Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters











Publication year range
1.
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
2.
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
3.
JACC Basic Transl Sci ; 1(1-2): 49-60, 2016.
Article in English | MEDLINE | ID: mdl-27127803

ABSTRACT

OBJECTIVES: The present study sought to investigate the mechanisms underlying the mitogenic function of telomerase and to test the hypothesis that everolimus, commonly used on drug-eluting stents, suppresses smooth muscle cells (SMC) proliferation by targeting telomerase. BACKGROUND: Proliferation of SMC during neointima formation is prevented by drug-eluting stents. Although the replicative capacity of mammalian cells is enhanced by telomerase expression, the contribution of telomerase to the proliferative response underlying neointima formation and its potential role as a pharmacological target remain to be investigated. METHODS: We first employed constitutive expression of telomerase reverse transcriptase (TERT) in cell systems to study transcriptional mechanisms by which telomerase activates a mitogenic program. Second, overexpression of telomerase in mice provided a model to study the role of telomerase as a drug target for the antiproliferative efficacy of everolimus. RESULTS: Inhibition of neointima formation by everolimus is lost in mice overexpressing TERT, indicating that repression of telomerase confers the antiproliferative efficacy of everolimus. Everolimus reduces TERT expression in SMC through an Ets-1-dependent inhibition of promoter activation. The inhibition of TERT-dependent SMC proliferation by everolimus occurred in the absence of telomere shortening but rather as a result of a G1→S phase arrest. Although everolimus failed to inhibit phosphorylation of the retinoblastoma protein as the gatekeeper of S-phase entry, it potently repressed downstream target genes. Using chromatin immunoprecipitation assays, we finally demonstrate that TERT induces E2F binding to S-phase gene promoters and supports histone acetylation, effects that are inhibited by everolimus and mediate its antiproliferative activity. CONCLUSIONS: These results characterize telomerase as a previously unrecognized target for the antiproliferative activity of everolimus. Our studies further identify a novel mitogenic pathway in SMC, which depends on the epigenetic activation of S-phase gene promoters by TERT.

4.
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
5.
FEBS Lett ; 588(24): 4825-30, 2014 Dec 20.
Article in English | MEDLINE | ID: mdl-25451221

ABSTRACT

The nuclear receptor NOR1 is an immediate-early response gene implicated in the transcriptional control of proliferation. Since the expression level of NOR1 is rapidly induced through cAMP response element binding (CREB) protein-dependent promoter activation, we investigated the contribution of histone acetylation to this transient induction. We demonstrate that NOR1 transcription is induced by histone deacetylase (HDAC) inhibition and by depletion of HDAC1 and HDAC3. HDAC inhibition activated the NOR1 promoter, increased histone acetylation and augmented the recruitment of phosphorylated CREB to the promoter. Furthermore, HDAC inhibition increased Ser133 phosphorylation of CREB and augmented NOR1 protein stability. These data outline previously unrecognized mechanisms of NOR1 regulation and illustrate a key role for histone acetylation in the rapid induction of NOR1.


Subject(s)
DNA-Binding Proteins/genetics , Epigenesis, Genetic , Histones/metabolism , Nerve Tissue Proteins/genetics , Orphan Nuclear Receptors/genetics , Acetylation/drug effects , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , Epigenesis, Genetic/drug effects , Histone Deacetylase Inhibitors/pharmacology , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , Protein Stability/drug effects , Rats
6.
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
7.
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
8.
Obesity (Silver Spring) ; 19(12): 2429-32, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21959341

ABSTRACT

Excessive accumulation of reactive oxygen species (ROS) in adipose tissue has been implicated in the development of insulin resistance and type 2 diabetes. However, emerging evidence suggests a physiologic role of ROS in cellular signaling and insulin sensitivity. In this study, we demonstrate that pharmacologic depletion of the antioxidant glutathione in mice prevents diet-induced obesity, increases energy expenditure and locomotor activity, and enhances insulin sensitivity. These observations support a beneficial role of ROS in glucose homeostasis and warrant further research to define the regulation of metabolism and energy balance by ROS.


Subject(s)
Diet/adverse effects , Energy Metabolism , Glutathione/metabolism , Insulin Resistance , Obesity/prevention & control , Reactive Oxygen Species/metabolism , Animals , Antioxidants/metabolism , Behavior, Animal , Blood Glucose/metabolism , Homeostasis , Locomotion , Mice , Mice, Inbred C57BL , Obesity/etiology , Signal Transduction
9.
J Biol Chem ; 286(41): 35485-35493, 2011 Oct 14.
Article in English | MEDLINE | ID: mdl-21868379

ABSTRACT

Members of the NR4A subgroup of the nuclear hormone receptor superfamily have emerged as key transcriptional regulators of proliferation and inflammation. NOR1 constitutes a ligand-independent transcription factor of this subgroup and induces cell proliferation; however, the transcriptional mechanisms underlying this mitogenic role remain to be defined. Here, we demonstrate that the F-box protein SKP2 (S phase kinase-associated protein 2), the substrate-specific receptor of the ubiquitin ligase responsible for the degradation of p27(KIP1) through the proteasome pathway, constitutes a direct transcriptional target for NOR1. Mitogen-induced Skp2 expression is silenced in vascular smooth muscle cells (VSMC) isolated from Nor1-deficient mice or transfected with Nor1 siRNA. Conversely, adenovirus-mediated overexpression of NOR1 induces Skp2 expression in VSMC and decreases protein abundance of its target p27. Transient transfection experiments establish that NOR1 transactivates the Skp2 promoter through a nerve growth factor-induced clone B response element (NBRE). Electrophoretic mobility shift and chromatin immunoprecipitation assays further revealed that NOR1 is recruited to this NBRE site in the Skp2 promoter in response to mitogenic stimulation. In vivo Skp2 expression is increased during the proliferative response underlying neointima formation, and this transcriptional induction depends on the expression of NOR1. Finally, we demonstrate that overexpression of Skp2 rescues the proliferative arrest of Nor1-deficient VSMC. Collectively, these results characterize Skp2 as a novel NOR1-regulated target gene and detail a previously unrecognized transcriptional cascade regulating mitogen-induced VSMC proliferation.


Subject(s)
Cell Proliferation , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Enzymologic , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/metabolism , Response Elements , S-Phase Kinase-Associated Proteins/biosynthesis , Animals , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , DNA-Binding Proteins/genetics , Mice , Mice, Knockout , Neointima/genetics , Neointima/metabolism , Nerve Tissue Proteins/genetics , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/genetics , S-Phase Kinase-Associated Proteins/genetics , Transcriptional Activation/genetics
10.
PLoS One ; 6(4): e18532, 2011 Apr 14.
Article in English | MEDLINE | ID: mdl-21533223

ABSTRACT

Aging constitutes a major independent risk factor for the development of type 2 diabetes and is accompanied by insulin resistance and adipose tissue dysfunction. One of the most important factors implicitly linked to aging and age-related chronic diseases is the accumulation of oxidative stress. However, the effect of increased oxidative stress on adipose tissue biology remains elusive. In this study, we demonstrate that aging in mice results in a loss of fat mass and the accumulation of oxidative stress in adipose tissue. In vitro, increased oxidative stress through glutathione depletion inhibits preadipocyte differentiation. This inhibition of adipogenesis is at least in part the result of reduced cell proliferation and an inhibition of G(1)→S-phase transition during the initial mitotic clonal expansion of the adipocyte differentiation process. While phosphorylation of the retinoblastoma protein (Rb) by cyclin/cdk complexes remains unaffected, oxidative stress decreases the expression of S-phase genes downstream of Rb. This silencing of S phase gene expression by increased oxidative stress is mediated through a transcriptional mechanism involving the inhibition of E2F recruitment and transactivation of its target promoters. Collectively, these data demonstrate a previously unrecognized role of oxidative stress in the regulation of adipogenesis which may contribute to age-associated adipose tissue dysfunction.


Subject(s)
Adipose Tissue/metabolism , Aging/metabolism , Oxidative Stress , 3T3-L1 Cells , Adipose Tissue/growth & development , Animals , Base Sequence , Body Composition , Chromatin Immunoprecipitation , DNA Primers , Glutathione/metabolism , Male , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction
11.
Arterioscler Thromb Vasc Biol ; 31(4): 851-60, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21233448

ABSTRACT

OBJECTIVE: Proliferation of smooth muscle cells (SMC) in response to vascular injury is central to neointimal vascular remodeling. There is accumulating evidence that histone acetylation constitutes a major epigenetic modification for the transcriptional control of proliferative gene expression; however, the physiological role of histone acetylation for proliferative vascular disease remains elusive. METHODS AND RESULTS: In the present study, we investigated the role of histone deacetylase (HDAC) inhibition in SMC proliferation and neointimal remodeling. We demonstrate that mitogens induce transcription of HDAC 1, 2, and 3 in SMC. Short interfering RNA-mediated knockdown of either HDAC 1, 2, or 3 and pharmacological inhibition of HDAC prevented mitogen-induced SMC proliferation. The mechanisms underlying this reduction of SMC proliferation by HDAC inhibition involve a growth arrest in the G(1) phase of the cell cycle that is due to an inhibition of retinoblastoma protein phosphorylation. HDAC inhibition resulted in a transcriptional and posttranscriptional regulation of the cyclin-dependent kinase inhibitors p21(Cip1) and p27(Kip). Furthermore, HDAC inhibition repressed mitogen-induced cyclin D1 mRNA expression and cyclin D1 promoter activity. As a result of this differential cell cycle-regulatory gene expression by HDAC inhibition, the retinoblastoma protein retains a transcriptional repression of its downstream target genes required for S phase entry. Finally, we provide evidence that these observations are applicable in vivo by demonstrating that HDAC inhibition decreased neointima formation and expression of cyclin D1 in a murine model of vascular injury. CONCLUSIONS: These findings identify HDAC as a critical component of a transcriptional cascade regulating SMC proliferation and suggest that HDAC might play a pivotal role in the development of proliferative vascular diseases, including atherosclerosis and in-stent restenosis.


Subject(s)
Cell Proliferation/drug effects , Epigenesis, Genetic/drug effects , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Hydroxylamines/pharmacology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Quinolines/pharmacology , Tunica Media/drug effects , Vascular System Injuries/drug therapy , Acetylation , Animals , Cell Cycle/drug effects , Cell Cycle Proteins/metabolism , Cells, Cultured , Chromatin Assembly and Disassembly/drug effects , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Disease Models, Animal , E2F Transcription Factors/metabolism , Histone Deacetylases/genetics , Histones/metabolism , Hyperplasia , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/injuries , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/pathology , Phosphorylation , RNA Interference , Rats , Retinoblastoma Protein/metabolism , Time Factors , Transcription, Genetic/drug effects , Tunica Media/enzymology , Tunica Media/injuries , Tunica Media/pathology , Vascular System Injuries/enzymology , Vascular System Injuries/pathology
12.
Arterioscler Thromb Vasc Biol ; 31(2): 245-52, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21106948

ABSTRACT

OBJECTIVE: Telomerase serves as a critical regulator of tissue renewal. Although telomerase activity is inducible in response to various environmental cues, it remains unknown whether telomerase is activated during the inflammatory remodeling underlying atherosclerosis formation. To address this question, we investigated in the present study the regulation of telomerase in macrophages and during atherosclerosis development in low-density lipoprotein receptor-deficient mice. METHODS AND RESULTS: We demonstrate that inflammatory stimuli activate telomerase in macrophages by inducing the expression of the catalytic subunit telomerase reverse transcriptase (TERT). Reporter and chromatin immunoprecipitation assays identified a previously unrecognized nuclear factor-κB (NF-κB) response element in the TERT promoter, to which NF-κB is recruited during inflammation. Inhibition of NF-κB signaling completely abolished the induction of TERT expression, characterizing TERT as a bona fide NF-κB target gene. Furthermore, functional experiments revealed that TERT deficiency results in a senescent cell phenotype. Finally, we demonstrate high levels of TERT expression in macrophages of human atherosclerotic lesions and establish that telomerase is activated during atherosclerosis development in low-density lipoprotein receptor-deficient mice. CONCLUSIONS: These results characterize TERT as a previously unrecognized NF-κB target gene in macrophages and demonstrate that telomerase is activated during atherosclerosis. This induction of TERT expression prevents macrophage senescence and may have important implications for the development of atherosclerosis.


Subject(s)
Atherosclerosis/metabolism , Inflammation/metabolism , Macrophages/enzymology , Telomerase/metabolism , Animals , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Cells, Cultured , Coronary Vessels/metabolism , Coronary Vessels/pathology , Coronary Vessels/physiopathology , Disease Models, Animal , Humans , Inflammation/pathology , Inflammation/physiopathology , Lipopolysaccharides/pharmacology , Lipoproteins, LDL/pharmacology , Macrophages/drug effects , Macrophages/pathology , Mice , Mice, Knockout , NF-kappa B/metabolism , Receptors, LDL/deficiency , Receptors, LDL/genetics , Telomerase/genetics , Tumor Necrosis Factor-alpha/pharmacology
13.
Arterioscler Thromb Vasc Biol ; 31(2): 253-60, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21088250

ABSTRACT

OBJECTIVE: Abdominal aortic aneurysms (AAA) are an age-related vascular disease and an important cause of morbidity and mortality. In this study, we sought to determine whether the catalytic component of telomerase, telomerase reverse transcriptase (TERT), modulates angiotensin (Ang) II-induced AAA formation. METHODS AND RESULTS: Low-density lipoprotein receptor-deficient (LDLr-/-) mice were lethally irradiated and reconstituted with bone marrow-derived cells from TERT-deficient (TERT-/-) mice or littermate wild-type mice. Mice were placed on a diet enriched in cholesterol, and AAA formation was quantified after 4 weeks of Ang II infusion. Repopulation of LDLr-/- mice with TERT-/- bone marrow-derived cells attenuated Ang II-induced AAA formation. TERT-deficient recipient mice revealed modest telomere attrition in circulating leukocytes at the study end point without any overt effect of the donor genotype on white blood cell counts. In mice repopulated with TERT-/- bone marrow, aortic matrix metalloproteinase-2 (MMP-2) activity was reduced, and TERT-/- macrophages exhibited decreased expression and activity of MMP-2 in response to stimulation with Ang II. Finally, we demonstrated in transient transfection studies that TERT overexpression activates the MMP-2 promoter in macrophages. CONCLUSIONS: TERT deficiency in bone marrow-derived macrophages attenuates Ang II-induced AAA formation in LDLr-/- mice and decreases MMP-2 expression. These results point to a previously unrecognized role of TERT in the pathogenesis of AAA.


Subject(s)
Angiotensin II/adverse effects , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/prevention & control , Macrophages/enzymology , Telomerase/deficiency , Animals , Bone Marrow Transplantation , Cell Movement , Cells, Cultured , Elastin/metabolism , Genotype , Macrophages/metabolism , Macrophages/pathology , Male , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Knockout , Models, Animal , Receptors, LDL/genetics , Receptors, LDL/metabolism , Telomerase/genetics , Telomere
14.
Circ Res ; 107(4): 501-11, 2010 Aug 20.
Article in English | MEDLINE | ID: mdl-20558821

ABSTRACT

RATIONALE: The orphan nuclear receptor NOR1 is a member of the evolutionary highly conserved and ligand-independent NR4A subfamily of the nuclear hormone receptor superfamily. Members of this subfamily have been characterized as early response genes regulating essential biological processes including inflammation and proliferation; however, the role of NOR1 in atherosclerosis remains unknown. OBJECTIVE: The goal of the present study was to determine the causal contribution of NOR1 to atherosclerosis development and to identify the mechanism by which this nuclear receptor participates in the disease process. METHODS AND RESULTS: In the present study, we demonstrate expression of NOR1 in endothelial cells of human atherosclerotic lesions. In response to inflammatory stimuli, NOR1 expression is rapidly induced in endothelial cells through a nuclear factor kappaB-dependent transactivation of the NOR1 promoter. Overexpression of NOR1 in human endothelial cells increased the expression of vascular cell adhesion molecule (VCAM)-1 and intercellular adhesion molecule-1, whereas NOR1 deficiency altered adhesion molecule expression in response to inflammatory stimuli. Transient transfection experiments and chromatin immunoprecipitation assays revealed that NOR1 induces VCAM-1 promoter activity by binding to a canonical response element for NR4A receptors in the VCAM-1 promoter. Further functional studies confirmed that NOR1 mediates monocyte adhesion by inducing VCAM-1 and intercellular adhesion molecule-1 expression in endothelial cells. Finally, we demonstrate that NOR1 deficiency reduces hypercholesterolemia-induced atherosclerosis formation in apoE(-/-) mice by decreasing the macrophage content of the lesion. CONCLUSIONS: In concert, these studies identify a novel pathway underlying monocyte adhesion and establish that NOR1 serves a previously unrecognized atherogenic role in mice by positively regulating monocyte recruitment to the vascular wall.


Subject(s)
Atherosclerosis/metabolism , DNA-Binding Proteins/deficiency , Monocytes/physiology , Nuclear Receptor Subfamily 4, Group A, Member 1/deficiency , Receptors, Steroid/deficiency , Receptors, Thyroid Hormone/deficiency , Animals , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Cell Adhesion/physiology , Cells, Cultured , Coronary Vessels/metabolism , Coronary Vessels/pathology , DNA-Binding Proteins/metabolism , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/metabolism , Response Elements/genetics , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
15.
Circulation ; 119(4): 577-86, 2009 Feb 03.
Article in English | MEDLINE | ID: mdl-19153266

ABSTRACT

BACKGROUND: The neuron-derived orphan receptor-1 (NOR1) belongs to the evolutionary highly conserved and most ancient NR4A subfamily of the nuclear hormone receptor superfamily. Members of this subfamily function as early-response genes regulating key cellular processes, including proliferation, differentiation, and survival. Although NOR1 has previously been demonstrated to be required for smooth muscle cell proliferation in vitro, the role of this nuclear receptor for the proliferative response underlying neointima formation and target genes trans-activated by NOR1 remain to be defined. METHODS AND RESULTS: Using a model of guidewire-induced arterial injury, we demonstrate decreased neointima formation in NOR1(-/-) mice compared with wild-type mice. In vitro, NOR1-deficient smooth muscle cells exhibit decreased proliferation as a result of a G(1)-->S phase arrest of the cell cycle and increased apoptosis in response to serum deprivation. NOR1 deficiency alters phosphorylation of the retinoblastoma protein by preventing mitogen-induced cyclin D1 and D2 expression. Conversely, overexpression of NOR1 induces cyclin D1 expression and the transcriptional activity of the cyclin D1 promoter in transient reporter assays. Gel shift and chromatin immunoprecipitation assays identified a putative response element for NR4A receptors in the cyclin D1 promoter, to which NOR1 is recruited in response to mitogenic stimulation. Finally, we provide evidence that these observations are applicable in vivo by demonstrating decreased cyclin D1 expression during neointima formation in NOR1-deficient mice. CONCLUSIONS: These experiments characterize cyclin D1 as an NOR1-regulated target gene in smooth muscle cells and demonstrate that NOR1 deficiency decreases neointima formation in response to vascular injury.


Subject(s)
DNA-Binding Proteins/genetics , Muscle, Smooth, Vascular/injuries , Muscle, Smooth, Vascular/physiology , Receptors, Steroid/genetics , Wound Healing/physiology , Animals , Aorta/cytology , Apoptosis/physiology , Cell Division/physiology , Cell Survival/physiology , Cells, Cultured , Coronary Vessels/cytology , Cyclin D1/genetics , Cyclin D2 , Cyclins/genetics , DNA-Binding Proteins/metabolism , E2F Transcription Factors/metabolism , Gene Expression/physiology , Humans , Mice , Mice, Mutant Strains , Muscle, Smooth, Vascular/cytology , Nuclear Receptor Subfamily 4, Group A, Member 1 , Phosphorylation/physiology , Promoter Regions, Genetic/physiology , Rats , Receptors, Steroid/metabolism , Retinoblastoma Protein/metabolism , Tunica Intima/cytology , Tunica Intima/injuries , Tunica Intima/physiology
16.
Circ Res ; 103(10): 1155-63, 2008 Nov 07.
Article in English | MEDLINE | ID: mdl-18818403

ABSTRACT

Peroxisome proliferator-activated receptor (PPAR)alpha, the molecular target for fibrates used to treat dyslipidemia, exerts pleiotropic effects on vascular cells. In vascular smooth muscle cells (VSMCs), we have previously demonstrated that PPARalpha activation suppresses G(1)-->S cell cycle progression by targeting the cyclin-dependent kinase inhibitor p16(INK4a) (p16). In the present study, we demonstrate that this inhibition of VSMC proliferation by PPARalpha is mediated through a p16-dependent suppression of telomerase activity, which has been implicated in key cellular functions including proliferation. PPARalpha activation inhibited mitogen-induced telomerase activity by repressing the catalytic subunit telomerase reverse transcriptase (TERT) through negative cross-talk with an E2F-1-dependent trans-activation of the TERT promoter. This trans-repression involved the recruitment of the retinoblastoma (RB) family proteins p107 and p130 to the TERT promoter resulting in impaired E2F-1 binding, an effect that was dependent on p16. The inhibition of cell proliferation by PPARalpha activation was lost in VSMCs following TERT overexpression or knockdown, pointing to a key role of telomerase as a target for the antiproliferative effects of PPARalpha. Finally, we demonstrate that PPARalpha agonists suppress telomerase activation during the proliferative response following vascular injury, indicating that these findings are applicable in vivo. In concert, these results demonstrate that the antiproliferative effects of PPARalpha in VSMCs depend on the suppression of telomerase activity by targeting the p16/RB/E2F transcriptional cascade.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/metabolism , G1 Phase/physiology , Gene Expression Regulation, Enzymologic/physiology , Myocytes, Smooth Muscle/enzymology , PPAR alpha/metabolism , S Phase/physiology , Telomerase/biosynthesis , Animals , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/genetics , Dyslipidemias/enzymology , Dyslipidemias/genetics , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , Enzyme Activation/drug effects , Enzyme Activation/physiology , Gene Expression Regulation, Enzymologic/drug effects , Mice , Mitogens/metabolism , Mitogens/pharmacology , PPAR alpha/agonists , Promoter Regions, Genetic/physiology , Rats , Retinoblastoma-Like Protein p107/genetics , Retinoblastoma-Like Protein p107/metabolism , Retinoblastoma-Like Protein p130/genetics , Retinoblastoma-Like Protein p130/metabolism , Telomerase/genetics , Transcription, Genetic/drug effects , Transcription, Genetic/physiology
17.
J Clin Invest ; 117(10): 2877-88, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17823662

ABSTRACT

Obesity is associated with a state of chronic, low-grade inflammation characterized by abnormal cytokine production and macrophage infiltration into adipose tissue, which may contribute to the development of insulin resistance. During immune responses, tissue infiltration by macrophages is dependent on the expression of osteopontin, an extracellular matrix protein and proinflammatory cytokine that promotes monocyte chemotaxis and cell motility. In the present study, we used a murine model of diet-induced obesity to examine the role of osteopontin in the accumulation of adipose tissue macrophages and the development of insulin resistance during obesity. Mice exposed to a high-fat diet exhibited increased plasma osteopontin levels, with elevated expression in macrophages recruited into adipose tissue. Obese mice lacking osteopontin displayed improved insulin sensitivity in the absence of an effect on diet-induced obesity, body composition, or energy expenditure. These mice further demonstrated decreased macrophage infiltration into adipose tissue, which may reflect both impaired macrophage motility and attenuated monocyte recruitment by stromal vascular cells. Finally, obese osteopontin-deficient mice exhibited decreased markers of inflammation, both in adipose tissue and systemically. Taken together, these results suggest that osteopontin may play a key role in linking obesity to the development of insulin resistance by promoting inflammation and the accumulation of macrophages in adipose tissue.


Subject(s)
Adipose Tissue/immunology , Insulin Resistance/immunology , Macrophages/immunology , Obesity/immunology , Osteopontin/physiology , Animals , Chemokine CCL2/metabolism , Chemotaxis/genetics , Dietary Fats/administration & dosage , Inflammation/genetics , Inflammation/immunology , Mice , Mice, Mutant Strains , Obesity/complications , Osteopontin/genetics
18.
Diabetes ; 56(6): 1662-70, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17360982

ABSTRACT

Osteopontin (OPN) is a proinflammatory cytokine implicated in the chemoattraction of monocytes and the development of atherosclerosis. Peroxisome proliferator-activated receptor (PPAR)alpha, a ligand-activated transcription factor with pleiotropic anti-inflammatory effects in macrophages, is the molecular target for fibrates, which are frequently used to treat dyslipidemia in patients with type 2 diabetes at high risk for cardiovascular disease. In the present study, we examined the regulation of OPN by PPARalpha agonists in macrophages and determined the effect of fibrate treatment on OPN plasma levels in patients with type 2 diabetes. Treatment of human macrophages with the PPARalpha ligands bezafibrate or WY14643 inhibited OPN expression. PPARalpha ligands suppressed OPN promoter activity, and an activator protein (AP)-1 consensus site conferred this repression. Overexpression of c-Fos and c-Jun reversed the inhibitory effect of PPARalpha ligands on OPN transcription, and, in chromatin immunoprecipitation assays, PPARalpha ligands inhibited c-Fos and phospho-c-Jun binding to the OPN promoter. Moreover, c-Fos and phospho-c-Jun protein expression was inhibited by PPARalpha agonists, indicating that PPARalpha ligands suppress OPN expression through negative cross talk with AP-1-dependent transactivation of the OPN promoter. This inhibitory effect of PPARalpha ligands on OPN expression was absent in PPARalpha-deficient macrophages, suggesting a receptor-mediated mechanism of OPN suppression. Finally, treatment of type 2 diabetic patients with bezafibrate significantly decreased OPN plasma levels. These results demonstrate a novel mechanism whereby PPARalpha ligands may impact macrophage inflammatory responses and decrease early proinflammatory markers for cardiovascular disease.


Subject(s)
Bezafibrate/pharmacology , Diabetes Mellitus, Type 2/blood , Gene Expression Regulation/drug effects , Macrophages/physiology , Osteopontin/genetics , PPAR alpha/agonists , Animals , Cell Line , Humans , Hypolipidemic Agents/pharmacology , Ligands , Mice , Monocytes/physiology , Osteopontin/blood , Plasmids , Polymerase Chain Reaction , Pyrimidines/pharmacology , Transfection
19.
J Biol Chem ; 281(44): 33467-76, 2006 Nov 03.
Article in English | MEDLINE | ID: mdl-16945922

ABSTRACT

Members of the nuclear hormone receptor superfamily function as key transcriptional regulators of inflammation and proliferation in cardiovascular diseases. In addition to the ligand-dependent peroxisome proliferator-activated receptors and liver X receptors, this family of transcription factors includes a large number of orphan receptors, and their role in vascular diseases remains to be investigated. The neuron-derived orphan receptor-1 (NOR1) belongs to the ligand-independent NR4A subfamily, which has been implicated in cell proliferation, differentiation, and apoptosis. In this study, we demonstrate NOR1 expression in vascular smooth muscle cells (SMC) of human atherosclerotic lesions. In response to mitogenic stimulation with platelet-derived growth factor (PDGF), SMC rapidly express NOR1 through an ERK-MAPK-dependent signaling pathway. 5'-deletion analysis, site-directed mutagenesis, and transactivation experiments demonstrate that PDGF-induced NOR1 expression is mediated through a cAMP-response element-binding protein (CREB)-dependent transactivation of the NOR1 promoter. Consequently, short interfering RNA-mediated depletion of CREB abolished PDGF-induced NOR1 expression in SMC. Furthermore, PDGF induced Ser-133 phosphorylation of CREB and subsequent binding to the CRE sites of the endogenous NOR1 promoter. Functional analysis demonstrated that PDGF induces NOR1 transactivation of its consensus NGFI-B-response elements (NBRE) in SMC. We finally demonstrate that SMC isolated from NOR1-deficient mice exhibit decreased cell proliferation and characterize cyclin D1 and D2 as NOR1 target genes in SMC. These experiments indicate that PDGF-induced NOR1 transcription in SMC is mediated through CREB-dependent transactivation of the NOR1 promoter and further demonstrate that NOR1 functions as a key transcriptional regulator of SMC proliferation.


Subject(s)
DNA-Binding Proteins/metabolism , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Nerve Tissue Proteins/metabolism , Platelet-Derived Growth Factor/pharmacology , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/metabolism , Animals , Atherosclerosis/metabolism , Cell Proliferation , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , DNA-Binding Proteins/genetics , Gene Expression Regulation , Humans , MAP Kinase Signaling System , Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Nerve Tissue Proteins/genetics , Phosphoserine/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , RNA, Messenger/genetics , Rats , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/genetics , Response Elements , Transcription, Genetic/genetics , Transcriptional Activation/genetics
20.
Circ Res ; 98(7): e50-9, 2006 Apr 14.
Article in English | MEDLINE | ID: mdl-16556873

ABSTRACT

Activation of the peroxisome proliferator-activated receptor (PPAR) gamma, the molecular target for insulin sensitizing thiazolidinediones used in patients with type 2 diabetes, inhibits vascular smooth muscle cell (VSMC) proliferation and prevents atherosclerosis and neointima formation. Emerging evidence indicates that telomerase controls key cellular functions including replicative lifespan, differentiation, and cell proliferation. In the present study, we demonstrate that ligand-induced and constitutive PPARgamma activation inhibits telomerase activity in VSMCs. Telomerase reverse transcriptase (TERT) confers the catalytic activity of telomerase, and PPARgamma ligands inhibit TERT expression through a receptor-dependent suppression of the TERT promoter. 5'-deletion analysis, site-directed mutagenesis, and transactivation studies using overexpression of Ets-1 revealed that suppression of TERT transcription by PPARgamma is mediated through negative cross-talk with Ets-1-dependent transactivation of the TERT promoter. Chromatin immunoprecipitation assays further demonstrated that PPARgamma ligands inhibit Ets-1 binding to the TERT promoter, which is mediated at least in part through an inhibition of Ets-1 expression by PPARgamma ligands. In VSMCs overexpressing TERT, the efficacy of PPARgamma ligands to inhibit cell proliferation is lost, indicating that TERT constitutes an important molecular target for the antiproliferative effects of PPARgamma ligands. Finally, we demonstrate that telomerase activation during the proliferative response after vascular injury is effectively inhibited by PPARgamma ligands. These findings provide a previously unrecognized mechanism for the antiproliferative effects of PPARgamma ligands and support the concept that PPARgamma ligands may constitute a novel therapeutic approach for the treatment of proliferative cardiovascular diseases.


Subject(s)
Muscle, Smooth, Vascular/physiology , PPAR gamma/physiology , Telomerase/antagonists & inhibitors , Animals , Aorta , Base Sequence , Cardiovascular Diseases/therapy , Cell Division/physiology , Cells, Cultured , DNA Primers , Enzyme Activation , Microscopy, Confocal , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/enzymology , Mutagenesis, Site-Directed , PPAR gamma/genetics , Rats , Recombinant Proteins/metabolism , Telomerase/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL