Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
Add more filters

Publication year range
1.
PLoS Genet ; 20(5): e1011279, 2024 May.
Article in English | MEDLINE | ID: mdl-38748723

ABSTRACT

The leiomodin (Lmod) family of actin-binding proteins play a critical role in muscle function, highlighted by the fact that mutations in all three family members (LMOD1-3) result in human myopathies. Mutations in the cardiac predominant isoform, LMOD2 lead to severe neonatal dilated cardiomyopathy. Most of the disease-causing mutations in the LMOD gene family are nonsense, or frameshift, mutations predicted to result in expression of truncated proteins. However, in nearly all cases of disease, little to no LMOD protein is expressed. We show here that nonsense-mediated mRNA decay, a cellular mechanism which eliminates mRNAs with premature termination codons, underlies loss of mutant protein from two independent LMOD2 disease-causing mutations. Furthermore, we generated steric-blocking oligonucleotides that obstruct deposition of the exon junction complex, preventing nonsense-mediated mRNA decay of mutant LMOD2 transcripts, thereby restoring mutant protein expression. Our investigation lays the initial groundwork for potential therapeutic intervention in LMOD-linked myopathies.


Subject(s)
Codon, Nonsense , Nonsense Mediated mRNA Decay , Humans , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/metabolism , Codon, Nonsense/genetics , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Mutation , Nonsense Mediated mRNA Decay/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
2.
Proc Natl Acad Sci U S A ; 120(47): e2315820120, 2023 Nov 21.
Article in English | MEDLINE | ID: mdl-37956287

ABSTRACT

Actin is a highly expressed protein in eukaryotic cells and is essential for numerous cellular processes. In particular, efficient striated muscle contraction is dependent upon the precise regulation of actin-based thin filament structure and function. Alterations in the lengths of actin-thin filaments can lead to the development of myopathies. Leiomodins and tropomodulins are members of an actin-binding protein family that fine-tune thin filament lengths, and their dysfunction is implicated in muscle diseases. An Lmod3 mutation [G326R] was previously identified in patients with nemaline myopathy (NM), a severe skeletal muscle disorder; this residue is conserved among Lmod and Tmod isoforms and resides within their homologous leucine-rich repeat (LRR) domain. We mutated this glycine to arginine in Lmod and Tmod to determine the physiological function of this residue and domain. This G-to-R substitution disrupts Lmod and Tmod's LRR domain structure, altering their binding interface with actin and destroying their abilities to regulate thin filament lengths. Additionally, this mutation renders Lmod3 nonfunctional in vivo. We found that one single amino acid is essential for folding of Lmod and Tmod LRR domains, and thus is essential for the opposing actin-regulatory functions of Lmod (filament elongation) and Tmod (filament shortening), revealing a mechanism underlying the development of NM.


Subject(s)
Actins , Myopathies, Nemaline , Humans , Actins/metabolism , Tropomodulin/genetics , Tropomodulin/metabolism , Myopathies, Nemaline/genetics , Myopathies, Nemaline/metabolism , Muscle Proteins/metabolism , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Sarcomeres/genetics , Sarcomeres/metabolism , Mutation , Muscle, Skeletal/metabolism
3.
PLoS Biol ; 18(9): e3000848, 2020 09.
Article in English | MEDLINE | ID: mdl-32898131

ABSTRACT

Improper lengths of actin-thin filaments are associated with altered contractile activity and lethal myopathies. Leiomodin, a member of the tropomodulin family of proteins, is critical in thin filament assembly and maintenance; however, its role is under dispute. Using nuclear magnetic resonance data and molecular dynamics simulations, we generated the first atomic structural model of the binding interface between the tropomyosin-binding site of cardiac leiomodin and the N-terminus of striated muscle tropomyosin. Our structural data indicate that the leiomodin/tropomyosin complex only forms at the pointed end of thin filaments, where the tropomyosin N-terminus is not blocked by an adjacent tropomyosin protomer. This discovery provides evidence supporting the debated mechanism where leiomodin and tropomodulin regulate thin filament lengths by competing for thin filament binding. Data from experiments performed in cardiomyocytes provide additional support for the competition model; specifically, expression of a leiomodin mutant that is unable to interact with tropomyosin fails to displace tropomodulin at thin filament pointed ends and fails to elongate thin filaments. Together with previous structural and biochemical data, we now propose a molecular mechanism of actin polymerization at the pointed end in the presence of bound leiomodin. In the proposed model, the N-terminal actin-binding site of leiomodin can act as a "swinging gate" allowing limited actin polymerization, thus making leiomodin a leaky pointed-end cap. Results presented in this work answer long-standing questions about the role of leiomodin in thin filament length regulation and maintenance.


Subject(s)
Actin Cytoskeleton/metabolism , Microfilament Proteins/chemistry , Microfilament Proteins/metabolism , Muscle Proteins/chemistry , Muscle Proteins/metabolism , Actin Capping Proteins/chemistry , Actin Capping Proteins/metabolism , Actin Cytoskeleton/chemistry , Actins/chemistry , Actins/metabolism , Animals , Animals, Newborn , Binding Sites , Cells, Cultured , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/metabolism , Humans , Mice , Models, Molecular , Molecular Dynamics Simulation , Myocardium/metabolism , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Interaction Domains and Motifs , Rats , Sarcomeres/metabolism
4.
Proc Natl Acad Sci U S A ; 116(50): 25126-25136, 2019 12 10.
Article in English | MEDLINE | ID: mdl-31757849

ABSTRACT

Cardiac protein homeostasis, sarcomere assembly, and integration of titin as the sarcomeric backbone are tightly regulated to facilitate adaptation and repair. Very little is known on how the >3-MDa titin protein is synthesized, moved, inserted into sarcomeres, detached, and degraded. Here, we generated a bifluorescently labeled knockin mouse to simultaneously visualize both ends of the molecule and follow titin's life cycle in vivo. We find titin mRNA, protein synthesis and degradation compartmentalized toward the Z-disk in adult, but not embryonic cardiomyocytes. Originating at the Z-disk, titin contributes to a soluble protein pool (>15% of total titin) before it is integrated into the sarcomere lattice. Titin integration, disintegration, and reintegration are stochastic and do not proceed sequentially from Z-disk to M-band, as suggested previously. Exchange between soluble and integrated titin depends on titin protein composition and differs between individual cardiomyocytes. Thus, titin dynamics facilitate embryonic vs. adult sarcomere remodeling with implications for cardiac development and disease.


Subject(s)
Myocytes, Cardiac/metabolism , Protein Kinases , Proteostasis/physiology , Animals , Mice , Mice, Transgenic , Microscopy , Protein Kinases/genetics , Protein Kinases/metabolism , Sarcomeres/metabolism , Single-Cell Analysis
5.
Proc Natl Acad Sci U S A ; 114(45): 11956-11961, 2017 11 07.
Article in English | MEDLINE | ID: mdl-29078393

ABSTRACT

Small heat shock protein HSPB7 is highly expressed in the heart. Several mutations within HSPB7 are associated with dilated cardiomyopathy and heart failure in human patients. However, the precise role of HSPB7 in the heart is still unclear. In this study, we generated global as well as cardiac-specific HSPB7 KO mouse models and found that loss of HSPB7 globally or specifically in cardiomyocytes resulted in embryonic lethality before embryonic day 12.5. Using biochemical and cell culture assays, we identified HSPB7 as an actin filament length regulator that repressed actin polymerization by binding to monomeric actin. Consistent with HSPB7's inhibitory effects on actin polymerization, HSPB7 KO mice had longer actin/thin filaments and developed abnormal actin filament bundles within sarcomeres that interconnected Z lines and were cross-linked by α-actinin. In addition, loss of HSPB7 resulted in up-regulation of Lmod2 expression and mislocalization of Tmod1. Furthermore, crossing HSPB7 null mice into an Lmod2 null background rescued the elongated thin filament phenotype of HSPB7 KOs, but double KO mice still exhibited formation of abnormal actin bundles and early embryonic lethality. These in vivo findings indicated that abnormal actin bundles, not elongated thin filament length, were the cause of embryonic lethality in HSPB7 KOs. Our findings showed an unsuspected and critical role for a specific small heat shock protein in directly modulating actin thin filament length in cardiac muscle by binding monomeric actin and limiting its availability for polymerization.


Subject(s)
Actin Cytoskeleton/metabolism , Cardiomyopathies/genetics , HSP27 Heat-Shock Proteins/genetics , Heart Defects, Congenital/genetics , Heart/embryology , Actin Cytoskeleton/genetics , Animals , Cytoskeletal Proteins/biosynthesis , Cytoskeletal Proteins/genetics , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Proteins/biosynthesis , Muscle Proteins/genetics , Myocardium/cytology , Myocytes, Cardiac/cytology , Organogenesis/genetics , Sarcomeres/metabolism , Tropomodulin/metabolism
6.
Genes Dev ; 26(2): 114-9, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22241783

ABSTRACT

Protein lysine methylation is one of the most widespread post-translational modifications in the nuclei of eukaryotic cells. Methylated lysines on histones and nonhistone proteins promote the formation of protein complexes that control gene expression and DNA replication and repair. In the cytoplasm, however, the role of lysine methylation in protein complex formation is not well established. Here we report that the cytoplasmic protein chaperone Hsp90 is methylated by the lysine methyltransferase Smyd2 in various cell types. In muscle, Hsp90 methylation contributes to the formation of a protein complex containing Smyd2, Hsp90, and the sarcomeric protein titin. Deficiency in Smyd2 results in the loss of Hsp90 methylation, impaired titin stability, and altered muscle function. Collectively, our data reveal a cytoplasmic protein network that employs lysine methylation for the maintenance and function of skeletal muscle.


Subject(s)
Cytoplasm/metabolism , HSP90 Heat-Shock Proteins/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Muscle, Skeletal/metabolism , Myofibrils/metabolism , Animals , Chick Embryo , Connectin , Cytoplasm/enzymology , Histone-Lysine N-Methyltransferase/genetics , Humans , Lysine/metabolism , Methylation , Muscle Proteins/metabolism , Myocardium/metabolism , Protein Kinases/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Zebrafish
7.
Circulation ; 137(6): 605-618, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29101288

ABSTRACT

BACKGROUND: Gap junction remodeling is well established as a consistent feature of human heart disease involving spontaneous ventricular arrhythmia. The mechanisms responsible for gap junction remodeling that include alterations in the distribution of, and protein expression within, gap junctions are still debated. Studies reveal that multiple transcriptional and posttranscriptional regulatory pathways are triggered in response to cardiac disease, such as those involving RNA-binding proteins. The expression levels of FXR1 (fragile X mental retardation autosomal homolog 1), an RNA-binding protein, are critical to maintain proper cardiac muscle function; however, the connection between FXR1 and disease is not clear. METHODS: To identify the mechanisms regulating gap junction remodeling in cardiac disease, we sought to identify the functional properties of FXR1 expression, direct targets of FXR1 in human left ventricle dilated cardiomyopathy (DCM) biopsy samples and mouse models of DCM through BioID proximity assay and RNA immunoprecipitation, how FXR1 regulates its targets through RNA stability and luciferase assays, and functional consequences of altering the levels of this important RNA-binding protein through the analysis of cardiac-specific FXR1 knockout mice and mice injected with 3xMyc-FXR1 adeno-associated virus. RESULTS: FXR1 expression is significantly increased in tissue samples from human and mouse models of DCM via Western blot analysis. FXR1 associates with intercalated discs, and integral gap junction proteins Cx43 (connexin 43), Cx45 (connexin 45), and ZO-1 (zonula occludens-1) were identified as novel mRNA targets of FXR1 by using a BioID proximity assay and RNA immunoprecipitation. Our findings show that FXR1 is a multifunctional protein involved in translational regulation and stabilization of its mRNA targets in heart muscle. In addition, introduction of 3xMyc-FXR1 via adeno-associated virus into mice leads to the redistribution of gap junctions and promotes ventricular tachycardia, showing the functional significance of FXR1 upregulation observed in DCM. CONCLUSIONS: In DCM, increased FXR1 expression appears to play an important role in disease progression by regulating gap junction remodeling. Together this study provides a novel function of FXR1, namely, that it directly regulates major gap junction components, contributing to proper cell-cell communication in the heart.


Subject(s)
Cardiomyopathy, Hypertrophic/metabolism , Gap Junctions/metabolism , Heart Ventricles/metabolism , RNA-Binding Proteins/metabolism , Tachycardia, Ventricular/etiology , Ventricular Function, Left , Ventricular Remodeling , Animals , Cardiomyopathy, Hypertrophic/complications , Cardiomyopathy, Hypertrophic/pathology , Cardiomyopathy, Hypertrophic/physiopathology , Cell Communication , Connexins/genetics , Connexins/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Gap Junctions/pathology , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Humans , Mice, Inbred C57BL , Mice, Knockout , Muscle Proteins/genetics , Muscle Proteins/metabolism , RNA-Binding Proteins/genetics , Rats , Risk Factors , Signal Transduction , Tachycardia, Ventricular/metabolism , Tachycardia, Ventricular/pathology , Tachycardia, Ventricular/physiopathology , Up-Regulation , Zonula Occludens-1 Protein/genetics , Zonula Occludens-1 Protein/metabolism
8.
Circulation ; 138(17): 1864-1878, 2018 10 23.
Article in English | MEDLINE | ID: mdl-29716942

ABSTRACT

BACKGROUND: Hypertrophic cardiomyocyte growth and dysfunction accompany various forms of heart disease. The mechanisms responsible for transcriptional changes that affect cardiac physiology and the transition to heart failure are not well understood. The intercalated disc (ID) is a specialized intercellular junction coupling cardiomyocyte force transmission and propagation of electrical activity. The ID is gaining attention as a mechanosensitive signaling hub and hotspot for causative mutations in cardiomyopathy. METHODS: Transmission electron microscopy, confocal microscopy, and single-molecule localization microscopy were used to examine changes in ID structure and protein localization in the murine and human heart. We conducted detailed cardiac functional assessment and transcriptional profiling of mice lacking myocardin-related transcription factor (MRTF)-A and MRTF-B specifically in adult cardiomyocytes to evaluate the role of mechanosensitive regulation of gene expression in load-induced ventricular remodeling. RESULTS: We found that MRTFs localize to IDs in the healthy human heart and accumulate in the nucleus in heart failure. Although mice lacking MRTFs in adult cardiomyocytes display normal cardiac physiology at baseline, pressure overload leads to rapid heart failure characterized by sarcomere disarray, ID disintegration, chamber dilation and wall thinning, cardiac functional decline, and partially penetrant acute lethality. Transcriptional profiling reveals a program of actin cytoskeleton and cardiomyocyte adhesion genes driven by MRTFs during pressure overload. Indeed, conspicuous remodeling of gap junctions at IDs identified by single-molecule localization microscopy may partially stem from a reduction in Mapre1 expression, which we show is a direct mechanosensitive MRTF target. CONCLUSIONS: Our study describes a novel paradigm in which MRTFs control an acute mechanosensitive signaling circuit that coordinates cross-talk between the actin and microtubule cytoskeleton and maintains ID integrity and cardiomyocyte homeostasis in heart disease.


Subject(s)
Heart Failure/metabolism , Hypertrophy, Left Ventricular/metabolism , Mechanotransduction, Cellular , Myocytes, Cardiac/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Aged , Animals , Animals, Newborn , COS Cells , Case-Control Studies , Chlorocebus aethiops , Connexin 43/genetics , Connexin 43/metabolism , Female , Gene Expression Regulation , Heart Failure/genetics , Heart Failure/pathology , Heart Failure/physiopathology , Humans , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Microscopy, Electron, Transmission , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Middle Aged , Myocytes, Cardiac/ultrastructure , NIH 3T3 Cells , Single Molecule Imaging , Trans-Activators/deficiency , Trans-Activators/genetics , Transcription Factors/deficiency , Transcription Factors/genetics , Ventricular Function, Left , Ventricular Remodeling
9.
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
10.
Proc Natl Acad Sci U S A ; 112(44): 13573-8, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26487682

ABSTRACT

Leiomodin 2 (Lmod2) is an actin-binding protein that has been implicated in the regulation of striated muscle thin filament assembly; its physiological function has yet to be studied. We found that knockout of Lmod2 in mice results in abnormally short thin filaments in the heart. We also discovered that Lmod2 functions to elongate thin filaments by promoting actin assembly and dynamics at thin filament pointed ends. Lmod2-KO mice die as juveniles with hearts displaying contractile dysfunction and ventricular chamber enlargement consistent with dilated cardiomyopathy. Lmod2-null cardiomyocytes produce less contractile force than wild type when plated on micropillar arrays. Introduction of GFP-Lmod2 via adeno-associated viral transduction elongates thin filaments and rescues structural and functional defects observed in Lmod2-KO mice, extending their lifespan to adulthood. Thus, to our knowledge, Lmod2 is the first identified mammalian protein that functions to elongate actin filaments in the heart; it is essential for cardiac thin filaments to reach a mature length and is required for efficient contractile force and proper heart function during development.


Subject(s)
Actin Cytoskeleton/metabolism , Cardiomyopathy, Dilated/metabolism , Cytoskeletal Proteins/metabolism , Muscle Proteins/metabolism , Myocardium/metabolism , Actin Cytoskeleton/genetics , Animals , Animals, Newborn , Cardiomyopathy, Dilated/embryology , Cardiomyopathy, Dilated/genetics , Cells, Cultured , Cytoskeletal Proteins/genetics , Fluorescence Recovery After Photobleaching , Genes, Lethal/genetics , Heart/embryology , Heart/physiopathology , Immunoblotting , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice, Knockout , Mice, Transgenic , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Muscle Contraction/genetics , Muscle Contraction/physiology , Muscle Proteins/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myocardium/pathology , Myocardium/ultrastructure , Sarcomeres/genetics , Sarcomeres/metabolism , Survival Analysis
11.
J Cell Sci ; 128(2): 239-50, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25431137

ABSTRACT

Precise regulation of thin filament length is essential for optimal force generation during muscle contraction. The thin filament capping protein tropomodulin (Tmod) contributes to thin filament length uniformity by regulating elongation and depolymerization at thin filament ends. The leiomodins (Lmod1-3) are structurally related to Tmod1-4 and also localize to actin filament pointed ends, but in vitro biochemical studies indicate that Lmods act instead as robust nucleators. Here, we examined the roles of Tmod4 and Lmod3 during Xenopus skeletal myofibrillogenesis. Loss of Tmod4 or Lmod3 resulted in severe disruption of sarcomere assembly and impaired embryonic movement. Remarkably, when Tmod4-deficient embryos were supplemented with additional Lmod3, and Lmod3-deficient embryos were supplemented with additional Tmod4, sarcomere assembly was rescued and embryonic locomotion improved. These results demonstrate for the first time that appropriate levels of both Tmod4 and Lmod3 are required for embryonic myofibrillogenesis and, unexpectedly, both proteins can function redundantly during in vivo skeletal muscle thin filament assembly. Furthermore, these studies demonstrate the value of Xenopus for the analysis of contractile protein function during de novo myofibril assembly.


Subject(s)
Embryo, Nonmammalian , Muscle Development/genetics , Muscle Proteins/biosynthesis , Tropomodulin/biosynthesis , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/ultrastructure , Animals , Gene Expression Regulation, Developmental , Microfilament Proteins , Muscle Contraction/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/embryology , Muscle, Skeletal/metabolism , Myocardium/metabolism , Myocardium/ultrastructure , Sarcomeres/genetics , Sarcomeres/ultrastructure , Tropomodulin/genetics , Xenopus laevis/embryology , Xenopus laevis/genetics
12.
Ann Neurol ; 79(6): 959-69, 2016 06.
Article in English | MEDLINE | ID: mdl-27074222

ABSTRACT

OBJECTIVE: Thin filament myopathies are among the most common nondystrophic congenital muscular disorders, and are caused by mutations in genes encoding proteins that are associated with the skeletal muscle thin filament. Mechanisms underlying muscle weakness are poorly understood, but might involve the length of the thin filament, an important determinant of force generation. METHODS: We investigated the sarcomere length-dependence of force, a functional assay that provides insights into the contractile strength of muscle fibers as well as the length of the thin filaments, in muscle fibers from 51 patients with thin filament myopathy caused by mutations in NEB, ACTA1, TPM2, TPM3, TNNT1, KBTBD13, KLHL40, and KLHL41. RESULTS: Lower force generation was observed in muscle fibers from patients of all genotypes. In a subset of patients who harbor mutations in NEB and ACTA1, the lower force was associated with downward shifted force-sarcomere length relations, indicative of shorter thin filaments. Confocal microscopy confirmed shorter thin filaments in muscle fibers of these patients. A conditional Neb knockout mouse model, which recapitulates thin filament myopathy, revealed a compensatory mechanism; the lower force generation that was associated with shorter thin filaments was compensated for by increasing the number of sarcomeres in series. This allowed muscle fibers to operate at a shorter sarcomere length and maintain optimal thin-thick filament overlap. INTERPRETATION: These findings might provide a novel direction for the development of therapeutic strategies for thin filament myopathy patients with shortened thin filament lengths. Ann Neurol 2016;79:959-969.


Subject(s)
Cytoskeleton/genetics , Muscle Proteins/genetics , Muscular Diseases/genetics , Muscular Diseases/physiopathology , Sarcomeres/genetics , Actins/genetics , Animals , Case-Control Studies , Cytoskeleton/physiology , Humans , Mice, Knockout , Muscle Contraction/genetics , Muscle Contraction/physiology , Muscle Proteins/metabolism , Muscle Proteins/physiology , Muscle, Skeletal/metabolism , Mutation , Sarcomeres/physiology
13.
Proc Natl Acad Sci U S A ; 111(51): E5527-36, 2014 Dec 23.
Article in English | MEDLINE | ID: mdl-25512505

ABSTRACT

CAPN3/p94/calpain-3, a calpain protease family member predominantly expressed in skeletal muscle, possesses unusually rapid and exhaustive autolytic activity. Mutations in the human CAPN3 gene impairing its protease functions cause limb-girdle muscular dystrophy type 2A (LGMD2A); yet, the connection between CAPN3's autolytic activity and the enzyme's function in vivo remain unclear. Here, we demonstrated that CAPN3 protease activity was reconstituted by intermolecular complementation (iMOC) between its two autolytic fragments. Furthermore, the activity of full-length CAPN3 active-site mutants was surprisingly rescued through iMOC with autolytic fragments containing WT amino acid sequences. These results provide evidence that WT CAPN3 can be formed by the iMOC of two different complementary CAPN3 mutants. The finding of iMOC-mediated restoration of calpain activity indicates a novel mechanism for the genotype-phenotype links in LGMD2A.


Subject(s)
Calpain/metabolism , Muscle Proteins/metabolism , Animals , COS Cells , Calpain/chemistry , Calpain/genetics , Chlorocebus aethiops , Humans , Muscle Proteins/chemistry , Muscle Proteins/genetics , Mutation , Proteolysis
14.
Proc Natl Acad Sci U S A ; 111(40): 14589-94, 2014 Oct 07.
Article in English | MEDLINE | ID: mdl-25246556

ABSTRACT

Titin, the largest protein known, forms a giant filament in muscle where it spans the half sarcomere from Z disk to M band. Here we genetically targeted a stretch of 14 immunoglobulin-like and fibronectin type 3 domains that comprises the I-band/A-band (IA) junction and obtained a viable mouse model. Super-resolution optical microscopy (structured illumination microscopy, SIM) and electron microscopy were used to study the thick filament length and titin's molecular elasticity. SIM showed that the IA junction functionally belongs to the relatively stiff A-band region of titin. The stiffness of A-band titin was found to be high, relative to that of I-band titin (∼ 40-fold higher) but low, relative to that of the myosin-based thick filament (∼ 70-fold lower). Sarcomere stretch therefore results in movement of A-band titin with respect to the thick filament backbone, and this might constitute a novel length-sensing mechanism. Findings disproved that titin at the IA junction is crucial for thick filament length control, settling a long-standing hypothesis. SIM also showed that deleting the IA junction moves the attachment point of titin's spring region away from the Z disk, increasing the strain on titin's molecular spring elements. Functional studies from the cellular to ex vivo and in vivo left ventricular chamber levels showed that this causes diastolic dysfunction and other symptoms of heart failure with preserved ejection fraction (HFpEF). Thus, our work supports titin's important roles in diastolic function and disease of the heart.


Subject(s)
Connectin/metabolism , Heart/physiology , Myocardium/metabolism , Sarcomeres/metabolism , Amino Acid Sequence , Animals , Biomechanical Phenomena , Blood Pressure/physiology , Blotting, Western , Cells, Cultured , Connectin/genetics , Echocardiography , Gene Expression Profiling , Linear Models , Mechanotransduction, Cellular , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Microscopy, Immunoelectron , Molecular Sequence Data , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Myocardium/cytology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Sarcomeres/ultrastructure , Sequence Homology, Amino Acid
15.
Arch Biochem Biophys ; 601: 32-41, 2016 07 01.
Article in English | MEDLINE | ID: mdl-26971467

ABSTRACT

Contractile perturbations downstream of Ca(2+) binding to troponin C, the so-called sarcomere-controlled mechanisms, represent the earliest indicators of energy remodeling in the diseased heart [1]. Central to cellular energy "sensing" is the adenosine monophosphate-activated kinase (AMPK) pathway, which is known to directly target myofilament proteins and alter contractility [2-6]. We previously showed that the upstream AMPK kinase, LKB1/MO25/STRAD, impacts myofilament function independently of AMPK [5]. Therefore, we hypothesized that the LKB1 complex associated with myofilament proteins and that alterations in energy signaling modulated targeting or localization of the LKB1 complex to the myofilament. Using an integrated strategy of myofilament mechanics, immunoblot analysis, co-immunoprecipitation, mass spectroscopy, and immunofluorescence, we showed that 1) LKB1 and MO25 associated with myofibrillar proteins, 2) cellular energy stress re-distributed AMPK/LKB1 complex proteins within the sarcomere, and 3) the LKB1 complex localized to the Z-Disk and interacted with cytoskeletal and energy-regulating proteins, including vinculin and ATP Synthase (Complex V). These data represent a novel role for LKB1 complex proteins in myofilament function and myocellular "energy" sensing in the heart.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Myocytes, Cardiac/metabolism , Myofibrils/metabolism , Protein Serine-Threonine Kinases/physiology , Troponin C/metabolism , AMP-Activated Protein Kinase Kinases , Animals , Calcium/metabolism , Cytoskeleton/metabolism , Gene Expression Regulation , Male , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence , Muscle Contraction , Rats , Rats, Sprague-Dawley , Sarcomeres/metabolism
16.
J Exp Biol ; 219(Pt 2): 146-52, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26792324

ABSTRACT

Efficient muscle contraction in skeletal muscle is predicated on the regulation of actin filament lengths. In one long-standing model that was prominent for decades, the giant protein nebulin was proposed to function as a 'molecular ruler' to specify the lengths of the thin filaments. This theory was questioned by many observations, including experiments in which the length of nebulin was manipulated in skeletal myocytes; this approach revealed that nebulin functions to stabilize filamentous actin, allowing thin filaments to reach mature lengths. In addition, more recent data, mostly from in vivo models and identification of new interacting partners, have provided evidence that nebulin is not merely a structural protein. Nebulin plays a role in numerous cellular processes including regulation of muscle contraction, Z-disc formation, and myofibril organization and assembly.


Subject(s)
Muscle Proteins/metabolism , Actin Cytoskeleton/metabolism , Animals , Disease , Humans , Muscle Proteins/chemistry , Muscle Proteins/genetics , Mutation/genetics , Protein Binding
17.
Am J Physiol Lung Cell Mol Physiol ; 308(10): L1025-38, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25795725

ABSTRACT

Paxillin is phosphorylated at multiple residues; however, the role of tyrosine phosphorylation of paxillin in endothelial barrier dysfunction and acute lung injury (ALI) remains unclear. We used siRNA and site-specific nonphosphorylable mutants of paxillin to abrogate the function of paxillin to determine its role in lung endothelial permeability and ALI. In vitro, lipopolysaccharide (LPS) challenge of human lung microvascular endothelial cells (HLMVECs) resulted in enhanced tyrosine phosphorylation of paxillin at Y31 and Y118 with no significant change in Y181 and significant barrier dysfunction. Knockdown of paxillin with siRNA attenuated LPS-induced endothelial barrier dysfunction and destabilization of VE-cadherin. LPS-induced paxillin phosphorylation at Y31 and Y118 was mediated by c-Abl tyrosine kinase, but not by Src and focal adhesion kinase. c-Abl siRNA significantly reduced LPS-induced endothelial barrier dysfunction. Transfection of HLMVECs with paxillin Y31F, Y118F, and Y31/118F double mutants mitigated LPS-induced barrier dysfunction and VE-cadherin destabilization. In vivo, the c-Abl inhibitor AG957 attenuated LPS-induced pulmonary permeability in mice. Together, these results suggest that c-Abl mediated tyrosine phosphorylation of paxillin at Y31 and Y118 regulates LPS-mediated pulmonary vascular permeability and injury.


Subject(s)
Acute Lung Injury/metabolism , Endothelium, Vascular/metabolism , Lipopolysaccharides/toxicity , Paxillin/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Acute Lung Injury/pathology , Animals , Antigens, CD , Cadherins , Capillary Permeability/drug effects , Capillary Permeability/genetics , Cells, Cultured , Endothelium, Vascular/pathology , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/metabolism , Gene Knockdown Techniques , Humans , Male , Mice , Paxillin/genetics , Phosphorylation/genetics , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Proto-Oncogene Proteins c-abl/genetics , Tyrphostins/pharmacology
18.
FASEB J ; 28(9): 3987-95, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24891520

ABSTRACT

Tropomodulin1 (Tmod1) is an actin-capping protein that plays an important role in actin filament pointed-end dynamics and length in striated muscle. No mechanisms have been identified to explain how Tmod1's functional properties are regulated. The purpose of this investigation was to explore the functional significance of the phosphorylation of Tmod1 at previously identified Thr54. Rat cardiomyocytes were assessed for phosphorylation of Tmod1 using Pro-Q Diamond staining and (32)P labeling. Green fluorescent protein-tagged phosphorylation-mimic (T54E) and phosphorylation-deficient (T54A) versions of Tmod1 were expressed in cultured cardiomyocytes, and the ability of these mutants to assemble and restrict actin lengths was observed. We report for the first time that Tmod1 is phosphorylated endogenously in cardiomyocytes, and phosphorylation at Thr54 causes a significant reduction in the ability of Tmod1 to assemble to the pointed end compared with that of the wild type (WT; 48 vs. 78%, respectively). In addition, overexpression of Tmod1-T54E restricts actin filament lengths by only ∼3%, whereas Tmod1-WT restricts the lengths significantly by ∼8%. Finally, Tmod1-T54E altered the actin filament-capping activity in polymerization assays. Taken together, our data suggest that pointed-end assembly and Tmod1's thin filament length regulatory function are regulated by its phosphorylation state.


Subject(s)
Actin Cytoskeleton/chemistry , Actin Cytoskeleton/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Peptide Fragments/metabolism , Tropomodulin/metabolism , Animals , Animals, Newborn , Blotting, Western , Cells, Cultured , Immunoprecipitation , Mutagenesis, Site-Directed , Mutation/genetics , Myocardium/cytology , Myocytes, Cardiac/cytology , Phosphorylation , Rats , Tropomodulin/genetics
19.
Methods ; 66(3): 370-9, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-23806641

ABSTRACT

Mutations in several sarcomeric proteins have been linked to various human myopathies. Therefore, having an in vivo developmental model available that develops quickly and efficiently is key for investigators to elucidate the critical steps, components and signaling pathways involved in building a myofibril; this is the pivotal foundation for deciphering disease mechanisms as well as the development of myopathy-related therapeutics. Although striated muscle cell culture studies have been extremely informative in providing clues to both the distribution and functions of sarcomeric proteins, myocytes in vivo develop in an irreproducible 3D environment. Xenopus laevis (frog) embryos are cost effective, compliant to protein level manipulations and develop relatively quickly (⩽ a week) in a petri dish, thus providing a powerful system for de novo myofibrillogenesis studies. Although fluorophore-conjugated phalloidin labeling is the gold standard approach for investigating actin-thin filament architecture, it is well documented that phalloidin-labeling can be challenging and inconsistent within Xenopus embryos. Therefore we highlight several techniques that can be utilized to preserve both antibody and fluorophore-conjugated phalloidin labeling within Xenopus embryos for high-resolution fluorescence microscopy.


Subject(s)
Microscopy, Fluorescence/methods , Sarcomeres/metabolism , Xenopus Proteins/analysis , Animals , Cryoultramicrotomy , Muscle Development , Phalloidine/analysis , Tissue Fixation , Xenopus laevis
20.
J Biol Chem ; 288(7): 4899-907, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23271735

ABSTRACT

Tropomodulin (Tmod) is an actin-capping protein that binds to the two tropomyosins (TM) at the pointed end of the actin filament to prevent further actin polymerization and depolymerization. Therefore, understanding the role of Tmod is very important when studying actin filament dependent processes such as muscle contraction and intracellular transport. The capping ability of Tmod is highly influenced by TM and is 1000-fold greater in the presence of TM. There are four Tmod isoforms (Tmod1-4), three of which, Tmod1, Tmod3, and Tmod4, are expressed in skeletal muscles. The affinity of Tmod1 to skeletal striated TM (stTM) is higher than that of Tmod3 and Tmod4 to stTM. In this study, we tested mutations in the TM-binding sites of Tmod1, using circular dichroism (CD) and prediction analysis (PONDR). The mutations R11K, D12N, and Q144K were chosen because they decreased the affinity of Tmod1 to stTM, making it similar to that of affinity of Tmod3 and Tmod4 to stTM. Significant reduction of inhibition of actin pointed-end polymerization in the presence of stTM was shown for Tmod1 (R11K/D12N/Q144K) as compared with WT Tmod1. When GFP-Tmod1 and mutants were expressed in primary chicken skeletal myocytes, decreased assembly of Tmod1 mutants was revealed. This indicates a direct correlation between TM-binding and the actin-capping abilities of Tmod. Our data confirmed the hypothesis that assembly of Tmod at the pointed-end of the actin filament depends on its TM-binding affinity.


Subject(s)
Gene Expression Regulation , Muscle Cells/cytology , Muscle, Skeletal/cytology , Tropomodulin/chemistry , Tropomodulin/genetics , Tropomyosin/chemistry , Actin Cytoskeleton/chemistry , Actins/chemistry , Amino Acid Sequence , Animals , Binding Sites , Chickens , Circular Dichroism , Mice , Microscopy, Fluorescence/methods , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Protein Binding , Protein Interaction Mapping/methods , Protein Isoforms , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL