascular Disorders/genetics , Cerebrovascular Disorders/pathology , Cerebrovascular Disorders/prevention & control , Cytokines/metabolism , Disease-Free Survival , Endarterectomy, Carotid , Female , Heart Diseases/genetics , Heart Diseases/pathology , Heart Diseases/prevention & control , Humans , Inflammation Mediators/metabolism , Kaplan-Meier Estimate , Macrophages/metabolism , Male , Middle Aged , RNA, Messenger/genetics , RNA, Messenger/metabolism , Risk Factors , Signal Transduction , T-Lymphocytes/metabolism , Time Factors , Toll-Like Receptor 7/drug effects , Toll-Like Receptor 7/genetics , Transcriptome , Treatment Outcome
11.
Sci Adv ; 2(7): e1600224, 2016 07.
Article in English | MEDLINE | ID: mdl-27532042

ABSTRACT

Atherosclerosis results from maladaptive inflammation driven primarily by macrophages, whose recruitment and proliferation drive plaque progression. In advanced plaques, macrophage death contributes centrally to the formation of plaque necrosis, which underlies the instability that promotes plaque rupture and myocardial infarction. Hence, targeting macrophage cell death pathways may offer promise for the stabilization of vulnerable plaques. Necroptosis is a recently discovered pathway of programmed cell necrosis regulated by RIP3 and MLKL kinases that, in contrast to apoptosis, induces a proinflammatory state. We show herein that necroptotic cell death is activated in human advanced atherosclerotic plaques and can be targeted in experimental atherosclerosis for both therapeutic and diagnostic interventions. In humans with unstable carotid atherosclerosis, expression of RIP3 and MLKL is increased, and MLKL phosphorylation, a key step in the commitment to necroptosis, is detected in advanced atheromas. Investigation of the molecular mechanisms underlying necroptosis showed that atherogenic forms of low-density lipoprotein increase RIP3 and MLKL transcription and phosphorylation-two critical steps in the execution of necroptosis. Using a radiotracer developed with the necroptosis inhibitor necrostatin-1 (Nec-1), we show that (123)I-Nec-1 localizes specifically to atherosclerotic plaques in Apoe (-/-) mice, and its uptake is tightly correlated to lesion areas by ex vivo nuclear imaging. Furthermore, treatment of Apoe (-/-) mice with established atherosclerosis with Nec-1 reduced lesion size and markers of plaque instability, including necrotic core formation. Collectively, our findings offer molecular insight into the mechanisms of macrophage cell death that drive necrotic core formation in atherosclerosis and suggest that this pathway can be used as both a diagnostic and therapeutic tool for the treatment of unstable atherosclerosis.


Subject(s)
Apoptosis , Atherosclerosis/diagnosis , Atherosclerosis/therapy , Amino Acid Chloromethyl Ketones/toxicity , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Apoptosis/drug effects , Atherosclerosis/veterinary , Bone Marrow Cells/cytology , Cells, Cultured , Cholesterol/blood , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Coronary Vessels/pathology , Humans , Imidazoles/chemistry , Imidazoles/therapeutic use , Indoles/chemistry , Indoles/therapeutic use , Interleukin-1beta/blood , Lipoproteins, LDL/toxicity , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Necrosis/therapy , Protein Kinases/genetics , Protein Kinases/metabolism , Reactive Oxygen Species/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
12.
Nat Commun ; 7: 12092, 2016 07 08.
Article in English | MEDLINE | ID: mdl-27386823

ABSTRACT

Coronary artery disease (CAD) is the leading cause of mortality and morbidity, driven by both genetic and environmental risk factors. Meta-analyses of genome-wide association studies have identified >150 loci associated with CAD and myocardial infarction susceptibility in humans. A majority of these variants reside in non-coding regions and are co-inherited with hundreds of candidate regulatory variants, presenting a challenge to elucidate their functions. Herein, we use integrative genomic, epigenomic and transcriptomic profiling of perturbed human coronary artery smooth muscle cells and tissues to begin to identify causal regulatory variation and mechanisms responsible for CAD associations. Using these genome-wide maps, we prioritize 64 candidate variants and perform allele-specific binding and expression analyses at seven top candidate loci: 9p21.3, SMAD3, PDGFD, IL6R, BMP1, CCDC97/TGFB1 and LMOD1. We validate our findings in expression quantitative trait loci cohorts, which together reveal new links between CAD associations and regulatory function in the appropriate disease context.


Subject(s)
Chromatin/chemistry , Coronary Artery Disease/genetics , Genetic Predisposition to Disease , Genome, Human , Genomics/methods , Quantitative Trait Loci , Alleles , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Chromatin/metabolism , Coronary Artery Disease/metabolism , Coronary Artery Disease/pathology , Coronary Vessels/metabolism , Coronary Vessels/pathology , Gene Expression Profiling , Gene Expression Regulation , Genome-Wide Association Study , Humans , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Primary Cell Culture , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism
13.
Arterioscler Thromb Vasc Biol ; 36(5): 972-83, 2016 05.
Article in English | MEDLINE | ID: mdl-26966274

ABSTRACT

OBJECTIVE: A recent genome-wide association study meta-analysis identified an intronic single nucleotide polymorphism in SMAD3, rs56062135C>T, the minor allele (T) which associates with protection from coronary artery disease. Relevant to atherosclerosis, SMAD3 is a key contributor to transforming growth factor-ß pathway signaling. Here, we seek to identify ≥1 causal coronary artery disease-associated single nucleotide polymorphisms at the SMAD3 locus and characterize mechanisms whereby the risk allele(s) contribute to coronary artery disease risk. APPROACH AND RESULTS: By genetic and epigenetic fine mapping, we identified a candidate causal single nucleotide polymorphism rs17293632C>T (D', 0.97; r(2), 0.94 with rs56062135) in intron 1 of SMAD3 with predicted functional effects. We show that the sequence encompassing rs17293632 acts as a strong enhancer in human arterial smooth muscle cells. The common allele (C) preserves an activator protein (AP)-1 site and enhancer function, whereas the protective (T) allele disrupts the AP-1 site and significantly reduces enhancer activity (P<0.001). Pharmacological inhibition of AP-1 activity upstream demonstrates that this allele-specific enhancer effect is AP-1 dependent (P<0.001). Chromatin immunoprecipitation experiments reveal binding of several AP-1 component proteins with preferential binding to the (C) allele. We show that rs17293632 is an expression quantitative trait locus for SMAD3 in blood and atherosclerotic plaque with reduced expression of SMAD3 in carriers of the protective allele. Finally, siRNA knockdown of SMAD3 in human arterial smooth muscle cells increases cell viability, consistent with an antiproliferative role. CONCLUSIONS: The coronary artery disease-associated rs17293632C>T single nucleotide polymorphism represents a novel functional cis-acting element at the SMAD3 locus. The protective (T) allele of rs17293632 disrupts a consensus AP-1 binding site in a SMAD3 intron 1 enhancer, reduces enhancer activity and SMAD3 expression, altering human arterial smooth muscle cell proliferation.


Subject(s)
Coronary Artery Disease/genetics , Coronary Artery Disease/prevention & control , Polymorphism, Single Nucleotide , Smad3 Protein/genetics , Binding Sites , Cell Proliferation , Cell Survival , Cells, Cultured , Coronary Artery Disease/diagnosis , Coronary Artery Disease/metabolism , Coronary Vessels/metabolism , Coronary Vessels/pathology , Enhancer Elements, Genetic , Genetic Predisposition to Disease , Genome-Wide Association Study , Haplotypes , Humans , Introns , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Phenotype , Plaque, Atherosclerotic , Promoter Regions, Genetic , Protective Factors , Quantitative Trait Loci , RNA Interference , Risk Factors , Smad3 Protein/metabolism , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Transfection
14.
Arterioscler Thromb Vasc Biol ; 36(5): 942-951, 2016 05.
Article in English | MEDLINE | ID: mdl-26941018

ABSTRACT

OBJECTIVE: Cholesterol homeostasis is fundamental to human health and is, thus, tightly regulated. MicroRNAs exert potent effects on biological pathways, including cholesterol metabolism, by repressing genes with related functions. We reasoned that this mode of pathway regulation could be exploited to identify novel genes involved in cholesterol homeostasis. APPROACH AND RESULTS: Here, we identify oxysterol-binding protein-like 6 (OSBPL6) as a novel target of 2 miRNA hubs regulating cholesterol homeostasis: miR-33 and miR-27b. Characterization of OSBPL6 revealed that it is transcriptionally regulated in macrophages and hepatocytes by liver X receptor and in response to cholesterol loading and in mice and nonhuman primates by Western diet feeding. OSBPL6 encodes the OSBPL-related protein 6 (ORP6), which contains dual membrane- and endoplasmic reticulum-targeting motifs. Subcellular localization studies showed that ORP6 is associated with the endolysosomal network and endoplasmic reticulum, suggesting a role for ORP6 in cholesterol trafficking between these compartments. Accordingly, knockdown of OSBPL6 results in aberrant clustering of endosomes and promotes the accumulation of free cholesterol in these structures, resulting in reduced cholesterol esterification at the endoplasmic reticulum. Conversely, ORP6 overexpression enhances cholesterol trafficking and efflux in macrophages and hepatocytes. Moreover, we show that hepatic expression of OSBPL6 is positively correlated with plasma levels of high-density lipoprotein cholesterol in a cohort of 200 healthy individuals, whereas its expression is reduced in human atherosclerotic plaques. CONCLUSIONS: These studies identify ORP6 as a novel regulator of cholesterol trafficking that is part of the miR-33 and miR-27b target gene networks that contribute to the maintenance of cholesterol homeostasis.


Subject(s)
Atherosclerosis/metabolism , MicroRNAs/metabolism , Receptors, Steroid/metabolism , 3' Untranslated Regions , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter 1/metabolism , Animals , Atherosclerosis/genetics , Atherosclerosis/pathology , Binding Sites , Biological Transport , Chlorocebus aethiops , Cholesterol/metabolism , Cholesterol, HDL/blood , Disease Models, Animal , Endoplasmic Reticulum/metabolism , HEK293 Cells , Hep G2 Cells , Hepatocytes/metabolism , Humans , Liver X Receptors/genetics , Liver X Receptors/metabolism , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Plaque, Atherosclerotic , Protein Binding , RNA Interference , Receptors, LDL/deficiency , Receptors, LDL/genetics , Receptors, Steroid/genetics , Transcription, Genetic , Transfection
15.
Circ Res ; 118(2): 230-40, 2016 Jan 22.
Article in English | MEDLINE | ID: mdl-26596284

ABSTRACT

RATIONALE: Genetic variation at the chromosome 9p21 cardiovascular risk locus has been associated with peripheral artery disease, but its mechanism remains unknown. OBJECTIVE: To determine whether this association is secondary to an increase in atherosclerosis, or it is the result of a separate angiogenesis-related mechanism. METHODS AND RESULTS: Quantitative evaluation of human vascular samples revealed that carriers of the 9p21 risk allele possess a significantly higher burden of immature intraplaque microvessels than carriers of the ancestral allele, irrespective of lesion size or patient comorbidity. To determine whether aberrant angiogenesis also occurs under nonatherosclerotic conditions, we performed femoral artery ligation surgery in mice lacking the 9p21 candidate gene, Cdkn2b. These animals developed advanced hindlimb ischemia and digital autoamputation, secondary to a defect in the capacity of the Cdkn2b-deficient smooth muscle cell to support the developing neovessel. Microarray studies identified impaired transforming growth factor ß (TGFß) signaling in cultured cyclin-dependent kinase inhibitor 2B (CDKN2B)-deficient cells, as well as TGFß1 upregulation in the vasculature of 9p21 risk allele carriers. Molecular signaling studies indicated that loss of CDKN2B impairs the expression of the inhibitory factor, SMAD-7, which promotes downstream TGFß activation. Ultimately, this manifests in the upregulation of a poorly studied effector molecule, TGFß1-induced-1, which is a TGFß-rheostat known to have antagonistic effects on the endothelial cell and smooth muscle cell. Dual knockdown studies confirmed the reversibility of the proposed mechanism, in vitro. CONCLUSIONS: These results suggest that loss of CDKN2B may not only promote cardiovascular disease through the development of atherosclerosis but may also impair TGFß signaling and hypoxic neovessel maturation.


Subject(s)
Atherosclerosis/enzymology , Cyclin-Dependent Kinase Inhibitor p15/metabolism , Muscle, Skeletal/blood supply , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Neovascularization, Physiologic , Signal Transduction , Transforming Growth Factor beta1/metabolism , Animals , Atherosclerosis/genetics , Atherosclerosis/mortality , Atherosclerosis/pathology , Carotid Arteries/enzymology , Carotid Arteries/pathology , Cell Hypoxia , Cells, Cultured , Chromosomes, Human, Pair 9 , Coronary Vessels/enzymology , Coronary Vessels/pathology , Cyclin-Dependent Kinase Inhibitor p15/deficiency , Cyclin-Dependent Kinase Inhibitor p15/genetics , Disease Models, Animal , Female , Genetic Predisposition to Disease , Hindlimb , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/physiopathology , Neovascularization, Pathologic , Phenotype , RNA Interference , Smad7 Protein/metabolism , Time Factors , Transfection , Transforming Growth Factor beta1/genetics
17.
PLoS Genet ; 11(5): e1005155, 2015 May.
Article in English | MEDLINE | ID: mdl-26020946

ABSTRACT

Recent genome wide association studies have identified a number of genes that contribute to the risk for coronary heart disease. One such gene, TCF21, encodes a basic-helix-loop-helix transcription factor believed to serve a critical role in the development of epicardial progenitor cells that give rise to coronary artery smooth muscle cells (SMC) and cardiac fibroblasts. Using reporter gene and immunolocalization studies with mouse and human tissues we have found that vascular TCF21 expression in the adult is restricted primarily to adventitial cells associated with coronary arteries and also medial SMC in the proximal aorta of mouse. Genome wide RNA-Seq studies in human coronary artery SMC (HCASMC) with siRNA knockdown found a number of putative TCF21 downstream pathways identified by enrichment of terms related to CAD, including "vascular disease," "disorder of artery," and "occlusion of artery," as well as disease-related cellular functions including "cellular movement" and "cellular growth and proliferation." In vitro studies in HCASMC demonstrated that TCF21 expression promotes proliferation and migration and inhibits SMC lineage marker expression. Detailed in situ expression studies with reporter gene and lineage tracing revealed that vascular wall cells expressing Tcf21 before disease initiation migrate into vascular lesions of ApoE-/- and Ldlr-/- mice. While Tcf21 lineage traced cells are distributed throughout the early lesions, in mature lesions they contribute to the formation of a subcapsular layer of cells, and others become associated with the fibrous cap. The lineage traced fibrous cap cells activate expression of SMC markers and growth factor receptor genes. Taken together, these data suggest that TCF21 may have a role regulating the differentiation state of SMC precursor cells that migrate into vascular lesions and contribute to the fibrous cap and more broadly, in view of the association of this gene with human CAD, provide evidence that these processes may be a mechanism for CAD risk attributable to the vascular wall.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/genetics , Cell Proliferation/genetics , Coronary Artery Disease/genetics , Myocytes, Smooth Muscle/pathology , Animals , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Cell Lineage/genetics , Coronary Artery Disease/pathology , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , High-Throughput Nucleotide Sequencing , Humans , Mice , Myoblasts/metabolism , Myoblasts/pathology , Myocytes, Smooth Muscle/metabolism , Stem Cells
18.
Circ Res ; 117(3): 266-78, 2015 Jul 17.
Article in English | MEDLINE | ID: mdl-26002865

ABSTRACT

RATIONALE: Therapeutically targeting macrophage reverse cholesterol transport is a promising approach to treat atherosclerosis. Macrophage energy metabolism can significantly influence macrophage phenotype, but how this is controlled in foam cells is not known. Bioinformatic pathway analysis predicts that miR-33 represses a cluster of genes controlling cellular energy metabolism that may be important in macrophage cholesterol efflux. OBJECTIVE: We hypothesized that cellular energy status can influence cholesterol efflux from macrophages, and that miR-33 reduces cholesterol efflux via repression of mitochondrial energy metabolism pathways. METHODS AND RESULTS: In this study, we demonstrated that macrophage cholesterol efflux is regulated by mitochondrial ATP production, and that miR-33 controls a network of genes that synchronize mitochondrial function. Inhibition of mitochondrial ATP synthase markedly reduces macrophage cholesterol efflux capacity, and anti-miR33 required fully functional mitochondria to enhance ABCA1-mediated cholesterol efflux. Specifically, anti-miR33 derepressed the novel target genes PGC-1α, PDK4, and SLC25A25 and boosted mitochondrial respiration and production of ATP. Treatment of atherosclerotic Apoe(-/-) mice with anti-miR33 oligonucleotides reduced aortic sinus lesion area compared with controls, despite no changes in high-density lipoprotein cholesterol or other circulating lipids. Expression of miR-33a/b was markedly increased in human carotid atherosclerotic plaques compared with normal arteries, and there was a concomitant decrease in mitochondrial regulatory genes PGC-1α, SLC25A25, NRF1, and TFAM, suggesting these genes are associated with advanced atherosclerosis in humans. CONCLUSIONS: This study demonstrates that anti-miR33 therapy derepresses genes that enhance mitochondrial respiration and ATP production, which in conjunction with increased ABCA1 expression, works to promote macrophage cholesterol efflux and reduce atherosclerosis.


Subject(s)
Adenosine Triphosphate/biosynthesis , Atherosclerosis/metabolism , Cholesterol/metabolism , Macrophages, Peritoneal/metabolism , Macrophages/metabolism , MicroRNAs/antagonists & inhibitors , Mitochondria/metabolism , Oligonucleotides, Antisense/therapeutic use , Amino Acid Transport Systems, Acidic/biosynthesis , Amino Acid Transport Systems, Acidic/genetics , Animals , Apolipoproteins E/deficiency , Atherosclerosis/genetics , Atherosclerosis/therapy , Base Sequence , Calcium-Binding Proteins/biosynthesis , Calcium-Binding Proteins/genetics , Cell Line , Gene Expression Regulation/drug effects , Genetic Therapy , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Mitochondrial Membrane Transport Proteins , Oligonucleotides, Antisense/pharmacology , Protein Serine-Threonine Kinases/genetics , Sequence Alignment , Sequence Homology, Nucleic Acid , Transcription Factors/biosynthesis , Transcription Factors/genetics
19.
PLoS One ; 10(3): e0122067, 2015.
Article in English | MEDLINE | ID: mdl-25807495

ABSTRACT

BACKGROUND: Podocyte foot process effacement accompanied by actin cytoskeleton rearrangements is a cardinal feature of many progressive human proteinuric diseases. RESULTS: By microarray profiling of mouse glomerulus, SCHIP1 emerged as one of the most highly enriched transcripts. We detected Schip1 protein in the kidney glomerulus, specifically in podocytes foot processes. Functionally, Schip1 inactivation in zebrafish by morpholino knock-down results in foot process disorganization and podocyte loss leading to proteinuria. In cultured podocytes Schip1 localizes to cortical actin-rich regions of lamellipodia, where it forms a complex with Nherf2 and ezrin, proteins known to participate in actin remodeling stimulated by PDGFß signaling. Mechanistically, overexpression of Schip1 in vitro causes accumulation of cortical F-actin with dissolution of transversal stress fibers and promotes cell migration in response to PDGF-BB stimulation. Upon actin disassembly by latrunculin A treatment, Schip1 remains associated with the residual F-actin-containing structures, suggesting a functional connection with actin cytoskeleton possibly via its interaction partners. A similar assay with cytochalasin D points to stabilization of cortical actin cytoskeleton in Schip1 overexpressing cells by attenuation of actin depolymerisation. CONCLUSIONS: Schip1 is a novel glomerular protein predominantly expressed in podocytes, necessary for the zebrafish pronephros development and function. Schip1 associates with the cortical actin cytoskeleton network and modulates its dynamics in response to PDGF signaling via interaction with the Nherf2/ezrin complex. Its implication in proteinuric diseases remains to be further investigated.


Subject(s)
Actin Cytoskeleton/metabolism , Carrier Proteins/metabolism , Cytoskeletal Proteins/metabolism , Phosphoproteins/metabolism , Podocytes/metabolism , Sodium-Hydrogen Exchangers/metabolism , Zebrafish Proteins/metabolism , Actin Cytoskeleton/drug effects , Actins/metabolism , Animals , Becaplermin , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cell Movement , Cells, Cultured , Cytochalasin D/metabolism , Fluorescence Resonance Energy Transfer , HEK293 Cells , Humans , Kidney Glomerulus/metabolism , Microscopy, Fluorescence , Oligonucleotides, Antisense/metabolism , Podocytes/cytology , Pronephros/metabolism , Proto-Oncogene Proteins c-sis/pharmacology , Pseudopodia/metabolism , RNA Interference , Signal Transduction/drug effects , Thiazolidines/pharmacology , Zebrafish/metabolism , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics
20.
PLoS Genet ; 10(3): e1004263, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24676100

ABSTRACT

Genome-wide association studies (GWAS) have identified chromosomal loci that affect risk of coronary heart disease (CHD) independent of classical risk factors. One such association signal has been identified at 6q23.2 in both Caucasians and East Asians. The lead CHD-associated polymorphism in this region, rs12190287, resides in the 3' untranslated region (3'-UTR) of TCF21, a basic-helix-loop-helix transcription factor, and is predicted to alter the seed binding sequence for miR-224. Allelic imbalance studies in circulating leukocytes and human coronary artery smooth muscle cells (HCASMC) showed significant imbalance of the TCF21 transcript that correlated with genotype at rs12190287, consistent with this variant contributing to allele-specific expression differences. 3' UTR reporter gene transfection studies in HCASMC showed that the disease-associated C allele has reduced expression compared to the protective G allele. Kinetic analyses in vitro revealed faster RNA-RNA complex formation and greater binding of miR-224 with the TCF21 C allelic transcript. In addition, in vitro probing with Pb2+ and RNase T1 revealed structural differences between the TCF21 variants in proximity of the rs12190287 variant, which are predicted to provide greater access to the C allele for miR-224 binding. miR-224 and TCF21 expression levels were anti-correlated in HCASMC, and miR-224 modulates the transcriptional response of TCF21 to transforming growth factor-ß (TGF-ß) and platelet derived growth factor (PDGF) signaling in an allele-specific manner. Lastly, miR-224 and TCF21 were localized in human coronary artery lesions and anti-correlated during atherosclerosis. Together, these data suggest that miR-224 interaction with the TCF21 transcript contributes to allelic imbalance of this gene, thus partly explaining the genetic risk for coronary heart disease associated at 6q23.2. These studies implicating rs12190287 in the miRNA-dependent regulation of TCF21, in conjunction with previous studies showing that this variant modulates transcriptional regulation through activator protein 1 (AP-1), suggests a unique bimodal level of complexity previously unreported for disease-associated variants.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Genome-Wide Association Study , MicroRNAs/genetics , Signal Transduction/genetics , 3' Untranslated Regions , Alleles , Asian People/genetics , Binding Sites , Cell Differentiation , Chromosomes, Human, Pair 6/genetics , Coronary Disease/etiology , Coronary Disease/genetics , Gene Expression Regulation , Humans , MicroRNAs/metabolism , Polymorphism, Single Nucleotide , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...