Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Sci Adv ; 10(11): eadk7160, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38489358

ABSTRACT

During development, cells make switch-like decisions to activate new gene programs specifying cell lineage. The mechanisms underlying these decisive choices remain unclear. Here, we show that the cardiovascular transcriptional coactivator myocardin (MYOCD) activates cell identity genes by concentration-dependent and switch-like formation of transcriptional condensates. MYOCD forms such condensates and activates cell identity genes at critical concentration thresholds achieved during smooth muscle cell and cardiomyocyte differentiation. The carboxyl-terminal disordered region of MYOCD is necessary and sufficient for condensate formation. Disrupting this region's ability to form condensates disrupts gene activation and smooth muscle cell reprogramming. Rescuing condensate formation by replacing this region with disordered regions from functionally unrelated proteins rescues gene activation and smooth muscle cell reprogramming. Our findings demonstrate that MYOCD condensate formation is required for gene activation during cardiovascular differentiation. We propose that the formation of transcriptional condensates at critical concentrations of cell type-specific regulators provides a molecular switch underlying the activation of key cell identity genes during development.


Subject(s)
Myocytes, Smooth Muscle , Transcription Factors , Cell Lineage/genetics , Cell Differentiation/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Myocytes, Smooth Muscle/metabolism , Transcriptional Activation
2.
Cardiovasc Res ; 120(1): 56-68, 2024 02 27.
Article in English | MEDLINE | ID: mdl-37890031

ABSTRACT

AIMS: RNA binding proteins play essential roles in mediating RNA splicing and are key post-transcriptional regulators in the heart. Our recent study demonstrated that RBPMS (RNA binding protein with multiple splicing) is crucial for cardiac development through modulating mRNA splicing, but little is known about its functions in the adult heart. In this study, we aim to characterize the post-natal cardiac function of Rbpms and its mechanism of action. METHODS AND RESULTS: We generated a cardiac-specific knockout mouse line and found that cardiac-specific loss of Rbpms caused severe cardiomyocyte contractile defects, leading to dilated cardiomyopathy and early lethality in adult mice. We showed by proximity-dependent biotin identification assay and mass spectrometry that RBPMS associates with spliceosome factors and other RNA binding proteins, such as RBM20, that are important in cardiac function. We performed paired-end RNA sequencing and RT-PCR and found that RBPMS regulates mRNA alternative splicing of genes associated with sarcomere structure and function, such as Ttn, Pdlim5, and Nexn, generating new protein isoforms. Using a minigene splicing reporter assay, we determined that RBPMS regulates target gene splicing through recognizing tandem intronic CAC motifs. We also showed that RBPMS knockdown in human induced pluripotent stem cell-derived cardiomyocytes impaired cardiomyocyte contraction. CONCLUSION: This study identifies RBPMS as an important regulator of cardiomyocyte contraction and cardiac function by modulating sarcomeric gene alternative splicing.


Subject(s)
Alternative Splicing , Induced Pluripotent Stem Cells , Animals , Humans , Mice , Connectin/metabolism , Induced Pluripotent Stem Cells/metabolism , Mice, Knockout , Myocytes, Cardiac/metabolism , RNA/metabolism , RNA Splicing , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
3.
Dev Cell ; 58(24): 2867-2880.e7, 2023 Dec 18.
Article in English | MEDLINE | ID: mdl-37972593

ABSTRACT

Cardiomyocytes are highly metabolic cells responsible for generating the contractile force in the heart. During fetal development and regeneration, these cells actively divide but lose their proliferative activity in adulthood. The mechanisms that coordinate their metabolism and proliferation are not fully understood. Here, we study the role of the transcription factor NFYa in developing mouse hearts. Loss of NFYa alters cardiomyocyte composition, causing a decrease in immature regenerative cells and an increase in trabecular and mature cardiomyocytes, as identified by spatial and single-cell transcriptome analyses. NFYa-deleted cardiomyocytes exhibited reduced proliferation and impaired mitochondrial metabolism, leading to cardiac growth defects and embryonic death. NFYa, interacting with cofactor SP2, activates genes linking metabolism and proliferation at the transcription level. Our study identifies a nodal role of NFYa in regulating prenatal cardiac growth and a previously unrecognized transcriptional control mechanism of heart metabolism, highlighting the importance of mitochondrial metabolism during heart development and regeneration.


Subject(s)
Myocytes, Cardiac , Transcription Factors , Animals , Mice , Cell Proliferation/physiology , Fetal Development , Fetal Heart/metabolism , Heart/physiology , Myocytes, Cardiac/metabolism , Transcription Factors/metabolism
4.
Nat Commun ; 14(1): 4333, 2023 07 19.
Article in English | MEDLINE | ID: mdl-37468485

ABSTRACT

Skeletal muscle fibers express distinct gene programs during development and maturation, but the underlying gene regulatory networks that confer stage-specific myofiber properties remain unknown. To decipher these distinctive gene programs and how they respond to neural activity, we generated a combined multi-omic single-nucleus RNA-seq and ATAC-seq atlas of mouse skeletal muscle development at multiple stages of embryonic, fetal, and postnatal life. We found that Myogenin, Klf5, and Tead4 form a transcriptional complex that synergistically activates the expression of muscle genes in developing myofibers. During myofiber maturation, the transcription factor Maf acts as a transcriptional switch to activate the mature fast muscle gene program. In skeletal muscles of mutant mice lacking voltage-gated L-type Ca2+ channels (Cav1.1), Maf expression and myofiber maturation are impaired. These findings provide a transcriptional atlas of muscle development and reveal genetic links between myofiber formation, maturation, and contraction.


Subject(s)
Muscle Fibers, Skeletal , Muscle, Skeletal , Mice , Animals , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Gene Expression Regulation , Transcription Factors/metabolism , Cell Differentiation
5.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Article in English | MEDLINE | ID: mdl-35101990

ABSTRACT

Emerging evidence indicates that a subset of RNA molecules annotated as noncoding contain short open reading frames that code for small functional proteins called microproteins, which have largely been overlooked due to their small size. To search for cardiac-expressed microproteins, we used a comparative genomics approach and identified mitolamban (Mtlbn) as a highly conserved 47-amino acid transmembrane protein that is abundantly expressed in the heart. Mtlbn localizes specifically to the inner mitochondrial membrane where it interacts with subunits of complex III of the electron transport chain and with mitochondrial respiratory supercomplexes. Genetic deletion of Mtlbn in mice altered complex III assembly dynamics and reduced complex III activity. Unbiased metabolomic analysis of heart tissue from Mtlbn knockout mice further revealed an altered metabolite profile consistent with deficiencies in complex III activity. Cardiac-specific Mtlbn overexpression in transgenic (TG) mice induced cardiomyopathy with histological, biochemical, and ultrastructural pathologic features that contributed to premature death. Metabolomic analysis and biochemical studies indicated that hearts from Mtlbn TG mice exhibited increased oxidative stress and mitochondrial dysfunction. These findings reveal Mtlbn as a cardiac-expressed inner mitochondrial membrane microprotein that contributes to mitochondrial electron transport chain activity through direct association with complex III and the regulation of its assembly and function.


Subject(s)
Cardiomyopathies/metabolism , Electron Transport Complex III/metabolism , Membrane Proteins/metabolism , Mitochondria, Heart/metabolism , Mitochondrial Proteins/metabolism , Myocardium/metabolism , Animals , Cardiomyopathies/genetics , Cells, Cultured , Electron Transport Complex III/genetics , Membrane Proteins/genetics , Mice , Mice, Knockout , Mitochondria, Heart/genetics , Mitochondrial Proteins/genetics , Organ Specificity
6.
Proc Natl Acad Sci U S A ; 118(23)2021 06 08.
Article in English | MEDLINE | ID: mdl-34088848

ABSTRACT

Homeothermic vertebrates produce heat in cold environments through thermogenesis, in which brown adipose tissue (BAT) increases mitochondrial oxidation along with uncoupling of the electron transport chain and activation of uncoupling protein 1 (UCP1). Although the transcription factors regulating the expression of UCP1 and nutrient oxidation genes have been extensively studied, only a few other proteins essential for BAT function have been identified. We describe the discovery of FAM195A, a BAT-enriched RNA binding protein, which is required for cold-dependent thermogenesis in mice. FAM195A knockout (KO) mice display whitening of BAT and an inability to thermoregulate. In BAT of FAM195A KO mice, enzymes involved in branched-chain amino acid (BCAA) metabolism are down-regulated, impairing their response to cold. Knockdown of FAM195A in brown adipocytes in vitro also impairs expression of leucine oxidation enzymes, revealing FAM195A to be a regulator of BCAA metabolism and a potential target for metabolic disorders.


Subject(s)
Adipocytes, Brown , Adipose Tissue, Brown , Cold Temperature , Intracellular Signaling Peptides and Proteins/metabolism , Thermogenesis , Amino Acids, Branched-Chain/genetics , Amino Acids, Branched-Chain/metabolism , Animals , Cell Line, Transformed , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Knockout
7.
Genes Dev ; 35(11-12): 835-840, 2021 06.
Article in English | MEDLINE | ID: mdl-33985971

ABSTRACT

Myocardin, a potent coactivator of serum response factor (SRF), competes with ternary complex factor (TCF) proteins for SRF binding to balance opposing mitogenic and myogenic gene programs in cardiac and smooth muscle. Here we identify a cardiac lncRNA transcribed adjacent to myocardin, named CARDINAL, which antagonizes SRF-dependent mitogenic gene transcription in the heart. CARDINAL-deficient mice show ectopic TCF/SRF-dependent mitogenic gene expression and decreased cardiac contractility in response to age and ischemic stress. CARDINAL forms a nuclear complex with SRF and inhibits TCF-mediated transactivation of the promitogenic gene c-fos, suggesting CARDINAL functions as an RNA cofactor for SRF in the heart.


Subject(s)
Gene Expression Regulation/genetics , Heart/physiology , Nuclear Proteins/metabolism , RNA, Long Noncoding/metabolism , Serum Response Factor/metabolism , Trans-Activators/metabolism , Age Factors , Animals , Disease Models, Animal , Gene Deletion , MEF2 Transcription Factors/metabolism , Mice , Mice, Inbred C57BL , Myocardial Contraction/genetics , Myocardial Infarction/genetics , Myocardial Infarction/physiopathology , Nuclear Proteins/genetics , RNA, Long Noncoding/genetics , Serum Response Factor/genetics , Trans-Activators/genetics , Transcriptional Activation
8.
Nat Cell Biol ; 23(5): 467-475, 2021 05.
Article in English | MEDLINE | ID: mdl-33941892

ABSTRACT

Direct cardiac reprogramming of fibroblasts to cardiomyocytes presents an attractive therapeutic strategy to restore cardiac function following injury. Cardiac reprogramming was initially achieved through overexpression of the transcription factors Gata4, Mef2c and Tbx5; later, Hand2 and Akt1 were found to further enhance this process1-5. Yet, staunch epigenetic barriers severely limit the ability of these cocktails to reprogramme adult fibroblasts6,7. We undertook a screen of mammalian gene regulatory factors to discover novel regulators of cardiac reprogramming in adult fibroblasts and identified the histone reader PHF7 as the most potent activating factor8. Mechanistically, PHF7 localizes to cardiac super enhancers in fibroblasts, and through cooperation with the SWI/SNF complex, it increases chromatin accessibility and transcription factor binding at these sites. Furthermore, PHF7 recruits cardiac transcription factors to activate a positive transcriptional autoregulatory circuit in reprogramming. Importantly, PHF7 achieves efficient reprogramming in the absence of Gata4. Here, we highlight the underexplored necessity of cardiac epigenetic readers, such as PHF7, in harnessing chromatin remodelling and transcriptional complexes to overcome critical barriers to direct cardiac reprogramming.


Subject(s)
GATA4 Transcription Factor/metabolism , Histones/metabolism , Signal Transduction/physiology , Ubiquitin-Protein Ligases/metabolism , Animals , Cellular Reprogramming , Fibroblasts/metabolism , GATA4 Transcription Factor/genetics , Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Regulatory Sequences, Nucleic Acid/physiology , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/genetics
10.
Elife ; 72018 10 09.
Article in English | MEDLINE | ID: mdl-30299255

ABSTRACT

Calcium (Ca2+) dysregulation is a hallmark of heart failure and is characterized by impaired Ca2+ sequestration into the sarcoplasmic reticulum (SR) by the SR-Ca2+-ATPase (SERCA). We recently discovered a micropeptide named DWORF (DWarf Open Reading Frame) that enhances SERCA activity by displacing phospholamban (PLN), a potent SERCA inhibitor. Here we show that DWORF has a higher apparent binding affinity for SERCA than PLN and that DWORF overexpression mitigates the contractile dysfunction associated with PLN overexpression, substantiating its role as a potent activator of SERCA. Additionally, using a well-characterized mouse model of dilated cardiomyopathy (DCM) due to genetic deletion of the muscle-specific LIM domain protein (MLP), we show that DWORF overexpression restores cardiac function and prevents the pathological remodeling and Ca2+ dysregulation classically exhibited by MLP knockout mice. Our results establish DWORF as a potent activator of SERCA within the heart and as an attractive candidate for a heart failure therapeutic.


Subject(s)
Calcium-Binding Proteins/metabolism , Cardiomyopathy, Dilated/physiopathology , Myocardial Contraction/drug effects , Peptides/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Animals , Disease Models, Animal , Gene Knockout Techniques , Heart Failure/prevention & control , LIM Domain Proteins/deficiency , Mice, Knockout , Muscle Proteins/deficiency
11.
Proc Natl Acad Sci U S A ; 115(36): E8430-E8439, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30127033

ABSTRACT

Twist transcription factors function as ancestral regulators of mesodermal cell fates in organisms ranging from Drosophila to mammals. Through lineage tracing of Twist2 (Tw2)-expressing cells with tamoxifen-inducible Tw2-CreERT2 and tdTomato (tdTO) reporter mice, we discovered a unique cell population that progressively contributes to cardiomyocytes (CMs), endothelial cells, and fibroblasts in the adult heart. Clonal analysis confirmed the ability of Tw2-derived tdTO+ (Tw2-tdTO+) cells to form CMs in vitro. Within the adult heart, Tw2-tdTO+ CMs accounted for ∼13% of total CMs, the majority of which resulted from fusion of Tw2-tdTO+ cells with existing CMs. Tw2-tdTO+ cells also contribute to cardiac remodeling after injury. We conclude that Tw2-tdTO+ cells participate in lifelong maintenance of cardiac function, at least in part through de novo formation of CMs and fusion with preexisting CMs, as well as in the genesis of other cellular components of the adult heart.


Subject(s)
Multipotent Stem Cells/metabolism , Myocardium/metabolism , Repressor Proteins/biosynthesis , Twist-Related Protein 1/biosynthesis , Animals , Drosophila melanogaster , Endothelial Cells/cytology , Endothelial Cells/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Mice , Mice, Transgenic , Multipotent Stem Cells/cytology , Myocardium/cytology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Repressor Proteins/genetics , Twist-Related Protein 1/genetics
12.
Cell Rep ; 23(13): 3701-3709, 2018 06 26.
Article in English | MEDLINE | ID: mdl-29949755

ABSTRACT

Micropeptide regulator of ß-oxidation (MOXI) is a conserved muscle-enriched protein encoded by an RNA transcript misannotated as non-coding. MOXI localizes to the inner mitochondrial membrane where it associates with the mitochondrial trifunctional protein, an enzyme complex that plays a critical role in fatty acid ß-oxidation. Isolated heart and skeletal muscle mitochondria from MOXI knockout mice exhibit a diminished ability to metabolize fatty acids, while transgenic MOXI overexpression leads to enhanced ß-oxidation. Additionally, hearts from MOXI knockout mice preferentially oxidize carbohydrates over fatty acids in an isolated perfused heart system compared to wild-type (WT) animals. MOXI knockout mice also exhibit a profound reduction in exercise capacity, highlighting the role of MOXI in metabolic control. The functional characterization of MOXI provides insight into the regulation of mitochondrial metabolism and energy homeostasis and underscores the regulatory potential of additional micropeptides that have yet to be identified.


Subject(s)
Fatty Acids/metabolism , Mitochondria, Muscle/metabolism , Mitochondrial Proteins/genetics , Amino Acid Sequence , Animals , Fatty Acids/chemistry , Humans , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mitochondria, Heart/metabolism , Mitochondrial Proteins/metabolism , Oxidation-Reduction , Sequence Alignment
13.
J Biol Chem ; 293(23): 8802-8811, 2018 06 08.
Article in English | MEDLINE | ID: mdl-29653945

ABSTRACT

Control of protein homeostasis is an essential cellular process that, when perturbed, can result in the deregulation or toxic accumulation of proteins. Owing to constant mechanical stress, striated muscle proteins are particularly prone to wear and tear and require several protein quality-control mechanisms to coordinate protein turnover and removal of damaged proteins. Kelch-like proteins, substrate adapters for the Cullin-3 (Cul3)-RING ligase (CRL3) complex, are emerging as critical regulators of striated muscle development and function, highlighting the importance of Cul3-mediated proteostasis in muscle function. To explore the role of Cul3-mediated proteostasis in striated muscle, here we deleted Cul3 specifically in either skeletal muscle (SkM-Cul3 KO) or cardiomyocytes (CM-Cul3 KO) of mice. The loss of Cul3 caused neonatal lethality and dramatic alterations in the proteome, which were unique to each striated muscle type. Many of the proteins whose expression was significantly changed in the SkM-Cul3 KO were components of the extracellular matrix and sarcomere, whereas proteins altered in the CM-Cul3 KO were involved in metabolism. These findings highlight the requirement for striated muscle-specific CRL3 activity and indicate how the CRL3 complex can control different nodes of protein interaction networks in different types of striated muscle. Further identification of Cul3 substrates, and how these substrates are targeted, may reveal therapeutic targets and treatment regimens for striated muscle diseases.


Subject(s)
Cullin Proteins/genetics , Gene Deletion , Muscle, Striated/pathology , Myocytes, Cardiac/pathology , Animals , Cells, Cultured , Cullin Proteins/metabolism , Gene Expression Regulation, Developmental , Metabolome , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Striated/embryology , Muscle, Striated/metabolism , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Protein Interaction Maps
14.
Elife ; 62017 08 09.
Article in English | MEDLINE | ID: mdl-28826497

ABSTRACT

Maintenance of muscle function requires assembly of contractile proteins into highly organized sarcomeres. Mutations in Kelch-like protein 41 (KLHL41) cause nemaline myopathy, a fatal muscle disorder associated with sarcomere disarray. We generated KLHL41 mutant mice, which display lethal disruption of sarcomeres and aberrant expression of muscle structural and contractile proteins, mimicking the hallmarks of the human disease. We show that KLHL41 is poly-ubiquitinated and acts, at least in part, by preventing aggregation and degradation of Nebulin, an essential component of the sarcomere. Furthermore, inhibition of KLHL41 poly-ubiquitination prevents its stabilization of nebulin, suggesting a unique role for ubiquitination in protein stabilization. These findings provide new insights into the molecular etiology of nemaline myopathy and reveal a mechanism whereby KLHL41 stabilizes sarcomeres and maintains muscle function by acting as a molecular chaperone. Similar mechanisms for protein stabilization likely contribute to the actions of other Kelch proteins.


Subject(s)
Cytoskeletal Proteins/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Myopathies, Nemaline/pathology , Sarcomeres/physiology , Ubiquitin/metabolism , Animals , Cytoskeletal Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Contraction , Muscle Proteins/genetics , Muscle, Skeletal/cytology , Mutation , Myopathies, Nemaline/genetics , Myopathies, Nemaline/metabolism , Ubiquitination
15.
Nat Cell Biol ; 19(3): 202-213, 2017 03.
Article in English | MEDLINE | ID: mdl-28218909

ABSTRACT

Skeletal muscle possesses remarkable regenerative potential due to satellite cells, an injury-responsive stem cell population located beneath the muscle basal lamina that expresses Pax7. By lineage tracing of progenitor cells expressing the Twist2 (Tw2) transcription factor in mice, we discovered a myogenic lineage that resides outside the basal lamina of adult skeletal muscle. Tw2+ progenitors are molecularly and anatomically distinct from satellite cells, are highly myogenic in vitro, and can fuse with themselves and with satellite cells. Tw2+ progenitors contribute specifically to type IIb/x myofibres during adulthood and muscle regeneration, and their genetic ablation causes wasting of type IIb myofibres. We show that Tw2 expression maintains progenitor cells in an undifferentiated state that is poised to initiate myogenesis in response to appropriate cues that extinguish Tw2 expression. Tw2-expressing myogenic progenitors represent a previously unrecognized, fibre-type-specific stem cell involved in postnatal muscle growth and regeneration.


Subject(s)
Aging/physiology , Muscle, Skeletal/metabolism , Repressor Proteins/metabolism , Stem Cells/metabolism , Twist-Related Protein 1/metabolism , Animals , Antigens, CD34/metabolism , Atrophy , Cardiotoxins/toxicity , Cell Lineage/drug effects , Cell Separation , Cells, Cultured , Gene Expression Profiling , Mice , Muscle Development/drug effects , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/drug effects , Myosins/metabolism , PAX7 Transcription Factor/metabolism , Regeneration/drug effects , Retroviridae/metabolism , Stem Cells/cytology , Tamoxifen/pharmacology
16.
Sci Signal ; 9(457): ra119, 2016 12 06.
Article in English | MEDLINE | ID: mdl-27923914

ABSTRACT

Micropeptides function as master regulators of calcium-dependent signaling in muscle. Sarco/endoplasmic reticulum Ca2+ ATPase (SERCA), the membrane pump that promotes muscle relaxation by taking up Ca2+ into the sarcoplasmic reticulum, is directly inhibited by three muscle-specific micropeptides: myoregulin (MLN), phospholamban (PLN), and sarcolipin (SLN). The widespread and essential function of SERCA across diverse cell types has raised questions as to how SERCA is regulated in cells that lack MLN, PLN, and SLN. We identified two transmembrane micropeptides, endoregulin (ELN) and another-regulin (ALN), that share key amino acids with their muscle-specific counterparts and function as direct inhibitors of SERCA pump activity. The distribution of transcripts encoding ELN and ALN mirrored that of SERCA isoform-encoding transcripts in nonmuscle cell types. Our findings identify additional members of the SERCA-inhibitory micropeptide family, revealing a conserved mechanism for the control of intracellular Ca2+ dynamics in both muscle and nonmuscle cell types.


Subject(s)
Calcium Signaling/drug effects , Peptides/chemistry , Peptides/pharmacology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Animals , COS Cells , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/pharmacology , Chlorocebus aethiops , Male , Mice , Muscle Proteins/chemistry , Muscle Proteins/pharmacology , Proteolipids/chemistry , Proteolipids/pharmacology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
17.
Proc Natl Acad Sci U S A ; 113(31): E4494-503, 2016 08 02.
Article in English | MEDLINE | ID: mdl-27418600

ABSTRACT

Innervation of skeletal muscle by motor neurons occurs through the neuromuscular junction, a cholinergic synapse essential for normal muscle growth and function. Defects in nerve-muscle signaling cause a variety of neuromuscular disorders with features of ataxia, paralysis, skeletal muscle wasting, and degeneration. Here we show that the nuclear zinc finger protein ZFP106 is highly enriched in skeletal muscle and is required for postnatal maintenance of myofiber innervation by motor neurons. Genetic disruption of Zfp106 in mice results in progressive ataxia and hindlimb paralysis associated with motor neuron degeneration, severe muscle wasting, and premature death by 6 mo of age. We show that ZFP106 is an RNA-binding protein that associates with the core splicing factor RNA binding motif protein 39 (RBM39) and localizes to nuclear speckles adjacent to spliceosomes. Upon inhibition of pre-mRNA synthesis, ZFP106 translocates with other splicing factors to the nucleolus. Muscle and spinal cord of Zfp106 knockout mice displayed a gene expression signature of neuromuscular degeneration. Strikingly, altered splicing of the Nogo (Rtn4) gene locus in skeletal muscle of Zfp106 knockout mice resulted in ectopic expression of NOGO-A, the neurite outgrowth factor that inhibits nerve regeneration and destabilizes neuromuscular junctions. These findings reveal a central role for Zfp106 in the maintenance of nerve-muscle signaling, and highlight the involvement of aberrant RNA processing in neuromuscular disease pathogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Muscle, Skeletal/metabolism , Muscular Atrophy/genetics , Wasting Syndrome/genetics , Adaptor Proteins, Signal Transducing/deficiency , Animals , COS Cells , Chlorocebus aethiops , Disease Models, Animal , Humans , Mice , Mice, Knockout , Mice, Transgenic , Motor Neurons/metabolism , Motor Neurons/pathology , Muscle Denervation , Muscle, Skeletal/innervation , Muscle, Skeletal/pathology , Muscular Atrophy/metabolism , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Wasting Syndrome/metabolism
18.
Development ; 143(15): 2853-61, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27385017

ABSTRACT

Myocardin-related transcription factors (MRTFs) play a central role in the regulation of actin expression and cytoskeletal dynamics. Stimuli that promote actin polymerization allow for shuttling of MRTFs to the nucleus where they activate serum response factor (SRF), a regulator of actin and other cytoskeletal protein genes. SRF is an essential regulator of skeletal muscle differentiation and numerous components of the muscle sarcomere, but the potential involvement of MRTFs in skeletal muscle development has not been examined. We explored the role of MRTFs in muscle development in vivo by generating mutant mice harboring a skeletal muscle-specific deletion of MRTF-B and a global deletion of MRTF-A. These double knockout (dKO) mice were able to form sarcomeres during embryogenesis. However, the sarcomeres were abnormally small and disorganized, causing skeletal muscle hypoplasia and perinatal lethality. Transcriptome analysis demonstrated dramatic dysregulation of actin genes in MRTF dKO mice, highlighting the importance of MRTFs in actin cycling and myofibrillogenesis. MRTFs were also shown to be necessary for the survival of skeletal myoblasts and for the efficient formation of intact myotubes. Our findings reveal a central role for MRTFs in sarcomere formation during skeletal muscle development and point to the potential involvement of these transcriptional co-activators in skeletal myopathies.


Subject(s)
Trans-Activators/metabolism , Transcription Factors/metabolism , Actins/metabolism , Animals , Female , Immunohistochemistry , Male , Mice , Mice, Knockout , Microscopy, Electron , Muscle Development/physiology , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Myoblasts/cytology , Myoblasts/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Serum Response Factor/genetics , Serum Response Factor/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Trans-Activators/genetics , Transcription Factors/genetics
19.
Mol Cell Biol ; 35(4): 649-61, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25487574

ABSTRACT

The cardiac conduction system coordinates electrical activation through a series of interconnected structures, including the atrioventricular node (AVN), the central connection point that delays impulse propagation to optimize cardiac performance. Although recent studies have uncovered important molecular details of AVN formation, relatively little is known about the transcriptional mechanisms that regulate AV delay, the primary function of the mature AVN. We identify here MyoR as a novel transcription factor expressed in Cx30.2(+) cells of the AVN. We show that MyoR specifically inhibits a Cx30.2 enhancer required for AVN-specific gene expression. Furthermore, we demonstrate that MyoR interacts directly with Gata4 to mediate transcriptional repression. Our studies reveal that MyoR contains two nonequivalent repression domains. While the MyoR C-terminal repression domain inhibits transcription in a context-dependent manner, the N-terminal repression domain can function in a heterologous context to convert the Hand2 activator into a repressor. In addition, we show that genetic deletion of MyoR in mice increases Cx30.2 expression by 50% and prolongs AV delay by 13%. Taken together, we conclude that MyoR modulates a Gata4-dependent regulatory circuit that establishes proper AV delay, and these findings may have wider implications for the variability of cardiac rhythm observed in the general population.


Subject(s)
Atrioventricular Node/metabolism , GATA4 Transcription Factor/metabolism , Signal Transduction/genetics , Transcription Factors/metabolism , Animals , Atrioventricular Node/cytology , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Binding Sites , COS Cells , Chlorocebus aethiops , Connexins/genetics , Connexins/metabolism , Embryo, Mammalian , Female , GATA4 Transcription Factor/genetics , Gene Expression Regulation , Genes, Reporter , Heart Rate/physiology , Luciferases/genetics , Luciferases/metabolism , Male , Mice , Mice, Transgenic , Protein Binding , Protein Interaction Domains and Motifs , Transcription Factors/genetics , Transcription, Genetic , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
20.
J Clin Invest ; 124(8): 3529-39, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24960163

ABSTRACT

Nemaline myopathy (NM) is a congenital myopathy that can result in lethal muscle dysfunction and is thought to be a disease of the sarcomere thin filament. Recently, several proteins of unknown function have been implicated in NM, but the mechanistic basis of their contribution to disease remains unresolved. Here, we demonstrated that loss of a muscle-specific protein, kelch-like family member 40 (KLHL40), results in a nemaline-like myopathy in mice that closely phenocopies muscle abnormalities observed in KLHL40-deficient patients. We determined that KLHL40 localizes to the sarcomere I band and A band and binds to nebulin (NEB), a protein frequently implicated in NM, as well as a putative thin filament protein, leiomodin 3 (LMOD3). KLHL40 belongs to the BTB-BACK-kelch (BBK) family of proteins, some of which have been shown to promote degradation of their substrates. In contrast, we found that KLHL40 promotes stability of NEB and LMOD3 and blocks LMOD3 ubiquitination. Accordingly, NEB and LMOD3 were reduced in skeletal muscle of both Klhl40-/- mice and KLHL40-deficient patients. Loss of sarcomere thin filament proteins is a frequent cause of NM; therefore, our data that KLHL40 stabilizes NEB and LMOD3 provide a potential basis for the development of NM in KLHL40-deficient patients.


Subject(s)
Muscle Proteins/deficiency , Myopathies, Nemaline/etiology , Myopathies, Nemaline/metabolism , Animals , Animals, Newborn , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Proteins/chemistry , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myopathies, Nemaline/pathology , Protein Interaction Domains and Motifs , Protein Stability , Proteolysis , Sarcomeres/metabolism , Sarcomeres/pathology , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL
...