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.
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.
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.

4.
Commun Biol ; 7(1): 7, 2024 01 02.
Article in English | MEDLINE | ID: mdl-38168645

ABSTRACT

Familial cardiomyopathy in pediatric stages is a poorly understood presentation of heart disease in children that is attributed to pathogenic mutations. Through exome sequencing, we report a homozygous variant in tropomodulin 1 (TMOD1; c.565C>T, p.R189W) in three individuals from two unrelated families with childhood-onset dilated and restrictive cardiomyopathy. To decipher the mechanism of pathogenicity of the R189W mutation in TMOD1, we utilized a wide array of methods, including protein analyses, biochemistry and cultured cardiomyocytes. Structural modeling revealed potential defects in the local folding of TMOD1R189W and its affinity for actin. Cardiomyocytes expressing GFP-TMOD1R189W demonstrated longer thin filaments than GFP-TMOD1wt-expressing cells, resulting in compromised filament length regulation. Furthermore, TMOD1R189W showed weakened activity in capping actin filament pointed ends, providing direct evidence for the variant's effect on actin filament length regulation. Our data indicate that the p.R189W variant in TMOD1 has altered biochemical properties and reveals a unique mechanism for childhood-onset cardiomyopathy.


Subject(s)
Actin Cytoskeleton , Cardiomyopathies , Child , Humans , Actin Cytoskeleton/metabolism , Actins/metabolism , Myocytes, Cardiac/metabolism , Mutation , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Tropomodulin/genetics , Tropomodulin/chemistry , Tropomodulin/metabolism
5.
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
6.
Cells ; 12(11)2023 05 23.
Article in English | MEDLINE | ID: mdl-37296576

ABSTRACT

As an essential component of the sarcomere, actin thin filament stems from the Z-disk extend toward the middle of the sarcomere and overlaps with myosin thick filaments. Elongation of the cardiac thin filament is essential for normal sarcomere maturation and heart function. This process is regulated by the actin-binding proteins Leiomodins (LMODs), among which LMOD2 has recently been identified as a key regulator of thin filament elongation to reach a mature length. Few reports have implicated homozygous loss of function variants of LMOD2 in neonatal dilated cardiomyopathy (DCM) associated with thin filament shortening. We present the fifth case of DCM due to biallelic variants in the LMOD2 gene and the second case with the c.1193G>A (p.W398*) nonsense variant identified by whole-exome sequencing. The proband is a 4-month male infant of Hispanic descent with advanced heart failure. Consistent with previous reports, a myocardial biopsy exhibited remarkably short thin filaments. However, compared to other cases of identical or similar biallelic variants, the patient presented here has an unusually late onset of cardiomyopathy during infancy. Herein, we present the phenotypic and histological features of this variant, confirm the pathogenic impact on protein expression and sarcomere structure, and discuss the current knowledge of LMOD2-related cardiomyopathy.


Subject(s)
Cardiomyopathies , Cardiomyopathy, Dilated , Infant, Newborn , Infant , Male , Humans , Cardiomyopathy, Dilated/genetics , Exome Sequencing , Homozygote , Heart
7.
Protein Sci ; 31(7): e4358, 2022 07.
Article in English | MEDLINE | ID: mdl-35762710

ABSTRACT

A transient increase in Ca2+ concentration in sarcomeres is essential for their proper function. Ca2+ drives striated muscle contraction via binding to the troponin complex of the thin filament to activate its interaction with the myosin thick filament. In addition to the troponin complex, the myosin essential light chain and myosin-binding protein C were also found to be Ca2+ sensitive. However, the effects of Ca2+ on the function of the tropomodulin family proteins involved in regulating thin filament formation have not yet been studied. Leiomodin, a member of the tropomodulin family, is an actin nucleator and thin filament elongator. Using pyrene-actin polymerization assay and transmission electron microscopy, we show that the actin nucleation activity of leiomodin is attenuated by Ca2+ . Using circular dichroism and nuclear magnetic resonance spectroscopy, we demonstrate that the mostly disordered, negatively charged region of leiomodin located between its first two actin-binding sites binds Ca2+ . We propose that Ca2+ binding to leiomodin results in the attenuation of its nucleation activity. Our data provide further evidence regarding the role of Ca2+ as an ultimate regulator of the ensemble of sarcomeric proteins essential for muscle function. SUMMARY STATEMENT: Ca2+ fluctuations in striated muscle sarcomeres modulate contractile activity via binding to several distinct families of sarcomeric proteins. The effects of Ca2+ on the activity of leiomodin-an actin nucleator and thin filament length regulator-have remained unknown. In this study, we demonstrate that Ca2+ binds directly to leiomodin and attenuates its actin nucleating activity. Our data emphasizes the ultimate role of Ca2+ in the regulation of the sarcomeric protein interactions.


Subject(s)
Actins , Tropomodulin , Actin Cytoskeleton , Muscle Contraction , Troponin
8.
Eur J Hum Genet ; 30(4): 450-457, 2022 04.
Article in English | MEDLINE | ID: mdl-35082396

ABSTRACT

Dilated cardiomyopathy (DCM) is characterized by cardiac enlargement and impaired ventricular contractility leading to heart failure. A single report identified variants in leiomodin-2 (LMOD2) as a cause of neonatally-lethal DCM. Here, we describe two siblings with DCM who died shortly after birth due to heart failure. Exome sequencing identified a homozygous LMOD2 variant in both siblings, (GRCh38)chr7:g.123656237G > A; NM_207163.2:c.273 + 1G > A, ablating the donor 5' splice-site of intron-1. Pre-mRNA splicing studies and western blot analysis on cDNA derived from proband cardiac tissue, MyoD-transduced proband skin fibroblasts and HEK293 cells transfected with LMOD2 gene constructs established variant-associated absence of canonically spliced LMOD2 mRNA and full-length LMOD2 protein. Immunostaining of proband heart tissue unveiled abnormally short actin-thin filaments. Our data are consistent with LMOD2 c.273 + 1G > A abolishing/reducing LMOD2 transcript expression by: (1) variant-associated perturbation in initiation of transcription due to ablation of the intron-1 donor; and/or (2) degradation of aberrant LMOD2 transcripts (resulting from use of alternative transcription start-sites or cryptic splice-sites) by nonsense-mediated decay. LMOD2 expression is critical for life and the absence of LMOD2 is associated with thin filament shortening and severe cardiac contractile dysfunction. This study describes the first splice-site variant in LMOD2 and confirms the role of LMOD2 variants in DCM.


Subject(s)
Cardiomyopathy, Dilated , Heart Failure , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Cardiomyopathy, Dilated/genetics , HEK293 Cells , Heart Failure/genetics , Heart Failure/metabolism , Homozygote , Humans , Infant, Newborn
9.
FEBS J ; 289(20): 6119-6131, 2022 10.
Article in English | MEDLINE | ID: mdl-34273242

ABSTRACT

Leiomodin is an important emerging regulator of thin filaments. As novel molecular, cellular, animal model, and human data accumulate, the mechanisms of its action become clearer. Structural studies played a significant part in understanding the functional significance of leiomodin's interacting partners and functional domains. In this review, we present the current state of knowledge on the structural and cellular properties of leiomodin which has led to two proposed mechanisms of its function. Although it is known that leiomodin is essential for life, numerous domains within leiomodin remain unstudied and as such, we outline future directions for investigations that we predict will provide evidence that leiomodin is a multifunctional protein.


Subject(s)
Actins , Tropomodulin , Actin Cytoskeleton/metabolism , Actins/metabolism , Animals , Binding Sites , Humans , Tropomodulin/metabolism , Tropomyosin/chemistry
10.
Stem Cell Reports ; 16(10): 2459-2472, 2021 10 12.
Article in English | MEDLINE | ID: mdl-34525378

ABSTRACT

The pathogenicity of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has been attributed to its ability to enter through the membrane-bound angiotensin-converting enzyme 2 (ACE2) receptor. Therefore, it has been heavily speculated that angiotensin-converting enzyme inhibitor (ACEI) or angiotensin receptor blocker (ARB) therapy may modulate SARS-CoV-2 infection. In this study, exposure of human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) and human endothelial cells (hECs) to SARS-CoV-2 identified significant differences in protein coding genes involved in immunity, viral response, and cardiomyocyte/endothelial structure. Specifically, transcriptome changes were identified in the tumor necrosis factor (TNF), interferon α/ß, and mitogen-activated protein kinase (MAPK) (hPSC-CMs) as well as nuclear factor kappa-B (NF-κB) (hECs) signaling pathways. However, pre-treatment of hPSC-CMs or hECs with two widely prescribed antihypertensive medications, losartan and lisinopril, did not affect the susceptibility of either cell type to SARS-CoV-2 infection. These findings demonstrate the toxic effects of SARS-CoV-2 in hPSC-CMs/hECs and, taken together with newly emerging multicenter trials, suggest that antihypertensive drug treatment alone does not alter SARS-CoV-2 infection.


Subject(s)
Antihypertensive Agents/pharmacology , COVID-19 Drug Treatment , Endothelial Cells/drug effects , Myocytes, Cardiac/drug effects , COVID-19/genetics , Cells, Cultured , Disease Susceptibility , Endothelial Cells/metabolism , Host-Pathogen Interactions/drug effects , Humans , Lisinopril/pharmacology , Losartan/pharmacology , Myocytes, Cardiac/metabolism , SARS-CoV-2/drug effects , SARS-CoV-2/physiology , Transcriptome/drug effects
11.
Trends Cell Biol ; 31(9): 708-711, 2021 09.
Article in English | MEDLINE | ID: mdl-34266732

ABSTRACT

Striated muscle is intricately designed to provide efficient and powerful muscle contractions. Recently, a long sought-after missing component of the thin filament pointed-end machinery was discovered: cyclase-associated protein 2 (CAP2). CAP2 was identified as a crucial contributor to actin polymerization, striated muscle development, and severe muscle disease when mutated.


Subject(s)
Actin Cytoskeleton , Actins , Actins/genetics , Humans , Muscle Contraction , Muscle Development , Muscle, Skeletal
12.
Commun Biol ; 4(1): 365, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33742108

ABSTRACT

The precise assembly of actin-based thin filaments is crucial for muscle contraction. Dysregulation of actin dynamics at thin filament pointed ends results in skeletal and cardiac myopathies. Here, we discovered adenylyl cyclase-associated protein 2 (CAP2) as a unique component of thin filament pointed ends in cardiac muscle. CAP2 has critical functions in cardiomyocytes as it depolymerizes and inhibits actin incorporation into thin filaments. Strikingly distinct from other pointed-end proteins, CAP2's function is not enhanced but inhibited by tropomyosin and it does not directly control thin filament lengths. Furthermore, CAP2 plays an essential role in cardiomyocyte maturation by modulating pre-sarcomeric actin assembly and regulating α-actin composition in mature thin filaments. Identification of CAP2's multifunctional roles provides missing links in our understanding of how thin filament architecture is regulated in striated muscle and it reveals there are additional factors, beyond Tmod1 and Lmod2, that modulate actin dynamics at thin filament pointed ends.


Subject(s)
Actin Cytoskeleton/metabolism , Actins/metabolism , Carrier Proteins/metabolism , Muscle Development , Myocytes, Cardiac/metabolism , Myofibrils/metabolism , Age Factors , Animals , Carrier Proteins/genetics , Cells, Cultured , Chick Embryo , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Rats, Sprague-Dawley , Signal Transduction , Tropomyosin/metabolism
13.
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
14.
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
15.
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
16.
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
17.
ACS Biomater Sci Eng ; 5(8): 3808-3816, 2019 Aug 12.
Article in English | MEDLINE | ID: mdl-33438421

ABSTRACT

Understanding how the mechanical microenvironment affects cardiomyocyte development is crucial to the creation of in vitro models for studying heart physiology and pathophysiology. This knowledge will also facilitate the design of biomaterials and tissue scaffolds utilized in the generation of functional tissue constructs for regenerative medicine and drug screening applications. Here, plasma lithography patterning of elastomeric substrates is exploited for creating microtissues composed of neonatal cardiomyocytes and investigating their attributes in different mechanical microenvironments. Restriction of the cellular outgrowth in line patterns results in cardiomyocytes developing into multicellular clusters and collectively adapting to geometric confinement and substrate stiffness. Immunofluorescence microscopy, video microscopy, and force spectroscopy show that the size and shape of the cardiomyocyte clusters, as well as sarcomere length, fiber alignment, beating amplitude, and beating frequency of the cardiomyocytes, are regulated by the microenvironmental cues. Computational analysis reveals that the mechanical stress at the cluster-substrate interface strongly correlates with the characteristics of the cardiomyocytes. Taken together, our results underscore a collective mechanoadaptation scheme in cardiac development.

18.
Mol Biol Cell ; 30(2): 268-281, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30462572

ABSTRACT

Missense mutations K15N and R21H in striated muscle tropomyosin are linked to dilated cardiomyopathy (DCM) and hypertrophic cardiomyopathy (HCM), respectively. Tropomyosin, together with the troponin complex, regulates muscle contraction and, along with tropomodulin and leiomodin, controls the uniform thin-filament lengths crucial for normal sarcomere structure and function. We used Förster resonance energy transfer to study effects of the tropomyosin mutations on the structure and kinetics of the cardiac troponin core domain associated with the Ca2+-dependent regulation of cardiac thin filaments. We found that the K15N mutation desensitizes thin filaments to Ca2+ and slows the kinetics of structural changes in troponin induced by Ca2+ dissociation from troponin, while the R21H mutation has almost no effect on these parameters. Expression of the K15N mutant in cardiomyocytes decreases leiomodin's thin-filament pointed-end assembly but does not affect tropomodulin's assembly at the pointed end. Our in vitro assays show that the R21H mutation causes a twofold decrease in tropomyosin's affinity for F-actin and affects leiomodin's function. We suggest that the K15N mutation causes DCM by altering Ca2+-dependent thin-filament regulation and that one of the possible HCM-causing mechanisms by the R21H mutation is through alteration of leiomodin's function.


Subject(s)
Actin Cytoskeleton/metabolism , Cardiomyopathies/genetics , Mutation/genetics , Tropomyosin/genetics , Actin Cytoskeleton/ultrastructure , Actins/metabolism , Adenosine Triphosphate/metabolism , Animals , Animals, Newborn , Calcium/metabolism , Humans , Hydrolysis , Mice , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...