Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters











Publication year range
1.
Am J Transl Res ; 5(4): 427-40, 2013.
Article in English | MEDLINE | ID: mdl-23724166

ABSTRACT

As a versatile regulatory mechanism, sumoylation has been found to be essential for ordered diverse cellular processes. However, the exact impact of sumoylation on endothelial function largely remained elusive. Here we investigated the role of small ubiquitin-like modifier 1 (SUMO1) mediated sumoylation in the regulation of endothelial function by examining its effect on angiogenesis and homeostatic responses. Adenoviral-mediated SUMO1 expression in porcine aortic endothelial cells (PAECs) dose-dependently promoted proliferation, migration and tube formation. In line with these results in PAECs, Matrigel plug assays in SUMO1 transgenic mice demonstrated a significant higher capacity for vascular neogenesis as compared with that of control littermates. Moreover, SUMO1 expression protected PAECs from serum starvation or H2O2-induced apoptosis. Mechanistic studies demonstrated that SUMO1 sumoylation modulates ERK1/2 activation and MMP13 expression as well as Jak2/STAT5 signaling to promote angiogenesis. SUMO1 sumoylation also suppressed NFκB and c-JUN transcriptional activity to provide protection for PAECs against oxidative stress-induced apoptosis. Given that sumoylation is a reversible process, dynamic regulation of the sumoylation function could be a novel strategy to modulate endothelial function in disease states.

2.
Breast Cancer Res ; 14(2): R52, 2012 Mar 19.
Article in English | MEDLINE | ID: mdl-22429491

ABSTRACT

INTRODUCTION: In this pre-clinical in vitro study conducted in estrogen receptor positive (ER+) breast cancer cells, we have characterized the effects of insulin-like growth factor I (IGF-1) on the cytostatic and cytotoxic action of antiestrogen treatment when used as a single agent or in combination with the antiprogestin mifepristone (MIF). Our goal was to identify new molecular targets to improve the efficacy of hormonal therapy in breast cancer patients that have a poor response to hormonal therapy, in part, due to high circulating levels of unbound insulinIGF-1. METHODS: IGF-1-mediated effects on cytostasis and apoptotic cell death were determined with cell counts conducted in the presence and absence of trypan blue; enzyme-linked immunosorbent assays to determine the intracellular levels of cleaved cytokeratin 18, a marker of epithelial cancer cell apoptosis; and immunoblot analysis to determine the levels of cleaved poly-ADP ribose polymerase (PARP) and lamin A that result from caspase-dependent apoptosis. Cytotoxicity was further characterized by determination of the levels of reactive oxygen species (ROS) and the percent of mitochondrial membrane depolarization in cell populations treated with the different hormones in the presence and absence of IGF-1. Small molecule inhibitors of the dual-specificity protein kinase MEK1, MEK1 siRNA, Bim siRNA, and vectors overexpressing MEK1 wild type and mutant, dominant negative cDNA were used to identify key IGF-1 downstream prosurvival effectors. RESULTS: IGF-1, at physiologically relevant levels, blocked the cytotoxic action(s) of the antiestrogens 4-hydroxytamoxifen (4-OHT) and tamoxifen (TAM) when used as single agents or in combination with the antiprogestin MIF. The antiapoptotic action of IGF-1 was mediated primarily through the action of MEK1. MEK1 expression reduced the levels of ROS and mitochondrial membrane depolarization induced by the hormonal treatments via a mechanism that involved the phosphorylation and proteasomal turnover of the proapoptotic BH3-only Bcl-2 family member Bim. Importantly, small-molecule inhibitors of MEK1 circumvented the prosurvival action of IGF-1 by restoring Bim to levels that more effectively mediated apoptosis in ER+ breast cancer cells. CONCLUSION: his study provides strong support for the use of MEK1 inhibitors in combination with hormonal therapy to effectively affect cytostasis and activate a Bim-dependent apoptotic pathway in ER+ breast cancer cells. We discuss that MEK1 blockade may be a particularly effective treatment for women with high circulating levels of IGF-1, which have been correlated to a poor prognosis.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Estrogen Receptor Modulators/pharmacology , Insulin-Like Growth Factor I/metabolism , MAP Kinase Kinase 1/metabolism , Membrane Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Bcl-2-Like Protein 11 , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Estrogen Antagonists/pharmacology , Female , Hormone Antagonists/pharmacology , Humans , Insulin-Like Growth Factor I/pharmacology , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/genetics , Mifepristone/pharmacology , Oxidative Stress/drug effects , Phosphorylation/drug effects , Reactive Oxygen Species/metabolism , Receptors, Estrogen/metabolism , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology
3.
Cell Mol Life Sci ; 68(6): 931-49, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20953658

ABSTRACT

In recent years the etiopathology of a number of debilitating diseases such as type 2 diabetes, arthritis, atherosclerosis, psoriasis, asthma, cystic fibrosis, sepsis, and ulcerative colitis has increasingly been linked to runaway cytokine-mediated inflammation. Cytokine-based therapeutic agents play a major role in the treatment of these diseases. However, the temporospatial changes in various cytokines are still poorly understood and attempts to date have focused on the inhibition of specific cytokines such as TNF-α. As an alternative approach, a number of preclinical studies have confirmed the therapeutic potential of targeting alpha7 nicotinic acetylcholine receptor-mediated anti-inflammatory effects through modulation of proinflammatory cytokines. This "cholinergic anti-inflammatory pathway" modulates the immune system through cholinergic mechanisms that act on alpha7 receptors expressed on macrophages and immune cells. If the preclinical findings translate into human efficacy this approach could potentially provide new therapies for treating a broad array of intractable diseases and conditions with inflammatory components.


Subject(s)
Cytokines/metabolism , Inflammation/metabolism , Models, Molecular , Nicotinic Agonists/metabolism , Receptors, Nicotinic/metabolism , Arthritis/complications , Asthma/complications , Colitis, Ulcerative/complications , Cystic Fibrosis/complications , Diabetes Mellitus, Type 2/complications , Humans , Inflammation/drug therapy , Inflammation/etiology , Models, Biological , Molecular Structure , Nicotinic Agonists/chemistry , Parkinson Disease/complications , Psoriasis/complications , Sepsis/complications , alpha7 Nicotinic Acetylcholine Receptor
4.
Pharm Res ; 28(2): 413-6, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20859658

ABSTRACT

Inflammatory disorders are characterized by the influx of immune cells into the vascular wall of veins and/or arteries in response to stimuli such as oxidized-LDL and various pathogens. These factors stimulate the local production of pro-inflammatory cytokines by macrophages and other cells that promote various inflammatory diseases such as atherosclerosis, Crohn's, Alzheimer's and diabetes. Numerous cytokines play a significant role in this process, though tumor necrosis factor (TNF) and various interleukins are thought to be among the most important regulators. These proinflammatory cytokines promote the above-described diseases by inducing endothelial cell dysfunction. In this brief commentary we will discuss some of the latest advances and discoveries in the treatment of these inflammatory diseases, making use of alpha7 nicotinic acetylcholine receptor (alpha7 nAChR) agonists.


Subject(s)
Inflammation/prevention & control , Inflammation/physiopathology , Nicotinic Agonists/pharmacology , Receptors, Nicotinic/metabolism , Animals , Cytokines/immunology , Endothelial Cells/drug effects , Humans , Interleukins/metabolism , Macrophages/metabolism , Mice , Nicotinic Agonists/chemistry , Nicotinic Agonists/therapeutic use , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism , alpha7 Nicotinic Acetylcholine Receptor
5.
Vascul Pharmacol ; 53(3-4): 160-8, 2010.
Article in English | MEDLINE | ID: mdl-20601126

ABSTRACT

Insulin resistance is an underlying mechanism of type 2 diabetes and its vascular complications. Recent evidence suggests that crosstalk between angiotensin II (Ang II) and the insulin signaling in vascular smooth muscle cell (VSMC) may contribute to cellular insulin resistance. We hypothesized that Ang II inhibits the anti-mitogenic pathways while enhancing the mitogenic pathways stimulated by insulin via activation of Protein Tyrosine Phosphatase-1B (PTP-1B) in VSMC. We found that Ang II significantly inhibited insulin-induced phosphorylation of tyrosine 608 of IRS-1 and serine 473 of Akt, a downstream member of anti-mitogenic pathway of insulin. In contrast, Ang II increased the serine phosphorylation of IRS-1 which was not affected by the presence of insulin. Activation of p42/p44 MAPK (a mitogenic pathway) induced by insulin was further enhanced by Ang II. Transfection of VSMC with PTP-1B antisense oligonucleotide markedly reduced the effects of Ang II on insulin signaling. Furthermore, an increase in VSMC growth was attenuated by PTP-1B antisense only in the presence of both Ang II and insulin. Finally, we also showed that Ang II-induced activation of PTP-1B in VSMC was PKA/JAK2 dependent. We conclude that Ang II modulates both anti-mitogenic and mitogenic pathways of insulin via the activation of PTP-1B.


Subject(s)
Angiotensin II/physiology , Insulin Resistance/physiology , Myocytes, Smooth Muscle/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/physiology , Animals , Aorta/metabolism , Cells, Cultured , Enzyme Activation , Insulin/pharmacology , Male , Phosphorylation , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology
6.
J Exp Biol ; 213(Pt 10): 1782-7, 2010 May.
Article in English | MEDLINE | ID: mdl-20435829

ABSTRACT

Cell-attached patches from isolated epithelial cells from larval bullfrog skin revealed a cation channel that was activated by applying suction (-1 kPa to -4.5 kPa) to the pipette. Activation was characterized by an initial large current spike that rapidly attenuated to a stable value and showed a variable pattern of opening and closing with continuing suction. Current-voltage plots demonstrated linear or inward rectification and single channel conductances of 44-56 pS with NaCl or KCl Ringer's solution as the pipette solution, and a reversal potential (-V(p)) of 20-40 mV. The conductance was markedly reduced with N-methyl-D-glucamide (NMDG)-Cl Ringer's solution in the pipette. Neither amiloride nor ATP, which are known to stimulate an apical cation channel in Ussing chamber preparations of larval frog skin, produced channel activation nor did these compounds affect the response to suction. Stretch activation was not affected by varying the pipette concentrations of Ca(2+) between 0 mmol l(-1) and 4 mmol l(-1) or by varying pH between 6.8 and 8.0. However, conductance was reduced with 4 mmol l(-1) Ca(2+). Western blot analysis of membrane homogenates from larval bullfrog and larval toad skin identified proteins that were immunoreactive with mammalian TRPC1 and TRPC5 (TRPC, canonical transient receptor potential channel) antibodies while homogenates of skin from newly metamorphosed bullfrogs were positive for TRPC1 and TRPC3/6/7 antibodies. The electrophysiological response of larval bullfrog skin resembles that of a stretch-activated cation channel characterized in Xenopus oocytes and proposed to be TRPC1. These results indicate this channel persists in all life stages of anurans and that TRP isoforms may be important for sensory functions of their skin.


Subject(s)
Ion Channel Gating/physiology , Rana catesbeiana/metabolism , Skin/metabolism , Stress, Mechanical , TRPC Cation Channels/metabolism , Animals , Blotting, Western , Calcium/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Hydrogen-Ion Concentration/drug effects , Ion Channel Gating/drug effects , Larva , Potassium Chloride/pharmacology , Skin/drug effects , Sodium Chloride/pharmacology
7.
PLoS One ; 5(3): e9593, 2010 Mar 09.
Article in English | MEDLINE | ID: mdl-20231889

ABSTRACT

Given the importance of Jak2 in cell signaling, a critical role for Jak2 in immune cells especially dendritic cells (DCs) has long been proposed. The exact function for Jak2 in DCs, however, remained poorly understood as Jak2 deficiency leads to embryonic lethality. Here we established Jak2 deficiency in adult Cre(+/+)Jak2(fl/fl) mice by tamoxifen induction. Loss of Jak2 significantly impaired DC development as manifested by reduced BMDC yield, smaller spleen size and reduced percentage of DCs in total splenocytes. Jak2 was also crucial for the capacity of DCs to mediate innate immune response. Jak2(-/-) DCs were less potent in response to inflammatory stimuli and showed reduced capacity to secrete proinflammatory cytokines such as TNFalpha and IL-12. As a result, Jak2(-/-) mice were defective for the early clearance of Listeria after infection. However, their potency to mediate adaptive immune response was not affected. Unlike DCs, Jak2(-/-) macrophages showed similar capacity secretion of proinflammatory cytokines, suggesting that Jak2 selectively modulates innate immune response in a DC-dependent manner. Consistent with these results, Jak2(-/-) mice were remarkably resistant to lethal dose of LPS-induced septic shock, a deadly sepsis characterized by the excessive innate immune response, and adoptive transfer of normal DCs restored their susceptibility to LPS-induced septic shock. Mechanistic studies revealed that Jak2/SATA5 signaling is pivotal for DC development and maturation, while the capacity for DCs secretion of proinflammatory cytokines is regulated by both Jak2/STAT5 and Jak2/STAT6 signaling.


Subject(s)
Dendritic Cells/cytology , Janus Kinase 2/metabolism , Lipopolysaccharides/metabolism , Animals , Immunity, Innate , Inflammation , Interleukin-12/metabolism , Macrophages/metabolism , Male , Mice , Mice, Transgenic , STAT5 Transcription Factor/metabolism , STAT6 Transcription Factor/metabolism , Spleen/metabolism , Tumor Necrosis Factor-alpha/metabolism
8.
J Pharmacol Exp Ther ; 332(1): 173-80, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19786623

ABSTRACT

Type 2 diabetes has become a pervasive public health problem. The etiology of the disease has not been fully defined but appears to involve abnormalities in peripheral and central nervous system pathways, as well as prominent inflammatory components. Because nicotinic acetylcholine receptors (nAChRs) are known to interact with anti-inflammatory pathways and have been implicated in control of appetite and body weight, as well as lipid and energy metabolism, we examined their role in modulating biological parameters associated with the disease. In a model of type 2 diabetes, the homozygous leptin-resistant db/db obese mouse, we measured the effects of a novel alpha7 nAChR-selective agonist [5-methyl-N-[2-(pyridin-3-ylmethyl)-1-azabicyclo[2.2.2]oct-3-yl]thiophene-2-carboxamide (TC-7020)] on body mass, glucose and lipid metabolism, and proinflammatory cytokines. Oral administration of TC-7020 reduced weight gain and food intake, reduced elevated glucose and glycated hemoglobin levels, and lowered elevated plasma levels of triglycerides and the proinflammatory cytokine tumor necrosis factor-alpha. These changes were reversed by the alpha7-selective antagonist methyllycaconitine, confirming the involvement of alpha7 nAChRs. Prevention of weight gain, decreased food intake, and normalization of glucose levels were also blocked by the Janus kinase 2 (JAK2) inhibitor alpha-cyano-(3,4-dihydroxy)-N-benzylcinnamide (AG-490), suggesting that these effects involve linkage of alpha7 nAChRs to the JAK2-signal transducer and activator of transcription 3 signaling pathway. The results show that alpha7 nAChRs play a central role in regulating biological parameters associated with diabetes and support the potential of targeting these receptors as a new therapeutic strategy for treatment.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Nicotinic Agonists/pharmacology , Obesity/prevention & control , Quinuclidines/pharmacology , Receptors, Nicotinic/metabolism , Thiophenes/pharmacology , Weight Gain/drug effects , Animals , Binding, Competitive , Blood Glucose/metabolism , Cell Line , Cloning, Molecular , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Type 2/blood , Electrophysiological Phenomena , Energy Metabolism/drug effects , Female , Humans , Ligands , Lipid Metabolism/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Structure , Nicotinic Agonists/chemistry , Obesity/blood , Obesity/metabolism , Oocytes/metabolism , Patch-Clamp Techniques , Quinuclidines/chemistry , Rats , Receptors, Leptin/genetics , Receptors, Nicotinic/biosynthesis , Receptors, Nicotinic/physiology , Thiophenes/chemistry , Tumor Necrosis Factor-alpha/blood , Xenopus laevis , alpha7 Nicotinic Acetylcholine Receptor
9.
Circ Res ; 105(10): 1013-22, 2009 Nov 06.
Article in English | MEDLINE | ID: mdl-19797171

ABSTRACT

RATIONALE: Obesity is a risk factor for cardiovascular dysfunction, yet the underlying factors driving this impaired function remain poorly understood. Insulin resistance is a common pathology in obese patients and has been shown to impair vascular function. Whether insulin resistance or obesity, itself, is causal remains unclear. OBJECTIVE: The present study tested the hypothesis that insulin resistance is the underlying mediator for impaired NO-mediated dilation in obesity by genetic deletion of the insulin-desensitizing enzyme protein tyrosine phosphatase (PTP)1B in db/db mice. METHODS AND RESULTS: The db/db mouse is morbidly obese, insulin-resistant, and has tissue-specific elevation in PTP1B expression compared to lean controls. In db/db mice, PTP1B deletion improved glucose clearance, dyslipidemia, and insulin receptor signaling in muscle and fat. Hepatic insulin signaling in db/db mice was not improved by deletion of PTP1B, indicating specific amelioration of peripheral insulin resistance. Additionally, obese mice demonstrate an impaired endothelium dependent and independent vasodilation to acetylcholine and sodium nitroprusside, respectively. This impairment, which correlated with increased superoxide in the db/db mice, was corrected by superoxide scavenging. Increased superoxide production was associated with increased expression of NAD(P)H oxidase 1 and its molecular regulators, Noxo1 and Noxa1. CONCLUSIONS: Deletion of PTP1B improved both endothelium dependent and independent NO-mediated dilation and reduced superoxide generation in db/db mice. PTP1B deletion did not affect any vascular function in lean mice. Taken together, these data reveal a role for peripheral insulin resistance as the mediator of vascular dysfunction in obesity.


Subject(s)
Endothelium, Vascular/enzymology , Gene Deletion , Gene Expression Regulation, Enzymologic , Insulin Resistance , Leptin/metabolism , Obesity/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Acetylcholine/pharmacology , Adaptor Proteins, Signal Transducing , Adipose Tissue/enzymology , Animals , Dyslipidemias/enzymology , Dyslipidemias/genetics , Glucose/genetics , Glucose/metabolism , Leptin/genetics , Mice , Mice, Inbred BALB C , Mice, Obese , Muscles/enzymology , NADH, NADPH Oxidoreductases/biosynthesis , NADH, NADPH Oxidoreductases/genetics , NADPH Oxidase 1 , Nitric Oxide/genetics , Nitric Oxide/metabolism , Nitroprusside/pharmacology , Obesity/genetics , Oxidation-Reduction/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Proteins/genetics , Proteins/metabolism , Superoxides/metabolism , Vasodilation/drug effects , Vasodilation/genetics , Vasodilator Agents/pharmacology
10.
Circulation ; 120(9): 753-63, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19687357

ABSTRACT

BACKGROUND: Obesity causes hypertension and sympathoactivation, a process proposed to be mediated by leptin. Protein tyrosine phosphatase 1B (PTP1B), a major new pharmaceutical target in the treatment of obesity and type II diabetes mellitus, constrains the metabolic actions of leptin, but the extent to which PTP1B regulates its cardiovascular effects is unclear. This study examined the hypothesis that PTP1B is a negative regulator of the cardiovascular effects of leptin. METHODS AND RESULTS: PTP1B knockout mice had lower body fat but higher mean arterial pressure (116+/-5 versus 105+/-5 mm Hg, P<0.05) than controls. Leptin infusion produced a greater anorexic effect in PTP1B knockout mice and a marked increase in mean arterial pressure (135+/-5 mm Hg) in PTP1B knockout mice only. The decrease in mean arterial pressure in response to ganglionic blockade was higher in PTP1B knockout mice (-38+/-3% versus -29+/-3%, P<0.05), which suggests increased sympathetic tone. PTP1B deletion blunted mean arterial pressure responses to phenylephrine injection (55+/-10% versus 93+/-7%, P<0.05). Phenylephrine-induced aortic contraction was reduced in PTP1B knockout mice (57.7+/-9% versus 96.3+/-12% of KCl, P<0.05), consistent with desensitization to chronically elevated sympathetic tone. Furthermore, PTP1B deletion significantly reduced gene expression of 3 alpha(1)-adrenergic receptor subtypes, consistent with blunted constriction to phenylephrine. CONCLUSIONS: These data indicate that PTP1B is a key regulator of the cardiovascular effects of leptin and that reduced vascular adrenergic reactivity provides a compensatory limit to the effects of leptin on mean arterial pressure.


Subject(s)
Hypertension/physiopathology , Leptin/metabolism , Obesity/physiopathology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Adrenergic alpha-Antagonists/pharmacology , Animals , Aorta/physiology , Blood Pressure/physiology , Hypertension/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Obesity/metabolism , Phenotype , Phenylephrine/pharmacology , Prazosin/pharmacology , Receptors, Adrenergic, alpha-1/genetics , Stress, Physiological/physiology , Sympathetic Nervous System/physiology , Vasoconstriction/drug effects , Vasoconstriction/physiology , Vasoconstrictor Agents/pharmacology
11.
Brain Res ; 1256: 1-7, 2009 Feb 23.
Article in English | MEDLINE | ID: mdl-19063868

ABSTRACT

Our laboratories have previously identified the alpha7 nAChR-JAK2 pathway as playing a central role in nicotine-induced neuroprotection. We have also reported that the angiotensin II (Ang II) AT(2) receptor induced activation of SHP-1 induces the tyrosine dephosphorylation of JAK2 that results in a complete neutralization of the alpha7 nAChR-JAK2 pro-survival cascade. In this study, we investigated the effects of inhibiting the alpha7 nAChR-JAK2 pro-survival cascade on the nicotine-induced production of the survival factor Bcl-2 and the transcriptional activation of NF-kappaB, AP-1, STAT1, STAT3, and STAT5. We report that nicotine induced the production of Bcl-2 and increased the transcriptional activation of NF-kappaB, AP-1, STAT1, and STAT3, and with the exception of AP-1, the other transcription factors (NF-kappaB, STAT1, and STAT3) were significantly reduced by JAK2 inhibition. We also demonstrate that, via transfection of either Bcl-2 antisense or NF-kappaB, STAT1 and STAT3 transcription factor decoys oligodeoxyribonucleotides into PC12 cells, nicotine induces its neuroprotection in PC12 cells via activation of the alpha7 nAChR-JAK2-(NF-kappaB; STAT3)-Bcl-2 pro-survival pathway. Finally, the neuroprotective nicotine-induced production of Bcl-2 appears to fully counteract the Abeta (1-42)-induced apoptosis of PC12 cells by blocking Abeta (1-42)-induced mitochondrial release of cytosolic cytochrome C.


Subject(s)
Apoptosis , Inflammation/physiopathology , Janus Kinase 2/metabolism , NF-kappa B/metabolism , Receptors, Nicotinic/physiology , STAT3 Transcription Factor/metabolism , Signal Transduction , Amyloid beta-Peptides/metabolism , Animals , Cytochromes c/metabolism , Enzyme Activation , Gene Expression , Janus Kinase 2/antagonists & inhibitors , Mitochondria/physiology , Neuroprotective Agents/pharmacology , Nicotine/pharmacology , PC12 Cells , Peptide Fragments/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , STAT1 Transcription Factor/metabolism , Transcription Factor AP-1/metabolism , Tyrphostins/pharmacology , alpha7 Nicotinic Acetylcholine Receptor
12.
Can J Physiol Pharmacol ; 86(6): 358-64, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18516099

ABSTRACT

Insulin resistance involves decreased phosphorylation of insulin receptor substrate (IRS) proteins and (or) Akt. In the vasculature, modulated Akt phosphorylation may cause impaired vasorelaxation via decreased eNOS activation. Diet-induced insulin resistance enhances endothelin-1(ET-1)-mediated vasoconstriction and prevents vasodilatation to insulin. Presently, we evaluated insulin-mediated vascular relaxation, assessed molecular markers of the insulin signaling pathway, and determined the involvement of ET-1 in response to insulin by using selective ETA- or ETB-receptor blockade in a lean model of type 2 diabetes. Dose-response curves to insulin (0.01-100 ng/mL) were generated with wire myograph using thoracic aorta rings from control Wistar or diabetic Goto-Kakizaki (GK) rats (n=3-11). Maximal relaxation (Rmax) to insulin was significantly impaired and insulin sensitivity was decreased in the GK group. Preincubation with 1 micromol/L BQ-123 or BQ-788 for ETA- and ETB-receptor blockade, respectively, resulted in improved insulin sensitivity. Immunoblotting for native and phosphorylated Akt and IRS-1 revealed a decrease in Akt activation in the GK group. In vivo hyperinsulinemic euglycemic clamp studies showed decreased glucose utilization in GK rats, indicative of insulin resistance. These findings provide evidence that vascular insulin resistance occurs in a nonobese model of diabetes and that both ET receptor subtypes are involved in vascular relaxation to insulin.


Subject(s)
Diabetes Mellitus, Type 2/physiopathology , Endothelin-1/physiology , Insulin/pharmacology , Vasodilation/drug effects , Adaptor Proteins, Signal Transducing/physiology , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Glucose Clamp Technique , In Vitro Techniques , Insulin/administration & dosage , Insulin/physiology , Male , Phosphorylation , Rats , Rats, Wistar , Receptor, Endothelin A/physiology , Receptor, Endothelin B/physiology , Receptor, Insulin/physiology , Signal Transduction , Vasodilation/physiology
13.
Arterioscler Thromb Vasc Biol ; 28(9): 1627-33, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18556569

ABSTRACT

OBJECTIVE: Increased formation of reactive oxygen species (ROS) has been identified as a causative factor in endothelial dysfunction by reducing NO bioavailability and uncoupling endothelial nitric oxide synthase (eNOS). However, the specific contribution of ROS to endothelial function is not well understood. METHODS AND RESULTS: A major source of intracellular ROS is the NADPH oxidase (Nox) family of enzymes. The goal of the current study was to directly assess the contribution of NADPH oxidase derived superoxide to eNOS function by expressing Nox5, a single gene product that constitutively produces superoxide within cells. Paradoxically, we found that instead of inhibiting eNOS, coexpression of Nox5 increased NO release from both bovine and human endothelial cells. To establish the functional significance of this observation in intact blood vessels, the endothelium of mouse aorta was transduced with Nox5 or control adenoviruses. Nox5 potently inhibited Ach-induced relaxation and potentiated contractile responses to phenylephrine. In precontracted aortae, acute exposure to superoxide dismutase induced significant vascular relaxation in vessels exposed to Nox5 versus control and unmasked the ability of Nox5 to activate eNOS in blood vessel endothelium. CONCLUSIONS: These findings suggest that ROS inhibit eNOS function via consumption of NO rather than direct inhibition of enzymatic activity.


Subject(s)
Endothelial Cells/enzymology , Endothelium, Vascular/enzymology , NADPH Oxidases/metabolism , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/metabolism , Superoxides/metabolism , Acetylcholine/pharmacology , Animals , COS Cells , Cattle , Cells, Cultured , Chlorocebus aethiops , Cyclic GMP/metabolism , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Enzyme Activation , Humans , Mice , Mice, Inbred C57BL , NADPH Oxidases/genetics , Nitric Oxide Synthase Type III/genetics , Phosphorylation , Superoxide Dismutase/metabolism , Transduction, Genetic , Transfection , Vasoconstriction , Vasoconstrictor Agents/pharmacology , Vasodilation , Vasodilator Agents/pharmacology
14.
Am J Physiol Heart Circ Physiol ; 293(2): H1291-9, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17526654

ABSTRACT

The JAK/STAT pathway is activated in vitro by angiotensin II (ANG II) and endothelin-1 (ET-1), which are implicated in the development of diabetic complications. We hypothesized that ANG II and ET-1 activate the JAK/STAT pathway in vivo to participate in the development of diabetic vascular complications. Using male Sprague-Dawley rats, we performed a time course study [days 7, 14, and 28 after streptozotocin (STZ) injection] to determine changes in phosphorylation of JAK2, STAT1, and STAT3 in thoracic aorta using standard Western blot techniques. On day 7 there was no change in phosphorylation of JAK2, STAT1, and STAT3. Phosphorylation of JAK2, STAT1, and STAT3 was significantly increased on days 14 and 28 and was inhibited by treatment with candesartan (AT(1) receptor antagonist, 10 mg x kg(-1) x day(-1) orally in drinking water), atrasentan (ET(A) receptor antagonist, 10 mg x kg(-1) x day(-1) orally in drinking water), and AG-490 (JAK2 inhibitor, 5 mg x kg(-1) x day(-1) intraperitoneally). On day 28, treatment with all inhibitors prevented the significant increase in systolic blood pressure (SBP; tail cuff) of STZ-induced diabetic rats (SBP: 157 +/- 9.0, 130 +/- 3.3, 128 +/- 6.8, and 131 +/- 10.4 mmHg in STZ, STZ-candesartan, STZ-atrasentan, and STZ-AG-490 rats, respectively). In isolated tissue bath studies, diabetic rats displayed impaired endothelium-dependent relaxation in aorta (maximal relaxation: 95.3 +/- 3.0, 92.6 +/- 7.4, 76.9 +/- 12.1, and 38.3 +/- 13.1% in sham, sham + AG-490, STZ + AG-490, and STZ rats, respectively). Treatment of rats with AG-490 restored endothelium-dependent relaxation in aorta from diabetic rats at 14 and 28 days of treatment. These results demonstrate that JAK2 activation in vivo participates in the development of vascular complications associated with STZ-induced diabetes.


Subject(s)
Angiotensin II/metabolism , Aorta, Thoracic/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetic Angiopathies/etiology , Endothelin-1/metabolism , Janus Kinase 2/metabolism , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/enzymology , Aorta, Thoracic/physiopathology , Atrasentan , Benzimidazoles/pharmacology , Biphenyl Compounds , Blood Glucose/metabolism , Blood Pressure , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/physiopathology , Diabetic Angiopathies/enzymology , Diabetic Angiopathies/metabolism , Diabetic Angiopathies/physiopathology , Endothelin A Receptor Antagonists , Enzyme Activation , Intracellular Signaling Peptides and Proteins/metabolism , Janus Kinase 2/antagonists & inhibitors , Male , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Protein Tyrosine Phosphatases/metabolism , Pyrrolidines/pharmacology , Rats , Receptor, Angiotensin, Type 1/metabolism , Receptor, Endothelin A/metabolism , Signal Transduction , Tetrazoles/pharmacology , Time Factors , Tyrphostins/pharmacology , Vasodilation
15.
Am J Physiol Renal Physiol ; 292(5): F1380-9, 2007 May.
Article in English | MEDLINE | ID: mdl-17213463

ABSTRACT

Experiments were performed to establish the pharmacological profile of purinoceptors and to identify the signal transduction pathways responsible for increases in intracellular calcium concentration ([Ca(2+)](i)) for cultured mouse mesangial cells. Mouse mesangial cells were loaded with fura 2 and examined using fluorescent spectrophotometry. Basal [Ca(2+)](i) averaged 102 +/- 2 nM (n = 346). One hundred micromolar concentrations of ATP, ADP, 2',3'-(benzoyl-4-benzoyl)-ATP (BzATP), ATP-gamma-S, and UTP in normal Ca(2+) medium evoked peak increases in [Ca(2+)](i) of 866 +/- 111, 236 +/- 18, 316 +/- 26, 427 +/- 37, and 808 +/- 73 nM, respectively. UDP or 2-methylthio-ATP (2MeSATP) failed to elicit significant increases in [Ca(2+)](i), whereas identical concentrations of adenosine, AMP, and alpha,beta-methylene ATP (alpha,beta-MeATP) had no detectable effect on [Ca(2+)](i). Removal of Ca(2+) from the extracellular medium had no significant effect on the peak increase in [Ca(2+)](i) induced by ATP, ADP, BzATP, ATP-gamma-S, or UTP compared with normal Ca(2+); however, Ca(2+)-free conditions did accelerate the rate of decline in [Ca(2+)](i) in cells treated with ATP and UTP. [Ca(2+)](i) was unaffected by membrane depolarization with 143 mM KCl. Western blot analysis for P2 receptors revealed expression of P2X(2), P2X(4), P2X(7), P2Y(2), and P2Y(4) receptors. No evidence of P2X(1) and P2X(3) receptor expression was detected, whereas RT-PCR analysis reveals mRNA expression for P2X(1), P2X(2), P2X(3), P2X(4), P2X(7), P2Y(2), and P2Y(4) receptors. These data indicate that receptor-specific P2 receptor activation increases [Ca(2+)](i) by stimulating calcium influx from the extracellular medium and through mobilization of Ca(2+) from intracellular stores in cultured mouse mesangial cells.


Subject(s)
Calcium/metabolism , Intracellular Membranes/metabolism , Mesangial Cells/metabolism , Receptors, Purinergic P2/physiology , Animals , Calcium Signaling , Cell Line , Culture Media/metabolism , Mice , Osmolar Concentration , Purinergic Agonists , Pyrimidines/metabolism , RNA, Messenger/metabolism , Receptors, Cell Surface/agonists , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stimulation, Chemical
16.
J Biol Chem ; 282(9): 6494-507, 2007 Mar 02.
Article in English | MEDLINE | ID: mdl-17164239

ABSTRACT

In contrast to other Nox isoforms, the activity of Nox5 does not require the presence of accessory proteins and is entirely dependent on the elevation of intracellular calcium. Previous studies have shown that the EC(50) of Nox5 for calcium is relatively high and raises the question of whether Nox5 can be sufficiently activated in cells that do not experience extreme elevations of intracellular calcium. In the current study, we have identified a novel mechanism governing the activity of Nox5. Exposure of cells expressing Nox5 to phorbol 12-myristate 13-acetate (PMA) resulted in a slow and sustained increase in ROS, which was markedly different from the rapid response to ionomycin. PMA greatly potentiated the activity of Nox5 in response to low concentrations of ionomycin. The ability of PMA to increase Nox5 activity was abolished by calcium chelation and was a direct effect on enzyme activity, since PMA increased the calcium sensitivity of Nox5 in a cell-free assay. PMA stimulated the time-dependent phosphorylation of Nox5 on Thr(494) and Ser(498). Mutation of these residues to alanine abolished both PMA-dependent phosphorylation and calcium sensitization. Conversely, mutation of Thr(494) and Ser(498) to glutamic acid produced a gain of function mutant that had increased activity at low concentrations of ionomycin. Within the cell, Nox5 was detected in detergent-resistant microdomains of the endoplasmic reticulum. In summary, the phosphorylation of Nox5 at key residues facilitates enzyme activation at lower levels of intracellular calcium and may provide an avenue for enzyme activation in response to a greater variety of extracellular stimuli.


Subject(s)
Calcium/analysis , Membrane Proteins/metabolism , NADPH Oxidases/metabolism , Amino Acid Substitution , Animals , COS Cells , Chlorocebus aethiops , Endoplasmic Reticulum/enzymology , Enzyme Activation , Humans , Membrane Proteins/genetics , NADPH Oxidase 5 , NADPH Oxidases/genetics , Phosphorylation , Reactive Oxygen Species/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Transfection
17.
Am J Physiol Renal Physiol ; 290(4): F762-8, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16527921

ABSTRACT

Excessive cellular growth is a major contributor to pathological changes associated with diabetic nephropathy. In particular, high glucose-induced growth of glomerular mesangial cells is a characteristic feature of diabetes-induced renal complications. Glomerular mesangial cells respond to traditional growth factors, although in diabetes this occurs in the context of an environment enriched in both circulating vasoactive mediators and high glucose. For example, the vasoactive peptide ANG II has been implicated in the pathogenesis of diabetic renal disease, and recent findings suggest that high glucose and ANG II activate intracellular signaling processes, including the polyol pathway and generation of reactive oxygen species. These pathways activate the Janus kinase (JAK)/signal transducers and activators of transcription (STAT) signaling cascades in glomerular mesangial cells. Activation of the JAK/STAT signaling cascade can stimulate excessive proliferation and growth of glomerular mesangial cells, contributing to diabetic nephropathy. This review focuses on some of the key elements in the diabetic microenvironment, especially high glucose and the accumulation of advanced glycoxidation end products and considers their impact on ANG II and other vasoactive peptide-mediated signaling events in vitro and in vivo.


Subject(s)
Diabetic Nephropathies/genetics , Diabetic Nephropathies/physiopathology , Glucose/metabolism , Protein-Tyrosine Kinases/metabolism , STAT Transcription Factors/metabolism , Signal Transduction , Angiotensin II/physiology , Glucose/pharmacokinetics , Humans , Kidney Glomerulus/pathology , Kidney Glomerulus/physiology
18.
Am J Physiol Renal Physiol ; 291(1): F116-21, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16449352

ABSTRACT

In the current study, we investigated the effect of simvastatin on the ability of high glucose (HG) and ANG II to activate the JAK2-STAT signaling cascade and induce glomerular mesangial cell (GMC) growth. We found that pretreatment with simvastatin significantly inhibited HG- and ANG II-induced collagen IV production, JAK2 activation, and phosphorylation of STAT1 and STAT3 in GMC. We also found that the activation of JAK2 by HG and ANG II was dependent on the Rho family of GTPases. Consistent with these in vitro results, both albumin protein excretion and phosphorylation of JAK2, STAT1, and STAT3 were attenuated in renal glomeruli by administration of simvastatin in a streptozotocin-induced rat model of HG diabetes. This study demonstrates that simvastatin blocks ANG II-induced activation of the JAK/STAT pathway in the diabetic environment, in vitro and in vivo, and, thereby, provides new insights into the molecular mechanisms underlying early diabetic nephropathy.


Subject(s)
Angiotensin II/physiology , Anticholesteremic Agents/pharmacology , Blood Glucose/physiology , Mesangial Cells/drug effects , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Simvastatin/pharmacology , Animals , Cells, Cultured , Collagen Type IV/metabolism , Diabetes Mellitus, Experimental/physiopathology , Diabetic Nephropathies/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Janus Kinase 2 , Male , Mesangial Cells/chemistry , Mesangial Cells/physiology , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Terpenes/pharmacology , rho GTP-Binding Proteins/physiology
19.
Vascul Pharmacol ; 43(5): 310-9, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16290054

ABSTRACT

Endothelin-1 (ET-1) and JAK2 are both implicated in diabetic complications. Therefore, we investigated whether ET-1 differentially activates JAK2 under conditions of normal (5 mM) and high (25 mM) glucose. We tested the hypothesis that reactive oxygen species mediate the activation of JAK2 in response to ET-1. In rat aortic vascular smooth muscle cells (VSMC), ET-1 (10 (- 7) M, 5 min) stimulated the activation of JAK2, which was further enhanced under high glucose conditions. Allopurinol (xanthine oxidase inhibitor, 1 microM) and l-NAME (nitric oxide synthase inhibitor, 1 mM) had no effect on ET-1-induced JAK2 activation, while apocynin (NAD(P)H oxidase inhibitor 100 microM) resulted in a significant inhibition of ET-1-induced JAK2 and MAPK activation. Overexpression of SOD did not inhibit ET-1-induced activation of JAK2, but catalase (50 units/mL) treatment resulted in complete inhibition. In vivo administration of apocynin (1.5 mM) resulted in a significant decrease ( 50%), while the ETA receptor antagonist ABT-627 completely inhibited phosphorylation of JAK2 in aortae from STZ-induced diabetic rats. Additionally, DHE staining of aortic sections was significantly reduced in diabetic rats treated with ABT-627. These data suggest that in VSMC, ET-1 via the ETA receptor, utilizes NAD(P)H oxidase to activate JAK2.


Subject(s)
Endothelin-1/pharmacology , Muscle, Smooth, Vascular/enzymology , NADPH Oxidases/metabolism , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins/physiology , Reactive Oxygen Species/metabolism , Adenoviridae/genetics , Animals , Aorta, Thoracic/cytology , Aorta, Thoracic/drug effects , Aorta, Thoracic/enzymology , Cell Separation , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Enzyme Activation/drug effects , Ethidium/analogs & derivatives , Fluorescent Dyes , Glucose/pharmacology , Immunoblotting , In Vitro Techniques , Janus Kinase 2 , Male , Mitogen-Activated Protein Kinases/physiology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , NADPH Oxidases/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL