Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
J Clin Med ; 12(13)2023 Jul 02.
Article in English | MEDLINE | ID: mdl-37445481

ABSTRACT

Much attention has been paid lately to harnessing the diagnostic and therapeutic potential of non-coding circular ribonucleic acids (circRNAs) and micro-RNAs (miRNAs) for the prevention and treatment of cardiovascular diseases. The genetic environment that contributes to atherosclerosis pathophysiology is immensely complex. Any potential therapeutic application of circRNAs must be assessed for risks, benefits, and off-target effects in both the short and long term. A search of the online PubMed database for publications related to circRNA and atherosclerosis from 2016 to 2022 was conducted. These studies were reviewed for their design, including methods for developing atherosclerosis and the effects of the corresponding atherosclerotic environment on circRNA expression. Investigated mechanisms were recorded, including associated miRNA, genes, and ultimate effects on cell mechanics, and inflammatory markers. The most investigated circRNAs were then further analyzed for redundant, disparate, and/or contradictory findings. Many disparate, opposing, and contradictory effects were observed across experiments. These include levels of the expression of a particular circRNA in atherosclerotic environments, attempted ascertainment of the in toto effects of circRNA or miRNA silencing on atherosclerosis progression, and off-target, cell-specific, and disease-specific effects. The high potential for detrimental and unpredictable off-target effects downstream of circRNA manipulation will likely render the practice of therapeutic targeting of circRNA or miRNA molecules not only complicated but perilous.

2.
Pharmacol Res Perspect ; 7(4): e00509, 2019 08.
Article in English | MEDLINE | ID: mdl-31372229

ABSTRACT

The novel clopidogrel conjugate, DT-678, is an effective inhibitor of platelets and thrombosis in preclinical studies. However, a comparison of the bleeding risk with DT-678 and currently approved P2Y12 antagonists has yet to be determined. The objective of this study was to evaluate the bleeding tendency of animals treated with clopidogrel, ticagrelor, and DT-678. Ninety-one New Zealand white rabbits were randomized to one of 13 treatment groups (n = 7). Platelet activation was assessed by flow cytometry and light transmission aggregometry before and after the administration of various doses of DT-678, clopidogrel, and ticagrelor. Tongue template bleeding times were also measured before and after drug treatment. Treatment with P2Y12 receptor antagonists caused a dose-dependent reduction in markers of platelet activation (P-selectin and integrin αIIbß3) and aggregation in response to adenosine diphosphate stimulation. At the same doses required for platelet inhibition, clopidogrel and ticagrelor significantly prolonged bleeding times, while DT-678 did not. DT-678 and the FDA-approved P2Y12 antagonists clopidogrel and ticagrelor are effective inhibitors of platelet activation and aggregation. However, unlike clopidogrel and ticagrelor, DT-678 did not prolong bleeding times at equally effective antiplatelet doses. The results suggest a more favorable benefit/risk ratio for DT-678 and potential utility as part of a dual antiplatelet therapy regimen.


Subject(s)
Disulfides/administration & dosage , Platelet Activation/drug effects , Purinergic P2Y Receptor Antagonists/administration & dosage , Animals , Bleeding Time , Clopidogrel/administration & dosage , Clopidogrel/chemistry , Clopidogrel/pharmacology , Disulfides/chemistry , Disulfides/pharmacology , Dose-Response Relationship, Drug , Purinergic P2Y Receptor Antagonists/pharmacology , Rabbits , Random Allocation , Ticagrelor/administration & dosage , Ticagrelor/pharmacology
3.
Mol Ther Nucleic Acids ; 16: 73-81, 2019 Jun 07.
Article in English | MEDLINE | ID: mdl-30852378

ABSTRACT

Cystic fibrosis (CF) is a lethal autosomal recessive disease caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. Nuclease-mediated precise gene editing (PGE) represents a promising therapy for CF, for which an efficient strategy that is free of viral vector, drug selection, and reporter enrichment (VDR free) is desirable. Here we compared different transfection methods (lipofectamine versus electroporation) and formats (plasmid DNA versus ribonucleoprotein) in delivering the CRISPR/Cas9 elements along with single-stranded oligodeoxynucleotides (ssODNs) to clinically relevant cells targeting major CFTR mutation loci. We demonstrate that, among different combinations, electroporation of CRISPR/Cas9 and guide RNA (gRNA) ribonucleoprotein (Cas9 RNP) is the most effective one. By using this VDR-free method, 4.8% to 27.2% efficiencies were achieved in creating dF508, G542X, and G551D mutations in a wild-type induced pluripotent stem cell (iPSC) line. When it is applied to a patient-derived iPSC line carrying the dF508 mutation, a greater than 20% precise correction rate was achieved. As expected, genetic correction leads to the restoration of CFTR function in iPSC-derived proximal lung organoids, as well as in a patient-derived adenocarcinoma cell line CFPAC-1. The present work demonstrates the feasibility of gene editing-based therapeutics toward monogenic diseases such as CF.

4.
Biochem Biophys Res Commun ; 409(1): 52-7, 2011 May 27.
Article in English | MEDLINE | ID: mdl-21549102

ABSTRACT

Rad is a member of a subclass of small GTP-binding proteins, the RGK family. In the present study we investigated the role of Rad protein in regulating cardiomyocyte viability. DNA fragmentation and TUNEL assays demonstrated that Rad promoted rat neonatal cardiomyocyte apoptosis. Rad silencing fully blocked serum deprivation induced apoptosis, indicating Rad is necessary for trigger cardiomyocyte apoptosis. Rad overexpression caused a dramatic decrease of the anti-apoptotic molecule Bcl-x(L), whereas Bcl-x(L) overexpression protected cardiomyocytes against Rad-induced apoptosis. Rad-triggered apoptosis was mediated by the activation of p38 MAPK. The p38 blocker SB203580 effectively protected cardiomyocytes against Rad-evoked apoptosis.


Subject(s)
Apoptosis , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/physiology , p38 Mitogen-Activated Protein Kinases/metabolism , ras Proteins/metabolism , Animals , Cell Survival , Cells, Cultured , Imidazoles/pharmacology , Myocytes, Cardiac/drug effects , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , RNA, Small Interfering/genetics , Rats , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , ras Proteins/genetics
5.
Endocrinology ; 149(8): 4086-94, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18467446

ABSTRACT

Nitration products of unsaturated fatty acids are formed via NO-dependent oxidative reactions and appear to be a new class of endogenous antiinflammatory mediators. Nitroalkene derivatives of nitrated linoleic acid (LNO(2)) and nitrated oleic acid (OA-NO(2)) alleviate inflammatory responses in macrophages, but the underlying mechanisms remain to be fully defined. Herein we report that LNO(2) and OA-NO(2) suppress proinflammatory signal transducer and activator of transcription (STAT) signaling in macrophages. In RAW264.7 cells, a murine macrophage cell line, LNO(2) and OA-NO(2) inhibited the lipopolysaccharide (LPS)-induced STAT1 phosphorylation and the STAT1-dependent transcriptional activity, thereby suppressing expression of its target gene such as iNOS and MCP-1. The nitroalkene-mediated inhibition of STAT1 activity was not affected by 2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl 3-oxide (a NO scavenger), GW9662 (a peroxisome proliferator-activated receptor-gamma-specific antagonist) or glutathione (an antioxidant), suggesting an underlying mechanism independent of NO, peroxisome proliferator-activated receptor-gamma, or thio-nitralkylation. In contrast, LNO(2) or OA-NO(2) alone up-regulated both mRNA and protein levels of MAPK phosphatase 1 (MKP-1) and strongly augmented the LPS-induced MKP-1 protein expression. Knockdown of MKP-1 by MKP-1 small interfering RNA enhanced the LPS-induced STAT1 phosphorylation, suggesting that MKP-1 acts as a negative regulator for LPS-induced STAT signaling. In addition, the nitroalkene-mediated inhibitory effects on STAT1 phosphorylation, iNOS expression, and MCP-1 secretion were also largely attenuated by the MKP-1 small interfering RNA approach. Taken together, our data demonstrate that nitroalkenes inhibit proinflammatory STAT signaling through inducting MKP-1 in macrophages.


Subject(s)
Alkenes/pharmacology , Dual Specificity Phosphatase 1/physiology , Lipopolysaccharides/pharmacology , Macrophages/drug effects , STAT1 Transcription Factor/metabolism , Animals , Cells, Cultured , Dual Specificity Phosphatase 1/metabolism , Inflammation/metabolism , Linoleic Acids/pharmacology , Macrophages/metabolism , Mice , Nitric Oxide/metabolism , Nitro Compounds/pharmacology , Oleic Acids/pharmacology , Phosphorylation/drug effects , Signal Transduction/drug effects , Sulfhydryl Compounds/metabolism , Sulfhydryl Compounds/pharmacology
6.
Cardiovasc Res ; 76(1): 141-8, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17631285

ABSTRACT

OBJECTIVE: Id2 (inhibitor of DNA-binding 2), a member of the helix-loop-helix family of transcription regulators, plays important roles in cell proliferation and differentiation. Recent reports have documented that Id2 is up-regulated during vascular lesion formation and overexpression of Id2 promotes vascular smooth muscle cell (VSMC) proliferation. However, the transcriptional regulation of Id2 gene expression in VSMC remains unexplored. METHODS AND RESULTS: Using Northern- and Western-blot analyses, we documented that interleukin-1beta (IL-1beta) induced Id2 gene expression in VSMC in a time- and dose-dependent manner. Overexpression of early growth response-1 (Egr-1) in VSMC induced Id2 expression while IL-1beta-induced Id2 expression was abrogated in VSMC by the Egr-1 repressor, NGFI-A binding protein 2 (NAB2), expressed from an adenovirus. Overexpression of Egr-1 transactivated the Id2 promoter in reporter assays dependent on the presence of intact putative Egr-1 binding sites as determined by mutagenesis. Finally, electrophoretic mobility shift assays (EMSA) demonstrated that the Egr-1 protein can bind the Egr-1 sites derived from the human Id2 promoter in vitro and chromatin immunoprecipitation identified the putative Egr-1 site between -723 to -712 as the functional Egr-1 binding site in vivo. CONCLUSIONS: Our data demonstrate that IL-1beta-induced Id2 expression in VSMC is mediated by the transcription factor Egr-1 in VSMC.


Subject(s)
Early Growth Response Protein 1/genetics , Gene Expression Regulation , Inhibitor of Differentiation Protein 2/genetics , Interleukin-1beta/pharmacology , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle/metabolism , Adenoviridae/genetics , Analysis of Variance , Binding Sites , Cells, Cultured , Chromatin Immunoprecipitation , Dose-Response Relationship, Drug , Early Growth Response Protein 1/antagonists & inhibitors , Electrophoretic Mobility Shift Assay , Gene Expression , Humans , Mutagenesis, Site-Directed , Myocytes, Smooth Muscle/drug effects , Promoter Regions, Genetic/genetics , RNA, Messenger/analysis , Repressor Proteins/genetics , Repressor Proteins/metabolism , Stimulation, Chemical , Transcriptional Activation
7.
Am J Physiol Heart Circ Physiol ; 293(1): H770-6, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17468336

ABSTRACT

Nitroalkenes, the nitration products of unsaturated fatty acids formed via NO-dependent oxidative reactions, have been demonstrated to exert strong biological actions in endothelial cells and monocytes/macrophages; however, little is known about their effects on vascular smooth muscle cells (VSMCs). The present study examined the role of nitro-linoleic acid (LNO(2)) in the regulation of VSMC proliferation. We observed that LNO(2) inhibited VSMC proliferation in a dose-dependent manner. In addition, LNO(2) induced growth arrest of VSMCs in the G(1)/S phase of the cell cycle with an upregulation of the cyclin-dependent kinase inhibitor p27(kip1). Furthermore, LNO(2) triggered nuclear factor-erythroid 2-related factor 2 (Nrf2) nuclear translocation and activation of the antioxidant-responsive element-driven transcriptional activity via impairing Kelch-like ECH-associating protein 1 (Keap1)-mediated negative control of Nrf2 activity in VSMCs. LNO(2) upregulated the expression of Nrf2 protein levels, but not mRNA levels, in VSMCs. A forced activation of Nrf2 led to an upregulation of p27(kip1) and growth inhibition of VSMCs. In contrast, knock down of Nrf2 using an Nrf2 siRNA approach reversed the LNO(2)-induced upregulation of p27(kip1) and inhibition of cellular proliferation in VSMCs. These studies provide the first evidence that nitroalkene LNO(2) inhibits VSMC proliferation through activation of the Keap1/Nrf2 signaling pathway, suggesting an important role of nitroalkenes in vascular biology.


Subject(s)
Cell Proliferation/drug effects , Linoleic Acids/administration & dosage , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/physiology , NF-E2-Related Factor 2/metabolism , Nitro Compounds/administration & dosage , Proteins/metabolism , Signal Transduction/physiology , Animals , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Dose-Response Relationship, Drug , Intracellular Signaling Peptides and Proteins , Kelch-Like ECH-Associated Protein 1 , Male , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
8.
Stem Cells Dev ; 16(1): 25-9, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17348803

ABSTRACT

Human embryonic stem (hES) cells hold great therapeutic potential for cell transplantation. To date, it remains uncertain whether undifferentiated hES cells can differentiate into cardiac lineage in vivo during myocardial infarction. Here we provide the first report that undifferentiated hES cells can survive in rat hearts during myocardial infarction without the formation of teratoma using undifferentiated green fluorescent protein (GFP)-transgenic hES cells. Using a laser-capture microscope to dissect the GFP-positive cell area from the hES-injected hearts, we documented the expression of human cardiac-specific genes, including GATA-4, Nkx-2.5, and cardiac troponin I. Taken together, our results demonstrate that undifferentiated hES cells can be driven to the cardiac lineage under the local injured environment in the heart, which may provide a potential method for regenerating de novo myocardium to treat myocardial infarction.


Subject(s)
Embryonic Stem Cells/cytology , Myocardial Infarction/therapy , Stem Cell Transplantation , Transplantation, Heterologous , Animals , Cell Differentiation , Disease Models, Animal , GATA4 Transcription Factor/genetics , Green Fluorescent Proteins/genetics , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/genetics , Humans , Microscopy, Confocal , Myocardial Infarction/pathology , Myocardium/pathology , Myocytes, Cardiac/cytology , Rats , Transcription Factors/genetics , Transgenes , Troponin I/genetics
9.
Curr Atheroscler Rep ; 9(3): 230-7, 2007 Sep.
Article in English | MEDLINE | ID: mdl-18241618

ABSTRACT

Peroxisome proliferator-activated receptors (PPAR) and liver X receptors (LXR) regulate a plethora of biologic processes and key metabolic and physiologic events. Deregulation of their transcription and activity is commonly associated with dyslipidemic disorders, diabetes, cancer, and cardiovascular disease. This review addresses recent advances in our understanding of the molecular mechanisms regulating transcription of these nuclear receptors. The heterogeneity of factors regulating their transcription and activity suggests intricate regulatory networks that determine their tissue expression pattern and their responses to pharmacologic agents. Understanding such mechanisms will facilitate unraveling their protective effects in disease as well as the design of effective targeted therapies.


Subject(s)
DNA-Binding Proteins/physiology , Peroxisome Proliferator-Activated Receptors/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Transcription, Genetic/physiology , Adipose Tissue/physiopathology , Atherosclerosis/physiopathology , CCAAT-Enhancer-Binding Proteins/physiology , DNA-Binding Proteins/genetics , Down-Regulation/physiology , Forkhead Transcription Factors/physiology , GATA Transcription Factors/physiology , Humans , Liver X Receptors , Obesity/physiopathology , Orphan Nuclear Receptors , Oxidative Stress/physiology , Peroxisome Proliferator-Activated Receptors/genetics , Peroxisome Proliferator-Activated Receptors/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Signal Transduction/physiology , Sirtuins/physiology , Smad Proteins/physiology , Transforming Growth Factor beta/physiology , Up-Regulation/physiology , Wnt Proteins/physiology
10.
J Biol Chem ; 281(47): 35686-98, 2006 Nov 24.
Article in English | MEDLINE | ID: mdl-16887803

ABSTRACT

Nitroalkene derivatives of linoleic acid (LNO2) and oleic acid (OA-NO2) are present; however, their biological functions remain to be fully defined. Herein, we report that LNO2 and OA-NO2 inhibit lipopolysaccharide-induced secretion of proinflammatory cytokines in macrophages independent of nitric oxide formation, peroxisome proliferator-activated receptor-gamma activation, or induction of heme oxygenase-1 expression. The electrophilic nature of fatty acid nitroalkene derivatives resulted in alkylation of recombinant NF-kappaB p65 protein in vitro and a similar reaction with p65 in intact macrophages. The nitroalkylation of p65 by fatty acid nitroalkene derivatives inhibited DNA binding activity and repressed NF-kappaB-dependent target gene expression. Moreover, nitroalkenes inhibited endothelial tumor necrosis factor-alpha-induced vascular cell adhesion molecule 1 expression and monocyte rolling and adhesion. These observations indicate that nitroalkenes such as LNO2 and OA-NO2, derived from reactions of unsaturated fatty acids and oxides of nitrogen, are a class of endogenous anti-inflammatory mediators.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Fatty Acids/chemistry , Signal Transduction , Animals , Bone Marrow Cells/metabolism , Heme Oxygenase (Decyclizing)/genetics , Macrophages/metabolism , Mice , Mice, Knockout , NF-kappa B/chemistry , Nitrogen/chemistry , PPAR gamma/metabolism , Protein Processing, Post-Translational , Recombinant Proteins/chemistry
11.
Vascul Pharmacol ; 45(1): 65-71, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16750656

ABSTRACT

Given the heterogeneous nature of metabolic dysfunctions associated with insulin resistance and type 2 diabetes (T2D), a single pharmaceutical cannot be expected to provide complication-free therapy in all patients. Thiazolidinediones (TZD) increase insulin sensitivity, reduce blood glucose and improve cardiovascular parameters. However, in addition to increasing fat mass, TZD have the potential in certain individuals to exacerbate underlying hepatosteatosis and diabetic cardiomyopathy. Pharmacogenetics should allow patient selection to maximize therapy and minimize risk. To this end, we have combined two genetically diverse inbred strains, NON/Lt and NZO/Lt, to produce a "negative heterosis" increasing the frequency of T2D in F1 males. As in humans with T2D, treatment of diabetic and hyperlipemic F1 males with rosiglitazone (Rosi), an agonist of peroxisome proliferator-activated gamma receptor (PPARgamma), reverses these disease phenotypes. However, the hybrid genome perturbed both major pathways for phosphatidylcholine (PC) biosynthesis in the liver, and effected remarkable alterations in the composition of cardiolipin in heart mitochondria. These metabolic defects severely exacerbated an underlying hepatosteatosis and increased levels of the adipokine, plasminogen activator inhibitor-1 (PAI-1), a risk factor for cardiovascular events. This model system demonstrates how the power of mouse genetics can be used to identify the metabolic signatures of individuals who may be prone to drug side effects.


Subject(s)
Cardiovascular System/drug effects , Diabetes Mellitus, Type 2/metabolism , Hypoglycemic Agents/adverse effects , Liver/drug effects , Phosphatidylcholines/metabolism , Thiazolidinediones/adverse effects , Animals , Cardiolipins/metabolism , Cardiovascular System/metabolism , Choline-Phosphate Cytidylyltransferase/antagonists & inhibitors , Choline-Phosphate Cytidylyltransferase/metabolism , Crosses, Genetic , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/genetics , Disease Models, Animal , Fatty Liver/chemically induced , Fatty Liver/metabolism , Gene Expression Regulation/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred NOD , Mice, Obese , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , PPAR gamma/agonists , PPAR gamma/metabolism , Phosphatidylethanolamine N-Methyltransferase/antagonists & inhibitors , Phosphatidylethanolamine N-Methyltransferase/metabolism , Rosiglitazone
12.
Am J Physiol Heart Circ Physiol ; 291(6): H2752-7, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16731635

ABSTRACT

Recent epidemiological studies have indicated that baseline C-reactive protein (CRP) levels may have value in prediction of cardiovascular risk. Using six tag single-nucleotide polymorphisms (SNPs) selected from our complete list of SNPs on the CRP gene, we investigated the association of CRP genotypes with plasma CRP levels and cardiovascular risk in the National Heart, Lung, and Blood Institute (NHLBI) Family Heart Study cohort (1,296 Caucasians, 48.5% male, 54.7 +/- 12.8 yr old). There was a significant trend toward association of CRP haplotypes with CRP levels (P = 0.045). SNP analysis indicated a highly significant association of SNP -757 (rs3093059, P = 0.0004) and SNP -286 (rs3091244, P = 0.0065) and a borderline association of SNP -7180 (rs1341665, P = 0.06) with CRP levels. Neither CRP haplotypes nor individual SNP genotypes were associated with intima-media thickness of the common carotid or internal carotid artery or the bifurcation of the carotid arteries. These results indicated a strong impact of local SNPs of the CRP gene on plasma CRP levels, but there was no direct evidence that these genetically controlled CRP elevations by local CRP SNPs contributed to cardiovascular disease phenotypes.


Subject(s)
C-Reactive Protein/genetics , Cardiovascular Diseases/blood , Cardiovascular Diseases/genetics , Genetic Predisposition to Disease/genetics , Polymorphism, Single Nucleotide , Adult , Aged , C-Reactive Protein/analysis , Cardiovascular Diseases/physiopathology , Cohort Studies , Cross-Sectional Studies , Female , Gene Expression Regulation , Genotype , Haplotypes/genetics , Humans , Male , Middle Aged , Risk Factors , United States
14.
J Biol Chem ; 280(51): 42464-75, 2005 Dec 23.
Article in English | MEDLINE | ID: mdl-16227625

ABSTRACT

Mass spectrometric analysis of human plasma and urine revealed abundant nitrated derivatives of all principal unsaturated fatty acids. Nitrated palmitoleic, oleic, linoleic, linolenic, arachidonic and eicosapentaenoic acids were detected in concert with their nitrohydroxy derivatives. Two nitroalkene derivatives of the most prevalent fatty acid, oleic acid, were synthesized (9- and 10-nitro-9-cis-octadecenoic acid; OA-NO2), structurally characterized and determined to be identical to OA-NO2 found in plasma, red cells, and urine of healthy humans. These regioisomers of OA-NO2 were quantified in clinical samples using 13C isotope dilution. Plasma free and esterified OA-NO2 concentrations were 619 +/- 52 and 302 +/- 369 nm, respectively, and packed red blood cell free and esterified OA-NO2 was 59 +/- 11 and 155 +/- 65 nm. The OA-NO2 concentration of blood is approximately 50% greater than that of nitrated linoleic acid, with the combined free and esterified blood levels of these two fatty acid derivatives exceeding 1 microm. OA-NO2 is a potent ligand for peroxisome proliferator activated receptors at physiological concentrations. CV-1 cells co-transfected with the luciferase gene under peroxisome proliferator-activated receptor (PPAR) response element regulation, in concert with PPARgamma, PPARalpha, or PPARdelta expression plasmids, showed dose-dependent activation of all PPARs by OA-NO2. PPARgamma showed the greatest response, with significant activation at 100 nm, while PPARalpha and PPARdelta were activated at approximately 300 nm OA-NO2. OA-NO2 also induced PPAR gamma-dependent adipogenesis and deoxyglucose uptake in 3T3-L1 preadipocytes at a potency exceeding nitrolinoleic acid and rivaling synthetic thiazo-lidinediones. These data reveal that nitrated fatty acids comprise a class of nitric oxide-derived, receptor-dependent, cell signaling mediators that act within physiological concentration ranges.


Subject(s)
Fatty Acids, Unsaturated/metabolism , Nitric Oxide/metabolism , Peroxisome Proliferator-Activated Receptors/metabolism , Signal Transduction , 3T3-L1 Cells , Animals , Fatty Acids, Unsaturated/blood , Fatty Acids, Unsaturated/urine , Humans , Ligands , Mice , Nuclear Magnetic Resonance, Biomolecular , Peroxisome Proliferator-Activated Receptors/genetics , Spectrometry, Mass, Electrospray Ionization , Transfection
15.
Am J Pathol ; 167(4): 1139-48, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16192648

ABSTRACT

C-reactive protein (CRP) is frequently deposited in the lesions of the arterial intima; however, the origin and pathological significance of CRP in these lesions are not completely understood. In this study, we measured CRP levels in the plasma of hypercholesterolemic rabbits and investigated CRP expression at both the mRNA and protein levels using rabbit and human atherosclerotic specimens. CRP levels were significantly elevated in both cholesterol-fed and Watanabe heritable hyperlipidemic rabbits, and CRP levels were clearly correlated with aortic atherosclerotic lesion size. Immunohistochemical staining coupled with Western blotting analysis revealed that CRP-immunoreactive proteins were found at all stages of atherosclerosis from the early to advanced lesions. CRP was present extracellularly and co-localized with apolipoprotein B but was rarely associated with the cytoplasm of macrophages and foam cells. Real-time reverse transcriptase-polymerase chain reaction analysis revealed that CRP mRNA in atherosclerotic lesions was barely detectable, and isolated macrophages did not express CRP mRNA, suggesting that CRP proteins found in the lesions were essentially derived from the circulation rather than synthesized de novo by vascular cells. These results suggest that there is a link between plasma CRP and the degree of atherosclerosis and that inhibition of plasma CRP may represent a therapeutic modality for the treatment of cardiovascular disease.


Subject(s)
Arteriosclerosis/metabolism , C-Reactive Protein/metabolism , Animals , Antibodies, Monoclonal/metabolism , Aorta/chemistry , Aorta/metabolism , Aorta/pathology , Arteriosclerosis/chemically induced , Arteriosclerosis/etiology , Arteriosclerosis/pathology , Blotting, Western , C-Reactive Protein/genetics , Cholesterol, Dietary/toxicity , Coronary Vessels/chemistry , Coronary Vessels/metabolism , Coronary Vessels/pathology , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rabbits , Reverse Transcriptase Polymerase Chain Reaction , Specific Pathogen-Free Organisms
16.
Stem Cells Dev ; 14(4): 367-77, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16137225

ABSTRACT

Human embryonic stem (hES) cells present a valuable source of cells with a vast therapeutic potential. However, the low efficiency of directed differentiation of hES cells remains a major obstacle in their uses for regenerative medicine. While differentiation may be controlled by the genetic manipulation, effective and efficient gene transfer into hES cells has been an elusive goal. Here, we show stable and efficient genetic manipulations of hES cells using lentiviral vectors. This method resulted in the establishment of stable gene expression without loss of pluripotency in hES cells. In addition, lentiviral vectors were effective in conveying the expression of an U6 promoter-driven small interfering RNA (siRNA), which was effective in silencing its specific target. Taken together, our results suggest that lentiviral gene delivery holds great promise for hES cell research and application.


Subject(s)
Embryo, Mammalian/cytology , Genetic Engineering/methods , Genetic Therapy/methods , Lentivirus/genetics , Stem Cells/cytology , Cell Differentiation , Cell Separation , Flow Cytometry , Genetic Vectors , Green Fluorescent Proteins/metabolism , Humans , Immunohistochemistry , Microscopy, Fluorescence , Models, Genetic , Myocytes, Cardiac/cytology , RNA, Small Interfering/metabolism , Regeneration , Reverse Transcriptase Polymerase Chain Reaction , Teratoma/metabolism , Teratoma/pathology , Transfection , Transgenes
17.
Life Sci ; 77(24): 3037-48, 2005 Oct 28.
Article in English | MEDLINE | ID: mdl-15970297

ABSTRACT

Peroxisome proliferator-activated receptor gamma (PPAR gamma), a member of the nuclear receptor family, has been implicated in the regulation of vascular smooth muscle cell (VSMC) growth; however, the underlying mechanisms are still not fully understood. We hypothesized that PPAR gamma functional deficiency may contribute to the enhanced proliferation of VSMC associated with hypertension in spontaneously hypertensive rats (SHR). We observed that PPAR gamma mRNA level in SHR VSMC was 3 approximately 4 fold higher than that from Wistar-Kyoto rats (WKY), but the protein expression levels of PPAR gamma are significantly lower in SHR than WKY VSMC, suggesting an impaired control of PPAR gamma protein expression in SHR VSMC. The deficiency of PPAR gamma protein expression in SHR VSMC was demonstrated by PPAR gamma reporter gene assays. Furthermore, the exaggerated growth of SHR VSMC was markedly attenuated by adenoviral PPAR gamma overexpression. Taken together, our results provided the first direct evidence that impaired expression of PPAR gamma protein contributes to the exaggerated growth of SHR VSMC.


Subject(s)
Arteries/metabolism , Cell Proliferation , Hypertension/metabolism , Muscle, Smooth, Vascular/metabolism , PPAR gamma/metabolism , Adenoviridae/genetics , Animals , Aorta/metabolism , Blood Pressure , Blotting, Northern , Blotting, Western , Cells, Cultured , Genes, Reporter , Hypertension/genetics , Hypertension/physiopathology , Immunoenzyme Techniques , Male , Muscle, Smooth, Vascular/cytology , PPAR gamma/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Transfection
18.
Circulation ; 111(8): 1071-7, 2005 Mar 01.
Article in English | MEDLINE | ID: mdl-15710763

ABSTRACT

BACKGROUND: Rad (Ras associated with diabetes) GTPase is a prototypic member of a new subfamily of Ras-related GTPases with unique structural features, although its physiological role remains largely unknown. In the present study, we characterized the Rad function in vascular smooth muscle cells (VSMCs) and the influence of adenovirus-mediated Rad (Ad-Rad) gene delivery on vascular remodeling after experimental angioplasty. METHODS AND RESULTS: We documented for the first time that neointimal formation using balloon-injured rat carotid arteries was associated with a significant increase in Rad expression as determined by immunohistochemistry and quantitative real-time reverse-transcriptase polymerase chain reaction. The levels of Rad expression in VSMCs were highly induced by platelet-derived growth factor and tumor necrosis factor-alpha. Morphometric analyses 14 days after injury revealed significantly diminished neointimal formation in the Ad-Rad-treated carotid arteries compared with Ad-GFP or PBS controls, whereas the mutated form of Rad GTPase, which can bind GDP but not GTP, increased neointimal formation. Overexpression of Rad significantly inhibited the attachment and migration of VSMCs. In addition, Rad expression dramatically reduced the formation of focal contacts and stress fibers in VSMCs by blocking the Rho/ROK signaling pathway. CONCLUSIONS: Our data clearly identified Rad GTPase as a novel and critical mediator that inhibits vascular lesion formation. Manipulation of the Rad signaling pathway may provide new therapeutic approaches that will limit vascular pathological remodeling.


Subject(s)
Cell Movement/physiology , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/physiology , Neovascularization, Pathologic/pathology , ras Proteins/physiology , Actins/antagonists & inhibitors , Animals , Aorta/cytology , Aorta/embryology , Carotid Arteries , Focal Adhesions/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Male , Muscle, Smooth, Vascular/enzymology , Neovascularization, Pathologic/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Stress Fibers/metabolism , Tunica Intima/metabolism , Tunica Intima/pathology , ras Proteins/biosynthesis , rho GTP-Binding Proteins/antagonists & inhibitors , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases
19.
Proc Natl Acad Sci U S A ; 102(7): 2340-5, 2005 Feb 15.
Article in English | MEDLINE | ID: mdl-15701701

ABSTRACT

Nitroalkene derivatives of linoleic acid (nitrolinoleic acid, LNO2) are formed via nitric oxide-dependent oxidative inflammatory reactions and are found at concentrations of approximately 500 nM in the blood of healthy individuals. We report that LNO2 is a potent endogenous ligand for peroxisome proliferator-activated receptor gamma (PPARgamma; Ki approximately 133 nM) that acts within physiological concentration ranges. This nuclear hormone receptor (PPARgamma) regulates glucose homeostasis, lipid metabolism, and inflammation. PPARgamma ligand activity is specific for LNO2)and not mediated by LNO2 decay products, NO donors, linoleic acid (LA), or oxidized LA. LNO2 is a significantly more robust PPARgamma ligand than other reported endogenous PPARgamma ligands, including lysophosphatidic acid (16:0 and 18:1), 15-deoxy-Delta12,14-PGJ2, conjugated LA and azelaoyl-phosphocholine. LNO2 activation of PPARgamma via CV-1 cell luciferase reporter gene expression analysis revealed a ligand activity that rivals or exceeds synthetic PPARgamma agonists such as rosiglitazone and ciglitazone, is coactivated by 9 cis-retinoic acid and is inhibited by the PPARgamma antagonist GW9662. LNO2 induces PPARgamma-dependent macrophage CD-36 expression, adipocyte differentiation, and glucose uptake also at a potency rivaling thiazolidinediones. These observations reveal that NO-mediated cell signaling reactions can be transduced by fatty acid nitration products and PPAR-dependent gene expression.


Subject(s)
Linoleic Acids/metabolism , PPAR gamma/metabolism , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/metabolism , Animals , Binding, Competitive , Cell Differentiation , Cell Line , Humans , Ligands , Linoleic Acids/chemistry , Mice , PPAR gamma/agonists , PPAR gamma/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction
20.
Stem Cells Dev ; 14(6): 671-5, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16433622

ABSTRACT

Human embryonic stem (hES) cells are undifferentiated and pluripotent cells that hold great therapeutic potential, but are hampered by our limited knowledge to promote specific cell differentiation. Here we provide the first report of the directed differentiation of hES cells into adipocytes. Embryoid bodies (EBs) derived from hES cells are shown to respond to factors that promote adipogenesis. Differentiated cells were observed that displayed the key features of adipocytes, i.e., expression of specific molecular markers, such as peroxisome proliferator-activated receptor gamma2 (PPARgamma2), adipocyte fatty acid binding protein (aP2) and adiponectin, the secretion of leptin, and the accumulation of lipid droplets in cytoplasm. Taken together, our results demonstrate that adipocytes derived from hES cells in vitro can provide a novel model system to study human adipogenesis and obesity.


Subject(s)
Adipocytes/physiology , Adipogenesis , Embryo, Mammalian/cytology , Stem Cells/physiology , Biomarkers , Cell Differentiation/drug effects , Cells, Cultured , Culture Media , Embryonic Development , Humans , Rosiglitazone , Thiazolidinediones/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...