Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 117
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Curr Cardiol Rep ; 25(5): 435-442, 2023 05.
Article in English | MEDLINE | ID: mdl-37052761

ABSTRACT

PURPOSE OF REVIEW: In this review, we discuss the prevalence of cardiovascular disease in people with type 1 diabetes. We outline key risk factors associated with increased cardiovascular event rates and discuss the prevalence and mechanisms underlying hyperlipidemia in people with type 1 diabetes. Finally, we summarize the evidence to support early and more aggressive lipid-lowering therapy in people with type 1 diabetes and review current guideline recommendations. RECENT FINDINGS: Comprehensive treatment of hyperglycemia, hypertension, and hyperlipidemia reduces adverse cardiovascular outcomes in people with type 2 diabetes. In contrast, evidence to support a comparable benefit of intensive cardiovascular risk factor management in people with type 1 diabetes is lacking from prospective, randomized trials and has only been shown in registries. Therefore, current treatment guidelines extrapolate prospective clinical trial evidence obtained in people with type 2 diabetes to provide similar treatment recommendations for people with type 1 and type 2 diabetes. Evidence supports the more aggressive treatment of cardiovascular risk factors in people with type 1 diabetes, who would likely benefit from early risk stratification and comprehensive risk factor management, including aggressive lipid-lowering therapy.


Subject(s)
Cardiovascular Diseases , Diabetes Mellitus, Type 1 , Diabetes Mellitus, Type 2 , Hyperlipidemias , Humans , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/etiology , Cardiovascular Diseases/prevention & control , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/drug therapy , Hyperlipidemias/complications , Hyperlipidemias/drug therapy , Prospective Studies , Lipids/therapeutic use
2.
Curr Cardiol Rep ; 23(7): 73, 2021 06 03.
Article in English | MEDLINE | ID: mdl-34081207

ABSTRACT

PURPOSE OF REVIEW: The purpose of this review is to provide an overview of the current quality of diabetes care in the USA, discuss barriers to guideline-recommended treatment adherence, and outline strategies for the improvement in care. RECENT FINDINGS: Current treatment guidelines highlight the importance of glycemic control, use of novel medications with proven cardiovascular efficacy, and multifactorial cardiovascular risk factor intervention for the treatment of diabetes and associated complications. Albeit proven evidence for these guidelines, the vast majority of patients with diabetes remain insufficiently treated. Interventions to improve outcomes require focus on care delivery systems, physician behavior, and patient-centered approaches. De-fragmenting care systems to form collaborative, multi-specialty teams, use of standardized and comprehensive treatment algorithms, development of quality assessment tools, avoiding physician therapeutic inertia, and addressing patient barriers, including lack of perceived benefit, insufficient diabetes education and access to care, and medication costs, represent key objectives to improve diabetes care and outcomes. Clinical research in standardized trials has proven the feasibility to reduce morbidity and mortality associated with diabetes. Implementing models of care to disseminate these encouraging research findings to the wider population and to overcome barriers to achieving guideline-recommended treatment goals should be the objective to improve our current quality of diabetes care in the USA.


Subject(s)
Diabetes Mellitus , Physicians , Diabetes Mellitus/drug therapy , Guideline Adherence , Humans , United States
4.
Arterioscler Thromb Vasc Biol ; 37(2): 301-311, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27932351

ABSTRACT

OBJECTIVE: Aberrant proliferation of smooth muscle cells (SMC) in response to injury induces pathological vascular remodeling during atherosclerosis and neointima formation. Telomerase is rate limiting for tissue renewal and cell replication; however, the physiological role of telomerase in vascular diseases remains to be determined. The goal of the present study was to determine whether telomerase reverse transcriptase (TERT) affects proliferative vascular remodeling and to define the molecular mechanism by which TERT supports SMC proliferation. APPROACH AND RESULTS: We first demonstrate high levels of TERT expression in replicating SMC of atherosclerotic and neointimal lesions. Using a model of guidewire-induced arterial injury, we demonstrate decreased neointima formation in TERT-deficient mice. Studies in SMC isolated from TERT-deficient and TERT overexpressing mice with normal telomere length established that TERT is necessary and sufficient for cell proliferation. TERT deficiency did not induce a senescent phenotype but resulted in G1 arrest albeit hyperphosphorylation of the retinoblastoma protein. This proliferative arrest was associated with stable silencing of the E2F1-dependent S-phase gene expression program and not reversed by ectopic overexpression of E2F1. Finally, chromatin immunoprecipitation and accessibility assays revealed that TERT is recruited to E2F1 target sites and promotes chromatin accessibility for E2F1 by facilitating the acquisition of permissive histone modifications. CONCLUSIONS: These data indicate a previously unrecognized role for TERT in neointima formation through epigenetic regulation of proliferative gene expression in SMC.


Subject(s)
Atherosclerosis/enzymology , Chromatin Assembly and Disassembly , E2F1 Transcription Factor/metabolism , Gene Silencing , Muscle, Smooth, Vascular/enzymology , Neointima , Telomerase/deficiency , Telomerase/metabolism , Vascular System Injuries/enzymology , Acetylation , Animals , Atherosclerosis/genetics , Atherosclerosis/pathology , Binding Sites , Cell Proliferation , Cells, Cultured , Disease Models, Animal , E2F1 Transcription Factor/genetics , Femoral Artery/enzymology , Femoral Artery/injuries , Femoral Artery/pathology , G1 Phase Cell Cycle Checkpoints , Genetic Predisposition to Disease , Histones/metabolism , Humans , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Muscle, Smooth, Vascular/injuries , Muscle, Smooth, Vascular/pathology , Phenotype , Phosphorylation , Protein Binding , RNA Interference , Retinoblastoma Protein/metabolism , Signal Transduction , Telomerase/genetics , Time Factors , Transfection , Vascular Remodeling , Vascular System Injuries/genetics , Vascular System Injuries/pathology
6.
BMC Cardiovasc Disord ; 17(1): 271, 2017 Oct 18.
Article in English | MEDLINE | ID: mdl-29047330

ABSTRACT

BACKGROUND: The NR4A3 orphan nuclear hormone receptor, NOR1, functions as a constitutively active transcription factor to regulate inflammation, proliferation, and cell survival during pathological vascular remodeling. Inflammatory processes represent key mechanisms leading to abdominal aortic aneurysm (AAA) formation. However, a role of NOR1 in AAA formation has not been investigated previously. METHODS: Inflammatory gene expression was analyzed in bone marrow-derived macrophages isolated from NOR1-deficient mice. Low-density lipoprotein receptor-deficient (LDLr-/-) mice were irradiated and reconstituted with hematopoietic stem cells obtained from NOR1-/- or wild-type littermate mice. Animals were infused with angiotensin II and fed a diet enriched in saturated fat to induce AAA formation. Quantification of AAA formation was performed by ultrasound and ex vivo measurements. RESULTS: Among 184 inflammatory genes that were analyzed, 36 genes were differentially regulated in LPS-treated NOR1-deficient macrophages. Albeit this difference in gene regulation, NOR1-deficiency in hematopoietic stem cells did not affect development of AAA formation in bone marrow-derived stem cell transplanted LDLr-deficient mice. CONCLUSION: NOR1 deletion induced differential inflammatory gene transcription in macrophages but did not influence AAA formation in mice.


Subject(s)
Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/metabolism , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Gene Deletion , Hematopoietic Stem Cells/metabolism , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Receptors, Steroid/deficiency , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/deficiency , Receptors, Thyroid Hormone/genetics , Animals , Inflammation/genetics , Inflammation/metabolism , Inflammation/prevention & control , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
7.
J Cell Physiol ; 231(6): 1276-82, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26505494

ABSTRACT

Telomerase reverse transcriptase (TERT) maintains telomeres and is rate limiting for replicative life span. While most somatic tissues silence TERT transcription resulting in telomere shortening, cells derived from cancer or cardiovascular diseases express TERT and activate telomerase. In the present study, we demonstrate that histone deacetylase (HDAC) inhibition induces TERT transcription and promoter activation. At the protein level in contrast, HDAC inhibition decreases TERT protein abundance through enhanced degradation, which decreases telomerase activity and induces senescence. Finally, we demonstrate that HDAC inhibition decreases TERT expression during vascular remodeling in vivo. These data illustrate a differential regulation of TERT transcription and protein stability by HDAC inhibition and suggest that TERT may constitute an important target for the anti-proliferative efficacy of HDAC inhibitors.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Hydroxylamines/pharmacology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Promoter Regions, Genetic , Quinolines/pharmacology , Telomerase/metabolism , Transcriptional Activation/drug effects , Animals , Cells, Cultured , Cellular Senescence/drug effects , Disease Models, Animal , Gene Expression Regulation , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/antagonists & inhibitors , Histone Deacetylase 2/metabolism , Histone Deacetylases/genetics , Mice, Inbred C57BL , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/pathology , Neointima , Proteolysis , RNA Interference , Rats , Telomerase/genetics , Transfection , Vascular Remodeling/drug effects , Vascular System Injuries/drug therapy , Vascular System Injuries/genetics , Vascular System Injuries/metabolism , Vascular System Injuries/pathology
8.
Am J Physiol Endocrinol Metab ; 311(1): E69-81, 2016 Jul 01.
Article in English | MEDLINE | ID: mdl-27166283

ABSTRACT

The NR4A orphan nuclear receptors function as early response genes to numerous stimuli. Our laboratory has previously demonstrated that overexpression of NR4A3 (NOR-1, MINOR) in 3T3-L1 adipocytes enhances insulin-stimulated glucose uptake. To assess the in vivo effect of NR4A3 on adipocytes, we generated transgenic mice with NR4A3 overexpression driven by the adipocyte fatty acid-binding protein (AP2) promoter (AP2-NR4A3 mice). We hypothesized that AP2-NR4A3 mice would display enhanced glucose tolerance and insulin sensitivity. However, AP2-NR4A3 mice exhibit metabolic impairment, including increased fasting glucose and insulin, impaired glucose tolerance, insulin resistance, decreased serum free fatty acids, and increased low-density lipoprotein-cholesterol. AP2-NR4A3 mice also display a significant reduction in serum epinephrine due to increased expression of catecholamine-catabolizing enzymes in adipose tissue, including monoamine oxidase-A. Furthermore, enhanced expression of monoamine oxidase-A is due to direct transcriptional activation by NR4A3. Finally, AP2-NR4A3 mice display cardiac and behavioral alterations consistent with chronically low circulating epinephrine levels. In conclusion, overexpression of NR4A3 in adipocytes produces a complex phenotype characterized by impaired glucose metabolism and low serum catecholamines due to enhanced degradation by adipose tissue.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Catecholamines/metabolism , DNA-Binding Proteins/genetics , Epinephrine/blood , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/genetics , Absorptiometry, Photon , Animals , Behavior, Animal , Blood Glucose/metabolism , Blotting, Western , Body Composition/genetics , Body Temperature , Cell Culture Techniques , Cholesterol, LDL/blood , Chromatin Immunoprecipitation , Energy Metabolism , Fatty Acid-Binding Proteins/genetics , Fatty Acids, Nonesterified/blood , Glucose Intolerance/genetics , Glucose Tolerance Test , Immunohistochemistry , Insulin/metabolism , Insulin Resistance/genetics , Lipolysis , Male , Metabolism , Mice , Mice, Transgenic , Monoamine Oxidase/metabolism , Promoter Regions, Genetic , Real-Time Polymerase Chain Reaction , Transcriptional Activation/genetics
9.
Clin Sci (Lond) ; 130(20): 1837-9, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27634843

ABSTRACT

Endothelial dysfunction and impaired vascular relaxation represent a common cause of microvascular disease in patients with diabetes. Although multiple mechanisms underlying altered endothelial cell function in diabetes have been described, there is currently no specific and approved pharmacological treatment. In this edition of Clinical Science, Morales-Cano et al. characterize voltage-dependent K(+) (Kv) channels as genes regulated by pharmacological activation of peroxisome proliferator-activated receptor-b/d (PPARb/d). Diabetes altered Kv channel function leading to impaired coronary artery relaxation, which was prevented by pharmacological activation of PPARb/d. These studies highlight an important mechanism of vascular dysfunction in diabetes and point to a potential approach for therapy, particularly considering that PPARb/d ligands have been developed and tested in small clinical trials.


Subject(s)
Diabetic Cardiomyopathies/metabolism , Muscle, Smooth, Vascular/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Coronary Vessels/physiopathology , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/physiopathology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Humans , Muscle, Smooth, Vascular/cytology , Vasodilation
10.
J Mol Cell Cardiol ; 81: 23-33, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25640159

ABSTRACT

Phosphodiesterase 4 (PDE4) activity mediates cAMP-dependent smooth muscle cell (SMC) activation following vascular injury. In this study we have investigated the effects of specific PDE4 inhibition with roflumilast on SMC proliferation and inflammatory activation in vitro and neointima formation following guide wire-induced injury of the femoral artery in mice in vivo. In vitro, roflumilast did not affect SMC proliferation, but diminished TNF-α induced expression of the vascular cell adhesion molecule 1 (VCAM-1). Specific activation of the cAMP effector Epac, but not PKA activation mimicked the effects of roflumilast on VCAM-1 expression. Consistently, the reduction of VCAM-1 expression was rescued following inhibition of Epac. TNF-α induced NFκB p65 translocation and VCAM-1 promoter activity were not altered by roflumilast in SMCs. However, roflumilast treatment and Epac activation repressed the induction of the activating epigenetic histone mark H3K4me2 at the VCAM-1 promoter, while PKA activation showed no effect. Furthermore, HDAC inhibition blocked the inhibitory effect of roflumilast on VCAM-1 expression. Both, roflumilast and Epac activation reduced monocyte adhesion to SMCs in vitro. Finally, roflumilast treatment attenuated femoral artery intima-media ratio by more than 50% after 4weeks. In summary, PDE4 inhibition regulates VCAM-1 through a novel Epac-dependent mechanism, which involves regulatory epigenetic components and reduces neointima formation following vascular injury. PDE4 inhibition and Epac activation might represent novel approaches for the treatment of vascular diseases, including atherosclerosis and in-stent restenosis.


Subject(s)
Aminopyridines/pharmacology , Benzamides/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Guanine Nucleotide Exchange Factors/genetics , Neointima/prevention & control , Phosphodiesterase 4 Inhibitors/pharmacology , Vascular Cell Adhesion Molecule-1/genetics , Vascular System Injuries/drug therapy , Animals , Cell Adhesion/drug effects , Cell Line , Cell Proliferation/drug effects , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Cyclopropanes/pharmacology , Femoral Artery/drug effects , Femoral Artery/injuries , Femoral Artery/metabolism , Gene Expression Regulation , Guanine Nucleotide Exchange Factors/metabolism , Histones/genetics , Histones/metabolism , Humans , Mice , Monocytes/cytology , Monocytes/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Neointima/genetics , Neointima/metabolism , Neointima/pathology , Rats , Signal Transduction , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Vascular System Injuries/genetics , Vascular System Injuries/metabolism , Vascular System Injuries/pathology
11.
Stem Cells ; 32(9): 2419-29, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24806827

ABSTRACT

The NR4A orphan nuclear receptor NOR1 functions as a constitutively active transcription factor regulating cellular inflammation and proliferation. In this study, we used bone marrow transplantation to determine the selective contribution of NOR1 expression in hematopoietic stem cells to the development of atherosclerosis. Reconstitution of lethally irradiated apoE(-/-) mice with NOR1-deficient hematopoietic stem cells accelerated atherosclerosis formation and macrophage recruitment following feeding a diet enriched in saturated fat. NOR1 deficiency in hematopoietic stem cells induced splenomegaly and monocytosis, specifically the abundance of inflammatory Ly6C(+) monocytes. Bone marrow transplantation studies further confirmed that NOR1 suppresses the proliferation of macrophage and dendritic progenitor (MDP) cells. Expression analysis identified RUNX1, a critical regulator of hematopoietic stem cell expansion, as a target gene suppressed by NOR1 in MDP cells. Finally, in addition to inducing Ly6C(+) monocytosis, NOR1 deletion increased the replicative rate of lesional macrophages and induced local foam cell formation within the atherosclerotic plaque. Collectively, our studies demonstrate that NOR1 deletion in hematopoietic stem cells accelerates atherosclerosis formation by promoting myelopoiesis in the stem cell compartment and by inducing local proatherogenic activities in the macrophage, including lesional macrophage proliferation and foam cell formation.


Subject(s)
DNA-Binding Proteins/deficiency , Hematopoietic Stem Cells/metabolism , Monocytes/metabolism , Nerve Tissue Proteins/deficiency , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , Receptors, Steroid/deficiency , Receptors, Thyroid Hormone/deficiency , Animals , Cell Proliferation/physiology , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , HEK293 Cells , Hematopoietic Stem Cells/cytology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Plaque, Atherosclerotic/genetics , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/genetics , Receptors, Thyroid Hormone/metabolism , Transfection
12.
Circ Res ; 110(11): e73-85, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22539767

ABSTRACT

RATIONALE: Abdominal aortic aneurysms (AAAs) exhibit marked sexual dimorphism with higher prevalence in men. Similarly, AAAs induced by angiotensin II (AngII) infusion into mice exhibit a higher prevalence in males. Testosterone promotes AAA pathology in adult male mice through regulation of angiotensin type 1A receptors (AT1aR) in abdominal aortas. However, mechanisms for sexual dimorphism of regional aortic angiotensin receptor expression and AAA formation are unknown. OBJECTIVE: To define the role of developmental testosterone exposures in sexual dimorphism of AAAs, we determined if exposure of neonatal female mice to testosterone confers adult susceptibility to AngII-induced AAAs. METHODS AND RESULTS: One-day-old female hypercholesterolemic mice were administered a single dose of either vehicle or testosterone. Neonatal testosterone administration increased abdominal aortic AT1aR mRNA abundance and promoted a striking increase in AngII-induced AAAs in adult females exhibiting low serum testosterone concentrations. AngII-induced atherosclerosis and ascending aortic aneurysms were also increased by testosterone administration to neonatal females. In contrast, neonatal testosterone administration in males had no effect on AngII-induced vascular pathologies. Deficiency of AT1aR in smooth muscle cells reduced effects of neonatal testosterone to promote AAAs in adult females but did not alter atherosclerosis or ascending aortic aneurysms. Testosterone increased AT1aR mRNA abundance and hydrogen peroxide generation in cultured abdominal aortic SMCs. Increased AT1aR mRNA abundance was maintained during progressive passaging of female smooth muscle cells. CONCLUSIONS: These data reveal an unrecognized role of transient sex hormone exposures during neonatal development as long-lasting mediators of regional aortic AT1aR expression and sexual dimorphism of AAAs.


Subject(s)
Angiotensin II/toxicity , Aorta, Abdominal/metabolism , Aortic Aneurysm, Abdominal/chemically induced , Receptor, Angiotensin, Type 1/metabolism , Testosterone Propionate/toxicity , Age Factors , Animals , Animals, Newborn , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/chemically induced , Atherosclerosis/metabolism , Cells, Cultured , Female , Hydrogen Peroxide/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , RNA, Messenger/metabolism , Receptor, Angiotensin, Type 1/deficiency , Receptor, Angiotensin, Type 1/genetics , Receptors, LDL/genetics , Receptors, LDL/metabolism , Risk Factors , Sex Characteristics , Sex Factors , Testosterone Propionate/administration & dosage , Up-Regulation
13.
Curr Atheroscler Rep ; 15(4): 319, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23423526

ABSTRACT

Epigenetics involve heritable and acquired changes in gene transcription that occur independently of the DNA sequence. Epigenetic mechanisms constitute a hierarchic upper-level of transcriptional control through complex modifications of chromosomal components and nuclear structures. These modifications include, for example, DNA methylation or post-translational modifications of core histones; they are mediated by various chromatin-modifying enzymes; and ultimately they define the accessibility of a transcriptional complex to its target DNA. Integrating epigenetic mechanisms into the pathophysiologic concept of complex and multifactorial diseases such as atherosclerosis may significantly enhance our understanding of related mechanisms and provide promising therapeutic approaches. Although still in its infancy, intriguing scientific progress has begun to elucidate the role of epigenetic mechanisms in vascular biology, particularly in the control of smooth muscle cell phenotypes. In this review, we will summarize epigenetic pathways in smooth muscle cells, focusing on mechanisms involved in the regulation of vascular remodeling.


Subject(s)
Atherosclerosis/genetics , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/physiology , Atherosclerosis/physiopathology , Cell Proliferation , DNA Methylation , Epigenesis, Genetic , Histones/metabolism , Humans , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/metabolism
14.
Curr Atheroscler Rep ; 15(5): 319, 2013 May.
Article in English | MEDLINE | ID: mdl-23630979

ABSTRACT

Epigenetics involve heritable and acquired changes in gene transcription that occur independently of the DNA sequence. Epigenetic mechanisms constitute a hierarchic upper-level of transcriptional control through complex modifications of chromosomal components and nuclear structures. These modifications include, for example, DNA methylation or post-translational modifications of core histones; they are mediated by various chromatin-modifying enzymes; and ultimately they define the accessibility of a transcriptional complex to its target DNA. Integrating epigenetic mechanisms into the pathophysiologic concept of complex and multifactorial diseases such as atherosclerosis may significantly enhance our understanding of related mechanisms and provide promising therapeutic approaches. Although still in its infancy, intriguing scientific progress has begun to elucidate the role of epigenetic mechanisms in vascular biology, particularly in the control of smooth muscle cell phenotypes. In this review, we will summarize epigenetic pathways in smooth muscle cells, focusing on mechanisms involved in the regulation of vascular remodeling.


Subject(s)
Atherosclerosis/genetics , Epigenesis, Genetic/genetics , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Chromatin Assembly and Disassembly , DNA Methylation , Gene Expression Regulation , Histones/metabolism , Humans , Protein Processing, Post-Translational
15.
Arterioscler Thromb Vasc Biol ; 32(2): 397-405, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22095985

ABSTRACT

OBJECTIVE: Peroxisome proliferator-activated receptor-γ (PPARγ) ligands attenuate angiotensin II (Ang II)-induced atherosclerosis through interactions with vascular smooth muscle cell (VSMC)-specific PPARγ in hypercholesterolemic mice. Therefore, the purpose of this study was to determine the mechanism of Ang II-mediated intracellular regulation of PPARγ in VSMCs. METHODS AND RESULTS: Incubation of cultured mouse aortic VSMCs with Ang II for 24 hours reduced abundance of PPARγ protein, mRNA, and transcriptional activity (P<0.001). This effect was attenuated by an angiotensin type 1 receptor antagonist, losartan. Ang II-induced PPARγ reduction was dependent on stimulation of transforming growth factor (TGF)-ß1 as demonstrated using either a neutralizing antibody or small interfering RNA (siRNA). Ang II-induced TGF-ß1 secretion was dependent on epidermal growth factor receptor kinase activation through reactive oxygen species production. Inhibition of p38 mitogen-activated protein kinase by SB203580 or siRNA inhibited both Ang II- and TGF-ß1-induced PPARγ reduction. Blockade of TGF-ß1 decreased p38 phosphorylation induced by Ang II. siRNA-mediated inhibition of histone deacetylase 3 attenuated p38-mediated reductions in PPARγ abundance. CONCLUSIONS: These findings suggest that Ang II decreases PPARγ abundance in cultured VSMCs via an angiotensin type 1 receptor-dependent secretion of TGF-ß1 via phosphorylation of p38 mitogen-activated protein kinase and histone deacetylase 3.


Subject(s)
Angiotensin II/pharmacology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , PPAR gamma/metabolism , Transforming Growth Factor beta1/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Aorta/cytology , Aorta/drug effects , Aorta/metabolism , Cells, Cultured , Enzyme Inhibitors/pharmacology , Histone Deacetylases/drug effects , Histone Deacetylases/metabolism , Imidazoles/pharmacology , Losartan/pharmacology , Male , Mice , Mice, Inbred C57BL , Models, Animal , Muscle, Smooth, Vascular/cytology , Pyridines/pharmacology , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Receptor, Angiotensin, Type 1/drug effects , Receptor, Angiotensin, Type 1/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
16.
J Am Soc Nephrol ; 23(11): 1835-46, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23085633

ABSTRACT

Osteopontin is a proinflammatory cytokine and monocyte chemoattractant implicated in the pathogenesis of diabetic nephropathy. Synthetic agonists for liver X receptors (LXRs) suppress the expression of proinflammatory genes, including osteopontin, but whether LXR activation modulates diabetic nephropathy is unknown. We administered the LXR agonist T0901317 to mice with streptozotocin-induced diabetes and evaluated its effects on diabetic nephropathy. The LXR agonist decreased urinary albumin excretion without altering blood glucose levels and substantially attenuated macrophage infiltration, mesangial matrix accumulation, and interstitial fibrosis. LXR activation suppressed the gene expression of inflammatory mediators, including osteopontin, in the kidney cortex. In vitro, LXR activation suppressed osteopontin expression in proximal tubular epithelial cells by inhibiting AP-1-dependent transcriptional activation of the osteopontin promoter. Taken together, these results suggest that inhibition of renal osteopontin by LXR agonists may have therapeutic potential for diabetic nephropathy.


Subject(s)
Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/metabolism , Hydrocarbons, Fluorinated/pharmacology , Orphan Nuclear Receptors/agonists , Osteopontin/antagonists & inhibitors , Sulfonamides/pharmacology , Animals , Cell Line , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Gene Expression/drug effects , Inflammation Mediators/metabolism , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Ligands , Liver X Receptors , Macrophages/drug effects , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Osteopontin/genetics , Osteopontin/metabolism , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factor AP-1/metabolism
17.
J Am Coll Cardiol ; 81(1): 1-12, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36351465

ABSTRACT

BACKGROUND: Supplements are commonly used by individuals with indications for lipid-lowering therapy, but evidence of their effectiveness to lower low-density lipoprotein cholesterol (LDL-C) is lacking, particularly when compared with statins. OBJECTIVES: The trial objective was to compare the efficacy of a low-dose statin with placebo and 6 common supplements in impacting lipid and inflammatory biomarkers. METHODS: This was a single-center, prospective, randomized, single-blind clinical trial among adults with no history of atherosclerotic cardiovascular disease (ASCVD), an LDL-C of 70 to 189 mg/dL, and an increased 10-year risk of ASCVD. Participants were randomized to rosuvastatin 5 mg daily, placebo, fish oil, cinnamon, garlic, turmeric, plant sterols, or red yeast rice. The primary endpoint was the percent change in LDL-C from baseline for rosuvastatin 5 mg daily compared with placebo and each supplement after 28 days. The primary endpoint was evaluated in a hierarchical fashion with rosuvastatin first compared with placebo, then each supplement in a prespecified order using analysis of covariance. RESULTS: A total of 190 participants completed the study. The percent LDL-C reduction with rosuvastatin was greater than all supplements and placebo (P < 0.001). The difference in LDL-C reduction with rosuvastatin compared with placebo was -35.2% (95% CI: -41.3% to -29.1%; P < 0.001). None of the dietary supplements demonstrated a significant decrease in LDL-C compared with placebo. Adverse event rates were similar across study groups. CONCLUSIONS: Among individuals with increased 10-year risk for ASCVD, rosuvastatin 5 mg daily lowered LDL-C significantly more than placebo, fish oil, cinnamon, garlic, turmeric, plant sterols, and red yeast rice. (Supplements, Placebo, or Rosuvastatin Study [SPORT]; NCT04846231).


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors , Phytosterols , Rosuvastatin Calcium , Cholesterol, LDL , Single-Blind Method , Prospective Studies , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Biomarkers , Dietary Supplements , Fish Oils , Treatment Outcome
18.
Diabetes Care ; 46(Suppl 1): S191-S202, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36507634

ABSTRACT

The American Diabetes Association (ADA) "Standards of Care in Diabetes" includes the ADA's current clinical practice recommendations and is intended to provide the components of diabetes care, general treatment goals and guidelines, and tools to evaluate quality of care. Members of the ADA Professional Practice Committee, a multidisciplinary expert committee, are responsible for updating the Standards of Care annually, or more frequently as warranted. For a detailed description of ADA standards, statements, and reports, as well as the evidence-grading system for ADA's clinical practice recommendations and a full list of Professional Practice Committee members, please refer to Introduction and Methodology. Readers who wish to comment on the Standards of Care are invited to do so at professional.diabetes.org/SOC.


Subject(s)
Diabetes Mellitus , Endocrinology , Renal Insufficiency, Chronic , Humans , Standard of Care , Diabetes Mellitus/therapy , Renal Insufficiency, Chronic/therapy , Societies, Medical , Reference Standards
19.
Diabetes Care ; 46(Suppl 1): S41-S48, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36507633

ABSTRACT

The American Diabetes Association (ADA) "Standards of Care in Diabetes" includes the ADA's current clinical practice recommendations and is intended to provide the components of diabetes care, general treatment goals and guidelines, and tools to evaluate quality of care. Members of the ADA Professional Practice Committee, a multidisciplinary expert committee, are responsible for updating the Standards of Care annually, or more frequently as warranted. For a detailed description of ADA standards, statements, and reports, as well as the evidence-grading system for ADA's clinical practice recommendations and a full list of Professional Practice Committee members, please refer to Introduction and Methodology. Readers who wish to comment on the Standards of Care are invited to do so at professional.diabetes.org/SOC.


Subject(s)
Diabetes Mellitus, Type 2 , Diabetes Mellitus , Endocrinology , Humans , Diabetes Mellitus, Type 2/prevention & control , Standard of Care , Comorbidity , Diabetes Mellitus/therapy , Societies, Medical , Reference Standards
20.
Diabetes Care ; 46(Suppl 1): S111-S127, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36507635

ABSTRACT

The American Diabetes Association (ADA) "Standards of Care in Diabetes" includes the ADA's current clinical practice recommendations and is intended to provide the components of diabetes care, general treatment goals and guidelines, and tools to evaluate quality of care. Members of the ADA Professional Practice Committee, a multidisciplinary expert committee, are responsible for updating the Standards of Care annually, or more frequently as warranted. For a detailed description of ADA standards, statements, and reports, as well as the evidence-grading system for ADA's clinical practice recommendations and a full list of Professional Practice Committee members, please refer to Introduction and Methodology. Readers who wish to comment on the Standards of Care are invited to do so at professional.diabetes.org/SOC.


Subject(s)
Diabetes Mellitus , Endocrinology , Humans , Standard of Care , Diabetes Mellitus/therapy , Societies, Medical , Reference Standards
SELECTION OF CITATIONS
SEARCH DETAIL