Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
1.
Arch Cardiovasc Dis ; 117(4): 283-296, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38490844

ABSTRACT

Atrial cardiomyopathy is defined as any complex of structural, architectural, contractile or electrophysiological changes affecting atria, with the potential to produce clinically relevant manifestations. Most of our knowledge about the mechanistic aspects of atrial cardiomyopathy is derived from studies investigating animal models of atrial fibrillation and atrial tissue samples obtained from individuals who have a history of atrial fibrillation. Several noninvasive tools have been reported to characterize atrial cardiomyopathy in patients, which may be relevant for predicting the risk of incident atrial fibrillation and its related outcomes, such as stroke. Here, we provide an overview of the pathophysiological mechanisms involved in atrial cardiomyopathy, and discuss the complex interplay of these mechanisms, including aging, left atrial pressure overload, metabolic disorders and genetic factors. We discuss clinical tools currently available to characterize atrial cardiomyopathy, including electrocardiograms, cardiac imaging and serum biomarkers. Finally, we discuss the clinical impact of atrial cardiomyopathy, and its potential role for predicting atrial fibrillation, stroke, heart failure and dementia. Overall, this review aims to highlight the critical need for a clinically relevant definition of atrial cardiomyopathy to improve treatment strategies.


Subject(s)
Atrial Appendage , Atrial Fibrillation , Cardiomyopathies , Stroke , Animals , Humans , Atrial Fibrillation/diagnosis , Heart Atria , Cardiomyopathies/diagnosis , Cardiomyopathies/therapy
4.
Cells ; 12(4)2023 02 11.
Article in English | MEDLINE | ID: mdl-36831249

ABSTRACT

BACKGROUND: The use of mesenchymal stem cells (MSCs) appears to be a promising therapeutic approach for cardiac repair after myocardial infarction. However, clinical trials have revealed the need to improve their therapeutic efficacy. Recent evidence demonstrated that mitochondria undergo spontaneous transfer from damaged cells to MSCs, resulting in the activation of the cytoprotective and pro-angiogenic functions of recipient MSCs. Based on these observations, we investigated whether the preconditioning of MSCs with mitochondria could optimize their therapeutic potential for ischemic heart disease. METHODS: Human MSCs were exposed to mitochondria isolated from human fetal cardiomyocytes. After 24 h, the effects of mitochondria preconditioning on the MSCs' function were analyzed both in vitro and in vivo. RESULTS: We found that cardiac mitochondria-preconditioning improved the proliferation and repair properties of MSCs in vitro. Mechanistically, cardiac mitochondria mediate their stimulatory effects through the production of reactive oxygen species, which trigger their own degradation in recipient MSCs. These effects were further confirmed in vivo, as the mitochondria preconditioning of MSCs potentiated their therapeutic efficacy on cardiac function following their engraftment into infarcted mouse hearts. CONCLUSIONS: The preconditioning of MSCs with the artificial transfer of cardiac mitochondria appears to be promising strategy to improve the efficacy of MSC-based cell therapy in ischemic heart disease.


Subject(s)
Mesenchymal Stem Cells , Myocardial Infarction , Myocardial Ischemia , Mice , Animals , Humans , Myocardial Ischemia/metabolism , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , Mitochondria, Heart/metabolism , Mesenchymal Stem Cells/metabolism
5.
J Am Coll Cardiol ; 80(23): 2205-2219, 2022 12 06.
Article in English | MEDLINE | ID: mdl-36456051

ABSTRACT

BACKGROUND: Patients with metabolic syndrome (MetS) have an increased risk of atrial fibrillation (AF). Impaired Ca2+ homeostasis and mitochondrial dysfunction have emerged as an arrhythmogenic substrate in both patients and animal models of MetS. Whether impaired mitochondrial Ca2+ handling underlies AF associated with MetS remains poorly explored. OBJECTIVES: The aim of this study was to determine the initial mechanisms related to AF susceptibility and mitochondrial dysfunction encountered in metabolic cardiomyopathy. METHODS: A total of 161 mice and 34 patients were studied. Mitochondrial Ca2+ and mitochondrial Ca2+ uniporter complex (MCUC) were investigated in right atrial tissue of patients with (n = 18) or without (n = 16) MetS and of C57Bl/6J mice fed with a high-fat sucrose diet (HFS) for 2 (n = 42) or 12 (n = 39) weeks. Susceptibility to AF was evaluated in isolated sinoatrial tissue and in vivo in mice. RESULTS: Increased expression of the MICUs subunits of the MCUC (1.00 ± 0.33 AU vs 1.29 ± 0.23 AU; P = 0.034) was associated with impaired mitochondrial Ca2+ uptake in patients (168.7 ± 31.3 nmol/min/mg vs 127.3 ± 18.4 nmol/min/mg; P = 0.026) and HFS mice (0.10 ± 0.04 ΔF/F0 × ms-1 vs 0.06 ± 0.03 ΔF/F0 × ms-1; P = 0.0086, and 0.15 ± 0.07 ΔF/F0 × ms-1 vs 0.046 ± 0.03 ΔF/F0 × ms-1; P = 0.0076 in 2- and 12-week HFS mice, respectively). HFS mice elicited a 70% increased susceptibility to AF. The MCUC agonist kaempferol restored MCUC activity in vitro and abolished the occurrence of AF in HFS mice. CONCLUSIONS: Impaired MCUC activity and mitochondrial Ca2+ homeostasis from the early stage of metabolic cardiomyopathy in mice lead to AF. Given that similar defects in cardiac mitochondrial Ca2+ handling are present in MetS patients, the modulation of the MCUC activity represents an attractive antiarrhythmic strategy.


Subject(s)
Atrial Appendage , Atrial Fibrillation , Metabolic Syndrome , Mice , Animals , Atrial Fibrillation/etiology , Calcium , Metabolic Syndrome/complications , Anti-Arrhythmia Agents , Mice, Inbred C57BL
6.
Redox Biol ; 56: 102431, 2022 10.
Article in English | MEDLINE | ID: mdl-35988446

ABSTRACT

YAP1 and TAZ are transcriptional co-activator proteins that play fundamental roles in many biological processes, from cell proliferation and cell lineage fate determination to tumorigenesis. We previously demonstrated that Limb Expression 1 (LIX1) regulates YAP1 and TAZ activity and controls digestive mesenchymal progenitor proliferation. However, LIX1 mode of action remains elusive. Here, we found that endogenous LIX1 is localized in mitochondria and is anchored to the outer mitochondrial membrane through S-palmitoylation of cysteine 84, a residue conserved in all LIX1 orthologs. LIX1 downregulation altered the mitochondrial ultrastructure, resulting in a significantly decreased respiration and attenuated production of mitochondrial reactive oxygen species (mtROS). Mechanistically, LIX1 knock-down impaired the stability of the mitochondrial proteins PHB2 and OPA1 that are found in complexes with mitochondrial-specific phospholipids and are required for cristae organization. Supplementation with unsaturated fatty acids counteracted the effects of LIX1 knock-down on mitochondrial morphology and ultrastructure and restored YAP1/TAZ signaling. Collectively, our data demonstrate that LIX1 is a key regulator of cristae organization, modulating mtROS level and subsequently regulating the signaling cascades that control fate commitment of digestive mesenchyme-derived cells.


Subject(s)
Cysteine , Mitochondria , Cysteine/metabolism , Mesoderm/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Phospholipids/metabolism , Reactive Oxygen Species/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
9.
Int J Cardiovasc Imaging ; 38(1): 79-89, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34905152

ABSTRACT

Prognosis of Duchenne muscular dystrophy (DMD) is related to cardiac dysfunction. Two dimensional-speckle tracking echocardiography (2D-STE) has recently emerged as a non-invasive functional biomarker for early detection of DMD-related cardiomyopathy. This study aimed to determine, in DMD children, the existence of left ventricle (LV) dyssynchrony using 2D-STE analysis. This prospective controlled study enrolled 25 boys with DMD (mean age 11.0 ± 3.5 years) with normal LV ejection fraction and 50 age-matched controls. Three measures were performed to assess LV mechanical dyssynchrony: the opposing-wall delays (longitudinal and radial analyses), the modified Yu index, and the time-to-peak delays of each segment. Feasibility and reproducibility of 2D-STE dyssynchrony were evaluated. All three mechanical dyssynchrony criteria were significantly higher in the DMD group than in healthy subjects: (1) opposing-wall delays in basal inferoseptal to basal anterolateral segments (61.4 ± 45.3 ms vs. 18.3 ± 50.4 ms, P < 0.001, respectively) and in mid inferoseptal to mid anterolateral segments (58.6 ± 35.3 ms vs. 42.4 ± 36.4 ms, P < 0.05, respectively), (2) modified Yu index (33.3 ± 10.1 ms vs. 28.5 ± 8.1 ms, P < 0.05, respectively), and (3) most of time-to-peak values, especially in basal and mid anterolateral segments. Feasibility was excellent and reliability was moderate to excellent, with ICC values ranging from 0.49 to 0.97. Detection of LV mechanical dyssynchrony using 2D-STE analysis is an easily and reproducible method in paediatric DMD. The existence of an early LV mechanical dyssynchrony visualized using 2D-STE analysis in children with DMD before the onset of cardiomyopathy represents a perspective for future paediatric drug trials in the DMD-related cardiomyopathy prevention.Clinical Trial Registration Clinicaltrials.gov NCT02418338. Post-hoc study, registered on April 16, 2015.


Subject(s)
Muscular Dystrophy, Duchenne , Ventricular Dysfunction, Left , Adolescent , Child , Echocardiography , Heart Ventricles/diagnostic imaging , Humans , Male , Muscular Dystrophy, Duchenne/complications , Muscular Dystrophy, Duchenne/diagnostic imaging , Predictive Value of Tests , Prospective Studies , Reproducibility of Results , Ventricular Dysfunction, Left/diagnostic imaging , Ventricular Dysfunction, Left/etiology
10.
Cell Death Dis ; 12(9): 824, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34471096

ABSTRACT

Cyclic adenosine monophosphate (cAMP) is a master regulator of mitochondrial metabolism but its precise mechanism of action yet remains unclear. Here, we found that a dietary saturated fatty acid (FA), palmitate increased intracellular cAMP synthesis through the palmitoylation of soluble adenylyl cyclase in cardiomyocytes. cAMP further induced exchange protein directly activated by cyclic AMP 1 (Epac1) activation, which was upregulated in the myocardium of obese patients. Epac1 enhanced the activity of a key enzyme regulating mitochondrial FA uptake, carnitine palmitoyltransferase 1. Consistently, pharmacological or genetic Epac1 inhibition prevented lipid overload, increased FA oxidation (FAO), and protected against mitochondrial dysfunction in cardiomyocytes. In addition, analysis of Epac1 phosphoproteome led us to identify two key mitochondrial enzymes of the the ß-oxidation cycle as targets of Epac1, the long-chain FA acyl-CoA dehydrogenase (ACADL) and the 3-ketoacyl-CoA thiolase (3-KAT). Epac1 formed molecular complexes with the Ca2+/calmodulin-dependent protein kinase II (CaMKII), which phosphorylated ACADL and 3-KAT at specific amino acid residues to decrease lipid oxidation. The Epac1-CaMKII axis also interacted with the α subunit of ATP synthase, thereby further impairing mitochondrial energetics. Altogether, these findings indicate that Epac1 disrupts the balance between mitochondrial FA uptake and oxidation leading to lipid accumulation and mitochondrial dysfunction, and ultimately cardiomyocyte death.


Subject(s)
Cyclic AMP/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Palmitic Acid/toxicity , Adenylyl Cyclases/metabolism , Amino Acid Sequence , Animals , Animals, Newborn , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Catecholamines/metabolism , Guanine Nucleotide Exchange Factors/chemistry , Humans , L-Lactate Dehydrogenase/metabolism , Lipoylation/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Models, Biological , Myocytes, Cardiac/metabolism , Oxidation-Reduction , Phosphoproteins/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , Solubility , Stress, Physiological/drug effects
11.
Circulation ; 143(6): 566-580, 2021 02 09.
Article in English | MEDLINE | ID: mdl-33272024

ABSTRACT

BACKGROUND: Ischemic cardiovascular diseases, particularly acute myocardial infarction (MI), is one of the leading causes of mortality worldwide. Indoleamine 2, 3-dioxygenase 1 (IDO) catalyzes 1 rate-limiting step of L-tryptophan metabolism, and emerges as an important regulator of many pathological conditions. We hypothesized that IDO could play a key role to locally regulate cardiac homeostasis after MI. METHODS: Cardiac repair was analyzed in mice harboring specific endothelial or smooth muscle cells or cardiomyocyte or myeloid cell deficiency of IDO and challenged with acute myocardial infarction. RESULTS: We show that kynurenine generation through IDO is markedly induced after MI in mice. Total genetic deletion or pharmacological inhibition of IDO limits cardiac injury and cardiac dysfunction after MI. Distinct loss of function of IDO in smooth muscle cells, inflammatory cells, or cardiomyocytes does not affect cardiac function and remodeling in infarcted mice. In sharp contrast, mice harboring endothelial cell-specific deletion of IDO show an improvement of cardiac function as well as cardiomyocyte contractility and reduction in adverse ventricular remodeling. In vivo kynurenine supplementation in IDO-deficient mice abrogates the protective effects of IDO deletion. Kynurenine precipitates cardiomyocyte apoptosis through reactive oxygen species production in an aryl hydrocarbon receptor-dependent mechanism. CONCLUSIONS: These data suggest that IDO could constitute a new therapeutic target during acute MI.


Subject(s)
Endothelial Cells/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/therapeutic use , Kynurenine/therapeutic use , Myocardial Infarction/drug therapy , Animals , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/pharmacology , Kynurenine/pharmacology , Mice , Myocardial Infarction/physiopathology
12.
Arch Cardiovasc Dis ; 113(10): 652-659, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32712201

ABSTRACT

The inflammatory response is frequent after acute myocardial infarction, and may worsen ischaemia-reperfusion injuries, leading to increased infarct size and poor prognosis. Therefore, inflammation may be a promising therapeutic target, and anti-inflammatory drugs appear to be potential additional treatments in this context. Among these treatments, colchicine-a well-known drug that has been used for centuries in clinical practice for rheumatism-may represent the ideal candidate. Indeed, colchicine exerts direct anti-inflammatory and pleiotropic effects, with potential anti-arrhythmic, anti-fibrotic and anti-atherosclerotic effects, which are particularly interesting in this population of patients. The effects of colchicine in the context of acute myocardial infarction were first studied in preclinical models, with a decrease in inflammation demonstrated in several in vitro and in vivo models. Moreover, a decrease in infarct size and positive effects on haemodynamic variables were also recently demonstrated in a mouse model. Regarding clinical studies, the positive effect of colchicine in stable coronary disease and atherosclerosis was assessed initially. More recently, the value of colchicine in acute myocardial infarction has been studied, showing a positive effect on inflammation and infarct size reduction. Finally, a randomised trial (the COLCOT study) has shown a reduction in outcomes in patients with acute coronary syndrome treated with colchicine.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Colchicine/therapeutic use , Inflammation Mediators/antagonists & inhibitors , Inflammation/drug therapy , Myocardial Infarction/drug therapy , Myocarditis/drug therapy , Animals , Anti-Inflammatory Agents/adverse effects , Colchicine/adverse effects , Humans , Inflammation/metabolism , Inflammation/pathology , Inflammation/physiopathology , Inflammation Mediators/metabolism , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocarditis/metabolism , Myocarditis/pathology , Myocarditis/physiopathology , Myocardium/metabolism , Myocardium/pathology , Signal Transduction , Treatment Outcome
13.
Redox Biol ; 34: 101554, 2020 07.
Article in English | MEDLINE | ID: mdl-32464499

ABSTRACT

INTRODUCTION: When confronted to stress or pathological conditions, the mitochondria overproduce reactive species that participate in the cellular dysfunction. These organelles are however difficult to target with antioxidants. A feature of mitochondria that can be used for this is the negatively charged compartments they form. Most of mitochondrion-targeting antioxidants are therefore cationic synthetic molecules. Our hypothesis is that such mitochondriotropic traits might also exists in natural molecules. AIM: We tested here whether sinapine, a natural phenolic antioxidant-bearing a permanent positive charge, can target mitochondria to modulate mitochondrial oxidative stress. METHODS: Experiments were performed in-vitro, in-cellulo, ex-vivo, and in-vivo, using cardiac tissue. The sinapic acid -lacking the positively-charged-choline-moiety present in sinapine-was used as a control. Sinapine entry into mitochondria was investigated in-vivo and in cardiomyocytes. We used fluorescent probes to detect cytosolic (H2DCFDA) and mitochondrial (DHR123) oxidative stress on cardiomyocytes induced with either hydrogen peroxide (H2O2) or antimycin A, respectively. Finally, ROS production was measured with DHE 10 min after ischemia-reperfusion (IR) on isolated heart, treated or not with sinapine, sinapic acid or with a known synthetic mitochondrion-targeted antioxidant (mitoTempo). RESULTS: We detected the presence of sinapine within mitochondria in-vitro, after incubation of isolated cardiomyocytes, and in-vivo, after oral treatment. The presence of sinapic acid was not detected in the mitochondria. Both the sinapine and the sinapic acid limited cytosolic oxidative stress in response to H2O2. Only sinapine was able to blunt oxidative stress resulting from antimycin A-induced mtROS. Both mitoTempo and sinapine improved cardiac functional recovery following IR. This was associated with lower ROS production within the cardiac tissue. CONCLUSION: Sinapine, a natural cationic hydrophilic phenol, commonly and substantially found in rapeseed species, effectively (i) enters within the mitochondria, (ii) selectively decreases the level of mitochondrial oxidative stress and, (iii) efficiently limits ROS production during cardiac ischemia-reperfusion.


Subject(s)
Hydrogen Peroxide , Myocytes, Cardiac , Antioxidants/metabolism , Antioxidants/pharmacology , Choline/analogs & derivatives , Coumaric Acids , Hydrogen Peroxide/metabolism , Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism
14.
Front Cell Dev Biol ; 8: 609493, 2020.
Article in English | MEDLINE | ID: mdl-33569379

ABSTRACT

Besides skeletal muscle dysfunction, Duchenne muscular dystrophy (DMD) exhibits a progressive cardiomyopathy characterized by an impaired calcium (Ca2+) homeostasis and a mitochondrial dysfunction. Here we aimed to determine whether sarco-endoplasmic reticulum (SR/ER)-mitochondria interactions and mitochondrial function were impaired in dystrophic heart at the early stage of the pathology. For this purpose, ventricular cardiomyocytes and mitochondria were isolated from 3-month-old dystrophin-deficient mice (mdx mice). The number of contacts points between the SR/ER Ca2+ release channels (IP3R1) and the porine of the outer membrane of the mitochondria, VDAC1, measured using in situ proximity ligation assay, was greater in mdx cardiomyocytes. Expression levels of IP3R1 as well as the mitochondrial Ca2+ uniporter (MCU) and its regulated subunit, MICU1, were also increased in mdx heart. MICU2 expression was however unchanged. Furthermore, the mitochondrial Ca2+ uptake kinetics and the mitochondrial Ca2+ content were significantly increased. Meanwhile, the Ca2+-dependent pyruvate dehydrogenase phosphorylation was reduced, and its activity significantly increased. In Ca2+-free conditions, pyruvate-driven complex I respiration was decreased whereas in the presence of Ca2+, complex I-mediated respiration was boosted. Further, impaired complex I-mediated respiration was independent of its intrinsic activity or expression, which remains unchanged but is accompanied by an increase in mitochondrial reactive oxygen species production. Finally, mdx mice were treated with the complex I modulator metformin for 1 month. Metformin normalized the SR/ER-mitochondria interaction, decreased MICU1 expression and mitochondrial Ca2+ content, and enhanced complex I-driven respiration. In summary, before any sign of dilated cardiomyopathy, the DMD heart displays an aberrant SR/ER-mitochondria coupling with an increase mitochondrial Ca2+ homeostasis and a complex I dysfunction. Such remodeling could be reversed by metformin providing a novel therapeutic perspective in DMD.

15.
Future Sci OA ; 7(2): FSO656, 2020 Nov 23.
Article in English | MEDLINE | ID: mdl-33437519

ABSTRACT

PURPOSE: To evaluate the impact of colchicine on sympathetic denervation after acute myocardial infarction (AMI). MATERIALS & METHODS: Ischemia/Reperfusion was induced in C57BL/6J male mice. Left coronary artery was ligated during 45 min followed by reperfusion. 400 µg/kg of colchicine or the placebo was administrated intraperitoneally 15 min before the reperfusion. RESULTS: Colchicine treatment significantly improved heart rate variability index after AMI. Colchicine prevented sympathetic denervation in the remote area (p = 0.04) but not in the scar area (p = 0.70). CONCLUSION: These results suggest promising protective pathway of colchicine after AMI.

16.
Mol Ther ; 28(1): 171-179, 2020 01 08.
Article in English | MEDLINE | ID: mdl-31607542

ABSTRACT

Recessive forms of catecholaminergic polymorphic ventricular tachycardia (CPVT) are induced by mutations in genes encoding triadin or calsequestrin, two proteins that belong to the Ca2+ release complex, responsible for intracellular Ca2+ release triggering cardiac contractions. To better understand the mechanisms of triadin-induced CPVT and to assay multiple therapeutic interventions, we used a triadin knockout mouse model presenting a CPVT-like phenotype associated with a decrease in calsequestrin protein level. We assessed different approaches to rescue protein expression and to correct intracellular Ca2+ release and cardiac function: pharmacological treatment with kifunensine or a viral gene transfer-based approach, using adeno-associated virus serotype 2/9 (AAV2/9) encoding the triadin or calsequestrin. We observed that the levels of triadin and calsequestrin are intimately linked, and that reduction of both proteins contributes to the CPVT phenotype. Different combinations of triadin and calsequestrin expression level were obtained using these therapeutic approaches. A full expression of each is not necessary to correct the phenotype; a fine-tuning of the relative re-expression of both triadin and calsequestrin is required to correct the CPVT phenotype and rescue the cardiac function. AAV-mediated gene delivery of calsequestrin or triadin and treatment with kifunensine are potential treatments for recessive forms of CPVT due to triadin mutations.


Subject(s)
Calsequestrin/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Muscle Proteins/metabolism , Tachycardia, Ventricular/metabolism , Alkaloids/therapeutic use , Animals , Arrhythmias, Cardiac/drug therapy , Calcium/metabolism , Calcium Signaling/genetics , Calsequestrin/genetics , Dependovirus , Disease Models, Animal , Genetic Therapy/methods , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Knockout , Muscle Proteins/genetics , Myocardial Contraction/drug effects , Myocardial Contraction/genetics , Myocytes, Cardiac/metabolism , Parvovirinae/genetics , Phenotype , Rats , Tachycardia, Ventricular/drug therapy , Tachycardia, Ventricular/pathology , Transduction, Genetic , Transfection
17.
J Am Soc Echocardiogr ; 32(3): 412-422, 2019 03.
Article in English | MEDLINE | ID: mdl-30679141

ABSTRACT

BACKGROUND: Prognosis of Duchenne muscular dystrophy (DMD) is related to cardiac dysfunction. Speckle-tracking echocardiographic (STE) imaging is emerging as a noninvasive functional biomarker to consider in the early detection of DMD-related cardiomyopathy. However, STE analysis has not been assessed in a prospectively controlled study, especially in presymptomatic children with DMD, and no study has used STE analysis in all three displacements (longitudinal, radial, and circumferential) and for both ventricles. METHODS: This prospective controlled study enrolled 108 boys, 36 of whom had DMD (mean age, 11 ± 3.8 years) and 72 of whom were age-matched control subjects in a 1:2 case-control design. Conventional echocardiographic variables were collected for the left and right ventricles. STE analyses were performed in the longitudinal, radial, and circumferential displacements for the left ventricle and in the free wall longitudinal displacement for the right ventricle. The effect of age on the evolution of two-dimensional strain in children with DMD was studied by adding an interaction term, DMD × age, in the models. RESULTS: Conventional echocardiographic measures were normal in both groups. Left ventricular (LV) ejection fraction ranged from 45% to 76% (mean, 63 ± 6%) in the DMD group and from 55% to 76% (mean, 64 ± 5%) in the control group. Global LV strain mean measures were significantly worse in the DMD group for the longitudinal (-16.8 ± 3.9% vs -20.6 ± 2.6%, P < .0001), radial (22.7 ± 11.3% vs 31.7 ± 14%, P = .002), and circumferential (-16.5 ± 3.8% vs -20.3 ± 3.1%, P < .0001) displacements. The decrease of global LV longitudinal strain with age in children with DMD was 0.34% per year more marked than that in control subjects. The LV inferolateral and anterolateral segments were specifically impaired, especially in the basal area. Right ventricular function evaluated using conventional echocardiography and STE analysis was normal and not different between children with DMD and control subjects. CONCLUSIONS: The existence of altered LV strain despite normal LV function in children with DMD represents an important perspective for future pediatric drug trials in DMD-related cardiomyopathy prevention.


Subject(s)
Cardiomyopathies/diagnosis , Echocardiography, Three-Dimensional/methods , Muscular Dystrophy, Duchenne/diagnosis , Stroke Volume/physiology , Adolescent , Cardiomyopathies/etiology , Cardiomyopathies/physiopathology , Child , Child, Preschool , Cross-Sectional Studies , Early Diagnosis , Female , Humans , Infant , Infant, Newborn , Male , Muscular Dystrophy, Duchenne/complications , Muscular Dystrophy, Duchenne/physiopathology , Prognosis , Prospective Studies , Reproducibility of Results
18.
Sci Signal ; 11(553)2018 10 23.
Article in English | MEDLINE | ID: mdl-30352948

ABSTRACT

Communication between the endoplasmic reticulum (ER) and mitochondria plays a pivotal role in Ca2+ signaling, energy metabolism, and cell survival. Dysfunction in this cross-talk leads to metabolic and neurodegenerative diseases. Wolfram syndrome is a fatal neurodegenerative disease caused by mutations in the ER-resident protein WFS1. Here, we showed that WFS1 formed a complex with neuronal calcium sensor 1 (NCS1) and inositol 1,4,5-trisphosphate receptor (IP3R) to promote Ca2+ transfer between the ER and mitochondria. In addition, we found that NCS1 abundance was reduced in WFS1-null patient fibroblasts, which showed reduced ER-mitochondria interactions and Ca2+ exchange. Moreover, in WFS1-deficient cells, NCS1 overexpression not only restored ER-mitochondria interactions and Ca2+ transfer but also rescued mitochondrial dysfunction. Our results describe a key role of NCS1 in ER-mitochondria cross-talk, uncover a pathogenic mechanism for Wolfram syndrome, and potentially reveal insights into the pathogenesis of other neurodegenerative diseases.


Subject(s)
Calcium/metabolism , Endoplasmic Reticulum/metabolism , Mitochondria/metabolism , Neuronal Calcium-Sensor Proteins/metabolism , Neuropeptides/metabolism , Wolfram Syndrome/metabolism , Animals , Ear, Inner/metabolism , Fibroblasts/metabolism , HEK293 Cells , Humans , Membrane Potential, Mitochondrial , Mice , Neurons/metabolism , Oxygen Consumption , RNA, Small Interfering/metabolism , Signal Transduction
19.
Cardiovasc Res ; 114(10): e68-e69, 2018 08 01.
Article in English | MEDLINE | ID: mdl-30052919
SELECTION OF CITATIONS
SEARCH DETAIL
...