Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Am J Respir Cell Mol Biol ; 69(2): 230-241, 2023 08.
Article in English | MEDLINE | ID: mdl-37163759

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is a clinical entity of increasing significance. COPD involves abnormalities of the airways and, in emphysema, parenchymal pulmonary destruction. Cardiovascular disease has emerged as a significant comorbidity to COPD. Heart failure with preserved ejection fraction (HFpEF) appears to be particularly associated with COPD-emphysema. Traditional treatments have shown limited efficacy in improving COPD-associated HFpEF. This lack of therapeutic efficacy highlights the need to identify potential mechanisms that link COPD-emphysema to HFpEF. Therefore, we aimed to study the delayed cardiac physiological impacts in a rat model with acute exacerbated emphysema. Emphysema was induced by four weekly 4 units elastase (ELA) intratracheal pulmonary instillations and exacerbation by one final additional lipolysaccharide (LPS) instillation in male Wistar rats. At 5 weeks after the ELA and LPS exposure, in vivo and ex vivo pulmonary and cardiac measurements were performed. Experimental exacerbated emphysema resulted in decreased pulmonary function and exercise intolerance. Histological analysis revealed parenchymal pulmonary destruction without signs of inflammation or cardiac fibrosis. In vivo cardiac functional analysis revealed diastolic dysfunction and tachycardia. Ex vivo analysis revealed a cellular cardiomyopathy with decreased myofilament Ca2+ sensitivity, cross-bridge cycling kinetics, and increased adrenergic PKA (protein kinase A)-dependent phosphorylation of troponin-I. Experimental exacerbated emphysema was associated with exercise intolerance that appeared to be secondary to increased ß-adrenergic tone and subsequent cardiac myofilament dysfunction. A ß1-receptor antagonist treatment (bisoprolol) started 24 hours after ELA-LPS instillation prevented in vivo and ex vivo diastolic dysfunction. These results suggest that novel treatment strategies targeted to the cardiac myofilament may be beneficial to combat exacerbated emphysema-associated HFpEF.


Subject(s)
Cardiomyopathies , Emphysema , Heart Failure , Pulmonary Disease, Chronic Obstructive , Pulmonary Emphysema , Male , Rats , Animals , Heart Failure/complications , Lipopolysaccharides , Stroke Volume/physiology , Rats, Wistar , Pulmonary Emphysema/pathology , Pulmonary Disease, Chronic Obstructive/pathology , Cardiomyopathies/complications
2.
Cardiovasc Drugs Ther ; 36(5): 793-803, 2022 10.
Article in English | MEDLINE | ID: mdl-34138361

ABSTRACT

CONTEXT: Duchenne muscular dystrophy (DMD) is associated with a progressive alteration in cardiac function. OBJECTIVE: The aim of this study was to detect early cardiac dysfunction using the high sensitive two-dimensional speckle-tracking echocardiography (2D strain) in mdx mouse model and to investigate the potential preventive effects of the S107 ryanodine receptor (RyR2) stabilizer on early onset of DMD-related cardiomyopathy. METHODS AND RESULTS: Conventional echocardiography and global and segmental left ventricle (LV) 2D strains were assessed in male mdx mice and control C57/BL10 mice from 2 to 12 months of age. Up to 12 months of age, mdx mice showed preserved myocardial function as assessed by conventional echocardiography. However, global longitudinal, radial, and circumferential LV 2D strains significantly declined in mdx mice compared to controls from the 9 months of age. Segmental 2D strain analysis found a predominant alteration in posterior, inferior, and lateral LV segments, with a more marked impairment with aging. Then, mdx mice were treated with S107 in the drinking water at a dose of 250 mg/L using two different protocols: earlier therapy from 2 to 6 months of age and later therapy from 6 to 9 months of age. The treatment with S107 was efficient only when administered earlier in very young animals (from 2 to 6 months of age) and prevented the segmental alterations seen in non-treated mdx mice. CONCLUSIONS: This is the first animal study to evaluate the therapeutic effect of a drug targeting early onset of DMD-related cardiomyopathy, using 2D strain echocardiography. Speckle-tracking analyses revealed early alterations of LV posterior segments that could be prevented by 4 months of RyR2 stabilization.


Subject(s)
Cardiomyopathies , Drinking Water , Muscular Dystrophy, Duchenne , Animals , Cardiomyopathies/diagnostic imaging , Cardiomyopathies/drug therapy , Cardiomyopathies/etiology , Male , Mice , Mice, Inbred mdx , Muscular Dystrophy, Duchenne/complications , Muscular Dystrophy, Duchenne/diagnosis , Muscular Dystrophy, Duchenne/drug therapy , Ryanodine Receptor Calcium Release Channel
3.
J Am Coll Cardiol ; 78(24): 2439-2453, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34886965

ABSTRACT

BACKGROUND: Duchenne muscular dystrophy is associated with progressive deterioration in left ventricular (LV) function. The golden retriever muscular dystrophy (GRMD) dog model recapitulates the pathology and clinical manifestations of Duchenne muscular dystrophy. Importantly, they develop progressive LV dysfunction starting at early age. OBJECTIVES: The authors tested the cardioprotective effect of chronic administration of the ARM036, a small molecule that stabilizes the closed conformation of the cardiac sarcoplasmic reticulum ryanodine receptor/calcium release channel (RyR2) in young GRMD-dogs. METHODS: Two-month-old GRMD-dogs were treated with ARM036 or placebo for 4 months. Healthy-dogs of the same genetic background served as controls. Cardiac function was evaluated by conventional and 2-dimensional speckle-tracking echocardiography. Cardiac cellular and molecular analyses were performed at 6 months old. RESULTS: Conventional echocardiography showed normal LV dimensions and ejection fraction in 6-month-old GRMD dogs. Interestingly, 2-dimensional speckle-tracking echocardiography revealed decreased global longitudinal strain and the presence of hypokinetic segments in placebo-treated GRMD dogs. Single-channel measurements revealed higher RyR2 open probability at low resting Ca2+ in GRMD cardiomyocytes than in controls. ARM036 prevented those in vivo and in vitro dysfunctions in GRMD dogs. Myofilament Ca2+-sensitivity was increased in permeabilized GRMD cardiomyocytes at short sarcomere length. ARM036 had no effect on this parameter. Cross-bridge cycling kinetics were altered in GRMD myocytes and recovered with ARM036 treatment, which coincided with the level of myosin binding protein-C-S glutathionylation. CONCLUSIONS: GRMD-dogs exhibit early LV dysfunction associated with altered myofilament contractile properties. These abnormalities were prevented pharmacologically by stabilizing RyR2 with ARM036.


Subject(s)
Muscular Dystrophy, Duchenne/complications , Ryanodine Receptor Calcium Release Channel/metabolism , Ventricular Dysfunction, Left/etiology , Ventricular Function, Left/physiology , Animals , Biopsy , Disease Models, Animal , Dogs , Echocardiography , Muscular Dystrophy, Duchenne/diagnosis , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myofibrils/metabolism , Myofibrils/pathology , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum/pathology , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/pathology
4.
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
5.
Int J Mol Sci ; 22(2)2021 Jan 11.
Article in English | MEDLINE | ID: mdl-33440843

ABSTRACT

Numerous protocols of cardiac differentiation have been established by essentially focusing on specific growth factors on human pluripotent stem cell (hPSC) differentiation efficiency. However, the optimal environmental factors to obtain cardiac myocytes in network are still unclear. The mesoderm germ layer differentiation is known to be enhanced by low oxygen exposure. Here, we hypothesized that low oxygen exposure enhances the molecular and functional maturity of the cardiomyocytes. We aimed at comparing the molecular and functional consequences of low (5% O2 or LOE) and high oxygen exposure (21% O2 or HOE) on cardiac differentiation of hPSCs in 2D- and 3D-based protocols. hPSC-CMs were differentiated through both the 2D (monolayer) and 3D (embryoid body) protocols using several lines. Cardiac marker expression and cell morphology were assessed. The mitochondrial localization and metabolic properties were evaluated. The intracellular Ca2+ handling and contractile properties were also monitored. The 2D cardiac monolayer can only be differentiated in HOE. The 3D cardiac spheroids containing hPSC-CMs in LOE further exhibited cardiac markers, hypertrophy, steadier SR Ca2+ release properties revealing a better SR Ca2+ handling, and enhanced contractile force. Preserved distribution of mitochondria and similar oxygen consumption by the mitochondrial respiratory chain complexes were also observed. Our results brought evidences that LOE is moderately beneficial for the 3D cardiac spheroids with hPSC-CMs exhibiting further maturity. In contrast, the 2D cardiac monolayers strictly require HOE.


Subject(s)
Cell Differentiation , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Oxygen/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Biomarkers , Calcium/metabolism , Cell Culture Techniques , Gene Expression , Humans , Mitochondria, Heart/metabolism , Sarcoplasmic Reticulum/metabolism , Spheroids, Cellular
6.
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
7.
Theranostics ; 10(9): 4006-4016, 2020.
Article in English | MEDLINE | ID: mdl-32226535

ABSTRACT

Sonic hedgehog (SHH) signaling pathway is involved in embryonic tissue patterning and development. Our previous work identified, in small rodent model of ischemia reperfusion, SHH as a specific efficient tool to reduce infarct size and subsequent arrhythmias by preventing ventricular repolarization abnormalities. The goal of the present study was to provide a proof of concept of the cardioprotective effect of SHH ligand in a porcine model of acute ischemia. Methods: The antiarrhythmic effect of SHH, either by a recombinant peptide (N-SHH) or shed membrane microparticles harboring SHH ligand (MPsSHH+), was evaluated in a first set of pigs following a short (25 min) coronary artery occlusion (CAO) followed by 24 hours-reperfusion (CAR) (Protocol A). The infarct-limiting effect was evaluated on a second set of pigs with 40 min of coronary artery occlusion followed by 24 hours reperfusion (Protocol B). Electrocardiogram (ECG) was recorded and arrhythmia's scores were evaluated. Area at risk and myocardial infarct size were quantified. Results: In protocol A, administration of N-SHH 15 min. after the onset of coronary occlusion significantly reduced the occurrence of ventricular fibrillation compared to control group. Evaluation of arrhythmic score showed that N-SHH treatment significantly reduced the overall occurrence of arrhythmias. In protocol B, massive infarction was observed in control animals. Either N-SHH or MPsSHH+ treatment reduced significantly the infarct size with a concomitant increase of salvaged area. The reduction in infarct size was both accompanied by a significant decrease in systemic biomarkers of myocardial injury, i.e., cardiac troponin I and fatty acid-binding protein and an increase of eNOS activation. Conclusions: We show for the first time in a large mammalian model that the activation of the SHH pathway by N-SHH or MPsSHH+ offers a potent protection of the heart to ischemia-reperfusion by preventing the reperfusion arrhythmias, reducing the infarct area and the circulating levels of biomarkers for myocardial injury. These data open up potentially theranostic prospects for patients suffering from myocardial infarction to prevent the occurrence of arrhythmias and reduce myocardial tissue damage.


Subject(s)
Anti-Arrhythmia Agents/therapeutic use , Arrhythmias, Cardiac/drug therapy , Heart/drug effects , Hedgehog Proteins/therapeutic use , Myocardial Infarction/drug therapy , Myocardial Ischemia/drug therapy , Myocardial Reperfusion Injury/drug therapy , Animals , Myocardium , Swine
8.
J Mol Cell Cardiol ; 131: 155-163, 2019 06.
Article in English | MEDLINE | ID: mdl-31051181

ABSTRACT

The mechanical and cellular relationships between systole and diastole during left ventricular (LV) dysfunction remain to be established. LV contraction-relaxation coupling was examined during LV hypertrophy induced by chronic hypertension. Chronically instrumented pigs received angiotensin II infusion for4weeks to induce chronic hypertension (133 ±â€¯7 mmHg vs 98 ±â€¯5 mmHg for mean arterial pressure at Day 28 vs 0, respectively) and LV hypertrophy. LV function was investigated with the instrumentation and echocardiography for LV twist-untwist assessment before and after dobutamine infusion. The cellular mechanisms were investigated by exploring the intracellular Ca2+ handling. At Day 28, pigs exhibited LV hypertrophy with LV diastolic dysfunction (impaired LV isovolumic relaxation, increased LV end-diastolic pressure, decreased and delayed LV untwisting rate) and LV systolic dysfunction (impaired LV isovolumic contraction and twist) although LV ejection fraction was preserved. Isolated cardiomyocytes exhibited altered shortening and lengthening. Interestingly, contraction-relaxation coupling remained preserved both in vivo and in vitro during LV hypertrophy. LV systolic and diastolic dysfunctions were associated to post-translational remodeling and dysfunction of the type 2 cardiac ryanodine receptor/Ca2+ release channel (RyR2), i.e., PKA hyperphosphorylation of RyR2, depletion of calstabin 2 (FKBP12.6), RyR2 leak and hypersensitivity of RyR2 to cytosolic Ca2+ during both contraction and relaxation phases. In conclusion, LV contraction-relaxation coupling remained preserved during chronic hypertension despite LV systolic and diastolic dysfunctions. This implies that LV diastolic dysfunction is accompanied by LV systolic dysfunction. At the cellular level, this is linked to sarcoplasmic reticulum Ca2+ leak through PKA-mediated RyR2 hyperphosphorylation and depletion of its stabilizing partner.


Subject(s)
Diastole/physiology , Hypertension/physiopathology , Systole/physiology , Animals , Blotting, Western , Echocardiography , Heart Rate/physiology , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/physiopathology , Immunoprecipitation , Ryanodine Receptor Calcium Release Channel/metabolism , Swine , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/physiopathology , Ventricular Function, Left/physiology
9.
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
10.
J Mol Cell Cardiol ; 114: 345-353, 2018 01.
Article in English | MEDLINE | ID: mdl-29275006

ABSTRACT

AIM: Duchenne Muscular Dystrophy (DMD) is associated with progressive depressed left ventricular (LV) function. However, DMD effects on myofilament structure and function are poorly understood. Golden Retriever Muscular Dystrophy (GRMD) is a dog model of DMD recapitulating the human form of DMD. OBJECTIVE: The objective of this study is to evaluate myofilament structure and function alterations in GRMD model with spontaneous cardiac failure. METHODS AND RESULTS: We have employed synchrotron X-rays diffraction to evaluate myofilament lattice spacing at various sarcomere lengths (SL) on permeabilized LV myocardium. We found a negative correlation between SL and lattice spacing in both sub-epicardium (EPI) and sub-endocardium (ENDO) LV layers in control dog hearts. In the ENDO of GRMD hearts this correlation is steeper due to higher lattice spacing at short SL (1.9µm). Furthermore, cross-bridge cycling indexed by the kinetics of tension redevelopment (ktr) was faster in ENDO GRMD myofilaments at short SL. We measured post-translational modifications of key regulatory contractile proteins. S-glutathionylation of cardiac Myosin Binding Protein-C (cMyBP-C) was unchanged and PKA dependent phosphorylation of the cMyBP-C was significantly reduced in GRMD ENDO tissue and more modestly in EPI tissue. CONCLUSIONS: We found a gradient of contractility in control dogs' myocardium that spreads across the LV wall, negatively correlated with myofilament lattice spacing. Chronic stress induced by dystrophin deficiency leads to heart failure that is tightly associated with regional structural changes indexed by increased myofilament lattice spacing, reduced phosphorylation of regulatory proteins and altered myofilament contractile properties in GRMD dogs.


Subject(s)
Cardiomyopathies/pathology , Muscular Dystrophy, Duchenne/pathology , Myofibrils/pathology , Animals , Calcium/metabolism , Disease Models, Animal , Dogs , Electrocardiography , Intracellular Space/metabolism , Muscular Dystrophy, Duchenne/diagnostic imaging , Myocardium/pathology , Myofibrils/metabolism , Phosphorylation , Sarcomeres/metabolism , Signal Transduction , Troponin/metabolism
11.
Biochim Biophys Acta Mol Basis Dis ; 1863(9): 2229-2239, 2017 09.
Article in English | MEDLINE | ID: mdl-28625916

ABSTRACT

Besides its role in calcium (Ca2+) homeostasis, the sarco-endoplamic reticulum (SR/ER) controls protein folding and is tethered to mitochondria. Under pathophysiological conditions the unfolded protein response (UPR) is associated with disturbance in SR/ER-mitochondria crosstalk. Here, we investigated whether ER stress altered SR/ER-mitochondria links, Ca2+ handling and muscle damage in WT (Wild Type) and mdx mice, the murine model of Duchenne Muscular Dystrophy (DMD). In WT mice, the SR/ER-mitochondria links were decreased in isolated FDB muscle fibers after injection of ER stress activator tunicamycin (TM). Ca2+ imaging revealed an increase of cytosolic Ca2+ transient and a decrease of mitochondrial Ca2+ uptake. The force generating capacity of muscle dropped after TM. This impaired contractile function was accompanied by an increase in autophagy markers and calpain-1 activation. Conversely, ER stress inhibitors restored SR/ER-mitochondria links, mitochondrial Ca2+ uptake and improved diaphragm contractility in mdx mice. Our findings demonstrated that ER stress-altered SR/ER-mitochondria links, disturbed Ca2+ handling and muscle function in WT and mdx mice. Thus, ER stress may open up a prospect of new therapeutic targets in DMD.


Subject(s)
Calcium Signaling , Calcium/metabolism , Endoplasmic Reticulum Stress , Mitochondria, Muscle/metabolism , Muscular Dystrophy, Duchenne/metabolism , Sarcoplasmic Reticulum/metabolism , Animals , Autophagy/genetics , Calpain/genetics , Calpain/metabolism , Disease Models, Animal , Male , Mice , Mice, Inbred mdx , Mitochondria, Muscle/genetics , Mitochondria, Muscle/pathology , Muscle Contraction/genetics , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Sarcoplasmic Reticulum/genetics , Sarcoplasmic Reticulum/pathology
12.
J Muscle Res Cell Motil ; 38(1): 17-24, 2017 02.
Article in English | MEDLINE | ID: mdl-28260211

ABSTRACT

Respiratory muscle contractile inactivity during mechanical ventilation (MV) induces diaphragm muscle weakness, a condition referred to as ventilator-induced diaphragmatic dysfunction (VIDD). Although VIDD pathophysiological mechanisms are still not fully understood, it has been recently suggested that remodeling of the sarcoplasmic reticulum (SR) calcium release channel/ryanodine receptors (RyR1) in the diaphragm is a proximal mechanism of VIDD. Here, we used piglets, a large animal model of VIDD that is more relevant to human pathophysiology, to determine whether RyR1 alterations are observed in the presence of diaphragm weakness. In piglets, diaphragm weakness induced by 72 h of respiratory muscle unloading was associated with SR RyR1 remodeling and abnormal resting SR Ca2+ leak in the diaphragm. Specifically, following controlled mechanical ventilation, diaphragm contractile function was reduced. Moreover, RyR1 macromolecular complexes were more oxidized, S-nitrosylated and phosphorylated at Ser-2844 and depleted of the stabilizing subunit calstabin1 compared with controls on adaptive support ventilation that maintains diaphragmatic contractile activity. Our study strongly supports the hypothesis that RyR1 is a potential therapeutic target in VIDD and the interest of using small molecule drugs to prevent RyR1-mediated SR Ca2+ leak induced by respiratory muscle unloading in patients who require controlled mechanical ventilation.


Subject(s)
Diaphragm/physiopathology , Respiration, Artificial , Respiratory Muscles/physiopathology , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Diaphragm/metabolism , Female , Models, Animal , Muscle Weakness/etiology , Muscle Weakness/metabolism , Muscle Weakness/physiopathology , Respiratory Muscles/metabolism , Swine , Ventilators, Mechanical
13.
Free Radic Biol Med ; 102: 229-239, 2017 01.
Article in English | MEDLINE | ID: mdl-27932075

ABSTRACT

Acute myocardial infarction leads to an increase in oxidative stress and lipid peroxidation. 4(RS)-4-F4t-Neuroprostane (4-F4t-NeuroP) is a mediator produced by non-enzymatic free radical peroxidation of the cardioprotective polyunsaturated fatty acid, docosahexaenoic acid (DHA). In this study, we investigated whether intra-cardiac delivery of 4-F4t-NeuroP (0.03mg/kg) prior to occlusion (ischemia) prevents and protects rat myocardium from reperfusion damages. Using a rat model of ischemic-reperfusion (I/R), we showed that intra-cardiac infusion of 4-F4t-NeuroP significantly decreased infarct size following reperfusion (-27%) and also reduced ventricular arrhythmia score considerably during reperfusion (-41%). Most notably, 4-F4t-NeuroP decreased ventricular tachycardia and post-reperfusion lengthening of QT interval. The evaluation of the mitochondrial homeostasis indicates a limitation of mitochondrial swelling in response to Ca2+ by decreasing the mitochondrial permeability transition pore opening and increasing mitochondria membrane potential. On the other hand, mitochondrial respiration measured by oxygraphy, and mitochondrial ROS production measured with MitoSox red® were unchanged. We found decreased cytochrome c release and caspase 3 activity, indicating that 4-F4t-NeuroP prevented reperfusion damages and reduced apoptosis. In conclusion, 4-F4t-NeuroP derived from DHA was able to protect I/R cardiac injuries by regulating the mitochondrial homeostasis.


Subject(s)
Docosahexaenoic Acids/administration & dosage , Mitochondria, Heart/drug effects , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/drug therapy , Neuroprostanes/administration & dosage , Animals , Docosahexaenoic Acids/metabolism , Heart/drug effects , Heart/physiopathology , Humans , Lipid Peroxidation/genetics , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardium/metabolism , Myocardium/pathology , Oxidative Stress/genetics , Protective Agents/administration & dosage , Rats , Reactive Oxygen Species/metabolism , Tachycardia, Ventricular/drug therapy , Tachycardia, Ventricular/metabolism , Tachycardia, Ventricular/pathology
14.
Proc Natl Acad Sci U S A ; 113(32): 9069-74, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27457930

ABSTRACT

Ventilator-induced diaphragmatic dysfunction (VIDD) refers to the diaphragm muscle weakness that occurs following prolonged controlled mechanical ventilation (MV). The presence of VIDD impedes recovery from respiratory failure. However, the pathophysiological mechanisms accounting for VIDD are still not fully understood. Here, we show in human subjects and a mouse model of VIDD that MV is associated with rapid remodeling of the sarcoplasmic reticulum (SR) Ca(2+) release channel/ryanodine receptor (RyR1) in the diaphragm. The RyR1 macromolecular complex was oxidized, S-nitrosylated, Ser-2844 phosphorylated, and depleted of the stabilizing subunit calstabin1, following MV. These posttranslational modifications of RyR1 were mediated by both oxidative stress mediated by MV and stimulation of adrenergic signaling resulting from the anesthesia. We demonstrate in the murine model that such abnormal resting SR Ca(2+) leak resulted in reduced contractile function and muscle fiber atrophy for longer duration of MV. Treatment with ß-adrenergic antagonists or with S107, a small molecule drug that stabilizes the RyR1-calstabin1 interaction, prevented VIDD. Diaphragmatic dysfunction is common in MV patients and is a major cause of failure to wean patients from ventilator support. This study provides the first evidence to our knowledge of RyR1 alterations as a proximal mechanism underlying VIDD (i.e., loss of function, muscle atrophy) and identifies RyR1 as a potential target for therapeutic intervention.


Subject(s)
Diaphragm/physiopathology , Respiration, Artificial/adverse effects , Ryanodine Receptor Calcium Release Channel/physiology , Animals , Calcium/metabolism , Humans , Mice , Muscle Contraction , Oxidative Stress , Receptors, Adrenergic, beta/physiology , Signal Transduction , Tacrolimus Binding Proteins/physiology , Ventilators, Mechanical/adverse effects
15.
Anesthesiology ; 117(3): 560-7, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22766523

ABSTRACT

BACKGROUND: Controlled mechanical ventilation is associated with ventilator-induced diaphragmatic dysfunction, which impedes weaning from mechanical ventilation. To design future clinical trials in humans, a better understanding of the molecular mechanisms using knockout models, which exist only in the mouse, is needed. The aims of this study were to ascertain the feasibility of developing a murine model of ventilator-induced diaphragmatic dysfunction and to determine whether atrophy, sarcolemmal injury, and the main proteolysis systems are activated under these conditions. METHODS: Healthy adult male C57/BL6 mice were assigned to three groups: (1) mechanical ventilation with end-expiratory positive pressure of 2-4 cm H2O for 6 h (n=6), (2) spontaneous breathing with continuous positive airway pressure of 2-4 cm H2O for 6 h (n=6), and (3) controls with no specific intervention (n=6). Airway pressure and hemodynamic parameters were monitored. Upon euthanasia, arterial blood gases and isometric contractile properties of the diaphragm and extensor digitorum longus were evaluated. Histology and immunoblotting for the main proteolysis pathways were performed. RESULTS: Hemodynamic parameters and arterial blood gases were comparable between groups and within normal physiologic ranges. Diaphragmatic but not extensor digitorum longus force production declined in the mechanical ventilation group (maximal force decreased by approximately 40%) compared with the control and continuous positive airway pressure groups. No histologic difference was found between groups. In opposition with the calpains, caspase 3 was activated in the mechanical ventilation group. CONCLUSION: Controlled mechanical ventilation for 6 h in the mouse is associated with significant diaphragmatic but not limb muscle weakness without atrophy or sarcolemmal injury and activates proteolysis.


Subject(s)
Diaphragm/physiopathology , Muscle Weakness/etiology , Ventilators, Mechanical/adverse effects , Animals , Carbon Dioxide/blood , Disease Models, Animal , Hemodynamics , Male , Mice , Mice, Inbred C57BL , Muscle Contraction , Respiration , Respiration, Artificial/adverse effects
16.
Am J Respir Crit Care Med ; 183(3): 364-71, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-20813887

ABSTRACT

RATIONALE: Diaphragmatic function is a major determinant of the ability to successfully wean patients from mechanical ventilation (MV). Paradoxically, MV itself results in a rapid loss of diaphragmatic strength in animals. However, very little is known about the time course or mechanistic basis for such a phenomenon in humans. OBJECTIVES: To determine in a prospective fashion the time course for development of diaphragmatic weakness during MV; and the relationship between MV duration and diaphragmatic injury or atrophy, and the status of candidate cellular pathways implicated in these phenomena. METHODS: Airway occlusion pressure (TwPtr) generated by the diaphragm during phrenic nerve stimulation was measured in short-term (0.5 h; n = 6) and long-term (>5 d; n = 6) MV groups. Diaphragmatic biopsies obtained during thoracic surgery (MV for 2-3 h; n = 10) and from brain-dead organ donors (MV for 24-249 h; n = 15) were analyzed for ultrastructural injury, atrophy, and expression of proteolysis-related proteins (ubiquitin, nuclear factor-κB, and calpains). MEASUREMENTS AND MAIN RESULTS: TwPtr decreased progressively during MV, with a mean reduction of 32 ± 6% after 6 days. Longer periods of MV were associated with significantly greater ultrastructural fiber injury (26.2 ± 4.8 vs. 4.7 ± 0.6% area), decreased cross-sectional area of muscle fibers (1,904 ± 220 vs. 3,100 ± 329 µm²), an increase of ubiquitinated proteins (+19%), higher expression of p65 nuclear factor-κB (+77%), and greater levels of the calcium-activated proteases calpain-1, -2, and -3 (+104%, +432%, and +266%, respectively) in the diaphragm. CONCLUSIONS: Diaphragmatic weakness, injury, and atrophy occur rapidly in critically ill patients during MV, and are significantly correlated with the duration of ventilator support.


Subject(s)
Diaphragm/injuries , Muscle Weakness/etiology , Respiration, Artificial/adverse effects , Adult , Calpain/analysis , Diaphragm/chemistry , Diaphragm/pathology , Diaphragm/physiopathology , Female , Humans , Male , Middle Aged , Muscle Weakness/pathology , Muscle Weakness/physiopathology , Muscular Atrophy/etiology , Muscular Atrophy/pathology , Muscular Atrophy/physiopathology , Time Factors , Transcription Factor RelA/analysis , Ubiquitinated Proteins/analysis , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...