Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Can J Physiol Pharmacol ; 101(12): 620-629, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37747059

ABSTRACT

The purpose of this study was to characterize the role of ß1-AR signaling and its cross-talk between cardiac renin-angiotensin system and thyroid-hormone-induced cardiac hypertrophy. T3 was administered at 0.5 mg·kg-1·day-1 for 10 days in ß1-KOT3 and WTT3 groups, while control groups received vehicle alone. Echocardiography and myocardial histology was performed; cardiac and serum ANGI/ANGII and ANP and cardiac levels of p-PKA, p-ERK1/2, p-p38-MAPK, p-AKT, p-4EBP1, and ACE were measured. WTT3 showed decreased IVSTd and increased LVEDD versus WTsal (p < 0.05). ß1-KOT3 exhibited lower LVEDD and higher relative IVSTd versus ß1-KOsal, the lowest levels of ejection fraction, and the highest levels of cardiomyocyte diameter (p < 0.05). Cardiac ANP levels decreased in WTT3 versus ß1-KOT3 (p < 0.05). Cardiac ACE expression was increased in T3-treated groups (p < 0.05). Phosphorylated-p38 MAPK levels were higher in WTT3 versus WTsal or ß1-KOT3, p-4EBP1 was elevated in ß1-KO animals, and p-ERK1/2 was up-regulated in ß1-KOT3. These findings suggest that ß1-AR signaling is crucial for TiCH.


Subject(s)
Cardiomyopathy, Restrictive , Mice , Animals , Cardiomyopathy, Restrictive/metabolism , Cardiomyopathy, Restrictive/pathology , Mice, Knockout , Myocardium/metabolism , Thyroid Hormones , Receptors, Adrenergic/metabolism , Angiotensin II/pharmacology
2.
ESC Heart Fail ; 9(6): 4330-4334, 2022 12.
Article in English | MEDLINE | ID: mdl-36056647

ABSTRACT

Herein, we describe histological mobilization of light chain cardiac amyloid documented by sequential left ventricular endomyocardial biopsies. These findings were associated with positive remodelling of cardiomyocytes and of restrictive cardiomyopathy resulting from 14 courses of chemotherapy over 17 years of time. Histological and ultrastructural findings of light chain cardiac amyloid removal led to increase in cardiomyocyte dimension and electrocardiogram voltages, reduction of biventricular wall thickness with improvement of left ventricular diastolic function, and NYHA class shifting from III to I.


Subject(s)
Cardiomyopathy, Restrictive , Humans , Cardiomyopathy, Restrictive/diagnosis , Cardiomyopathy, Restrictive/metabolism , Myocytes, Cardiac/metabolism , Myocardium/pathology , Amyloid/metabolism , Biopsy
3.
Eur J Pharmacol ; 918: 174791, 2022 Mar 05.
Article in English | MEDLINE | ID: mdl-35093323

ABSTRACT

It is well-established that cardiac fibrosis contributes to cardiac dysfunction and adverse outcomes. However, the underlying mechanisms remain elusive, warranting further studies to develop new therapeutic strategies. It has been suggested that loureirin B can ameliorate the progression of fibrotic diseases. This study investigated the effects of loureirin B on cardiac fibrosis and explored the underlying mechanisms. Transverse aortic constriction (TAC) was performed to induce cardiac fibrosis in mice. Loureirin B (10 mg/kg/day) or saline was continuously delivered via subcutaneous osmotic mini-pumps. Cardiac fibroblasts (CFs) were treated with angiotensin II (Ang II, 100 nM, 24 h) to simulate fibrosis in vitro. Immunochemistry, echocardiography, and Sircol collagen assays were conducted to evaluate the cardioprotective effects. Quantitative real-time polymerase chain reaction, Western blot, and transfection techniques were performed to elucidate the mechanisms. Results showed that loureirin B prevented cardiac fibrosis and improved cardiac function in mice subjected to TAC. Treatment with loureirin B inhibited the elevation of inflammatory factors (interleukin-1ß, interleukin-6, and tumor necrosis factor-α), transforming growth factor-ß1 (TGF-ß1), and Pin1 induced by TAC. Furthermore, loureirin B treatment inhibited the increased fibroblast activation and collagen synthesis induced by Ang II in CFs. In addition, loureirin B inhibited increased expression of TGF-ß1 and Pin1 induced by Ang II or TAC. Mechanistically, overexpression of Pin1 induced increased TGF-ß1 expression and blocked the anti-fibrotic effects in Ang II-induced CFs treated with loureirin B. Loureirin B ameliorated cardiac fibrosis and dysfunction both in vitro and in vivo probably through the Pin1/TGF-ß1 signaling pathway.


Subject(s)
Myocardium , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Resins, Plant/pharmacology , Transforming Growth Factor beta1/metabolism , Animals , Cardiomyopathy, Restrictive/drug therapy , Cardiomyopathy, Restrictive/metabolism , Cardiotonic Agents/pharmacology , Cell Proliferation/drug effects , Collagen/metabolism , Disease Models, Animal , Echocardiography/methods , Fibrosis , Immunochemistry , Mice , Myocardium/metabolism , Myocardium/pathology , Signal Transduction/drug effects
4.
Sci Rep ; 11(1): 12861, 2021 06 18.
Article in English | MEDLINE | ID: mdl-34145321

ABSTRACT

DCBLD2 encodes discodin, CUB and LCCL domain-containing protein 2, a type-I transmembrane receptor that is involved in intracellular receptor signalling pathways and the regulation of cell growth. In this report, we describe a 5-year-old female who presented severe clinical features, including restrictive cardiomyopathy, developmental delay, spasticity and dysmorphic features. Trio-whole-exome sequencing and segregation analysis were performed to identify the genetic cause of the disease within the family. A novel homozygous nonsense variant in the DCBLD2 gene (c.80G > A, p.W27*) was identified as the most likely cause of the patient's phenotype. This nonsense variant falls in the extracellular N-terminus of DCBLD2 and thus might affect proper protein function of the transmembrane receptor. A number of in vitro investigations were performed on the proband's skin fibroblasts compared to normal fibroblasts, which allowed a comprehensive assessment resulting in the functional characterization of the identified DCBLD2 nonsense variant in different cellular processes. Our data propose a significant association between the identified variant and the observed reduction in cell proliferation, cell cycle progression, intracellular ROS, and Ca2 + levels, which would likely explain the phenotypic presentation of the patient as associated with lethal restrictive cardiomyopathy.


Subject(s)
Abnormalities, Multiple/genetics , Cardiomyopathy, Restrictive/genetics , Codon, Nonsense , Developmental Disabilities/genetics , Genetic Predisposition to Disease , Homozygote , Membrane Proteins/genetics , Abnormalities, Multiple/diagnosis , Alleles , Calcium/metabolism , Cardiomyopathy, Restrictive/diagnosis , Cardiomyopathy, Restrictive/metabolism , Cell Cycle/genetics , Child, Preschool , Consanguinity , Developmental Disabilities/diagnosis , Developmental Disabilities/metabolism , Facies , Female , Genetic Association Studies/methods , Genome, Mitochondrial , Genomics/methods , Humans , Magnetic Resonance Angiography , Phenotype , Radiography, Thoracic , Reactive Oxygen Species/metabolism , Exome Sequencing
5.
Eur J Clin Invest ; 51(12): e13640, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34129696

ABSTRACT

OBJECTIVE: To investigate the potential association between neutrophil degranulation and patterns of myocardial dysfunction in a cohort of patients with type 2 diabetes mellitus (T2DM). BACKGROUND: Two distinct phenotypes of diabetic cardiomyopathy have been described: a restrictive phenotype with diastolic dysfunction (restrictive/DD) and a dilative phenotype with systolic dysfunction (dilative/SD). However, the underlying determinants of these two patterns are not yet recognized. METHODS: In this single-centre, observational, cross-sectional study, 492 patients were recruited. Ultrasonographic measurements were performed by two experienced sonographers, blinded to the clinical data of the participants. Serum biomarkers of neutrophil degranulation were measured by enzyme-linked immunosorbent sandwich assay (ELISA). RESULTS: After adjustment for confounders, resistin, myeloperoxidase, matrix metalloproteinase 8 and matrix metalloproteinase 9/tissue inhibitor of metalloproteinases 1 complex were positively associated with the restrictive/DD pattern compared with the normal pattern. Similarly, MPO was positively associated with the dilative/SD pattern compared with the normal pattern, and resistin was negatively associated with the dilative/SD pattern compared with the restrictive/DD pattern. CONCLUSIONS: Neutrophil degranulation is associated with the restrictive/DD echocardiographic pattern in patients with T2DM, but not with the normal pattern and dilative/SD patterns. Neutrophils could have a pivotal role in the pathogenesis of myocardial dysfunction, and particularly diastolic dysfunction, in patients with T2DM.


Subject(s)
Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Restrictive/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetic Cardiomyopathies/metabolism , Neutrophil Activation , Aged , Biomarkers/metabolism , Cardiomyopathy, Dilated/diagnostic imaging , Cardiomyopathy, Dilated/etiology , Cardiomyopathy, Dilated/physiopathology , Cardiomyopathy, Restrictive/diagnostic imaging , Cardiomyopathy, Restrictive/etiology , Cardiomyopathy, Restrictive/physiopathology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/physiopathology , Diabetic Cardiomyopathies/diagnostic imaging , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/physiopathology , Echocardiography , Female , Heart Failure, Diastolic/diagnostic imaging , Heart Failure, Diastolic/etiology , Heart Failure, Diastolic/metabolism , Heart Failure, Diastolic/physiopathology , Heart Failure, Systolic/diagnostic imaging , Heart Failure, Systolic/etiology , Heart Failure, Systolic/metabolism , Heart Failure, Systolic/physiopathology , Humans , Male , Matrix Metalloproteinase 8/metabolism , Matrix Metalloproteinase 9/metabolism , Middle Aged , Peroxidase/metabolism , Resistin/metabolism , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-2/metabolism
6.
Nat Commun ; 12(1): 3575, 2021 06 11.
Article in English | MEDLINE | ID: mdl-34117258

ABSTRACT

An amino acid exchange (P209L) in the HSPB8 binding site of the human co-chaperone BAG3 gives rise to severe childhood cardiomyopathy. To phenocopy the disease in mice and gain insight into its mechanisms, we generated humanized transgenic mouse models. Expression of human BAG3P209L-eGFP in mice caused Z-disc disintegration and formation of protein aggregates. This was accompanied by massive fibrosis resulting in early-onset restrictive cardiomyopathy with increased mortality as observed in patients. RNA-Seq and proteomics revealed changes in the protein quality control system and increased autophagy in hearts from hBAG3P209L-eGFP mice. The mutation renders hBAG3P209L less soluble in vivo and induces protein aggregation, but does not abrogate hBAG3 binding properties. In conclusion, we report a mouse model mimicking the human disease. Our data suggest that the disease mechanism is due to accumulation of hBAG3P209L and mouse Bag3, causing sequestering of components of the protein quality control system and autophagy machinery leading to sarcomere disruption.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cardiomyopathy, Restrictive/genetics , Cardiomyopathy, Restrictive/metabolism , Animals , Autophagy , Binding Sites , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Cardiomyopathies/therapy , Cardiomyopathy, Restrictive/therapy , Child , Disease Models, Animal , Gene Expression Regulation , Genetic Therapy , Heart , Heat-Shock Proteins , Humans , Mice , Mice, Transgenic , Molecular Chaperones/metabolism , Mutation , Protein Binding , Proteomics , Sarcomeres/metabolism
7.
Med Mol Morphol ; 54(3): 289-295, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34057638

ABSTRACT

Restrictive cardiomyopathy (RCM) is a rare primary myocardial disease, and its pathological features are yet to be determined. Restrictive cardiomyopathy with MHY7 mutation was diagnosed in a 65-year-old Japanese woman. Electron microscopy of a myocardial biopsy revealed electron-dense materials resulting from focal myocyte degeneration and necrosis as well as tubular structures and pseudo-inclusion bodies in some nuclei. These features may be associated with the pathogenesis of RCM.


Subject(s)
Cardiac Myosins/genetics , Cardiomyopathy, Restrictive/pathology , Muscle Cells/pathology , Mutation, Missense , Myosin Heavy Chains/genetics , Aged , Biopsy , Cardiomyopathy, Restrictive/genetics , Cardiomyopathy, Restrictive/metabolism , Female , Humans , Muscle Cells/ultrastructure , Pedigree
8.
PLoS One ; 15(3): e0229227, 2020.
Article in English | MEDLINE | ID: mdl-32182250

ABSTRACT

TNNI3 encoding cTnI, the inhibitory subunit of the troponin complex, is the main target for mutations leading to restrictive cardiomyopathy (RCM). Here we investigate two cTnI-R170G/W amino acid replacements, identified in infantile RCM patients, which are located in the regulatory C-terminus of cTnI. The C-terminus is thought to modulate the function of the inhibitory region of cTnI. Both cTnI-R170G/W strongly enhanced the Ca2+-sensitivity of skinned fibres, as is typical for RCM-mutations. Both mutants strongly enhanced the affinity of troponin (cTn) to tropomyosin compared to wildtype cTn, whereas binding to actin was either strengthened (R170G) or weakened (R170W). Furthermore, the stability of reconstituted thin filaments was reduced as revealed by electron microscopy. Filaments containing R170G/W appeared wavy and showed breaks. Decoration of filaments with myosin subfragment S1 was normal in the presence of R170W, but was irregular with R170G. Surprisingly, both mutants did not affect the Ca2+-dependent activation of reconstituted cardiac thin filaments. In the presence of the N-terminal fragment of cardiac myosin binding protein C (cMyBPC-C0C2) cooperativity of thin filament activation was increased only when the filaments contained wildtype cTn. No effect was observed in the presence of cTn containing R170G/W. cMyBPC-C0C2 significantly reduced binding of wildtype troponin to actin/tropomyosin, but not of both mutant cTn. Moreover, we found a direct troponin/cMyBPC-C0C2 interaction using microscale thermophoresis and identified cTnI and cTnT, but not cTnC as binding partners for cMyBPC-C0C2. Only cTn containing cTnI-R170G showed a reduced affinity towards cMyBPC-C0C2. Our results suggest that the RCM cTnI variants R170G/W impair the communication between thin and thick filament proteins and destabilize thin filaments.


Subject(s)
Amino Acid Substitution , Cardiomyopathy, Restrictive/genetics , Myocardium/metabolism , Sarcomeres/metabolism , Troponin I/genetics , Actins/metabolism , Animals , Calcium/metabolism , Cardiomyopathy, Restrictive/metabolism , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Child, Preschool , Guinea Pigs , Humans , Microscopy, Electron , Models, Biological , Protein Binding , Tropomyosin/metabolism
9.
ESC Heart Fail ; 5(4): 724-726, 2018 08.
Article in English | MEDLINE | ID: mdl-29741282

ABSTRACT

Most individuals with cardiomyopathy associated with variants of the LMNA (lamin A) gene present with cardiac conduction abnormalities followed by dilated cardiomyopathy and cardiac failure; some also have skeletal muscle weakness. In this report, an individual with restrictive cardiomyopathy presenting with conduction defects followed by cardiac dysfunction of a restrictive nature eventually requiring cardiac transplantation is described. Subsequently, progressive skeletal muscle weakness became evident. The finding of a new LMNA pathologic gene variant in this patient increases the options for genetic testing of individuals with restrictive cardiomyopathy.


Subject(s)
Cardiomyopathy, Restrictive/genetics , DNA/genetics , Lamin Type A/genetics , Mutation, Missense , Myocardium/pathology , Cardiomyopathy, Restrictive/diagnosis , Cardiomyopathy, Restrictive/metabolism , DNA Mutational Analysis , Humans , Lamin Type A/metabolism , Male , Middle Aged , Myocardium/metabolism , Pedigree , Phenotype
10.
Sci Rep ; 7(1): 16809, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29196671

ABSTRACT

Light chain amyloidosis (AL), the most common systemic amyloidosis, is caused by the overproduction and the aggregation of monoclonal immunoglobulin light chains (LC) in target organs. Due to genetic rearrangement and somatic hypermutation, virtually, each AL patient presents a different amyloidogenic LC. Because of such complexity, the fine molecular determinants of LC aggregation propensity and proteotoxicity are, to date, unclear; significantly, their decoding requires investigating large sets of cases. Aiming to achieve generalizable observations, we systematically characterised a pool of thirteen sequence-diverse full length LCs. Eight amyloidogenic LCs were selected as responsible for severe cardiac symptoms in patients; five non-amyloidogenic LCs were isolated from patients affected by multiple myeloma. Our comprehensive approach (consisting of spectroscopic techniques, limited proteolysis, and X-ray crystallography) shows that low fold stability and high protein dynamics correlate with amyloidogenic LCs, while hydrophobicity, structural rearrangements and nature of the LC dimeric association interface (as observed in seven crystal structures here presented) do not appear to play a significant role in defining amyloid propensity. Based on the structural and biophysical data, our results highlight shared properties driving LC amyloid propensity, and these data will be instrumental for the design of synthetic inhibitors of LC aggregation.


Subject(s)
Cardiomyopathy, Restrictive/metabolism , Immunoglobulin Light Chains/chemistry , Immunoglobulin Light Chains/metabolism , Immunoglobulin Light-chain Amyloidosis/metabolism , Multiple Myeloma/metabolism , Aged , Cardiomyopathy, Restrictive/genetics , Cardiomyopathy, Restrictive/urine , Crystallography, X-Ray , Female , Humans , Hydrophobic and Hydrophilic Interactions , Immunoglobulin Light Chains/urine , Immunoglobulin Light-chain Amyloidosis/genetics , Immunoglobulin Light-chain Amyloidosis/urine , Male , Middle Aged , Multiple Myeloma/genetics , Multiple Myeloma/urine , Protein Folding , Protein Stability , Proteolysis
11.
Cardiovasc Res ; 113(10): 1124-1136, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28371863

ABSTRACT

AIMS: The E143K (Glu → Lys) mutation in the myosin essential light chain has been associated with restrictive cardiomyopathy (RCM) in humans, but the mechanisms that underlie the development of defective cardiac function are unknown. Using transgenic E143K-RCM mice, we sought to determine the molecular and cellular triggers of E143K-induced heart remodelling. METHODS AND RESULTS: The E143K-induced abnormalities in cardiac function and morphology observed by echocardiography and invasive haemodynamics were paralleled by augmented active and passive tension measured in skinned papillary muscle fibres compared with wild-type (WT)-generated force. In vitro, E143K-myosin had increased duty ratio and binding affinity to actin compared with WT-myosin, increased actin-activated ATPase activity and slower rates of ATP-dependent dissociation of the acto-myosin complex, indicating an E143K-induced myosin hypercontractility. E143K was also observed to reduce the level of myosin regulatory light chain phosphorylation while that of troponin-I remained unchanged. Small-angle X-ray diffraction data showed a decrease in the filament lattice spacing (d1,0) with no changes in the equatorial reflections intensity ratios (I1,1/I1,0) in E143K vs. WT skinned papillary muscles. The hearts of mutant-mice demonstrated ultrastructural defects and fibrosis that progressively worsened in senescent animals and these changes were hypothesized to contribute to diastolic disturbance and to mild systolic dysfunction. Gene expression profiles of E143K-hearts supported the histopathology results and showed an upregulation of stress-response and collagen genes. Finally, proteomic analysis evidenced RCM-dependent metabolic adaptations and higher energy demands in E143K vs. WT hearts. CONCLUSIONS: As a result of the E143K-induced myosin hypercontractility, the hearts of RCM mice model exhibited cardiac dysfunction, stiff ventricles and physiological, morphologic, and metabolic remodelling consistent with the development of RCM. Future efforts should be directed toward normalization of myosin motor function and the use of myosin-specific therapeutics to avert the hypercontractile state of E143K-myosin and prevent pathological cardiac remodelling.


Subject(s)
Cardiomyopathy, Restrictive/genetics , Mutation , Myocardial Contraction/genetics , Myocytes, Cardiac/pathology , Myosin Light Chains/genetics , Sarcomeres/pathology , Ventricular Function, Left/genetics , Ventricular Myosins/genetics , Ventricular Remodeling/genetics , Actins/metabolism , Adenosine Triphosphate/metabolism , Animals , Cardiomyopathy, Restrictive/metabolism , Cardiomyopathy, Restrictive/pathology , Cardiomyopathy, Restrictive/physiopathology , Collagen/metabolism , Disease Models, Animal , Energy Metabolism , Female , Fibrosis , Genetic Predisposition to Disease , Humans , Male , Mice, Transgenic , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/ultrastructure , Myosin Light Chains/metabolism , Phenotype , Phosphorylation , Sarcomeres/metabolism , Sarcomeres/ultrastructure , Ventricular Myosins/metabolism
12.
J Biol Chem ; 291(41): 21817-21828, 2016 Oct 07.
Article in English | MEDLINE | ID: mdl-27557662

ABSTRACT

The cardiac troponin I (cTnI) R145W mutation is associated with restrictive cardiomyopathy (RCM). Recent evidence suggests that this mutation induces perturbed myofilament length-dependent activation (LDA) under conditions of maximal protein kinase A (PKA) stimulation. Some cardiac disease-causing mutations, however, have been associated with a blunted response to PKA-mediated phosphorylation; whether this includes LDA is unknown. Endogenous troponin was exchanged in isolated skinned human myocardium for recombinant troponin containing either cTnI R145W, PKA/PKC phosphomimetic charge mutations (S23D/S24D and T143E), or various combinations thereof. Myofilament Ca2+ sensitivity of force, tension cost, LDA, and single myofibril activation/relaxation parameters were measured. Our results show that both R145W and T143E uncouple the impact of S23D/S24D phosphomimetic on myofilament function, including LDA. Molecular dynamics simulations revealed a marked reduction in interactions between helix C of cTnC (residues 56, 59, and 63), and cTnI (residue 145) in the presence of either cTnI RCM mutation or cTnI PKC phosphomimetic. These results suggest that the RCM-associated cTnI R145W mutation induces a permanent structural state that is similar to, but more extensive than, that induced by PKC-mediated phosphorylation of cTnI Thr-143. We suggest that this structural conformational change induces an increase in myofilament Ca2+ sensitivity and, moreover, uncoupling from the impact of phosphorylation of cTnI mediated by PKA at the Ser-23/Ser-24 target sites. The R145W RCM mutation by itself, however, does not impact LDA. These perturbed biophysical and biochemical myofilament properties are likely to significantly contribute to the diastolic cardiac pump dysfunction that is seen in patients suffering from a restrictive cardiomyopathy that is associated with the cTnI R145W mutation.


Subject(s)
Cardiomyopathy, Restrictive , Molecular Dynamics Simulation , Mutation, Missense , Sarcomeres , Troponin I , Amino Acid Substitution , Cardiomyopathy, Restrictive/genetics , Cardiomyopathy, Restrictive/metabolism , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Humans , Male , Protein Kinase C/chemistry , Protein Kinase C/genetics , Protein Kinase C/metabolism , Sarcomeres/chemistry , Sarcomeres/genetics , Sarcomeres/metabolism , Structure-Activity Relationship , Troponin I/chemistry , Troponin I/genetics , Troponin I/metabolism
13.
Arch Biochem Biophys ; 573: 69-76, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25813360

ABSTRACT

Diastolic dysfunction refers to an impaired relaxation and an abnormality in ventricular blood filling during diastole while systolic function is preserved. Cardiac myofibril hypersensitivity to Ca(2+) is a major factor that causes impaired relaxation of myocardial cells. The present study investigates the effect of the green tea extract catechins on myofibril calcium desensitization and restoration of diastolic function in a restrictive cardiomyopathy (RCM) mouse model with cardiac troponin mutations. Wild type (WT) and RCM mice were treated daily with catechin (epigallocatechin-3-gallate, EGCg, 50 mg/kg body weight) for 3 months. Echocardiography and cell based assays were performed to measure cardiac structure and flow-related variables including chamber dimensions, fraction shortening, trans-mitral flow patterns in the experimental mice. In addition, myocyte contractility and calcium dynamics were measured in WT and RCM cardiomyocytes treated in vitro with 5 µM EGCg. Our data indicated that RCM mice treated with EGCg showed an improved diastolic function while systolic function remained unchanged. At the cellular level, sarcomere relaxation and calcium decay were accelerated in RCM myocardial cells treated with EGCg. These results suggest that catechin is effective in reversing the impaired relaxation in RCM myocardial cells and rescuing the RCM mice with diastolic dysfunction.


Subject(s)
Calcium/metabolism , Cardiomyopathy, Restrictive/metabolism , Catechin/analogs & derivatives , Diastole/drug effects , Animals , Cardiomyopathy, Restrictive/pathology , Cardiomyopathy, Restrictive/physiopathology , Catechin/pharmacology , Cell Size/drug effects , Electrocardiography , Mice, Transgenic , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Sarcomeres/drug effects , Sarcomeres/physiology , Troponin I/genetics
15.
Arch Biochem Biophys ; 552-553: 3-10, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24326031

ABSTRACT

The C-terminal end-segment of Troponin I (TnI) corresponding to the last 27-33 amino acids is the most conserved structure of TnI and interacts with tropomyosin in a Ca(2+)-regulated manner, suggesting a role in muscle relaxation. Mutations in the C-terminal end-segment of cardiac TnI cause restrictive cardiomyopathy. Here we demonstrate that mouse cardiac TnI containing R193H or R205H mutation have significantly conformational changes in the region interfacing with troponin T (TnT) and increased binding affinity for TnT. These restrictive cardiomyopathy mutations also exhibit increased binding affinity for troponin C at pCa 4. The effects of R193H mutation were more profound than that of R205H. Tertiary troponin complex was reconstituted using the TnI mutants and a mini TnT lacking tropomyosin-binding sites to examine the interaction between the C-terminal end-segment of TnI and tropomyosin. The results showed that, R193H, but not R205H, caused a moderate but statistically significant increase in the binding affinity for tropomyosin at pCa 9. Similar trend was observed at pCa 5.5 but not pCa 4. These results provide novel evidence for the function of the C-terminal end-segment of TnI, where mutations with conformational effects alter TnI's interaction with other troponin subunits and tropomyosin to cause diastolic dysfunction.


Subject(s)
Cardiomyopathy, Restrictive/genetics , Point Mutation , Troponin C/metabolism , Troponin I/genetics , Troponin I/metabolism , Troponin T/metabolism , Animals , Calcium/metabolism , Cardiomyopathy, Restrictive/metabolism , Mice , Protein Binding , Protein Conformation , Troponin I/chemistry
16.
J Mol Cell Cardiol ; 53(3): 446-57, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22683325

ABSTRACT

Restrictive cardiomyopathy (RCM) has been linked to mutations in the thin filament regulatory protein cardiac troponin I (cTnI). As the pathogenesis of RCM from genotype to clinical phenotype is not fully understood, transgenic (Tg) mice were generated with cardiac specific expression of an RCM-linked missense mutation (R193H) in cTnI. R193H Tg mouse hearts with 15% stoichiometric replacement had smaller hearts and significantly elevated end diastolic pressures (EDP) in vivo. Using a unique carbon microfiber-based assay, membrane intact R193H adult cardiac myocytes generated higher passive tensions across a range of physiologic sarcomere lengths resulting in significant Ca(2+) independent cellular diastolic tone that was manifest in vivo as elevated organ-level EDP. Sarcomere relaxation and Ca(2+) decay was uncoupled in isolated R193H Tg adult myocytes due to the increase in myofilament Ca(2+) sensitivity of tension, decreased passive compliance of the sarcomere, and adaptive in vivo changes in which phospholamban (PLN) content was decreased. Further evidence of Ca(2+) and mechanical uncoupling in R193H Tg myocytes was demonstrated by the biphasic response of relaxation to increased pacing frequency versus the negative staircase seen with Ca(2+) decay. In comparison, non-transgenic myocyte relaxation closely paralleled the accelerated Ca(2+) decay. Ca(2+) transient amplitude was also significantly blunted in R193H Tg myocytes despite normal mechanical shortening resulting in myocyte hypercontractility when compared to non-transgenics. These results identify for the first time that a single point mutation in cTnI, R193H, directly causes elevated EDP due to a myocyte intrinsic loss of compliance independent of Ca(2+) cycling or altered cardiac morphology. The compound influence of impaired relaxation and elevated EDP represents a clinically severe form of diastolic dysfunction similar to the hemodynamic state documented in RCM patients.


Subject(s)
Cardiomyopathy, Restrictive/metabolism , Cardiomyopathy, Restrictive/physiopathology , Excitation Contraction Coupling , Myocardial Contraction , Troponin I/metabolism , Animals , Calcium/metabolism , Cardiomyopathy, Restrictive/genetics , Disease Models, Animal , Mice , Mice, Transgenic , Mutation , Myocardial Contraction/genetics , Myocytes, Cardiac/metabolism , Sarcomeres/metabolism , Troponin I/genetics
17.
Mol Endocrinol ; 26(5): 809-18, 2012 May.
Article in English | MEDLINE | ID: mdl-22403173

ABSTRACT

Cardiac injury induces myocardial expression of the thyroid hormone inactivating type 3 deiodinase (D3), which in turn dampens local thyroid hormone signaling. Here, we show that the D3 gene (Dio3) is a tissue-specific imprinted gene in the heart, and thus, heterozygous D3 knockout (HtzD3KO) mice constitute a model of cardiac D3 inactivation in an otherwise systemically euthyroid animal. HtzD3KO newborns have normal hearts but later develop restrictive cardiomyopathy due to cardiac-specific increase in thyroid hormone signaling, including myocardial fibrosis, impaired myocardial contractility, and diastolic dysfunction. In wild-type littermates, treatment with isoproterenol-induced myocardial D3 activity and an increase in the left ventricular volumes, typical of cardiac remodeling and dilatation. Remarkably, isoproterenol-treated HtzD3KO mice experienced a further decrease in left ventricular volumes with worsening of the diastolic dysfunction and the restrictive cardiomyopathy, resulting in congestive heart failure and increased mortality. These findings reveal crucial roles for Dio3 in heart function and remodeling, which may have pathophysiologic implications for human restrictive cardiomyopathy.


Subject(s)
Cardiomyopathy, Restrictive/metabolism , Iodide Peroxidase/metabolism , Myocardium/enzymology , Animals , Animals, Newborn , Cardiomyopathy, Restrictive/pathology , Cardiomyopathy, Restrictive/physiopathology , Cardiotonic Agents/administration & dosage , Cardiotonic Agents/pharmacology , Dose-Response Relationship, Drug , Gene Expression Profiling , Gene Expression Regulation/drug effects , Heart/drug effects , Heart/growth & development , Heart/physiopathology , Heart Failure/etiology , Infusions, Intravenous , Iodide Peroxidase/genetics , Isoproterenol/administration & dosage , Isoproterenol/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Proteins/genetics , Muscle Proteins/metabolism , Myocardium/metabolism , Myocardium/pathology , RNA, Messenger/metabolism , Ventricular Remodeling
18.
Toxicol Mech Methods ; 22(5): 330-5, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22394340

ABSTRACT

In recent years, diabetes and its associated complications have come to represent a major public health concern. It is a complex disease characterized by multiple metabolic derangements and is known to impair cardiac function by disrupting the balance between pro-oxidants and antioxidants at the cellular level. The subsequent generation of reactive oxygen species (ROS) and accompanying oxidative stress are hallmarks of the molecular mechanisms responsible for cardiovascular disease. Among several oxidative stress-mediated mechanisms that have been proposed, ROS-mediated oxidative stress has received the most attention. ROS have been shown to interact with proteins, lipids, and DNA, causing damage to the cellular macromolecules and subsequently, deterioration of cellular function. Induction of thioredoxin-1 (Trx1) gene expression has been demonstrated to protect the diabetic myocardium from dysfunction by reducing oxidative stress and enhancing the expression of heme oxygenase-1 (HO-1) and vascular endothelial growth factor (VEGF). The failure of antioxidants to consistently demonstrate clinical benefit necessitates further investigation of the role of oxidative stress in diabetes-mediated cardiovascular disease.


Subject(s)
Cardiomyopathy, Dilated/etiology , Cardiomyopathy, Hypertrophic/etiology , Cardiomyopathy, Restrictive/etiology , Diabetic Cardiomyopathies/etiology , Oxidative Stress , Animals , Antioxidants/administration & dosage , Antioxidants/therapeutic use , Cardiomyopathy, Dilated/drug therapy , Cardiomyopathy, Dilated/immunology , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Hypertrophic/drug therapy , Cardiomyopathy, Hypertrophic/immunology , Cardiomyopathy, Hypertrophic/metabolism , Cardiomyopathy, Restrictive/drug therapy , Cardiomyopathy, Restrictive/immunology , Cardiomyopathy, Restrictive/metabolism , Diabetic Cardiomyopathies/drug therapy , Diabetic Cardiomyopathies/immunology , Diabetic Cardiomyopathies/metabolism , Heme Oxygenase-1/biosynthesis , Heme Oxygenase-1/genetics , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Thioredoxins/genetics , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics
20.
J Biol Chem ; 286(32): 28435-43, 2011 Aug 12.
Article in English | MEDLINE | ID: mdl-21680742

ABSTRACT

Two Drosophila myosin II point mutations (D45 and Mhc(5)) generate Drosophila cardiac phenotypes that are similar to dilated or restrictive human cardiomyopathies. Our homology models suggest that the mutations (A261T in D45, G200D in Mhc(5)) could stabilize (D45) or destabilize (Mhc(5)) loop 1 of myosin, a region known to influence ADP release. To gain insight into the molecular mechanism that causes the cardiomyopathic phenotypes to develop, we determined whether the kinetic properties of the mutant molecules have been altered. We used myosin subfragment 1 (S1) carrying either of the two mutations (S1(A261T) and S1(G200D)) from the indirect flight muscles of Drosophila. The kinetic data show that the two point mutations have an opposite effect on the enzymatic activity of S1. S1(A261T) is less active (reduced ATPase, higher ADP affinity for S1 and actomyosin subfragment 1 (actin · S1), and reduced ATP-induced dissociation of actin · S1), whereas S1(G200D) shows increased enzymatic activity (enhanced ATPase, reduced ADP affinity for both S1 and actin · S1). The opposite changes in the myosin properties are consistent with the induced cardiac phenotypes for S1(A261T) (dilated) and S1(G200D) (restrictive). Our results provide novel insights into the molecular mechanisms that cause different cardiomyopathy phenotypes for these mutants. In addition, we report that S1(A261T) weakens the affinity of S1 · ADP for actin, whereas S1(G200D) increases it. This may account for the suppression (A261T) or enhancement (G200D) of the skeletal muscle hypercontraction phenotype induced by the troponin I held-up(2) mutation in Drosophila.


Subject(s)
Actins , Adenosine Diphosphate/metabolism , Cardiomyopathy, Restrictive , Drosophila Proteins , Models, Cardiovascular , Myosin Type II , Point Mutation , Actins/genetics , Actins/metabolism , Actomyosin/genetics , Actomyosin/metabolism , Animals , Cardiomyopathy, Restrictive/genetics , Cardiomyopathy, Restrictive/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster , Humans , Muscles/metabolism , Myosin Type II/genetics , Myosin Type II/metabolism , Phenotype , Troponin I/genetics , Troponin I/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...