Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters











Publication year range
1.
NPJ Regen Med ; 9(1): 21, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39285234

ABSTRACT

Neonatal dilated cardiomyopathy (DCM) is a poorly understood muscular disease of the heart. Several homozygous biallelic variants in LMOD2, the gene encoding the actin-binding protein Leiomodin 2, have been identified to result in severe DCM. Collectively, LMOD2-related cardiomyopathies present with cardiac dilation and decreased heart contractility, often resulting in neonatal death. Thus, it is evident that Lmod2 is essential to normal human cardiac muscle function. This study aimed to understand the underlying pathophysiology and signaling pathways related to the first reported LMOD2 variant (c.1193 G > A, p.Trp398*). Using patient-specific human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) and a mouse model harboring the homologous mutation to the patient, we discovered dysregulated actin-thin filament lengths, altered contractility and calcium handling properties, as well as alterations in the serum response factor (SRF)-dependent signaling pathway. These findings reveal that LMOD2 may be regulating SRF activity in an actin-dependent manner and provide a potential new strategy for the development of biologically active molecules to target LMOD2-related cardiomyopathies.

2.
Sci Adv ; 10(11): eadk1890, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38478604

ABSTRACT

Muscle contraction is a regulated process driven by the sliding of actin-thin filaments over myosin-thick filaments. Lmod2 is an actin filament length regulator and essential for life since human mutations and complete loss of Lmod2 in mice lead to dilated cardiomyopathy and death. To study the little-known role of Lmod2 in skeletal muscle, we created a mouse model with Lmod2 expressed exclusively in the heart but absent in skeletal muscle. Loss of Lmod2 in skeletal muscle results in decreased force production in fast- and slow-twitch muscles. Soleus muscle from rescued Lmod2 knockout mice have shorter thin filaments, increased Lmod3 levels, and present with a myosin fiber type switch from fast myosin heavy chain (MHC) IIA to the slower MHC I isoform. Since Lmod2 regulates thin-filament length in slow-twitch but not fast-twitch skeletal muscle and force deficits were observed in both muscle types, this work demonstrates that Lmod2 regulates skeletal muscle contraction, independent of its role in thin-filament length regulation.


Subject(s)
Muscle Contraction , Sarcomeres , Animals , Humans , Mice , Cytoskeletal Proteins/genetics , Heart , Mice, Knockout , Muscle Contraction/physiology , Muscle, Skeletal/physiology , Myosins
3.
Hum Mol Genet ; 33(12): 1036-1054, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38493359

ABSTRACT

Nemaline myopathy (NM) is a rare congenital neuromuscular disorder characterized by muscle weakness and hypotonia, slow gross motor development, and decreased respiratory function. Mutations in at least twelve genes, all of each encode proteins that are either components of the muscle thin filament or regulate its length and stability, have been associated with NM. Mutations in Nebulin (NEB), a giant filamentous protein localized in the sarcomere, account for more than 50% of NM cases. At present, there remains a lack of understanding of whether NEB genotype influences nebulin function and NM-patient phenotypes. In addition, there is a lack of therapeutically tractable models that can enable drug discovery and address the current unmet treatment needs of patients. To begin to address these gaps, here we have characterized five new zebrafish models of NEB-related NM. These mutants recapitulate most aspects of NEB-based NM, showing drastically reduced survival, defective muscle structure, reduced contraction force, shorter thin filaments, presence of electron-dense structures in myofibers, and thickening of the Z-disks. This study represents the first extensive investigation of an allelic series of nebulin mutants, and thus provides an initial examination in pre-clinical models of potential genotype-phenotype correlations in human NEB patients. It also represents the first utilization of a set of comprehensive outcome measures in zebrafish, including correlation between molecular analyses, structural and biophysical investigations, and phenotypic outcomes. Therefore, it provides a rich source of data for future studies exploring the NM pathomechanisms, and an ideal springboard for therapy identification and development for NEB-related NM.


Subject(s)
Alleles , Disease Models, Animal , Muscle Proteins , Muscle, Skeletal , Mutation , Myopathies, Nemaline , Phenotype , Sarcomeres , Zebrafish , Myopathies, Nemaline/genetics , Myopathies, Nemaline/pathology , Myopathies, Nemaline/physiopathology , Zebrafish/genetics , Animals , Muscle Proteins/genetics , Muscle Proteins/metabolism , Sarcomeres/genetics , Sarcomeres/metabolism , Sarcomeres/pathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Humans , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
4.
Biophys Rep (N Y) ; 4(1): 100147, 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38404534

ABSTRACT

In vitro motility (IVM) assays allow for the examination of the basic interaction between cytoskeletal filaments with molecular motors and the influence many physiological factors have on this interaction. Examples of factors that can be studied include changes in ADP and pH that emulate fatigue, altered phosphorylation that can occur with disease, and mutations within myofilament proteins that cause disease. While IVM assays can be analyzed manually, the main limitation is the ability to extract accurate data rapidly from videos collected without individual bias. While programs have been created in the past to enable data extraction, many are now out of date or require the use of proprietary software. Here, we report the generation of a Python-based tracking program, Philament, which automatically extracts data on instantaneous and average velocities, and allows for fully automated analysis of IVM recordings. The data generated are presented in an easily accessible spreadsheet-based, comma-separated values file. Philament also contains a novel method of quantifying the smoothness of filament motion. By fitting curves to standard deviations of velocity and average velocities, the influence of different experimental conditions can be compared relative to one another. This comparison provides a qualitative measure of protein interactions where steeper slopes indicate more unstable interactions and shallower slopes indicate more stable interactions within the myofilament. Overall, Philament's automation of IVM analysis provides easier entry into the field of cardiovascular mechanics and enables users to create a truly high-throughput experimental data analysis.

5.
J Mol Cell Cardiol ; 165: 103-114, 2022 04.
Article in English | MEDLINE | ID: mdl-35031281

ABSTRACT

Titin's C-zone is an inextensible segment in titin, comprised of 11 super-repeats and located in the cMyBP-C-containing region of the thick filament. Previously we showed that deletion of titin's super-repeats C1 and C2 (TtnΔC1-2 model) results in shorter thick filaments and contractile dysfunction of the left ventricular (LV) chamber but that unexpectedly LV diastolic stiffness is normal. Here we studied the contraction-relaxation kinetics from the time-varying elastance of the LV and intact cardiomyocyte, cellular work loops of intact cardiomyocytes, Ca2+ transients, cross-bridge kinetics, and myofilament Ca2+ sensitivity. Intact cardiomyocytes of TtnΔC1-2 mice exhibit systolic dysfunction and impaired relaxation. The time-varying elastance at both LV and single-cell levels showed that activation kinetics are normal in TtnΔC1-2 mice, but that relaxation is slower. The slowed relaxation is, in part, attributable to an increased myofilament Ca2+ sensitivity and slower early Ca2+ reuptake. Cross-bridge dynamics showed that cross-bridge kinetics are normal but that the number of force-generating cross-bridges is reduced. In vivo sarcomere length (SL) measurements revealed that in TtnΔC1-2 mice the operating SL range of the LV is shifted towards shorter lengths. This normalizes the apparent cell and LV diastolic stiffness but further reduces systolic force as systole occurs further down on the ascending limb of the force-SL relation. We propose that the reduced working SLs reflect titin's role in regulating diastolic stiffness by altering the number of sarcomeres in series. Overall, our study reveals that thick filament length regulation by titin's C-zone is critical for normal cardiac function.


Subject(s)
Myofibrils , Sarcomeres , Animals , Connectin/genetics , Mice , Muscle Contraction , Myocytes, Cardiac , Protein Kinases/genetics , Sarcomeres/physiology
6.
PLoS One ; 15(1): e0226138, 2020.
Article in English | MEDLINE | ID: mdl-31899774

ABSTRACT

A novel cardiac-specific transgenic mouse model was generated to identify the physiological consequences of elongated thin filaments during post-natal development in the heart. Remarkably, increasing the expression levels in vivo of just one sarcomeric protein, Lmod2, results in ~10% longer thin filaments (up to 26% longer in some individual sarcomeres) that produce up to 50% less contractile force. Increasing the levels of Lmod2 in vivo (Lmod2-TG) also allows us to probe the contribution of Lmod2 in the progression of cardiac myopathy because Lmod2-TG mice present with a unique cardiomyopathy involving enlarged atrial and ventricular lumens, increased heart mass, disorganized myofibrils and eventually, heart failure. Turning off of Lmod2 transgene expression at postnatal day 3 successfully prevents thin filament elongation, as well as gross morphological and functional disease progression. We show here that Lmod2 has an essential role in regulating cardiac contractile force and function.


Subject(s)
Actin Cytoskeleton/pathology , Cardiomyopathies/physiopathology , Cytoskeletal Proteins/physiology , Heart Failure/etiology , Muscle Proteins/physiology , Muscle, Skeletal/pathology , Sarcomeres/pathology , Animals , Animals, Newborn , Female , Heart Failure/pathology , Male , Mice , Mice, Transgenic , Muscle Contraction
7.
Sci Adv ; 5(9): eaax2066, 2019 09.
Article in English | MEDLINE | ID: mdl-31517052

ABSTRACT

Neonatal heart failure is a rare, poorly-understood presentation of familial dilated cardiomyopathy (DCM). Exome sequencing in a neonate with severe DCM revealed a homozygous nonsense variant in leiomodin 2 (LMOD2, p.Trp398*). Leiomodins (Lmods) are actin-binding proteins that regulate actin filament assembly. While disease-causing mutations in smooth (LMOD1) and skeletal (LMOD3) muscle isoforms have been described, the cardiac (LMOD2) isoform has not been previously associated with human disease. Like our patient, Lmod2-null mice have severe early-onset DCM and die before weaning. The infant's explanted heart showed extraordinarily short thin filaments with isolated cardiomyocytes displaying a large reduction in maximum calcium-activated force production. The lack of extracardiac symptoms in Lmod2-null mice, and remarkable morphological and functional similarities between the patient and mouse model informed the decision to pursue cardiac transplantation in the patient. To our knowledge, this is the first report of aberrant cardiac thin filament assembly associated with human cardiomyopathy.


Subject(s)
Actin Cytoskeleton , Cardiomyopathy, Dilated , Codon, Nonsense , Cytoskeletal Proteins , Muscle Proteins , Myocardium , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/pathology , Animals , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Female , Humans , Infant , Infant, Newborn , Male , Mice , Mice, Mutant Strains , Muscle Proteins/genetics , Muscle Proteins/metabolism , Myocardium/metabolism , Myocardium/pathology
8.
J Mol Cell Cardiol ; 122: 88-97, 2018 09.
Article in English | MEDLINE | ID: mdl-30102883

ABSTRACT

Leiomodin-2 (Lmod2) is a striated muscle-specific actin binding protein that is implicated in assembly of thin filaments. The necessity of Lmod2 in the adult mouse and role it plays in the mechanics of contraction are unknown. To answer these questions, we generated cardiac-specific conditional Lmod2 knockout mice (cKO). These mice die within a week of induction of the knockout with severe left ventricular systolic dysfunction and little change in cardiac morphology. Cardiac trabeculae isolated from cKO mice have a significant decrease in maximum force production and a blunting of myofilament length-dependent activation. Thin filaments are non-uniform and substantially reduced in length in cKO hearts, affecting the functional overlap of the thick and thin filaments. Remarkably, we also found that Lmod2 levels are directly linked to thin filament length and cardiac function in vivo, with a low amount (<20%) of Lmod2 necessary to maintain cardiac function. Thus, Lmod2 plays an essential role in maintaining proper cardiac thin filament length in adult mice, which in turn is necessary for proper generation of contractile force. Dysregulation of thin filament length in the absence of Lmod2 contributes to heart failure.


Subject(s)
Cytoskeletal Proteins/genetics , Heart Failure/genetics , Muscle Contraction/genetics , Muscle Proteins/genetics , Myofibrils/pathology , Analysis of Variance , Animals , Calcium/metabolism , Echocardiography , Gene Knockout Techniques , Heart Failure/pathology , Linear Models , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/metabolism , Sarcomeres/pathology , Ventricular Dysfunction, Left/diagnostic imaging
9.
Arch Biochem Biophys ; 647: 84-92, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29626422

ABSTRACT

Calcium regulation of cardiac muscle contraction is controlled by the thin-filament proteins troponin and tropomyosin bound to actin. In the absence of calcium, troponin-tropomyosin inhibits myosin-interactions on actin and induces muscle relaxation, whereas the addition of calcium relieves the inhibitory constraint to initiate contraction. Many mutations in thin filament proteins linked to cardiomyopathy appear to disrupt this regulatory switching. Here, we tested perturbations caused by mutant tropomyosins (E40K, DCM; and E62Q, HCM) on intra-filament interactions affecting acto-myosin interactions including those induced further by myosin association. Comparison of wild-type and mutant human α-tropomyosin (Tpm1.1) behavior was carried out using in vitro motility assays and molecular dynamics simulations. Our results show that E62Q tropomyosin destabilizes thin filament off-state function by increasing calcium-sensitivity, but without apparent affect on global tropomyosin structure by modifying coiled-coil rigidity. In contrast, the E40K mutant tropomyosin appears to stabilize the off-state, demonstrates increased tropomyosin flexibility, while also decreasing calcium-sensitivity. In addition, the E40K mutation reduces thin filament velocity at low myosin concentration while the E62Q mutant tropomyosin increases velocity. Corresponding molecular dynamics simulations indicate specific residue interactions that are likely to redefine underlying molecular regulatory mechanisms, which we propose explain the altered contractility evoked by the disease-causing mutations.


Subject(s)
Cardiomyopathies/genetics , Myosins/metabolism , Point Mutation , Tropomyosin/genetics , Troponin/metabolism , Calcium/metabolism , Cardiomyopathies/metabolism , Humans , Molecular Dynamics Simulation , Protein Conformation , Protein Interaction Maps , Tropomyosin/chemistry , Tropomyosin/metabolism
10.
J Mol Biol ; 428(11): 2446-2461, 2016 06 05.
Article in English | MEDLINE | ID: mdl-27107639

ABSTRACT

An "invariant proline" separates the myosin S1 head from its S2 tail and is proposed to be critical for orienting S1 during its interaction with actin, a process that leads to muscle contraction. Mutation of the invariant proline to leucine (P838L) caused dominant restrictive cardiomyopathy in a pediatric patient (Karam et al., Congenit. Heart Dis. 3:138-43, 2008). Here, we use Drosophila melanogaster to model this mutation and dissect its effects on the biochemical and biophysical properties of myosin, as well as on the structure and physiology of skeletal and cardiac muscles. P838L mutant myosin isolated from indirect flight muscles of transgenic Drosophila showed elevated ATPase and actin sliding velocity in vitro. Furthermore, the mutant heads exhibited increased rotational flexibility, and there was an increase in the average angle between the two heads. Indirect flight muscle myofibril assembly was minimally affected in mutant homozygotes, and isolated fibers displayed normal mechanical properties. However, myofibrils degraded during aging, correlating with reduced flight abilities. In contrast, hearts from homozygotes and heterozygotes showed normal morphology, myofibrillar arrays, and contractile parameters. When P838L was placed in trans to Mhc(5), an allele known to cause cardiac restriction in flies, it did not yield the constricted phenotype. Overall, our studies suggest that increased rotational flexibility of myosin S1 enhances myosin ATPase and actin sliding. Moreover, instability of P838L myofibrils leads to decreased function during aging of Drosophila skeletal muscle, but not cardiac muscle, despite the strong evolutionary conservation of the P838 residue.


Subject(s)
Cardiomyopathy, Restrictive/genetics , Drosophila melanogaster/genetics , Mutation/genetics , Myosin Subfragments/genetics , Proline/genetics , Actins/genetics , Animals , Drosophila melanogaster/metabolism , Flight, Animal/physiology , Muscle Contraction/genetics , Muscle, Skeletal/metabolism , Myocardium/metabolism , Myofibrils/genetics , Myosin Heavy Chains/genetics , Myosins/genetics , Phenotype
11.
Proc Natl Acad Sci U S A ; 113(8): 2306-11, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26858417

ABSTRACT

The Frank-Starling mechanism of the heart is due, in part, to modulation of myofilament Ca(2+) sensitivity by sarcomere length (SL) [length-dependent activation (LDA)]. The molecular mechanism(s) that underlie LDA are unknown. Recent evidence has implicated the giant protein titin in this cellular process, possibly by positioning the myosin head closer to actin. To clarify the role of titin strain in LDA, we isolated myocardium from either WT or homozygous mutant (HM) rats that express a giant splice isoform of titin, and subjected the muscles to stretch from 2.0 to 2.4 µm of SL. Upon stretch, HM compared with WT muscles displayed reduced passive force, twitch force, and myofilament LDA. Time-resolved small-angle X-ray diffraction measurements of WT twitching muscles during diastole revealed stretch-induced increases in the intensity of myosin (M2 and M6) and troponin (Tn3) reflections, as well as a reduction in cross-bridge radial spacing. Independent fluorescent probe analyses in relaxed permeabilized myocytes corroborated these findings. X-ray electron density reconstruction revealed increased mass/ordering in both thick and thin filaments. The SL-dependent changes in structure observed in WT myocardium were absent in HM myocardium. Overall, our results reveal a correlation between titin strain and the Frank-Starling mechanism. The molecular basis underlying this phenomenon appears not to involve interfilament spacing or movement of myosin toward actin but, rather, sarcomere stretch-induced simultaneous structural rearrangements within both thin and thick filaments that correlate with titin strain and myofilament LDA.


Subject(s)
Connectin/physiology , Heart/physiology , Animals , Calcium Signaling , Connectin/chemistry , Connectin/genetics , Models, Cardiovascular , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/physiology , Myocardial Contraction/genetics , Myocardial Contraction/physiology , Myocardium/metabolism , Myofibrils/physiology , Myosins/metabolism , Rats , Rats, Mutant Strains , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Scattering, Small Angle , Stress, Mechanical , Troponin C/genetics , Troponin C/metabolism , X-Ray Diffraction
12.
Nat Commun ; 6: 6018, 2015 Jan 19.
Article in English | MEDLINE | ID: mdl-25598495

ABSTRACT

Atrial fibrillation (AF) is the most common supraventricular arrhythmia that, for unknown reasons, is linked to intense endurance exercise. Our studies reveal that 6 weeks of swimming or treadmill exercise improves heart pump function and reduces heart-rates. Exercise also increases vulnerability to AF in association with inflammation, fibrosis, increased vagal tone, slowed conduction velocity, prolonged cardiomyocyte action potentials and RyR2 phosphorylation (CamKII-dependent S2814) in the atria, without corresponding alterations in the ventricles. Microarray results suggest the involvement of the inflammatory cytokine, TNFα, in exercised-induced atrial remodelling. Accordingly, exercise induces TNFα-dependent activation of both NFκB and p38MAPK, while TNFα inhibition (with etanercept), TNFα gene ablation, or p38 inhibition, prevents atrial structural remodelling and AF vulnerability in response to exercise, without affecting the beneficial physiological changes. Our results identify TNFα as a key factor in the pathology of intense exercise-induced AF.


Subject(s)
Atrial Fibrillation/metabolism , Atrial Fibrillation/physiopathology , Heart Atria/metabolism , Heart Atria/physiopathology , Physical Exertion/physiology , Tumor Necrosis Factor-alpha/metabolism , Animals , Heart Rate/physiology , Male , Mice , NF-kappa B/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
13.
J Appl Physiol (1985) ; 117(12): 1471-7, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25324513

ABSTRACT

Familial hypertrophic cardiomyopathy (HCM) is associated with mutations in sarcomeric proteins, including the myosin regulatory light chain (RLC). Here we studied the impact of three HCM mutations located in the NH2 terminus of the RLC on the molecular mechanism of ß-myosin heavy chain (MHC) cross-bridge mechanics using the in vitro motility assay. To generate mutant ß-myosin, native RLC was depleted from porcine cardiac MHC and reconstituted with mutant (A13T, F18L, and E22K) or wild-type (WT) human cardiac RLC. We characterized the mutant myosin force and motion generation capability in the presence of a frictional load. Compared with WT, all three mutants exhibited reductions in maximal actin filament velocity when tested under low or no frictional load. The actin-activated ATPase showed no significant difference between WT and HCM-mutant-reconstituted myosins. The decrease in velocity has been attributed to a significantly increased duty cycle, as was measured by the dependence of actin sliding velocity on myosin surface density, for all three mutant myosins. These results demonstrate a mutation-induced alteration in acto-myosin interactions that may contribute to the pathogenesis of HCM.


Subject(s)
Cardiac Myosins/metabolism , Cardiomyopathy, Hypertrophic, Familial/genetics , Cardiomyopathy, Hypertrophic, Familial/metabolism , Mutation , Myocardial Contraction , Myocytes, Cardiac/metabolism , Myosin Heavy Chains/metabolism , Myosin Light Chains/genetics , Myosin Light Chains/metabolism , Animals , Cardiomyopathy, Hypertrophic, Familial/physiopathology , Genetic Predisposition to Disease , Humans , Kinetics , Phenotype , Protein Binding , Protein Interaction Domains and Motifs , Recombinant Proteins/metabolism , Swine
14.
Arch Biochem Biophys ; 564: 89-99, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25241052

ABSTRACT

Striated muscle contraction is regulated by an interaction network connecting the effects of troponin, Ca(2+), and myosin-heads to the azimuthal positioning of tropomyosin along thin filaments. Many missense mutations, located at the actin-tropomyosin interface, however, reset the regulatory switching mechanism either by weakening or strengthening residue-specific interactions, leading to hyper- or hypo-contractile pathologies. Here, we compute energy landscapes for the actin-tropomyosin interface and quantify contributions of single amino acid residues to actin-tropomyosin binding. The method is a useful tool to assess effects of actin and tropomyosin mutations, potentially relating initial stages of myopathy to alterations in thin filament stability and regulation. Landscapes for mutant filaments linked to hyper-contractility provide a simple picture that describes a decrease in actin-tropomyosin interaction energy. Destabilizing the blocked (relaxed)-state parallels previously noted enhanced Ca(2+)-sensitivity conferred by these mutants. Energy landscapes also identify post-translational modifications that can rescue regulatory imbalances. For example, cardiomyopathy-associated E62Q tropomyosin mutation weakens actin-tropomyosin interaction, but phosphorylation of neighboring S61 rescues the binding-deficit, results confirmed experimentally by in vitro motility assays. Unlike results on hyper-contractility-related mutants, landscapes for tropomyosin mutants tied to hypo-contractility do not present a straightforward picture. These mutations may affect other components of the regulatory network, e.g., troponin-tropomyosin signaling.


Subject(s)
Actin Cytoskeleton/chemistry , Calcium/chemistry , Cardiomyopathies , Genetic Diseases, Inborn , Mutation, Missense , Tropomyosin/chemistry , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Amino Acid Substitution , Calcium/metabolism , Humans , Signal Transduction/genetics , Tropomyosin/genetics , Tropomyosin/metabolism
15.
J Mol Cell Cardiol ; 74: 318-29, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24992035

ABSTRACT

We have examined, for the first time, the effects of the familial hypertrophic cardiomyopathy (HCM)-associated Lys104Glu mutation in the myosin regulatory light chain (RLC). Transgenic mice expressing the Lys104Glu substitution (Tg-MUT) were generated and the results were compared to Tg-WT (wild-type human ventricular RLC) mice. Echocardiography with pulse wave Doppler in 6month-old Tg-MUT showed early signs of diastolic disturbance with significantly reduced E/A transmitral velocities ratio. Invasive hemodynamics in 6month-old Tg-MUT mice also demonstrated a borderline significant prolonged isovolumic relaxation time (Tau) and a tendency for slower rate of pressure decline, suggesting alterations in diastolic function in Tg-MUT. Six month-old mutant animals had no LV hypertrophy; however, at >13months they displayed significant hypertrophy and fibrosis. In skinned papillary muscles from 5 to 6month-old mice a mutation induced reduction in maximal tension and slower muscle relaxation rates were observed. Mutated cross-bridges showed increased rates of binding to the thin filaments and a faster rate of the power stroke. In addition, ~2-fold lower level of RLC phosphorylation was observed in the mutant compared to Tg-WT. In line with the higher mitochondrial content seen in Tg-MUT hearts, the MUT-myosin ATPase activity was significantly higher than WT-myosin, indicating increased energy consumption. In the in vitro motility assay, MUT-myosin produced higher actin sliding velocity under zero load, but the velocity drastically decreased with applied load in the MUT vs. WT myosin. Our results suggest that diastolic disturbance (impaired muscle relaxation, lower E/A) and inefficiency of energy use (reduced contractile force and faster ATP consumption) may underlie the Lys104Glu-mediated HCM phenotype.


Subject(s)
Cardiomyopathy, Hypertrophic/genetics , Mutation , Myocytes, Cardiac/metabolism , Myosin Light Chains/genetics , Papillary Muscles/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Animals , Calcium/metabolism , Cardiomyopathy, Hypertrophic/diagnostic imaging , Cardiomyopathy, Hypertrophic/metabolism , Cardiomyopathy, Hypertrophic/pathology , Diastole , Gene Expression Regulation , Heart Rate , Male , Mice , Mice, Transgenic , Molecular Sequence Data , Muscle Relaxation , Myocardial Contraction , Myocytes, Cardiac/pathology , Myosin Light Chains/metabolism , Papillary Muscles/diagnostic imaging , Papillary Muscles/pathology , Primary Cell Culture , Signal Transduction , Tissue Culture Techniques , Ultrasonography, Doppler, Pulsed
16.
Biophys J ; 103(6): 1275-84, 2012 Sep 19.
Article in English | MEDLINE | ID: mdl-22995500

ABSTRACT

The demembranated (skinned) muscle fiber preparation is widely used to investigate muscle contraction because the intracellular ionic conditions can be precisely controlled. However, plasma membrane removal results in a loss of osmotic regulation, causing abnormal hydration of the myofilament lattice and its proteins. We investigated the structural and functional consequences of varied myofilament lattice spacing and protein hydration on cross-bridge rates of force development and detachment in Drosophila melanogaster indirect flight muscle, using x-ray diffraction to compare the lattice spacing of dissected, osmotically compressed skinned fibers to native muscle fibers in living flies. Osmolytes of different sizes and exclusion properties (Dextran T-500 and T-10) were used to differentially alter lattice spacing and protein hydration. At in vivo lattice spacing, cross-bridge attachment time (t(on)) increased with higher osmotic pressures, consistent with a reduced cross-bridge detachment rate as myofilament protein hydration decreased. In contrast, in the swollen lattice, t(on) decreased with higher osmotic pressures. These divergent responses were reconciled using a structural model that predicts t(on) varies inversely with thick-to-thin filament surface distance, suggesting that cross-bridge rates of force development and detachment are modulated more by myofilament lattice geometry than protein hydration. Generalizing these findings, our results suggest that cross-bridge cycling rates slow as thick-to-thin filament surface distance decreases with sarcomere lengthening, and likewise, cross-bridge cycling rates increase during sarcomere shortening. Together, these structural changes may provide a mechanism for altering cross-bridge performance throughout a contraction-relaxation cycle.


Subject(s)
Drosophila melanogaster/cytology , Drosophila melanogaster/physiology , Flight, Animal , Muscle, Skeletal/metabolism , Myofibrils/metabolism , Actins/metabolism , Animals , Biomechanical Phenomena , Dextrans/pharmacology , Drosophila melanogaster/metabolism , Kinetics , Muscle, Skeletal/cytology , Muscle, Skeletal/physiology , Myofibrils/drug effects , Myosins/metabolism , Osmosis/drug effects , Surface Properties
17.
Circ Res ; 109(9): 1024-1030, 2011 Oct 14.
Article in English | MEDLINE | ID: mdl-21903937

ABSTRACT

RATIONALE: Baseline contractility of mouse hearts is modulated in a phosphatidylinositol 3-kinase-γ-dependent manner by type 4 phosphodiesterases (PDE4), which regulate cAMP levels within microdomains containing the sarcoplasmic reticulum (SR) calcium ATPase type 2a (SERCA2a). OBJECTIVE: The goal of this study was to determine whether PDE4D regulates basal cardiac contractility. METHODS AND RESULTS: At 10 to 12 weeks of age, baseline cardiac contractility in PDE4D-deficient (PDE4D(-/-)) mice was elevated mice in vivo and in Langendorff perfused hearts, whereas isolated PDE4D(-/-) cardiomyocytes showed increased whole-cell Ca2+ transient amplitudes and SR Ca2+content but unchanged L-type calcium current, compared with littermate controls (WT). The protein kinase A inhibitor R(p)-adenosine-3',5' cyclic monophosphorothioate (R(p)-cAMP) lowered whole-cell Ca2+ transient amplitudes and SR Ca2+ content in PDE4D(-/-) cardiomyocytes to WT levels. The PDE4 inhibitor rolipram had no effect on cardiac contractility, whole-cell Ca2+ transients, or SR Ca2+ content in PDE4D(-/-) preparations but increased these parameters in WT myocardium to levels indistinguishable from those in PDE4D(-/-). The functional changes in PDE4D(-/-) myocardium were associated with increased PLN phosphorylation but not cardiac ryanodine receptor phosphorylation. Rolipram increased PLN phosphorylation in WT cardiomyocytes to levels indistinguishable from those in PDE4D(-/-) cardiomyocytes. In murine and failing human hearts, PDE4D coimmunoprecipitated with SERCA2a but not with cardiac ryanodine receptor. CONCLUSIONS: PDE4D regulates basal cAMP levels in SR microdomains containing SERCA2a-PLN, but not L-type Ca2+ channels or ryanodine receptor. Because whole-cell Ca2+ transient amplitudes are reduced in failing human myocardium, these observations may have therapeutic implications for patients with heart failure.


Subject(s)
Calcium Channels, L-Type/physiology , Calcium/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Myocardial Contraction/physiology , Myocytes, Cardiac/metabolism , Sarcoplasmic Reticulum/metabolism , Animals , Calcium-Binding Proteins/metabolism , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Female , Heart Ventricles/metabolism , Heart Ventricles/pathology , Humans , Male , Mice , Mice, Knockout , Models, Animal , Myocytes, Cardiac/pathology , Phosphatidylinositol 3-Kinases/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
18.
Proc Natl Acad Sci U S A ; 108(33): 13576-81, 2011 Aug 16.
Article in English | MEDLINE | ID: mdl-21825130

ABSTRACT

Rapid electrical conduction in the His-Purkinje system tightly controls spatiotemporal activation of the ventricles. Although recent work has shed much light on the regulation of early specification and morphogenesis of the His-Purkinje system, less is known about how transcriptional regulation establishes impulse conduction properties of the constituent cells. Here we show that Iroquois homeobox gene 3 (Irx3) is critical for efficient conduction in this specialized tissue by antithetically regulating two gap junction-forming connexins (Cxs). Loss of Irx3 resulted in disruption of the rapid coordinated spread of ventricular excitation, reduced levels of Cx40, and ectopic Cx43 expression in the proximal bundle branches. Irx3 directly represses Cx43 transcription and indirectly activates Cx40 transcription. Our results reveal a critical role for Irx3 in the precise regulation of intercellular gap junction coupling and impulse propagation in the heart.


Subject(s)
Bundle of His/physiology , Heart Conduction System , Homeodomain Proteins/physiology , Purkinje Fibers/physiology , Transcription Factors/physiology , Animals , Connexin 43/genetics , Connexins/genetics , Gap Junctions , Gene Expression Regulation , Genes, Homeobox , Heart Ventricles , Mice , Transcription, Genetic
19.
Biophys J ; 100(7): 1737-46, 2011 Apr 06.
Article in English | MEDLINE | ID: mdl-21463587

ABSTRACT

The N-terminal extension and phosphorylation of the myosin regulatory light chain (RLC) independently improve Drosophila melanogaster flight performance. Here we examine the functional and structural role of the RLC in chemically skinned fibers at various thick and thin filament lattice spacings from four transgenic Drosophila lines: rescued null or control (Dmlc2(+)), truncated N-terminal extension (Dmlc2(Δ2-46)), disrupted myosin light chain kinase phosphorylation sites (Dmlc2(S66A,S67A)), and dual mutant (Dmlc2(Δ2-46; S66A,S67A)). The N-terminal extension truncation and phosphorylation sites disruption mutations decreased oscillatory power output and the frequency of maximum power output in maximally Ca(2+)-activated fibers compressed to near in vivo inter-thick filament spacing, with the phosphorylation sites disruption mutation having a larger affect. The diminished power output parameters with the N-terminal extension truncation and phosphorylation sites disruption mutations were due to the reduction of the number of strongly-bound cross-bridges and rate of myosin force production, with the larger parameter reductions in the phosphorylation sites disruption mutation additionally related to reduced myosin attachment time. The phosphorylation and N-terminal extension-dependent boost in cross-bridge kinetics corroborates previous structural data, which indicate these RLC attributes play a complementary role in moving and orienting myosin heads toward actin target sites, thereby increasing fiber and whole fly power generation.


Subject(s)
Actin Cytoskeleton/metabolism , Drosophila melanogaster/metabolism , Myosin Light Chains/chemistry , Myosin Light Chains/metabolism , Actin Cytoskeleton/chemistry , Animals , Biomechanical Phenomena , Elastic Modulus , Flight, Animal , Muscle Fibers, Skeletal/metabolism , Phosphorylation , Viscosity , X-Ray Diffraction
20.
Am J Physiol Heart Circ Physiol ; 300(6): H2155-60, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21460195

ABSTRACT

The cellular mechanism underlying the Frank-Starling law of the heart is myofilament length-dependent activation. The mechanism(s) whereby sarcomeres detect changes in length and translate this into increased sensitivity to activating calcium has been elusive. Small-angle X-ray diffraction studies have revealed that the intact myofilament lattice undergoes numerous structural changes upon an increase in sarcomere length (SL): lattice spacing and the I(1,1)/I(1,0) intensity ratio decreases, whereas the M3 meridional reflection intensity (I(M3)) increases, concomitant with increases in diastolic and systolic force. Using a short (∼10 ms) X-ray exposure just before electrical stimulation, we were able to obtain detailed structural information regarding the effects of external osmotic compression (with mannitol) and obtain SL on thin intact electrically stimulated isolated rat right ventricular trabeculae. We show that over the same incremental increases in SL, the relative changes in systolic force track more closely to the relative changes in myosin head orientation (as reported by I(M3)) than to the relative changes in lattice spacing. We conclude that myosin head orientation before activation determines myocardial sarcomere activation levels and that this may be the dominant mechanism for length-dependent activation.


Subject(s)
Actin Cytoskeleton/diagnostic imaging , Heart/physiology , Myosin Heavy Chains/chemistry , Myosins/chemistry , Stroke Volume/physiology , Actin Cytoskeleton/physiology , Animals , Electric Stimulation , Male , Models, Animal , Myocardial Contraction/physiology , Myocardium/metabolism , Myosin Heavy Chains/metabolism , Myosins/metabolism , Radiography , Rats , Rats, Inbred Strains , Sarcomeres/diagnostic imaging , Sarcomeres/physiology , X-Ray Diffraction
SELECTION OF CITATIONS
SEARCH DETAIL