Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Vasc Cell ; 7: 3, 2015.
Article in English | MEDLINE | ID: mdl-25914805

ABSTRACT

BACKGROUND: Recent evidence suggests that a majority of RNAs in the genome do not code for proteins. They are located in the sense (S) or antisense (AS) orientation and, to date, the functional significance of these non-coding RNAs (ncRNAs) is poorly understood. Here, we examined the relationship between S and AS transcripts in the regulation of a key angiogenesis gene, Delta-like 4 (Dll4). METHODS: Rapid Amplification of cDNA Ends (RACE) method was used to identify natural antisense transcripts in the Dll4 gene locus in murine and human endothelial cells, referred to as Dll4 Anti-Sense (Dll4-AS). Messenger RNA (mRNA) levels of Dll4 and Dll4-AS were quantified by real-time PCR. The function of Dll4-AS was investigated by overexpression and knocking down of Dll4-AS. RESULTS: Dll4-AS comprises of three isoforms that map proximal to the Dll4 promoter region. Expression patterns of Dll4-AS isoforms vary among different endothelial cell lines, but are always congruent with those of Dll4. A dual promoter element in the Dll4 locus has been identified that controls the expression of both transcripts. Both Dll4-AS and Dll4 are sensitive to cellular density in that higher cellular density favors their expression. Exogenous Dll4 stimuli such as VEGF, FGF and Notch signaling inhibitor altered both DLL4-AS and DLL4 expression suggesting co-regulation of the transcripts. Also, knocking down of Dll4-AS results in down-regulation of Dll4 expression. As a consequence, endothelial cell proliferation and migration increases in vitro, and sprout formation increases. The regulation of Dll4 by Dll4-AS was also conserved in vivo. CONCLUSION: A novel form of non-coding RNA-mediated regulation at the Dll4 locus contributes to vascular developmental processes such as cell proliferation, migration and sprouting.

2.
BMC Biochem ; 15: 27, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25519881

ABSTRACT

BACKGROUND: The mitogen-activated protein kinases (MAPKs) pathway is critical for cellular signaling, and proteins such as phosphatases that regulate this pathway are important for normal tissue development. Based on our previous work on dual specificity phosphatase-5 (DUSP5), and its role in embryonic vascular development and disease, we hypothesized that mutations in DUSP5 will affect its function. RESULTS: In this study, we tested this hypothesis by generating full-length glutathione-S-transferase-tagged DUSP5 and serine 147 proline mutant (S147P) proteins from bacteria. Light scattering analysis, circular dichroism, enzymatic assays and molecular modeling approaches have been performed to extensively characterize the protein form and function. We demonstrate that both proteins are active and, interestingly, the S147P protein is hypoactive as compared to the DUSP5 WT protein in two distinct biochemical substrate assays. Furthermore, due to the novel positioning of the S147P mutation, we utilize computational modeling to reconstruct full-length DUSP5 and S147P to predict a possible mechanism for the reduced activity of S147P. CONCLUSION: Taken together, this is the first evidence of the generation and characterization of an active, full-length, mutant DUSP5 protein which will facilitate future structure-function and drug development-based studies.


Subject(s)
Dual-Specificity Phosphatases/biosynthesis , Amino Acid Sequence , Amino Acid Substitution , Catalytic Domain , Dual-Specificity Phosphatases/chemistry , Dual-Specificity Phosphatases/genetics , Extracellular Signal-Regulated MAP Kinases/chemistry , Humans , Molecular Dynamics Simulation , Molecular Sequence Data , Protein Binding , Protein Biosynthesis
3.
Biol Open ; 4(1): 48-61, 2014 Dec 12.
Article in English | MEDLINE | ID: mdl-25505152

ABSTRACT

In this study, we have identified a novel member of the AMPK family, namely Sucrose non-fermenting related kinase (Snrk), that is responsible for maintaining cardiac metabolism in mammals. SNRK is expressed in the heart, and brain, and in cell types such as endothelial cells, smooth muscle cells and cardiomyocytes (CMs). Snrk knockout (KO) mice display enlarged hearts, and die at postnatal day 0. Microarray analysis of embryonic day 17.5 Snrk hearts, and blood profile of neonates display defect in lipid metabolic pathways. SNRK knockdown CMs showed altered phospho-acetyl-coA carboxylase and phospho-AMPK levels similar to global and endothelial conditional KO mouse. Finally, adult cardiac conditional KO mouse displays severe cardiac functional defects and lethality. Our results suggest that Snrk is essential for maintaining cardiac metabolic homeostasis, and shows an autonomous role for SNRK during mammalian development.

4.
Curr Biol ; 23(5): 430-5, 2013 Mar 04.
Article in English | MEDLINE | ID: mdl-23434281

ABSTRACT

Mechanical tension is an ever-present physiological stimulus essential for the development and homeostasis of locomotory, cardiovascular, respiratory, and urogenital systems. Tension sensing contributes to stem cell differentiation, immune cell recruitment, and tumorigenesis. Yet, how mechanical signals are transduced inside cells remains poorly understood. Here, we identify chaperone-assisted selective autophagy (CASA) as a tension-induced autophagy pathway essential for mechanotransduction in muscle and immune cells. The CASA complex, comprised of the molecular chaperones Hsc70 and HspB8 and the cochaperone BAG3, senses the mechanical unfolding of the actin-crosslinking protein filamin. Together with the chaperone-associated ubiquitin ligase CHIP, the complex initiates the ubiquitin-dependent autophagic sorting of damaged filamin to lysosomes for degradation. Autophagosome formation during CASA depends on an interaction of BAG3 with synaptopodin-2 (SYNPO2). This interaction is mediated by the BAG3 WW domain and facilitates cooperation with an autophagosome membrane fusion complex. BAG3 also utilizes its WW domain to engage in YAP/TAZ signaling. Via this pathway, BAG3 stimulates filamin transcription to maintain actin anchoring and crosslinking under mechanical tension. By integrating tension sensing, autophagosome formation, and transcription regulation during mechanotransduction, the CASA machinery ensures tissue homeostasis and regulates fundamental cellular processes such as adhesion, migration, and proliferation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Autophagy , Mechanotransduction, Cellular , Molecular Chaperones/metabolism , Acyltransferases , Animals , Apoptosis Regulatory Proteins , Humans , Jurkat Cells , Male , Mice , Microfilament Proteins/metabolism , Phosphoproteins/metabolism , Rats , Stress, Mechanical , Transcription Factors/metabolism , YAP-Signaling Proteins
5.
J Muscle Res Cell Motil ; 34(1): 61-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23225103

ABSTRACT

The assembly of striated muscle myofibrils is a multistep process in which a variety of proteins is involved. One of the first and most important steps in myofibrillogenesis is the arrangement of thin myofilaments into ordered I-Z-I brushes, requiring the coordinated activity of numerous actin binding proteins. The early expression of myopodin prior to sarcomeric α-actinin, as well as its binding to actin, α-actinin and filamin indicate an important role for this protein in actin cytoskeleton remodelling with the precise function of myopodin in this process yet remaining to be resolved. While myopodin was previously described as a protein capable of cross-linking actin filaments into thick bundles upon transient transfections, it has remained unclear whether myopodin alone is capable of bundling actin, or if additional proteins are involved. We have therefore investigated the in vitro actin binding properties of myopodin. High speed cosedimentation assays with skeletal muscle actin confirmed direct binding of myopodin to F-actin and showed that this interaction is mediated by at least two independent actin binding sites, found in all myopodin isoforms identified to date. Furthermore, low-speed cosedimentation assays revealed that not only full length myopodin, but also the fragment containing only the second binding site, bundles microfilaments in the absence of accessory proteins. Ultrastructural analysis demonstrated that this bundling activity resembled that of α-actinin. Biochemical experiments revealed that bundling was not achieved by myopodin's ability to dimerize, indicating the presence of two individual F-actin binding sites within the second binding segment. Thus full length myopodin contains at least three F-actin binding sites. These data provide further understanding of the mechanisms by which myopodin contributes to actin reorganization during myofibril assembly.


Subject(s)
Actin Cytoskeleton/metabolism , Actinin/metabolism , Actins/metabolism , Protein Multimerization , Animals , Binding Sites , Chickens/metabolism , Chromatography, Gel , Cloning, Molecular , Electrophoresis, Polyacrylamide Gel , Escherichia coli/genetics , Escherichia coli/metabolism , Humans , Multiprotein Complexes/metabolism , Muscle, Skeletal/metabolism , Protein Binding , Two-Hybrid System Techniques
6.
J Appl Physiol (1985) ; 114(2): 252-61, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23172031

ABSTRACT

The present study examined the role of the dual-specificity protein phosphatase-5 (DUSP-5) in the pressure-induced myogenic responses of organ-cultured cerebral arterial segments. In these studies, we initially compared freshly isolated and organ-cultured cerebral arterial segments with respect to responses to step increases in intravascular pressure, vasodilator and vasoconstrictor stimuli, activities of the large-conductance arterial Ca(2+)-activated K(+) (K(Ca)) single-channel current, and stable protein expression of DUSP-5 enzyme. The results demonstrate maintained pressure-dependent myogenic vasoconstriction, DUSP-5 protein expression, endothelium-dependent and -independent dilations, agonist-induced constriction, and unitary K(Ca) channel conductance in organ-cultured cerebral arterial segments similar to that in freshly isolated cerebral arteries. Furthermore, using a permeabilization transfection technique in organ-cultured cerebral arterial segments, gene-specific small interfering RNA (siRNA) induced knockdown of DUSP-5 mRNA and protein, which were associated with enhanced pressure-dependent cerebral arterial myogenic constriction and increased phosphorylation of PKC-ßII. In addition, siRNA knockdown of DUSP-5 reduced levels of phosphorylated ROCK and ERK1 with no change in the level of phosphorylated ERK2. Pharmacological inhibition of ERK1/2 phosphorylation significantly attenuated pressure-induced myogenic constriction in cerebral arteries. The findings within the present studies illustrate that DUSP-5, native in cerebral arterial muscle cells, appears to regulate signaling of pressure-dependent myogenic cerebral arterial constriction, which is crucial for the maintenance of constant cerebral blood flow to the brain.


Subject(s)
Cerebral Arteries/physiology , Cerebrovascular Circulation/physiology , Dual-Specificity Phosphatases/physiology , Muscle Development/physiology , Muscle, Smooth, Vascular/physiology , Vasoconstriction/physiology , Animals , Brain/physiology , Cells, Cultured , Cerebral Arteries/cytology , Dual-Specificity Phosphatases/drug effects , Dual-Specificity Phosphatases/genetics , Male , Models, Animal , Muscle, Smooth, Vascular/cytology , Patch-Clamp Techniques , Potassium Channels, Calcium-Activated/physiology , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , Regional Blood Flow/physiology , Signal Transduction/physiology
7.
Nat Med ; 18(5): 766-73, 2012 May.
Article in English | MEDLINE | ID: mdl-22466703

ABSTRACT

Alternative splicing has a major role in cardiac adaptive responses, as exemplified by the isoform switch of the sarcomeric protein titin, which adjusts ventricular filling. By positional cloning using a previously characterized rat strain with altered titin mRNA splicing, we identified a loss-of-function mutation in the gene encoding RNA binding motif protein 20 (Rbm20) as the underlying cause of pathological titin isoform expression. The phenotype of Rbm20-deficient rats resembled the pathology seen in individuals with dilated cardiomyopathy caused by RBM20 mutations. Deep sequencing of the human and rat cardiac transcriptome revealed an RBM20-dependent regulation of alternative splicing. In addition to titin (TTN), we identified a set of 30 genes with conserved splicing regulation between humans and rats. This network is enriched for genes that have previously been linked to cardiomyopathy, ion homeostasis and sarcomere biology. Our studies emphasize the key role of post-transcriptional regulation in cardiac function and provide mechanistic insights into the pathogenesis of human heart failure.


Subject(s)
Cardiomyopathy, Dilated/genetics , Muscle Proteins/genetics , Protein Kinases/genetics , RNA Splicing , RNA-Binding Proteins/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Base Sequence , Connectin , Humans , LIM Domain Proteins/genetics , Molecular Sequence Data , Mutation , RNA-Binding Proteins/physiology , Rats , Rats, Inbred BN , Rats, Inbred F344
8.
Eur J Cell Biol ; 89(9): 681-92, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20554076

ABSTRACT

Here we introduce myopodin as a novel filamin C binding partner. Corroborative yeast two-hybrid and biochemical analyses indicate that the central part of myopodin that shows high homology to the closely related protein synaptopodin and that is common to all its currently known or predicted variants interacts with filamin C immunoglobulin-like domains 20-21. A detailed characterization of the previously described interaction between myopodin and alpha-actinin demonstrates for the first time that myopodin contains three independent alpha-actinin-binding sites. Newly developed myopodin-specific antibodies reveal expression at the earliest stages of in vitro differentiation of human skeletal muscle cells preceding the expression of sarcomeric alpha-actinin. Myopodin colocalizes with filamin and alpha-actinin during all stages of muscle development. By contrast, colocalization with its previously identified binding partner zyxin is restricted to early developmental stages. Genetic and cellular analyses of skeletal muscle provided direct evidence for an alternative transcriptional start site in exon three, corroborating the expression of a myopodin variant lacking the PDZ domain encoded by exons 1 and 2 in skeletal muscle. We conclude that myopodin is a multiadapter protein of the sarcomeric Z-disc that links nascent myofibrils to the sarcolemma via zyxin, and might play a role in early assembly and stabilization of the Z-disc. Mutations in FLNC, ACTN2 and several other genes encoding Z-disc-related proteins cause myopathy and cardiomyopathy. Its localization and its association with the myopathy-associated proteins filamin C and alpha-actinin make myopodin an interesting candidate for a muscle disease gene.


Subject(s)
Actinin/metabolism , Contractile Proteins/metabolism , Microfilament Proteins/metabolism , Sarcomeres/metabolism , Adult , Contractile Proteins/genetics , Filamins , Humans , Immunoprecipitation , Microfilament Proteins/genetics , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Two-Hybrid System Techniques
9.
Cardiovasc Res ; 85(4): 739-50, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-19843512

ABSTRACT

AIMS: Xin is a striated muscle-specific F-actin binding protein that has been implicated in cardiomyopathies. In cardiomyocytes, Xin is localized at intercalated discs (IDs). Mice lacking only two of the three Xin isoforms (XinAB(-/-) mice) develop severe cardiac hypertrophy. To further investigate the function of Xin variants in the mammalian heart, we generated XinABC(-/-) mice deficient in all Xin isoforms. METHODS AND RESULTS: XinABC(-/-) mice showed a very mild phenotype: heart weight, heart weight to tibia length ratios, and cardiac dimensions were not altered. Increased perivascular fibrosis was only observed in hearts of young XinABC(-/-) mice. Striking differences were revealed in isolated cardiomyocytes: XinABC(-/-) cells demonstrated a significantly increased number of non-terminally localized ID-like structures. Furthermore, resting sarcomere length was increased, sarcomere shortening, peak shortening at 0.5-1 Hz, and the duration of shortening were decreased, and shortening and relengthening velocities were accelerated at frequencies above 4 Hz in XinABC(-/-) cardiomyocytes. ECG showed a significantly shorter HV interval and a trend towards shorter QRS interval in XinABC(-/-) mice, suggesting a faster conduction velocity of the ventricular-specific conduction system. In human cardiac tissue, expression of XinC protein was detected solely in samples from patients with cardiac hypertrophy. CONCLUSION: Total Xin deficiency leads to topographical ID alterations, premature fibrosis and subtle changes in contractile behaviour; this is a milder cardiac phenotype than that observed in XinAB(-/-) mice, which still can express XinC. Together with the finding that XinC is detected solely in cardiomyopathic human tissues, this suggests that its expression is responsible for the stronger dominant phenotype in XinAB(-/-) mice. Furthermore, it indicates that XinC may be involved in the development of human cardiac hypertrophy.


Subject(s)
Cardiomegaly/physiopathology , Cardiomyopathies/physiopathology , DNA-Binding Proteins/genetics , Myocytes, Cardiac/physiology , Nuclear Proteins/genetics , Severity of Illness Index , Actins/metabolism , Adult , Aged , Animals , Atrial Fibrillation/metabolism , Atrial Fibrillation/pathology , Atrial Fibrillation/physiopathology , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Electrocardiography , Female , Gene Expression/physiology , Gene Library , Humans , Isomerism , Male , Mice , Mice, Mutant Strains , Middle Aged , Myocardial Contraction/physiology , Myocytes, Cardiac/pathology , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Phenotype , Sarcomeres/pathology
10.
Dev Biol ; 318(1): 92-101, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18436206

ABSTRACT

Mechanical instability of skeletal muscle cells is the major cause of congenital muscular dystrophy. Here we show that the zebrafish lost-contact mutant, that lacks a functional integrin-linked kinase (ilk) gene, suffers from mechanical instability of skeletal muscle fibres. With genetic and morpholino knock-down experiments we demonstrate that: 1) laminin, itgalpha7, Ilk and beta-parvin are all critical for mechanical stability in skeletal muscles. 2) Ilk acts redundantly with the dystrophin/dystroglycan adhesion complex in maintaining mechanical stability of skeletal muscles. 3) Ilk protein is recruited to the myotendinous junctions, which requires the ECM component laminin and the presence of itgalpha7 in the sarcolemma. 4) Ilk, unexpectedly, is dispensable for formation of the adhesion complex. Ilk, however, is required for strengthening the adhesion of the muscle fibre with the ECM and this activity requires the presence of a functional kinase domain in Ilk. 5) We identified a novel interaction between Ilk and the mechanical stretch sensor protein MLP. Thus, Ilk is an essential intracellular component downstream of laminin and itgalpha7, providing strengthening of skeletal muscle fibre adhesion with the ECM and therefore qualified as a novel candidate gene for congenital muscular dystrophy.


Subject(s)
Antigens, CD/metabolism , Cell Adhesion/physiology , Extracellular Matrix/metabolism , Integrin alpha Chains/metabolism , Muscle, Skeletal , Protein Serine-Threonine Kinases/metabolism , Zebrafish Proteins/metabolism , Zebrafish , Actinin/genetics , Actinin/metabolism , Animals , Antigens, CD/classification , Antigens, CD/genetics , Cytoskeleton/metabolism , Extracellular Matrix/genetics , Humans , Integrin alpha Chains/classification , Integrin alpha Chains/genetics , Laminin/genetics , Laminin/metabolism , Lim Kinases/genetics , Lim Kinases/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/embryology , Muscle, Skeletal/metabolism , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/metabolism , Paxillin/genetics , Paxillin/metabolism , Phenotype , Phylogeny , Protein Serine-Threonine Kinases/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Two-Hybrid System Techniques , Zebrafish/anatomy & histology , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins/genetics
11.
Circulation ; 116(5): 515-25, 2007 Jul 31.
Article in English | MEDLINE | ID: mdl-17646580

ABSTRACT

BACKGROUND: Extracellular matrix proteins, such as laminins, and endothelial cells are known to influence cardiomyocyte performance; however, the underlying molecular mechanisms remain poorly understood. METHODS AND RESULTS: We used a forward genetic screen in zebrafish to identify novel genes required for myocardial function and were able to identify the lost-contact (loc) mutant, which encodes a nonsense mutation in the integrin-linked kinase (ilk) gene. This loc/ilk mutant is associated with a severe defect in cardiomyocytes and endothelial cells that leads to severe myocardial dysfunction. Additional experiments revealed the epistatic regulation between laminin-alpha4 (Lama4), integrin, and Ilk, which led us to screen for mutations in the human ILK and LAMA4 genes in patients with severe dilated cardiomyopathy. We identified 2 novel amino acid residue-altering mutations (2828C>T [Pro943Leu] and 3217C>T [Arg1073X]) in the integrin-interacting domain of the LAMA4 gene and 1 mutation (785C>T [Ala262Val]) in the ILK gene. Biacore quantitative protein/protein interaction data, which have been used to determine the equilibrium dissociation constants, point to the loss of integrin-binding capacity in case of the Pro943Leu (Kd=5+/-3 micromol/L) and Arg1073X LAMA4 (Kd=1+/-0.2 micromol/L) mutants compared with the wild-type LAMA4 protein (Kd=440+/-20 nmol/L). Additional functional data point to the loss of endothelial cells in affected patients as a direct consequence of the mutant genes, which ultimately leads to heart failure. CONCLUSIONS: This is the first report on mutations in the laminin, integrin, and ILK system in human cardiomyopathy, which has consequences for endothelial cells as well as for cardiomyocytes, thus providing a new genetic basis for dilated cardiomyopathy in humans.


Subject(s)
Cardiomyopathy, Dilated/genetics , Endothelial Cells/pathology , Laminin/genetics , Mutation, Missense , Myocytes, Cardiac/pathology , Point Mutation , Protein Serine-Threonine Kinases/genetics , Adult , Amino Acid Substitution , Animals , COS Cells , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Cell Adhesion , Chlorocebus aethiops , Chromosome Mapping , Codon, Nonsense , DNA Mutational Analysis , Embryo, Nonmammalian/pathology , Epigenesis, Genetic , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Female , Heart/embryology , Heart Failure/etiology , Heart Failure/pathology , Humans , Integrins/metabolism , Laminin/physiology , Male , Middle Aged , Models, Molecular , Myocardium/pathology , Oligonucleotides, Antisense/toxicity , Pedigree , Protein Binding , Protein Conformation , Protein Interaction Mapping , Protein Serine-Threonine Kinases/physiology , Protein Structure, Tertiary , Transfection , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/physiology
12.
Exp Cell Res ; 312(11): 2154-67, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-16631741

ABSTRACT

Filamin c is the predominantly expressed filamin isoform in striated muscles. It is localized in myofibrillar Z-discs, where it binds FATZ and myotilin, and in myotendinous junctions and intercalated discs. Here, we identify Xin, the protein encoded by the human gene 'cardiomyopathy associated 1' (CMYA1) as filamin c binding partner at these specialized structures where the ends of myofibrils are attached to the sarcolemma. Xin directly binds the EVH1 domain proteins Mena and VASP. In the adult heart, Xin and Mena/VASP colocalize with filamin c in intercalated discs. In cultured cardiomyocytes, the proteins also localize in the nonstriated part of myofibrils, where sarcomeres are assembled and an extensive reorganization of the actin cytoskeleton occurs. Unusual intraexonic splicing events result in the existence of three Xin isoforms that associate differentially with its ligands. The identification of the complex filamin c-Xin-Mena/VASP provides a first glance on the role of Xin in the molecular mechanisms involved in developmental and adaptive remodeling of the actin cytoskeleton during cardiac morphogenesis and sarcomere assembly.


Subject(s)
Alternative Splicing , Cell Adhesion Molecules/genetics , Contractile Proteins/metabolism , Cytoskeletal Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphoproteins/genetics , Animals , Animals, Newborn , Base Sequence , Binding Sites/genetics , Blotting, Western , Cell Adhesion Molecules/analysis , Cell Adhesion Molecules/metabolism , Cells, Cultured , Contractile Proteins/analysis , Cytoskeletal Proteins/analysis , Cytoskeletal Proteins/genetics , Exons , Filamins , Genetic Variation , Humans , Microfilament Proteins/analysis , Models, Biological , Molecular Sequence Data , Myocytes, Cardiac/chemistry , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Phosphoproteins/analysis , Phosphoproteins/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary/genetics , Rats
13.
J Cell Sci ; 117(Pt 22): 5257-68, 2004 Oct 15.
Article in English | MEDLINE | ID: mdl-15454575

ABSTRACT

Xin is a protein that is expressed during early developmental stages of cardiac and skeletal muscles. Immunolocalization studies indicated a peripheral localization in embryonic mouse heart, where Xin localizes with beta-catenin and N-cadherin. In adult tissues, Xin is found primarily in the intercalated discs of cardiomyocytes and the myotendinous junctions of skeletal muscle cells, both specialized attachment sites of the myofibrillar ends to the sarcolemma. A large part of the Xin protein consists of unique 16 amino acid repeats with unknown function. We have investigated the characteristics of the Xin repeats by transfection experiments and actin-binding assays and ascertained that, upon expression in cultured cells, these repeats bind to and stabilize the actin-based cytoskeleton. In vitro co-sedimentation assays with skeletal muscle actin indicated that they not only directly bind actin filaments, but also have the capability of arranging microfilaments into networks that sediment upon low-speed centrifugation. Very similar repeats were also found in 'Xin-repeat protein 2' (XIRP2), a novel protein that seems to be expressed mainly in striated muscles. Human XIRP2 contains 28 Xin repeats with properties identical to those of Xin. We conclude that the Xin repeats define a novel, repetitive actin-binding motif present in at least two different muscle proteins. These Xin-repeat proteins therefore constitute the first two members of a novel family of actin-binding proteins.


Subject(s)
Actins/chemistry , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Actins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cadherins/metabolism , Cell Line , Cell Line, Tumor , Circular Dichroism , Cytoskeletal Proteins/metabolism , Focal Adhesions , Humans , LIM Domain Proteins , Microscopy, Fluorescence , Molecular Sequence Data , Muscle, Skeletal/metabolism , Mutation , Peptides/chemistry , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Sequence Homology, Amino Acid , Thiazoles/chemistry , Thiazoles/pharmacology , Thiazolidines , Trans-Activators/metabolism , Transfection , Tropomyosin/chemistry , beta Catenin
SELECTION OF CITATIONS
SEARCH DETAIL