Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
J Neurosci ; 43(47): 8058-8072, 2023 11 22.
Article in English | MEDLINE | ID: mdl-37748861

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease affecting motor neurons. Recently, genome-wide association studies identified KIF5A as a new ALS-causing gene. KIF5A encodes a protein of the kinesin-1 family, allowing the anterograde transport of cargos along the microtubule rails in neurons. In ALS patients, mutations in the KIF5A gene induce exon 27 skipping, resulting in a mutated protein with a new C-terminal region (KIF5A Δ27). To understand how KIF5A Δ27 underpins the disease, we developed an ALS-associated KIF5A Drosophila model. When selectively expressed in motor neurons, KIF5A Δ27 alters larval locomotion as well as morphology and synaptic transmission at neuromuscular junctions in both males and females. We show that the distribution of mitochondria and synaptic vesicles is profoundly disturbed by KIF5A Δ27 expression. That is consistent with the numerous KIF5A Δ27-containing inclusions observed in motor neuron soma and axons. Moreover, KIF5A Δ27 expression leads to motor neuron death and reduces life expectancy. Our in vivo model reveals that a toxic gain of function underlies the pathogenicity of ALS-linked KIF5A mutant.SIGNIFICANCE STATEMENT Understanding how a mutation identified in patients with amyotrophic lateral sclerosis (ALS) causes the disease and the loss of motor neurons is crucial to fight against this disease. To this end, we have created a Drosophila model based on the motor neuron expression of the KIF5A mutant gene, recently identified in ALS patients. KIF5A encodes a kinesin that allows the anterograde transport of cargos. This model recapitulates the main features of ALS, including alterations of locomotion, synaptic neurotransmission, and morphology at neuromuscular junctions, as well as motor neuron death. KIF5A mutant is found in cytoplasmic inclusions, and its pathogenicity is because of a toxic gain of function.


Subject(s)
Amyotrophic Lateral Sclerosis , Neurodegenerative Diseases , Male , Animals , Female , Humans , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Kinesins/genetics , Kinesins/metabolism , Genome-Wide Association Study , Neurodegenerative Diseases/metabolism , Motor Neurons/metabolism , Neuromuscular Junction/metabolism , Mutation/genetics , Drosophila/metabolism , Inclusion Bodies/metabolism
2.
Int J Mol Sci ; 22(5)2021 Mar 04.
Article in English | MEDLINE | ID: mdl-33806419

ABSTRACT

Arterial smooth muscle exhibits rhythmic oscillatory contractions called vasomotion and believed to be a protective mechanism against tissue hypoperfusion or hypoxia. Oscillations of vascular tone depend on voltage and follow oscillations of the membrane potential. Voltage-gated sodium channels (Nav), responsible for the initiation and propagation of action potentials in excitable cells, have also been evidenced both in animal and human vascular smooth muscle cells (SMCs). For example, they contribute to arterial contraction in rats, but their physiopathological relevance has not been established in human vessels. In the present study, we investigated the functional role of Nav in the human artery. Experiments were performed on human uterine arteries obtained after hysterectomy and on SMCs dissociated from these arteries. In SMCs, we recorded a tetrodotoxin (TTX)-sensitive and fast inactivating voltage-dependent INa current. Various Nav genes, encoding α-subunit isoforms sensitive (Nav 1.2; 1.3; 1.7) and resistant (Nav 1.5) to TTX, were detected both in arterial tissue and in SMCs. Nav channels immunostaining showed uniform distribution in SMCs and endothelial cells. On arterial tissue, we recorded variations of isometric tension, ex vivo, in response to various agonists and antagonists. In arterial rings placed under hypoxic conditions, the depolarizing agent KCl and veratridine, a specific Nav channels agonist, both induced a sustained contraction overlaid with rhythmic oscillations of tension. After suppression of sympathetic control either by blocking the release of catecholamine or by antagonizing the target adrenergic response, rhythmic activity persisted while the sustained contraction was abolished. This rhythmic activity of the arteries was suppressed by TTX but, in contrast, only attenuated by antagonists of calcium channels, Na+/Ca2+ exchanger, Na+/K+-ATPase and the cardiac Nav channel. These results highlight the role of Nav as a novel key element in the vasomotion of human arteries. Hypoxia promotes activation of Nav channels involved in the initiation of rhythmic oscillatory contractile activity.


Subject(s)
Arteries/metabolism , Hypoxia/metabolism , Muscle Contraction/physiology , Voltage-Gated Sodium Channels/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Adult , Animals , Arteries/drug effects , Calcium Channels/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Humans , Middle Aged , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Sodium-Calcium Exchanger/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Tetrodotoxin/pharmacology
3.
Int J Mol Sci ; 22(9)2021 May 10.
Article in English | MEDLINE | ID: mdl-34068508

ABSTRACT

Duchenne muscular dystrophy (DMD) is a devastating condition shortening the lifespan of young men. DMD patients suffer from age-related dilated cardiomyopathy (DCM) that leads to heart failure. Several molecular mechanisms leading to cardiomyocyte death in DMD have been described. However, the pathological progression of DMD-associated DCM remains unclear. In skeletal muscle, a dramatic decrease in stem cells, so-called satellite cells, has been shown in DMD patients. Whether similar dysfunction occurs with cardiac muscle cardiovascular progenitor cells (CVPCs) in DMD remains to be explored. We hypothesized that the number of CVPCs decreases in the dystrophin-deficient heart with age and disease state, contributing to DCM progression. We used the dystrophin-deficient mouse model (mdx) to investigate age-dependent CVPC properties. Using quantitative PCR, flow cytometry, speckle tracking echocardiography, and immunofluorescence, we revealed that young mdx mice exhibit elevated CVPCs. We observed a rapid age-related CVPC depletion, coinciding with the progressive onset of cardiac dysfunction. Moreover, mdx CVPCs displayed increased DNA damage, suggesting impaired cardiac muscle homeostasis. Overall, our results identify the early recruitment of CVPCs in dystrophic hearts and their fast depletion with ageing. This latter depletion may participate in the fibrosis development and the acceleration onset of the cardiomyopathy.


Subject(s)
Cardiomyopathy, Dilated/genetics , Dystrophin/genetics , Muscular Dystrophy, Duchenne/genetics , Myocardium/metabolism , Proto-Oncogene Proteins c-kit/genetics , Aging/genetics , Aging/pathology , Animals , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Cardiovascular System/metabolism , Cardiovascular System/pathology , DNA Damage/genetics , Disease Models, Animal , Dystrophin/deficiency , Gene Expression Regulation/genetics , Humans , Mice , Mice, Inbred mdx/genetics , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Stem Cells/metabolism , Stem Cells/pathology
4.
J Muscle Res Cell Motil ; 38(1): 3-16, 2017 02.
Article in English | MEDLINE | ID: mdl-28224334

ABSTRACT

Cardiac hypertrophy (CH) is an adaptive process that exists in two distinct forms and allows the heart to adequately respond to an organism's needs. The first form of CH is physiological, adaptive and reversible. The second is pathological, irreversible and associated with fibrosis and cardiomyocyte death. CH involves multiple molecular mechanisms that are still not completely defined but it is now accepted that physiological CH is associated more with the PI3-K/Akt pathway while the main signaling cascade activated in pathological CH involves the Calcineurin-NFAT pathway. It was recently demonstrated that the TRPM4 channel may act as a negative regulator of pathological CH by regulating calcium entry and thus the Cn-NFAT pathway. In this study, we examined if the TRPM4 channel is involved in the physiological CH process. We evaluated the effects of 4 weeks endurance training on the hearts of Trpm4 +/+ and Trpm4 -/- mice. We identified an elevated functional expression of the TRPM4 channel in cardiomyocytes after endurance training suggesting a potential role for the channel in physiological CH. We then observed that Trpm4 +/+ mice displayed left ventricular hypertrophy after endurance training associated with enhanced cardiac function. By contrast, Trpm4 -/- mice did not develop these adaptions. While Trpm4 -/- mice did not develop gross cardiac hypertrophy, the cardiomyocyte surface area was larger and associated with an increase of Tunel positive cells. Endurance training in Trpm4 +/+ mice did not increase DNA fragmentation in the heart. Endurance training in Trpm4 +/+ mice was associated with activation of the classical physiological CH Akt pathway while Trpm4 -/- favored the Calcineurin pathway. Calcium studies demonstrated that TRPM4 channel negatively regulates calcium entry providing support for activation of the Cn-NFAT pathway in Trpm4 -/- mice. In conclusion, we provide evidence for the functional expression of TRPM4 channel in response to endurance training. This expression may help to maintain the balance between physiological and pathological hypertrophy.


Subject(s)
Atrial Remodeling/physiology , Physical Endurance/physiology , TRPM Cation Channels/genetics , Animals , Cardiomegaly , Male , Mice , TRPM Cation Channels/metabolism
5.
Sci Rep ; 14(1): 19540, 2024 08 22.
Article in English | MEDLINE | ID: mdl-39174694

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a devastating paralytic disorder caused by the death of motoneurons. Several mutations in the KIF5A gene have been identified in patients with ALS. Some mutations affect the splicing sites of exon 27 leading to its deletion (Δ27 mutation). KIF5A Δ27 is aggregation-prone and pathogenic for motoneurons due to a toxic gain of function. Another mutation found to be enriched in ALS patients is a proline/leucine substitution at position 986 (P986L mutation). Bioinformatic analyses strongly suggest that this variant is benign. Our study aims to conduct functional studies in Drosophila to classify the KIF5A P986L variant. When expressed in motoneurons, KIF5A P986L does not modify the morphology of larval NMJ or the synaptic transmission. In addition, KIF5A P986L is uniformly distributed in axons and does not disturb mitochondria distribution. Locomotion at larval and adult stages is not affected by KIF5A P986L. Finally, both KIF5A WT and P986L expression in adult motoneurons extend median lifespan compared to control flies. Altogether, our data show that the KIF5A P986L variant is not pathogenic for motoneurons and may represent a hypomorphic allele, although it is not causative for ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Kinesins , Motor Neurons , Animals , Kinesins/genetics , Kinesins/metabolism , Motor Neurons/metabolism , Motor Neurons/pathology , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/metabolism , Mutation , Humans , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila , Neuromuscular Junction/metabolism , Neuromuscular Junction/pathology , Drosophila melanogaster/genetics , Synaptic Transmission/genetics , Disease Models, Animal , Axons/metabolism , Axons/pathology , Larva/genetics , Larva/metabolism
6.
Cancers (Basel) ; 13(9)2021 Apr 27.
Article in English | MEDLINE | ID: mdl-33925547

ABSTRACT

Diffuse grade II IDH-mutant gliomas are slow-growing brain tumors that progress into high-grade gliomas. They present intratumoral cell heterogeneity, and no reliable markers are available to distinguish the different cell subtypes. The molecular mechanisms underlying the formation of this cell diversity is also ill-defined. Here, we report that SOX9 and OLIG1 transcription factors, which specifically label astrocytes and oligodendrocytes in the normal brain, revealed the presence of two largely nonoverlapping tumoral populations in IDH1-mutant oligodendrogliomas and astrocytomas. Astrocyte-like SOX9+ cells additionally stained for APOE, CRYAB, ID4, KCNN3, while oligodendrocyte-like OLIG1+ cells stained for ASCL1, EGFR, IDH1, PDGFRA, PTPRZ1, SOX4, and SOX8. GPR17, an oligodendrocytic marker, was expressed by both cells. These two subpopulations appear to have distinct BMP, NOTCH1, and MAPK active pathways as stainings for BMP4, HEY1, HEY2, p-SMAD1/5 and p-ERK were higher in SOX9+ cells. We used primary cultures and a new cell line to explore the influence of NOTCH1 activation and BMP treatment on the IDH1-mutant glioma cell phenotype. This revealed that NOTCH1 globally reduced oligodendrocytic markers and IDH1 expression while upregulating APOE, CRYAB, HEY1/2, and an electrophysiologically-active Ca2+-activated apamin-sensitive K+ channel (KCNN3/SK3). This was accompanied by a reduction in proliferation. Similar effects of NOTCH1 activation were observed in nontumoral human oligodendrocytic cells, which additionally induced strong SOX9 expression. BMP treatment reduced OLIG1/2 expression and strongly upregulated CRYAB and NOGGIN, a negative regulator of BMP. The presence of astrocyte-like SOX9+ and oligodendrocyte-like OLIG1+ cells in grade II IDH1-mutant gliomas raises new questions about their role in the pathology.

7.
Prog Biophys Mol Biol ; 159: 105-117, 2021 01.
Article in English | MEDLINE | ID: mdl-33031824

ABSTRACT

The transient receptor potential Melastatin 4 (TRPM4) channel is a calcium-activated non-selective cation channel expressed widely. In the heart, using a knock-out mouse model, the TRPM4 channel has been shown to be involved in multiple processes, including ß-adrenergic regulation, cardiac conduction, action potential duration and hypertrophic adaptations. This channel was recently shown to be involved in stress-induced cardiac arrhythmias in a mouse model overexpressing TRPM4 in ventricular cardiomyocytes. However, the link between TRPM4 channel expression in ventricular cardiomyocytes, the hypertrophic response to stress and/or cellular arrhythmias has yet to be elucidated. In this present study, we induced pathological hypertrophy in response to myocardial infarction using a mouse model of Trpm4 gene invalidation, and demonstrate that TRPM4 is essential for survival. We also demonstrate that the TRPM4 is required to activate both the Akt and Calcineurin pathways. Finally, using two hypertrophy models, either a physiological response to endurance training or a pathological response to myocardial infarction, we show that TRPM4 plays a role in regulating transient calcium amplitudes and leads to the development of cellular arrhythmias potentially in cooperation with the Sodium-calcium exchange (NCX). Here, we report two functions of the TRPM4 channel: first its role in adaptive hypertrophy, and second its association with NCX could mediate transient calcium amplitudes which trigger cellular arrhythmias.


Subject(s)
Heart Ventricles/metabolism , Hypertrophy/metabolism , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , TRPM Cation Channels/metabolism , Animals , Arrhythmias, Cardiac/metabolism , Biomechanical Phenomena/physiology , Calcineurin/metabolism , Calcium/metabolism , Echocardiography , Electrocardiography , Mice , Mice, Knockout , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Sodium/metabolism
8.
Cells ; 9(12)2020 11 27.
Article in English | MEDLINE | ID: mdl-33260927

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal neurological disorder characterized by the progressive degeneration of upper and lower motoneurons. Despite motoneuron death being recognized as the cardinal event of the disease, the loss of glial cells and interneurons in the brain and spinal cord accompanies and even precedes motoneuron elimination. In this review, we provide striking evidence that the degeneration of astrocytes and oligodendrocytes, in addition to inhibitory and modulatory interneurons, disrupt the functionally coherent environment of motoneurons. We discuss the extent to which the degeneration of glial cells and interneurons also contributes to the decline of the motor system. This pathogenic cellular network therefore represents a novel strategic field of therapeutic investigation.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Motor Neurons/pathology , Animals , Astrocytes/pathology , Humans , Interneurons/pathology , Neuroglia/pathology , Oligodendroglia/pathology
9.
Food Chem Toxicol ; 141: 111403, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32387306

ABSTRACT

The prevalence of metabolic syndrome (MetS), elevating cardiovascular risks, is increasing worldwide, with no available global therapeutic options. The intake of plain, mineral or biocompatible modified waters was shown to prevent some MetS features. This study was designed to analyze, in mice fed a high fat and sucrose diet (HFSD), the effects on MetS features of the daily intake of a reverse osmosed, weakly remineralized, water (OW) and of an OW dynamized by a physical processing (ODW), compared to tap water (TW). The HFSD was effective at inducing major features of MetS such as obesity, hepatic steatosis and inflammation, blood dyslipidemia, systemic glucose intolerance and muscle insulin resistance. Compared to TW, OW intake decreased hepatic fibrosis and inflammation, and mitigated hepatic steatosis and dyslipidemia. ODW intake further improved skeletal muscle insulin sensitivity and systemic glucose tolerance. This study highlights the deleterious metabolic impacts of the daily intake of TW, in combination with a high energy diet, and its possible involvement in MetS prevalence increase. In addition, it demonstrates that biocompatible modified water may be promising non-pharmaceutical, cost-effective tools for nutritional approaches in the treatment of MetS.


Subject(s)
Biocompatible Materials , Diet, High-Fat , Drinking Water , Metabolic Syndrome/prevention & control , Obesity/etiology , Animals , Basal Metabolism , Biomarkers/metabolism , Insulin Resistance , Lipogenesis , Liver Glycogen/metabolism , Male , Metabolic Syndrome/complications , Metabolic Syndrome/metabolism , Mice , Mice, Inbred C57BL , Obesity/complications
10.
Br J Pharmacol ; 177(19): 4448-4463, 2020 10.
Article in English | MEDLINE | ID: mdl-32608017

ABSTRACT

BACKGROUND AND PURPOSE: The artemisinin derivative, artemether, has antimalarial activity with potential neurotoxic and cardiotoxic effects. Artemether in nanocapsules (NC-ATM) is more efficient than free artemether for reducing parasitaemia and increasing survival of Plasmodium berghei-infected mice. NCs also prevent prolongation of the QT interval of the ECG. Here, we assessed cellular cardiotoxicity of artemether and how this toxicity was prevented by nanoencapsulation. EXPERIMENTAL APPROACH: Mice were treated with NC-ATM orally (120 mg·kg-1 twice daily) for 4 days. Other mice received free artemether, blank NCs, and vehicle for comparison. We measured single-cell contraction, intracellular Ca2+ transient using fluorescent Indo-1AM Ca2+ dye, and electrical activity using the patch-clamp technique in freshly isolated left ventricular myocytes. The acute effect of free artemether was also tested on cardiomyocytes of untreated animals. KEY RESULTS: Artemether prolonged action potentials (AP) upon acute exposure (at 0.1, 1, and 10 µM) of cardiomyocytes from untreated mice or after in vivo treatment. This prolongation was unrelated to blockade of K+ currents, increased Ca2+ currents or promotion of a sustained Na+ current. AP lengthening was abolished by the NCX inhibitor SEA-0400. Artemether promoted irregular Ca2+ transients during pacing and spontaneous Ca2+ events during resting periods. NC-ATM prevented all effects. Blank NCs had no effects compared with vehicle. CONCLUSION AND IMPLICATIONS: Artemether induced NCX-dependent AP lengthening (explaining QTc prolongation) and disrupted Ca2+ handling, both effects increasing pro-arrhythmogenic risks. NCs prevented these adverse effects, providing a safe alternative to the use of artemether alone, especially to treat malaria.


Subject(s)
Calcium , Myocytes, Cardiac , Action Potentials , Animals , Arrhythmias, Cardiac , Artemether , Calcium/metabolism , Mice , Myocytes, Cardiac/metabolism , Patch-Clamp Techniques , Sodium-Calcium Exchanger
11.
Article in English | MEDLINE | ID: mdl-32656189

ABSTRACT

Duchenne muscular dystrophy (DMD) is a severe genetic disorder characterized by the lack of functional dystrophin. DMD is associated with progressive dilated cardiomyopathy, eventually leading to heart failure as the main cause of death in DMD patients. Although several molecular mechanisms leading to the DMD cardiomyocyte (DMD-CM) death were described, mostly in mouse model, no suitable human CM model was until recently available together with proper clarification of the DMD-CM phenotype and delay in cardiac symptoms manifestation. We obtained several independent dystrophin-deficient human pluripotent stem cell (hPSC) lines from DMD patients and CRISPR/Cas9-generated DMD gene mutation. We differentiated DMD-hPSC into cardiac cells (CC) creating a human DMD-CC disease model. We observed that mutation-carrying cells were less prone to differentiate into CCs. DMD-CCs demonstrated an enhanced cell death rate in time. Furthermore, ion channel expression was altered in terms of potassium (Kir2.1 overexpression) and calcium handling (dihydropyridine receptor overexpression). DMD-CCs exhibited increased time of calcium transient rising compared to aged-matched control, suggesting mishandling of calcium release. We observed mechanical impairment (hypocontractility), bradycardia, increased heart rate variability, and blunted ß-adrenergic response connected with remodeling of ß-adrenergic receptors expression in DMD-CCs. Overall, these results indicated that our DMD-CC models are functionally affected by dystrophin-deficiency associated and recapitulate functional defects and cardiac wasting observed in the disease. It offers an accurate tool to study human cardiomyopathy progression and test therapies in vitro.

12.
Biochim Biophys Acta Mol Basis Dis ; 1865(1): 230-242, 2019 01.
Article in English | MEDLINE | ID: mdl-30463691

ABSTRACT

Heart failure with preserved ejection fraction (HFpEF) is a common clinical syndrome associated with high morbidity and mortality. Therapeutic options are limited due to a lack of knowledge of the pathology and its evolution. We investigated the cellular phenotype and Ca2+ handling in hearts recapitulating HFpEF criteria. HFpEF was induced in a portion of male Wistar rats four weeks after abdominal aortic banding. These animals had nearly normal ejection fraction and presented elevated blood pressure, lung congestion, concentric hypertrophy, increased LV mass, wall stiffness, impaired active relaxation and passive filling of the left ventricle, enlarged left atrium, and cardiomyocyte hypertrophy. Left ventricular cell contraction was stronger and the Ca2+ transient larger. Ca2+ cycling was modified with a RyR2 mediated Ca2+ leak from the sarcoplasmic reticulum and impaired Ca2+ extrusion through the Sodium/Calcium exchanger (NCX), which promoted an increase in diastolic Ca2+. The Sarcoplasmic/endoplasmic reticulum Ca2+ ATPase (SERCA2a) and NCX protein levels were unchanged. The phospholamban (PLN) to SERCA2a ratio was augmented in favor of an inhibitory effect on the SERCA2a activity. Conversely, PLN phosphorylation at the calmodulin-dependent kinase II (CaMKII)-specific site (PLN-Thr17), which promotes SERCA2A activity, was increased as well, suggesting an adaptive compensation of Ca2+ cycling. Altogether our findings show that cardiac remodeling in hearts with a HFpEF status differs from that known for heart failure with reduced ejection fraction. These data also underscore the interdependence between systolic and diastolic "adaptations" of Ca2+ cycling with complex compensative interactions between Ca2+ handling partner and regulatory proteins.


Subject(s)
Calcium/metabolism , Heart Failure/metabolism , Myocytes, Cardiac/metabolism , Stroke Volume , Animals , Calcium-Binding Proteins/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Disease Models, Animal , Heart Ventricles/metabolism , Homeodomain Proteins/metabolism , Hypertension/metabolism , Male , Rats , Rats, Wistar , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Sodium-Calcium Exchanger/metabolism , Ventricular Dysfunction, Left/metabolism
13.
J Mol Cell Cardiol ; 44(6): 1002-1015, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18482733

ABSTRACT

Diabetes is associated with increased risk of diastolic dysfunction, heart failure, QT prolongation and rhythm disturbances independent of age, hypertension or coronary artery disease. Although these observations suggest electrical remodeling in the heart with diabetes, the relationship between the metabolic and the functional derangements is poorly understood. Exploiting a mouse model (MHC-PPARalpha) with cardiac-specific overexpression of the peroxisome proliferator-activated receptor alpha (PPARalpha), a key driver of diabetes-related lipid metabolic dysregulation, the experiments here were aimed at examining directly the link(s) between alterations in cardiac fatty acid metabolism and the functioning of repolarizing, voltage-gated K(+) (Kv) channels. Electrophysiological experiments on left (LV) and right (RV) ventricular myocytes isolated from young (5-6 week) MHC-PPARalpha mice revealed marked K(+) current remodeling: I(to,f) densities are significantly (P<0.01) lower, whereas I(ss) densities are significantly (P<0.001) higher in MHC-PPARalpha, compared with age-matched wild type (WT), LV and RV myocytes. Consistent with the observed reductions in I(to,f) density, expression of the KCND2 (Kv4.2) transcript is significantly (P<0.001) lower in MHC-PPARalpha, compared with WT, ventricles. Western blot analyses revealed that expression of the Kv accessory protein, KChIP2, is also reduced in MHC-PPARalpha ventricles in parallel with the decrease in Kv4.2. Although the properties of the endogenous and the "augmented" I(ss) suggest a role(s) for two pore domain K(+) channel (K2P) pore-forming subunits, the expression levels of KCNK2 (TREK1), KCNK3 (TASK1) and KCNK5 (TASK2) in MHC-PPARalpha and WT ventricles are not significantly different. The molecular mechanisms underlying I(to,f) and I(ss) remodeling in MHC-PPARalpha ventricular myocytes, therefore, are distinct.


Subject(s)
Cardiomyopathies/metabolism , Diabetes Complications/metabolism , Membrane Potentials , Myocardium/metabolism , PPAR alpha/biosynthesis , Potassium Channels, Voltage-Gated/metabolism , Ventricular Remodeling , Animals , Cardiomyopathies/etiology , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Diabetes Complications/genetics , Diabetes Complications/pathology , Disease Models, Animal , Electrophysiologic Techniques, Cardiac , Fatty Acids/genetics , Fatty Acids/metabolism , Gene Expression , Heart Ventricles/metabolism , Heart Ventricles/pathology , Humans , Kv Channel-Interacting Proteins/metabolism , Membrane Potentials/genetics , Mice , Mice, Transgenic , Myocardium/pathology , Myocytes, Cardiac/metabolism , Organ Specificity/genetics , PPAR alpha/genetics , Potassium/metabolism , Potassium Channels, Voltage-Gated/genetics , Ventricular Remodeling/genetics
14.
Mol Biol Cell ; 16(5): 2414-23, 2005 May.
Article in English | MEDLINE | ID: mdl-15758029

ABSTRACT

In the adult, the heart rate is driven by spontaneous and repetitive depolarizations of pacemaker cells to generate a firing of action potentials propagating along the conduction system and spreading into the ventricles. In the early embryo before E9.5, the pacemaker ionic channel responsible for the spontaneous depolarization of cells is not yet functional. Thus the mechanisms that initiate early heart rhythm during cardiogenesis are puzzling. In the absence of a functional pacemaker ionic channel, the oscillatory nature of inositol 1,4,5-trisphosphate (InsP3)-induced intracellular Ca2+ signaling could provide an alternative pacemaking mechanism. To test this hypothesis, we have engineered pacemaker cells from embryonic stem (ES) cells, a model that faithfully recapitulates early stages of heart development. We show that InsP3-dependent shuttle of free Ca2+ in and out of the endoplasmic reticulum is essential for a proper generation of pacemaker activity during early cardiogenesis and fetal life.


Subject(s)
Calcium Signaling/physiology , Fetal Heart/embryology , Fetal Heart/metabolism , Heart Conduction System/embryology , Heart Conduction System/metabolism , Inositol 1,4,5-Trisphosphate/metabolism , Animals , Calcium Channels/genetics , Calcium Channels/metabolism , Calcium Signaling/drug effects , Calreticulin/genetics , Calreticulin/metabolism , Clone Cells , DNA, Complementary/genetics , Endoplasmic Reticulum/metabolism , Inositol 1,4,5-Trisphosphate/pharmacology , Inositol 1,4,5-Trisphosphate Receptors , Mice , Models, Cardiovascular , Myoblasts, Cardiac/drug effects , Myoblasts, Cardiac/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
15.
Circ Res ; 96(4): 451-8, 2005 Mar 04.
Article in English | MEDLINE | ID: mdl-15662035

ABSTRACT

Voltage-gated K+ (Kv) channel accessory (beta) subunits associate with pore-forming Kv alpha subunits and modify the properties and/or cell surface expression of Kv channels in heterologous expression systems. There is very little presently known, however, about the functional role(s) of Kv beta subunits in the generation of native cardiac Kv channels. Exploiting mice with a targeted disruption of the Kvbeta1 gene (Kvbeta1-/-), the studies here were undertaken to explore directly the role of Kvbeta1 in the generation of ventricular Kv currents. Action potential waveforms and peak Kv current densities are indistinguishable in myocytes isolated from the left ventricular apex (LVA) of Kvbeta1-/- and wild-type (WT) animals. Analysis of Kv current waveforms, however, revealed that mean+/-SEM I(to,f) density is significantly (P< or =0.01) lower in Kvbeta1-/- (21.0+/-0.9 pA/pF; n=68), than in WT (25.3+/-1.4 pA/pF; n=42), LVA myocytes, and that mean+/-SEM I(K,slow) density is significantly (P< or =0.01) higher in Kvbeta1-/- (19.1+/-0.9 pA/pF; n=68), compared with WT (15.9+/-0.7 pA/pF; n=42), LVA cells. Pharmacological studies demonstrated that the TEA-sensitive component of I(K,slow), I(K,slow2,) is selectively increased in Kvbeta1-/- LVA myocytes. In parallel with the alterations in I(to,f) and I(K,slow2) densities, Kv4.3 expression is decreased and Kv2.1 expression is increased in Kvbeta1-/- ventricles. Taken together, these results demonstrate that Kvbeta1 differentially regulates the functional cell surface expression of myocardial I(to,f) and I(K,slow2) channels.


Subject(s)
Myocardium/metabolism , Myocytes, Cardiac/metabolism , Potassium Channels, Calcium-Activated/physiology , Potassium Channels, Voltage-Gated/physiology , Potassium/metabolism , Action Potentials , Alternative Splicing , Amino Acid Sequence , Animals , Base Sequence , Calcium-Binding Proteins/metabolism , Cell Membrane/metabolism , Exons , Heart Ventricles/cytology , Heart Ventricles/metabolism , Introns , Ion Channel Gating/drug effects , Kv Channel-Interacting Proteins , Kv1.1 Potassium Channel , Kv1.2 Potassium Channel , Kv1.4 Potassium Channel , Kv1.5 Potassium Channel , Large-Conductance Calcium-Activated Potassium Channels , Membrane Proteins/metabolism , Mice , Mice, Knockout , Molecular Sequence Data , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Potassium Channels, Calcium-Activated/biosynthesis , Potassium Channels, Calcium-Activated/chemistry , Potassium Channels, Calcium-Activated/deficiency , Potassium Channels, Calcium-Activated/genetics , Potassium Channels, Voltage-Gated/biosynthesis , Potassium Channels, Voltage-Gated/chemistry , Potassium Channels, Voltage-Gated/drug effects , Potassium Channels, Voltage-Gated/genetics , Potassium Channels, Voltage-Gated/metabolism , Protein Interaction Mapping , Protein Subunits , Sequence Alignment , Shab Potassium Channels , Shal Potassium Channels
16.
Circ Res ; 97(12): 1342-50, 2005 Dec 09.
Article in English | MEDLINE | ID: mdl-16293790

ABSTRACT

Previous studies have demonstrated a role for voltage-gated K+ (Kv) channel alpha subunits of the Kv4 subfamily in the generation of rapidly inactivating/recovering cardiac transient outward K+ current, I(to,f), channels. Biochemical studies suggest that mouse ventricular I(to,f) channels reflect the heteromeric assembly of Kv4.2 and Kv4.3 with the accessory subunits, KChIP2 and Kvbeta1, and that Kv4.2 is the primary determinant of regional differences in (mouse ventricular) I(to,f) densities. Interestingly, the phenotypic consequences of manipulating I(to,f) expression in different mouse models are distinct. In the experiments here, the effects of the targeted deletion of Kv4.2 (Kv4.2(-/-)) were examined. Unexpectedly, voltage-clamp recordings from Kv4.2(-/-) ventricular myocytes revealed that I(to,f) is eliminated. In addition, the slow transient outward K+ current, I(to,s), and the Kv1.4 protein (which encodes I(to,s)) are upregulated in Kv4.2(-/-) ventricles. Although Kv4.3 mRNA/protein expression is not measurably affected, KChIP2 expression is markedly reduced in Kv4.2(-/-) ventricles. Similar to Kv4.3, expression of Kvbeta1, as well as Kv1.5 and Kv2.1, is similar in wild-type and Kv4.2(-/-) ventricles. In addition, and in marked contrast to previous findings in mice expressing a truncated Kv4.2 transgene, the elimination I(to,f) in Kv4.2(-/-) mice does not result in ventricular hypertrophy. Taken together, these findings demonstrate not only an essential role for Kv4.2 in the generation of mouse ventricular I(to,f) channels but also that the loss of I(to,f) per se does not have overt pathophysiological consequences.


Subject(s)
Cardiomegaly/etiology , Heart Ventricles/pathology , Myocytes, Cardiac/physiology , Shal Potassium Channels/physiology , Ventricular Remodeling , Animals , Electrocardiography , Kv Channel-Interacting Proteins/physiology , Mice , Mice, Knockout , Shal Potassium Channels/genetics
17.
Sci Rep ; 7: 44998, 2017 03 28.
Article in English | MEDLINE | ID: mdl-28349937

ABSTRACT

Chagas disease is a neglected parasitic disease caused by the protozoan Trypanosoma cruzi. New antitrypanosomal options are desirable to prevent complications, including a high rate of cardiomyopathy. Recently, a natural substance, lychnopholide, has shown therapeutic potential, especially when encapsulated in biodegradable polymeric nanocapsules. However, little is known regarding possible adverse effects of lychnopholide. Here we show that repeated-dose intravenous administration of free lychnopholide (2.0 mg/kg/day) for 20 days caused cardiopathy and mortality in healthy C57BL/6 mice. Echocardiography revealed concentric left ventricular hypertrophy with preserved ejection fraction, diastolic dysfunction and chamber dilatation at end-stage. Single cardiomyocytes presented altered contractility and Ca2+ handling, with spontaneous Ca2+ waves in diastole. Acute in vitro lychnopholide application on cardiomyocytes from healthy mice also induced Ca2+ handling alterations with abnormal RyR2-mediated diastolic Ca2+ release. Strikingly, the encapsulation of lychnopholide prevented the cardiac alterations induced in vivo by the free form repeated doses. Nanocapsules alone had no adverse cardiac effects. Altogether, our data establish lychnopholide presented in nanocapsule form more firmly as a promising new drug candidate to cure Chagas disease with minimal cardiotoxicity. Our study also highlights the potential of nanotechnology not only to improve the efficacy of a drug but also to protect against its adverse effects.


Subject(s)
Biocompatible Materials , Cardiotonic Agents/pharmacology , Lactones/adverse effects , Nanocapsules , Polymers , Sesquiterpenes/adverse effects , Trypanocidal Agents/adverse effects , Trypanosoma cruzi/drug effects , Animals , Biocompatible Materials/chemistry , Calcium/metabolism , Calcium Signaling/drug effects , Cardiotonic Agents/chemistry , Cardiotoxicity , Chagas Disease/diagnosis , Chagas Disease/drug therapy , Chagas Disease/mortality , Chagas Disease/parasitology , Echocardiography , Male , Mice , Molecular Imaging , Mortality , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Nanocapsules/chemistry , Polymers/chemistry
18.
Circ Res ; 90(5): 586-93, 2002 Mar 22.
Article in English | MEDLINE | ID: mdl-11909823

ABSTRACT

Previous studies have demonstrated a role for Kv4 alpha subunits in the generation of the fast transient outward K+ current, I(to,f), in the mammalian myocardium. The experiments here were undertaken to explore the role of homomeric/heteromeric assembly of Kv4.2 and Kv4.3 and of the Kv channel accessory subunit, KChIP2, in the generation of mouse ventricular I(to,f). Western blots reveal that the expression of Kv4.2 parallels the regional heterogeneity in I(to,f) density, whereas Kv4.3 and KChIP2 are uniformly expressed in adult mouse ventricles. Antisense oligodeoxynucleotides (AsODNs) targeted against Kv4.2 or Kv4.3 selectively attenuate I(to,f) in mouse ventricular cells. Adenoviral-mediated coexpression of Kv4.2 and Kv4.3 in HEK-293 cells and in mouse ventricular myocytes produces transient outward K+ currents with properties distinct from those produced on expression of Kv4.2 or Kv4.3 alone, and the gating properties of the heteromeric Kv4.2/Kv4.3 channels in ventricular cells are more similar to native I(to,f) than are the homomeric Kv4.2 or Kv4.3 channels. Biochemical studies reveal that Kv4.2, Kv4.3, and KChIP2 coimmunoprecipitate from adult mouse ventricles. In addition, most of the Kv4.2 and KChIP2 are associated with Kv4.3 in situ. Taken together, these results demonstrate that functional mouse ventricular I(to,f) channels are heteromeric, comprising Kv4.2/Kv4.3 alpha subunits and KChIP2. The results here also suggest that Kv4.2 is the primary determinant of the regional heterogeneity in I(to,f) expression in adult mouse ventricle.


Subject(s)
Heart Ventricles/metabolism , Myocardium/metabolism , Potassium Channels, Voltage-Gated , Potassium Channels/metabolism , Potassium/metabolism , Protein Subunits , Animals , Blotting, Western , Calcium-Binding Proteins/antagonists & inhibitors , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Culture Techniques , Heart Ventricles/cytology , Ion Channel Gating/drug effects , Kv Channel-Interacting Proteins , Membrane Potentials/drug effects , Mice , Myocardium/cytology , Oligonucleotides, Antisense/pharmacology , Patch-Clamp Techniques , Potassium Channel Blockers , Potassium Channels/genetics , Precipitin Tests , Protein Structure, Quaternary/physiology , Shal Potassium Channels
19.
Circ Res ; 92(2): 234-42, 2003 Feb 07.
Article in English | MEDLINE | ID: mdl-12574152

ABSTRACT

Although electrophysiological remodeling occurs in various myocardial diseases, the underlying molecular mechanisms are poorly understood. cDNA microarrays containing probes for a large population of mouse genes encoding ion channel subunits ("IonChips") were developed and exploited to investigate remodeling of ion channel transcripts associated with altered thyroid status in adult mouse ventricle. Functional consequences of hypo- and hyperthyroidism were evaluated with patch-clamp and ECG recordings. Hypothyroidism decreased heart rate and prolonged QTc duration. Opposite changes were observed in hyperthyroidism. Microarray analysis revealed that hypothyroidism induces significant reductions in KCNA5, KCNB1, KCND2, and KCNK2 transcripts, whereas KCNQ1 and KCNE1 expression is increased. In hyperthyroidism, in contrast, KCNA5 and KCNB1 expression is increased and KCNQ1 and KCNE1 expression is decreased. Real-time RT-PCR validated these results. Consistent with microarray analysis, Western blot experiments confirmed those modifications at the protein level. Patch-clamp recordings revealed significant reductions in I(to,f) and I(K,slow) densities, and increased I(Ks) density in hypothyroid myocytes. In addition to effects on K+ channel transcripts, transcripts for the pacemaker channel HCN2 were decreased and those encoding the alpha1C Ca2+ channel (CaCNA1C) were increased in hypothyroid animals. The expression of Na+, Cl-, and inwardly rectifying K+ channel subunits, in contrast, were unaffected by thyroid hormone status. Taken together, these data demonstrate that thyroid hormone levels selectively and differentially regulate transcript expression for at least nine ion channel alpha- and beta-subunits. Our results also document the potential of cDNA microarray analysis for the simultaneous examination of ion channel transcript expression levels in the diseased/remodeled myocardium.


Subject(s)
Heart Ventricles/physiopathology , Hyperthyroidism/physiopathology , Hypothyroidism/physiopathology , Ion Channels/biosynthesis , Ion Channels/genetics , Animals , Body Weight , Electrocardiography , Electrophysiologic Techniques, Cardiac , Gene Expression Profiling , Heart Rate/physiology , Heart Ventricles/pathology , Male , Mice , Mice, Inbred C57BL , Myocardium/chemistry , Myocardium/metabolism , Myocardium/pathology , Oligonucleotide Array Sequence Analysis , Organ Size , Patch-Clamp Techniques , Potassium Channels, Voltage-Gated/biosynthesis , Potassium Channels, Voltage-Gated/genetics , RNA, Messenger/analysis , RNA, Messenger/metabolism , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction
20.
Sci Rep ; 5: 17969, 2015 Dec 10.
Article in English | MEDLINE | ID: mdl-26655634

ABSTRACT

Ranolazine is a recently developed drug used for the treatment of patients with chronic stable angina. It is a selective inhibitor of the persistent cardiac Na(+) current (INa), and is known to reduce the Na(+)-dependent Ca(2+) overload that occurs in cardiomyocytes during ischemia. Vascular effects of ranolazine, such as vasorelaxation,have been reported and may involve multiple pathways. As voltage-gated Na(+) channels (Nav) present in arteries play a role in contraction, we hypothesized that ranolazine could target these channels. We studied the effects of ranolazine in vitro on cultured aortic smooth muscle cells (SMC) and ex vivo on rat aortas in conditions known to specifically activate or promote INa. We observed that in the presence of the Nav channel agonist veratridine, ranolazine inhibited INa and intracellular Ca(2+) calcium increase in SMC, and arterial vasoconstriction. In arterial SMC, ranolazine inhibited the activity of tetrodotoxin-sensitive voltage-gated Nav channels and thus antagonized contraction promoted by low KCl depolarization. Furthermore, the vasorelaxant effects of ranolazine, also observed in human arteries and independent of the endothelium, involved antagonization of the α1-adrenergic receptor. Combined α1-adrenergic antagonization and inhibition of SMCs Nav channels could be involved in the vascular effects of ranolazine.


Subject(s)
Adrenergic alpha-1 Receptor Antagonists/pharmacology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Ranolazine/pharmacology , Receptors, Adrenergic, alpha-1/metabolism , Sodium Channel Blockers/pharmacology , Voltage-Gated Sodium Channels/metabolism , Action Potentials/drug effects , Animals , Aorta , Humans , Muscle Contraction/drug effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Rats , Uterine Artery/drug effects , Uterine Artery/metabolism , Vasodilation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL