Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Br J Pharmacol ; 2024 May 08.
Article in English | MEDLINE | ID: mdl-38720437

ABSTRACT

Noncoding RNAs (ncRNAs) are pivotal for various pathological processes, impacting disease progression. The potential for leveraging ncRNAs to prevent or treat atherosclerosis and associated cardiovascular diseases is of great significance, especially given the increasing prevalence of atherosclerosis in an ageing and sedentary population. Together, these diseases impose a substantial socio-economic burden, demanding innovative therapeutic solutions. This review explores the potential of ncRNAs in atherosclerosis treatment. We commence by examining approaches for identifying and characterizing atherosclerosis-associated ncRNAs. We then delve into the functional aspects of ncRNAs in atherosclerosis development and progression. Additionally, we review current RNA and RNA-targeting molecules in development or under approval for clinical use, offering insights into their pharmacological potential. The importance of improved ncRNA delivery strategies is highlighted. Finally, we suggest avenues for advanced research to accelerate the use of ncRNAs in treating atherosclerosis and mitigating its societal impact.

2.
Mol Ther Nucleic Acids ; 35(2): 102173, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38617973

ABSTRACT

Epigenetic processes involving long non-coding RNAs regulate endothelial gene expression. However, the underlying regulatory mechanisms causing endothelial dysfunction remain to be elucidated. Enhancer of zeste homolog 2 (EZH2) is an important rheostat of histone H3K27 trimethylation (H3K27me3) that represses endothelial targets, but EZH2 RNA binding capacity and EZH2:RNA functional interactions have not been explored in post-ischemic angiogenesis. We used formaldehyde/UV-assisted crosslinking ligation and sequencing of hybrids and identified a new role for maternally expressed gene 3 (MEG3). MEG3 formed the predominant RNA:RNA hybrid structures in endothelial cells. Moreover, MEG3:EZH2 assists recruitment onto chromatin. By EZH2-chromatin immunoprecipitation, following MEG3 depletion, we demonstrated that MEG3 controls recruitment of EZH2/H3K27me3 onto integrin subunit alpha4 (ITGA4) promoter. Both MEG3 knockdown or EZH2 inhibition (A-395) promoted ITGA4 expression and improved endothelial cell migration and adhesion to fibronectin in vitro. The A-395 inhibitor re-directed MEG3-assisted chromatin remodeling, offering a direct therapeutic benefit by increasing endothelial function and resilience. This approach subsequently increased the expression of ITGA4 in arterioles following ischemic injury in mice, thus promoting arteriogenesis. Our findings show a context-specific role for MEG3 in guiding EZH2 to repress ITGA4. Novel therapeutic strategies could antagonize MEG3:EZH2 interaction for pre-clinical studies.

4.
Cells ; 12(14)2023 07 12.
Article in English | MEDLINE | ID: mdl-37508497

ABSTRACT

Current clinical data show that, despite constant efforts to develop novel therapies and clinical approaches, atherosclerotic cardiovascular diseases (ASCVD) are still one of the leading causes of death worldwide. Advanced and unstable atherosclerotic plaques most often trigger acute coronary events that can lead to fatal outcomes. However, despite the fact that different plaque phenotypes may require different treatments, current approaches to prognosis, diagnosis, and classification of acute coronary syndrome do not consider the diversity of plaque phenotypes. Long non-coding RNAs (lncRNAs) represent an important class of molecules that are implicated in epigenetic control of numerous cellular processes. Here we review the latest knowledge about lncRNAs' influence on plaque development and stability through regulation of immune response, lipid metabolism, extracellular matrix remodelling, endothelial cell function, and vascular smooth muscle function, with special emphasis on pro-atherogenic and anti-atherogenic lncRNA functions. In addition, we present current challenges in the research of lncRNAs' role in atherosclerosis and translation of the findings from animal models to humans. Finally, we present the directions for future lncRNA-oriented research, which may ultimately result in patient-oriented therapeutic strategies for ASCVD.


Subject(s)
Atherosclerosis , Plaque, Atherosclerotic , RNA, Long Noncoding , Animals , Humans , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Atherosclerosis/genetics , Atherosclerosis/metabolism , Endothelial Cells/metabolism
5.
J Mol Endocrinol ; 70(1)2023 01 01.
Article in English | MEDLINE | ID: mdl-36222832

ABSTRACT

Leading a research group as an early career researcher (ECR) in academia presents many challenges. First, it imposes many additional pressures on individuals, causing fear of missing out on a great opportunity that could advance your career. Together, the unsettling nature of short-term or temporary contracts, lack of guidance and the imposter syndrome can trigger a crisis in future leadership. Most leadership positions at universities are held by senior colleagues. ECRs have modest input in decision-making, due to a requirement for specific leadership training and experience with oversight that precedes suitable decision-making. The turbulence of the unprecedented world COVID-19 crisis has been felt disproportionally by many researchers, intensely by those with caring responsibilities. In the current academic climate, navigating either between your postdoctoral or fellowship project, leading others, taking strategic project directions, mentoring or networking may feel like too much. This editorial expresses views on the current state of the matter in academia with suggestions for helpful strategies to employ to meet research endpoints. It also addresses some challenges that new principal investigators and academic leaders may face due to external or institutional change, and provides some tangible advice with action points.


Subject(s)
COVID-19 , Leadership , Research Personnel , Humans , Career Mobility , Pandemics
6.
Noncoding RNA ; 8(1)2022 Jan 10.
Article in English | MEDLINE | ID: mdl-35076559

ABSTRACT

We are delighted to share with you our seventh Journal Club and highlight some of the most interesting papers published recently [...].

7.
Noncoding RNA ; 7(4)2021 Sep 23.
Article in English | MEDLINE | ID: mdl-34698214

ABSTRACT

The vascular endothelium comprises the interface between the circulation and the vessel wall and, as such, is under the dynamic regulation of vascular signalling, nutrients, and hypoxia. Understanding the molecular drivers behind endothelial cell (EC) and vascular smooth muscle cell (VSMC) function and dysfunction remains a pivotal task for further clinical progress in tackling vascular disease. A newly emerging era in vascular biology with landmark deep sequencing approaches has provided us with the means to profile diverse layers of transcriptional regulation at a single cell, chromatin, and epigenetic level. This review describes the roles of major vascular long non-coding RNA (lncRNAs) in the epigenetic regulation of EC and VSMC function and discusses the recent progress in their discovery, detection, and functional characterisation. We summarise new findings regarding lncRNA-mediated epigenetic mechanisms-often regulated by hypoxia-within the vascular endothelium and smooth muscle to control vascular homeostasis in health and disease. Furthermore, we outline novel molecular techniques being used in the field to delineate the lncRNA subcellular localisation and interaction with proteins to unravel their biological roles in the epigenetic regulation of vascular genes.

9.
EMBO Rep ; 21(7): e48192, 2020 07 03.
Article in English | MEDLINE | ID: mdl-32337819

ABSTRACT

Autophagy is an essential cellular quality control process that has emerged as a critical one for vascular homeostasis. Here, we show that trichoplein (TCHP) links autophagy with endothelial cell (EC) function. TCHP localizes to centriolar satellites, where it binds and stabilizes PCM1. Loss of TCHP leads to delocalization and proteasome-dependent degradation of PCM1, further resulting in degradation of PCM1's binding partner GABARAP. Autophagic flux under basal conditions is impaired in THCP-depleted ECs, and SQSTM1/p62 (p62) accumulates. We further show that TCHP promotes autophagosome maturation and efficient clearance of p62 within lysosomes, without affecting their degradative capacity. Reduced TCHP and high p62 levels are detected in primary ECs from patients with coronary artery disease. This phenotype correlates with impaired EC function and can be ameliorated by NF-κB inhibition. Moreover, Tchp knock-out mice accumulate of p62 in the heart and cardiac vessels correlating with reduced cardiac vascularization. Taken together, our data reveal that TCHP regulates endothelial cell function via an autophagy-mediated mechanism.


Subject(s)
Adaptor Proteins, Signal Transducing , Autophagy , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins , Centrioles/metabolism , Endothelial Cells/metabolism , Humans , Mice , NF-kappa B , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/metabolism
10.
Mol Ther ; 26(12): 2823-2837, 2018 12 05.
Article in English | MEDLINE | ID: mdl-30274787

ABSTRACT

MicroRNAs regulate endothelial function and angiogenesis, but their implication in pericyte biology remains undetermined. A PCR array, covering a panel of 379 human microRNAs, showed microRNA-532-5p to be one of the most differentially modulated by hypoxia, which was confirmed by qPCR in both skeletal muscle and adventitial pericytes. Furthermore, microRNA-532-5p was upregulated in murine muscular pericytes early after experimentally induced ischemia, decreasing below baseline after reperfusion. Transfection of human pericytes with anti-microRNA, microRNA-mimic, or controls indicates microRNA-532-5p modulates pro-angiogenic activity via transcriptional regulation of angiopoietin-1. Tie-2 blockade abrogated the ability of microRNA-532-5p-overexpressing pericytes to promote endothelial network formation in vitro. However, angiopoietin-1 is not a direct target of microRNA-532-5p. In silico analysis of microRNA-532-5p inhibitory targets associated with angiopoietin-1 transcription indicated three potential candidates, BACH1, HIF1AN, and EGLN1. Binding of microRNA-532-5p to the BACH1 3' UTR was confirmed by luciferase assay. MicroRNA-532-5p silencing increased BACH1, while a microRNA-532-5p mimic decreased expression. Silencing of BACH1 modulated angiopoietin-1 gene and protein expression. ChIP confirmed BACH1 transcriptional regulation of angiopoietin-1 promoter. Finally, microRNA-532-5p overexpression increased pericyte coverage in an in vivo Matrigel assay, suggesting its role in vascular maturation. This study provides a new mechanistic understanding of the transcriptional program orchestrating angiopoietin-1/Tie-2 signaling in human pericytes.


Subject(s)
Angiopoietin-1/genetics , Basic-Leucine Zipper Transcription Factors/genetics , Gene Expression Regulation , MicroRNAs/genetics , Pericytes/metabolism , RNA Interference , Autocrine Communication , Biomarkers , Gene Expression Profiling , Genes, Reporter , Humans , Hypoxia , Paracrine Communication , Phenotype , Transcriptome
11.
Mol Ther ; 26(8): 1996-2007, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29843955

ABSTRACT

Transforming growth factor beta (TGF-ß) is crucial for regulation of the endothelial cell (EC) homeostasis. Perturbation of TGF-ß signaling leads to pathological conditions in the vasculature, causing cardiovascular disease and fibrotic disorders. The TGF-ß pathway is critical in endothelial-to-mesenchymal transition (EndMT), but a gap remains in our understanding of the regulation of TGF-ß and related signaling in the endothelium. This study applied a gain- and loss-of function approach and an in vivo model of skin wound healing to demonstrate that miR-148b regulates TGF-ß signaling and has a key role in EndMT, targeting TGFB2 and SMAD2. Overexpression of miR-148b increased EC migration, proliferation, and angiogenesis, whereas its inhibition promoted EndMT. Cytokine challenge decreased miR-148b levels in ECs while promoting EndMT through the regulation of SMAD2. Finally, in a mouse model of skin wound healing, delivery of miR-148b mimics promoted wound vascularization and accelerated closure. In contrast, inhibition of miR-148b enhanced EndMT in wounds, resulting in impaired wound closure that was reversed by SMAD2 silencing. Together, these results demonstrate for the first time that miR-148b is a key factor controlling EndMT and vascularization. This opens new avenues for therapeutic application of miR-148b in vascular and tissue repair.


Subject(s)
MicroRNAs/genetics , Neovascularization, Physiologic , Signal Transduction , Skin/injuries , Wound Healing , Animals , Cell Movement , Disease Models, Animal , Epithelial-Mesenchymal Transition , Female , Human Umbilical Vein Endothelial Cells , Humans , Mice , Skin/metabolism , Smad2 Protein/metabolism , Transforming Growth Factor beta , Transforming Growth Factor beta2/metabolism
12.
Oxid Med Cell Longev ; 2016: 3565127, 2016.
Article in English | MEDLINE | ID: mdl-27247702

ABSTRACT

The aging process worsens the human body functions at multiple levels, thus causing its gradual decrease to resist stress, damage, and disease. Besides changes in gene expression and metabolic control, the aging rate has been associated with the production of high levels of Reactive Oxygen Species (ROS) and/or Reactive Nitrosative Species (RNS). Specific increases of ROS level have been demonstrated as potentially critical for induction and maintenance of cell senescence process. Causal connection between ROS, aging, age-related pathologies, and cell senescence is studied intensely. Senescent cells have been proposed as a target for interventions to delay the aging and its related diseases or to improve the diseases treatment. Therapeutic interventions towards senescent cells might allow restoring the health and curing the diseases that share basal processes, rather than curing each disease in separate and symptomatic way. Here, we review observations on ROS ability of inducing cell senescence through novel mechanisms that underpin aging processes. Particular emphasis is addressed to the novel mechanisms of ROS involvement in epigenetic regulation of cell senescence and aging, with the aim to individuate specific pathways, which might promote healthy lifespan and improve aging.


Subject(s)
Aging/genetics , Aging/metabolism , Cellular Senescence , Humans , Reactive Oxygen Species
13.
Nat Commun ; 6: 8024, 2015 Aug 13.
Article in English | MEDLINE | ID: mdl-26268439

ABSTRACT

The communication between vascular endothelial cells (ECs) and pericytes in the microvasculature is fundamental for vascular growth and homeostasis; however, these processes are disrupted by diabetes. Here we show that modulation of p75(NTR) expression in ECs exposed to high glucose activates transcription of miR-503, which negatively affects pericyte function. p75(NTR) activates NF-κB to bind the miR-503 promoter and upregulate miR-503 expression in ECs. NF-κB further induces activation of Rho kinase and shedding of endothelial microparticles carrying miR-503, which transfer miR-503 from ECs to vascular pericytes. The integrin-mediated uptake of miR-503 in the recipient pericytes reduces expression of EFNB2 and VEGFA, resulting in impaired migration and proliferation. We confirm operation of the above mechanisms in mouse models of diabetes, in which EC-derived miR-503 reduces pericyte coverage of capillaries, increased permeability and impaired post-ischaemic angiogenesis in limb muscles. Collectively, our data demonstrate that miR-503 regulates pericyte-endothelial crosstalk in microvascular diabetic complications.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Endothelial Cells/metabolism , MicroRNAs/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Nerve Growth Factor/metabolism , Animals , Cells, Cultured , Diabetes Mellitus, Experimental/genetics , Gene Expression Regulation/physiology , Hindlimb/blood supply , Humans , Ischemia , Male , Mice , Mice, Knockout , MicroRNAs/genetics , NF-kappa B/genetics , NF-kappa B/metabolism , Nerve Tissue Proteins/genetics , Oligonucleotide Array Sequence Analysis , Pericytes/physiology , Receptors, Nerve Growth Factor/genetics , Transcription, Genetic/physiology
14.
Arterioscler Thromb Vasc Biol ; 35(3): 664-74, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25614281

ABSTRACT

OBJECTIVE: Gestational diabetes mellitus (GDM) produces fetal hyperglycemia with increased lifelong risks for the exposed offspring of cardiovascular and other diseases. Epigenetic mechanisms induce long-term gene expression changes in response to in utero environmental perturbations. Moreover, microRNAs (miRs) control the function of endothelial cells (ECs) under physiological and pathological conditions and can target the epigenetic machinery. We investigated the functional and expressional effect of GDM on human fetal ECs of the umbilical cord vein (HUVECs). We focused on miR-101 and 1 of its targets, enhancer of zester homolog-2 (EZH2), which trimethylates the lysine 27 of histone 3, thus repressing gene transcription. EZH2 exists as isoforms α and ß. APPROACH AND RESULTS: HUVECs were prepared from GDM or healthy pregnancies and tested in apoptosis, migration, and Matrigel assays. GDM-HUVECs demonstrated decreased functional capacities, increased miR-101 expression, and reduced EZH2- ß and trimethylation of histone H3 on lysine 27 levels. MiR-101 inhibition increased EZH2 expression and improved GDM-HUVEC function. Healthy HUVECs were exposed to high or normal d-glucose concentration for 48 hours and then tested for miR-101 and EZH2 expression. Similar to GDM, high glucose increased miR-101 expression. Chromatin immunoprecipitation using an antibody for EZH2 followed by polymerase chain reaction analyses for miR-101 gene promoter regions showed that both GDM and high glucose concentration reduced EZH2 binding to the miR-101 locus in HUVECs. Moreover, EZH2-ß overexpression inhibited miR-101 promoter activity in HUVECs. CONCLUSIONS: GDM impairs HUVEC function via miR-101 upregulation. EZH2 is both a transcriptional inhibitor and a target gene of miR-101 in HUVECs, and it contributes to some of the miR-101-induced defects of GDM-HUVECs.


Subject(s)
Diabetes, Gestational/enzymology , Human Umbilical Vein Endothelial Cells/enzymology , MicroRNAs/metabolism , Polycomb Repressive Complex 2/metabolism , Apoptosis , Binding Sites , Case-Control Studies , Cell Movement , Cell Survival , Cells, Cultured , Diabetes, Gestational/genetics , Diabetes, Gestational/pathology , Diabetes, Gestational/physiopathology , Enhancer of Zeste Homolog 2 Protein , Female , Gestational Age , Glucose/metabolism , Histones/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Methylation , Neovascularization, Physiologic , Phenotype , Polycomb Repressive Complex 2/genetics , Pregnancy , Promoter Regions, Genetic , RNA Interference , Signal Transduction , Time Factors , Transcription, Genetic , Transfection , Up-Regulation
15.
Mol Ther ; 23(1): 32-42, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25189741

ABSTRACT

Epigenetic mechanisms may regulate the expression of pro-angiogenic genes, thus affecting reparative angiogenesis in ischemic limbs. The enhancer of zest homolog-2 (EZH2) induces thtrimethylation of lysine 27 on histone H3 (H3K27me3), which represses gene transcription. We explored (i) if EZH2 expression is regulated by hypoxia and ischemia; (ii) the impact of EZH2 on the expression of two pro-angiogenic genes: eNOS and BDNF; (iii) the functional effect of EZH2 inhibition on cultured endothelial cells (ECs); (iv) the therapeutic potential of EZH2 inhibition in a mouse model of limb ischemia (LI). EZH2 expression was increased in cultured ECs exposed to hypoxia (control: normoxia) and in ECs extracted from mouse ischemic limb muscles (control: absence of ischemia). EZH2 increased the H3K27me3 abundance onto regulatory regions of eNOS and BDNF promoters. In vitro RNA silencing or pharmacological inhibition by 3-deazaneplanocin (DZNep) of EZH2 increased eNOS and BDNF mRNA and protein levels and enhanced functional capacities (migration, angiogenesis) of ECs under either normoxia or hypoxia. In mice with experimentally induced LI, DZNep increased angiogenesis in ischaemic muscles, the circulating levels of pro-angiogenic hematopoietic cells and blood flow recovery. Targeting EZH2 for inhibition may open new therapeutic avenues for patients with limb ischemia.


Subject(s)
Epigenesis, Genetic , Hypoxia/genetics , Ischemia/genetics , Neovascularization, Physiologic/drug effects , Polycomb Repressive Complex 2/genetics , Adenosine/analogs & derivatives , Adenosine/pharmacology , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Cell Hypoxia , Enhancer of Zeste Homolog 2 Protein , Femoral Artery/surgery , Hindlimb/blood supply , Hindlimb/drug effects , Hindlimb/surgery , Histones/genetics , Histones/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Hypoxia/drug therapy , Hypoxia/metabolism , Hypoxia/pathology , Ischemia/drug therapy , Ischemia/metabolism , Male , Mice , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Polycomb Repressive Complex 2/antagonists & inhibitors , Polycomb Repressive Complex 2/metabolism , Primary Cell Culture , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Transcription, Genetic
16.
Biochem Pharmacol ; 86(1): 146-53, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23415904

ABSTRACT

11ß-Hydroxysteroid dehydrogenase 1 (11ßHSD1; EC 1.1.1.146) generates active glucocorticoids from inert 11-keto metabolites. However, it can also metabolize alternative substrates, including 7ß-hydroxy- and 7-keto-cholesterol (7ßOHC, 7KC). This has been demonstrated in vitro but its consequences in vivo are uncertain. We used genetically modified mice to investigate the contribution of 11ßHSD1 to the balance of circulating levels of 7KC and 7ßOHC in vivo, and dissected in vitro the kinetics of the interactions between oxysterols and glucocorticoids for metabolism by the mouse enzyme. Circulating levels of 7KC and 7ßOHC in mice were 91.3±22.3 and 22.6±5.7 nM respectively, increasing to 1240±220 and 406±39 nM in ApoE(-/-) mice receiving atherogenic western diet. Disruption of 11ßHSD1 in mice increased (p<0.05) the 7KC/7ßOHC ratio in plasma (by 20%) and also in isolated microsomes (2 fold). The 7KC/7ßOHC ratio was similarly increased when NADPH generation was restricted by disruption of hexose-6-phosphate dehydrogenase. Reduction and oxidation of 7-oxysterols by murine 11ßHSD1 proceeded more slowly and substrate affinity was lower than for glucocorticoids. in vitro 7ßOHC was a competitive inhibitor of oxidation of corticosterone (Ki=0.9 µM), whereas 7KC only weakly inhibited reduction of 11-dehydrocorticosterone. However, supplementation of 7-oxysterols in cultured cells, secondary to cholesterol loading, preferentially slowed reduction of glucocorticoids, rather than oxidation. Thus, in mouse, 11ßHSD1 influenced the abundance and balance of circulating and tissue levels of 7ßOHC and 7KC, promoting reduction of 7KC. In health, 7-oxysterols are unlikely to regulate glucocorticoid metabolism. However, in hyperlipidaemia, 7-oxysterols may inhibit glucocorticoid metabolism and modulate signaling through corticosteroid receptors.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Hydroxycholesterols/metabolism , Ketocholesterols/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , Animals , Carbohydrate Dehydrogenases/genetics , Catalytic Domain , Computer Simulation , Glucocorticoids/metabolism , HEK293 Cells , Humans , Kinetics , Male , Mice , Mice, Transgenic , Microsomes, Liver/metabolism , Models, Molecular , Oxidation-Reduction
17.
FASEB J ; 27(4): 1519-31, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23303209

ABSTRACT

11ß-Hydroxysteroid dehydrogenase type-1 (11ß-HSD1) converts inert cortisone into active cortisol, amplifying intracellular glucocorticoid action. 11ß-HSD1 deficiency improves cardiovascular risk factors in obesity but exacerbates acute inflammation. To determine the effects of 11ß-HSD1 deficiency on atherosclerosis and its inflammation, atherosclerosis-prone apolipoprotein E-knockout (ApoE-KO) mice were treated with a selective 11ß-HSD1 inhibitor or crossed with 11ß-HSD1-KO mice to generate double knockouts (DKOs) and challenged with an atherogenic Western diet. 11ß-HSD1 inhibition or deficiency attenuated atherosclerosis (74-76%) without deleterious effects on plaque structure. This occurred without affecting plasma lipids or glucose, suggesting independence from classical metabolic risk factors. KO plaques were not more inflamed and indeed had 36% less T-cell infiltration, associated with 38% reduced circulating monocyte chemoattractant protein-1 (MCP-1) and 36% lower lesional vascular cell adhesion molecule-1 (VCAM-1). Bone marrow (BM) cells are key to the atheroprotection, since transplantation of DKO BM to irradiated ApoE-KO mice reduced atherosclerosis by 51%. 11ß-HSD1-null macrophages show 76% enhanced cholesterol ester export. Thus, 11ß-HSD1 deficiency reduces atherosclerosis without exaggerated lesional inflammation independent of metabolic risk factors. Selective 11ß-HSD1 inhibitors promise novel antiatherosclerosis effects over and above their benefits for metabolic risk factors via effects on BM cells, plausibly macrophages.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/deficiency , Atherosclerosis/metabolism , Bone Marrow/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 1/antagonists & inhibitors , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Atherosclerosis/genetics , Bone Marrow/drug effects , Glucocorticoids/metabolism , Mice , Mice, Knockout , Risk Factors , Vascular Cell Adhesion Molecule-1/metabolism
18.
Biochimie ; 95(3): 548-55, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22940536

ABSTRACT

The atherogenic 7-oxysterols, 7-ketocholesterol (7-KC) and 7ß-hydroxycholesterol (7ßOHC), can directly impair arterial function. Inter-conversion of 7-KC and 7ßOHC has recently been shown as a novel role for the glucocorticoid-metabolizing enzyme 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1). Since this enzyme is expressed in vascular smooth muscle cells, we addressed the hypothesis that inter-conversion of 7-KC and 7ßOHC by 11ß-HSD1 may contribute to regulation of arterial function. Incubation (4-24 h) of aortic rings with either 7-KC (25 µM) or 7ßOHC (20 µM) had no effect on endothelium-dependent (acetylcholine) or -independent (sodium nitroprusside) relaxation. In contrast, exposure to 7-KC (but not to 7ßOHC) attenuated noradrenaline-induced contraction (E(max)) after 4 h (0.78 ± 0.28 vs 0.40 ± 0.08 mN/mm; p < 0.05) and 24 h (2.28 ± 0.34 vs 1.56 ± 0.48 mN/mm; p < 0.05). Both 7-oxysterols were detected by GCMS in the aortic wall of chow-fed C57Bl6/J mice, with concentrations of 7-KC (1.41 ± 0.81 ng/mg) higher (p = 0.05) than 7ßOHC (0.16 ± 0.06 ng/mg). In isolated mouse aortic rings 11ß-HSD1 was shown to act as an oxo-reductase, inter-converting 7-KC and 7ßOHC. This activity was lost in aorta from 11ß-HSD1(-/-) mice, which had low oxysterol levels. Renal homogenates from 11ß-HSD1(-/-) mice were used to confirm that the type 2 isozyme of 11ß-HSD does not inter-convert 7-KC and 7ßOHC. These results demonstrate that 7-KC has greater effects than 7ßOHC on vascular function, and that 11ß-HSD1 can inter-convert 7-KC and 7ßOHC in the arterial wall, contributing to the regulation of 7-oxysterol levels and potentially influencing vascular function. This mechanism may be important in the cardioprotective effects of 11ß-HSD1 inhibitors.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Aorta/metabolism , Hydroxycholesterols/metabolism , Ketocholesterols/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 1/deficiency , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , Animals , Aorta/drug effects , Aorta/physiology , Gene Deletion , Hydroxycholesterols/pharmacology , In Vitro Techniques , Ketocholesterols/pharmacology , Male , Mice
19.
Arterioscler Thromb Vasc Biol ; 32(12): e149-60, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23065828

ABSTRACT

OBJECTIVE: The p75 neurotrophin receptor (p75(NTR)) contributes to diabetes mellitus-induced defective postischemic neovascularization. The interleukin-33 receptor ST2 is expressed as transmembrane (ST2L) and soluble (sST2) isoforms. Here, we studied the following: (1) the impact of p75(NTR) in the healing of ischemic and diabetic calf wounds; (2) the link between p75(NTR) and ST2; and (3) circulating sST2 levels in critical limb ischemia (CLI) patients. METHODS AND RESULTS: Diabetes mellitus was induced in p75(NTR) knockout (p75KO) mice and wild-type (WT) littermates by streptozotocin. Diabetic and nondiabetic p75KO and WT mice received left limb ischemia induction and a full-thickness wound on the ipsilateral calf. Diabetes mellitus impaired wound closure and angiogenesis and increased ST2 expression in WT, but not in p75KO wounds. In cultured endothelial cells, p75(NTR) promoted ST2 (both isoforms) expression through p38(MAPK)/activating transcription factor 2 pathway activation. Next, sST2 was measured in the serum of patients with CLI undergoing either revascularization or limb amputation and in the 2 nondiabetic groups (with CLI or nonischemic individuals). Serum sST2 increased in diabetic patients with CLI and was directly associated with higher mortality at 1 year from revascularization. CONCLUSIONS: p75(NTR) inhibits the healing of ischemic lower limb wounds in diabetes mellitus and promotes ST2 expression. Circulating sST2 predicts mortality in diabetic CLI patients.


Subject(s)
Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus/mortality , Ischemia/physiopathology , Lower Extremity/blood supply , Nerve Tissue Proteins/physiology , Receptors, Cell Surface/metabolism , Receptors, Interleukin/metabolism , Receptors, Nerve Growth Factor/physiology , Activating Transcription Factor 2/metabolism , Aged , Aged, 80 and over , Animals , Biomarkers/metabolism , Cells, Cultured , Diabetes Complications/complications , Diabetes Mellitus/metabolism , Diabetes Mellitus/physiopathology , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Disease Models, Animal , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Female , Humans , Interleukin-1 Receptor-Like 1 Protein , Ischemia/etiology , Male , Mice , Mice, Knockout , Middle Aged , Nerve Tissue Proteins/pharmacology , Predictive Value of Tests , Receptors, Nerve Growth Factor/deficiency , Receptors, Nerve Growth Factor/genetics , Streptozocin/adverse effects , Wound Healing/physiology , p38 Mitogen-Activated Protein Kinases/metabolism
20.
Article in English | MEDLINE | ID: mdl-22236027

ABSTRACT

Vascular dysfunction is a common consequence of diabetes mellitus. Stable propagation of gene expression from cell to cell generation during development of diseases (like diabetes) is regulated by epigenetic mechanisms. These are heritable patterns of gene expression that cannot solely be explained by changes in DNA sequence. Recent evidence shows that diabetes-induced epigenetic changes can affect gene expression in vascular endothelial cells and vascular smooth muscles cells. Such effects further influence inflammatory and insulin production pathways in these cells and thus ensure a long-term memory, whereby epigenetic changes are maintained even long after restoring normo-glycaemic conditions by appropriate therapeutic approaches. This review focuses on the epigenetic marks, which endure on the vascular chromatin under diabetic conditions.


Subject(s)
Chromatin/metabolism , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 2/complications , Diabetic Angiopathies/genetics , Epigenesis, Genetic , Acetylation , Animals , DNA Methylation , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Female , Histones/metabolism , Humans , Mice , Muscle, Smooth, Vascular/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...