Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Am J Respir Cell Mol Biol ; 69(4): 391-403, 2023 10.
Article in English | MEDLINE | ID: mdl-37290041

ABSTRACT

Acute respiratory distress syndrome (ARDS) is a lung disease characterized by acute onset of noncardiogenic pulmonary edema, hypoxemia, and respiratory insufficiency. The current treatment for ARDS is mainly supportive in nature, providing a critical need for targeted pharmacological management. We addressed this medical problem by developing a pharmacological treatment for pulmonary vascular leakage, a culprit of alveolar damage and lung inflammation. Our novel therapeutic target is the microtubule accessory factor EB3 (end binding protein 3), which contributes to pulmonary vascular leakage by amplifying pathological calcium signaling in endothelial cells in response to inflammatory stimuli. EB3 interacts with IP3R3 (inositol 1,4,5-trisphosphate receptor 3) and orchestrates calcium release from endoplasmic reticulum stores. Here, we designed and tested the therapeutic benefits of a 14-aa peptide named CIPRI (cognate IP3 receptor inhibitor), which disrupted EB3-IP3R3 interaction in vitro and in lungs of mice challenged with endotoxin. Treatment with CIPRI or depletion of IP3R3 in lung microvascular endothelial monolayers mitigated calcium release from endoplasmic reticulum stores and prevented a disassembly of vascular endothelial cadherin junctions in response to the proinflammatory mediator α-thrombin. Furthermore, intravenous administration of CIPRI in mice mitigated inflammation-induced lung injury, blocked pulmonary microvascular leakage, prevented activation of NFAT (nuclear factor of activated T cells) signaling, and reduced production of proinflammatory cytokines in the lung tissue. CIPRI also improved survival of mice from endotoxemia and polymicrobial sepsis. Together, these data demonstrate that targeting EB3-IP3R3 interaction with a cognate peptide is a promising strategy to address hyperpermeability of microvessels in inflammatory lung diseases.


Subject(s)
Pulmonary Edema , Respiratory Distress Syndrome , Mice , Animals , Endothelial Cells/metabolism , Calcium/metabolism , Respiratory Distress Syndrome/metabolism , Lung/pathology , Pulmonary Edema/pathology , Carrier Proteins/metabolism , Microtubule-Associated Proteins/metabolism
2.
J Clin Invest ; 131(18)2021 09 15.
Article in English | MEDLINE | ID: mdl-34324437

ABSTRACT

Ischemic cardiomyopathy is associated with an increased risk of sudden death, activation of the unfolded protein response (UPR), and reductions in multiple cardiac ion channels. When activated, the protein kinase-like ER kinase (PERK) branch of the UPR reduces protein translation and abundance. We hypothesized that PERK inhibition could prevent ion channel downregulation and reduce arrhythmia risk after myocardial infarct (MI). MI induced in mice by coronary artery ligation resulted in reduced ion channel levels, ventricular tachycardia (VT), and prolonged corrected intervals between the Q and T waves on the ECGs (QTc). Protein levels of major cardiac ion channels were decreased. MI cardiomyocytes showed significantly prolonged action potential duration and decreased maximum upstroke velocity. Cardiac-specific PERK KO reduced electrical remodeling in response to MI, with shortened QTc intervals, fewer VT episodes, and higher survival rates. Pharmacological PERK inhibition had similar effects. In conclusion, we found that activated PERK during MI contributed to arrhythmia risk by the downregulation of select cardiac ion channels. PERK inhibition prevented these changes and reduced arrhythmia risk. These results suggest that ion channel downregulation during MI is a fundamental arrhythmia mechanism and that maintenance of ion channel levels is antiarrhythmic.


Subject(s)
Arrhythmias, Cardiac/prevention & control , Myocardial Infarction/complications , Myocardial Infarction/metabolism , Unfolded Protein Response/physiology , eIF-2 Kinase/antagonists & inhibitors , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/metabolism , Down-Regulation , Female , Heart Disease Risk Factors , Humans , Indoles/pharmacology , Ion Channels/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Cardiovascular , Protein Kinase Inhibitors/pharmacology , Unfolded Protein Response/drug effects , eIF-2 Kinase/deficiency , eIF-2 Kinase/genetics
3.
J Am Heart Assoc ; 7(8)2018 04 07.
Article in English | MEDLINE | ID: mdl-29627768

ABSTRACT

BACKGROUND: Heart failure (HF) is associated with increased arrhythmia risk and triggered activity. Abnormal Ca2+ handling is thought to underlie triggered activity, and mitochondria participate in Ca2+ homeostasis. METHODS AND RESULTS: A model of nonischemic HF was induced in C57BL/6 mice by hypertension. Computer simulations were performed using a mouse ventricular myocyte model of HF. Isoproterenol-induced premature ventricular contractions and ventricular fibrillation were more prevalent in nonischemic HF mice than sham controls. Isolated myopathic myocytes showed decreased cytoplasmic Ca2+ transients, increased mitochondrial Ca2+ transients, and increased action potential duration at 90% repolarization. The alteration of action potential duration at 90% repolarization was consistent with in vivo corrected QT prolongation and could be explained by augmented L-type Ca2+ currents, increased Na+-Ca2+ exchange currents, and decreased total K+ currents. Of myopathic ventricular myocytes, 66% showed early afterdepolarizations (EADs) compared with 17% of sham myocytes (P<0.05). Intracellular application of 1 µmol/L Ru360, a mitochondrial Ca2+ uniporter-specific antagonist, could reduce mitochondrial Ca2+ transients, decrease action potential duration at 90% repolarization, and ameliorate EADs. Furthermore, genetic knockdown of mitochondrial Ca2+ uniporters inhibited mitochondrial Ca2+ uptake, reduced Na+-Ca2+ exchange currents, decreased action potential duration at 90% repolarization, suppressed EADs, and reduced ventricular fibrillation in nonischemic HF mice. Computer simulations showed that EADs promoted by HF remodeling could be abolished by blocking either the mitochondrial Ca2+ uniporter or the L-type Ca2+ current, consistent with the experimental observations. CONCLUSIONS: Mitochondrial Ca2+ handling plays an important role in EADs seen with nonischemic cardiomyopathy and may represent a therapeutic target to reduce arrhythmic risk in this condition.


Subject(s)
Arrhythmias, Cardiac/etiology , Cardiomyopathies/metabolism , Heart Ventricles/metabolism , Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , Action Potentials/physiology , Animals , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/physiopathology , Cardiomyopathies/complications , Cardiomyopathies/pathology , Disease Models, Animal , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Male , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/pathology , Sodium-Calcium Exchanger/metabolism
4.
Int J Cardiol ; 249: 282-286, 2017 Dec 15.
Article in English | MEDLINE | ID: mdl-28916354

ABSTRACT

BACKGROUND: Our previous studies showed that in ischemic and nonischemic heart failure (HF), the voltage-gated cardiac Na+ channel α subunit (SCN5A) mRNA is abnormally spliced to produce two truncated transcript variants (E28C and D) that activate the unfolded protein response (UPR). We tested whether SCN5A post-transcriptional regulation was abnormal in hypertrophic cardiomyopathy (HCM). MATERIAL AND METHODS: Human heart tissue was obtained from HCM patients. The changes in relative abundances of SCN5A, its variants, splicing factors RBM25 and LUC7A, and PERK, a major effector of the UPR, were analyzed by real time RT-PCR and the expression changes were confirmed by Western Blot. RESULTS: We found reduced full-length transcript, increased SCN5A truncation variants and activation of UPR in HCM when compared to control hearts. In these patients, real time RT-PCR revealed that HCM patients had decreased SCN5A mRNA to 27.8±4.07% of control (P<0.01) and an increased abundance of E28C and E28D (3.4±0.3 and 2.8±0.3-fold, respectively, P<0.05). PERK mRNA increased 8.2±3.1 fold (P<0.01) in HCM patients. Western blot confirmed a significant increase of PERK. CONCLUSIONS: These data suggested that the full-length SCN5A was reduced in patients with HCM. This reduction was accompanied by abnormal SCN5A pre-mRNA splicing and UPR activation. These changes may contribute to the arrhythmic risk in HCM.


Subject(s)
Cardiomyopathy, Hypertrophic/metabolism , NAV1.5 Voltage-Gated Sodium Channel/metabolism , RNA Splicing/physiology , RNA, Messenger/metabolism , Adult , Aged , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/pathology , Female , Humans , Male , Middle Aged , NAV1.5 Voltage-Gated Sodium Channel/genetics , RNA, Messenger/genetics
5.
Heart Rhythm ; 14(3): 440-447, 2017 03.
Article in English | MEDLINE | ID: mdl-27989687

ABSTRACT

BACKGROUND: The reduced form of nicotinamide adenine dinucleotide (NADH) increases in cardiomyopathy, activates protein kinase C (PKC), up-regulates mitochondrial reactive oxygen species (mitoROS), and down-regulates the cardiac Na+ channel (NaV1.5). OBJECTIVE: The purpose of this study was to determine how NADH signals down-regulation of NaV1.5. METHODS: Isolated mouse cardiomyocytes were used for patch-clamp recording and for monitoring mitoROS with MitoSOX Red. HEK293 cells were used for transient transfections. HEK293 cells stably expressing human NaV1.5 were used for single channel recording, whole-cell patch-clamp recording, activity measurements of phospholipase C and phospholipase D (PLD), channel protein purification, and co-immunoprecipitation with PKC isoforms. HL-1 cells were used for mitochondria isolation. RESULTS: NADH enhanced PLD activity (1.6- ± 0.1-fold, P <.01) and activated PKCδ. Activated PKCδ translocated to mitochondria and up-regulated mitoROS (2.8- ± 0.3-fold, P <.01) by enhancing the activities of mitochondrial complexes I, II, and IV (1.1- to 1.5-fold, P <.01). PKCδ also interacted with NaV1.5 to down-regulate Na+ current (INa). Reduction in INa by activated PKCδ was prevented by antioxidants and by mutating the known PKC phosphorylation site S1503. At the single channel level, the mechanism of current reduction by PKC and recovery by protein kinase A was a change in single channel conductance. CONCLUSION: NADH activated PKCδ by enhancing PLD activity. PKCδ modulated both mitoROS and NaV1.5. PKCδ elevated mitoROS by enhancing mitochondrial oxidative phosphorylation complex activities. PKCδ-mediated channel phosphorylation and mitoROS were both required to down-regulate NaV1.5 and alter single channel conductance.


Subject(s)
Mitochondria, Heart/physiology , Myocytes, Cardiac/physiology , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Protein Kinase C/metabolism , Animals , Arrhythmias, Cardiac/metabolism , Cells, Cultured , Down-Regulation , Mice , NAD/metabolism , Phosphorylation , Reactive Oxygen Species/metabolism
7.
J Am Coll Cardiol ; 63(21): 2261-9, 2014 Jun 03.
Article in English | MEDLINE | ID: mdl-24703920

ABSTRACT

OBJECTIVES: The aim of this study was to determine the association of SCN5A cardiac sodium (Na(+)) channel mRNA splice variants in white blood cells (WBCs) with risk of arrhythmias in heart failure (HF). BACKGROUND: HF is associated with upregulation of two cardiac SCN5A mRNA splice variants that encode prematurely truncated, nonfunctional Na(+) channels. Because circulating WBCs demonstrate similar SCN5A splicing patterns, we hypothesized that these WBC-derived splice variants might further stratify patients with HF who are at risk for arrhythmias. METHODS: Simultaneously obtained myocardial core samples and WBCs were compared for SCN5A variants C (VC) and D (VD). Circulating variant levels were compared among patients with HF, divided into three groups: HF without an implantable cardioverter-defibrillator (ICD), HF with an ICD without appropriate intervention, and HF with an ICD with appropriate intervention. RESULTS: Myocardial tissue-derived SCN5A variant expression levels strongly correlated with circulating WBC samples for both VC and VD variants (r = 0.78 and 0.75, respectively). After controlling for covariates, patients with HF who had received an appropriate ICD intervention had higher expression levels of both WBC-derived SCN5A variants compared with patients with HF with ICDs who had not received appropriate ICD intervention (odds ratio, 3.25; 95% CI, 1.64-6.45; p = 0.001). Receiver operating characteristic analysis revealed that circulating SCN5A variant levels were highly associated with the risk for appropriate ICD intervention (area under the curve ≥0.97). CONCLUSIONS: Circulating expression levels of SCN5A variants were strongly associated with myocardial tissue levels. Furthermore, circulating variant levels were correlative with arrhythmic risk as measured by ICD events in an HF population within 1 year. (Sodium Channel Splicing in Heart Failure Trial [SOCS-HEFT]; NCT01185587).


Subject(s)
Defibrillators, Implantable , Heart Failure/blood , Heart Failure/therapy , NAV1.5 Voltage-Gated Sodium Channel/blood , Protein Isoforms/blood , RNA, Messenger/blood , Adult , Aged , Aged, 80 and over , Biomarkers/blood , Cross-Sectional Studies , Defibrillators, Implantable/adverse effects , Electric Countershock/adverse effects , Female , Heart Failure/genetics , Humans , Male , Middle Aged , Myocardium/metabolism , Myocardium/pathology , NAV1.5 Voltage-Gated Sodium Channel/genetics , Pilot Projects , Protein Isoforms/genetics , RNA, Messenger/genetics , Risk Factors
8.
Circ Arrhythm Electrophysiol ; 6(5): 1018-24, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24036084

ABSTRACT

BACKGROUND: Human heart failure (HF) increases alternative mRNA splicing of the type V, voltage-gated cardiac Na+ channel α-subunit (SCN5A), generating variants encoding truncated, nonfunctional channels that are trapped in the endoplasmic reticulum. In this work, we tested whether truncated Na+ channels activate the unfolded protein response (UPR), contributing to SCN5A electric remodeling in HF. METHODS AND RESULTS: UPR and SCN5A were analyzed in human ventricular systolic HF tissue samples and human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Cells were exposed to angiotensin II (AngII) and hypoxia, known activators of abnormal SCN5A mRNA splicing, or were induced to overexpress SCN5A variants. UPR effectors, protein kinase R-like ER kinase (PERK), calreticulin, and CHOP, were increased in human HF tissues. Induction of SCN5A variants with AngII or hypoxia or the expression of exogenous variants induced the UPR with concomitant downregulation of Na+ current. PERK activation destabilized SCN5A and, surprisingly, Kv4.3 channel mRNAs but not transient receptor potential cation channel M7 (TRPM7) channel mRNA. PERK inhibition prevented the loss of full-length SCN5A and Kv4.3 mRNA levels resulting from expressing Na+ channel mRNA splice variants. CONCLUSIONS: UPR can be initiated by Na+ channel mRNA splice variants and is involved in the reduction of cardiac Na+ current during human HF. Because the effect is not entirely specific to the SCN5A transcript, the UPR may play an important role in downregulation of multiple cardiac genes in HF.


Subject(s)
Heart Failure, Systolic/metabolism , Myocytes, Cardiac/metabolism , Sodium Channels/metabolism , Unfolded Protein Response/physiology , Angiotensin II/pharmacology , Blotting, Western , CCAAT-Enhancer-Binding Proteins/metabolism , Calreticulin/metabolism , Electrophysiologic Techniques, Cardiac , Endoplasmic Reticulum/metabolism , Heart Failure, Systolic/physiopathology , Humans , NAV1.5 Voltage-Gated Sodium Channel/metabolism , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Transfection , eIF-2 Kinase/metabolism
9.
Circ Arrhythm Electrophysiol ; 6(3): 623-31, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23559673

ABSTRACT

BACKGROUND: Previously, we showed that a mouse model (ACE8/8) of cardiac renin-angiotensin system activation has a high rate of spontaneous ventricular tachycardia and sudden cardiac death secondary to a reduction in connexin43 level. Angiotensin-II activation increases reactive oxygen species (ROS) production, and ACE8/8 mice show increased cardiac ROS. We sought to determine the source of ROS and whether ROS played a role in the arrhythmogenesis. METHODS AND RESULTS: Wild-type and ACE8/8 mice with and without 2 weeks of treatment with L-NIO (NO synthase inhibitor), sepiapterin (precursor of tetrahydrobiopterin), MitoTEMPO (mitochondria-targeted antioxidant), TEMPOL (a general antioxidant), apocynin (nicotinamide adenine dinucleotide phosphate oxidase inhibitor), allopurinol (xanthine oxidase inhibitor), and ACE8/8 crossed with P67 dominant negative mice to inhibit the nicotinamide adenine dinucleotide phosphate oxidase were studied. Western blotting, detection of mitochondrial ROS by MitoSOX Red, electron microscopy, immunohistochemistry, fluorescent dye diffusion technique for functional assessment of connexin43, telemetry monitoring, and in vivo electrophysiology studies were performed. Treatment with MitoTEMPO reduced sudden cardiac death in ACE8/8 mice (from 74% to 18%; P<0.005), decreased spontaneous ventricular premature beats, decreased ventricular tachycardia inducibility (from 90% to 17%; P<0.05), diminished elevated mitochondrial ROS to the control level, prevented structural damage to mitochondria, resulted in 2.6-fold increase in connexin43 level at the gap junctions, and corrected gap junction conduction. None of the other antioxidant therapies prevented ventricular tachycardia and sudden cardiac death in ACE8/8 mice. CONCLUSIONS: Mitochondrial oxidative stress plays a central role in angiotensin II-induced gap junction remodeling and arrhythmia. Mitochondria-targeted antioxidants may be effective antiarrhythmic drugs in cases of renin-angiotensin system activation.


Subject(s)
Antioxidants/pharmacology , Connexin 43/metabolism , Death, Sudden, Cardiac/etiology , Mitochondria/metabolism , Oxidative Stress/physiology , Tachycardia, Ventricular/drug therapy , Acetophenones/pharmacology , Animals , Connexin 43/drug effects , Cyclic N-Oxides/pharmacology , Disease Models, Animal , Mice , Mice, Inbred Strains , NADPH Oxidases/pharmacology , Random Allocation , Reactive Oxygen Species/metabolism , Renin-Angiotensin System/drug effects , Risk Factors , Sensitivity and Specificity , Spin Labels , Tachycardia, Ventricular/physiopathology
10.
Alcohol Alcohol ; 48(2): 131-7, 2013.
Article in English | MEDLINE | ID: mdl-22878590

ABSTRACT

AIMS: Excessive alcohol use in the form of binge drinking is associated with many adverse medical outcomes. Using an animal model, the primary objective of this study was to determine the effects of repeated episodes of binge drinking on myocardial structure, blood pressure (BP) and activation of mitogen-activated protein kinases (MAPKs). The effects of carvedilol, a beta-adrenergic blocker, were also examined in this animal model of binge drinking. METHODS: Rats were randomized into three groups: control, binge and binge + carvedilol (20 mg/kg). Animals received intragastric administration of 5 g ethanol/kg in the morning × 4 days (Monday-Thursday) followed by no ethanol on Friday-Sunday. Animals were maintained on the protocol for 5 weeks. BP was measured using radiotelemetry methods. Animals underwent echocardiography at baseline, 2.5 and 5 weeks. Myocardial MAPKs were analyzed at 5 weeks using western blot techniques. RESULTS: Over the course of 5 weeks, binge drinking was associated with significant transient increases in BP that were greater at 4 and 5 weeks compared with earlier time points. Carvedilol treatment significantly attenuated the binge-induced transient increases in BP at 4 and 5 weeks. No significant changes were found in echocardiographic parameters at any time period; however, binge drinking was associated with increased phosphorylation of p38 MAPK, which was blocked by carvedilol treatment. CONCLUSION: Repeated episodes of binge drinking result in progressive and transient increases in BP, no change in myocardial structure and differential regulation of MAPK activation.


Subject(s)
Binge Drinking/enzymology , Binge Drinking/physiopathology , Mitogen-Activated Protein Kinases/metabolism , Adrenergic Antagonists/pharmacology , Adrenergic Antagonists/therapeutic use , Animals , Binge Drinking/drug therapy , Blood Pressure/drug effects , Blood Pressure/physiology , Carbazoles/pharmacology , Carbazoles/therapeutic use , Carvedilol , Enzyme Activation/drug effects , Enzyme Activation/physiology , Gastric Mucosa/drug effects , Gastric Mucosa/enzymology , Gastric Mucosa/physiopathology , Male , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Pilot Projects , Propanolamines/pharmacology , Propanolamines/therapeutic use , Random Allocation , Rats , Rats, Sprague-Dawley , Secondary Prevention , Time Factors
11.
J Mol Cell Cardiol ; 54: 25-34, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23123323

ABSTRACT

Cardiomyopathy is associated with cardiac Na(+) channel downregulation that may contribute to arrhythmias. Previously, we have shown that elevated intracellular NADH causes a decrease in cardiac Na(+) current (I(Na)) signaled by an increase in mitochondrial reactive oxygen species (ROS). In this study, we tested whether the NADH-mitochondria ROS pathway was involved in the reduction of I(Na) in a nonischemic cardiomyopathic model and correlated the findings with myopathic human hearts. Nonischemic cardiomyopathy was induced in C57BL/6 mice by hypertension after unilateral nephrectomy, deoxycorticosterone acetate (DOCA) pellet implantation, and salt water substitution. Sham operated mice were used as controls. After six weeks, heart tissue and ventricular myocytes isolated from mice were utilized for whole cell patch clamp recording, NADH/NAD(+) level measurements, and mitochondrial ROS monitoring with confocal microscopy. Human explanted hearts were studied using optical mapping. Compared to the sham mice, the arterial blood pressure was higher, the left ventricular volume was significantly enlarged (104.7±3.9 vs. 87.9±6.1 µL, P<0.05), and the ejection fraction was reduced (37.1±1.8% vs. 49.4±3.7%, P<0.05) in DOCA mice. Both the whole cell and cytosolic NADH level were increased (279±70% and 123±2% of sham, respectively, P<0.01), I(Na) was decreased (60±10% of sham, P<0.01), and mitochondrial ROS overproduction was observed (2.9±0.3-fold of sham, P<0.01) in heart tissue and myocytes of myopathic mice vs. sham. Treatment of myocytes with NAD(+) (500 µM), mitoTEMPO (10 µM), chelerythrine (50 µM), or forskolin (5 µM) restored I(Na) back to the level of sham. Injection of NAD(+) (100mg/kg) or mitoTEMPO (0.7 mg/kg) twice (at 24h and 1h before myocyte isolation) to animals also restored I(Na). All treatments simultaneously reduced mitochondrial ROS levels to that of controls. CD38 was found to transduce the extracellular NAD(+) signal. Correlating with the mouse model, failing human hearts showed a reduction in conduction velocity that improved with NAD(+). Nonischemic cardiomyopathy was associated with elevated NADH level, PKC activation, mitochondrial ROS overproduction, and a concomitant decrease in I(Na). Reducing mitochondrial ROS by application of NAD(+), mitoTEMPO, PKC inhibitors, or PKA activators, restored I(Na). NAD(+) improved conduction velocity in human myopathic hearts.


Subject(s)
Cardiomyopathies/metabolism , Mitochondria, Heart/physiology , NAV1.5 Voltage-Gated Sodium Channel/metabolism , ADP-ribosyl Cyclase 1/metabolism , Action Potentials/drug effects , Animals , Benzophenanthridines/pharmacology , Colforsin/pharmacology , Down-Regulation , Heart Conduction System/drug effects , Heart Conduction System/physiopathology , Humans , In Vitro Techniques , Membrane Glycoproteins/metabolism , Membrane Potentials/drug effects , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , NAD/metabolism , NAD/pharmacology , NAV1.5 Voltage-Gated Sodium Channel/genetics , Organophosphorus Compounds/pharmacology , Oxidative Stress , Patch-Clamp Techniques , Piperidines/pharmacology , Reactive Oxygen Species/metabolism
12.
J Mol Cell Cardiol ; 56: 44-54, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23247392

ABSTRACT

Despite the increasing prevalence of heart failure with preserved left ventricular function, there are no specific treatments, partially because the mechanism of impaired relaxation is incompletely understood. Evidence indicates that cardiac relaxation may depend on nitric oxide (NO), generated by NO synthase (NOS) requiring the co-factor tetrahydrobiopterin (BH(4)). Recently, we reported that hypertension-induced diastolic dysfunction was accompanied by cardiac BH(4) depletion, NOS uncoupling, a depression in myofilament cross-bridge kinetics, and S-glutathionylation of myosin binding protein C (MyBP-C). We hypothesized that the mechanism by which BH(4) ameliorates diastolic dysfunction is by preventing glutathionylation of MyBP-C and thus reversing changes of myofilament properties that occur during diastolic dysfunction. We used the deoxycorticosterone acetate (DOCA)-salt mouse model, which demonstrates mild hypertension, myocardial oxidative stress, and diastolic dysfunction. Mice were divided into two groups that received control diet and two groups that received BH(4) supplement for 7days after developing diastolic dysfunction at post-operative day 11. Mice were assessed by echocardiography. Left ventricular papillary detergent-extracted fiber bundles were isolated for simultaneous determination of force and ATPase activity. Sarcomeric protein glutathionylation was assessed by immunoblotting. DOCA-salt mice exhibited diastolic dysfunction that was reversed after BH(4) treatment. Diastolic sarcomere length (DOCA-salt 1.70±0.01 vs. DOCA-salt+BH(4) 1.77±0.01µm, P<0.001) and relengthening (relaxation constant, τ, DOCA-salt 0.28±0.02 vs. DOCA-salt+BH(4) 0.08±0.01, P<0.001) were also restored to control by BH(4) treatment. pCa(50) for tension increased in DOCA-salt compared to sham but reverted to sham levels after BH(4) treatment. Maximum ATPase rate and tension cost (ΔATPase/ΔTension) decreased in DOCA-salt compared to sham, but increased after BH(4) treatment. Cardiac MyBP-C glutathionylation increased in DOCA-salt compared to sham, but decreased with BH(4) treatment. MyBP-C glutathionylation correlated with the presence of diastolic dysfunction. Our results suggest that by depressing S-glutathionylation of MyBP-C, BH(4) ameliorates diastolic dysfunction by reversing a decrease in cross-bridge turnover kinetics. These data provide evidence for modulation of cardiac relaxation by post-translational modification of myofilament proteins.


Subject(s)
Biopterins/analogs & derivatives , Cardiovascular Agents/administration & dosage , Heart Failure, Diastolic/drug therapy , Myofibrils/physiology , Adenosine Triphosphatases/metabolism , Administration, Oral , Animals , Biopterins/administration & dosage , Carrier Proteins/metabolism , Cells, Cultured , Desoxycorticosterone/pharmacology , Diastole/drug effects , Dietary Supplements , Glutathione/metabolism , Heart Failure, Diastolic/diagnostic imaging , Heart Failure, Diastolic/physiopathology , Mice , Myofibrils/drug effects , Myofibrils/enzymology , Oxidative Stress , Protein Processing, Post-Translational , Stroke Volume/drug effects , Ultrasonography
13.
Circ Res ; 110(6): 841-50, 2012 Mar 16.
Article in English | MEDLINE | ID: mdl-22343711

ABSTRACT

RATIONALE: Previously, we demonstrated that a deoxycorticosterone acetate (DOCA)-salt hypertensive mouse model produces cardiac oxidative stress and diastolic dysfunction with preserved systolic function. Oxidative stress has been shown to increase late inward sodium current (I(Na)), reducing the net cytosolic Ca(2+) efflux. OBJECTIVE: Oxidative stress in the DOCA-salt model may increase late I(Na), resulting in diastolic dysfunction amenable to treatment with ranolazine. METHODS AND RESULTS: Echocardiography detected evidence of diastolic dysfunction in hypertensive mice that improved after treatment with ranolazine (E/E':sham, 31.9 ± 2.8, sham+ranolazine, 30.2 ± 1.9, DOCA-salt, 41.8 ± 2.6, and DOCA-salt+ranolazine, 31.9 ± 2.6; P=0.018). The end-diastolic pressure-volume relationship slope was elevated in DOCA-salt mice, improving to sham levels with treatment (sham, 0.16 ± 0.01 versus sham+ranolazine, 0.18 ± 0.01 versus DOCA-salt, 0.23 ± 0.2 versus DOCA-salt+ranolazine, 0.17 ± 0.0 1 mm Hg/L; P<0.005). DOCA-salt myocytes demonstrated impaired relaxation, τ, improving with ranolazine (DOCA-salt, 0.18 ± 0.02, DOCA-salt+ranolazine, 0.13 ± 0.01, sham, 0.11 ± 0.01, sham+ranolazine, 0.09 ± 0.02 seconds; P=0.0004). Neither late I(Na) nor the Ca(2+) transients were different from sham myocytes. Detergent extracted fiber bundles from DOCA-salt hearts demonstrated increased myofilament response to Ca(2+) with glutathionylation of myosin binding protein C. Treatment with ranolazine ameliorated the Ca(2+) response and cross-bridge kinetics. CONCLUSIONS: Diastolic dysfunction could be reversed by ranolazine, probably resulting from a direct effect on myofilaments, indicating that cardiac oxidative stress may mediate diastolic dysfunction through altering the contractile apparatus.


Subject(s)
Acetanilides/pharmacology , Calcium/metabolism , Diastole/drug effects , Heart Failure, Diastolic/drug therapy , Myocytes, Cardiac/drug effects , Myofibrils/drug effects , Piperazines/pharmacology , Acetanilides/blood , Animals , Desoxycorticosterone/toxicity , Diastole/physiology , Disease Models, Animal , Enzyme Inhibitors/blood , Enzyme Inhibitors/pharmacology , Heart Failure, Diastolic/chemically induced , Heart Failure, Diastolic/physiopathology , Mice , Mineralocorticoids/toxicity , Myocardial Contraction/drug effects , Myocardial Contraction/physiology , Myocytes, Cardiac/physiology , Myofibrils/metabolism , Oxidative Stress/physiology , Piperazines/blood , Ranolazine , Sodium/metabolism , Ventricular Dysfunction, Left/chemically induced , Ventricular Dysfunction, Left/drug therapy , Ventricular Dysfunction, Left/physiopathology
14.
Eur J Heart Fail ; 10(11): 1057-64, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18815071

ABSTRACT

AIM: To determine the effects of cigarette smoke (CS) exposure on the expression/activation of mitogen-activated protein kinases (MAPKs) (extracellular signal-regulated kinase [ERK1/2], p38-kinase [p38] and c-Jun NH2-terminal protein kinase [JNK]), norepinephrine (NE) levels and myocardial structure and function. METHODS: Rats were randomised to two groups: CS-exposed (n=12) or room air (CON) (n=10). After 5 weeks, the animals underwent echocardiography with pulse-wave Doppler flow measurements. Hearts were removed for microscopy and Western blot analysis. RESULTS: CS exposure was associated with significant increases in NE urinary levels and larger ventricular dimensions (mm) (CON=left ventricular end diastolic dimension [LVEDD] 7.99+/-0.10, LV end systolic dimension [LVESD] 4.55+/-0.20, CS=LVEDD 8.3+/-0.10, LVESD 5.3+/-0.09, p=0.026, p=0.003). There was also evidence of systolic dysfunction in the CS-exposed group compared to the CON group (fractional shortening %, CON=43+/-2, CS=36+/-.09, p=0.010). In CS-exposed hearts, significant increases in phosphorylated p38/total p38 (0.975+/-0.05) and phosphorylated ERK1/2/totalERK1/2 (1.919+/-0.050) were found compared to CON hearts (0.464+/-0.008, 0.459+/-0.050, respectively). No significant differences were found in JNK levels between the groups. CONCLUSIONS: Increased NE levels and MAPK activation are associated with CS-related left ventricular remodelling.


Subject(s)
Heart Ventricles/enzymology , Mitogen-Activated Protein Kinases/metabolism , Smoking/adverse effects , Tobacco Smoke Pollution/adverse effects , Ventricular Remodeling/drug effects , Animals , Blotting, Western , Disease Models, Animal , Echocardiography , Enzyme Activation/drug effects , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Male , Microscopy, Electron , Mitogen-Activated Protein Kinases/drug effects , Myocardium/enzymology , Myocardium/ultrastructure , Radioimmunoassay , Rats , Rats, Sprague-Dawley
15.
Am J Hypertens ; 20(1): 53-61, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17198912

ABSTRACT

BACKGROUND: Early treatment with captopril prevents the development of hypertension by inhibiting the generation of angiotensin II and smooth muscle contraction. Although smooth muscle contraction is regulated by myosin light chain phosphorylation (MLC-P), the role of MLC-P in captopril effects in hypertension has not been described. Therefore, we treated spontaneously hypertensive rats (SHR) with captopril and investigated the effects of this agent on downstream signaling. METHODS: Male SHR (n = 12) were treated with captopril (3.7 mmol/L in drinking water) beginning in utero and continuing up to 12 weeks of age. Age- and sex-matched untreated SHR and Wistar-Kyoto (WKY) rats were used as controls. Rats were split into three subgroups and were sacrificed at 12, 18, or 24 weeks of age. Systolic blood pressure, left ventricular weight, and body weight were measured. Mesenteric arteries were removed for histologic and biochemical studies. RESULTS: At 12 weeks, captopril significantly decreased systolic blood pressure (from 198 +/- 10 to 125+/-16 mm Hg), reduced left ventricular weight-to-body weight ratios (from 2.94 +/- 0.06 to 2.17 +/- 0.08 mg/g), and prevented vascular remodeling in mesenteric arteries in SHR. Ras expression, extracellular receptor kinase phosphorylation (ERK-P), myosin light chain kinase (MLCK) expression, and MLC-P were all significantly increased in mesenteric arteries in untreated SHR compared with WKY rats. Early captopril treatment in SHR significantly inhibited Ras and MLCK expression at all ages and decreased ERK-P and MLC-P at 12 and 18 weeks in mesenteric arteries. CONCLUSIONS: These data demonstrate that the antihypertensive effects of captopril are correlated with inhibition of Ras-regulated ERK activation, MLCK expression, and MLC-P.


Subject(s)
Angiotensin II/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Hypertension/metabolism , Myosin Light Chains/metabolism , ras Proteins/metabolism , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Animals , Captopril/therapeutic use , Female , Hypertension/drug therapy , Hypertrophy, Left Ventricular/prevention & control , Male , Mesenteric Arteries/metabolism , Phosphorylation , Rats , Rats, Inbred SHR
16.
Mol Biol Cell ; 17(9): 4039-50, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16822834

ABSTRACT

Regulation of gene transcription in vascular smooth muscle cells (VSMCs) by serum response factor (SRF) plays a crucial role in vascular development and in the pathophysiology of vascular diseases. Nevertheless, the regulation of specific genes by SRF in vascular diseases is poorly understood. Therefore, we investigated the regulation of smooth muscle myosin light chain kinase (smMLCK) by using spontaneously hypertensive rats (SHR) as an experimental model. We found that smMLCK expression in blood vessels increases during the development of hypertension and is always greater in blood vessels from SHR compared with normotensive rats. Analysis of the DNA sequences of the promoters isolated from SHR and normotensive rats revealed that SHR contain a 12-base pair insertion adjacent to the CArG box. This insertion increases SRF binding to the CArG box and positively regulates SRF-dependent promoter activity. The increase in smMLCK expression was blocked by dominant-negative SRF, dominant-negative Ras, or antisense oligonucleotides to ERK. In vivo, inhibiting MEK decreased smMLCK expression and blood pressure in SHR partly by decreasing SRF binding to the smMLCK promoter. These data provide novel insight into the regulation of smMLCK expression at the molecular level and demonstrate the importance of SRF in regulating smMLCK promoter activity in SHR.


Subject(s)
Gene Expression Regulation, Enzymologic , Hypertension/enzymology , Mutagenesis, Insertional , Myosin-Light-Chain Kinase/genetics , Myosin-Light-Chain Kinase/metabolism , Promoter Regions, Genetic/genetics , Serum Response Factor/metabolism , Animals , Base Sequence , Blood Pressure/physiology , COS Cells , Cells, Cultured , Chlorocebus aethiops , Hypertension/physiopathology , Introns/genetics , Mice , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Molecular Sequence Data , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/enzymology , Myosin Light Chains/metabolism , Myosin-Light-Chain Kinase/antagonists & inhibitors , Phosphorylation , Protein Binding , Proto-Oncogene Proteins p21(ras)/metabolism , Rats , Rats, Inbred SHR , Signal Transduction
17.
Eur J Cancer ; 42(7): 948-57, 2006 May.
Article in English | MEDLINE | ID: mdl-16574402

ABSTRACT

We have previously shown that ML-7, which inhibits myosin light chain kinase (MLCK), induces apoptosis in transformed and non-transformed cells. We have extended these studies and found that ML-7 stimulates the ability of etoposide to induce apoptosis in Mm5MT mouse mammary adenocarcinoma cells and Mat-Ly-Lu rat prostate cancer cells in vitro. ML-7 was also found to have a chemopreventive effect using an in vitro mouse mammary organ culture model. In vivo experiments demonstrated that ML-7 retards the growth of mammary tumours in mice and prostate tumours in rats. Moreover, ML-7 significantly stimulates the ability of etoposide to prevent the growth of established mammary tumours in mice and prostate tumours in rats. These results provide evidence for the efficacy of ML-7 as an adjuvant to etoposide in these models and warrants further development.


Subject(s)
Adenocarcinoma/pathology , Azepines/pharmacology , Mammary Neoplasms, Experimental/pathology , Myosin-Light-Chain Kinase/antagonists & inhibitors , Naphthalenes/pharmacology , Prostatic Neoplasms/pathology , Adenocarcinoma/drug therapy , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis/drug effects , Cell Division , Chemotherapy, Adjuvant , Etoposide/therapeutic use , Humans , Male , Mammary Neoplasms, Experimental/drug therapy , Mice , Mice, Inbred BALB C , Prostatic Neoplasms/drug therapy , Random Allocation , Rats
18.
Mol Cell Biol ; 25(14): 6259-66, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15988034

ABSTRACT

Previous short-term studies have correlated an increase in the phosphorylation of the 20-kDa light chain of myosin II (MLC20) with blebbing in apoptotic cells. We have found that this increase in MLC20 phosphorylation is rapidly followed by MLC20 dephosphorylation when cells are stimulated with various apoptotic agents. MLC20 dephosphorylation is not a consequence of apoptosis because MLC20 dephosphorylation precedes caspase activation when cells are stimulated with a proapoptotic agent or when myosin light chain kinase (MLCK) is inhibited pharmacologically or by microinjecting an inhibitory antibody to MLCK. Moreover, blocking caspase activation increased cell survival when MLCK is inhibited or when cells are treated with tumor necrosis factor alpha. Depolymerizing actin filaments or detaching cells, processes that destabilize the cytoskeleton, or inhibiting myosin ATPase activity also resulted in MLC20 dephosphorylation and cell death. In vivo experiments showed that inhibiting MLCK increased the number of apoptotic cells and retarded the growth of mammary cancer cells in mice. Thus, MLC20 dephosphorylation occurs during physiological cell death and prolonged MLC20 dephosphorylation can trigger apoptosis.


Subject(s)
Apoptosis , Myosin Light Chains/metabolism , Myosin Type II/metabolism , Myosin-Light-Chain Kinase/antagonists & inhibitors , Animals , Antibodies/immunology , Antibodies/pharmacology , Caspase 3 , Caspase Inhibitors , Caspases/metabolism , Cells, Cultured , Cytoskeleton/metabolism , Humans , Mice , Myosin-Light-Chain Kinase/immunology , Phosphorylation
SELECTION OF CITATIONS
SEARCH DETAIL
...