Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Exp Physiol ; 109(4): 549-561, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38461483

ABSTRACT

Changes in myonuclear architecture and positioning are associated with exercise adaptations and ageing. However, data on the positioning and number of myonuclei following exercise are inconsistent. Additionally, whether myonuclear domains (MNDs; i.e., the theoretical volume of cytoplasm within which a myonucleus is responsible for transcribing DNA) and myonuclear positioning are altered with age remains unclear. The aim of this investigation was to investigate relationships between age and activity status and myonuclear domains and positioning. Vastus lateralis muscle biopsies from younger endurance-trained (YT) and older endurance-trained (OT) individuals were compared with age-matched untrained counterparts (YU and OU; OU samples were acquired during surgical operation). Serial, optical z-slices were acquired throughout isolated muscle fibres and analysed to give three-dimensional coordinates for myonuclei and muscle fibre dimensions. The mean cross-sectional area (CSA) of muscle fibres from OU individuals was 33%-53% smaller compared with the other groups. The number of nuclei relative to fibre CSA was 90% greater in OU compared with YU muscle fibres. Additionally, scaling of MND volume with fibre size was altered in older untrained individuals. The myonuclear arrangement, in contrast, was similar across groups. Fibre CSA and most myonuclear parameters were significantly associated with age in untrained individuals, but not in trained individuals. These data indicate that regular endurance exercise throughout the lifespan might better preserve the size of muscle fibres in older age and maintain the relationship between fibre size and MND volumes. Inactivity, however, might result in reduced muscle fibre size and altered myonuclear parameters.


Subject(s)
Aging , Muscle Fibers, Skeletal , Humans , Aged , Muscle Fibers, Skeletal/physiology , Cell Nucleus , Quadriceps Muscle , Exercise Therapy , Muscle, Skeletal
2.
Cardiovasc Res ; 119(11): 2074-2088, 2023 09 05.
Article in English | MEDLINE | ID: mdl-37067297

ABSTRACT

AIMS: Nuclear envelope integrity is essential for the compartmentalization of the nucleus and cytoplasm. Importantly, mutations in genes encoding nuclear envelope (NE) and associated proteins are the second highest cause of familial dilated cardiomyopathy. One such NE protein that causes cardiomyopathy in humans and affects mouse heart development is Lem2. However, its role in the heart remains poorly understood. METHODS AND RESULTS: We generated mice in which Lem2 was specifically ablated either in embryonic cardiomyocytes (Lem2 cKO) or in adult cardiomyocytes (Lem2 iCKO) and carried out detailed physiological, tissue, and cellular analyses. High-resolution episcopic microscopy was used for three-dimensional reconstructions and detailed morphological analyses. RNA-sequencing and immunofluorescence identified altered pathways and cellular phenotypes, and cardiomyocytes were isolated to interrogate nuclear integrity in more detail. In addition, echocardiography provided a physiological assessment of Lem2 iCKO adult mice. We found that Lem2 was essential for cardiac development, and hearts from Lem2 cKO mice were morphologically and transcriptionally underdeveloped. Lem2 cKO hearts displayed high levels of DNA damage, nuclear rupture, and apoptosis. Crucially, we found that these defects were driven by muscle contraction as they were ameliorated by inhibiting myosin contraction and L-type calcium channels. Conversely, reducing Lem2 levels to ∼45% in adult cardiomyocytes did not lead to overt cardiac dysfunction up to 18 months of age. CONCLUSIONS: Our data suggest that Lem2 is critical for integrity at the nascent NE in foetal hearts, and protects the nucleus from the mechanical forces of muscle contraction. In contrast, the adult heart is not detectably affected by partial Lem2 depletion, perhaps owing to a more established NE and increased adaptation to mechanical stress. Taken together, these data provide insights into mechanisms underlying cardiomyopathy in patients with mutations in Lem2 and cardio-laminopathies in general.


Subject(s)
Nuclear Envelope , Nuclear Proteins , Animals , Humans , Mice , DNA Damage , Heart , Mutation , Myocytes, Cardiac/metabolism , Nuclear Envelope/genetics , Nuclear Envelope/metabolism , Nuclear Proteins/genetics
3.
J Cell Physiol ; 237(1): 696-705, 2022 01.
Article in English | MEDLINE | ID: mdl-34322871

ABSTRACT

The transcriptional demands of skeletal muscle fibres are high and require hundreds of nuclei (myonuclei) to produce specialised contractile machinery and multiple mitochondria along their length. Each myonucleus spatially regulates gene expression in a finite volume of cytoplasm, termed the myonuclear domain (MND), which positively correlates with fibre cross-sectional area (CSA). Endurance training triggers adaptive responses in skeletal muscle, including myonuclear accretion, decreased MND sizes and increased expression of the transcription co-activator peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α). Previous work has shown that overexpression of PGC-1α in skeletal muscle regulates mitochondrial biogenesis, myonuclear accretion and MND volume. However, whether PGC-1α is critical for these processes in adaptation to endurance training remained unclear. To test this, we evaluated myonuclear distribution and organisation in endurance-trained wild-type mice and mice lacking PGC-1α in skeletal muscle (PGC-1α mKO). Here, we show a differential myonuclear accretion response to endurance training that is governed by PGC-1α and is dependent on muscle fibre size. The positive relationship of MND size and muscle fibre CSA trended towards a stronger correlation in PGC-1a mKO versus control after endurance training, suggesting that myonuclear accretion was slightly affected with increasing fibre CSA in PGC-1α mKO. However, in larger fibres, the relationship between MND and CSA was significantly altered in trained versus sedentary PGC-1α mKO, suggesting that PGC-1α is critical for myonuclear accretion in these fibres. Accordingly, there was a negative correlation between the nuclear number and CSA, suggesting that in larger fibres myonuclear numbers fail to scale with CSA. Our findings suggest that PGC-1α is an important contributor to myonuclear accretion following moderate-intensity endurance training. This may contribute to the adaptive response to endurance training by enabling a sufficient rate of transcription of genes required for mitochondrial biogenesis.


Subject(s)
Endurance Training , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Animals , Cell Nucleus/metabolism , Humans , Mice , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Int J Biochem Cell Biol ; 137: 106035, 2021 08.
Article in English | MEDLINE | ID: mdl-34242685

ABSTRACT

The nucleus provides a physical and selective chemical boundary to segregate the genome from the cytoplasm. The contents of the nucleus are surrounded by the nuclear envelope, which acts as a hub of mechanosensation, transducing forces from the external cytoskeleton to the nucleus, thus impacting on nuclear morphology, genome organisation, gene transcription and signalling pathways. Muscle tissues such as the heart are unique in that they actively generate large contractile forces, resulting in a distinctive mechanical environment which impacts nuclear properties, function and mechanosensing. In light of this, mutations that affect the function of the nuclear envelope (collectively known as nuclear envelopathies and laminopathies) disproportionately result in striated muscle diseases, which include dilated and arrhythmogenic cardiomyopathies. Here we review the nucleus and its role in mechanotransduction, as well as associated defects that lead to cardiac dysfunction.


Subject(s)
Cell Nucleus/metabolism , Heart Diseases/pathology , Mechanotransduction, Cellular , Nuclear Proteins/metabolism , Animals , Cell Nucleus/genetics , Heart Diseases/genetics , Heart Diseases/metabolism , Humans , Nuclear Proteins/genetics
5.
Clin Sci (Lond) ; 134(16): 2177-2187, 2020 08 28.
Article in English | MEDLINE | ID: mdl-32844998

ABSTRACT

Skeletal muscle weakness is an important determinant of age-related declines in independence and quality of life but its causes remain unclear. Accelerated ageing syndromes such as Hutchinson-Gilford Progerin Syndrome, caused by mutations in genes encoding nuclear envelope proteins, have been extensively studied to aid our understanding of the normal biological ageing process. Like several other pathologies associated with genetic defects to nuclear envelope proteins including Emery-Dreifuss muscular dystrophy, Limb-Girdle muscular dystrophy and congenital muscular dystrophy, these disorders can lead to severe muscle dysfunction. Here, we first describe the structure and function of nuclear envelope proteins, and then review the mechanisms by which mutations in genes encoding nuclear envelope proteins induce premature ageing diseases and muscle pathologies. In doing so, we highlight the potential importance of such genes in processes leading to skeletal muscle weakness in old age.


Subject(s)
Aging , Membrane Proteins/genetics , Muscle Weakness/genetics , Muscle, Skeletal/metabolism , Mutation , Nuclear Envelope/metabolism , Humans , Lamin Type A/genetics , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Dystrophies/genetics , Nuclear Proteins/genetics
6.
Methods Mol Biol ; 1840: 251-281, 2018.
Article in English | MEDLINE | ID: mdl-30141050

ABSTRACT

The linker of nucleoskeleton and cytoskeleton (LINC) complex mediates intracellular cross talk between the nucleus and the cytoplasm. In striated muscle, the LINC complex provides structural support to the myocyte nucleus and plays an essential role in regulating gene expression and mechanotransduction. A wide range of cardiac and skeletal myopathies have been linked to mutations in LINC complex proteins. Studies utilizing tissue-specific knockout and mutant mouse models have revealed important insights into the roles of the LINC complex in striated muscle. In this chapter, we describe several feasible approaches for generating striated muscle-specific gene knockout and mutant mouse models to study LINC complex protein function in cardiac and skeletal muscle. The experimental procedures used for phenotyping and analysis of LINC complex knockout mice are also described.


Subject(s)
Cytoskeleton/metabolism , Multiprotein Complexes/metabolism , Muscle, Striated/metabolism , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Alleles , Amino Acid Substitution , Animals , Biomarkers , CRISPR-Cas Systems , Echocardiography , Fluorescent Antibody Technique , Gene Targeting , Genotype , Mice , Mice, Knockout , Mice, Transgenic
7.
Biophys Rev ; 10(4): 1033-1051, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29869195

ABSTRACT

The linker of nucleoskeleton and cytoskeleton (LINC) complex couples the nuclear lamina to the cytoskeleton. The LINC complex and its associated proteins play diverse roles in cells, ranging from genome organization, nuclear morphology, gene expression, to mechanical stability. The importance of a functional LINC complex is highlighted by the large number of mutations in genes encoding LINC complex proteins that lead to skeletal and cardiac myopathies. In this review, the structure, function, and interactions between components of the LINC complex will be described. Mutations that are known to cause cardiomyopathy in patients will be discussed alongside their respective mouse models. Furthermore, future challenges for the field and emerging technologies to investigate LINC complex function will be discussed.

8.
Cardiovasc Res ; 114(3): 378-388, 2018 Mar 01.
Article in English | MEDLINE | ID: mdl-29040414

ABSTRACT

AIMS: Luma is a recently discovered, evolutionarily conserved protein expressed in mammalian heart, which is associated with the LInker of Nucleoskeleton and Cytoskeleton (LINC) complex. The LINC complex structurally integrates the nucleus and the cytoplasm and plays a critical role in mechanotransduction across the nuclear envelope. Mutations in several LINC components in both humans and mice result in various cardiomyopathies, implying they play essential, non-redundant roles. A single amino acid substitution of serine 358 to leucine (S358L) in Luma is the unequivocal cause of a distinct form of arrhythmogenic cardiomyopathy. However, the role of Luma in heart has remained obscure. In addition, it also remains to be determined how the S358L mutation in Luma leads to cardiomyopathy. METHODS AND RESULTS: To determine the role of Luma in the heart, we first determined the expression pattern of Luma in mouse heart. Luma was sporadically expressed in cardiomyocytes throughout the heart, but was highly and uniformly expressed in cardiac fibroblasts and vascular smooth muscle cells. We also generated germline null Luma mice and discovered that germline null mutants were viable and exhibited normal cardiac function. Luma null mice also responded normally to pressure overload induced by transverse aortic constriction. In addition, localization and expression of other LINC complex components in both cardiac myocytes and fibroblasts was unaffected by global loss of Luma. Furthermore, we also generated and characterized Luma S358L knock-in mice, which displayed normal cardiac function and morphology. CONCLUSION: Our data suggest that Luma is dispensable for murine cardiac development and function and that the Luma S358L mutation alone may not cause cardiomyopathy in mice.


Subject(s)
Heart/embryology , Membrane Proteins/metabolism , Myocardium/metabolism , Animals , Arrhythmogenic Right Ventricular Dysplasia/genetics , Arrhythmogenic Right Ventricular Dysplasia/metabolism , Cells, Cultured , Cytoskeleton/metabolism , Female , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Genetic Predisposition to Disease , Heart/physiopathology , Humans , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/physiopathology , Male , Mechanotransduction, Cellular , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Morphogenesis , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Smooth Muscle/metabolism , Nuclear Matrix/metabolism
9.
J Clin Invest ; 127(8): 3189-3200, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28737513

ABSTRACT

Defective protein quality control (PQC) systems are implicated in multiple diseases. Molecular chaperones and co-chaperones play a central role in functioning PQC. Constant mechanical and metabolic stress in cardiomyocytes places great demand on the PQC system. Mutation and downregulation of the co-chaperone protein BCL-2-associated athanogene 3 (BAG3) are associated with cardiac myopathy and heart failure, and a BAG3 E455K mutation leads to dilated cardiomyopathy (DCM). However, the role of BAG3 in the heart and the mechanisms by which the E455K mutation leads to DCM remain obscure. Here, we found that cardiac-specific Bag3-KO and E455K-knockin mice developed DCM. Comparable phenotypes in the 2 mutants demonstrated that the E455K mutation resulted in loss of function. Further experiments revealed that the E455K mutation disrupted the interaction between BAG3 and HSP70. In both mutants, decreased levels of small heat shock proteins (sHSPs) were observed, and a subset of proteins required for cardiomyocyte function was enriched in the insoluble fraction. Together, these observations suggest that interaction between BAG3 and HSP70 is essential for BAG3 to stabilize sHSPs and maintain cardiomyocyte protein homeostasis. Our results provide insight into heart failure caused by defects in BAG3 pathways and suggest that increasing BAG3 protein levels may be of therapeutic benefit in heart failure.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins/genetics , Cardiomyopathies/metabolism , Heat-Shock Proteins/metabolism , Mutation , Animals , Cardiomyopathies/genetics , Coculture Techniques , Echocardiography , HSP70 Heat-Shock Proteins/metabolism , Heart Failure/metabolism , Kaplan-Meier Estimate , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Chaperones/metabolism , Myocytes, Cardiac/metabolism , Phenotype
10.
J Cell Biol ; 216(7): 1915-1924, 2017 07 03.
Article in English | MEDLINE | ID: mdl-28533284

ABSTRACT

The position of the nucleus in a cell is controlled by interactions between the linker of nucleoskeleton and cytoskeleton (LINC) complex and the cytoskeleton. Defects in nuclear positioning and abnormal aggregation of nuclei occur in many muscle diseases and correlate with muscle dysfunction. Nesprin 1, which includes multiple isoforms, is an integral component of the LINC complex, critical for nuclear positioning and anchorage in skeletal muscle, and is thought to provide an essential link between nuclei and actin. However, previous studies have yet to identify which isoform is responsible. To elucidate this, we generated a series of nesprin 1 mutant mice. We showed that the actin-binding domains of nesprin 1 were dispensable, whereas nesprin 1α2, which lacks actin-binding domains, was crucial for postnatal viability, nuclear positioning, and skeletal muscle function. Furthermore, we revealed that kinesin 1 was displaced in fibers of nesprin 1α2-knockout mice, suggesting that this interaction may play an important role in positioning of myonuclei and functional skeletal muscle.


Subject(s)
Cell Nucleus/metabolism , Muscle Fibers, Skeletal/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Actins/metabolism , Animals , Binding Sites , Cell Nucleus/pathology , Cytoskeletal Proteins , Genotype , Kinesins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Muscle Fibers, Skeletal/pathology , Mutation , Myofibrils/metabolism , Myofibrils/pathology , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Phenotype , Protein Binding , Protein Interaction Domains and Motifs , Signal Transduction
11.
JCI Insight ; 1(16): e89908, 2016 Oct 06.
Article in English | MEDLINE | ID: mdl-27734035

ABSTRACT

Adipose tissue is a key endocrine organ that governs systemic homeostasis. PPARγ is a master regulator of adipose tissue signaling that plays an essential role in insulin sensitivity, making it an important therapeutic target. The selective PPARγ agonist rosiglitazone (RSG) has been used to treat diabetes. However, adverse cardiovascular effects have seriously hindered its clinical application. Experimental models have revealed that PPARγ activation increases cardiac hypertrophy. RSG stimulates cardiac hypertrophy and oxidative stress in cardiomyocyte-specific PPARγ knockout mice, implying that RSG might stimulate cardiac hypertrophy independently of cardiomyocyte PPARγ. However, candidate cell types responsible for RSG-induced cardiomyocyte hypertrophy remain unexplored. Utilizing cocultures of adipocytes and cardiomyocytes, we found that stimulation of PPARγ signaling in adipocytes increased miR-200a expression and secretion. Delivery of miR-200a in adipocyte-derived exosomes to cardiomyocytes resulted in decreased TSC1 and subsequent mTOR activation, leading to cardiomyocyte hypertrophy. Treatment with an antagomir to miR-200a blunted this hypertrophic response in cardiomyocytes. In vivo, specific ablation of PPARγ in adipocytes was sufficient to blunt hypertrophy induced by RSG treatment. By delineating mechanisms by which RSG elicits cardiac hypertrophy, we have identified pathways that mediate the crosstalk between adipocytes and cardiomyocytes to regulate cardiac remodeling.


Subject(s)
Adipocytes/metabolism , Cardiomegaly/genetics , MicroRNAs/genetics , Myocytes, Cardiac/metabolism , PPAR gamma/metabolism , 3T3 Cells , Animals , Cardiomegaly/chemically induced , Cells, Cultured , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , PPAR gamma/antagonists & inhibitors , PPAR gamma/genetics , Rosiglitazone , TOR Serine-Threonine Kinases/metabolism , Thiazolidinediones/adverse effects , Tuberous Sclerosis Complex 1 Protein , Tumor Suppressor Proteins/metabolism
12.
J Clin Invest ; 125(4): 1708-12, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25705887

ABSTRACT

Arrhythmogenic cardiomyopathy (AC) is associated with mutations in genes encoding intercalated disc proteins and ultimately results in sudden cardiac death. A subset of patients with AC have the autosomal recessive cardiocutaneous disorder Naxos disease, which is caused by a 2-base pair deletion in the plakoglobin-encoding gene JUP that results in a truncated protein with reduced expression. In mice, cardiomyocyte-specific plakoglobin deficiency recapitulates many aspects of human AC, and overexpression of the truncated Naxos-associated plakoglobin also results in an AC-like phenotype; therefore, it is unclear whether Naxos disease results from loss or gain of function consequent to the plakoglobin mutation. Here, we generated 2 knockin mouse models in which endogenous Jup was engineered to express the Naxos-associated form of plakoglobin. In one model, Naxos plakoglobin bypassed the nonsense-mediated mRNA decay pathway, resulting in normal levels of the truncated plakoglobin. Moreover, restoration of Naxos plakoglobin to WT levels resulted in normal heart function. Together, these data indicate that a gain of function in the truncated form of the protein does not underlie the clinical phenotype of patients with Naxos disease and instead suggest that insufficiency of the truncated Naxos plakoglobin accounts for disease manifestation. Moreover, these results suggest that increasing levels of truncated or WT plakoglobin has potential as a therapeutic approach to Naxos disease.


Subject(s)
Arrhythmogenic Right Ventricular Dysplasia/genetics , Desmoplakins/genetics , Hair Diseases/genetics , Keratoderma, Palmoplantar/genetics , Myocytes, Cardiac/pathology , gamma Catenin/physiology , Animals , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/physiopathology , Arrhythmogenic Right Ventricular Dysplasia/pathology , Codon, Nonsense , Fibrosis , Frameshift Mutation , Gene Knock-In Techniques , Genes, Lethal , Hair Diseases/pathology , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Humans , Keratoderma, Palmoplantar/pathology , Mice , Myocardial Contraction , Myocardium/pathology , Myocytes, Cardiac/metabolism , Nonsense Mediated mRNA Decay , Peptide Fragments/physiology , Phenotype , RNA Stability , RNA, Messenger/metabolism , Sequence Deletion , Wnt Signaling Pathway , gamma Catenin/chemistry , gamma Catenin/deficiency , gamma Catenin/genetics
13.
Nat Commun ; 6: 6135, 2015 Jan 22.
Article in English | MEDLINE | ID: mdl-25609142

ABSTRACT

Integrin activation, which is regulated by allosteric changes in receptor conformation, enables cellular responses to the chemical, mechanical and topological features of the extracellular microenvironment. A global view of how activation state converts the molecular composition of the region proximal to integrins into functional readouts is, however, lacking. Here, using conformation-specific monoclonal antibodies, we report the isolation of integrin activation state-dependent complexes and their characterization by mass spectrometry. Quantitative comparisons, integrating network, clustering, pathway and image analyses, define multiple functional protein modules enriched in a conformation-specific manner. Notably, active integrin complexes are specifically enriched for proteins associated with microtubule-based functions. Visualization of microtubules on micropatterned surfaces and live cell imaging demonstrate that active integrins establish an environment that stabilizes microtubules at the cell periphery. These data provide a resource for the interrogation of the global molecular connections that link integrin activation to adhesion signalling.


Subject(s)
Integrins/metabolism , Microtubules/metabolism , Proteomics/methods , Allosteric Site , Antibodies, Monoclonal/chemistry , Cerebral Cortex/metabolism , Cluster Analysis , Dimethylpolysiloxanes/chemistry , Fibroblasts/metabolism , Foreskin/metabolism , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Integrin beta1/metabolism , K562 Cells , Male , Mass Spectrometry , Microscopy, Fluorescence , Protein Binding , Protein Conformation , Proteome , Signal Transduction
14.
J Cell Sci ; 127(Pt 12): 2672-82, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24706950

ABSTRACT

Crosstalk between the microtubule (MT) and actin cytoskeletons is fundamental to many cellular processes including cell polarisation and cell motility. Previous work has shown that members of the growth-arrest-specific 2 (GAS2) family mediate the crosstalk between filamentous actin (F-actin) and MTs, but the molecular basis of this process remained unclear. By using fluorescence microscopy, we demonstrate that three members of this family, GAS2-like 1, GAS2-like 2 and GAS2-like 3 (G2L1, G2L2 and G2L3, also known as GAS2L1, GAS2L2 and GAS2L3, respectively) are differentially involved in mediating the crosstalk between F-actin and MTs. Although all localise to actin and MTs, only the exogenous expression of G2L1 and G2L2 influenced MT stability, dynamics and guidance along actin stress fibres. Biochemical analysis and live-cell imaging revealed that their functions are largely due to the association of these proteins with MT plus-end-binding proteins that bind to SxIP or SxLP motifs located at G2L C-termini. Our findings lead to a model in which end-binding (EB) proteins play a key role in mediating actin-MT crosstalk.


Subject(s)
Actins/metabolism , Microfilament Proteins/physiology , Microtubule-Associated Proteins/physiology , Microtubules/metabolism , Stress Fibers/metabolism , Animals , CHO Cells , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Conserved Sequence , Cricetinae , Cricetulus , Humans , Mice , Microtubule-Associated Proteins/metabolism , NIH 3T3 Cells , Protein Binding , Protein Sorting Signals , Protein Transport
15.
PLoS Genet ; 10(2): e1004114, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24586179

ABSTRACT

Recent interest has focused on the importance of the nucleus and associated nucleoskeleton in regulating changes in cardiac gene expression in response to biomechanical load. Mutations in genes encoding proteins of the inner nuclear membrane and nucleoskeleton, which cause cardiomyopathy, also disrupt expression of a biomechanically responsive gene program. Furthermore, mutations in the outer nuclear membrane protein Nesprin 1 and 2 have been implicated in cardiomyopathy. Here, we identify for the first time a role for the outer nuclear membrane proteins, Nesprin 1 and Nesprin 2, in regulating gene expression in response to biomechanical load. Ablation of both Nesprin 1 and 2 in cardiomyocytes, but neither alone, resulted in early onset cardiomyopathy. Mutant cardiomyocytes exhibited altered nuclear positioning, shape, and chromatin positioning. Loss of Nesprin 1 or 2, or both, led to impairment of gene expression changes in response to biomechanical stimuli. These data suggest a model whereby biomechanical signals are communicated from proteins of the outer nuclear membrane, to the inner nuclear membrane and nucleoskeleton, to result in changes in gene expression required for adaptation of the cardiomyocyte to changes in biomechanical load, and give insights into etiologies underlying cardiomyopathy consequent to mutations in Nesprin 1 and 2.


Subject(s)
Cardiomyopathies/genetics , Myocardium/metabolism , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Animals , Biomechanical Phenomena , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cell Nucleus/metabolism , Cytoskeletal Proteins , Gene Expression Regulation , Humans , Mice , Mutation , Myocytes, Cardiac/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Envelope/genetics , Nuclear Envelope/metabolism , Nuclear Matrix/metabolism , Nuclear Proteins/metabolism
16.
Circ Res ; 114(3): 538-48, 2014 Jan 31.
Article in English | MEDLINE | ID: mdl-24481844

ABSTRACT

The linker of nucleoskeleton and cytoskeleton (LINC) complex, composed of proteins within the inner and the outer nuclear membranes, connects the nuclear lamina to the cytoskeleton. The importance of this complex has been highlighted by the discovery of mutations in genes encoding LINC complex proteins, which cause skeletal or cardiac myopathies. Herein, this review summarizes structure, function, and interactions of major components of the LINC complex, highlights how mutations in these proteins may lead to cardiac disease, and outlines future challenges in the field.


Subject(s)
Cytoskeleton/chemistry , Cytoskeleton/physiology , Heart Diseases/physiopathology , Myocytes, Cardiac/physiology , Nuclear Matrix/chemistry , Nuclear Matrix/physiology , Plakins/chemistry , Plakins/physiology , Animals , Cytoskeleton/pathology , Heart Diseases/pathology , Humans , Myocytes, Cardiac/chemistry , Myocytes, Cardiac/pathology , Nuclear Matrix/pathology
17.
J Biol Chem ; 286(28): 24987-95, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21561867

ABSTRACT

The microtubule (MT) and actin cytoskeletons are fundamental to cell integrity, because they control a host of cellular activities, including cell division, growth, polarization, and migration. Proteins involved in mediating the cross-talk between MT and actin cytoskeletons are key to many cellular processes and play important physiological roles. We identified a new member of the GAS2 family of MT-actin cross-linking proteins, named G2L3 (GAS2-like 3). We show that GAS2-like 3 is widely conserved throughout evolution and is ubiquitously expressed in human tissues. GAS2-like 3 interacts with filamentous actin and MTs via its single calponin homology type 3 domain and C terminus, respectively. Interestingly, the role of the putative MT-binding GAS2-related domain is to modulate the binding of GAS2-like 3 to both filamentous actin and MTs. This is in contrast to GAS2-related domains found in related proteins, where it functions as a MT-binding domain. Furthermore, we show that tubulin acetylation drives GAS2-like 3 localization to MTs and may provide functional insights into the role of GAS2-like 3.


Subject(s)
Actin Cytoskeleton/metabolism , Microfilament Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Actin Cytoskeleton/genetics , Animals , Evolution, Molecular , Gene Expression Regulation/physiology , HEK293 Cells , HeLa Cells , Humans , Mice , Microfilament Proteins/genetics , Microtubule-Associated Proteins/genetics , Microtubules/genetics , NIH 3T3 Cells , Organ Specificity/physiology , Protein Binding , Protein Structure, Tertiary
SELECTION OF CITATIONS
SEARCH DETAIL
...