An increase in bradykinin has been suggested to contribute to the enhanced insulin sensitivity observed in the presence of ACE inhibitors. To investigate a potential direct, nonvascular effect on an insulin target tissue, the effect of bradykinin on glucose uptake and insulin signaling was studied in primary rat adipocytes. Whereas basal glucose uptake was not altered, bradykinin augmented insulin-stimulated glucose uptake twofold, which was blocked by HOE-140, a bradykinin B2 receptor antagonist. The bradykinin effect on glucose uptake was nitric oxide (NO) dependent, mimicked by NO donors and absent in adipocytes from endothelial NO synthase−/− mice. Investigation of insulin signaling revealed that bradykinin enhanced insulin receptor substrate-1 (IRS-1) Tyr phosphorylation, Akt/protein kinase B phosphorylation, and GLUT4 translocation. In contrast, insulin-stimulated extracellular signal–regulated kinase1/2 and Jun NH2-terminal kinase (JNK) activation were decreased in the presence of bradykinin, accompanied by decreased IRS-1 Ser307 phosphorylation. Furthermore, bradykinin did not enhance insulin action in the presence of the JNK inhibitor, SP-600125, or in adipocytes from JNK1−/− mice. These data indicate that bradykinin enhances insulin sensitivity in adipocytes via an NO-dependent pathway that acts by modulating the feedback inhibition of insulin signaling at the level of IRS-1.

Insulin resistance is associated with obesity and type 2 diabetes (1). Factors implicated in the pathogenesis of insulin resistance include nutrient excess, elevated levels of tumor necrosis factor (TNF)-α, resistin, decreased adiponectin, and hyperinsulinemia (rev. in 2,3). Insulin resistance has also been associated with dyslipidemia and hypertension (4). Although these associations have been coined the “metabolic syndrome” (5), the cellular and molecular mechanisms of these linkages are not completely understood.

In the context of hypertension, a number of trials have been conducted in subjects at high risk for the development of cardiovascular disease. The Heart Outcomes Prevention Evaluation (HOPE) study demonstrated that therapy with ramipril, an ACE inhibitor (ACEI), decreased cardiovascular events by 35% and, unexpectedly, significantly reduced the incidence of type 2 diabetes (6). Given the strong association between hypertension and insulin resistance, one possible explanation is that ACE inhibition improves insulin sensitivity. There have been several reports of hypoglycemia concomitant with apparent potentiation of the action of hypoglycemic agents in individuals with type 2 diabetes who received ACEIs (7). Furthermore, ACE inhibition enhanced insulin sensitivity in vivo in dogs and humans (8,9).

Blood pressure is regulated by the renin-angiotensin system (RAS) and the kallikrein-kinin pathway (KKP) (10). The main product of the RAS is angiotensin II (AngII), which is produced in a reaction catalyzed by ACE. AngII is a vasopressor and has been shown to contribute to hypertension and atherogenesis. Bradykinin is the major active peptide of the KKP, which counterregulates the RAS. Bradykinin acts by binding to G-protein–coupled receptors, the B1 and B2 receptor, and signals predominantly through the constitutively expressed B2 receptor to mediate most of its actions in vascular tissue via the activation of endothelial nitric oxide (NO) synthase (eNOS) through a Ca2+-dependent pathway (11,12). Inhibition of ACE, which is also called kininase II, not only blocks the production of AngII, but also elevates the circulating levels of bradykinin (13).

Both the RAS and KKP are present in the circulation as well as in many tissues (10,14). The question of whether decreased AngII or elevated bradykinin mediates the enhanced insulin sensitivity observed in the presence of ACEIs has been addressed in part by the use of AngII receptor blockers (ARBs). In rodent studies, ARBs were not as effective as ACEIs in increasing insulin action (15), and infusion of bradykinin improved insulin action in Zucker fa/fa rats (16). These data point to bradykinin as playing a major role. Although many effects of bradykinin have been attributed to actions on vascular tissue and blood flow (17), the possibility of direct effects on insulin target tissues has not been excluded and its mechanism has not been elucidated. Thus, we tested the effect of bradykinin on insulin sensitivity in isolated rat adipocytes using glucose uptake as a metabolic outcome and explored the interaction of the bradykinin and insulin signal transduction pathways.

Dulbecco’s modified Eagle’s medium was purchased from Invitrogen (Burlington, ON, Canada) and collagenase type I from Worthington (Lakewood, NJ). Bio-Rad protein assay reagent was from Bio-Rad (Hercules, CA) and LumiGlo chemiluminescent substrate kit from KPL (Gaithersburg, MD). The bradykinin assay kit was obtained from Peninsula Laboratories (San Carlos, CA). The Jun NH2-terminal kinase (JNK) activity immunoassay kit was from Calbiochem (San Diego, CA). Anti-bradykinin B2 receptor, anti-Akt/protein kinase B (PKB), anti-Akt2/PKBβ, anti–phospho-Akt/PKB (Thr308), anti–phospho-Akt/PKB (Ser473), anti-p38, anti–extracellular signal–regulated kinase (ERK)1/2, anti–phospho-ERK1/2 (Thr202/Tyr204), anti-JNK, anti–phospho-JNK (Thr183/Tyr185), and anti-pY antibodies were from Cell Signaling (Beverley, MA). Anti-neuronal NO synthase (nNOS), anti-eNOS, and anti-inducible NO synthase (iNOS) antibodies were from BD Transduction Laboratories (San Diego, CA). Anti–insulin receptor substrate-1 (IRS-1), anti–phospho-IRS-1 (Ser307), and anti-p85 antibodies were from Upstate (Lake Placid, NY). Monoclonal anti-hemagglutinin antibody was from Covance (Princeton, NJ). Omapatrilat was kindly provided by Bristol-Myers Squibb (St. Laurent, QC, Canada). Rhodamine red–conjugated anti-IgG1 antibody was from Jackson Immunoresearch (West Grove, PA). Paraformaldehyde was from Fisher Scientific (Ottawa, ON, Canada). Vectashield was from Vector Laboratories (Burlingame, CA). All other chemicals were from Sigma Aldrich (Oakville, ON).

Animal protocols were approved by the Animal Care Committee of the Toronto General Hospital and carried out in accordance with the guidelines of the Canadian Council of Animal Care. Male Sprague-Dawley rats (180–220 g) were obtained from Charles River (St. Constant, QC, Canada). The following strains of male mice (7 weeks old) were obtained from The Jackson Laboratories (Bar Harbor, ME): eNOS−/−, nNOS−/−, JNK1−/−, JNK2−/−, and controls B6129SF2/J and C57BL/6J. Animals were given rodent chow and water ad libitum, exposed to a 12-h light/dark cycle, and acclimatized for 1 week before experiments.

Isolation of epididymal fat pads.

Male Sprague-Dawley rats or male mice were killed, epididymal fat pads were removed, and adipocytes were isolated as described (18). After collagenase digestion, adipocytes were filtered through 20-μ mesh and washed three times with 3% BSA–Krebs-Ringer bicarbonate HEPES buffer (118 mmol/l NaCl, 5 mmol/l KCl, 1.2 mmol/l MgSO4, 2.5 mmol/l CaCl2, 1.2 mmol/l KH2PO4, 5 mmol/l NaHCO3, 30 mmol/l HEPES, and 1 mmol/l pyruvate, pH 7.4).

2-deoxy-d-[3H]-glucose uptake assay.

Isolated adipocytes were incubated with various concentrations of bradykinin and/or NO inhibitors/scavengers/donors for the times indicated. Basal and insulin-stimulated (100 nmol/l for 30 min at 37°C) rates of glucose uptake were assayed as described (18).

Western blotting.

Adipocytes were solubilized by adding 200 μl of lysis buffer (50 mmol/l Tris-HCl, pH 7.5, 1 mmol/l EDTA, 1 mmol/l EGTA, 0.5 mmol/l sodium orthovanadate, 0.1% 2-mercaptoethanol, 1% Triton X-100, 50 mmol/l sodium fluoride, 5 mmol/l sodium pyrophosphate, 10 mmol/l sodium β-glycerophosphate, 0.1 mmol/l phenylmethylsulfonyl fluoride, 1 μg/ml aprotinin, 1 μg/ml pepstatin, 1 μg/ml leupeptin, and 1 μg/ml okadaic acid) per milliliter of cells followed by homogenization. After centrifugation, the fat was removed, and the infranatant was stored at −70°C. Laemmli sample buffer (2×) with 0.006% bromphenol blue was added to equal aliquots of protein (Bradford assay) and boiled for 5 min. For mitogen-activated protein kinase (MAPK) analysis, cellular protein was extracted using boiling Laemmli sample buffer. For GLUT4 analysis, urea sample buffer was used (6.86 mol/l urea, 4.29% SDS, 43 mmol/l Tris-HCl, and 300 mmol/l dithiothreitol). For immunoprecipitation of Akt2/PKBβ, 750 μg of whole cell lysates were incubated with 4 μg of anti-Akt2 antibody at 4°C overnight, followed by addition of 50 μl of protein A-agarose beads for 2 h, and the samples were centrifuged and washed three times. After separation by SDS-PAGE, proteins were transferred onto nitrocellulose membranes, which were blocked with 5% milk- or BSA–Tris-buffered saline with 0.1% Tween for 1 h at room temperature. Membranes were incubated overnight with primary antibodies, washed with Tris-buffered saline with 0.1% Tween and incubated with horseradish peroxidase–conjugated secondary antibody for 1 h at room temperature. Membranes were washed, reacted with LumiGlo chemiluminescence, and exposed to film.

Subcellular fractionation of rat adipocytes.

Adipocytes were fractionated as described previously (19). Briefly, cells were washed, homogenized (250 mmol/l sucrose, 2 mmol/l EDTA, 2.5 mmol/l Tris-HCl, pH 7.4, 10 μg/ml aprotinin, 10 μg/ml leupeptin, and 100 μmol/l phenylmethylsulfonyl fluoride) and then centrifuged at 13,100g for 20 min at 4°C to remove the fat cake. The infranatant was centrifuged at 31,000g for 1 h to yield the low-density microsomes (LDMs). The pellet from the first spin was layered over a sucrose cushion and centrifuged at 75,000g for 1 h. The interphase was removed and spun at 39,000g for 20 min to yield the plasma membrane. The purity and yield of LDM and plasma membrane were similar in control and bradykinin-treated cells. The relative amounts of LDM and plasma membrane GLUT4 were determined by normalizing the intensities of the bands from the immunoblots for the protein yield of each fraction.

Immunofluorescence and confocal microscopy.

Primary adipocytes were electroporated as described previously (20). Briefly, 400 μl of adipocyte suspension (lipocrit of 60%) were added to an equal volume of Dulbecco’s modified Eagle’s medium containing 7.5 μg of pQBI25 plasmid encoding hemagglutinin–GLUT4–green fluorescent protein (GFP) (kindly provided by S.W. Cushman, Yale University, New Haven, CT). Using the Gene Pulser XCell System (BioRad), cells were electroporated in 0.4-cm gap-width cuvettes with two pulses of 25 μF at 800 V followed by one pulse of 1,050 μF at 200 V. Eighteen hours after transfection, the adipocytes were washed, treated or not treated with bradykinin (1 μmol/l) for 1 h and/or insulin (100 nmol/l) for 30 min, and incubated with mouse monoclonal anti-hydroxylamine antibody (1:150) and donkey anti-mouse IgG antibody conjugated with rhodamine red (1:100), fixed in 4% paraformaldehyde in PBS, and mounted on glass slides using Vectashield mounting medium as described (21). Fluorescence was observed using the Leica TCS SP2 Confocal Laser Scanning Microscope and analyzed with Leica Confocal Software.

Induction of iNOS.

iNOS was induced with a single injection of lipopolysaccharide (LPS) (4 mg/kg) (22). Eight hours later, total cell proteins (100 μg) were separated by SDS-PAGE and immunoblotted for iNOS (1:350).

Quantification of concentration of bradykinin.

Media in which the adipocytes were incubated were collected at various times after addition of bradykinin. The bradykinin concentration was determined by an enzyme immunoassay (Peninsula).

Quantification of JNK activity.

Adipocytes, treated or not treated with bradykinin and/or insulin, were lysed. JNK activity was then determined using the kit as described by the manufacturer (Calbiochem).

Statistical analysis.

Values shown are means ± SE. Results were analyzed by ANOVA, and differences were deemed significant at P < 0.05.

Bradykinin enhances insulin-stimulated glucose uptake in rat adipocytes via the B2 receptor.

Whereas treatment of primary rat adipocytes for 30 min with 0–10 μmol/l bradykinin had no effect on basal 2-deoxyglucose (2DG) uptake, insulin-stimulated 2DG uptake was enhanced in a dose-dependent manner, peaking at 1 μmol/l ([pmol · 5 × 108 cells−1 · 3 min−1] control basal 35.4 ± 2.8, insulin 246.2 ± 47.7, bradykinin basal 33.7 ± 5.4, and bradykinin + insulin 374.8 ± 12.7; P < 0.05, insulin vs. bradykinin + insulin) (Fig. 1A). The effect of bradykinin was maintained at 1 and 6 h (Fig. 1B). To determine the duration and reversibility of the effect of bradykinin, the adipocytes were washed after 1 h of incubation. Insulin-stimulated 2DG uptake remained enhanced for 1 h after washout, but this effect was no longer significant at 2 h (Fig. 1C).

Although effective bradykinin concentrations are in the nanomolar range in vivo, bradykinin is subject to degradation by cell-associated kininases. Indeed, the concentration of bradykinin markedly decreased upon incubation with adipocytes from 1 μmol/l to 49 ± 3 nmol/l after 1 h and to 608 ± 52 pmol/l after 6 h. To examine the effect of kininase inhibition, adipocytes were pretreated with either the ACEI captopril (1 μmol/l) or the ACEI/neutral endopeptidase inhibitor omapatrilat (1 μmol/l) for 1 h before the addition of bradykinin. Both captopril and omapatrilat shifted the dose-response curve to the left so that as little as 10 nmol/l bradykinin significantly enhanced insulin-stimulated 2DG uptake. (Fig. 1A).

The presence of the B2 receptor in rat adipocytes was demonstrated by immunoblotting (Fig. 2A). In addition, pretreatment of adipocytes with the B2 receptor antagonist HOE-140 (250 μmol/l) blocked the enhancement by bradykinin of insulin-stimulated glucose uptake (Fig. 2B).

Bradykinin action on insulin-stimulated 2DG uptake involves NO and eNOS.

Bradykinin leads to NO production in endothelial cells, which in turn signals its vasodilatory actions (1113). To investigate whether bradykinin also acts via the generation of NO in adipocytes, we first immunoblotted the three isoforms of NO synthase (NOS): eNOS, nNOS, and iNOS. Under standard conditions, isolated rat adipocytes expressed primarily eNOS, with no nNOS and barely detectable levels of iNOS (Fig. 3A). However, iNOS could be induced by LPS administration after 8 h (Fig. 3A). To probe the role of NO, adipocytes were incubated with and without bradykinin in the presence and absence of either the NOS inhibitor Nω-nitro-l-arginine methyl ester (l-NAME) (1 mmol/l) or the NO scavenger 2-(4-carboxyphenyl)-4,5-dihydro-4,4,5,5-tetramethyl-1H-imidazol-1-oxyl-3-oxide potassium salt (PTIO) (250 μmol/l), followed by assessment of basal and insulin-stimulated 2DG uptake. Both l-NAME and PTIO blocked the enhancement by bradykinin of insulin-stimulated 2DG uptake (Fig. 3B). To test whether NO generation was sufficient to mimic the effect of bradykinin, adipocytes were incubated for 30 min with and without insulin in the absence and presence of one of three NO donors: sodium nitroprusside (SNP), 3-morpholino-sydnonimine, and hydroxylamine. These significantly enhanced insulin-stimulated 2DG uptake comparable to the effect of bradykinin (Fig. 3C). Insulin-stimulated 2DG uptake remained elevated after NO donors for up to 6 h, similar to bradykinin (data not shown). The above data indicate that NO is the mediator of the enhanced insulin-stimulated 2DG uptake by bradykinin.

The eNOS isoform is required for bradykinin action in adipocytes.

The requirement for NO synthesis and the detection of eNOS led us to test the effect of bradykinin in adipocytes lacking eNOS or nNOS. Bradykinin enhanced insulin-stimulated 2DG uptake in adipocytes from nNOS−/− mice and control mice to a similar extent (Fig. 4A). However, although adipocytes from eNOS−/− mice had a slightly increased basal and decreased insulin-stimulated 2DG uptake compared with controls, bradykinin did not alter either basal or insulin-stimulated glucose uptake (Fig. 4B). Because the defect in the action of bradykinin in eNOS−/− mice was presumed to be due to an inability to generate NO, it was predicted that the adipocytes would respond to an NO donor. Indeed, treatment of eNOS−/− adipocytes with hydroxylamine (500 μmol/l) augmented insulin-stimulated glucose transport (Fig. 4B). Thus, the presence of eNOS is critical for bradykinin to enhance insulin-stimulated glucose uptake.

Bradykinin augments insulin-stimulated GLUT4 translocation.

To explore the mechanism by which bradykinin increases insulin-stimulated glucose uptake, GLUT4 translocation was determined. Whereas bradykinin alone minimally increased plasma membrane GLUT4 (NS), it significantly augmented the effect of insulin to induce translocation (GLUT4 content in the plasma membrane as a percentage of total GLUT4 from densitometric analysis: control basal 21.5 ± 4.9, control + insulin 55.6 ± 7.2, bradykinin basal 27.8 ± 8.0, and bradykinin + insulin 83.5 ± 3.9%; P < 0.05 vs. control + insulin) (Fig. 5B).

To confirm these findings, adipocytes were transfected with a hemagglutinin epitope–tagged GLUT4-GFP–expressing cDNA. The hemagglutinin tag is inserted in the exofacial loop of GLUT4. Thus, in nonpermeabilized cells, anti-hemagglutinin will bind only to GLUT4 that is inserted into the plasma membrane (20,21). Imaging confirmed that bradykinin-enhanced GLUT4 translocation (mean fluorescence intensity representing cell surface hemagglutinin-GLUT4-GFP [arbitrary units]: control basal 1.0 ± 0, control + insulin 1.99 ± 0.13, bradykinin basal 1.17 ± 0.015, and bradykinin + insulin 2.57 ± 0.08; P < 0.04 vs. control + insulin (n = 3) (Fig. 5C).

Bradykinin augments insulin signaling in rat adipocytes.

Recruitment of GLUT4 to the plasma membrane by insulin is mediated by a signaling pathway that involves Tyr phosphorylation of IRS-1 and activation of phosphatidylinositol 3-kinase (PI3K) and Akt/PKB (23). Insulin stimulation led to an increase in IRS-1 pY and p85 association (Fig. 6A). Treatment with bradykinin alone did not alter basal levels but significantly enhanced these effects of insulin (Fig. 6A). Similarly, although bradykinin alone had no effect on Akt/PKB phosphorylation on Thr308 and Ser473, insulin-stimulated phosphorylation was significantly augmented (Fig. 6B). Of the three isoforms of Akt/PKB, Akt1/PKBα, Akt2/PKBβ, and Akt3/PKBγ (24), Akt2/PKBβ is involved in insulin-stimulated GLUT4 translocation to the plasma membrane (25,26). Thus, Akt2/PKBβ was immunoprecipitated from whole cell lysates, separated by SDS-PAGE, and immunoblotted for total Akt2/PKBβ and Ser473. Bradykinin significantly enhanced insulin-stimulated Akt2/PKBβ pSer473 phosphorylation (Fig. 6C).

Bradykinin decreases insulin-stimulated phosphorylation of ERK1/2 (p42/p44 MAPK) and JNK.

Apart from the PI3K-Akt/PKB signaling pathway, insulin also leads to the activation of the MAPK pathway (23,27). Thus, we examined the effect of bradykinin ± insulin on the phosphorylation levels of p38 MAPK, ERK1/2 (p42/p44 MAPK), and JNK using phospho-specific antibodies that recognize their activated forms. Insulin did not stimulate the phosphorylation of p38 MAPK in adipocytes, consistent with previous reports (28) (data not shown), whereas the phosphorylation of ERK1/2 and p54 JNK was increased. It was noted that expression of p46 JNK appeared very weak to undetectable in adipocyte lysates. Unexpectedly, in contrast to the findings with Akt/PKB, bradykinin decreased the insulin-stimulated phosphorylation of both ERK1/2 and p54 JNK (Fig. 7A). The functional consequence of decreased phosphorylation was confirmed by the finding of decreased kinase activity in JNK immunoprecipitates (phospho-c-Jun corrected for total JNK [arbitrary units]: control basal 1.0, control + insulin 1.69 ± 0.01, bradykinin basal 0.81 ± 0.06, and bradykinin + insulin 0.96 ± 0.08; P = 0.014, control + insulin vs. bradykinin + insulin) (Fig. 7B).

These somewhat surprising results suggested a potential mechanism by which bradykinin could increase insulin signaling to glucose transport. It has been reported that IRS-1 may be a substrate of JNK (27) and ERK1/2 (29). In particular, phosphorylation of Ser307 of IRS-1 by JNK has been found to be associated with decreased insulin-stimulated IRS-1 Tyr phosphorylation and insulin resistance (27,30). Exposure to bradykinin significantly decreased IRS-1 Ser307 phosphorylation in response to insulin, accompanied by an increase in IRS-1 pY (Fig. 7C). Phosphorylation of Ser616 of IRS-1 by ERK1/2 also reduces insulin signaling (29). Bradykinin decreased insulin-stimulated phosphorylation of IRS-1 Ser616 (Fig. 7B). These data suggested that inhibition of JNK and/or ERK activity and subsequent IRS-1 Ser307 and Ser616 phosphorylation, respectively, could mediate the enhancement of insulin signaling by bradykinin.

To test whether inhibition of either JNK or ERK could replicate the effect of bradykinin to enhance glucose uptake, adipocytes were preincubated with either the MEK inhibitor PD-98059 or the JNK inhibitor SP-600125. Inhibition of JNK but not of MEK increased insulin-stimulated 2DG uptake comparable to bradykinin. Coincubation of the JNK inhibitor with bradykinin did not result in any further increase in insulin-stimulated 2DG uptake (Fig. 7C).

The JNK1 isoform appears to be responsible for the phosphorylation of IRS-1 Ser307 and induction of insulin resistance. Thus, JNK1−/− but not JNK2−/− mice are insulin sensitive and resistant to high-fat diet–induced insulin resistance (31,32). Adipocytes from JNK1−/− mice showed increased insulin-stimulated glucose uptake, and this was not enhanced by bradykinin. In contrast, JNK2−/− adipocytes displayed sensitivity to insulin and bradykinin similar to that of controls (Fig. 7D). Taken together, these data suggest that bradykinin enhances insulin sensitivity by inhibition of JNK activation.

Bradykinin signals via NO to decrease insulin-stimulated JNK phosphorylation.

To confirm the role of NO in bradykinin-mediated inhibition of JNK, adipocytes were treated with either bradykinin or the NO donor, hydroxylamine, in the presence and absence of insulin. Both bradykinin and hydroxylamine significantly reduced insulin-stimulated JNK phosphorylation (Fig. 8A). In addition, compared with controls, the basal level of JNK phosphorylation was significantly greater in adipocytes from eNOS−/− mice, and did not decrease upon bradykinin treatment (Fig. 8B). These data indicate that signaling via NO is involved in bradykinin-mediated inhibition of JNK phosphorylation and activation.

Bradykinin has been implicated as a vasoactive factor associated with increased insulin sensitivity (9,15,16). Both ACEIs and bradykinin improved insulin action in skeletal muscle of Zucker fatty rats in vivo (16,33) and insulin-stimulated 2DG uptake ex vivo and in vitro (3436). In the present study, addition of bradykinin directly to isolated adipocytes increased insulin-stimulated 2DG uptake in a dose- and time-dependent manner. This effect of bradykinin was mediated through its B2 receptor, in agreement with one previous study (35), and is consistent with the observation that mice lacking the B2 receptor display insulin resistance (37). In another study, HOE-140, the B2 receptor antagonist, also blunted the ability of ACEIs to improve insulin sensitivity (36). Together, these findings strongly suggest that the major mechanism by which ACEI improve insulin action is via increased levels of bradykinin rather than decreased production of AngII.

Our results demonstrate that NO is required for bradykinin to enhance insulin action in adipocytes. A number of studies investigating the putative role of NO in insulin action have focused on the vascular system. Treatment of rats with the NOS inhibitor l-NG-monomethyl-l-arginine resulted in hypertension and insulin resistance (17). Furthermore, mice with a targeted deletion of eNOS (eNOS−/−) also manifest hypertension and insulin resistance (38). Thus, it has been postulated that NO improves insulin action in muscle in vivo as a result of increased blood flow (17,39). In the present study, we examined the role of NO independent of the vasculature. In isolated rat adipocytes, three different NO donors replicated the effect of bradykinin to enhance the action of insulin. In addition, inhibition of NOS or scavenging NO blocked the ability of bradykinin to augment insulin-stimulated 2DG uptake. In contrast to these data, one previous study reported that in 3T3-L1 adipocytes, a NO donor stimulated glucose uptake independent of insulin and its downstream effectors, IRS-1 and Akt/PKB (40). The concentration of SNP used in that study was >150-fold that used here (50 mmol/l vs. 300–400 μmol/l). Either the cell culture model or the NO concentration or both may have contributed to the discrepancies observed.

The importance of eNOS as a putative mediator of bradykinin action in adipocytes was demonstrated by the finding that bradykinin failed to increase insulin-stimulated 2DG uptake in adipocytes from eNOS−/− mice in contrast to wild-type and nNOS−/− mice. However, the NO donor hydroxylamine significantly enhanced insulin-stimulated glucose uptake in these adipocytes. Thus, NO production by eNOS is critical for bradykinin to enhance insulin sensitivity in adipocytes.

As opposed to eNOS, iNOS can produce large amounts of NO over long periods of time, which may be detrimental (41). Chronic exposure to high concentrations of NO or induction of iNOS by cytokine treatment of cultured skeletal muscle cells has been associated with insulin resistance (42), and iNOS−/− mice are protected from insulin resistance induced by a high-fat diet (43). In agreement with these reports, we found that incubation of isolated adipocytes with very high concentrations of NO donors (greater than those in Fig. 3C) resulted in decreased insulin-stimulated 2DG uptake (data not shown).

Investigation of the mechanism of cross-talk between bradykinin and the insulin signaling pathway revealed that the enhancement of insulin-stimulated glucose transport by bradykinin was associated with increases of 1) GLUT4 translocation from LDMs to plasma membrane, 2) IRS-1 pY, 3) IRS-1 association with the p85 regulatory subunit of PI3K, and 4) phosphorylation of Thr308 and Ser473 of Akt/PKB. These findings extend previous reports of increased Tyr phosphorylation of IRS-1 and GLUT4 translocation by insulin in the presence of bradykinin (35,44).

As part of the investigation of insulin signaling in the presence of bradykinin, we examined the MAPK family of enzymes. Bradykinin itself can activate ERK1/2 in mesangial cells (45), SW-480 cells (46), and HEK 293 cells (47). In contrast to these reports and our findings of enhanced IRS-1 and Akt/PKB phosphorylation, bradykinin caused a reduction in insulin-stimulated phosphorylation of both ERK1/2 and JNK. The effect of bradykinin on the MAPK family of enzymes in adipocytes had not been previously reported. As seen in Fig. 7A, phosphorylation of ERK1/2 and JNK was detectable in unstimulated cells, probably due to the release of a variety of inflammatory mediators, including TNF-α and interleukin-1α and interleukin-6, by the adipocyte isolation procedure (48). These basal levels of phosphorylation were reduced but not entirely abolished by washing and preincubating the cells in 3% BSA–Krebs-Ringer bicarbonate HEPES buffer, pH 7.4, for 4–5 h at 37°C before experiments.

There is evidence that JNK activation and subsequent phosphorylation of Ser307 of IRS-1 results in decreased Tyr phosphorylation of IRS-1 and impaired insulin signaling (27,30). These events may occur in response to cytokines such as TNF-α (30), to free fatty acids (49), and to insulin itself (30). In the present study, we observed that insulin-stimulated phosphorylation of Ser307 and Ser616, a site of ERK phosphorylation, of IRS-1 was decreased by bradykinin. Whereas inhibition of MEK had no effect on glucose uptake, inhibition of JNK potentiated insulin-stimulated 2DG uptake to an extent similar to bradykinin. To confirm these findings, we examined glucose uptake in adipocytes isolated from JNK1−/− mice, which have enhanced insulin sensitivity associated with reduced IRS-1 Ser307 phosphorylation (31). These adipocytes exhibited enhanced insulin sensitivity and, in contrast to JNK2−/− and control strains, there was no potentiation of insulin-stimulated 2DG uptake by bradykinin. Inhibitor of IκB-kinase β is an inflammatory cytokine-activated kinase that also has been implicated in insulin resistance by promoting IRS-1 Ser phosphorylation (50,51). However, we did not detect any change in inhibitor of IκB-kinase β phosphorylation (phospho-specific antibodies from cell signaling) upon treatment with insulin and/or bradykinin under the conditions of these experiments (not shown).

The link between bradykinin-mediated NO signaling to enhance glucose uptake and to inhibit JNK was confirmed by the observation that the NO donor hydroxylamine also blunted insulin-stimulated JNK phosphorylation. Furthermore, adipocytes from eNOS−/− mice displayed an elevated basal level of phosphorylated JNK, which was not reduced by bradykinin. Together, these data indicate that bradykinin potentiates insulin-stimulated glucose uptake via the production of NO and subsequent inhibition of JNK activation. Although the mechanism by which NO inhibits JNK activity is not clear, some of its actions are exerted by S-nitrosylation of proteins. Endogenous NO has been reported to suppress JNK through such a thiol-redox mechanism (52), suggesting one possible mechanism for bradykinin action.

In summary, this study demonstrates that bradykinin significantly enhances insulin-stimulated glucose transport in adipocytes via a signaling pathway that involves eNOS and the generation of NO independent of blood flow. Investigation of the mechanism of cross-talk revealed that bradykinin decreases insulin stimulation of JNK, which is associated with reduced IRS-1 Ser307 phosphorylation and increased insulin stimulation of IRS-1 Tyr phosphorylation, p85 association, Akt/PKB phosphorylation, and GLUT4 translocation. These data present a novel mechanism that may account for the sensitization to insulin observed in the presence of ACEIs and bradykinin signaling. Elucidating the role of this regulatory pathway in normal physiology, in insulin-resistant states, and in the presence of currently used pharmacological agents such as ACEIs and ARBs will be of interest.

FIG. 1.

Bradykinin augments insulin-stimulated glucose uptake in rat adipocytes. A: Isolated rat adipocytes were pretreated with captopril, omapatrilat, or vehicle for 1 h, followed by bradykinin ± insulin for 30 min, and 2DG uptake was assayed. B: Adipocytes were incubated with bradykinin or vehicle for 1 h or 6 h ± insulin for the final 30 min. C: Adipocytes were incubated ± bradykinin for 1 h and washed, and 2DG uptake ± insulin was assayed at t = 0, 1, and 2 h after washing. Values are means ± SE (n = 5). *P < 0.02 vs. insulin without bradykinin.

FIG. 1.

Bradykinin augments insulin-stimulated glucose uptake in rat adipocytes. A: Isolated rat adipocytes were pretreated with captopril, omapatrilat, or vehicle for 1 h, followed by bradykinin ± insulin for 30 min, and 2DG uptake was assayed. B: Adipocytes were incubated with bradykinin or vehicle for 1 h or 6 h ± insulin for the final 30 min. C: Adipocytes were incubated ± bradykinin for 1 h and washed, and 2DG uptake ± insulin was assayed at t = 0, 1, and 2 h after washing. Values are means ± SE (n = 5). *P < 0.02 vs. insulin without bradykinin.

Close modal
FIG. 2.

Bradykinin increases insulin-stimulated glucose uptake via the B2 receptor (B2R). A: Protein (20 μg) from adipocyte and cultured vascular smooth muscle cell (VSMC) lysates was immunoblotted for B2 receptor and actin. B: Adipocytes were pretreated with the B2 receptor antagonist HOE-140 for 20 min, followed by bradykinin ± insulin. Values are means ± SE (n = 5). *P < 0.02 vs. insulin without bradykinin.

FIG. 2.

Bradykinin increases insulin-stimulated glucose uptake via the B2 receptor (B2R). A: Protein (20 μg) from adipocyte and cultured vascular smooth muscle cell (VSMC) lysates was immunoblotted for B2 receptor and actin. B: Adipocytes were pretreated with the B2 receptor antagonist HOE-140 for 20 min, followed by bradykinin ± insulin. Values are means ± SE (n = 5). *P < 0.02 vs. insulin without bradykinin.

Close modal
FIG. 3.

NO is required for bradykinin action. A: Adipocyte lysates from saline-injected rats were immunoblotted for eNOS, nNOS, and iNOS and from LPS-injected rats for iNOS. Representative immunoblot (n = 3). B: Adipocytes were preincubated with l-NAME, PTIO, or vehicle for 1 h and then with bradykinin ± insulin (n = 6). C: Adipocytes were incubated (30 min) with 3-morpholino-sydnonimine (SIN-1), SNP, and hydroxylamine (HA) ± insulin. Values are means ± SE (n = 5). *P < 0.05 vs. insulin without bradykinin.

FIG. 3.

NO is required for bradykinin action. A: Adipocyte lysates from saline-injected rats were immunoblotted for eNOS, nNOS, and iNOS and from LPS-injected rats for iNOS. Representative immunoblot (n = 3). B: Adipocytes were preincubated with l-NAME, PTIO, or vehicle for 1 h and then with bradykinin ± insulin (n = 6). C: Adipocytes were incubated (30 min) with 3-morpholino-sydnonimine (SIN-1), SNP, and hydroxylamine (HA) ± insulin. Values are means ± SE (n = 5). *P < 0.05 vs. insulin without bradykinin.

Close modal
FIG. 4.

Requirement of eNOS for bradykinin action on insulin-stimulated glucose uptake. Adipocytes were isolated from control (nNOS+/+) and nNOS−/− control (eNOS+/+) mice (A) and eNOS−/− mice (B) and treated with vehicle, bradykinin, or hydroxylamine (HA) (30 min) ± insulin. Values are means ± SE (n = 4). A: *P < 0.05 vs. control insulin or nNOS−/− insulin without bradykinin. B: *P < 0.05 vs. control insulin without bradykinin or HA; **P < 0.01 vs. eNOS−/− insulin with or without bradykinin; #P < 0.04 vs. basal eNOS−/−.

FIG. 4.

Requirement of eNOS for bradykinin action on insulin-stimulated glucose uptake. Adipocytes were isolated from control (nNOS+/+) and nNOS−/− control (eNOS+/+) mice (A) and eNOS−/− mice (B) and treated with vehicle, bradykinin, or hydroxylamine (HA) (30 min) ± insulin. Values are means ± SE (n = 4). A: *P < 0.05 vs. control insulin or nNOS−/− insulin without bradykinin. B: *P < 0.05 vs. control insulin without bradykinin or HA; **P < 0.01 vs. eNOS−/− insulin with or without bradykinin; #P < 0.04 vs. basal eNOS−/−.

Close modal
FIG. 5.

Bradykinin enhances insulin-stimulated GLUT4 translocation. Adipocytes were treated with bradykinin for 1 h ± insulin (final 40 min), and LDM and plasma membrane fractions were isolated. A: Representative immunoblot. B: The total amount of GLUT4 (LDM + plasma membrane) was set at 100%. Values are means ± SE (n = 5) and represent the percentage of total GLUT4 in each fraction. *P < 0.02 vs. insulin without bradykinin. C: Adipocytes were transfected with a hemagglutinin-GLUT4-GFP expression vector by electroporation. After overnight incubation, cells were treated with and without bradykinin ± insulin. A representative set of cells is shown. RhR, rhodamine red signal (see text for details).

FIG. 5.

Bradykinin enhances insulin-stimulated GLUT4 translocation. Adipocytes were treated with bradykinin for 1 h ± insulin (final 40 min), and LDM and plasma membrane fractions were isolated. A: Representative immunoblot. B: The total amount of GLUT4 (LDM + plasma membrane) was set at 100%. Values are means ± SE (n = 5) and represent the percentage of total GLUT4 in each fraction. *P < 0.02 vs. insulin without bradykinin. C: Adipocytes were transfected with a hemagglutinin-GLUT4-GFP expression vector by electroporation. After overnight incubation, cells were treated with and without bradykinin ± insulin. A representative set of cells is shown. RhR, rhodamine red signal (see text for details).

Close modal
FIG. 6.

Bradykinin augments insulin-stimulated IRS-1 and Akt/PKB phosphorylation. Adipocytes were treated as in the legend to Fig. 5. A and B: Representative immunoblots and means ± SE (n = 4–5). C: Akt2/PKBβ was immunoprecipitated from whole cell lysates. Representative immunoblots of Akt2/PKBβ and pSer473-Akt2/PKBβ. (n = 3). The intensities of the bands from phospho-specific immunoblots were corrected for those of total kinases. Values are means ± SE. *P < 0.03 vs. insulin without bradykinin.

FIG. 6.

Bradykinin augments insulin-stimulated IRS-1 and Akt/PKB phosphorylation. Adipocytes were treated as in the legend to Fig. 5. A and B: Representative immunoblots and means ± SE (n = 4–5). C: Akt2/PKBβ was immunoprecipitated from whole cell lysates. Representative immunoblots of Akt2/PKBβ and pSer473-Akt2/PKBβ. (n = 3). The intensities of the bands from phospho-specific immunoblots were corrected for those of total kinases. Values are means ± SE. *P < 0.03 vs. insulin without bradykinin.

Close modal
FIG. 7.

Bradykinin decreases ERK1/2 and JNK phosphorylation. Adipocytes were treated as in the legend to Fig. 5. A: Representative immunoblots and means ± SE (n = 5). B: Immunoprecipitated JNK activity was assayed, and total JNK and phospho–c-Jun were immunoblotted (n = 5) (see text for quantification). C: Representative immunoblots and means ± SE (n = 3), except pSer616-IRS-1 (n = 1). A and C: The intensities of the phospho-specific antibody bands were corrected for total kinases. *P < 0.04 vs. insulin without bradykinin. D: Adipocytes were pretreated with either SP-600125, PD-98059, or vehicle, followed by bradykinin ± insulin. 2DG uptake was assayed. Values are means ± SE (n = 4). *P < 0.03 vs. insulin without bradykinin. E: Adipocytes from JNK1−/−, control (JNK1+/+), JNK2−/−, and C57BL/6J mice were treated with bradykinin ± insulin. Values are mean ± SE (n = 4). *P < 0.04 vs. control insulin.

FIG. 7.

Bradykinin decreases ERK1/2 and JNK phosphorylation. Adipocytes were treated as in the legend to Fig. 5. A: Representative immunoblots and means ± SE (n = 5). B: Immunoprecipitated JNK activity was assayed, and total JNK and phospho–c-Jun were immunoblotted (n = 5) (see text for quantification). C: Representative immunoblots and means ± SE (n = 3), except pSer616-IRS-1 (n = 1). A and C: The intensities of the phospho-specific antibody bands were corrected for total kinases. *P < 0.04 vs. insulin without bradykinin. D: Adipocytes were pretreated with either SP-600125, PD-98059, or vehicle, followed by bradykinin ± insulin. 2DG uptake was assayed. Values are means ± SE (n = 4). *P < 0.03 vs. insulin without bradykinin. E: Adipocytes from JNK1−/−, control (JNK1+/+), JNK2−/−, and C57BL/6J mice were treated with bradykinin ± insulin. Values are mean ± SE (n = 4). *P < 0.04 vs. control insulin.

Close modal
FIG. 8.

NO decreases insulin-stimulated JNK phosphorylation. A: Adipocytes were treated with bradykinin or hydroxylamine (HA) (500 mmol/l) ± insulin. Representative immunoblots of JNK and phospho-JNK. (n = 3). *P < 0.05 vs. insulin without bradykinin; **P < 0.03 vs. basal without bradykinin. B: Adipocytes from control (eNOS+/+) and eNOS−/− mice were treated as in the legend to Fig. 7. Representative immunoblots of JNK and phospho-JNK. Intensities of phosphorylated bands were corrected for total kinases. Values are means ± SE (n = 3). *P < 0.04 vs. insulin without bradykinin; **P < 0.04 vs. basal control (eNOS+/+).

FIG. 8.

NO decreases insulin-stimulated JNK phosphorylation. A: Adipocytes were treated with bradykinin or hydroxylamine (HA) (500 mmol/l) ± insulin. Representative immunoblots of JNK and phospho-JNK. (n = 3). *P < 0.05 vs. insulin without bradykinin; **P < 0.03 vs. basal without bradykinin. B: Adipocytes from control (eNOS+/+) and eNOS−/− mice were treated as in the legend to Fig. 7. Representative immunoblots of JNK and phospho-JNK. Intensities of phosphorylated bands were corrected for total kinases. Values are means ± SE (n = 3). *P < 0.04 vs. insulin without bradykinin; **P < 0.04 vs. basal control (eNOS+/+).

Close modal

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked “advertisement” in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

This work was funded by a grant from the Canadian Diabetes Association (to I.G.F.) and by Grant FRN 38009 from the Canadian Institutes of Health Research (to I.G.F.). K.M.B. was supported by a Canadian Diabetes Association Doctoral Scholarship and a Banting and Best Diabetes Centre–Novo Nordisk Studentship.

1.
Beck-Nielsen H, Groop LC: Metabolic and genetic characterization of prediabetic states: sequence of events leading to non-insulin-dependent diabetes mellitus.
J Clin Invest
94
:
1714
–1721,
1994
2.
Virkamaki A, Ueki K, Kahn CR: Protein-protein interaction in insulin signaling and the molecular mechanisms of insulin resistance.
J Clin Invest
103
:
931
–943,
1999
3.
Kershaw EE, Flier JS: Adipose tissue as an endocrine organ.
J Clin Endocrinol Metab
89
:
2548
–2556,
2004
4.
Natali A, Ferrannini E: Hypertension, insulin resistance, and the metabolic syndrome.
Endocrinol Metab Clin North Am
33
:
417
–429,
2004
5.
Dandona P, Aljada A, Chaudhuri A, Mohanty P, Garg R: Metabolic syndrome: a comprehensive perspective based on interactions between obesity, diabetes, and inflammation.
Circulation
111
:
1448
–1454,
2005
6.
Yusuf S, Sleight P, Pogue J, Bosch J, Davies R, Dagenais G: Effects of an angiotensin-converting-enzyme inhibitor, ramipril, on cardiovascular events in high-risk patients: the Heart Outcomes Prevention Evaluation Study Investigators.
N Engl J Med
342
:
145
–153,
2000
7.
McMurray J, Fraser DM: Captopril, enalapril, and blood glucose.
Lancet
1
:
1035
,
1986
8.
Torlone E, Rambotti AM, Perriello G, Botta G, Santeusanio F, Brunetti P, Bolli GB: ACE-inhibition increases hepatic and extrahepatic sensitivity to insulin in patients with type 2 (non-insulin-dependent) diabetes mellitus and arterial hypertension.
Diabetologia
34
:
119
–125,
1991
9.
Uehara M, Kishikawa H, Isami S, Kisanuki K, Ohkubo Y, Miyamura N, Miyata T, Yano T, Shichiri M: Effect on insulin sensitivity of angiotensin converting enzyme inhibitors with or without a sulphydryl group: bradykinin may improve insulin resistance in dogs and humans.
Diabetologia
37
:
300
–307,
1994
10.
Takahashi N, Hagaman JR, Kim HS, Smithies O: Minireview: computer simulations of blood pressure regulation by the renin-angiotensin system.
Endocrinology
144
:
2184
–2190,
2003
11.
Regoli D, Calo G, Rizzi A, Bogoni G, Gobeil F, Campobasso C, Mollica G, Beani L: Bradykinin receptors and receptor ligands (with special emphasis on vascular receptors).
Regul Pept
65
:
83
–89,
1996
12.
Harris MB, Ju H, Venema VJ, Liang H, Zou R, Michell BJ, Chen ZP, Kemp BE, Venema RC: Reciprocal phosphorylation and regulation of endothelial nitric-oxide synthase in response to bradykinin stimulation.
J Biol Chem
276
:
16587
–16591,
2001
13.
Farhy RD, Carretero OA, Ho KL, Scicli AG: Role of kinins and nitric oxide in the effects of angiotensin converting enzyme inhibitors on neointima formation.
Circ Res
72
:
1202
–1210,
1993
14.
Karlsson C, Lindell K, Ottosson M, Sjostrom L, Carlsson B, Carlsson LM: Human adipose tissue expresses angiotensinogen and enzymes required for its conversion to angiotensin II.
J Clin Endocrinol Metab
83
:
3925
–3929,
1998
15.
Carvalho CR, Thirone AC, Gontijo JA, Velloso LA, Saad MJ: Effect of captopril, losartan, and bradykinin on early steps of insulin action.
Diabetes
46
:
1950
–1957,
1997
16.
Henriksen EJ, Jacob S, Augustin HJ, Dietze GJ: Glucose transport activity in insulin-resistant rat muscle: effects of angiotensin-converting enzyme inhibitors and bradykinin antagonism.
Diabetes
45 (Suppl. 1)
:
S125
–S128,
1996
17.
Baron AD, Zhu JS, Marshall S, Irsula O, Brechtel G, Keech C: Insulin resistance after hypertension induced by the nitric oxide synthesis inhibitor l-NMMA in rats.
Am J Physiol
269
:
E709
–E715,
1995
18.
Tang S, Le-Tien H, Goldstein BJ, Shin P, Lai R, Fantus IG: Decreased in situ insulin receptor dephosphorylation in hyperglycemia-induced insulin resistance in rat adipocytes.
Diabetes
50
:
83
–90,
2001
19.
Jarett L: Subcellular fractionation of adipocytes.
Methods Enzymol
31
:
60
–71,
1974
20.
Chen H, Wertheimer SJ, Lin CH, Katz SL, Amrein KE, Burn P, Quon MJ: Protein-tyrosine phosphatases PTP1B and syp are modulators of insulin-stimulated translocation of GLUT4 in transfected rat adipose cells.
J Biol Chem
272
:
8026
–8031,
1997
21.
Dawson K, Aviles-Hernandez A, Cushman SW, Malide D: Insulin-regulated trafficking of dual-labeled glucose transporter 4 in primary rat adipose cells.
Biochem Biophys Res Commun
287
:
445
–454,
2001
22.
Ribiere C, Jaubert AM, Gaudiot N, Sabourault D, Marcus ML, Boucher JL, Denis-Henriot D, Giudicelli Y: White adipose tissue nitric oxide synthase: a potential source for NO production.
Biochem Biophys Res Commun
222
:
706
–712,
1996
23.
Saltiel A, Kahn, CR.: Insulin signaling and regulation of glucose and lipid metabolism.
Nature
414
:
799
–806,
2001
24.
Marte BM, Downward J: PKB/Akt: connecting phosphoinositide 3-kinase to cell survival and beyond.
Trends Biochem Sci
22
:
355
–358,
1997
25.
Cho H, Mu J, Kim JK, Thorvaldsen JL, Chu Q, Crenshaw EB 3rd, Kaestner KH, Bartolomei MS, Shulman GI, Birnbaum MJ: Insulin resistance and a diabetes mellitus-like syndrome in mice lacking the protein kinase Akt2 (PKB β).
Science
292
:
1728
–1731,
2001
26.
Bae SS, Cho H, Mu J, Birnbaum MJ: Isoform-specific regulation of insulin-dependent glucose uptake by Akt/protein kinase B.
J Biol Chem
278
:
49530
–49536,
2003
27.
Aguirre V, Uchida T, Yenush L, Davis R, White MF: The c-Jun NH2-terminal kinase promotes insulin resistance during association with insulin receptor substrate-1 and phosphorylation of Ser307.
J Biol Chem
275
:
9047
–9054,
2000
28.
Carlson CJ, Koterski S, Sciotti RJ, Poccard GB, Rondinone CM: Enhanced basal activation of mitogen-activated protein kinases in adipocytes from type 2 diabetes: potential role of p38 in the downregulation of GLUT4 expression.
Diabetes
52
:
634
–641,
2003
29.
De Fea K, Roth RA: Modulation of insulin receptor substrate-1 tyrosine phosphorylation and function by mitogen-activated protein kinase.
J Biol Chem
272
:
31400
–31406,
1997
30.
Rui L, Aguirre V, Kim JK, Shulman GI, Lee A, Corbould A, Dunaif A, White MF: Insulin/IGF-1 and TNF-α stimulate phosphorylation of IRS-1 at inhibitory Ser307 via distinct pathways.
J Clin Invest
107
:
181
–189,
2001
31.
Hirosumi J, Tuncman G, Chang L, Gorgun CZ, Uysal KT, Maeda K, Karin M, Hotamisligil GS: A central role for JNK in obesity and insulin resistance.
Nature
420
:
333
–336,
2002
32.
Jaeschke A, Czech MP, Davis RJ: An essential role of the JIP1 scaffold protein for JNK activation in adipose tissue.
Genes Dev
18
:
1976
–1980,
2004
33.
Henriksen EJ, Jacob S, Kinnick TR, Youngblood EB, Schmit MB, Dietze GJ: ACE inhibition and glucose transport in insulin resistant muscle: roles of bradykinin and nitric oxide.
Am J Physiol
277
:
R332
–R336,
1999
34.
Shiuchi T, Cui TX, Wu L, Nakagami H, Takeda-Matsubara Y, Iwai M, Horiuchi M: ACE inhibitor improves insulin resistance in diabetic mouse via bradykinin and NO.
Hypertension
40
:
329
–334,
2002
35.
Isami S, Kishikawa H, Araki E, Uehara M, Kaneko K, Shirotani T, Todaka M, Ura S, Motoyoshi S, Matsumoto K, Miyamura N, Shichiri M: Bradykinin enhances GLUT4 translocation through the increase of insulin receptor tyrosine kinase in primary adipocytes: evidence that bradykinin stimulates the insulin signalling pathway.
Diabetologia
39
:
412
–420,
1996
36.
Kudoh A, Matsuki A: Effects of angiotensin-converting enzyme inhibitors on glucose uptake.
Hypertension
36
:
239
–244,
2000
37.
Duka I, Shenouda S, Johns C, Kintsurashvili E, Gavras I, Gavras H: Role of the B2 receptor of bradykinin in insulin sensitivity.
Hypertension
38
:
1355
–1360,
2001
38.
Duplain H, Burcelin R, Sartori C, Cook S, Egli M, Lepori M, Vollenweider P, Pedrazzini T, Nicod P, Thorens B, Scherrer U: Insulin resistance, hyperlipidemia, and hypertension in mice lacking endothelial nitric oxide synthase.
Circulation
104
:
342
–345,
2001
39.
Sondergaard HM, Bottcher M, Schmitz O, Nielsen TT, Botker HE: Insulin-stimulated myocardial glucose uptake and the relation to perfusion and the nitric oxide system.
J Vasc Res
41
:
38
–45,
2004
40.
Tanaka T, Nakatani K, Morioka K, Urakawa H, Maruyama N, Kitagawa N, Katsuki A, Araki-Sasaki R, Hori Y, Gabazza EC, Yano Y, Wada H, Nobori T, Sumida Y, Adachi Y: Nitric oxide stimulates glucose transport through insulin-independent GLUT4 translocation in 3T3–L1 adipocytes.
Eur J Endocrinol
149
:
61
–67,
2003
41.
Xie Q, Nathan C: The high-output nitric oxide pathway: role and regulation.
J Leukoc Biol
56
:
576
–582,
1994
42.
Bedard S, Marcotte B, Marette A: Cytokines modulate glucose transport in skeletal muscle by inducing the expression of inducible nitric oxide synthase.
Biochem J
325
:
487
–493,
1997
43.
Perreault M, Marette A: Targeted disruption of inducible nitric oxide synthase protects against obesity-linked insulin resistance in muscle.
Nat Med
7
:
1138
–1143,
2001
44.
Miyata T, Taguchi T, Uehara M, Isami S, Kishikawa H, Kaneko K, Araki E, Shichiri M: Bradykinin potentiates insulin-stimulated glucose uptake and enhances insulin signal through the bradykinin B2 receptor in dog skeletal muscle and rat L6 myoblasts.
Eur J Endocrinol
138
:
344
–352,
1998
45.
Jaffa AA, Miller BS, Rosenzweig SA, Naidu PS, Velarde V, Mayfield RK: Bradykinin induces tubulin phosphorylation and nuclear translocation of MAP kinase in mesangial cells.
Am J Physiol
273
:
F916
–F924,
1997
46.
Graness A, Adomeit A, Heinze R, Wetzker R, Liebmann C: A novel mitogenic signaling pathway of bradykinin in the human colon carcinoma cell line SW-480 involves sequential activation of a Gq/11 protein, phosphatidylinositol 3-kinase β, and protein kinase Cε.
J Biol Chem
273
:
32016
–32022,
1998
47.
Naraba H, Ueno A, Kosugi Y, Yoshimura M, Murakami M, Kudo I, Oh-ishi S: Agonist stimulation of B1 and B2 kinin receptors causes activation of the MAP kinase signaling pathway, resulting in the translocation of AP-1 in HEK 293 cells.
FEBS Lett
435
:
96
–100,
1998
48.
Ruan H, Zarnowski MJ, Cushman SW, Lodish HF: Standard isolation of primary adipose cells from mouse epididymal fat pads induces inflammatory mediators and down-regulates adipocyte genes.
J Biol Chem
278
:
47585
–47593,
2003
49.
Yu C, Chen Y, Cline GW, Zhang D, Zong H, Wang Y, Bergeron R, Kim JK, Cushman SW, Cooney GJ, Atcheson B, White MF, Kraegen EW, Shulman GI: Mechanism by which fatty acids inhibit insulin activation of insulin receptor substrate-1 (IRS-1)-associated phosphatidylinositol 3-kinase activity in muscle.
J Biol Chem
277
:
50230
–50236,
2002
50.
Yuan M, Konstantopoulos N, Lee J, Hansen L, Li AZW, Karin M, Shoelson SE: Reversal of obesity-and diet-induced insulin resistance with salicylates or targeted disruption of IKKβ.
Science
293
:
1673
–1677,
2001
51.
Gao Z, Hwang D, Bataille F, LeFevre M, York D, Quon MJ, Ye J: Serine phosphorylation of insulin receptor substrate 1 by inhibitor κB kinase complex.
J Biol Chem
277
:
48115
–18121,
2002
52.
Park HS, Huh SH, Kim MS, Lee SH, Choi EJ: Nitric oxide negatively regulates c-Jun N-terminal kinase/stress-activated protein kinase by means of S-nitrosylation.
Proc Natl Acad Sci U S A
97
:
14382
–14387,
2000