Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Endocrinology ; 162(8)2021 08 01.
Article in English | MEDLINE | ID: mdl-34037749

ABSTRACT

Endothelial insulin receptors (Insr) promote sprouting angiogenesis, although the underpinning cellular and molecular mechanisms are unknown. Comparing mice with whole-body insulin receptor haploinsufficiency (Insr+/-) against littermate controls, we found impaired limb perfusion and muscle capillary density after inducing hind-limb ischemia; this was in spite of increased expression of the proangiogenic growth factor Vegfa. Insr+/- neonatal retinas exhibited reduced tip cell number and branching complexity during developmental angiogenesis, which was also found in separate studies of mice with endothelium-restricted Insr haploinsufficiency. Functional responses to vascular endothelial growth factor A (VEGF-A), including in vitro angiogenesis, were also impaired in aortic rings and pulmonary endothelial cells from Insr+/- mice. Human umbilical vein endothelial cells with shRNA-mediated knockdown of Insr also demonstrated impaired functional angiogenic responses to VEGF-A. VEGF-A signaling to Akt and endothelial nitric oxide synthase was intact, but downstream signaling to extracellular signal-reduced kinase 1/2 (ERK1/2) was impaired, as was VEGF receptor-2 (VEGFR-2) internalization, which is required specifically for signaling to ERK1/2. Hence, endothelial insulin receptors facilitate the functional response to VEGF-A during angiogenic sprouting and are required for appropriate signal transduction from VEGFR-2 to ERK1/2.


Subject(s)
Endothelium, Vascular/metabolism , MAP Kinase Signaling System , Neovascularization, Physiologic , Receptor, Insulin/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic , Vascular Endothelial Growth Factor Receptor-2/metabolism
2.
Sci Rep ; 9(1): 16876, 2019 11 14.
Article in English | MEDLINE | ID: mdl-31727906

ABSTRACT

Glucose and hypotonicity induced cell swelling stimulate insulin release from pancreatic ß-cells but the mechanisms are poorly understood. Recently, Piezo1 was identified as a mechanically-activated nonselective Ca2+ permeable cationic channel in a range of mammalian cells. As cell swelling induced insulin release could be through stimulation of Ca2+ permeable stretch activated channels, we hypothesised a role for Piezo1 in cell swelling induced insulin release. Two rat ß-cell lines (INS-1 and BRIN-BD11) and freshly-isolated mouse pancreatic islets were studied. Intracellular Ca2+ measurements were performed using the fura-2 Ca2+ indicator dye and ionic current was recorded by whole cell patch-clamp. Piezo1 agonist Yoda1, a competitive antagonist of Yoda1 (Dooku1) and an inactive analogue of Yoda1 (2e) were used as chemical probes. Piezo1 mRNA and insulin secretion were measured by RT-PCR and ELISA respectively. Piezo1 mRNA was detected in both ß-cell lines and mouse islets. Yoda1 evoked Ca2+ entry was inhibited by Yoda1 antagonist Dooku1 as well as other Piezo1 inhibitors gadolinium and ruthenium red, and not mimicked by 2e. Yoda1, but not 2e, stimulated Dooku1-sensitive insulin release from ß-cells and pancreatic islets. Hypotonicity and high glucose increased intracellular Ca2+ and enhanced Yoda1 Ca2+ influx responses. Yoda1 and hypotonicity induced insulin release were significantly inhibited by Piezo1 specific siRNA. Pancreatic islets from mice with haploinsufficiency of Piezo1 released less insulin upon exposure to Yoda1. The data show that Piezo1 channel agonist induces insulin release from ß-cell lines and mouse pancreatic islets suggesting a role for Piezo1 in cell swelling induced insulin release. Hence Piezo1 agonists have the potential to be used as enhancers of insulin release.


Subject(s)
Calcium/metabolism , Glucose/pharmacology , Insulin-Secreting Cells/drug effects , Insulin/metabolism , Ion Channels/genetics , Membrane Proteins/genetics , Animals , Biological Transport/drug effects , Cell Line, Tumor , Gadolinium/pharmacology , Gene Expression Regulation , Glucose/metabolism , Heterozygote , Insulin Secretion/genetics , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Ion Channels/antagonists & inhibitors , Ion Channels/metabolism , Mechanotransduction, Cellular , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Pyrazines/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Ruthenium Red/pharmacology , Thiadiazoles/pharmacology , Tissue Culture Techniques
3.
Diab Vasc Dis Res ; 16(2): 160-170, 2019 03.
Article in English | MEDLINE | ID: mdl-30295509

ABSTRACT

Insulin and insulin-like growth factor-1 stimulate specific responses in arteries, which may be disrupted by diet-induced obesity. We examined (1) temporal effects of high-fat diet compared to low-fat diet in mice on insulin receptor, insulin-like growth factor-1 receptor, insulin receptor/insulin-like growth factor-1 receptor hybrid receptor expression and insulin/insulin-like growth factor-1-mediated Akt phosphorylation in aorta; and (2) effects of high-fat diet on insulin and insulin-like growth factor-1-mediated Akt phosphorylation and vascular tone in resistance arteries. Medium-term high-fat diet (5 weeks) decreased insulin-like growth factor-1 receptor expression and increased hybrid expression (~30%) only. After long-term (16 weeks) high-fat diet, insulin receptor expression was reduced by ~30%, insulin-like growth factor-1 receptor expression decreased a further ~40% and hybrid expression increased a further ~60%. Independent correlates of hybrid receptor expression were high-fat diet, duration of high-fat diet and plasma insulin-like growth factor-1 (all p < 0.05). In aorta, insulin was a more potent activator of Akt than insulin-like growth factor-1, whereas in resistance arteries, insulin-like growth factor-1 was more potent than insulin. High-fat diet blunted insulin-mediated vasorelaxation ( p < 0.01) but had no effect on insulin-like growth factor-1-mediated vasorelaxation in resistance arteries. Our findings support the possibility that hybrid receptor level is influenced by nutritional and metabolic cues. Moreover, vessel-dependent effects of insulin and insulin-like growth factor-1 on vascular tone and Akt activation may have implications in treating obesity-related vascular disease.


Subject(s)
Aorta/drug effects , Insulin/pharmacology , Mesenteric Arteries/drug effects , Obesity/enzymology , Proto-Oncogene Proteins c-akt/metabolism , Receptor, IGF Type 1/metabolism , Vascular Resistance/drug effects , Animals , Antigens, CD/metabolism , Aorta/enzymology , Cells, Cultured , Diet, Fat-Restricted , Diet, High-Fat , Disease Models, Animal , Enzyme Activation , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Insulin-Like Growth Factor I/pharmacology , Male , Mesenteric Arteries/enzymology , Mesenteric Arteries/physiopathology , Mice, Inbred C57BL , Obesity/blood , Obesity/physiopathology , Phosphorylation , Receptor, IGF Type 1/genetics , Receptor, Insulin/metabolism , Receptors, Somatomedin/metabolism , Signal Transduction/drug effects , Vasodilation/drug effects
4.
Endocrinology ; 159(8): 2917-2925, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29796592

ABSTRACT

Reduced systemic insulin signaling promotes endothelial dysfunction and diminished endogenous vascular repair. We investigated whether restoration of endothelial insulin receptor expression could rescue this phenotype. Insulin receptor knockout (IRKO) mice were crossed with mice expressing a human insulin receptor endothelial cell-specific overexpression (hIRECO) to produce IRKO-hIRECO progeny. No metabolic differences were noted between IRKO and IRKO-hIRECO mice in glucose and insulin tolerance tests. In contrast with control IRKO littermates, IRKO-hIRECO mice exhibited normal blood pressure and aortic vasodilatation in response to acetylcholine, comparable to parameters noted in wild type littermates. These phenotypic changes were associated with increased basal- and insulin-stimulated nitric oxide production. IRKO-hIRECO mice also demonstrated normalized endothelial repair after denuding arterial injury, which was associated with rescued endothelial cell migration in vitro but not with changes in circulating progenitor populations or culture-derived myeloid angiogenic cells. These data show that restoration of endothelial insulin receptor expression alone is sufficient to prevent the vascular dysfunction caused by systemically reduced insulin signaling.


Subject(s)
Aorta/metabolism , Blood Glucose/metabolism , Endothelium, Vascular/metabolism , Haploinsufficiency/genetics , Receptor, Insulin/genetics , Vasodilation/genetics , Acetylcholine/pharmacology , Animals , Antigens, CD/genetics , Aorta/physiopathology , Blood Pressure , Cell Movement , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Glucose Tolerance Test , Humans , In Vitro Techniques , Male , Mice , Mice, Knockout , Mice, Transgenic , Nitric Oxide/metabolism , Receptor, Insulin/metabolism , Signal Transduction , Vasodilation/drug effects , Vasodilator Agents/pharmacology
5.
Endocrinology ; 159(2): 696-709, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29186427

ABSTRACT

Insulin resistance is associated with impaired endothelial regeneration in response to mechanical injury. We recently demonstrated that insulinlike growth factor-binding protein-1 (IGFBP1) ameliorated insulin resistance and increased nitric oxide generation in the endothelium. In this study, we hypothesized that IGFBP1 would improve endothelial regeneration and restore endothelial reparative functions in the setting of insulin resistance. In male mice heterozygous for deletion of insulin receptors, endothelial regeneration after femoral artery wire injury was enhanced by transgenic expression of human IGFBP1 (hIGFBP1). This was not explained by altered abundance of circulating myeloid angiogenic cells. Incubation of human endothelial cells with hIGFBP1 increased integrin expression and enhanced their ability to adhere to and repopulate denuded human saphenous vein ex vivo. In vitro, induction of insulin resistance by tumor necrosis factor α (TNFα) significantly inhibited endothelial cell migration and proliferation. Coincubation with hIGFBP1 restored endothelial migratory and proliferative capacity. At the molecular level, hIGFBP1 induced phosphorylation of focal adhesion kinase, activated RhoA and modulated TNFα-induced actin fiber anisotropy. Collectively, the effects of hIGFBP1 on endothelial cell responses and acceleration of endothelial regeneration in mice indicate that manipulating IGFBP1 could be exploited as a putative strategy to improve endothelial repair in the setting of insulin resistance.


Subject(s)
Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Insulin Resistance , Insulin-Like Growth Factor Binding Protein 1/metabolism , Animals , Cell Movement , Endothelial Cells/cytology , Female , Focal Adhesion Protein-Tyrosine Kinases/genetics , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Insulin-Like Growth Factor Binding Protein 1/genetics , Integrins/genetics , Integrins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
6.
Arterioscler Thromb Vasc Biol ; 34(9): 2051-8, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25012136

ABSTRACT

OBJECTIVES: Defective endothelial regeneration predisposes to adverse arterial remodeling and is thought to contribute to cardiovascular disease in type 2 diabetes mellitus. We recently demonstrated that the type 1 insulin-like growth factor receptor (IGF1R) is a negative regulator of insulin sensitivity and nitric oxide bioavailability. In this report, we examined partial deletion of the IGF1R as a potential strategy to enhance endothelial repair. APPROACH AND RESULTS: We assessed endothelial regeneration after wire injury in mice and abundance and function of angiogenic progenitor cells in mice with haploinsufficiency of the IGF1R (IGF1R(+/-)). Endothelial regeneration after arterial injury was accelerated in IGF1R(+/-) mice. Although the yield of angiogenic progenitor cells was lower in IGF1R(+/-) mice, these angiogenic progenitor cells displayed enhanced adhesion, increased secretion of insulin-like growth factor-1, and enhanced angiogenic capacity. To examine the relevance of IGF1R manipulation to cell-based therapy, we transfused IGF1R(+/-) bone marrow-derived CD117(+) cells into wild-type mice. IGF1R(+/-) cells accelerated endothelial regeneration after arterial injury compared with wild-type cells and did not alter atherosclerotic lesion formation. CONCLUSIONS: Haploinsufficiency of the IGF1R is associated with accelerated endothelial regeneration in vivo and enhanced tube forming and adhesive potential of angiogenic progenitor cells in vitro. Partial deletion of IGF1R in transfused bone marrow-derived CD117(+) cells enhanced their capacity to promote endothelial regeneration without altering atherosclerosis. Our data suggest that manipulation of the IGF1R could be exploited as novel therapeutic approach to enhance repair of the arterial wall after injury.


Subject(s)
Carotid Artery Diseases/prevention & control , Endothelium, Vascular/physiology , Femoral Artery/injuries , Hematopoietic Stem Cells/physiology , Neovascularization, Physiologic/physiology , Receptor, IGF Type 1/physiology , Animals , Aorta, Thoracic/pathology , Apolipoproteins E/deficiency , Carotid Artery Diseases/etiology , Carotid Artery Diseases/genetics , Cell Adhesion , Endothelium, Vascular/metabolism , Female , Gene Expression Regulation , Genotype , Hematopoietic Stem Cell Transplantation , Male , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type III/metabolism , Phenotype , Phosphorylation , Protein Processing, Post-Translational , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor, IGF Type 1/deficiency , Receptor, IGF Type 1/genetics , Regeneration
7.
Stem Cells ; 32(10): 2714-23, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24916783

ABSTRACT

Recent data suggest reduced indices of vascular repair in South Asian men, a group at increased risk of cardiovascular events. Outgrowth endothelial cells (OEC) represent an attractive tool to study vascular repair in humans and may offer potential in cell-based repair therapies. We aimed to define and manipulate potential mechanisms of impaired vascular repair in South Asian (SA) men. In vitro and in vivo assays of vascular repair and angiogenesis were performed using OEC derived from SA men and matched European controls, prior defining potentially causal molecular mechanisms. SA OEC exhibited impaired colony formation, migration, and in vitro angiogenesis, associated with decreased expression of the proangiogenic molecules Akt1 and endothelial nitric oxide synthase (eNOS). Transfusion of European OEC into immunodeficient mice after wire-induced femoral artery injury augmented re-endothelialization, in contrast with SA OEC and vehicle; SA OEC also failed to promote angiogenesis after induction of hind limb ischemia. Expression of constitutively active Akt1 (E17KAkt), but not green fluorescent protein control, in SA OEC increased in vitro angiogenesis, which was abrogated by a NOS antagonist. Moreover, E17KAkt expressing SA OEC promoted re-endothelialization of wire-injured femoral arteries, and perfusion recovery of ischemic limbs, to a magnitude comparable with nonmanipulated European OEC. Silencing Akt1 in European OEC recapitulated the functional deficits noted in SA OEC. Reduced signaling via the Akt/eNOS axis is causally linked with impaired OEC-mediated vascular repair in South Asian men. These data prove the principle of rescuing marked reparative dysfunction in OEC derived from these men.


Subject(s)
Blood Vessels/pathology , Endothelial Cells/cytology , Endothelial Cells/enzymology , Proto-Oncogene Proteins c-akt/metabolism , Wound Healing , Adult , Animals , Asia , Demography , Endothelial Cells/drug effects , Gene Silencing , Humans , Insulin/pharmacology , Male , Mice, Nude , Phosphorylation/drug effects , Risk Factors , White People , Wound Healing/drug effects
8.
Diabetes ; 62(6): 1981-9, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23382450

ABSTRACT

Circadian rhythms are integral to the normal functioning of numerous physiological processes. Evidence from human and mouse studies suggests that loss of rhythm occurs in obesity and cardiovascular disease and may be a neglected contributor to pathophysiology. Obesity has been shown to impair the circadian clock mechanism in liver and adipose tissue but its effect on cardiovascular tissues is unknown. We investigated the effect of diet-induced obesity in C57BL6J mice upon rhythmic transcription of clock genes and diurnal variation in vascular and metabolic systems. In obesity, clock gene function and physiological rhythms were preserved in the vasculature but clock gene transcription in metabolic tissues and rhythms of glucose tolerance and insulin sensitivity were blunted. The most pronounced attenuation of clock rhythm occurred in adipose tissue, where there was also impairment of clock-controlled master metabolic genes and both AMPK mRNA and protein. Across tissues, clock gene disruption was associated with local inflammation but diverged from impairment of insulin signaling. We conclude that vascular tissues are less sensitive to pathological disruption of diurnal rhythms during obesity than metabolic tissues and suggest that cellular disruption of clock gene rhythmicity may occur by mechanisms shared with inflammation but distinct from those leading to insulin resistance.


Subject(s)
Circadian Rhythm/physiology , Insulin Resistance/physiology , Obesity/metabolism , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Circadian Rhythm/genetics , Diet, High-Fat/adverse effects , Immunoblotting , Male , Mice , Obesity/etiology , Obesity/immunology , Polymerase Chain Reaction
9.
Biochem Biophys Res Commun ; 420(4): 828-33, 2012 Apr 20.
Article in English | MEDLINE | ID: mdl-22465119

ABSTRACT

Coronary heart disease (CHD) is a condition characterized by increased levels of proinflammatory cytokines, including tumor necrosis factor-α (TNF-α). TNF-α can induce vascular endothelial cell (EC) and smooth muscle cell (SMC) dysfunction, central events in development of neointimal lesions. The reduced incidence of CHD in young women is believed to be due to the protective effects of estradiol (E2). We therefore investigated the effects of TNF-α on human neointima formation and SMC/EC functions and any modulatory effects of E2. Saphenous vein (SV) segments were cultured in the presence of TNF-α (10 ng/ml), E2 (2.5 nM) or both in combination. Neointimal thickening was augmented by incubation with TNF-α, an effect that was abolished by co-culture with E2. TNF-α increased SV-SMC proliferation in a concentration-dependent manner that was optimal at 10 ng/ml (1.5-fold increase), and abolished by E2 at all concentrations studied (1-50 nM). Surprisingly, E2 itself at low concentrations (1 and 5 nM) stimulated SV-SMC proliferation to a level comparable to that of TNF-α alone. SV-EC migration was significantly impaired by TNF-α (42% of control), and co-culture with E2 partially restored the ability of SV-EC to migrate and repair the wound. In contrast, TNF-α increased SV-SMC migration by 1.7-fold, an effect that was completely reversed by co-incubation with E2. Finally, TNF-α potently induced ICAM-1 and VCAM-1 expression in both SV-EC and SV-SMC. However there was no modulation by E2 in either cell-type. In conclusion, TNF-α induced SV neointima formation, increased SMC proliferation and migration, impaired SV-EC migration and increased expression of adhesion molecules. E2 exerted distinct cell-type and function-specific modulation, the mechanisms underlying which are worthy of further detailed study.


Subject(s)
Endothelium, Vascular/drug effects , Estradiol/pharmacology , Muscle, Smooth, Vascular/drug effects , Neointima/pathology , Tumor Necrosis Factor-alpha/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Endothelium, Vascular/pathology , Female , Humans , Intercellular Adhesion Molecule-1/biosynthesis , Muscle, Smooth, Vascular/pathology , Vascular Cell Adhesion Molecule-1/biosynthesis
10.
Clin Exp Pharmacol Physiol ; 36(5-6): 478-86, 2009 May.
Article in English | MEDLINE | ID: mdl-19673929

ABSTRACT

1. Thiazolidinediones (TZDs) are peroxisome proliferator-activated receptor (PPAR) gamma agonists that are used to lower insulin resistance in Type 2 diabetic patients. Although TZDs exhibit beneficial effects on the vasculature, their effects on the heart are less clear and are the subject of current clinical debate. Thiazolidinediones have been reported to reduce adverse myocardial remodelling, a pathology in which cardiac myofibroblasts (CMF) are pivotal. 2. The aim of the present study was to investigate whether TZDs modulate specific human CMF functions of importance to the myocardial remodelling process and to determine whether any of these effects were mediated via PPARgamma activation. 3. Immunoblotting of cultured human CMF homogenates revealed strong expression of PPARgamma (approximately 50 kDa). Three different TZDs (ciglitazone, rosiglitazone and troglitazone) and the endogenous PPARgamma ligand 15-deoxy-delta(12,14)-prostaglandin J(2) (15d-PGJ(2)) inhibited CMF proliferation (cell number and expression of proliferating cell nuclear antigen) in a concentration-dependent manner (range 0.1-10 micromol/L) with similar potencies. This antiproliferative effect of TZDs was not reversed by the PPARgamma antagonists GW9662 or T0070907 (10-25 micromol/L). None of the TZDs or 15d-PGJ(2) affected cell migration or invasion (Boyden chamber assays without or with Matrigel barrier), matrix metalloproteinase-2 or -9 secretion (gelatin zymography) or the actin cytoskeleton (rhodamine/phalloidin fluorescent confocal microscopy). 4. In conclusion, TZDs reduce human CMF proliferation via a PPARgamma-independent mechanism. Although TZDs do not inhibit CMF invasion, their antiproliferative activity may contribute to the ability of this class of drugs to modulate adverse myocardial remodelling.


Subject(s)
Myocytes, Cardiac/drug effects , PPAR gamma/physiology , Thiazolidinediones/pharmacology , 3T3-L1 Cells , Animals , Cell Adhesion/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Cytoskeleton/physiology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/physiology , Humans , Hypoglycemic Agents/pharmacology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , PPAR gamma/agonists , PPAR gamma/metabolism , Signal Transduction/drug effects
11.
Cardiovasc Res ; 76(1): 81-90, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17612514

ABSTRACT

OBJECTIVE: In addition to direct effects on myocardial cell function, tumor necrosis factor alpha (TNFalpha) contributes to adverse cardiac remodeling by increasing production of other pro-inflammatory cytokines [e.g. interleukin (IL)-1 and IL-6]. Both statins and thiazolidinediones (TZDs) have beneficial effects on cardiac remodeling, possibly due to their anti-inflammatory properties. The present study examined the mechanisms by which TNFalpha stimulates expression of pro-inflammatory cytokines in cultured human cardiac fibroblasts and determined the effects of statin or TZD treatment. METHODS: Human cardiac fibroblasts were cultured from biopsies of right atrial appendages. Cytokine mRNA expression and secretion was measured using quantitative real-time RT-PCR and ELISA. Activation of signaling pathways was determined by immunoblotting with phospho-specific antibodies. RESULTS: TNFalpha (0.1-10 ng/ml) stimulated IL-6, IL-1alpha and IL-1beta mRNA expression in cardiac fibroblasts in a concentration-dependent manner. Pharmacological inhibitors and receptor-neutralizing antibodies established that both TNFalpha-induced IL-6 and IL-1beta expression was mediated via the TNFRI receptor and p38 mitogen-activated protein kinase (MAPK), phosphoinositide 3-kinase (PI3K)/Akt and nuclear factor (NF)-kappaB pathways. In contrast, TNFalpha-induced IL-1alpha expression required both TNFRI and TNFRII subtypes and p38 MAPK and PI3K/Akt pathways, but was negatively regulated by the NF-kappaB pathway. Neither statins (simvastatin, fluvastatin) nor TZDs (ciglitazone, rosiglitazone, troglitazone) had inhibitory effects on TNFalpha-induced IL-6 secretion or IL-1alpha/beta mRNA expression; indeed, cytokine expression was increased in response to TZDs. CONCLUSIONS: Our data provide important insights into the regulation of pro-inflammatory cytokine expression in human cardiac fibroblasts and suggest that the myocardial anti-inflammatory effects of statins and TZDs are not due to inhibition of TNFalpha-induced IL-1 or IL-6 expression by cardiac fibroblasts.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Interleukins/biosynthesis , Myocytes, Cardiac/immunology , Signal Transduction/drug effects , Thiazolidinediones/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Analysis of Variance , Antibodies, Monoclonal/pharmacology , Cells, Cultured , Chromans/pharmacology , Coronary Disease/immunology , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay/methods , Fatty Acids, Monounsaturated/pharmacology , Fluvastatin , Humans , Indoles/pharmacology , Interleukin-1alpha/analysis , Interleukin-1alpha/biosynthesis , Interleukin-1alpha/genetics , Interleukin-1beta/analysis , Interleukin-1beta/biosynthesis , Interleukin-1beta/genetics , Interleukin-6/analysis , Interleukin-6/biosynthesis , Interleukin-6/genetics , Myocytes, Cardiac/drug effects , Receptors, Tumor Necrosis Factor, Type I/immunology , Receptors, Tumor Necrosis Factor, Type II/immunology , Reverse Transcriptase Polymerase Chain Reaction/methods , Rosiglitazone , Simvastatin/pharmacology , Stimulation, Chemical , Troglitazone
SELECTION OF CITATIONS
SEARCH DETAIL
...