Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters

Publication year range
1.
Am J Physiol Cell Physiol ; 324(6): C1223-C1235, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37125775

ABSTRACT

Dilated cardiomyopathy caused by mutations in LMNA, encoding A-type lamins (i.e., LMNA cardiomyopathy), is characterized by a left ventricle enlargement and ultimately results in poor cardiac contractility associated with conduction defects. Despite current strategies to aggressively manage the symptoms, the disorder remains a common cause of sudden death and heart failure with decreased ejection fraction. Patient care includes cardioverter defibrillator implantation but the last therapeutic option remains cardiac transplantation. A-type lamins are intermediate filaments and are the main components of the nuclear lamina, a meshwork underlying the inner nuclear membrane, which plays an essential role in both maintaining the nuclear structure and organizing the cytoskeletal structures within the cell. Cytoskeletal proteins function as scaffold to resist external mechanical stress. An increasing amount of evidence demonstrates that LMNA mutations can lead to disturbances in several structural and cytoskeletal components of the cell such as microtubules, actin cytoskeleton, and intermediate filaments. Collectively, this review focuses on the significance of these cytoskeletal modulators and emphasizes their potential therapeutic role in LMNA cardiomyopathy. Indeed, molecular tuning of cytoskeletal dynamics has been successfully used in preclinical models and provides adequate grounds for a therapeutic approach for patients with LMNA cardiomyopathy.


Subject(s)
Cardiomyopathies , Lamin Type A , Humans , Lamin Type A/genetics , Lamin Type A/metabolism , Cardiomyopathies/genetics , Cardiomyopathies/therapy , Cardiomyopathies/metabolism , Cytoskeleton/genetics , Cytoskeleton/metabolism , Microtubules/metabolism , Mutation/genetics
2.
Hum Mol Genet ; 29(24): 3919-3934, 2021 02 25.
Article in English | MEDLINE | ID: mdl-33388782

ABSTRACT

Mutations in the lamin A/C gene (LMNA), which encodes A-type lamins, cause several diseases called laminopathies, the most common of which is dilated cardiomyopathy with muscular dystrophy. The role of Ca2+ regulation in these diseases remain poorly understood. We now show biochemical remodeling of the ryanodine receptor (RyR)/intracellular Ca2+ release channel in heart samples from human subjects with LMNA mutations, including protein kinase A-catalyzed phosphorylation, oxidation and depletion of the stabilizing subunit calstabin. In the LmnaH222P/H222P murine model of Emery-Dreifuss muscular dystrophy caused by LMNA mutation, we demonstrate an age-dependent biochemical remodeling of RyR2 in the heart and RyR1 in skeletal muscle. This RyR remodeling is associated with heart and skeletal muscle dysfunction. Defective heart and muscle function are ameliorated by treatment with a novel Rycal small molecule drug (S107) that fixes 'leaky' RyRs. SMAD3 phosphorylation is increased in hearts and diaphragms of LmnaH222P/H222P mice, which enhances NADPH oxidase binding to RyR channels, contributing to their oxidation. There is also increased generalized protein oxidation, increased calcium/calmodulin-dependent protein kinase II-catalyzed phosphorylation of RyRs and increased protein kinase A activity in these tissues. Our data show that RyR remodeling plays a role in cardiomyopathy and skeletal muscle dysfunction caused by LMNA mutation and identify these Ca2+ channels as a potential therapeutic target.


Subject(s)
Cardiomyopathies/pathology , Disease Models, Animal , Heart/physiopathology , Lamin Type A/genetics , Muscular Dystrophies/pathology , Mutation , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Calcium Signaling , Cardiomyopathies/etiology , Cardiomyopathies/metabolism , Female , Homeostasis , Humans , Male , Mice , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophies/etiology , Muscular Dystrophies/metabolism , Ryanodine Receptor Calcium Release Channel/genetics
3.
Methods ; 190: 3-12, 2021 06.
Article in English | MEDLINE | ID: mdl-32278808

ABSTRACT

What if the next generation of successful treatments was hidden in the current pharmacopoeia? Identifying new indications for existing drugs, also called the drug repurposing or drug rediscovery process, is a highly efficient and low-cost strategy. First reported almost a century ago, drug repurposing has emerged as a valuable therapeutic option for diseases that do not have specific treatments and rare diseases, in particular. This review focuses on Hutchinson-Gilford progeria syndrome (HGPS), a rare genetic disorder that induces accelerated and precocious aging, for which drug repurposing has led to the discovery of several potential treatments over the past decade.


Subject(s)
Progeria , Humans , Lamin Type A/genetics , Pharmaceutical Preparations , Progeria/drug therapy , Progeria/genetics
4.
Hum Mol Genet ; 28(13): 2237-2244, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31220270

ABSTRACT

Autosomal Emery-Dreifuss muscular dystrophy (EDMD) is caused by mutations in the lamin A/C gene (LMNA) encoding A-type nuclear lamins, intermediate filament proteins of the nuclear envelope. Classically, the disease manifests as scapulo-humero-peroneal muscle wasting and weakness, early joint contractures and dilated cardiomyopathy with conduction blocks; however, variable skeletal muscle involvement can be present. Previously, we and other demonstrated altered activity of signaling pathways in hearts and striated muscles of LmnaH222P/H222P mice, a model of autosomal EDMD. We showed that blocking their activation improved cardiac function. However, the evaluation of the benefit of these treatments on the whole organism is suffering from a better knowledge of the performance in mouse models. We show in the present study that LmnaH222P/H222P mice display a significant loss of lean mass, consistent with the dystrophic process. This is associated with altered VO2 peak and respiratory exchange ratio. These results showed for the first time that LmnaH222P/H222P mice have decreased performance and provided a new useful means for future therapeutic interventions on this model of EDMD.


Subject(s)
Lamin Type A/genetics , Muscular Dystrophy, Emery-Dreifuss/genetics , Animals , Body Composition , Disease Models, Animal , Male , Mice , Mice, Transgenic , Muscular Dystrophy, Emery-Dreifuss/metabolism , Muscular Dystrophy, Emery-Dreifuss/physiopathology , Mutation , Ventricular Function, Left , Weight Loss
5.
Hum Mol Genet ; 28(24): 4043-4052, 2019 12 15.
Article in English | MEDLINE | ID: mdl-29893868

ABSTRACT

Mutations in the lamin A/C gene (LMNA) cause an autosomal dominant inherited form of dilated cardiomyopathy associated with cardiac conduction disease (hereafter referred to as LMNA cardiomyopathy). Compared with other forms of dilated cardiomyopathy, mutations in LMNA are responsible for a more aggressive clinical course owing to a high rate of malignant ventricular arrhythmias. Gap junctions are intercellular channels that allow direct communication between neighboring cells, which are involved in electrical impulse propagation and coordinated contraction of the heart. For gap junctions to properly control electrical synchronization in the heart, connexin-based hemichannels must be correctly targeted to intercalated discs, Cx43 being the major connexin in the working myocytes. We here showed an altered distribution of Cx43 in a mouse model of LMNA cardiomyopathy. However, little is known on the molecular mechanisms of Cx43 remodeling in pathological context. We now show that microtubule cytoskeleton alteration and decreased acetylation of α-tubulin lead to remodeling of Cx43 in LMNA cardiomyopathy, which alters the correct communication between cardiomyocytes, ultimately leading to electrical conduction disturbances. Preventing or reversing this process could offer a strategy to repair damaged heart. Stabilization of microtubule cytoskeleton using Paclitaxel improved intraventricular conduction defects. These results indicate that microtubule cytoskeleton contributes to the pathogenesis of LMNA cardiomyopathy and that drugs stabilizing the microtubule may be beneficial for patients.


Subject(s)
Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Connexin 43/metabolism , Lamin Type A/genetics , Paclitaxel/pharmacology , Acetylation/drug effects , Animals , Cardiac Conduction System Disease/genetics , Cardiomyopathies/pathology , Connexin 43/genetics , Cytoskeleton/metabolism , Cytoskeleton/pathology , Gap Junctions/drug effects , Gap Junctions/metabolism , Gap Junctions/pathology , Lamin Type A/metabolism , Male , Mice , Mice, Knockout , Microtubules/metabolism , Microtubules/pathology , Mutation , Myocardium/pathology , Myocytes, Cardiac/pathology
6.
FASEB J ; 34(2): 2987-3005, 2020 02.
Article in English | MEDLINE | ID: mdl-31908029

ABSTRACT

The expression of α-cardiac actin, a major constituent of the cytoskeleton of cardiomyocytes, is dramatically decreased in a mouse model of dilated cardiomyopathy triggered by inducible cardiac-specific serum response factor (Srf) gene disruption that could mimic some forms of human dilated cardiomyopathy. To investigate the consequences of the maintenance of α-cardiac actin expression in this model, we developed a new transgenic mouse based on Cre/LoxP strategy, allowing together the induction of SRF loss and a compensatory expression of α-cardiac actin. Here, we report that maintenance of α-cardiac actin within cardiomyocytes temporally preserved cytoarchitecture from adverse cardiac remodeling through a positive impact on both structural and transcriptional levels. These protective effects were accompanied in vivo by the decrease of ROS generation and protein carbonylation and the downregulation of NADPH oxidases NOX2 and NOX4. We also show that ectopic expression of α-cardiac actin protects HEK293 cells against oxidative stress induced by H2 O2 . Oxidative stress plays an important role in the development of cardiac remodeling and contributes also to the pathogenesis of heart failure. Taken together, these findings indicate that α-cardiac actin could be involved in the regulation of oxidative stress that is a leading cause of adverse remodeling during dilated cardiomyopathy development.


Subject(s)
Actins/metabolism , Cardiomyopathy, Dilated/metabolism , Myocytes, Cardiac/metabolism , Oxidative Stress , Actins/genetics , Animals , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Dilated/prevention & control , Disease Models, Animal , Female , Humans , Hydrogen Peroxide/pharmacology , Male , Mice , Mice, Transgenic , Myocytes, Cardiac/pathology , NADPH Oxidase 2/genetics , NADPH Oxidase 2/metabolism , NADPH Oxidase 4/genetics , NADPH Oxidase 4/metabolism
7.
Hum Mol Genet ; 27(19): 3353-3360, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29982513

ABSTRACT

Cardiomyopathy caused by lamin A/C gene (LMNA) mutations (hereafter referred as LMNA cardiomyopathy) is an anatomic and pathologic condition associated with muscular and electrical dysfunction of the heart, often leading to heart failure-related disability. There is currently no specific therapy available for patients that target the molecular pathophysiology of LMNA cardiomyopathy. We showed here an increase in oxidative stress levels in the hearts of mice carrying LMNA mutation, associated with a decrease of the key cellular antioxidant glutathione (GHS). Oral administration of N-acetyl cysteine, a GHS precursor, led to a marked improvement of GHS content, a decrease in oxidative stress markers including protein carbonyls and an improvement of left ventricular structure and function in a model of LMNA cardiomyopathy. Collectively, our novel results provide therapeutic insights into LMNA cardiomyopathy.


Subject(s)
Acetylcysteine/administration & dosage , Cardiomyopathy, Dilated/genetics , Heart Failure/genetics , Lamin Type A/genetics , Acetylcysteine/metabolism , Animals , Antioxidants/administration & dosage , Antioxidants/metabolism , Cardiomyopathy, Dilated/drug therapy , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/physiopathology , Disease Models, Animal , Glutathione/metabolism , Heart/drug effects , Heart/physiopathology , Heart Failure/drug therapy , Heart Failure/metabolism , Heart Failure/pathology , Heart Ventricles/drug effects , Heart Ventricles/physiopathology , Humans , Mice , Mutation , Myocardium/pathology , Oxidative Stress/drug effects
8.
Hum Mol Genet ; 27(22): 3870-3880, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30053027

ABSTRACT

Cardiomyopathy caused by lamin A/C gene (LMNA) mutations (hereafter referred as LMNA cardiomyopathy) is an anatomic and pathologic condition associated with muscle and electrical dysfunction of the heart, often leading to heart failure-related disability. There is currently no specific therapy available for patients that target the molecular pathophysiology of LMNA cardiomyopathy. Recent studies suggested that nicotinamide adenine dinucleotide (NAD+) cellular content could be a critical determinant for heart function. Biosynthesis of NAD+ from vitamin B3 (known as salvage pathways) is the primary source of NAD+. We showed here that NAD+ salvage pathway was altered in the heart of mouse and human carrying LMNA mutation, leading to an alteration of one of NAD+ co-substrate enzymes, PARP-1. Oral administration of nicotinamide riboside, a natural NAD+ precursor and a pyridine-nucleoside form of vitamin B3, leads to a marked improvement of the NAD+ cellular content, an increase of PARylation of cardiac proteins and an improvement of left ventricular structure and function in a model of LMNA cardiomyopathy. Collectively, our results provide mechanistic and therapeutic insights into dilated cardiomyopathy caused by LMNA mutations.


Subject(s)
Cardiomyopathies/genetics , Heart/physiopathology , Lamin Type A/genetics , NAD/genetics , Poly (ADP-Ribose) Polymerase-1/genetics , Animals , Cardiomyopathies/physiopathology , Disease Models, Animal , Heart Failure/genetics , Heart Failure/physiopathology , Heart Ventricles/metabolism , Heart Ventricles/physiopathology , Humans , Mice , Mutation , NAD/biosynthesis , Niacinamide/genetics , Niacinamide/metabolism , Poly ADP Ribosylation/genetics , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/physiopathology
9.
Hum Mol Genet ; 27(17): 3060-3078, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29878125

ABSTRACT

Hyper-activation of extracellular signal-regulated kinase (ERK) 1/2 contributes to heart dysfunction in cardiomyopathy caused by mutations in the lamin A/C gene (LMNA cardiomyopathy). The mechanism of how this affects cardiac function is unknown. We show that active phosphorylated ERK1/2 directly binds to and catalyzes the phosphorylation of the actin depolymerizing factor cofilin-1 on Thr25. Cofilin-1 becomes active and disassembles actin filaments in a large array of cellular and animal models of LMNA cardiomyopathy. In vivo expression of cofilin-1, phosphorylated on Thr25 by endogenous ERK1/2 signaling, leads to alterations in left ventricular function and cardiac actin. These results demonstrate a novel role for cofilin-1 on actin dynamics in cardiac muscle and provide a rationale on how increased ERK1/2 signaling leads to LMNA cardiomyopathy.


Subject(s)
Actins/metabolism , Cardiomyopathy, Dilated/pathology , Cofilin 1/metabolism , Lamin Type A/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mutation , Actins/genetics , Adolescent , Adult , Animals , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/metabolism , Case-Control Studies , Cofilin 1/genetics , Female , Heart/physiology , Humans , Lamin Type A/metabolism , Male , Mice , Middle Aged , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Phosphorylation , Signal Transduction , Young Adult
10.
Biochem Biophys Res Commun ; 529(3): 861-867, 2020 08 27.
Article in English | MEDLINE | ID: mdl-32540097

ABSTRACT

The cytoskeleton is a complex network interlinking filaments that extend throughout the cytoplasm from the nucleus to the plasma membrane. Three major types of filaments are found in the cytoskeleton: actin filaments, microtubules, and intermediate filaments. They play a key role in the ability of cells to both resist mechanical stress and generate force. However, the precise involvement of intermediate filament proteins in these processes remains unclear. Here, we focused on nuclear A-type lamins, which are connected to the cytoskeleton via the Linker of Nucleoskeleton and Cytoskeleton (LINC) complex. Using micro-constriction rheology, we investigated the impact of A-type lamins (p.H222P) mutation on the mechanical properties of muscle cells. We demonstrate that the expression of point mutation of lamin A in muscle cells increases cellular stiffness compared with cells expressing wild type lamin A and that the chemical agent selumetinib, an inhibitor of the ERK1/2 signaling, reversed the mechanical alterations in mutated cells. These results highlight the interplay between A-type lamins and mechano-signaling, which are supported by cell biology measurements.


Subject(s)
Lamin Type A/genetics , Muscle Fibers, Skeletal/cytology , Point Mutation , Animals , Biomechanical Phenomena , Cell Line , Lamin Type A/metabolism , MAP Kinase Signaling System , Mice , Muscle Fibers, Skeletal/metabolism
11.
Hum Mol Genet ; 26(2): 333-343, 2017 01 15.
Article in English | MEDLINE | ID: mdl-28069793

ABSTRACT

Cardiomyopathy caused by lamin A/C gene (LMNA) mutations (hereafter referred as LMNA cardiomyopathy) is characterized by cardiac conduction abnormalities and left ventricular systolic dysfunction predisposing to heart failure. Previous cardiac transcriptional profiling of LmnaH222P/H222P mouse, a small animal model of LMNA cardiomyopathy, suggested decreased WNT/ß-catenin signalling. We confirmed decreased WNT/ß-catenin signalling in the hearts of these mice by demonstrating decreased ß-catenin and WNT proteins. This was correlated with increased expression of soluble Frizzled-related proteins that modulate the WNT/ß-catenin signalling pathway. Hearts of LmnaH222P/H222P mice also demonstrated lowered expression of the gap junction connexin 43. Activation of WNT/ß-catenin activity with 6-bromoindirubin-3'-oxime improved cardiac contractility and ameliorated intraventricular conduction defects in LmnaH222P/H222P mice, which was associated with increased expression of myocardial connexin 43. These results indicate that decreased WNT/ß-catenin contributes to the pathophysiology of LMNA cardiomyopathy and that drugs activating ß-catenin may be beneficial in affected individuals.


Subject(s)
Cardiomyopathy, Dilated/genetics , Connexin 43/genetics , Lamin Type A/genetics , beta Catenin/genetics , Animals , Cardiomyopathy, Dilated/drug therapy , Cardiomyopathy, Dilated/physiopathology , Connexin 43/biosynthesis , Disease Models, Animal , Gene Expression Regulation/drug effects , Glycoproteins/biosynthesis , Glycoproteins/genetics , Heart Failure/drug therapy , Heart Failure/genetics , Heart Failure/physiopathology , Humans , Indoles/administration & dosage , Intracellular Signaling Peptides and Proteins , Mice , Mutation , Oximes/administration & dosage , Ventricular Dysfunction, Left/drug therapy , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/physiopathology , Wnt Proteins/genetics , Wnt Signaling Pathway/drug effects , beta Catenin/biosynthesis
12.
Curr Opin Neurol ; 32(5): 728-734, 2019 10.
Article in English | MEDLINE | ID: mdl-31460960

ABSTRACT

PURPOSE OF REVIEW: Emery-Dreifuss muscular dystrophy (EDMD) is caused by mutations in EMD encoding emerin and LMNA encoding A-type lamins, proteins of the nuclear envelope. In the past decade, there has been an extraordinary burst of research on the nuclear envelope. Discoveries resulting from this basic research have implications for better understanding the pathogenesis and developing treatments for EDMD. RECENT FINDINGS: Recent clinical research has confirmed that EDMD is one of several overlapping skeletal muscle phenotypes that can result from mutations in EMD and LMNA with dilated cardiomyopathy as a common feature. Basic research on the nuclear envelope has provided new insights into how A-type lamins and emerin function in force transmission throughout the cell, which may be particularly important in striated muscle. Much of the recent research has focused on the heart and LMNA mutations. Prevalence and outcome studies have confirmed the relative severity of cardiac disease. Robust mouse models of EDMD caused by LMNA mutations has allowed for further insight into pathogenic mechanisms and potentially beneficial therapeutic approaches. SUMMARY: Recent clinical and basic research on EDMD is gradually being translated to clinical practice and possibly novel therapies.


Subject(s)
Lamin Type A/metabolism , Membrane Proteins/metabolism , Muscular Dystrophy, Emery-Dreifuss/genetics , Mutation , Nuclear Envelope/metabolism , Nuclear Proteins/metabolism , Animals , Humans , Lamin Type A/genetics , Membrane Proteins/genetics , Mice , Muscle, Skeletal/metabolism , Muscular Dystrophy, Emery-Dreifuss/metabolism , Nuclear Proteins/genetics , Phenotype
13.
Hum Mol Genet ; 25(11): 2220-2233, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27131347

ABSTRACT

Cardiomyopathy caused by lamin A/C gene mutations (LMNA cardiomyopathy) is characterized by increased myocardial fibrosis, which impairs left ventricular relaxation and predisposes to heart failure, and cardiac conduction abnormalities. While we previously discovered abnormally elevated extracellular signal-regulated kinase 1/2 (ERK1/2) activities in heart in LMNA cardiomyopathy, its role on the development of myocardial fibrosis remains unclear. We now showed that transforming growth factor (TGF)-ß/Smad signaling participates in the activation of ERK1/2 signaling in LMNA cardiomyopathy. ERK1/2 acts on connective tissue growth factor (CTGF/CCN2) expression to mediate the myocardial fibrosis and left ventricular dysfunction. Studies in vivo demonstrate that inhibiting CTGF/CCN2 using a specific antibody decreases myocardial fibrosis and improves the left ventricular dysfunction. Together, these findings show that cardiac ERK1/2 activity is modulated in part by TGF-ß/Smad signaling, leading to altered activation of CTGF/CCN2 to mediate fibrosis and alter cardiac function. This identifies a novel mechanism in the development of LMNA cardiomyopathy.


Subject(s)
Cardiomyopathies/genetics , Connective Tissue Growth Factor/genetics , Fibrosis/genetics , Lamin Type A/genetics , Transforming Growth Factor beta/genetics , Animals , Cardiomyopathies/pathology , Fibrosis/pathology , Humans , MAP Kinase Signaling System/genetics , Mice , Mice, Knockout , Myocardium/metabolism , Myocardium/pathology , Smad Proteins/genetics , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/pathology
14.
Am J Physiol Heart Circ Physiol ; 314(6): H1192-H1202, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29451818

ABSTRACT

Mutations in genes encoding components of the sarcomere cause cardiomyopathy, which is often associated with abnormal Ca2+ sensitivity of muscle contraction. We have previously shown that a heart-specific myosin light chain phosphatase small subunit (hHS-M21) increases the Ca2+ sensitivity of muscle contraction. The aim of the present study was to investigate the function of hHS-M21 in vivo and the causative role of abnormal Ca2+ sensitivity in cardiomyopathy. We generated transgenic mice with cardiac-specific overexpression of hHS-M21. We confirmed that hHS-M21 increased the Ca2+ sensitivity of cardiac muscle contraction in vivo, which was not followed by an increased phosphorylation of myosin light chain 2 isoforms. hHS-M21 transgenic mice developed severe systolic dysfunction with myocardial fibrosis and degeneration of cardiomyocytes in association with sinus bradycardia and atrioventricular conduction defect. The contractile dysfunction and cardiac fibrosis were improved by treatment with the Rho kinase inhibitor fasudil. Our findings suggested that the overexpression of hHS-M21 results in cardiac dysfunction and conduction disturbance via non-myosin light chain 2 phosphorylation-dependent regulation. NEW & NOTEWORTHY The present study is the first to develop mice with transgenic overexpression of a heart-specific myosin light chain phosphatase small subunit (hHS-M21) and to examine the effects of hHS-M21 on cardiac function. Elevation of hHS-M21 induced heart failure with myocardial fibrosis and degeneration of cardiomyocytes accompanied by supraventricular arrhythmias.


Subject(s)
Arrhythmias, Cardiac/enzymology , Heart Failure/enzymology , Myocytes, Cardiac/enzymology , Myosin-Light-Chain Phosphatase/metabolism , Animals , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/pathology , Arrhythmias, Cardiac/physiopathology , Calcium Signaling , Cardiac Myosins/metabolism , Cardiomyopathies/enzymology , Cardiomyopathies/genetics , Cardiomyopathies/physiopathology , Disease Models, Animal , Fibrosis , Genetic Predisposition to Disease , Heart Failure/genetics , Heart Failure/pathology , Heart Failure/physiopathology , Heart Rate , Humans , Mice, Inbred C57BL , Mice, Transgenic , Myocardial Contraction , Myocytes, Cardiac/pathology , Myosin Light Chains/metabolism , Myosin-Light-Chain Phosphatase/genetics , Phenotype , Phosphorylation , Protein Subunits , Up-Regulation , Ventricular Dysfunction, Left/enzymology , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/physiopathology , Ventricular Function, Left , Ventricular Remodeling , rho-Associated Kinases/metabolism
15.
Bioorg Med Chem ; 25(3): 1004-1013, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28011205

ABSTRACT

Signaling mediated by extracellular signal-regulated kinases 1 and 2 (ERK1/2) is involved in numerous cellular processes. Mitogen-activated protein kinase kinases (MEK1/2) catalyze the phosphorylation of ERK1/2, converting it into an active kinase that regulates the expression of numerous genes and cellular processes. Inhibitors of MEK1/2 have demonstrated preclinical and clinical efficacy in certain cancers and types of cardiomyopathy. We report the synthesis of a novel, allosteric, macrocyclic MEK1/2 inhibitor that potently inhibits ERK1/2 activity in cultured cells and tissues of mice after systemic administration. Mice with dilated cardiomyopathy caused by a lamin A/C gene mutation have abnormally increased cardiac ERK1/2 activity. In these mice, this novel MEK1/2 inhibitor is well tolerated, improves left ventricular systolic function, decreases left ventricular fibrosis, has beneficial effects on skeletal muscle structure and pathology and prolongs survival. The novel MEK1/2 inhibitor described herein may therefore find clinical utility in the treatment of this rare cardiomyopathy, other types of cardiomyopathy and cancers in humans.


Subject(s)
Cardiomyopathy, Dilated/drug therapy , Disease Models, Animal , Lamin Type A/genetics , Macrocyclic Compounds/pharmacology , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Animals , Cardiomyopathy, Dilated/genetics , Dose-Response Relationship, Drug , Macrocyclic Compounds/administration & dosage , Macrocyclic Compounds/chemistry , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 1/metabolism , Molecular Structure , Mutation , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship
16.
Semin Cell Dev Biol ; 29: 107-15, 2014 May.
Article in English | MEDLINE | ID: mdl-24440603

ABSTRACT

Lamins A and C, encoded by LMNA, are constituent of the nuclear lamina, a meshwork of proteins underneath the nuclear envelope first described as scaffolding proteins of the nucleus. Since the discovery of LMNA mutations in highly heterogeneous human disorders (including cardiac and muscular dystrophies, lipodystrophies and progeria), the number of functions described for lamin A/C has expanded. Lamin A/C is notably involved in the regulation of chromatin structure and gene transcription, and in the resistance of cells to mechanical stress. This review focuses on studies performed on knock-out and knock-in Lmna mouse models, which have led to decipher some of the lamin A/C functions in striated muscles and to the first preclinical trials of pharmaceutical therapies.


Subject(s)
Cardiomyopathies/genetics , Lamin Type A/genetics , Muscle, Striated/pathology , Muscular Dystrophies/genetics , Nuclear Lamina/genetics , Actin Cytoskeleton/pathology , Animals , Cardiomyopathies/drug therapy , Cardiomyopathies/pathology , Desmin/genetics , Disease Models, Animal , Gene Expression Regulation/genetics , Gene Knock-In Techniques , Humans , Mice , Mice, Knockout , Muscle, Striated/cytology , Nuclear Envelope , Vimentin/genetics
17.
Hum Mol Genet ; 23(1): 1-11, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-23933734

ABSTRACT

Mutations in the lamin A/C gene (LMNA) encoding A-type nuclear lamins cause dilated cardiomyopathy with variable muscular dystrophy. These mutations enhance mitogen-activated protein kinase signaling in the heart and pharmacological inhibition of extracellular signal-regulated kinase (ERK) 1 and 2 improves cardiac function in Lmna(H222P/H222P) mice. In the current study, we crossed mice lacking ERK1 to Lmna(H222P/H222P) mice and examined cardiac performance and survival. Male Lmna(H222P/H222P)/Erk1(-/-) mice lacking ERK1 had smaller left ventricular end systolic diameters and increased fractional shortening (FS) at 16 weeks of age than Lmna(H222P/H222P/)Erk1(+/+) mice. Their mean survival was also significantly longer. However, the improved cardiac function was abrogated at 20 weeks of age concurrent with an increased activity of ERK2. Lmna(H222P/H222P)/Erk1(-/-) mice treated with an inhibitor of ERK1/2 activation had smaller left ventricular diameters and increased FS at 20 weeks of age. These results provide genetic evidence that ERK1 and ERK2 contribute to the development of cardiomyopathy caused by LMNA mutations and reveal interplay between these isoenzymes in maintaining a combined pathological activity in heart.


Subject(s)
Cardiomyopathies/genetics , Cardiomyopathies/physiopathology , Lamin Type A/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Animals , Benzimidazoles/pharmacology , Cardiomyopathies/pathology , Disease Models, Animal , Female , Gene Expression Regulation , Heart Ventricles/drug effects , Heart Ventricles/physiopathology , Humans , Lamin Type A/metabolism , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/genetics , Mutation
18.
Hum Mol Genet ; 21(19): 4325-33, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22773734

ABSTRACT

We previously interrogated the transcriptome in heart tissue from Lmna(H222P/H222P) mice, a mouse model of cardiomyopathy caused by lamin A/C gene (LMNA) mutation, and found that the extracellular signal-regulated kinase 1/2 and Jun N-terminal kinase branches of the mitogen-activated protein (MAP) kinase signaling pathway were abnormally hyperactivated prior to the onset of significant cardiac impairment. We have now used an alternative gene expression analysis tool to reanalyze this transcriptome and identify hyperactivation of a third branch of the MAP kinase cascade, p38α signaling. Biochemical analysis of hearts from Lmna(H222P/H222P) mice showed enhanced p38α activation prior to and after the onset of heart disease as well as in hearts from human subjects with cardiomyopathy caused by LMNA mutations. Treatment of Lmna(H222P/H222P) mice with the p38α inhibitor ARRY-371797 prevented left ventricular dilatation and deterioration of fractional shortening compared with placebo-treated mice but did not block the expression of collagen genes involved in cardiac fibrosis. These results demonstrate that three different branches of the MAP kinase signaling pathway with overlapping consequences are involved in the pathogenesis of cardiomyopathy caused by LMNA mutations. They further suggest that pharmacological inhibition of p38α may be useful in the treatment of this disease.


Subject(s)
Cardiomyopathy, Dilated/enzymology , Ethylenediamines/pharmacology , Indazoles/pharmacology , Lamin Type A/genetics , Mitogen-Activated Protein Kinase 14/metabolism , Mutation, Missense , Signal Transduction , Adolescent , Animals , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/metabolism , Cells, Cultured , Female , Humans , Lamin Type A/metabolism , Male , Mice , Mice, Transgenic , Middle Aged , Mitogen-Activated Protein Kinase 14/genetics , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/metabolism , Young Adult
19.
Biochem Biophys Res Commun ; 452(4): 958-61, 2014 Oct 03.
Article in English | MEDLINE | ID: mdl-25218145

ABSTRACT

BACKGROUND: Mutations in the LMNA gene encoding A-type nuclear lamins can cause dilated cardiomyopathy with or without skeletal muscular dystrophy. Previous studies have shown abnormally increased extracellular signal-regulated kinase 1/2 activity in hearts of Lmna(H222P/H222P) mice, a small animal model. Inhibition of this abnormal signaling activity with a mitogen-activated protein kinase kinase 1/2 (MEK1/2) inhibitor has beneficial effects on heart function and survival in these mice. However, such treatment has not been examined relative to any standard of care intervention for dilated cardiomyopathy or heart failure. We therefore examined the effects of an angiotensin II converting enzyme (ACE) inhibitor on left ventricular function in Lmna(H222P/H222P) mice and assessed if adding a MEK1/2 inhibitor would provide added benefit. METHODS: Male Lmna(H222P/H222P) mice were treated with the ACE inhibitor benazepril, the MEK1/2 inhibitor selumetinib or both. Transthoracic echocardiography was used to measure left ventricular diameters and fractional shortening was calculated. RESULTS: Treatment of Lmna(H222P/H222P) mice with either benazepril or selumetinib started at 8weeks of age, before the onset of detectable left ventricular dysfunction, lead to statistically significantly increased fractional shortening compared to placebo at 16weeks of age. There was a trend towards a great value for fractional shortening in the selumetinib-treated mice. When treatment was started at 16weeks of age, after the onset of left ventricular dysfunction, the addition of selumetinib treatment to benazepril lead to a statistically significant increase in left ventricular fractional shortening at 20weeks of age. CONCLUSIONS: Both ACE inhibition and MEK1/2 inhibition have beneficial effects on left ventricular function in Lmna(H222P/H222P) mice and both drugs together have a synergistic benefit when initiated after the onset of left ventricular dysfunction. These results provide further preclinical rationale for a clinical trial of a MEK1/2 inhibitor in addition to standard of care in patients with dilated cardiomyopathy caused by LMNA mutations.


Subject(s)
Angiotensin II/metabolism , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Cardiomyopathy, Dilated/drug therapy , Cardiomyopathy, Dilated/physiopathology , Lamin Type A/genetics , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Animals , Benzazepines , Benzimidazoles , Cardiomyopathy, Dilated/diagnosis , Cardiomyopathy, Dilated/pathology , Male , Mice , Mutation/genetics , Treatment Outcome
20.
J Biol Chem ; 287(48): 40513-24, 2012 Nov 23.
Article in English | MEDLINE | ID: mdl-23048029

ABSTRACT

BACKGROUND: Mutations in LMNA gene cause cardiomyopathy, for which mechanistic insights are lacking. RESULTS: Dusp4 expression is enhanced in hearts with LMNA cardiomyopathy, and its overexpression in mice causes it by activating AKT-mTOR signaling that impairs autophagy. CONCLUSIONS: Dusp4 causes cardiac dysfunction and may contribute to the development of LMNA cardiomyopathy. SIGNIFICANCE: Revealing pathogenic mechanisms of LMNA cardiomyopathy is essential for the development of mechanism-based therapies. Mutations in the lamin A/C gene (LMNA) cause a diverse spectrum of diseases, the most common of which is dilated cardiomyopathy often with skeletal muscular dystrophy. Lamin A and C are fundamental components of the nuclear lamina, a dynamic meshwork of intermediate filaments lining the nuclear envelope inner membrane. Prevailing evidence suggests that the nuclear envelope functions as a signaling node and that abnormality in the nuclear lamina leads to dysregulated signaling pathways that underlie disease pathogenesis. We previously showed that activated ERK1/2 in hearts of a mouse model of LMNA cardiomyopathy (Lmna(H222P/H222P) mice) contributes to disease, but the complete molecular pathogenesis remains poorly understood. Here we uncover a pathogenic role of dual specificity phosphatase 4 (Dusp4), which is transcriptionally induced by ERK1/2. Dusp4 is highly expressed in the hearts of Lmna(H222P/H222P) mice, and transgenic mice with cardiac-selective overexpression of Dusp4 display heart dysfunction similar to LMNA cardiomyopathy. In both primary tissue and cell culture models, overexpression of Dusp4 positively regulates AKT-mTOR signaling, resulting in impaired autophagy. These findings identify a pathogenic role of Dusp4 in LMNA cardiomyopathy.


Subject(s)
Cardiomyopathies/enzymology , Dual-Specificity Phosphatases/metabolism , Lamin Type A/genetics , Mitogen-Activated Protein Kinase Phosphatases/metabolism , Mutation, Missense , Animals , Autophagy , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Cardiomyopathies/physiopathology , Dual-Specificity Phosphatases/genetics , Female , Heart/physiopathology , Humans , Lamin Type A/metabolism , MAP Kinase Signaling System , Male , Mice , Mice, Inbred CBA , Mice, Transgenic , Mitogen-Activated Protein Kinase Phosphatases/genetics , Myocardium/enzymology , Myocardium/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL