Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Publication year range
1.
Clin Exp Pharmacol Physiol ; 42(2): 179-85, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25399833

ABSTRACT

Exercise training reverses atherosclerotic risk factors associated with metabolic syndrome and obesity. The aim of the present study was to determine the molecular anti-inflammatory, anti-oxidative and anti-atherogenic effects in aorta from rats with high-fat diet-induced obesity. Male Sprague-Dawley rats were placed on a high-fat (HFD) or control (CD) diet for 12 weeks. The HFD rats were then divided into four groups: (i) sedentary HFD-fed rats (HFD-S); (ii) exercise trained (motor treadmill 5 days/week, 60 min/day, 12 weeks) HFD-fed rats (HFD-Ex); (iii) modified diet (HFD to CD) sedentary rats (HF/CD-S); and (iv) an exercise-trained modified diet group (HF/CD-Ex). Tissue levels of NADPH oxidase (activity and expression), NADPH oxidase (Nox) 1, Nox2, Nox4, p47(phox) , superoxide dismutase (SOD)-1, angiotensin AT1 and AT2 receptors, phosphorylated mitogen-activated protein kinase (MAPK; extracellular signal-regulated kinase (ERK) 1/2, stress-activated protein kinase (SAPK)/c-Jun N-terminal kinase (JNK)) and vascular cell adhesion molecule-1 (VCAM-1) were determined in the aorta. Plasma cytokines (tumour necrosis factor (TNF)-α and interleukin (IL)-6) levels were also measured. Obesity was accompanied by increases in NADPH oxidase activity, p47(phox) translocation, Nox4 and VCAM-1 protein expression, MAPK (ERK1/2, SAPK/JNK) phosphorylation and plasma TNF-α and IL-6 levels. Exercise training and switching from the HFD to CD reversed almost all these molecular changes. In addition, training increased aortic SOD-1 protein expression and decreased ERK1/2 phosphorylation. These findings suggest that protective effects of exercise training on atherosclerotic risk factors induced by obesity are associated with downregulation of NADPH oxidase, ERK1/2 and SAPK/JNK activity and increased SOD-1 expression.


Subject(s)
Coronary Artery Disease/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System/physiology , NADPH Oxidases/metabolism , Obesity/metabolism , Physical Conditioning, Animal/physiology , Protein Kinases/metabolism , Animals , Diet, High-Fat/adverse effects , Down-Regulation/physiology , Interleukin-6/metabolism , Male , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation/physiology , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/metabolism , Risk Factors , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Tumor Necrosis Factor-alpha/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
2.
Cell Signal ; 25(11): 2163-75, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23838006

ABSTRACT

We demonstrated a role for the Mg(2+) transporter TRPM7, a bifunctional protein with channel and α-kinase domains, in aldosterone signaling. Molecular mechanisms underlying this are elusive. Here we investigated the function of TRPM7 and its α-kinase domain on Mg(2+) and pro-inflammatory signaling by aldosterone. Kidney cells (HEK-293) expressing wild-type human TRPM7 (WThTRPM7) or constructs in which the α-kinase domain was deleted (ΔKinase) or rendered inactive with a point mutation in the ATP binding site of the α-kinase domain (K1648R) were studied. Aldosterone rapidly increased [Mg(2+)]i and stimulated NADPH oxidase-derived generation of reactive oxygen species (ROS) in WT hTRPM7 and TRPM7 kinase dead mutant cells. Translocation of annexin-1 and calpain-II and spectrin cleavage (calpain target) were increased by aldosterone in WT hTRPM7 cells but not in α-kinase-deficient cells. Aldosterone stimulated phosphorylation of MAP kinases and increased expression of pro-inflammatory mediators ICAM-1, Cox-2 and PAI-1 in Δkinase and K1648R cells, effects that were inhibited by eplerenone (mineralocorticoid receptor (MR) blocker). 2-APB, a TRPM7 channel inhibitor, abrogated aldosterone-induced Mg(2+) responses in WT hTRPM7 and mutant cells. In 2-APB-treated ΔKinase and K1648R cells, aldosterone-stimulated inflammatory responses were unchanged. These data indicate that aldosterone stimulates Mg(2+) influx and ROS production in a TRPM7-sensitive, kinase-insensitive manner, whereas activation of annexin-1 requires the TRPM7 kinase domain. Moreover TRPM7 α-kinase modulates inflammatory signaling by aldosterone in a TRPM7 channel/Mg(2+)-independent manner. Our findings identify novel mechanisms for non-genomic actions of aldosterone involving differential signaling through MR-activated TRPM7 channel and α-kinase.


Subject(s)
Aldosterone/metabolism , Magnesium/metabolism , Protein Kinases/genetics , Signal Transduction , TRPM Cation Channels/genetics , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Annexin A1/metabolism , Binding Sites , Boron Compounds/pharmacology , Calpain/metabolism , Eplerenone , Gene Expression Regulation , HEK293 Cells , Humans , Ion Transport , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Phosphorylation , Protein Binding , Protein Kinases/deficiency , Protein Serine-Threonine Kinases , Protein Structure, Tertiary , Reactive Oxygen Species/metabolism , Spectrin/metabolism , Spironolactone/analogs & derivatives , Spironolactone/pharmacology , TRPM Cation Channels/antagonists & inhibitors , TRPM Cation Channels/metabolism
3.
Cardiovasc Res ; 91(4): 720-31, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21576132

ABSTRACT

AIMS: We demonstrated c-Src activation as a novel non-genomic signalling pathway for aldosterone in vascular smooth muscle cells (VSMCs). Here, we investigated molecular mechanisms and biological responses of this phenomenon, focusing on the role of lipid rafts/caveolae and platelet-derived growth factor receptor (PDGFR) in c-Src-regulated proinflammatory responses by aldosterone. METHODS AND RESULTS: Studies were performed in cultured VSMCs from Wistar-Kyoto (WKY) rats and caveolin-1 knockout (Cav 1(-/-)) and wild-type mice. Aldosterone stimulation increased c-Src phosphorylation and trafficking to lipid rafts/caveolae. Cholesterol depletion with methyl-ß-cyclodextrin abrogated aldosterone-induced phosphorylation of c-Src and its target, Pyk2. Aldosterone effects were recovered by cholesterol reload. Aldosterone-induced c-Src and cortactin phosphorylation was reduced in caveolin-1-silenced and Cav 1(-/-) VSMCs. PDGFR is phosphorylated by aldosterone within cholesterol-rich fractions of VSMCs. AG1296, a PDGFR inhibitor, prevented c-Src phosphorylation and translocation to cholesterol-rich fractions. Aldosterone induced an increase in adhesion molecule protein content and promoted monocyte adhesion to VSMCs, responses that were inhibited an by cholesterol depletion, caveolin-1 deficiency, AG1296 and PP2, a c-Src inhibitor. Mineralocorticoid receptor (MR) content in flotillin-2-rich fractions and co-immunoprecipitation with c-Src and PDGFR increased upon aldosterone stimulation, indicating MR-lipid raft/signalling association. CONCLUSION: We demonstrate that aldosterone-mediated c-Src trafficking/activation and proinflammatory signalling involve lipid rafts/caveolae via PDGFR.


Subject(s)
Aldosterone/pharmacology , Cholesterol/physiology , Inflammation/chemically induced , Membrane Microdomains/physiology , Muscle, Smooth, Vascular/drug effects , Protein-Tyrosine Kinases/physiology , Receptors, Platelet-Derived Growth Factor/physiology , Animals , CSK Tyrosine-Protein Kinase , Caveolae/physiology , Mice , Mice, Knockout , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Phosphorylation , Protein Transport , Rats , Rats, Inbred WKY , Receptors, Mineralocorticoid/physiology , src-Family Kinases
4.
Hypertension ; 51(4): 915-21, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18268139

ABSTRACT

Hyperaldosteronism is associated with hypertension, cardiovascular fibrosis, and electrolyte disturbances, including hypomagnesemia. Mechanisms underlying aldosterone-mediated Mg(2+) changes are unclear, but the novel Mg(2+) transporters TRPM6 and TRPM7 may be important. We examined whether aldosterone influences renal TRPM6/7 and the TRPM7 downstream target annexin-1 and tested the hypothesis that Mg(2+) administration ameliorates aldosterone-induced cardiovascular and renal injury and prevents aldosterone-associated hypertension. C57B6 mice were studied (12 weeks, n=8 to 9/group); (1) control group (0.2% dietary Mg(2+)), (2) Mg(2+) group (0.75% dietary Mg(2+)), (3) aldosterone group (Aldo, 400 microg/kg/min and 0.9% NaCl drinking water), and (4) Aldo+Mg(2+) group. Blood pressure was unaltered by aldosterone and was similar in all groups throughout the experiment. Serum Na(+) was increased and serum K(+) and Mg(2+) decreased in the Aldo group. Aldo mice had hypomagnesuria and proteinuria, and renal, cardiac, and aortic fibrosis, which were normalized by Mg(2+) supplementation. Renal and cardiovascular expression of interleukin-6, VCAM1 and COX2 was increased in the Aldo group. Magnesium attenuated renal and cardiac interleukin-6 content and decreased renal VCAM1 and cardiac COX2 expression (P<0.05). Aldosterone decreased expression of renal TRPM7 and the downstream target annexin-1 (P<0.05) without effect on TRPM6. Whereas Mg(2+) increased mRNA expression of TRPM6 and TRPM7, it had no effect on TRPM7 and annexin-1 protein content. Our data demonstrate that aldosterone mediates blood pressure-independent renal and cardiovascular fibrosis and inflammation through Mg(2+)-sensitive pathways. We suggest that altered Mg(2+) metabolism in hyperaldosteronism may relate to TRPM7 downregulation and that Mg(2+) protects against cardiovascular and renal damaging actions of aldosterone.


Subject(s)
Aldosterone/pharmacology , Kidney/pathology , Magnesium/pharmacology , Nephritis/pathology , TRPM Cation Channels/genetics , Animals , Blood Pressure/drug effects , Body Weight , Down-Regulation/drug effects , Down-Regulation/immunology , Fibrosis , Gene Expression/physiology , Hyperaldosteronism/chemically induced , Hyperaldosteronism/immunology , Hyperaldosteronism/pathology , Kidney/immunology , Magnesium/blood , Magnesium/urine , Male , Mice , Mice, Inbred C57BL , Nephritis/immunology , Organ Size , Sodium/blood , Sodium/urine , TRPM Cation Channels/metabolism
5.
Hypertension ; 51(2): 500-6, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18195161

ABSTRACT

We demonstrated previously that, in mice with chronic angiotensin II-dependent hypertension, gp91phox-containing NADPH oxidase is not involved in the development of high blood pressure, despite being important in redox signaling. Here we sought to determine whether a gp91phox homologue, Nox1, may be important in blood pressure elevation and activation of redox-sensitive pathways in a model in which the renin-angiotensin system is chronically upregulated. Nox1-deficient mice and transgenic mice expressing human renin (TTRhRen) were crossed, and 4 genotypes were generated: control, TTRhRen, Nox1-deficient, and TTRhRen Nox1-deficient. Blood pressure and oxidative stress (systemic and renal) were increased in TTRhRen mice (P<0.05). This was associated with increased NADPH oxidase activation. Nox1 deficiency had no effect on the development of hypertension in TTRhRen mice. Phosphorylation of c-Src, mitogen-activated protein kinases, and focal adhesion kinase was significantly increased 2- to 3-fold in kidneys from TTRhRen mice. Activation of c-Src, p38 mitogen-activated protein kinase, c-Jun N-terminal kinase, and focal adhesion kinase but not of extracellular signal regulated kinase 1/2 or extracellular signal regulated kinase 5, was reduced in TTRhRen/Nox1-deficient mice (P<0.05). Expression of procollagen III was increased in TTRhRen and TTRhRen/Nox1-deficient mice versus control mice, whereas vascular cell adhesion molecule-1 was only increased in TTRhRen mice. Our findings demonstrate that, in Nox1-deficient TTRhRen mice, blood pressure is elevated despite reduced NADPH oxidase activation, decreased oxidative stress, and attenuated redox signaling. Our results suggest that Nox1-containing NADPH oxidase plays a key role in the modulation of systemic and renal oxidative stress and redox-dependent signaling but not in the elevation of blood pressure in a model of chronic angiotensin II-dependent hypertension.


Subject(s)
Blood Pressure , Hypertension/physiopathology , Kidney/metabolism , NADH, NADPH Oxidoreductases/deficiency , Signal Transduction , Angiotensin II , Animals , CSK Tyrosine-Protein Kinase , Chronic Disease , Female , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Hypertension/chemically induced , Hypertension/metabolism , Male , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinases/metabolism , NADH, NADPH Oxidoreductases/metabolism , NADPH Oxidase 1 , Oxidation-Reduction , Oxidative Stress , Phosphorylation , Procollagen/metabolism , Protein Isoforms/metabolism , Protein-Tyrosine Kinases/metabolism , Renin/genetics , Renin/metabolism , Systole , Vascular Cell Adhesion Molecule-1/metabolism , src-Family Kinases
6.
Am J Physiol Regul Integr Comp Physiol ; 290(1): R73-8, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16109804

ABSTRACT

Intracellular Mg2+ depletion has been implicated in vascular dysfunction in hypertension. We demonstrated that transient receptor potential melastatin 7 (TRPM7) cation channels mediate Mg2+ influx in VSMCs. Whether this plays a role in [Mg2+]i deficiency in hypertension is unclear. Here, we tested the hypothesis that downregulation of TRPM7 and its homologue TRPM6 is associated with reduced [Mg2+]i and that ANG II negatively regulates TRPM6/7 in vascular smooth muscle cells (VSMCs) from spontaneously hypertensive rats (SHR). Cultured VSMCs from Wistar Kyoto (WKY) and SHR were studied. mRNA and protein expression of TRPM6 and TRPM7 were assessed by RT-PCR and immunoblotting, respectively. Translocation of annexin-1, specific TRPM7 substrate, was measured as an index of TRPM7 activation. [Mg2+]i was determined using mag fura-2. VSMCs from WKY and SHR express TRPM6 and TRPM7. Basal TRPM6 expression was similar in WKY and SHR, but basal TRPM7 content was lower in VSMCs from SHR vs. WKY. This was associated with significantly reduced [Mg2+]i in SHR cells (P < 0.01). ANG II time-dependently increased TRPM6 expression, with similar responses in WKY and SHR. ANG II significantly increased TRPM7 expression in WKY (P < 0.05), but not in SHR. Annexin-1 translocation was reduced 1.5-2-fold in SHR vs. WKY. Our findings demonstrate that TRPM6 and TRPM7 are differentially regulated in VSMCs from SHR and WKY. Whereas TRPM6 is unaltered in SHR, expression of TRPM7 is blunted. This was associated with attenuated annexin-1 translocation and decreased VSMC [Mg2+]i in SHR. Downregulation of TRPM7, but not TRPM6, may play a role in altered Mg2+ homeostasis in VSMCs from SHR.


Subject(s)
Angiotensin II/pharmacology , Gene Expression Regulation/drug effects , Hypertension/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , TRPM Cation Channels/metabolism , Animals , Annexin A1/metabolism , Annexins/metabolism , Cells, Cultured , Down-Regulation , Magnesium/metabolism , Protein Transport , RNA, Messenger/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , TRPM Cation Channels/genetics
7.
Clin Sci (Lond) ; 110(2): 243-53, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16271043

ABSTRACT

We have demonstrated recently [Callera, Touyz, Teixeira, Muscara, Carvalho, Fortes, Schiffrin and Tostes (2003) Hypertension 42, 811-817] that increased vascular oxidative stress in DOCA (deoxycorticosterone acetate)-salt rats is associated with activation of the ET (endothelin) system via ETA receptors. The exact source of ET-1-mediated oxidative stress remains unclear. The aim of the present study was to investigate whether ET-1 increases generation of ROS (reactive oxygen species) in DOCA-salt hypertension through NADPH-oxidase-dependent mechanisms. Xanthine oxidase, eNOS (endothelial nitric oxide synthase) and COX-2 (cyclo-oxygenase-2) were also examined as potential ET-1 sources of ROS as well as mitochondrial respiration. DOCA-salt and control UniNX (uninephrectomized) rats were treated with the ETA antagonist BMS182874 (40 mg.day(-1).kg(-1) of body weight) or vehicle. Plasma TBARS (thiobarbituric acid-reacting substances) were increased in DOCA-salt compared with UniNX rats. Activity of NADPH and xanthine oxidases in aorta, mesenteric arteries and heart was increased in DOCA-salt rats. BMS182874 decreased plasma TBARS levels without influencing NADPH and xanthine oxidase activities in DOCA-salt rats. Increased p22(phox) protein expression and increased p47(phox) membrane translocation in arteries from DOCA-salt by rats were not affected by BMS182874 treatment. Increased eNOS and COX-2 expression, also observed in aortas from DOCA-salt rats, was unaltered by BMS182874. Increased mitochondrial generation of ROS in DOCA-salt rats was normalized by BMS182874. ETA antagonism also increased the expression of mitochondrial MnSOD (manganese superoxide dismutase) in DOCA-salt rats. In conclusion, activation of NADPH oxidase does not seem to be the major source of oxidative stress induced by ET-1/ETA in DOCA-salt hypertension, which also appears to be independent of increased activation of xanthine oxidase or eNOS/COX-2 overexpression. Mitochondria may play a role in ET-1-driven oxidative stress, as evidenced by increased mitochondrial-derived ROS in this model of hypertension.


Subject(s)
Antihypertensive Agents/therapeutic use , Dansyl Compounds/therapeutic use , Endothelin-1/metabolism , Hypertension/metabolism , NADPH Oxidases , Animals , Aorta , Desoxycorticosterone , Endothelin A Receptor Antagonists , Endothelium, Vascular/metabolism , Male , Mesenteric Arteries , Mitochondria/metabolism , Models, Animal , Myocardium , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress , Rats , Rats, Wistar , Receptor, Endothelin A/metabolism , Sodium Chloride, Dietary , Superoxide Dismutase/metabolism , Thiobarbituric Acid Reactive Substances/metabolism , Xanthine Oxidase/metabolism
8.
Hypertension ; 46(4): 1032-8, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16157790

ABSTRACT

Aldosterone plays an important role in the pathogenesis of hypertension. We previously demonstrated that nongenomic signaling by aldosterone in vascular smooth muscle cells occurs through c-Src-dependent pathways. Here we tested the hypothesis that upregulation of c-Src by aldosterone plays a role in increased mitogen-activated protein (MAP) kinase activation, [3H]-proline incorporation, and NADPH-driven generation of reactive oxygen species, thereby inducing cell growth, collagen production, and inflammation, respectively, in vascular smooth muscle cells from spontaneously hypertensive rats. The time course of c-Src phosphorylation by aldosterone was shifted to the left in vascular myocytes from hypertensive animals. Aldosterone rapidly increased phosphorylation of p38 MAP kinase and extracellular signal-regulated kinase with significantly greater effects in cells from spontaneously hypertensive rats versus control cells (P<0.05). Aldosterone increased NADPH oxidase activity with significantly greater responses in vascular smooth muscle cells from hypertensive animals (P<0.05). These events were associated with enhanced [3H]proline incorporation (index of collagen synthesis) in cells from spontaneously hypertensive rats (P<0.05). The NADPH oxidase activity increase, collagen synthesis, c-Src, and MAP kinase phosphorylation induced by aldosterone were significantly reduced by eplerenone (selective mineralocorticoid receptor blocker) and PP2 (selective c-Src inhibitor). In conclusion, nongenomic signaling by exogenous aldosterone, mediated through c-Src, is increased in vascular smooth muscle cells from spontaneously hypertensive rats. Upregulation of c-Src signaling may be important in the profibrotic and proinflammatory actions of aldosterone in this genetic model of hypertension.


Subject(s)
Aldosterone/pharmacology , Hypertension/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , Rats, Inbred SHR/metabolism , Signal Transduction/drug effects , Animals , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Male , NADPH Oxidases/metabolism , Phosphorylation/drug effects , Proline/metabolism , Rats , Rats, Inbred WKY , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Rev. bras. hipertens ; 12(2): 84-88, abr.-jun. 2005. tab, graf
Article in Portuguese | LILACS | ID: lil-421619

ABSTRACT

Os efeitos produzidos por fármacos ou medicamentos utilizados no tratamento da hipertensão arterial são decorrentes de sua interação com componentes macromoleculares das células, como receptores, enzimas, proteínas transportadoras, canais iônicos. A ativação de um receptor modifica a função de componentes interligados ao mesmo, iniciando uma série de alterações intracelulares, como ativação de proteínas efetoras e geração de mensageiros secundários, que irão determinar mudanças na função celular. Existem quatro famílias de receptores: (1) acoplados a proteínas G; (2) nucleares; (3) com atividade enzimática intrínseca e (4) tipo canais iônicos. Os mensageiros secundários mais conhecidos são AMPc, GMPc, IP 3' DAG, íons Ca2+ e óxido nítrico. Os alvos celulares para os medicamentos utilizados no tratamento da hipertensão arterial são apresentados no texto


Subject(s)
Cardiovascular System , Hypertension/drug therapy , Membrane Transport Proteins , Receptors, Cytoplasmic and Nuclear , Antihypertensive Agents
SELECTION OF CITATIONS
SEARCH DETAIL
...