21 research outputs found

    Drag reduction by polyethylene glycol in the tail arterial bed of normotensive and hypertensive rats

    Get PDF
    This study was designed to evaluate the effect of drag reducer polymers (DRP) on arteries from normotensive (Wistar) and spontaneously hypertensive rats (SHR). Polyethylene glycol (PEG 4000 at 5000 ppm) was perfused in the tail arterial bed with (E+) and without endothelium (E-) from male, adult Wistar (N = 14) and SHR (N = 13) animals under basal conditions (constant flow at 2.5 mL/min). In these preparations, flow-pressure curves (1.5 to 10 mL/min) were constructed before and 1 h after PEG 4000 perfusion. Afterwards, the tail arterial bed was fixed and the internal diameters of the arteries were then measured by microscopy and drag reduction was assessed based on the values of wall shear stress (WSS) by computational simulation. In Wistar and SHR groups, perfusion of PEG 4000 significantly reduced pulsatile pressure (Wistar/E+: 17.5 ± 2.8; SHR/E+: 16.3 ± 2.7%), WSS (Wistar/E+: 36; SHR/E+: 40%) and the flow-pressure response. The E- reduced the effects of PEG 4000 on arteries from both groups, suggesting that endothelial damage decreased the effect of PEG 4000 as a DRP. Moreover, the effects of PEG 4000 were more pronounced in the tail arterial bed from SHR compared to Wistar rats. In conclusion, these data demonstrated for the first time that PEG 4000 was more effective in reducing the pressure-flow response as well as WSS in the tail arterial bed of hypertensive than of normotensive rats and these effects were amplified by, but not dependent on, endothelial integrity. Thus, these results show an additional mechanism of action of this polymer besides its mechanical effect through the release and/or bioavailability of endothelial factors

    Effects of small doses of ouabain on the arterial blood pressure of anesthetized hypertensive and normotensive rats

    Get PDF
    Ouabain increases vascular resistance and may induce hypertension by inhibiting the Na+ pump. The effects of 0.18 and 18 µg/kg, and 1.8 mg/kg ouabain pretreatment on the phenylephrine (PHE; 0.1, 0.25 and 0.5 µg, in bolus)-evoked pressor responses were investigated using anesthetized normotensive (control and uninephrectomized) and hypertensive (1K1C and DOCA-salt treated) rats. Treatment with 18 µg/kg ouabain increased systolic and diastolic blood pressure in all groups studied. However, the magnitude of this increase was larger for the hypertensive 1K1C and DOCA-salt rats than for normotensive animals, while the pressor effect of 0.18 µg/kg ouabain was greater only in DOCA-salt rats. A very large dose (1.8 mg/kg) produced toxic effects on the normotensive control but not on uninephrectomized or 1K1C rats. Rat tail vascular beds were perfused to analyze the effects of 10 nM ouabain on the pressor response to PHE. In all animals, 10 nM ouabain increased the PHE pressor response, but this increase was larger in hypertensive DOCA-salt rats than in normotensive and 1K1C rats. Results suggested that a) increases in diastolic blood pressure induced by 18 µg/kg ouabain were larger in hypertensive than normotensive rats; b) in DOCA-salt rats, smaller ouabain doses had a stronger effect than in other groups; c) hypertensive and uninephrectomized rats were less sensitive to toxic doses of ouabain, and d) after treatment with 10 nM ouabain isolated tail vascular beds from DOCA-salt rats were more sensitive to the pressor effect of PHE than those from normotensive and 1K1C hypertensive rats. These data suggest that very small doses of ouabain, which might produce nanomolar plasma concentrations, enhance pressor reactivity in DOCA-salt hypertensive rats, supporting the idea that endogenous ouabain may contribute to the increase and maintenance of vascular tone in hypertension

    Isoproterenol Induces Vascular Oxidative Stress And Endothelial Dysfunction Via A Giα-coupled β2-adrenoceptor Signaling Pathway

    No full text
    Objective: Sustained β-adrenergic stimulation is a hallmark of sympathetic hyperactivity in cardiovascular diseases. It is associated with oxidative stress and altered vasoconstrictor tone. This study investigated the β-adrenoceptor subtype and the signaling pathways implicated in the vascular effects of β-adrenoceptor overactivation. Methods and Results: Mice lacking the β1- or β2-adrenoceptor subtype (β1KO, β2KO) and wild-type (WT) were treated with isoproterenol (ISO, 15 μg.g-1.day-1, 7 days). ISO significantly enhanced the maximal vasoconstrictor response (Emax) of the aorta to phenylephrine in WT ( +34%) and β1KO mice (+35%) but not in β2KO mice. The nitric oxide synthase (NOS) inhibitor L-NAME abolished the differences in phenylephrine response between the groups, suggesting that ISO impaired basal NO availability in the aorta of WT and β1KO mice. Superoxide dismutase (SOD), pertussis toxin (PTx) or PD 98,059 (p-ERK 1/2 inhibitor) incubation reversed the hypercontractility of aortic rings from ISO-treated WT mice; aortic contraction of ISO-treated β2KO mice was not altered. Immunoblotting revealed increased aortic expression of Giα-3 protein (+50%) and phosphorylated ERK1/2 (+90%) and decreased eNOS dimer/monomer ratio in ISO-treated WT mice. ISO enhanced the fluorescence response to dihydroethidium (+100%) in aortas from WT mice, indicating oxidative stress that was normalized by SOD, PTx and L-NAME. The ISO effects were abolished in β2KO mice. Conclusions: The β2-adrenoceptor/Giα signaling pathway is implicated in the enhanced vasoconstrictor response and eNOS uncoupling-mediated oxidative stress due to ISO treatment. Thus, long-term β2-AR activation might results in endothelial dysfunction. © 2014 Davel et al.93Parati, G., Esler, M., The human sympathetic nervous system: Its relevance in hypertension and heart failure (2012) Eur Heart J, 33, pp. 1058-1066Bristow, M.R., Gilbert, E.M., Abraham, W.T., Adams, K.F., Fowler, M.B., Hershberger, R.E., Kubo, S.H., Shusterman, N., Carvedilol produces dose-related improvements in left ventricular function and survival in subjects with chronic heart failure (1996) Circulation, 94 (11), pp. 2807-2816Carll, A.P., Willis, M.S., Lust, R.M., Costa, D.L., Farraj, A.K., Merits of non-invasive rat models of left ventricular heart failure (2011) Cardiovasc Toxicol, 11, pp. 91-112Banerjee, S.K., Sood, S., Dinda, A.K., Das, T.K., Maulik, S.K., Chronic oral administration of raw garlic protects against isoproterenol-induced myocardial necrosis in rat (2003) Comparative Biochemistry and Physiology - C Toxicology and Pharmacology, 136 (4), pp. 377-386. , DOI 10.1016/j.cca.2003.10.011Murray, D.R., Mummidi, S., Valente, A.J., Yoshida, T., Somanna, N.K., β2 adrenergic activation induces the expression of IL-18 binding protein, a potent inhibitor of isoproterenol induced cardiomyocyte hypertrophy in vitro and myocardial hypertrophy in vivo (2012) J Mol Cell Cardiol, 52, pp. 206-218Murray, D.R., Prabhu, S.D., Chandrasekar, B., Chronic β-adrenergic stimulation induces myocardial proinflammatory cytokine expression (2000) Circulation, 101 (20), pp. 2338-2341Davel, A.P., Fukuda, L.E., Sá, L.L., Munhoz, C.D., Scavone, C., Effects of isoproterenol treatment for 7 days on inflammatory mediators in the rat aorta (2008) Am J Physiol Heart Circ Physiol, 295, pp. 211-219Davel, A.P.C., Kawamoto, E.M., Scavone, C., Vassallo, D.V., Rossoni, L.V., Changes in vascular reactivity following administration of isoproterenol for 1 week: A role for endothelial modulation (2006) British Journal of Pharmacology, 148 (5), pp. 629-639. , DOI 10.1038/sj.bjp.0706749, PII 0706749Kim, H.K., Park, W.S., Warda, M., Park, S.Y., Ko, E.A., Beta-adrenergic overstimulation impaired vascular contractility via actin-cytoskeleton disorganization in rabbit cerebral artery (2012) PLoS One, 7, pp. e43884Morisco, C., Zebrowski, D.C., Vatner, D.E., Vatner, S.F., Sadoshima, J., Beta-adrenergic cardiac hypertrophy is mediated primarily by the beta1-subtype in the rat heart (2001) Journal of Molecular and Cellular Cardiology, 33 (3), pp. 561-573. , DOI 10.1006/jmcc.2000.1332Patterson, A.J., Zhu, W., Chow, A., Agrawal, R., Kosek, J., Protecting the myocardium: A role for the β2-adrenergic receptor in the heart (2004) Crit Care Med, 32, pp. 1041-1048Srivastava, S., Chandrasekar, B., Gu, Y., Luo, J., Hamid, T., Hill, B.G., Prabhu, S.D., Downregulation of CuZn-superoxide dismutase contributes to b-adrenergic receptor-mediated oxidative stress in the heart (2007) Cardiovascular Research, 74 (3), pp. 445-455. , DOI 10.1016/j.cardiores.2007.02.016, PII S0008636307000673Ryall, J.G., Schertzer, J.D., Murphy, K.T., Allen, A.M., Lynch, G.S., Chronic β2- adrenoceptor stimulation impairs cardiac relaxation via reduced SR Ca2+- ATPase protein and activity (2008) Am J Physiol Heart Circ Physiol, 294, pp. H2587-H2595Au, D.H., Udris, E.M., Fan, V.S., Curtis, J.R., McDonell, M.B., Fihn, S.D., Risk of mortality and heart failure exacerbations associated with inhaled beta-adrenoceptor agonists among patients with known left ventricular systolic dysfunction (2003) Chest, 123 (6), pp. 1964-1969. , DOI 10.1378/chest.123.6.1964Xu, Q., Dalic, A., Fang, L., Kiriazis, H., Ritchie, R.H., Myocardial oxidative stress contributes to transgenic β2-adrenoceptor activation-induced cardiomyopathy and heart failure (2011) Br J Pharmacol, 162, pp. 1012-1028Flacco, N., Segura, V., Perez-Aso, M., Estrada, S., Seller, J.F., Different β-adrenoceptor subtypes coupling to cAMP or NO/cGMP pathways: Implications in the relaxant response of rat conductance and resistance vessels (2013) Br J Pharmacol, 169, pp. 413-425Chruscinski, A., Brede, M.E., Meinel, L., Lohse, M.J., Kobilka, B.K., Differential distribution of β-adrenergic receptor subtypes in blood vessels of knockout mice lacking β1- or β2- adrenergic receptors (2001) Mol Pharmacol, 60, pp. 955-962Daaka, Y., Luttrell, L.M., Lefkowitz, R.J., Switching of the coupling of the β2-adrenergic receptor to different G proteins by protein kinase A (1997) Nature, 6, pp. 88-91Rohrer, D.K., Desai, K.H., Jasper, J.R., Stevens, M.E., Regula, D.P., Targeted disruption of the mouse β1-adrenergic receptor gene: Developmental and cardiovascular effects (1996) Proc Natl Acad Sci USA, 93, pp. 7375-7380Chruscinski, A.J., Rohrer, D.K., Schauble, E., Desai, K.H., Bernstein, D., Targeted disruption of the β2-adrenergic receptor gene (1999) J Biol Chem, 274, pp. 16694-16700Davel, A.P., Ceravolo, G.S., Wenceslau, C.F., Carvalho, M.H., Brum, P.C., Increased vascular contractility and oxidative stress in β2-adrenoceptor knockout mice: The role of NADPH oxidase (2012) J Vasc Res, 49, pp. 342-352Baloǧlu, E., Kiziltepe, O., Gürdal, H., The role of Gi proteins in reduced vasorelaxation response to beta-adrenoceptor agonists in rat aorta during maturation (2007) Eur J Pharmacol, 564, pp. 167-173Lembo, G., Iaccarino, G., Vecchione, C., Barbato, E., Morisco, C., Insulin enhances endothelial α2-adrenergic vasorelaxation by a pertussis toxin mechanism (1997) Hypertension, 30, pp. 1128-1134. , 1997Cai, S., Khoo, J., Mussa, S., Alp, N.J., Channon, K.M., Endothelial nitric oxide synthase dysfunction in diabetic mice: Importance of tetrahydrobiopterin in eNOS dimerisation (2005) Diabetologia, 48 (9), pp. 1933-1940. , DOI 10.1007/s00125-005-1857-5Lobato, N.S., Neves, K.B., Filgueira, F.P., Fortes, Z.B., Carvalho, M.H., The adipokine chemerin augments vascular reactivity to contractile stimuli via activation of the MEK-ERK1/2 pathway (2012) Life Sci, 91, pp. 600-606Fukuda, L.E., Davel, A.P., Verissimo-Filho, S., Lopes, L.R., Cachofeiro, V., Fenofibrate and pioglitazone do not ameliorate the altered vascular reactivity in aorta of isoproterenol-treated rats (2008) J Cardiovasc Pharmacol, 52, pp. 413-421Ogut, O., Brozovich, F.V., The potential role of MLC phosphatase and MAPK signalling in the pathogenesis of vascular dysfunction in heart failure (2008) J Cell Mol Med, 12, pp. 2158-2164Negrão, C.E., Hamilton, M.A., Fonarow, G.C., Hage, A., Moriguchi, J.D., Impaired endothelium-mediated vasodilation is not the principal cause of vasoconstriction in heart failure (2000) Am J Physiol Heart Circ Physiol, 278, pp. H168-H174Davel, A.P., Wenceslau, C.F., Akamine, E.H., Xavier, F.E., Couto, G.K., Endothelial dysfunction in cardiovascular and endocrine-metabolic diseases: An update (2011) Braz J Med Biol Res, 44, pp. 920-932Xu, J., Li, N., Dai, D.Z., Yu, F., Dai, Y., The endothelin receptor antagonist CPU0213 is more effective than aminoguanidine to attenuate isoproterenol-induced vascular abnormality by suppressing overexpression of NADPH oxidase, ETA, ETB, and MMP9 in the vasculature (2008) J Cardiovasc Pharmacol, 52, pp. 42-48Beckman, J.S., Beckman, T.W., Chen, J., Marshall, P.A., Freeman, B.A., Apparent hydroxyl radical production by peroxynitrite: Implications for endothelial injury from nitric oxide and superoxide (1990) Proc Natl Acad Sci USA, 87, pp. 1620-1624Mian, K.B., Martin, W., Differential sensitivity of basal and acetylcholine-stimulated activity of nitric oxide to destruction by superoxide anion in rat aorta (1995) Br J Pharmacol, 115, pp. 993-1000Ma, X.-I., Weyrich, A.S., Lefer, D.J., Lefer, A.M., Diminished basal nitric oxide release after myocardial ischemia and reperfusion promotes neutrophil adherence to coronary endothelium (1993) Circulation Research, 72 (2), pp. 403-412Ferro, A., Coash, M., Yamamoto, T., Rob, J., Ji, Y., Queen, L., Nitric oxide-dependent beta2-adrenergic dilatation of rat aorta is mediated through activation of both protein kinase A and Akt (2004) British Journal of Pharmacology, 143 (3), pp. 397-403. , DOI 10.1038/sj.bjp.0705933Pourageaud, F., Leblais, V., Bellance, N., Marthan, R., Muller, B., Role of β2- adrenoceptors (β-AR), but not β1-, β3-AR and endothelial nitric oxide, in beta- AR-mediated relaxation of rat intrapulmonary artery (2005) Naunyn Schmiedebergs Arch Pharmacol, 372, pp. 14-23Queen, L.R., Ji, Y., Xu, B., Young, L., Yao, K., Mechanisms underlying β2-adrenoceptor-mediated nitric oxide generation by human umbilical vein endothelial (2006) J Physiol, 576, pp. 585-594Xiao, R.P., Avdonin, P., Zhou, Y.Y., Cheng, H., Akhter, S.A., Coupling of β2-adrenoceptor to Gi proteins and its physiological relevance in murine cardiac myocytes (1999) Circ Res, 84, pp. 43-52Banquet, S., Delannoy, E., Agouni, A., Dessy, C., Lacomme, S., Role of G(i/o)-Src kinase-PI3K/Akt pathway and caveolin-1 in β2-adrenoceptor coupling to endothelial NO synthase in mouse pulmonary artery (2011) Cell Signal, 23, pp. 1136-1143Itoh, H., Toyama, R., Kozasa, T., Tsukamoto, T., Matsuoka, M., Presence of three distinct molecular species of Gi protein alpha subunit. Structure of rat cDNAs and human genomic DNAs (1988) J Biol Chem, 263, pp. 6656-6664Wong, Y.H., Conklin, B.R., Bourne, H.R., Gz-mediated hormonal inhibition of cyclic AMP accumulation (1992) Science, 255, pp. 339-342Li, Y., Anand-Srivastava, M.B., Inactivation of enhanced expression of Gi proteins by pertussis toxin attenuates the development of high blood pressure in spontaneously hypertensive rats (2002) Circulation Research, 91 (3), pp. 247-254. , DOI 10.1161/01.RES.0000029969.39875.4BSaha, S., Li, Y., Lappas, G., Anand-Srivastava, M.B., Activation of natriuretic peptide receptor-C attenuates the enhanced oxidative stress in vascular smooth muscle cells from spontaneously hypertensive rats: Implication of Gialpha protein (2008) J Mol Cell Cardiol, 44, pp. 336-344Anand-Srivastava, M.B., Enhanced expression of inhibitory guanine nucleotide regulatory protein in spontaneously hypertensive rats. Relationship to adenylate cyclase inhibition (1992) Biochem J, 288, pp. 79-85Ge, C., Garcia, R., Anand-Srivastava, M.B., Enhanced expression of Gialpha protein and adenylyl cyclase signaling in aortas from 1 kidney 1 clip hypertensive rats (2006) Canadian Journal of Physiology and Pharmacology, 84 (7), pp. 739-746. , DOI 10.1139/Y05-123Zemancíková, A., Török, J., Zicha, J., Kunes, J., Inactivation of G(i) proteins by pertussis toxin diminishes the effectiveness of adrenergic stimuli in conduit arteries from spontaneously hypertensive rats (2008) Physiol Res, 57, pp. 299-302Zou, M.-H., Shi, C., Cohen, R.A., Oxidation of the zinc-thiolate complex and uncoupling of endothelial nitric oxide synthase by peroxynitrite (2002) Journal of Clinical Investigation, 109 (6), pp. 817-826. , DOI 10.1172/JCI200214442Antoniades, C., Shirodaria, C., Leeson, P., Antonopoulos, A., Warrick, N., Association of plasma asymmetrical dimethylarginine (ADMA) with elevated vascular superoxide production and endothelial nitric oxide synthase uncoupling: Implications for endothelial function in human atherosclerosis (2009) Eur Heart J, 30, pp. 1142-1150Antoniades, C., Shirodaria, C., Warrick, N., Cai, S., De Bono, J., Lee, J., Leeson, P., Channon, K.M., 5-Methyltetrahydrofolate rapidly improves endothelial function and decreases superoxide production in human vessels: Effects on vascular tetrahydrobiopterin availability and endothelial nitric oxide synthase coupling (2006) Circulation, 114 (11), pp. 1193-1201. , DOI 10.1161/CIRCULATIONAHA.106.612325, PII 0000301720060912000012Kim, N., Kim, H., Youm, J.B., Sun, P.W., Warda, M., Ko, J.-H., Han, J., Site specific differential activation of ras/raf/ERK signaling in rabbit isoproterenol-induced left ventricular hypertrophy (2006) Biochimica et Biophysica Acta - Molecular Cell Research, 1763 (10), pp. 1067-1075. , DOI 10.1016/j.bbamcr.2006.08.002, PII S0167488906002096Cheng, J., Wu, C.C., Gotlinger, K.H., Zhang, F., Falck, J.R., 20-Hydroxy- 5,8,11,14-eicosatetraenoic acid mediates endothelial dysfunction via IkappaB kinase-dependent endothelial nitric-oxide synthase uncoupling (2010) J Pharmacol Exp Ther, 332, pp. 57-65Bauersachs, J., Bouloumie, A., Fraccarollo, D., Hu, K., Busse, R., Ertl, G., Endothelial dysfunction in chronic myocardial infarction despite increased vascular endothelial nitric oxide synthase and soluble guanylate cyclase expression: Role of enhanced vascular superoxide production (1999) Circulation, 100 (3), pp. 292-298Moens, A.L., Kietadisorn, R., Lin, J.Y., Kass, D., Targeting endothelial and myocardial dysfunction with tetrahydrobiopterin (2011) J Mol Cell Cardiol, 5, pp. 559-563Kietadisorn, R., Juni, R.P., Moens, A.L., Tackling endothelial dysfunction by modulating NOS uncoupling: New insights into its pathogenesis and therapeutic possibilities (2012) Am J Physiol Endocrinol Metab, 302, pp. E481-E495Molenaar, P., Smolich, J.J., Russell, F.D., McMartin, L.R., Summers, R.J., Differential regulation of beta-1 and beta-2 adrenoceptors in guinea pig atrioventricular conducting system after chronic (-)-isoproterenol infusion (1990) Journal of Pharmacology and Experimental Therapeutics, 255 (1), pp. 393-400Soltysinska, E., Thiele, S., Olesen, S.P., Osadchii, O.E., Chronic sympathetic activation promotes downregulation of b-adrenoceptor-mediated effects in the guinea pig heart independently of structural remodeling and systolic dysfunction (2011) Pflugers Arch, 462, pp. 529-543Shenoy, S.K., Drake, M.T., Nelson, C.D., Houtz, D.A., Xiao, K., β-arrestin-dependent, G protein-independent ERK1/2 activation by the β2 adrenergic receptor (2006) J Biol Chem, 281, pp. 1261-1273Gava, A.L., Peotta, V.A., Cabral, A.M., Meyrelles, S.S., Vasquez, E.C., Decreased baroreflex sensitivity in isoproterenol-treated mice with cardiac hypertrophy (2004) Autonomic Neuroscience: Basic and Clinical, 114 (1-2), pp. 47-54. , DOI 10.1016/j.autneu.2004.07.003, PII S1566070204001614A randomized trial of beta-blockade in heart failure. The Cardiac Insufficiency Bisoprolol Study (CIBIS) (1994) Circulation, 90, pp. 1765-1773. , CIBIS Investigators and CommitteesEffect of metoprolol CR/XL in chronic heart failure: Metoprolol CR/XL Randomised Intervention Trial in Congestive Heart Failure (MERIT-HF) (1999) Lancet, 353, pp. 2001-2007. , MERIT-HF Study GroupPacker, M., Bristow, M.R., Cohn, J.N., Colucci, W.S., Fowler, M.B., Gilbert, E.M., Shusterman, N.H., The effect of carvedilol on morbidity and mortality in patients with chronic heart failure (1996) New England Journal of Medicine, 334 (21), pp. 1349-1355. , DOI 10.1056/NEJM199605233342101Hothersall, J.D., Black, J., Caddick, S., Vinter, J.G., Tinker, A., The design, synthesis and pharmacological characterization of novel β2-adrenoceptor antagonists (2011) Br J Pharmacol, 164, pp. 317-33

    Effects of ouabain on vascular reactivity

    No full text
    Ouabain is an endogenous substance occurring in the plasma in the nanomolar range, that has been proposed to increase vascular resistance and induce hypertension. This substance acts on the a-subunit of Na+,K+-ATPase inhibiting the Na+-pump activity. In the vascular smooth muscle this effect leads to intracellular Na+ accumulation that reduces the activity of the Na+/Ca2+ exchanger and to an increased vascular tone. It was also suggested that circulating ouabain, even in the nanomolar range, sensitizes the vascular smooth muscle to vasopressor substances. We tested the latter hypothesis by studying the effects of ouabain in the micromolar and nanomolar range on phenylephrine (PE)-evoked pressor responses. The experiments were performed in normotensive and hypertensive rats in vivo, under anesthesia, and in perfused rat tail vascular beds. The results showed that ouabain pretreatment increased the vasopressor responses to PE in vitro and in vivo. This sensitization after ouabain treatment was also observed in hypertensive animals which presented an enhanced vasopressor response to PE in comparison to normotensive animals. It is suggested that ouabain at nanomolar concentrations can sensitize vascular smooth muscle to vasopressor stimuli possibly contributing to increased tone in hypertensio

    Time course of training-induced microcirculatory changes and of vegf expression in skeletal muscles of spontaneously hypertensive female rats

    Get PDF
    Exercise-induced vessel changes modulate arterial pressure (AP) in male spontaneously hypertensive rats (SHR). Vascular endothelial growth factor (VEGF) is important for angiogenesis of skeletal muscle. The present study evaluated the time course of VEGF and angiogenesis after short- and long-term exercise training of female SHR and Wistar Kyoto (WKY) rats, 8-9 weeks (200-250 g). Rats were allocated to daily training or remained sedentary for 3 days (N = 23) or 13 weeks (N = 23). After training, the carotid artery was catheterized for AP measurements. Locomotor (tibialis anterior and gracilis) and non-locomotor skeletal muscles (temporalis) were harvested and prepared for histologic and protein expression analyses. Training increased treadmill performance by all groups (SHR = 28%, WKY = 64%, 3 days) and (SHR = 141%, WKY = 122%, 13 weeks). SHR had higher values of AP than WKY (174 ± 4 vs 111 ± 2 mmHg) that were not altered by training. Three days of running increased VEGF expression (SHR = 28%, WKY = 36%) simultaneously with an increase in capillary-to-fiber ratio in gracilis muscle (SHR = 19%, WKY = 15%). In contrast, 13 weeks of training increased gracilis capillary-to-fiber ratio (SHR = 18%, WKY = 19%), without simultaneous changes in VEGF expression. Training did not change VEGF expression and capillarity of temporalis muscle. We conclude that training stimulates time- and tissue-dependent VEGF protein expression, independent of pressure levels. VEGF triggers angiogenesis in locomotor skeletal muscle shortly after the exercise starts, but is not involved in the maintenance of capillarity after long-term exercise in female rats

    Enhanced Endothelium-dependent Relaxation Of Rat Pulmonary Artery Following β-adrenergic Overstimulation: Involvement Of The No/cgmp/vasp Pathway

    No full text
    Aims: The aim of this study was to investigate whether β-adrenoceptor (β-AR) overstimulation induced by in vivo treatment with isoproterenol (ISO) alters vascular reactivity and nitric oxide (NO) production and signaling in pulmonary arteries. Main methods: Vehicle or ISO (0.3mgkg-1 day-1) was administered daily to male Wistar rats. After 7days, the jugular vein was cannulated to assess right ventricular (RV) systolic pressure (SP) and end diastolic pressure (EDP). The extralobar pulmonary arteries were isolated to evaluate the relaxation responses, protein expression (Western blot), NO production (diaminofluorescein-2 fluorescence), and cyclic guanosine 3',5'-monophosphate (cGMP) levels (enzyme immunoassay kit). Key findings: ISO treatment induced RV hypertrophy; however, no differences in RV-SP and EDP were observed. The pulmonary arteries from the ISO-treated group showed enhanced relaxation to acetylcholine that was abolished by the NO synthase (NOS) inhibitor Nω-nitro-l-arginine methyl ester (l-NAME); whereas relaxation elicited by sodium nitroprusside, ISO, metaproterenol, mirabegron, or KCl was not affected by ISO treatment. ISO-treated rats displayed enhanced endothelial NOS (eNOS) and vasodilator-stimulated phosphoprotein (VASP) expression in the pulmonary arteries, while phosphodiesterase-5 protein expression decreased. ISO treatment increased NO and cGMP levels and did not induce eNOS uncoupling. Significance: The present data indicate that β-AR overactivation enhances the endothelium-dependent relaxation of pulmonary arteries. This effect was linked to an increase in eNOS-derived NO production, cGMP formation and VASP content and to a decrease in phosphodiesterase-5 expression. Therefore, elevated NO bioactivity through cGMP/VASP signaling could represent a protective mechanism of β-AR overactivation on pulmonary circulation

    Increased Vascular Contractility And Oxidative Stress In β 2-adrenoceptor Knockout Mice: The Role Of Nadph Oxidase

    No full text
    Background/Aims: β 2-adrenoceptor (β 2-AR) activation induces smooth muscle relaxation and endothelium-derived nitric oxide (NO) release. However, whether endogenous basal β 2-AR activity controls vascular redox status and NO bioavailability is unclear. Thus, we aimed to evaluate vascular reactivity in mice lacking functional β 2- AR (β 2KO), focusing on the role of NO and superoxide anion. Methods and Results: Isolated thoracic aortas from β 2KO and wild-type mice (WT) were studied. β 2KO aortas exhibited an enhanced contractile response to phenylephrine compared to WT. Endothelial removal and L-NAME incubation increased phenylephrine-induced contraction, abolishing the differences between β 2KO and WT mice. Basal NO availability was reduced in aortas from β 2KO mice. Incubation of β 2KO aortas with superoxide dismutase or NADPH inhibitor apocynin restored the enhanced contractile response to phenylephrine to WT levels. β 2KO aortas exhibited oxidative stress detected by enhanced dihydroethidium fluorescence, which was normalized by apocynin. Protein expression of eNOS was reduced, while p47 phox expression was enhanced in β 2KO aortas. Conclusions: The present results demonstrate for the first time that enhanced NADPH-derived superoxide anion production is associated with reduced NO bioavailability in aortas of β 2KO mice. This study extends the knowledge of the relevance of the endogenous activity of β 2-AR to the maintenance of the vascular physiology. © 2012 S. Karger AG, Basel.494342352Tanaka, Y., Horinouchi, T., Koike, K., New insights into β-adrenoceptors in smooth muscle: Distribution of receptor subtypes and molecular mechanisms triggering muscle relaxation (2005) Clinical and Experimental Pharmacology and Physiology, 32 (7), pp. 503-514Steinberg, S.F., Jaffe, E.A., Bilezikian, J.P., Endothelial cells contain adrenoceptors (1984) Naunyn-Schmiedebergs Arch Pharmacol, 325, pp. 310-313Molenaar, P., Malta, E., Jones, C.R., Buxton, B.F., Summers, R.J., Autoradiographic localization and function of adrenoceptors on the human internal mammary artery and saphenous vein (1988) Br J Pharmacol, 95, pp. 225-233Gray, D.W., Marshall, I., Novel signal transduction pathway mediating endothelium-dependent- adrenoceptor vasorelaxation in rat thoracic aorta (1992) Br J Pharmacol, 107, pp. 684-690Ferro, A., Coash, M., Yamamoto, T., Rob, J., Ji, Y., Queen, L., Nitric oxide-dependent β 2-adrenergic dilatation of rat aorta is mediated through activation of both protein kinase A and Akt (2004) British Journal of Pharmacology, 143 (3), pp. 397-403. , DOI 10.1038/sj.bjp.0705933Graves, J., Poston, L., Beta-adrenoceptor agonist mediated relaxation of rat isolated resistance arteries: A role for the endothelium and nitric oxide (1993) British Journal of Pharmacology, 108 (3), pp. 631-637Priest, R.M., Hucks, D., Ward, J.P.T., Noradrenaline, β-adrenoceptor mediated vasorelaxation and nitric oxide in large and small pulmonary arteries of the rat (1997) British Journal of Pharmacology, 122 (7), pp. 1375-1384Pourageaud, F., Leblais, V., Bellance, N., Marthan, R., Muller, B., Role of,2-adrenoceptors (AR), but not,1-,3-AR and endothelial nitric oxide, in AR-mediated relaxation of rat intrapulmonary artery (2005) Naunyn Schmiedebergs Arch Pharmacol, 372, pp. 14-23Akimoto, Y., Horinouchi, T., Shibano, M., Matsushita, M., Yamashita, Y., Okamoto, T., Yamaki, F., Koike, K., Nitric oxide (NO) primarily accounts for endothelium-dependent component of α-adrenoceptor-activated smooth muscle relaxation of mouse aorta in response to isoprenaline (2002) Journal of Smooth Muscle Research, 38 (4-5), pp. 87-99. , DOI 10.1540/jsmr.38.87Leblais, V., Delannoy, E., Fresquet, F., Begueret, H., Bellance, N., Banquet, S., Allieres, C., Muller, B., Adrenergic relaxation in pulmonary arteries: Preservation of the endothelial nitric oxide-dependent β 2 component in pulmonary hypertension (2008) Cardiovascular Research, 77 (1), pp. 202-210. , DOI 10.1093/cvr/cvm008Dawes, M., Chowienczyk, P.J., Ritter, J.M., Effects of inhibition of the L-arginine/nitric oxide pathway on vasodilation caused by β-adrenergic agonists in human forearm (1997) Circulation, 95 (9), pp. 2293-2297Chruscinski, A., Brede, M.E., Meinel, L., Lohse, M.J., Kobilka, B.K., Hein, L., Differential distribution of β-adrenergic receptor subtypes in blood vessels of knockout mice lacking β 1- or β 2- adrenergic receptors (2001) Molecular Pharmacology, 60 (5), pp. 955-962Rascado, R.R., Bendhack, L.M., Activation of α 2-adrenoceptors is necessary to induce nitric oxide release in isoprenaline-induced relaxation (2005) Vascular Pharmacology, 42 (2), pp. 63-68. , DOI 10.1016/j.vph.2005.01.008Alexander, S.P.H., Mathie, A., Peters, J.A., Guide to Receptors and Channels (GRAC), 4th edition (2009) Br J Pharmacol, 158, pp. S1-S254Ferro, A., Queen, L.R., Priest, R.M., Xu, B., Ritter, J.M., Poston, L., Ward, J.P.T., Activation of nitric oxide synthase by β 2-adrenoceptors in human umbilical vein endothelium in vitro (1999) British Journal of Pharmacology, 126 (8), pp. 1872-1880. , DOI 10.1038/sj.bjp.0702512Queen, L.R., Ji, Y., Xu, B., Young, L., Yao, K., Wyatt, A.W., Rowlands, D.J., Ferro, A., Mechanisms underlying β 2-adrenoceptor-mediated nitric oxide generation by human umbilical vein endothelial cells (2006) Journal of Physiology, 576 (2), pp. 585-594. , DOI 10.1113/jphysiol.2006.115998Gillissen, A., Wickenburg, D., Van Zwoll, D., Schultze-Werninghaus, G., Beta-2-agonists have antioxidant function in vitro. 2. The effect of beta-2-agonists on oxidant-mediated cytotoxicity and on superoxide anion generated by human polymorphonuclear leukocytes (1997) Respiration, 64 (1), pp. 23-28Mirza, Z.N., Kato, M., Kimura, H., Tachibana, A., Fujiu, T., Suzuki, M., Mochizuki, H., Morikawa, A., Fenoterol inhibits superoxide anion generation by human polymorphonuclear leukocytes via β 2-adrenoceptor-dependent and -independent mechanisms (2002) Annals of Allergy, Asthma and Immunology, 88 (5), pp. 494-500Trombetta, I.C., Batalha, L.T., Rondon, M.U.P.B., Laterza, M.C., Frazzatto, E., Alves, M.J.N.N., Santos, A.C., Negrao, C.E., Gly 16 + Glu 27 β 2-adrenoceptor polymorphisms cause increased forearm blood flow responses to mental stress and handgrip in humans (2005) Journal of Applied Physiology, 98 (3), pp. 787-794. , DOI 10.1152/japplphysiol.00503.2004Garovic, V.D., Joyner, M.J., Dietz, N.M., Boerwinkle, E., Turner, S.T., 2-Adrenergic receptor polymorphism and nitric oxide-dependent forearm blood flow responses to isoproterenol in humans (2003) Journal of Physiology, 546 (2), pp. 583-589. , DOI 10.1113/jphysiol.2002.031138Chruscinski, A.J., Rohrer, D.K., Schauble, E., Desai, K.H., Bernstein, D., Kobilka, B.K., Targeted disruption of the 2-adrenergic receptor gene (1999) J Biol Chem, 274, pp. 16694-16700Rolim, N.P.L., Medeiros, A., Rosa, K.T., Mattos, K.C., Irigoyen, M.C., Krieger, E.M., Krieger, J.E., Brum, P.C., Exercise training improves the net balance of cardiac Ca 2+ handling protein expression in heart failure (2007) Physiological Genomics, 29 (3), pp. 246-252. , http://physiolgenomics.physiology.org/cgi/reprint/29/3/246, DOI 10.1152/physiolgenomics.00188.2006Rabelo, L.A., Cortes, S.F., Alvarez-Leite, J.I., Lemos, V.S., Endothelium dysfunction in LDL receptor knockout mice: A role for H 2O 2 (2003) British Journal of Pharmacology, 138 (7), pp. 1215-1220. , DOI 10.1038/sj.bjp.0705164Davel, A.P.C., Kawamoto, E.M., Scavone, C., Vassallo, D.V., Rossoni, L.V., Changes in vascular reactivity following administration of isoproterenol for 1 week: A role for endothelial modulation (2006) British Journal of Pharmacology, 148 (5), pp. 629-639. , DOI 10.1038/sj.bjp.0706749, PII 0706749Rossoni, L.V., Salaices, M., Marin, J., Vassallo, D.V., Alonso, M.J., Alterations in phenylephrine-induced contractions and the vascular expression of Na +,K +-ATPase in ouabain-induced hypertension (2002) British Journal of Pharmacology, 135 (3), pp. 771-781Davel, A.P., Fukuda, L.E., Sá, L.L., Munhoz, C.D., Scavone, C., Sanz-Rosa, D., Cachofeiro, V., Rossoni, L.V., Effects of isoproterenol treatment for 7 days on inflammatory mediators in the rat aorta (2008) Am J Physiol Heart Circ Physiol, 295, pp. 211-219Iaccarino, G., Ciccarelli, M., Sorriento, D., Galasso, G., Campanile, A., Santulli, G., Cipolletta, E., Trimarco, B., Ischemic neoangiogenesis enhanced by, 2-adrenergic receptor overexpression: A novel role for the endothelial adrenergic system (2005) Circ Res, 97, pp. 1182-1189Van Hove, C.E., Van Der Donckt, C., Herman, A.G., Bult, H., Fransen, P., Vasodilator efficacy of nitric oxide depends on mechanisms of intracellular calcium mobilization in mouse aortic smooth muscle cells (2009) Br J Pharmacol, 158, pp. 920-930Isenovic, E., Walsh, M.F., Muniyappa, R., Bard, M., Diglio, C.A., Sowers, J.R., Phosphatidylinositol 3-kinase may mediate isoproterenol-induced vascular relaxation in part through nitric oxide production (2002) Metabolism: Clinical and Experimental, 51 (3), pp. 380-386. , DOI 10.1053/meta.2002.30525Rohrer, D.K., Chruscinski, A., Schauble, E.H., Bernstein, D., Kobilka, B.K., Cardiovascular and metabolic alterations in mice lacking both beta1-and beta2-adrenergic receptors (1999) J Biol Chem, 274, pp. 16701-16708Alvarez, Y., Briones, A.M., Hernanz, R., Perez-Giron, J.V., Alonso, M.J., Salaices, M., Role of NADPH oxidase and iNOS in vasoconstrictor responses of vessels from hypertensive and normotensive rats (2008) British Journal of Pharmacology, 153 (5), pp. 926-935. , DOI 10.1038/sj.bjp.0707575, PII 0707575Giachini, F.R., Sullivan, J.C., Lima, V.V., Carneiro, F.S., Fortes, Z.B., Pollock, D.M., Carvalho, M.H., Tostes, R.C., Extracellular signalregulated kinase 1/2 activation, via downregulation of mitogen-activated protein kinase phosphatase 1, mediates sex differences in desoxycorticosterone acetate-salt hypertension vascular reactivity (2010) Hypertension, 55, pp. 172-179Birukov, K.G., Cyclic stretch, reactive oxygen species, and vascular remodeling (2009) Antioxid Redox Signal., 11, pp. 1651-1667Duerrschmidt, N., Stielow, C., Muller, G., Pagano, P.J., Morawietz, H., NO-mediated regulation of NAD(P)H oxidase by laminar shear stress in human endothelial cells (2006) Journal of Physiology, 576 (2), pp. 557-567. , DOI 10.1113/jphysiol.2006.111070Selemidis, S., Dusting, G.J., Peshavariya, H., Kemp-Harper, B.K., Drummond, G.R., Nitric oxide suppresses NADPH oxidase-dependent superoxide production by S-nitrosylation in human endothelial cells (2007) Cardiovascular Research, 75 (2), pp. 349-358. , DOI 10.1016/j.cardiores.2007.03.030, PII S0008636307001873Lassegue, B., Griendling, K.K., NADPH oxidases: Functions and pathologies in the vasculature (2010) Arterioscler Thromb Vasc Biol, 30, pp. 653-661Ximenes, V.F., Kanegae, M.P.P., Rissato, S.R., Galhiane, M.S., The oxidation of apocynin catalyzed by myeloperoxidase: Proposal for NADPH oxidase inhibition (2007) Archives of Biochemistry and Biophysics, 457 (2), pp. 134-141. , DOI 10.1016/j.abb.2006.11.010, PII S0003986106004498Touyz, R.M., Apocynin, NADPH oxidase, and vascular cells: A complex matter (2008) Hypertension, 51 (2), pp. 172-174. , DOI 10.1161/HYPERTENSIONAHA.107.103200, PII 0000426820080200000003Burger, D., Montezano, A.C., Nishigaki, N., He, Y., Carter, A., Touyz, R.M., Endothelial microparticle formation by angiotensin II is mediated via Ang II receptor type I/NADPH oxidase/Rho kinase pathways targeted to lipid rafts (2011) Arterioscler Thromb Vasc Biol, 31, pp. 1898-1907Barbieri, S.S., Cavalca, V., Eligini, S., Brambilla, M., Caiani, A., Tremoli, E., Colli, S., Apocynin prevents cyclooxygenase 2 expression in human monocytes through NADPH oxidase and glutathione redox-dependent mechanisms (2004) Free Radical Biology and Medicine, 37 (2), pp. 156-165. , DOI 10.1016/j.freeradbiomed.2004.04.020, PII S0891584904003417MacMillan-Crow, L.A., Cruthirds, D.L., Manganese superoxide dismutase in disease (2001) Free Radical Res, 34, pp. 325-336Takahata, Y., Takarada, T., Iemata, M., Yamamoto, T., Nakamura, Y., Kodama, A., Yoneda, Y., Functional expression of, 2-adrenergic receptors responsible for protection against oxidative stress through promotion of glutathione synthesis after Nrf2 upregulation in undifferentiated mesenchymal C3H10T1/2 stem cells (2009) J Cell Physiol, 218, pp. 268-275Xu, Q., Dalic, A., Fang, L., Kiriazis, H., Ritchie, R.H., Sim, K., Gao, X.M., Du, X.J., Myocardial oxidative stress contributes to transgenic 2-adrenoceptor activation-induced cardiomyopathy and heart failure (2011) Br J Pharmacol, 162, pp. 1012-102

    Pancreatic Islets Isolated From β2 Adrenergic Receptor Knockout Mice Show Reduced Insulin Secretion In Response To Nutrients [ilhotas Pancreaśticas Isoladas De Camundongos Com Deleção Do Receptor Adrenérgico β2 Apresenta Reduzida Secreção De Insulina Em Resposta A Nutrientes]

    No full text
    Activation of β2 adrenergic receptors by catecholamine or catecholamine-mimetic substances may enhance insulin secretion. We herein investigated KCl- and nutrient-stimulated insulin secretion in pancreatic islets isolated from β2 knockout (β2KO) mice. β2KO mice showed reduced body weight, fasting hypoglycaemia associate to a similar fasting insulinemia compared to control. β2KO mice also showed reduced glucose tolerance despite the higher sensitivity to insulin. Glucose-induced insulin secretion was impaired in pancreatic islets isolated from β2KO mice. Leucine-induced (20mM) insulin secretion was diminished in pancreatic islets isolated from β2KO mice when compared to control one. The depolarizing effect of KCl on insulin secretion was also impaired in pancreatic islets from β2KO mice. These results suggested a possible role of β2 adrenergic receptors on nutrient-induced insulin secretion.353437443Ahrén, B., Autonomic regulation of islet hormone secretion-implications for health and disease (2000) Diabetologia, 43 (4), pp. 393-410Ahrén, B., Lundquist, I., Effects of selective and non-selective β-adrenergic agents on insulin secretion in vivo (1981) European Journal of Endocrinology, 71 (1), pp. 93-104Asensio, C., Jimenez, M., Rohner-Jeanrenaud, F., The lack of beta-adrenoceptors results in enhanced insulin sensitivity in mice exhibiting increased adiposity and glucose intolerance (2005) Diabetes, 54 (12), pp. 3490-3495Ashcroft, F.M., ATP-sensitive potassium channelopathies: Focus on insulin secretion (2005) The Journal of Clinical Investigation, 115 (8), pp. 2047-2058Berlan, M., Dang, T.L., The role of beta and alpha adrenergic receptors in the lipolytic effect of catecholamines on dog adipocytes (1978) Journal De Physiologie, 74 (6), pp. 601-608Blachier, F., Leclerq-Meyer, V., Marchand, J., Marchand, J., Woussen-Cole, M.C., Mathias, P.C., Sener, A., Malaisse, W.J., Stimulus-secretion coupling of arginine-induced insulin release. Functional response of islets to L-arginine and L-ornithine (1989) Biochimica Et Biophysica Acta, 1013 (2), pp. 144-151Bonen, A., MeGeney, L.A., McCarthy, S.C., McDermott, J.C., Tan, M.H., Epinephrine administration stimulates GLUT4 translocation but reduces glucose transport in muscle (1992) Biochemical and Biophysical Research Communications, 187 (2), pp. 685-691Brum, P.C., Rolim, N.P., Bacurau, A.V., Medeiros, A., Neurohumoral activation in heart failure: The role of adrenergic receptors (2006) Anais Da Academia Brasileira De Ciências, 78 (3), pp. 485-503Carpinelli, A.R., Malaisse, W.J., Regulation of 86Rb outflow from pancreatic islets: The dual effect of nutrient secretagogues (1981) The Journal of Physiololgy, 315, pp. 143-156Carvalho, E., Jansson, P.A., Axelsen, M., Eriksson, J.W., Huang, X., Groop, L., Rondinone, C., Smith, U., Low cellular IRS 1 gene and protein expression predict insulin resistance and NIDDM (1999) The FASEB Journal, 13 (15), pp. 2173-2178Charles, S., Tamagawa, T., Henquin, J.C., A single mechanism for the stimulation of insulin release and 86Rb+ efflux from rat islets by cationic amino acids (1982) The Biochemical Journal, 208 (2), pp. 301-308Chruscinski, A.J., Rohrer, D.K., Schauble, E., Desai, K.H., Bernstein, D., Kobilka, B.K., Targeted disruption of the beta2 adrenergic receptor gene (1999) Journal of Biological Chemistry, 274 (24), pp. 16694-16700Doronin, S., Wang, H., Malbon, C.C., Insulin stimulates phosphorylation of β2-adrenergic receptor by the insulin receptor, creating a potent feedback inhibitor of its tyrosine kinase (2002) Journal of Biological Chemistry, 277 (12), pp. 10698-10703Fisher, R.A., Kumar, R., Hanahan, D.J., Olson, M.S., Effects of beta-adrenergic stimulation on 1- O-hexadecyl-2-acetyl-sn-glycero-3-phosphocholine-mediated vasoconstriction and glycogenolysis in the perfused rat liver (1986) Jounal of Biological Chemistry, 261 (19), pp. 8817-8823Graphpad, P.R.I.S.M., (2003) User's Guide. Version 4.00 For Windows, , San Diego: GraphPad Software. IncGylfe, E., Comparison of the effects of leucines, non- metabolizable leucine analogues and other insulin secretagogues on the activity of glutamate dehydrogenase (1976) Acta Diabetologica Latina, 13 (1-2), pp. 20-24Han, X.X., Bonen, A., Epinephrine translocates GLUT-4 but inhibits insulin-stimulated glucose transport in rat muscle (1998) American Journal of Physiology, 274 (4), pp. 700-707Henquin, J.C., Meissner, H.P., Effects of amino acids on membrane potential and 86Rb+ fluxes in pancreatic β-cells (1981) American Journal of Physiology, 240 (3), pp. 245-252Henquin, J.C., Ravier, M.A., Nenquin, M., Jonas, J.C., Gilon, P., Hierarchy of the β-cell signals controlling insulin secretion (2003) European Journal of Clinical Investigation, 33 (9), pp. 742-750Hermans, M.P., Schmeer, W., Henquin, J.C., The permissive effect of glucose, tolbutamide and high K+ on arginine stimulation of insluin release in isolated mouse islets (1987) Diabetologia, 30 (8), pp. 659-665Jones, J.P., Dohm, G.L., Regulation of glucose transporter GLUT4 and hexokinase II gene transcription by insulin and epinephrine (1997) American Journal of Physiology, 273 (4), pp. E682-E687Kurose, T., Seino, Y., Nishi, S., Tsuji, K., Taminato, T., Tsuda, K., Imura, H., Mechanism of sympathetic neural regulation of insulin, somatostatin, and glucagon secretion (1990) American Journal of Physiology, 258 (1), pp. E220-E227Latorraca, M.Q., Carneiro, E.M., Mello, M.A., Boschero, A.C., Reduced insulin secretion in response to nutrients in islets from malnourished young rats is associated with a diminished calcium uptake (1999) The Journal of Nutrition of Biochemistry, 10 (1), pp. 37-43Laurent, D., Petersen, K.F., Russel, R.R., Cline, G.W., Shulman, G.I., Effect of epinephrine on muscle glycogenolysis and insulin-stimulated muscle glycogen synthesis in humans (1998) American Journal of Physiology, 274 (1), pp. E130-E138Macdonald, P.E., Joseph, J.W., Rorsman, P., Glucose-sensing mechanisms in pancreatic β-cells Philosophical Transactions of the Royal Society of London. Series B, Biological Sciences, 360 (1464), pp. 2211-2225Marçal, A.C., Camporez, J.P.G., Lima-Salgado, T.M., Cintra, D.E., Akamine, E.H., Ribeiro, L.M., Almeida, F.N., Carvalho, C.R.O., Changes in food intake, metabolic parameters and insulin resistance are induced by an isoenergetic, medium- chain fatty acid diet and are associated with modifications in insulin signalling in isolated rat pancreatic islets (2012) British Journal of Nutrition, 27, pp. 1-12Marçal, A.C., Grassiolli, S., Da Rocha, D.N., Puzzi, M.A., Gravena, C., Scomparin, D.X., De Freitas, M.P.C., The dual effect of isoproterenol on insulin release is suppressed in pancreatic islets from hypothalamic obese rats (2006) Endocrine, 29 (3), pp. 445-449Medeiros, A., Rolim, N.P., Oliveira, R.S., Rosa, K.T., Mattos, K.C., Casarini, D.E., Irigoyen, M.C., Brum, P.C., Exercise training delays cardiac dysfunction and prevents calcium handling abnormalities in sympathetic hyperactivity-induced heart failure mice (2008) The Journal of Applied Physiology, 104 (1), pp. 103-109Miller, R.E., Pancreatic neuroendocrinology: Peripheral neural mechanisms in the regulation of the islets of Langerhans (1981) Endocrine Reviews, 2 (4), pp. 471-494Narimiya, M., Yamada, H., Matsuba, I., Ikeda, Y., Tanese, T., Abe, M., Adrenergic modulation of insulin and glucagon secretion from the isolated perfused rat pancreas (1981) Endocrinologia Japonica, 28 (3), pp. 281-292Nishimura, H., Saltis, J., Habberfield, A.D., Garty, N.B., Greenberg, A.S., Cushman, S.W., Londos, C., Simpson, I.A., Phosphorylation state of the GLUT4 isoform of the glucose transporter in subfractions of the rat adipose cell: Effects of insulin, adenosine, and isoproterenol (1991) Proceedings of the National Academy of Sciences of the United States of America, 88 (24), pp. 11500-11504Panten, U., Kriegstein, E., Poses, W., Schönborn, J., Hasselblatt, A., Effects of L- Leucine and α-ketoisocaproic acid upon insulin secretion and metabolism of isolated pancreatic islets (1972) FEBS Letters, 20 (2), pp. 225-228Peterhoff, M., Sieg, A., Brede, M., Chao, C.M., Hein, L., Ulrich, S., Inhibition of insulin secretion via distinct signaling pathways in alpha2- adrenoceptor knockout mice (2003) European Journal of Endocrinology, 149 (4), pp. 343-350Rogers, P., Webb, G.B., Estimation of body fat in normal and obese mice (1980) The British Journal of Nutrition, 43 (1), pp. 83-86Saltiel, A.R., Kahn, C.R., Insulin signalling and the regulation of glucose and lipid metabolism (2001) Nature, 414 (6865), pp. 799-806Sener, A., Malaisse, W.J., L-Leucine and a nonmetabolized analogue activate pancreatic islet glutamate dhydrogenase (1980) Nature, 288 (5787), pp. 187-189Sjoholm, A., Alpha-adrenergic inhibition of rat pancreatic beta-cell replication and insulin secretion is mediated through a pertussis toxin-sensitive G-protein regulating islet cAMP content (1991) Biochemical and Biophysical Research Communications, 180 (1), pp. 152-155Smith, P.A., Sakura, H., Coles, B., Gummerson, N., Proks, P., Ashcroft, F.M., Electrogenic arginine transport mediates stimulus-secretion coupling im mouse pancreatic β-cells (1997) The Journal of Physiology, 499, pp. 625-635Weinhaus, A.J., Poronnik, P., Tuch, B.E., Cook, D.I., Mechanisms of arginine-induced increase in cytosolic calcium concentration in the beta-cell line NIT-1 (1997) Diabetologia, 40 (4), pp. 374-382Yamauchi, T., Tobe, K., Tamemoto, H., Ueki, K., Kaburagi, Y., Yamamoto-Honda, R., Takahashi, Y., Kadowaki, T., Insulin signalling and insulin actions in the muscles and livers of insulin-resistant, insulin receptor substrate 1-deficient mice (1996) Molecular and Cellular Biology, 16 (6), pp. 3074-3084Yang, J., Hodel, A., Holman, G.D., Insulin and isoproterenol have opposing roles in the maintenance of cytosol pH and optimal fusion of GLUT4 vesicles with the plasma membrane (2002) Jounal of Biological Chemistry, 277 (8), pp. 6559-6566White, M.F., Kahn, C.R., The insulin signaling system (1994) Journal of Biological Chemistry, 269 (1), pp. 1-4Zanquetta, M.M., Nascimento, M.E., Mori, R.C., D'Agord, S.B., Young, M.E., Machado, U.F., Participation of beta-adrenergic activity in modulation of GLUT4 expression during fasting and refeeding in rats (2006) Metabolism, 55 (11), pp. 1538-154
    corecore