Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Eur J Clin Invest ; 54(5): e14172, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38293760

RESUMO

BACKGROUND: Glucocorticoid (GR) and mineralocorticoid (MR) receptors are highly expressed in cardiac tissue, and both can be activated by corticosteroids. MR activation, in acute myocardial infarction (AMI), worsens cardiac function, and increase NHE activity contributing to the deleterious process. In contrast, effects of GR activation are not fully understood, probably because of the controversial scenario generated by using different doses or potencies of corticosteroids. AIMS: We tested the hypothesis that an acute dose of hydrocortisone (HC), a low-potency glucocorticoid, in a murine model of AMI could be cardioprotective by regulating NHE1 activity, leading to a decrease in oxidative stress. MATERIALS AND METHODS: Isolated hearts from Wistar rats were subjected to regional ischemic protocol. HC (10 nmol/L) was added to the perfusate during early reperfusion. Infarct size and oxidative stress were determined. Isolated papillary muscles from non-infarcted hearts were used to evaluate HC effect on sodium-proton exchanger 1 (NHE1) by analysing intracellular pH recovery from acute transient acidosis. RESULTS: HC treatment decreased infarct size, improved cardiac mechanics, reduced oxidative stress after AMI, while restoring the decreased level of the pro-fusion mitochondrial protein MFN-2. Co-treatment with the GR-blocker Mifepristone avoided these effects. HC reduced NHE1 activity by increasing the NHE1 pro-inhibiting Ser648 phosphorylation site and its upstream kinase AKT. HC restored the decreased AKT phosphorylation and anti-apoptotic BCL-2 protein expression detected after AMI. CONCLUSIONS: Our results provide the first evidence that acute HC treatment during early reperfusion induces cardioprotection against AMI, associated with a non-genomic HC-triggered NHE1 inhibition by AKT and antioxidant action that might involves mitochondrial dynamics improvement.


Assuntos
Infarto do Miocárdio , Traumatismo por Reperfusão , Ratos , Camundongos , Animais , Miocárdio/metabolismo , Antioxidantes/farmacologia , Antioxidantes/metabolismo , Hidrocortisona/farmacologia , Hidrocortisona/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Glucocorticoides/farmacologia , Glucocorticoides/metabolismo , Ratos Wistar , Trocadores de Sódio-Hidrogênio , Infarto do Miocárdio/prevenção & controle , Infarto do Miocárdio/metabolismo , Traumatismo por Reperfusão/metabolismo
2.
Life Sci ; 312: 121219, 2023 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-36435222

RESUMO

Two alkalinizing mechanisms coexist in cardiac myocytes to maintain intracellular pH: sodium/bicarbonate cotransporter (electroneutral isoform NBCn1 and electrogenic isoform NBCe1) and sodium/proton exchanger (NHE1). Dysfunction of these transporters has previously been reported to be responsible for the development of cardiovascular diseases. The aim of this study was to evaluate the contribution of the downregulation of the NBCe1 to the development of cardiac hypertrophy. To specifically reduce NBCe1 expression, we cloned shRNA into a cardiotropic adeno-associated vector (AAV9-shNBCe1). After 28 days of being injected with AAV9-shNBCe1, the expression and the activity of NBCe1 in the rat heart were reduced. Strikingly, downregulation of NBCe1 causes significant hypertrophic heart growth, lengthening of the action potential in isolated myocytes, an increase in the duration of the QT interval and an increase in the frequency of Ca2+ waves without any significant changes in Ca2+ transients. An increased compensatory expression of NBCn1 and NHE1 was also observed. We conclude that reduction of NBCe1 is sufficient to induce cardiac hypertrophy and modify the electrical features of the rat heart.


Assuntos
Bicarbonatos , Simportadores de Sódio-Bicarbonato , Ratos , Animais , Simportadores de Sódio-Bicarbonato/genética , Simportadores de Sódio-Bicarbonato/metabolismo , Bicarbonatos/metabolismo , Cardiomegalia/genética , Cardiomegalia/metabolismo , Sódio/metabolismo , Isoformas de Proteínas/metabolismo , Concentração de Íons de Hidrogênio
3.
Biochim Biophys Acta Gen Subj ; 1867(2): 130278, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36410610

RESUMO

It has been demonstrated that supplementation with the two main omega 3 polyunsaturated fatty acids (ω3 FAs), docosahexaenoic acid (DHA), and eicosapentaenoic acid (EPA), leads to modifications in the cardiac physiology. ω3 FAs can affect the membrane's lipid composition, as well as proteins' location and/or function. The Na+/H+ exchanger (NHE1) is an integral membrane protein involved in the maintenance of intracellular pH and its hyperactivity has been associated with the development of various cardiovascular diseases such as cardiac hypertrophy. Our aim was to determine the effect of ω3 FAs on systolic blood pressure (SBP), lipid profiles, NHE1 activity, and cardiac function in spontaneously hypertensive rats (SHR) using Wistar rats (W) as normotensive control. After weaning, the rats received orally ω3 FAs (200 mg/kg body mass/day/ 4 months). We measured SBP, lipid profiles, and different echocardiography parameters, which were used to calculate cardiac hypertrophy index, systolic function, and ventricular geometry. The rats were sacrificed, and ventricular cardiomyocytes were obtained to measure NHE1 activity. While the treatment with ω3 FAs did not affect the SBP, lipid analysis of plasma revealed a significant decrease in omega-6/omega-3 ratio, correlated with a significant reduction in left ventricular mass index in SHR. The NHE1 activity was significantly higher in SHR compared with W. While in W the NHE1 activity was similar in both groups, a significant decrease in NHE1 activity was detected in SHRs supplemented with ω3 FAs, reaching values comparable with W. Altogether, these findings revealed that diet supplementation with ω3 FAs since early age prevents the development of cardiac hypertrophy in SHR, perhaps by decreasing NHE1 activity, without altering hemodynamic overload.


Assuntos
Ácidos Graxos Ômega-3 , Ratos , Animais , Ratos Wistar , Ácidos Graxos Ômega-3/farmacologia , Ácidos Docosa-Hexaenoicos , Ácido Eicosapentaenoico , Ratos Endogâmicos SHR , Cardiomegalia/tratamento farmacológico , Cardiomegalia/prevenção & controle
4.
Neuroscience ; 465: 154-165, 2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-33957206

RESUMO

Transient receptor potential ankyrin 1 (TRPA1) channel is expressed in a subset of nociceptive neurons. This channel integrates several nociceptive signals. Particularly, it is modulated by intracellular pH (pHi). Na+/H+ exchanger 1 (NHE1) contributes to the maintenance of pHi in nociceptors. However, it is currently unknown whether the interaction between TRPA1 and NHE1 contributes to the nociceptive processing. Thus, the purpose of this study was to assess the functional interaction between NHE1 and TRPA1 in small dorsal root ganglion (DRG) neurons from primary culture obtained from adult rats. Moreover, we also evaluated their possible interaction in acute and inflammatory pain. Zoniporide (selective NHE1 inhibitor) reduced pHi and increased intracellular calcium in a concentration-dependent fashion in DRG neurons. Zoniporide and allyl isothiocyanate (AITC, TRPA1 agonist) increased calcium transients in the same DRG neuron, whereas that A-967079 (TRPA1 antagonist) prevented the effect of zoniporide in DRG neurons. Repeated AITC induced TRPA1 desensitization and this effect was prevented by zoniporide. Both NHE1 and TRPA1 were localized at the membrane surface of DRG neurons in culture. Local peripheral zoniporide enhanced AITC-induced pronociception and this effect was prevented by A-967079. Likewise, zoniporide potentiated Complete Freund's Adjuvant (CFA)-induced hypersensitivity, effect which was prevented by A-967079 in vivo. CFA paw injection increased TRPA1 and decresed NHE1 protein expression in DRG. These results suggest a functional interaction between NHE1 and TRPA1 in DRG neurons in vitro. Moreover, data suggest that this interaction participates in acute and inflamatory pain conditions in vivo.


Assuntos
Gânglios Espinais , Canais de Potencial de Receptor Transitório , Animais , Neurônios , Nociceptividade , Ratos , Trocador 1 de Sódio-Hidrogênio , Canal de Cátion TRPA1
5.
Front Cardiovasc Med ; 8: 617519, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33693035

RESUMO

The cardiac Na+/H+ exchanger (NHE1) is a membrane glycoprotein fundamental for proper cell functioning due its multiple housekeeping tasks, including regulation of intracellular pH, Na+ concentration, and cell volume. In the heart, hyperactivation of NHE1 has been linked to the development of different pathologies. Several studies in animal models that reproduce the deleterious effects of ischemia/reperfusion injury or cardiac hypertrophy have conclusively demonstrated that NHE1 inhibition provides cardioprotection. Unfortunately, NHE1 inhibitors failed to reproduce these effects in the clinical arena. The reasons for those discrepancies are not apparent yet. However, a reasonable clue to consider would be that drugs that completely abolish the exchanger activity, including that its essential housekeeping function may not be the best therapeutic approach. Therefore, interventions tending to specifically reduce its hyperactive state without affecting its basal activity emerge as a novel potential gold standard. In this regard, a promising goal seems to be the modulation of the phosphorylation state of the cytosolic tail of the exchanger. Recent own experiments demonstrated that Sildenafil, a phosphodiesterase 5A inhibitor drug that has been widely used for the treatment of erectile dysfunction is able to decrease NHE1 phosphorylation, and hence reduce its hyperactivity. In connection, growing evidence demonstrates cardioprotective properties of Sildenafil against different cardiac pathologies, with the distinctive characteristic of directly affecting cardiac tissue without altering blood pressure. This mini-review was aimed to focus on the regulation of NHE1 activity by Sildenafil. For this purpose, experimental data reporting Sildenafil effects in different animal models of heart disease will be discussed.

6.
Front Cardiovasc Med ; 8: 622583, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33718450

RESUMO

The stretch of cardiac muscle increases developed force in two phases. The first phase occurs immediately after stretch and is the expression of the Frank-Starling mechanism, while the second one or slow force response (SFR) occurs gradually and is due to an increase in the calcium transient amplitude. An important step in the chain of events leading to the SFR generation is the increased production of reactive oxygen species (ROS) leading to redox sensitive ERK1/2, p90RSK, and NHE1 phosphorylation/activation. Conversely, suppression of ROS production blunts the SFR. The purpose of this study was to explore whether overexpression of the ubiquitously expressed antioxidant molecule thioredoxin-1 (TRX1) affects the SFR development and NHE1 phosphorylation. We did not detect any change in basal phopho-ERK1/2, phopho-p90RSK, and NHE1 expression in mice with TRX1 overexpression compared to wild type (WT). Isolated papillary muscles from WT or TRX1-overexpressing mice were stretched from 92 to 98% of its maximal length. A prominent SFR was observed in WT mice that was completely canceled in TRX1 animals. Interestingly, myocardial stretch induced a significant increase in NHE1 phosphorylation in WT mice that was not detected in TRX1-overexpressing mice. These novel results suggest that magnification of cardiac antioxidant defense power by overexpression of TRX1 precludes NHE1 phosphorylation/activation after stretch, consequently blunting the SFR development.

7.
Eur J Pharmacol ; 891: 173724, 2021 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-33152335

RESUMO

Previously, we have shown that an increased cGMP-activated protein Kinase (PKG) activity after phosphodiesterase 5 (PDE5) inhibition by Sildenafil (SIL), leads to myocardial Na+/H+ exchanger (NHE1) inhibition preserving its basal homeostatic function. Since NHE1 is hyperactive in the hypertrophied myocardium of spontaneous hypertensive rats (SHR), while its inhibition was shown to prevent and revert this pathology, the current study was aimed to evaluate the potential antihypertrophic effect of SIL on adult SHR myocardium. We initially tested the inhibitory capability of SIL on NHE1 in isolated cardiomyocytes of SHR by comparing H+ efflux during the recovery from an acid load. After confirmed that effect, eight-month-old SHR were chronically treated for one month with SIL through drinking water. Compared to their littermate controls, SIL-treated rats presented a decreased NHE1 activity, which correlated with a reduction in its phosphorylation level assigned to activation of a PKG-p38 MAP kinase-PP2A signaling pathway. Moreover, treated animals showed a decreased oxidative stress that appears to be a consequence of a decreased mitochondrial NHE1 phosphorylation. Treated SHR showed a significant reduction in the pro-hypertrophic phosphatase calcineurin, despite slight tendency to decrease hypertrophy was detected. When SIL treatment was prolonged to three months, a significant decrease in myocardial hypertrophy and interstitial fibrosis that correlated with a lower myocardial stiffness was observed. In conclusion, the current study provides evidence concerning the ability of SIL to revert established cardiac hypertrophy in SHR, a clinically relevant animal model that resembles human essential hypertension.


Assuntos
Cardiomegalia/prevenção & controle , Miócitos Cardíacos/efeitos dos fármacos , Músculos Papilares/efeitos dos fármacos , Inibidores da Fosfodiesterase 5/farmacologia , Citrato de Sildenafila/farmacologia , Trocador 1 de Sódio-Hidrogênio/metabolismo , Animais , Cardiomegalia/enzimologia , Cardiomegalia/etiologia , Cardiomegalia/fisiopatologia , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Modelos Animais de Doenças , Regulação para Baixo , Fibrose , Hipertensão/complicações , Masculino , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/patologia , Músculos Papilares/enzimologia , Músculos Papilares/fisiopatologia , Fosforilação , Proteína Fosfatase 2/metabolismo , Ratos Endogâmicos SHR , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
8.
Front Cardiovasc Med ; 7: 614279, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33553262

RESUMO

Current evidence reveals that cardiac mineralocorticoid receptor (MR) activation following myocardial stretch plays an important physiological role in adapting developed force to sudden changes in hemodynamic conditions. Its underlying mechanism involves a previously unknown nongenomic effect of the MR that triggers redox-mediated Na+/H+ exchanger (NHE1) activation, intracellular Na+ accumulation, and a consequent increase in Ca2+ transient amplitude through reverse Na+/Ca2+ exchange. However, clinical evidence assigns a detrimental role to MR activation in the pathogenesis of severe cardiac diseases such as congestive heart failure. This mini review is meant to present and briefly discuss some recent discoveries about locally triggered cardiac MR signals with the objective of shedding some light on its physiological but potentially pathological consequences in the heart.

9.
Biochem Pharmacol ; 170: 113667, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31622577

RESUMO

Pathological cardiac hypertrophy (PCH) can be triggered by epidermal growth factor receptor (EGFR) transactivation. Progression of PCH can be prevented by inhibition of hyperactive Na+/H+ exchanger isoform 1 (NHE1). We first aimed, to limit PCH of spontaneously hypertensive rats (SHR) by specific and localized silencing of cardiac EGFR, and second to study the connection of its activation pathway with cardiac NHE1 activity. Short hairpin RNA (shRNA) against EGFR was delivered with a lentivirus (l-shEGFR) in the cardiac left ventricle (LV) wall. Protein expression was analyzed by immunoblots, and NHE1 activity was indirectly measured in isolated papillary muscles by rate of pHi recovery from transient acidification. EGFR protein expression in the LV was reduced compared to the group injected with l-shSCR (Scrambled sequence) without changes in ErbB2 or ErbB4. Hypertrophic parameters together with cardiomyocytes cross sectional area were reduced in animals injected with l-shEGFR. Echocardiographic analysis exhibited a reduced fractional shortening in the l-shSCR group 30 days following treatment that was not observed in l-shEGFR group. l-shEGFR treated rats presented a reduced basal production of reactive oxygen species and decreased lipid peroxidation. NHE1 activity was significantly diminished in hearts with a partial EGFR silencing, without modification of its protein expression. We conclude that specifically silencing cardiac EGFR expression prevents progression of PCH through a pathway that involves a decrease in the NHE1 activity. Lentiviral vectors prove to be a valuable tool for long term expression of shRNA, bringing the possibility to extend its use in clinical area.


Assuntos
Cardiomegalia/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Inativação Gênica/fisiologia , Trocador 1 de Sódio-Hidrogênio/metabolismo , Animais , Cardiomegalia/patologia , Receptores ErbB/antagonistas & inibidores , Células HEK293 , Humanos , Masculino , Ratos , Ratos Endogâmicos SHR , Trocador 1 de Sódio-Hidrogênio/antagonistas & inibidores
10.
J Mol Cell Cardiol ; 136: 53-63, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31518570

RESUMO

Heart failure is the leading cause of death among diabetic people. Cellular and molecular entities leading to diabetic cardiomyopathy are, however, poorly understood. Coupling of cardiac carbonic anhydrase II (CAII) and Na+/H+ exchanger 1 (NHE1) to form a transport metabolon was analyzed in obese type 2 diabetic mice (ob-/-) and control heterozygous littermates (ob+/-). Echocardiography showed elevated systolic interventricular septum thickness and systolic posterior wall thickness in ob-/- mice at 9 and 16 weeks. ob-/- mice showed increased left ventricular (LV) weight/tibia length ratio and increased cardiomyocyte cross sectional area as compared to controls, indicating cardiac hypertrophy. Immunoblot analysis showed increased CAII expression in LV samples of ob-/-vs. ob+/- mice, and augmented Ser703 phosphorylation on NHE1 in ob-/- hearts. Reciprocal co-immunoprecipitation analysis showed strong association of CAII and NHE1 in LV samples of ob-/- mice. NHE1-dependent rate of intracellular pH (pHi) normalization after transient acid loading of isolated cardiomyocytes was higher in ob-/- mice vs. ob+/-. NHE transport activity was also augmented in cultured H9C2 rat cardiomyoblasts treated with high glucose/high palmitate, and it was normalized after CA inhibition. We conclude that the NHE1/CAII metabolon complex is exacerbated in diabetic cardiomyopathy of ob-/- mice, which may lead to perturbation of pHi and [Na+] and [Ca2+] handling in these diseased hearts.


Assuntos
Anidrase Carbônica II/metabolismo , Cardiomegalia/patologia , Diabetes Mellitus Tipo 2/complicações , Trocador 1 de Sódio-Hidrogênio/metabolismo , Animais , Anidrase Carbônica II/antagonistas & inibidores , Inibidores da Anidrase Carbônica/farmacologia , Cardiomegalia/diagnóstico por imagem , Cardiomegalia/metabolismo , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Eletrocardiografia , Etoxzolamida/farmacologia , Feminino , Ventrículos do Coração/patologia , Concentração de Íons de Hidrogênio , Camundongos Mutantes , Camundongos Transgênicos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Fosforilação , Processamento de Proteína Pós-Traducional , Ratos , Serina/metabolismo
11.
Cell Physiol Biochem ; 52(2): 172-185, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30816666

RESUMO

BACKGROUND/AIMS: Myocardial stretch increases cardiac force in two consecutive phases: The first one due to Frank-Starling mechanism, followed by the gradually developed slow force response (SFR). The latter is the mechanical counterpart of an autocrine/paracrine mechanism involving the release of angiotensin II (Ang II) and endothelin (ET) leading to Na⁺/H⁺ exchanger 1 (NHE-1) phosphorylation and activation. Since previous evidence indicates that p38-MAP kinase (p38-MAPK) negatively regulates the Ang II-induced NHE1 activation in vascular smooth muscle and the positive inotropic effect of ET in the heart, we hypothesized that this kinase might modulate the magnitude of the SFR to stretch. METHODS: Experiments were performed in isolated rat papillary muscles subjected to sudden stretch from 92 to 98% of its maximal length, in the absence or presence of the p38-MAPK inhibitor SB202190, or its inactive analogous SB202474. Western blot technique was used to determine phosphorylation level of p38-MAPK, ERK1/2, p90RSK and NHE-1 (previously immunoprecipitated with NHE-1 polyclonal antibody). Dual specificity phosphatase 6 (DUSP6) expression was evaluated by RT-PCR and western blot. Additionally, the Na⁺-dependent intracellular pH recovery from an ammonium prepulse-induced acid load was used to asses NHE-1 activity. RESULTS: The SFR was larger under p38-MAPK inhibition (SB202190), effect that was not observed in the presence of an inactive analogous (SB202474). Myocardial stretch activated p38-MAPK, while pre-treatment with SB202190 precluded this effect. Inhibition of p38-MAPK increased stretched-induced NHE-1 phosphorylation and activity, key event in the SFR development. Consistently, p38-MAPK inhibition promoted a greater increase in ERK1/2-p90RSK phosphorylation/activation after myocardial stretch, effect that may certainly be responsible for the observed increase in NHE-1 phosphorylation under this condition. Myocardial stretch induced up-regulation of the DUSP6, which specifically dephosphorylates ERK1/2, effect that was blunted by SB202190. CONCLUSION: Taken together, our data support the notion that p38-MAPK activation after myocardial stretch restricts the SFR by limiting ERK1/2-p90RSK phosphorylation, and consequently NHE-1 phosphorylation/activity, through a mechanism that involves DUSP6 up-regulation.


Assuntos
Fosfatase 6 de Especificidade Dupla/biossíntese , Regulação Enzimológica da Expressão Gênica , Sistema de Sinalização das MAP Quinases , Contração Miocárdica , Miocárdio/enzimologia , Regulação para Cima , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Imidazóis/farmacologia , Fosforilação/efeitos dos fármacos , Piridinas/farmacologia , Ratos , Ratos Wistar , Trocador 1 de Sódio-Hidrogênio/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
12.
Eur J Pharmacol ; 849: 96-105, 2019 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-30721701

RESUMO

Since the original description as potent antianginal compounds, phosphodiesterase 5A inhibitors have continuously increased their possible therapeutic applications. In the heart, Sildenafil was shown to protect against an ischemic insult by decreasing cardiac Na+/H+ exchanger (NHE1) activity, action that was mediated by protein kinase G. p38 mitogen activated protein kinase (p38MAPK) activation was described in cardiac ischemia, but its precise role remains elusive. It has been shown that p38MAPK is activated by protein kinase G (PKG) in certain non-cardiac tissues, while in others modulates NHE1 activity. Current study was aimed to seek the role of p38MAPK in the Sildenafil-triggered pathway leading to NHE1 inhibition in myocardium. Rat isolated papillary muscles were used to evaluate NHE1 activity during intracellular pH recovery from an acidic load. Protein kinases phosphorylation (activation) was determined by western blot. Sustained acidosis promoted NHE1 hyperactivity by enhancing Ser703 phosphorylation, effect that was blunted by Sildenafil. p38MAPK inhibition reversed the effect of Sildenafil on NHE1. Activation of p38MAPK, by Sodium Arsenite or Anisomycin, mimicked the inhibitory effect of Sildenafil on the exchanger. Consistently, Sildenafil induced p38MAPK phosphorylation/activation during acidosis. Neither Sildenafil nor p38MAPK inhibition affected extracellular signal-regulated kinases 1/2 phosphorylation, kinases upstream NHE1. Furthermore, inhibition of NHE1 after p38MAPK activation was precluded by preventing the activation of protein phosphatase 2A with Okadaic Acid. Taken together, these results suggest that activation of p38MAPK is a necessary step to trigger the inhibitory effect of Sildenafil on cardiac NHE1 activity, thorough a mechanism that involves protein phosphatase 2A-mediated exchanger dephosphorylation.


Assuntos
Coração/efeitos dos fármacos , Miocárdio/metabolismo , Citrato de Sildenafila/farmacologia , Trocadores de Sódio-Hidrogênio/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Acidose/enzimologia , Acidose/metabolismo , Acidose/patologia , Animais , Ativação Enzimática/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Miocárdio/citologia , Miocárdio/patologia , Fosforilação/efeitos dos fármacos , Proteína Fosfatase 2/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Wistar , Trocador 1 de Sódio-Hidrogênio/metabolismo
13.
Exp Mol Pathol ; 107: 1-9, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30664842

RESUMO

Chronic activation of the renin angiotensin system (RAS) favors several cardiac diseases, among which myocardial hypertrophy occupies an outstanding place. In this context, the hyperactivity of the cardiac Na+/H+ (NHE-1) exchanger plays a key role. The pathologic remodeling of the myocardium constitutes an independent risk factor for morbidity and mortality with continuously increasing healthcare cost. Therefore, the development of better therapeutic strategies emerges as highly mandatory. Our goal was to prevent angiotensin II (ANGII)-induced cardiac hypertrophy by NHE-1 gene silencing in Wistar rats. The intramyocardial injection of a lentivirus coding a specific small interference RNA (l-shNHE1) significantly reduced NHE-1 expression exclusively in the heart (~ 50%) and prevented cardiac remodeling in rats exposed to chronic infusion of ANG II (heart weigh/tibia length: 24,03 ±â€¯0,7915 mg/mm vs 28,45 ±â€¯0,9779 mg/mm and collagen volume fraction 2526 ±â€¯0,5003 vs 5982 ±â€¯1043 in l-shNHE1 + ANGII and ANGII, respectively). Interestingly, this was accompanied by an improvement in cardiac function determined by echocardiography even though blood pressure remained elevated (Fractional shortening 0,5960 ±â€¯0,4228 vs -0,9567 ±â€¯0,06888 and blood pressure at the end of ANGII treatment 141,2 ±â€¯6117 mmHg vs 134,1 ±â€¯6723 mmHg; in l-shNHE1 + ANGII and ANGII, respectively). ANGII infusion increased myocardial NADPH oxidase activity but the l-shNHE1 injection prevented oxidative stress as revealed by the normalization of lipid peroxidation (T-BARS 12,40 ±â€¯2887.vs 23,05 ±â€¯1537 in l-shNHE1 + ANGII and ANGII, respectively). These results allow as to propose the partial silencing of the cardiac NHE-1 through lentiviral injection as a promising tool in the prevention of ANGII-induced cardiac hypertrophy.


Assuntos
Angiotensina II/metabolismo , Técnicas de Silenciamento de Genes/métodos , Trocador 1 de Sódio-Hidrogênio/antagonistas & inibidores , Remodelação Ventricular/fisiologia , Animais , Cardiomegalia/metabolismo , Masculino , Ratos , Ratos Wistar
14.
BMC Nephrol ; 19(1): 179, 2018 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-30005635

RESUMO

BACKGROUND: Angiotensin II (Ang II) contributes to the progression of renal diseases associated with proteinuria and glomerulosclerosis mainly by inducing podocyte apoptosis. In the present study, we investigated whether the chronic effects of Ang II via AT1 receptor (AT1R) would result in endoplasmic reticulum (ER) stress/PKC-delta/p38 MAPK stimulation, and consequently podocyte apoptosis. METHODS: Wistar rats were treated with Ang II (200 ng·kg-1·min-1, 42 days) and or losartan (10 mg·kg-1·day-1, 14 days). Immortalized mouse podocyte were treated with 1 µM Ang II and/or losartan (1 µM) or SB203580 (0.1 µM) (AT1 receptor antagonist and p38 MAPK inhibitor) for 24 h. Kidney sections and cultured podocytes were used to evaluate protein expression by immunofluorescence and immunoblotting. Apoptosis was evaluated by flow cytometry and intracellular pH (pHi) was analyzed using microscopy combined with the fluorescent probe BCECF/AM. RESULTS: Compared with controls, Ang II via AT1R increased chaperone GRP 78/Bip protein expression in rat glomeruli (p < 0.001) as well as in podocyte culture (p < 0.01); increased phosphorylated eIf2-α (p < 0.05), PKC-delta (p < 0.01) and p38 MAPK (p < 0.001) protein expression. Furthermore, Ang II induced p38 MAPK-mediated late apoptosis and increased the Bax/Bcl-2 ratio (p < 0.001). Simultaneously, Ang II via AT1R induced p38 MAPK-NHE1-mediated increase of pHi recovery rate after acid loading. CONCLUSION: Together, our results indicate that Ang II-induced podocyte apoptosis is associated with AT1R/ER stress/PKC-delta/p38 MAPK axis and enhanced NHE1-mediated pHi recovery rate.


Assuntos
Angiotensina II/toxicidade , Estresse do Retículo Endoplasmático/fisiologia , Podócitos/metabolismo , Proteína Quinase C-delta/metabolismo , Trocador 1 de Sódio-Hidrogênio/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Linhagem Celular Transformada , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Camundongos , Podócitos/efeitos dos fármacos , Isoformas de Proteínas/metabolismo , Distribuição Aleatória , Ratos , Ratos Wistar
15.
Acta Physiol (Oxf) ; 224(2): e13092, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-31595734

RESUMO

AIM: Myocardial Na+/H+ exchanger-1 (NHE-1) hyperactivity and oxidative stress are interrelated phenomena playing pivotal roles in the development of pathological cardiac hypertrophy and heart failure. Exercise training is effective to convert pathological into physiological hypertrophy in the spontaneously hypertensive rats (SHR), and IGF-1-key humoral mediator of exercise training-inhibits myocardial NHE-1, at least in normotensive rats. Therefore, we hypothesize that IGF-1 by hampering NHE-1 hyperactivity and oxidative stress should exert a cardioprotective effect in the SHR. METHODS: NHE-1 activity [proton efflux ( JH+ ) mmol L-1 min-1], expression and phosphorylation; H2O2 production; superoxide dismutase (SOD) activity; contractility and calcium transients were measured in SHR hearts in the presence/absence of IGF-1. RESULTS: IGF-1 significantly decreased NHE-1 activity ( JH+ at pHi 6.95: 1.39 ± 0.32, n = 9 vs C 3.27 ± 0.3, n = 20, P < .05); effect prevented by AG1024, an antagonist of IGF-1 receptor (2.7 ± 0.4, n = 7); by the PI3K inhibitor wortmannin (3.14 ± 0.41, n = 7); and the AKT inhibitor MK2206 (3.37 ± 0.43, n = 14). Moreover, IGF-1 exerted an antioxidant effect revealed by a significant reduction in H2O2 production accompanied by an increase in SOD activity. In addition, IGF-1 improved cardiomyocyte contractility as evidenced by an increase in sarcomere shortening and a decrease in the relaxation constant, underlined by an increase in the amplitude and rate of decay of the calcium transients. CONCLUSION: IGF-1 exerts a cardioprotective role on the hypertrophied hearts of the SHR, in which the inhibition of NHE-1 hyperactivity, as well as the positive inotropic and antioxidant effects, emerges as key players.


Assuntos
Fator de Crescimento Insulin-Like I/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Trocador 1 de Sódio-Hidrogênio/metabolismo , Animais , Cardiomegalia , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos , Miocárdio/metabolismo , Ratos , Ratos Endogâmicos SHR , Ratos Wistar , Trocador 1 de Sódio-Hidrogênio/antagonistas & inibidores , Trocador 1 de Sódio-Hidrogênio/genética
16.
Vet Immunol Immunopathol ; 191: 68-73, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28895869

RESUMO

Cytoplasmic pH homeostasis is required for an appropriate response in polymorphonuclear neutrophils (PMNs). In these cells, chemotaxis and reactive oxygen species (ROS) production are reduced by the use of Na+/H+ exchanger (NHE-1) inhibitors, but these results are mainly obtained using amiloride, a non-selective NHE-1 inhibitor. In bovine PMNs, the role of NHE-1 in functional responses has not been confirmed yet. The aim of this study was to determine the role of NHE-1 using amiloride and zoniporide in pH regulation, ROS production, matrix metalloproteinase 9 (MMP-9) release and calcium flux in bovine PMNs induced by the platelet activation factor (PAF), additionally we evaluated the presence of NHE-1 and NHE-2 mRNA Our data show the presence only of NHE-1 but not NHE-2 in bovine PMNs. Amiloride or zoniporide inhibited the intracellular alkalization induced by PAF without affecting calcium flux. Amiloride diminished ROS production and MMP-9 release, while zoniporide enhanced ROS production without change the MMP-9 release induced by PAF. Our work led us to conclude that changes in intracellular pH induced by PAF are regulated by NHE-1 in bovine neutrophils, but the effects of amiloride on ROS production and MMP-9 release induced by PAF are not NHE-1 dependent.


Assuntos
Amilorida/farmacologia , Metaloproteinase 9 da Matriz/metabolismo , Neutrófilos/efeitos dos fármacos , Fator de Ativação de Plaquetas/antagonistas & inibidores , Explosão Respiratória/efeitos dos fármacos , Trocador 1 de Sódio-Hidrogênio/antagonistas & inibidores , Animais , Cálcio/metabolismo , Bovinos , Feminino , Guanidinas/farmacologia , Neutrófilos/enzimologia , Neutrófilos/metabolismo , Fator de Ativação de Plaquetas/farmacologia , Reação em Cadeia da Polimerase/veterinária , Pirazóis/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Trocador 1 de Sódio-Hidrogênio/metabolismo
17.
J Appl Physiol (1985) ; 118(9): 1154-60, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25749446

RESUMO

We examined the effect of specific and local silencing of sodium/hydrogen exchanger isoform 1 (NHE1) with a small hairpin RNA delivered by lentivirus (L-shNHE1) in the cardiac left ventricle (LV) wall of spontaneously hypertensive rats, to reduce cardiac hypertrophy. Thirty days after the lentivirus was injected, NHE1 protein expression was reduced 53.3 ± 3% in the LV of the L-shNHE1 compared with the control group injected with L-shSCR (NHE1 scrambled sequence), without affecting its expression in other organs, such as liver and lung. Hypertrophic parameters as LV weight-to-body weight and LV weight-to-tibia length ratio were significantly reduced in animals injected with L-shNHE1 (2.32 ± 0.5 and 19.30 ± 0.42 mg/mm, respectively) compared with L-shSCR-injected rats (2.68 ± 0.06 and 21.53 ± 0.64 mg/mm, respectively). Histochemical analysis demonstrated a reduction of cardiomyocytes cross-sectional area in animals treated with L-shNHE1 compared with L-shSCR (309,81 ± 20,86 vs. 424,52 ± 21 µm(2), P < 0.05). Echocardiography at the beginning and at the end of the treatment showed that shNHE1 expression for 30 days induced 9% reduction of LV mass. Also, animals treated with L-shNHE1 exhibited a reduced LV wall thickness without changing LV diastolic dimension and arterial pressure, indicating an increased parietal stress. In addition, midwall shortening was not modified, despite the increased wall tension, suggesting an improvement of cardiac function. Chronic shNHE1 expression in the heart emerges as a possible methodology to reduce pathological cardiac hypertrophy, avoiding potentially undesired effects caused from a body-wide inhibition of NHE1.


Assuntos
Cardiomegalia/genética , Cardiomegalia/patologia , Inativação Gênica/fisiologia , Miocárdio/metabolismo , Trocadores de Sódio-Hidrogênio/genética , Animais , Pressão Arterial/genética , Pressão Arterial/fisiologia , Linhagem Celular , Diástole/genética , Diástole/fisiologia , Ecocardiografia/métodos , Células HEK293 , Ventrículos do Coração/metabolismo , Ventrículos do Coração/patologia , Humanos , Masculino , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , RNA Interferente Pequeno/genética , Ratos , Ratos Endogâmicos SHR/genética , Ratos Endogâmicos SHR/fisiologia , Trocador 1 de Sódio-Hidrogênio
18.
Cardiovasc Pathol ; 24(4): 236-40, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25670255

RESUMO

BACKGROUND: Reactive oxygen species, such as superoxide, are being increasingly recognized as key components of a vast array of signaling pathways. Angiotensin II is a well-recognized stimulus for superoxide production through NADPH oxidase activation and opening of the mitochondrial ATP-sensitive potassium channels (mKATP). A role for this mechanism has been proposed to explain several physiological effects of the peptide. The aim of this study was to evaluate the involvement of this mechanism in the inotropic response to 100nmol/L angiotensin II. METHODS: Sarcomere shortening and intracellular pH (BCECF-epifluorescence technique) were evaluated in isolated cat ventricular myocytes placed in a perfusion chamber on the stage of an inverted microscope. Myocardial superoxide production was evaluated by the lucigenin quimioluminiscence method. RESULTS: Angiotensin II (100nmol/L) increased~70% sarcomere shortening, effect that was only partially prevented by NADPH oxidase inhibition, mKATP channel blockade or inhibition of the cardiac Na(+)/H(+) exchanger (NHE-1). Moreover, angiotensin II stimulates NHE-1 activity by a NADPH oxidase-dependent mechanism. Myocardial superoxide production was also increased by angiotensin II, and this action was completely prevented either by NADPH oxidase inhibition or mKATP channel blockade. CONCLUSIONS: The positive inotropic response to 100nmol/L angiotensin II is due to both ROS/NHE-1 dependent and independent pathways, this being a point of divergence with the signaling previously described to be triggered by lower concentrations of angiotensin II (i.e.: 1nmol/L).


Assuntos
Angiotensina II/farmacologia , Cardiotônicos/farmacologia , Ventrículos do Coração/efeitos dos fármacos , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Superóxidos/metabolismo , Animais , Gatos , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Ventrículos do Coração/metabolismo , Concentração de Íons de Hidrogênio , Miócitos Cardíacos/metabolismo , NADPH Oxidases/antagonistas & inibidores , NADPH Oxidases/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/metabolismo , Sarcômeros/efeitos dos fármacos , Sarcômeros/metabolismo , Transdução de Sinais/efeitos dos fármacos , Trocadores de Sódio-Hidrogênio/antagonistas & inibidores , Trocadores de Sódio-Hidrogênio/metabolismo
19.
J Mol Cell Cardiol ; 76: 186-95, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25240639

RESUMO

BACKGROUND: The involvement of NHE-1 hyperactivity, critical for pathological cardiac hypertrophy (CH), in physiological CH has not been elucidated yet. Stimulation of NHE-1 increases intracellular Na(+) and Ca(2+) favouring calcineurin activation. Since myocardial stretch, an activator of NHE-1, is common to both types of CH, we speculate that NHE-1 hyperactivity may also happen in physiological CH. However, calcineurin activation is characteristic only for pathological hypertrophy. We hypothesize that an inhibitory AKT-dependent mechanism prevents NHE-1 hyperactivity in the setup of physiological CH. METHODS: Physiological CH was induced in rats by swimming (90 min/day, 12 weeks) or in cultured isolated cardiomyocytes with IGF-1 (10 nmol/L). RESULTS: Training induced eccentric CH development (left ventricular weight/tibial length: 22.0±0.3 vs. 24.3±0.7 mg/mm; myocyte cross sectional area: 100±3.2 vs. 117±4.1 %; sedentary (Sed) and swim-trained (Swim) respectively; p<0.05] with decreased myocardial stiffness and collagen deposition [1.7±0.05 % (Sed) vs. 1.4±0.09 % (Swim); p<0.05]. Increased phosphorylation of AKT, ERK1/2, p90(RSK) and NHE-1 at the consensus site for ERK1/2-p90(RSK) were detected in the hypertrophied hearts (P-AKT: 134±10 vs. 100±5; P-ERK1/2: 164±17 vs. 100±18; P-p90(RSK): 160±18 vs. 100±9; P-NHE-1 134±10 vs. 100±10; % in Swim vs. Sed respectively; p<0.05). No significant changes were detected neither in calcineurin activation [calcineurin Aß 100±10 (Sed) vs. 96±12 (Swim)], nor NFAT nuclear translocation [100±3.11 (Sed) vs. 95±9.81 % (Swim)] nor NHE-1 expression [100±8.5 (Sed) vs. 95±6.7 % (Swim)]. Interestingly, the inhibitory phosphorylation of the NHE-1 consensus site for AKT was increased in the hypertrophied myocardium (151.6±19.4 (Swim) vs. 100±9.5 % (Sed); p<0.05). In isolated cardiomyocytes 24 hours IGF-1 increased cell area (114±1.3 %; p<0.05) and protein/DNA content (115±3.9 %, p<0.05), effects not abolished by NHE-1 inhibition with cariporide (114±3 and 117±4.4 %, respectively). IGF-1 significantly decreased NHE-1 activity during pHi recovery from sustained intracellular acidosis (JH+ at pHi 6.8: 4.08±0.74 and 9.09±1.21 mmol/L/min, IGF-1 vs. control; p<0.05), and abolished myocardial slow force response, the mechanical counterpart of stretch-induced NHE-1 activation. CONCLUSIONS: NHE-1 hyperactivity seems not to be involved in physiological CH development, contrary to what characterizes pathological CH. We propose that AKT, through an inhibitory phosphorylation of the NHE-1, prevents its stretch-induced activation. This posttranslational modification emerges as an adaptive mechanism that avoids NHE-1 hyperactivity preserving its housekeeping functioning.


Assuntos
Coração/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Trocadores de Sódio-Hidrogênio/metabolismo , Animais , Tamanho Celular , Células Cultivadas , Fator de Crescimento Insulin-Like I/fisiologia , Masculino , Mecanotransdução Celular , Miócitos Cardíacos/metabolismo , Ratos Wistar , Natação
20.
Eur J Pharmacol ; 737: 125-32, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-24858370

RESUMO

Previous results show that prolonged treatment with EMD-87580 (EMD) NHE-1 blocker attenuates and reverses postinfarction remodelling. Our aim was to evaluate the effects of the treatment of EMD compared to ischemic postconditioning (IPO) in a model of regional ischemia. Isolated hearts were subjected to 40-min coronary occlusion followed by 60-min reperfusion (IC). Other hearts were treated with EMD 5µM during the first 10min of reperfusion or submitted to one cycle of 2min of reperfusion and 2min of ischemia as IPO protocol. Infarct sizes (IS), postischemic myocardial and vascular functions were assessed. The concentration of thiobarbituric reactive substances (TBARS), reduced glutathione (GSH) and expression of phosphorylated forms of ERK1/2, Akt, GSK-3ß, eNOS were analyzed. MnSOD cytosolic activity - as an index of mitochondrial permeability - was also measured. EMD treatment and IPO decreased IS~50% and significantly improved the postischemic recovery of contractility and coronary perfusion. TBARS decreased and GSH increased after interventions compared to the values observed in IC hearts. MnSOD cytosolic activity increased in IC group and was significantly attenuated by EMD and abolished in IPO hearts. The content of P-ERK1/2 increased whereas P-Akt, P-GSK-3ß and P-eNOS decreased in IC hearts. EMD treatment and IPO reversed these changes. The present data show that EMD treatment at the beginning of reperfusion-similarly to IPO- limited infarct size and attenuated the postischemic impairment of myocardial function through reactive oxygen species-mediated ERK1/2/Akt/GSK-3ß/eNOS pathways.


Assuntos
Cardiotônicos/farmacologia , Guanidinas/farmacologia , Pós-Condicionamento Isquêmico , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Traumatismo por Reperfusão Miocárdica/terapia , Sulfonas/farmacologia , Animais , Cardiotônicos/uso terapêutico , Guanidinas/uso terapêutico , Masculino , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Estresse Oxidativo/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Ratos , Ratos Wistar , Trocadores de Sódio-Hidrogênio/antagonistas & inibidores , Sulfonas/uso terapêutico , Disfunção Ventricular Esquerda/tratamento farmacológico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA