Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Nature ; 499(7458): 306-11, 2013 Jul 18.
Article in English | MEDLINE | ID: mdl-23868260

ABSTRACT

Aberrant neovascularization contributes to diseases such as cancer, blindness and atherosclerosis, and is the consequence of inappropriate angiogenic signalling. Although many regulators of pathogenic angiogenesis have been identified, our understanding of this process is incomplete. Here we explore the transcriptome of retinal microvessels isolated from mouse models of retinal disease that exhibit vascular pathology, and uncover an upregulated gene, leucine-rich alpha-2-glycoprotein 1 (Lrg1), of previously unknown function. We show that in the presence of transforming growth factor-ß1 (TGF-ß1), LRG1 is mitogenic to endothelial cells and promotes angiogenesis. Mice lacking Lrg1 develop a mild retinal vascular phenotype but exhibit a significant reduction in pathological ocular angiogenesis. LRG1 binds directly to the TGF-ß accessory receptor endoglin, which, in the presence of TGF-ß1, results in promotion of the pro-angiogenic Smad1/5/8 signalling pathway. LRG1 antibody blockade inhibits this switch and attenuates angiogenesis. These studies reveal a new regulator of angiogenesis that mediates its effect by modulating TGF-ß signalling.


Subject(s)
Endothelium, Vascular/metabolism , Glycoproteins/physiology , Retinal Neovascularization/metabolism , Signal Transduction , Transforming Growth Factor beta1/metabolism , Animals , Cells, Cultured , Endothelium, Vascular/cytology , Glycoproteins/genetics , Glycoproteins/metabolism , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Transforming Growth Factor beta/metabolism , Retinal Neovascularization/genetics , Retinal Vessels/metabolism , Transforming Growth Factor beta1/pharmacology
2.
Am J Pathol ; 180(1): 399-409, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22067912

ABSTRACT

Retinal pathologies are frequently accompanied by retinal vascular responses, including the formation of new vessels by angiogenesis (neovascularization). Pathological vascular changes may also include less well characterized traits of vascular remodeling that are non-neovascular, such as vessel pruning and the emergence of dilated and tortuous vessel phenotypes (telangiectasis). The molecular mechanisms underlying neovascular growth versus non-neovascular remodeling are poorly understood. We therefore undertook to identify novel regulators of non-neovascular remodeling in the retina by using the dystrophic Royal College of Surgeons (RCS) rat and the retinal dystrophy 1 (RD1) mouse, both of which display pronounced non-neovascular remodeling. Gene expression profiling of isolated retinal vessels from these mutant rodent models and wild-type controls revealed 60 differentially expressed genes. These included the genes for apelin (Apln) and for its receptor (Aplnr), both of which were strongly up-regulated in the mutants. Crossing RD1 mice into an Apln-null background substantially reduced vascular telangiectasia. In contrast, Apln gene deletion had no effect in two models of neovascular pathology [laser-induced choroidal neovascularization and the very low density lipoprotein receptor (Vldlr)-knockout mouse]. These findings suggest that in these models apelin has minimal effect on sprouting retinal angiogenesis, but contributes significantly to pathogenic non-neovascular remodeling.


Subject(s)
Intercellular Signaling Peptides and Proteins/physiology , Retinal Degeneration/physiopathology , Retinal Vessels/metabolism , Adipokines , Animals , Apelin , Choroidal Neovascularization/genetics , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/physiopathology , Gene Expression , Gene Silencing , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Microvessels/metabolism , Mutation/genetics , Rats , Retina , Retinal Degeneration/genetics , Retinal Degeneration/metabolism , Retinal Telangiectasis/prevention & control , Up-Regulation
3.
Arterioscler Thromb Vasc Biol ; 31(3): 657-64, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21148427

ABSTRACT

OBJECTIVE: RhoJ/TCL was identified by our group as an endothelial-expressed Rho GTPase. The aim of this study was to determine its tissue distribution, subcellular localization, and function in endothelial migration and tube formation. METHODS AND RESULTS: Using in situ hybridization, RhoJ was localized to endothelial cells in a set of normal and cancerous tissues and in the vasculature of mouse embryos; endogenous RhoJ was localized to focal adhesions by immunofluorescence. The proangiogenic factor vascular endothelial growth factor activated RhoJ in endothelial cells. Using either small interfering (si)RNA-mediated knockdown of RhoJ expression or overexpression of constitutively active RhoJ (daRhoJ), RhoJ was found to positively regulate endothelial motility and tubule formation. Downregulating RhoJ expression increased focal adhesions and stress fibers in migrating cells, whereas daRhoJ overexpression resulted in the converse. RhoJ downregulation resulted in increased contraction of a collagen gel and increased phospho-myosin light chain, indicative of increased actomyosin contractility. Pharmacological inhibition of Rho-kinase (which phosphorylates myosin light chain) or nonmuscle myosin II reversed the defective tube formation and migration of RhoJ knockdown cells. CONCLUSIONS: RhoJ is endothelial-expressed in vivo, activated by vascular endothelial growth factor, localizes to focal adhesions, regulates endothelial cell migration and tube formation, and modulates actomyosin contractility and focal adhesion numbers.


Subject(s)
Actomyosin/metabolism , Cell Movement , Endothelial Cells/enzymology , Focal Adhesions/metabolism , GTP Phosphohydrolases/metabolism , Neovascularization, Physiologic , rho GTP-Binding Proteins/metabolism , Animals , Cell Movement/drug effects , Cell Shape , Cells, Cultured , Endothelial Cells/drug effects , Fluorescent Antibody Technique , GTP Phosphohydrolases/genetics , Humans , In Situ Hybridization , Mice , Myosin Light Chains/metabolism , Neovascularization, Physiologic/drug effects , Phosphorylation , Protein Kinase Inhibitors/pharmacology , RNA Interference , Stress Fibers/metabolism , Transfection , Vascular Endothelial Growth Factor A/metabolism , rho GTP-Binding Proteins/genetics , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
4.
Data Brief ; 45: 108770, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36533287

ABSTRACT

This article presents data concerning STX18-AS1, a long noncoding RNA gene identified from a Genome-wide association study of Atrial Septal Defect (ASD). The data describes its expression patterns in human tissues and functions in regulating cardiomyocyte differentiation in vitro. STX18-AS1 is a lncRNA with a higher abundance in developing tissues, including hearts. Its transcription distribution within the embryonic hearts during key heart septation stages supports STX18-AS1's association with risk SNPs for ASD. The CRISPR stem cell pool in which STX18-AS1 was knocked down, showed reduced CM differentiation efficiency and lower expression of key cardiac transcriptional factors. This indicated its regulative role in supporting the lineage specification from cardiac mesoderm into cardiac progenitors and cardiomyocytes. These data can benefit the understanding of human embryonic heart developmental biology, and the time-course changes of cardiac transcriptional factors during in vitro cardiomyocyte differentiation from human embryonic stem cells.

5.
Circ Res ; 102(5): 562-70, 2008 Mar 14.
Article in English | MEDLINE | ID: mdl-18202311

ABSTRACT

Growth, maturation, and integrity of the blood vessel network require extensive communication between the endothelial cells, which line the vascular lumen, and associated mural cells, namely vascular smooth muscle cells and pericytes. Pericytes extend long processes, make direct contact with the capillary endothelium, and promote vascular quiescence by suppressing angiogenic sprouting. Vascular smooth muscle cells are highly contractile, extracellular matrix-secreting cells that cover arteries and veins and provide them with mechanical stability and elasticity. In the damaged blood vessel wall, for example in atherosclerotic lesions, vascular smooth muscle cells lose their differentiated state and acquire a highly mitotic, so-called "synthetic" phenotype, which is thought to promote pathogenesis. Among other factors, extracellular matrix molecules and integrin family cell-matrix receptors may regulate this phenotypic transition. Here we show that the inactivation of the gene encoding the integrin beta1 subunit (Itgb1) with a Cre-loxP approach in mice leads to mural cell defects and postnatal lethality. Integrin beta1-deficient vascular smooth muscle cells display several hallmarks of the synthetic phenotype: Cell proliferation is enhanced, whereas differentiation and their ability to support blood vessels are compromised. Similarly, mutant pericytes are poorly spread but present in larger numbers. Our analysis of this mutant model shows that integrin beta1-mediated cell-matrix adhesion is a major determinant of the mural cell phenotype.


Subject(s)
Blood Vessels/physiology , Cell Adhesion/physiology , Integrin beta1/physiology , Muscle, Smooth, Vascular/metabolism , Animals , Blood Vessels/cytology , Cell Differentiation/genetics , Cell Movement/genetics , Cell Proliferation , Cells, Cultured , Extracellular Matrix/metabolism , Flow Cytometry , Gene Targeting , Integrin beta1/genetics , Mice , Mice, Transgenic , Muscle, Smooth, Vascular/cytology
6.
Br J Pharmacol ; 176(20): 3956-3971, 2019 10.
Article in English | MEDLINE | ID: mdl-31328787

ABSTRACT

BACKGROUND AND PURPOSE: The Hippo pathway has emerged as a potential therapeutic target to control pathological cardiac remodelling. The core components of the Hippo pathway, mammalian Ste-20 like kinase 1 (Mst1) and mammalian Ste-20 like kinase 2 (Mst2), modulate cardiac hypertrophy, apoptosis, and fibrosis. Here, we study the effects of pharmacological inhibition of Mst1/2 using a novel inhibitor XMU-MP-1 in controlling the adverse effects of pressure overload-induced hypertrophy. EXPERIMENTAL APPROACH: We used cultured neonatal rat cardiomyocytes (NRCM) and C57Bl/6 mice with transverse aortic constriction (TAC) as in vitro and in vivo models, respectively, to test the effects of XMU-MP-1 treatment. We used luciferase reporter assays, western blots and immunofluorescence assays in vitro, with echocardiography, qRT-PCR and immunohistochemical methods in vivo. KEY RESULTS: XMU-MP-1 treatment significantly increased activity of the Hippo pathway effector yes-associated protein and inhibited phenylephrine-induced hypertrophy in NRCM. XMU-MP-1 improved cardiomyocyte survival and reduced apoptosis following oxidative stress. In vivo, mice 3 weeks after TAC, were treated with XMU-MP-1 (1 mg·kg-1 ) every alternate day for 10 further days. XMU-MP-1-treated mice showed better cardiac contractility than vehicle-treated mice. Cardiomyocyte cross-sectional size and expression of the hypertrophic marker, brain natriuretic peptide, were reduced in XMU-MP-1-treated mice. Improved heart function in XMU-MP-1-treated mice with TAC, was accompanied by fewer TUNEL positive cardiomyocytes and lower levels of fibrosis, suggesting inhibition of cardiomyocyte apoptosis and decreased fibrosis. CONCLUSIONS AND IMPLICATIONS: The Hippo pathway inhibitor, XMU-MP-1, reduced cellular hypertrophy and improved survival in cultured cardiomyocytes and, in vivo, preserved cardiac function following pressure overload.


Subject(s)
Myocytes, Cardiac/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Sulfonamides/pharmacology , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Cells, Cultured , Hippo Signaling Pathway , Male , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Pressure , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Sprague-Dawley , Sulfonamides/chemistry , Benzenesulfonamides
7.
Mol Biol Cell ; 16(7): 3411-24, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15843432

ABSTRACT

The vertebrate proteins Nesprin-1 and Nesprin-2 (also referred to as Enaptin and NUANCE) together with ANC-1 of Caenorhabditis elegans and MSP-300 of Drosophila melanogaster belong to a novel family of alpha-actinin type actin-binding proteins residing at the nuclear membrane. Using biochemical techniques, we demonstrate that Nesprin-2 binds directly to emerin and the C-terminal common region of lamin A/C. Selective disruption of the lamin A/C network in COS7 cells, using a dominant negative lamin B mutant, resulted in the redistribution of Nesprin-2. Furthermore, using lamin A/C knockout fibroblasts we show that lamin A/C is necessary for the nuclear envelope localization of Nesprin-2. In normal skin where lamin A/C is differentially expressed, strong Nesprin-2 expression was found in all epidermal layers, including the basal layer where only lamin C is present. This indicates that lamin C is sufficient for proper Nesprin-2 localization at the nuclear envelope. Expression of dominant negative Nesprin-2 constructs and knockdown studies in COS7 cells revealed that the presence of Nesprin-2 at the nuclear envelope is necessary for the proper localization of emerin. Our data imply a scaffolding function of Nesprin-2 at the nuclear membrane and suggest a potential involvement of this multi-isomeric protein in human disease.


Subject(s)
Lamin Type A/biosynthesis , Microfilament Proteins/biosynthesis , Nerve Tissue Proteins/biosynthesis , Nuclear Envelope/metabolism , Nuclear Proteins/biosynthesis , Animals , Blotting, Western , COS Cells , Caenorhabditis elegans , Cell Line, Tumor , Cell Nucleus/metabolism , Cytoplasm/metabolism , Drosophila melanogaster , Genes, Dominant , Glutathione Transferase/metabolism , Humans , Immunoblotting , Immunohistochemistry , Immunoprecipitation , In Vitro Techniques , Membrane Proteins/metabolism , Microscopy, Fluorescence , Models, Biological , Models, Genetic , Mutation , Plasmids/metabolism , Protein Binding , Protein Structure, Tertiary , RNA, Small Interfering/metabolism , Thymopoietins/metabolism , Transfection , Two-Hybrid System Techniques
9.
Nat Protoc ; 10(10): 1459-73, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26334866

ABSTRACT

The mouse fetal metatarsal provides a unique tool for studying angiogenesis. In comparison with other commonly used in vitro or ex vivo angiogenesis assays, vessel outgrowth from mouse fetal metatarsals is more representative of sprouting angiogensis in vivo. It allows the analysis of blood vessel growth, and the mechanisms underpinning this process, in a multicellular microenvironment that drives the formation of a robust and complex vascular network in the absence of exogenous growth factors. By labeling different constituents of the vascular structure, it is possible to perform 3D rendering of the spatial interplay between different cellular components and to carry out quantitative analysis of vessel outgrowth. High-resolution imaging permits the visualization of fine structural and cellular details. As the assay involves the use of fetal tissues, it is possible to follow new blood vessel formation in genetically modified mice that are perinatally lethal. The entire process takes 9-13 d. A detailed description of how to set up and perform the assay is described here.


Subject(s)
Culture Techniques/methods , Metatarsal Bones/blood supply , Neovascularization, Pathologic/pathology , Animals , Disease Models, Animal , Female , Fetus , Fluorescent Antibody Technique , Metatarsal Bones/pathology , Mice , Microvessels/pathology , Neovascularization, Pathologic/genetics , Staining and Labeling
10.
Nat Commun ; 6: 7286, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26129894

ABSTRACT

During angiogenesis, Rho-GTPases influence endothelial cell migration and cell-cell adhesion; however it is not known whether they control formation of vessel lumens, which are essential for blood flow. Here, using an organotypic system that recapitulates distinct stages of VEGF-dependent angiogenesis, we show that lumen formation requires early cytoskeletal remodelling and lateral cell-cell contacts, mediated through the RAC1 guanine nucleotide exchange factor (GEF) DOCK4 (dedicator of cytokinesis 4). DOCK4 signalling is necessary for lateral filopodial protrusions and tubule remodelling prior to lumen formation, whereas proximal, tip filopodia persist in the absence of DOCK4. VEGF-dependent Rac activation via DOCK4 is necessary for CDC42 activation to signal filopodia formation and depends on the activation of RHOG through the RHOG GEF, SGEF. VEGF promotes interaction of DOCK4 with the CDC42 GEF DOCK9. These studies identify a novel Rho-family GTPase activation cascade for the formation of endothelial cell filopodial protrusions necessary for tubule remodelling, thereby influencing subsequent stages of lumen morphogenesis.


Subject(s)
GTPase-Activating Proteins/physiology , Neovascularization, Pathologic , Neovascularization, Physiologic , Pseudopodia/physiology , Animals , Cytoskeleton/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Mice, Inbred C57BL , Mice, Knockout , Vascular Endothelial Growth Factor A/metabolism , cdc42 GTP-Binding Protein/metabolism , rho GTP-Binding Proteins/metabolism
11.
Eur J Cell Biol ; 91(8): 614-28, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22555292

ABSTRACT

Emery-Dreifuss muscular dystrophy (EDMD) is a late onset-disease characterized by skeletal muscle wasting and heart defects with associated risk of sudden death. The autosomal dominant form of the disease is caused by mutations in the LMNA gene encoding LaminA and C, the X-linked form results from mutations in the gene encoding the inner nuclear membrane protein Emerin (STA). Both Emerin and LaminA/C interact with the nuclear envelope proteins Nesprin-1 and -2 and mutations in genes encoding C-terminal isoforms of Nesprin-1 and -2 have also been implicated in EDMD. Here we analyse primary fibroblasts from patients affected by either Duchenne muscular dystrophy (DMD) or Emery-Dreifuss muscular dystrophy/Charcot-Marie-Tooth syndrome (EDMD/CMT) that in addition to the disease causing mutations harbour mutations in the Nesprin-1 gene and in the SUN1 and SUN2 gene, respectively. SUN proteins together with the Nesprins form the core of the LINC complex which connects the nucleus with the cytoskeleton. The mutations are accompanied by changes in cell adhesion, cell migration, senescence, and stress response, as well as in nuclear shape and nuclear envelope composition which are changes characteristic for laminopathies. Our results point to a potential influence of mutations in components of the LINC complex on the clinical outcome and the molecular pathology in the patients.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Fibroblasts/pathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Emery-Dreifuss/genetics , Cell Adhesion , Cell Movement , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Nucleus/pathology , Cell Nucleus Shape , Cellular Senescence , Charcot-Marie-Tooth Disease/metabolism , Charcot-Marie-Tooth Disease/pathology , Cytoskeletal Proteins , Female , Fibroblasts/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lamin Type A/genetics , Lamin Type A/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Emery-Dreifuss/metabolism , Muscular Dystrophy, Emery-Dreifuss/pathology , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Primary Cell Culture , Stress, Physiological , Transfection , Wound Healing
12.
Curr Biol ; 19(8): 668-74, 2009 Apr 28.
Article in English | MEDLINE | ID: mdl-19345098

ABSTRACT

During new blood vessel formation, the cessation of angiogenic sprouting is necessary for the generation of functional vasculature. How sprouting is halted is not known, but it is contemporaneous with the development of stable intercellular junctions [1]. We report that VE-cadherin, which is responsible for endothelial adherens junction organization [2, 3], plays a crucial role in the cessation of sprouting. Abrogating VE-cadherin function in an organotypic angiogenesis assay and in zebrafish embryos stimulates sprouting. We show that VE-cadherin signals to Rho-kinase-dependent myosin light-chain 2 phosphorylation, leading to actomyosin contractility [4], which regulates the distribution of VE-cadherin at cell-cell junctions. VE-cadherin antagonizes VEGFR2 signaling, and consequently, inhibition of VE-cadherin, Rho-kinase, or actomyosin contractility leads to VEGF-driven, Rac1-dependent sprouting. These findings suggest a novel mechanism by which cell-cell adhesion suppresses Rac1-dependent migration and sprouting by increasing actomyosin contractility at cell junctions.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Cardiac Myosins/metabolism , Cell Communication/physiology , Myosin Light Chains/metabolism , Signal Transduction/physiology , Zebrafish/anatomy & histology , Zebrafish/embryology , Actins/metabolism , Animals , Antigens, CD/genetics , Cadherins/genetics , Cardiac Myosins/genetics , Cell Line , Endothelial Cells/cytology , Endothelial Cells/physiology , Gene Knockdown Techniques , Heterocyclic Compounds, 4 or More Rings/metabolism , Humans , Intercellular Junctions/metabolism , Myosin Light Chains/genetics , Myosins/metabolism , Neovascularization, Physiologic , Phosphorylation , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Zebrafish/physiology , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
13.
J Cell Sci ; 118(Pt 15): 3419-30, 2005 Aug 01.
Article in English | MEDLINE | ID: mdl-16079285

ABSTRACT

Nesprins form a novel class of nuclear envelope-anchored spectrin-repeat proteins. We show that a direct association of their highly conserved C-terminal luminal domain with the inner nuclear membrane protein Sun1 mediates their nuclear envelope localisation. In Nesprin-1 and Nesprin-2 the conserved C-terminal amino acids PPPX are essential for the interaction with a C-terminal region in Sun1. In fact, Sun1 is required for the proper nuclear envelope localisation of Nesprin-2 as shown using dominant-negative mutants and by knockdown of Sun1 expression. Sun1 itself does not require functional A-type lamins for its localisation at the inner nuclear membrane in mammalian cells. Our findings propose a conserved nuclear anchorage mechanism between Caenorhabditis elegans and mammals and suggest a model in which Sun1 serves as a ;structural bridge' connecting the nuclear interior with the actin cytoskeleton.


Subject(s)
Microfilament Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Envelope/metabolism , Nuclear Proteins/metabolism , Animals , COS Cells , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Cell Nucleus/metabolism , Chlorocebus aethiops , Cloning, Molecular , Cytoskeletal Proteins , HeLa Cells , Humans , Intracellular Membranes/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C3H , Mice, Knockout , Microfilament Proteins/genetics , Microtubule-Associated Proteins/genetics , Models, Biological , Nerve Tissue Proteins/genetics , Nuclear Envelope/classification , Nuclear Envelope/ultrastructure , Nuclear Proteins/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
14.
Exp Cell Res ; 295(2): 330-9, 2004 May 01.
Article in English | MEDLINE | ID: mdl-15093733

ABSTRACT

Enaptin belongs to a family of recently identified giant proteins that associate with the F-actin cytoskeleton as well as the nuclear membrane. It is composed of an N-terminal alpha-actinin type actin-binding domain (ABD) followed by a long coiled coil rod and a transmembrane domain at the C-terminus. The ABD binds to F-actin in vivo and in vitro and leads to bundle formation. The human Enaptin gene spreads over 515 kb and gives rise to several splicing isoforms (Nesprin-1, Myne-1, Syne-1, CPG2). The longest assembled cDNA encompasses 27,669 bp and predicts a 1014 kDa protein. Antibodies against the ABD of Enaptin localise the protein at F-actin-rich structures throughout the cell and in focal contacts as well as at the nuclear envelope. In COS7 cells, the protein is also present within the nuclear compartment. With the discovery of the actin-binding properties of Enaptin and the highly homologous Nuance, we define a family of proteins that integrate the cytoskeleton with the nucleoskeleton.


Subject(s)
Actins/metabolism , Cell Membrane/metabolism , Cytoskeleton/metabolism , Actinin/metabolism , Alternative Splicing , Animals , Antibodies/metabolism , Base Sequence , COS Cells , Cell Compartmentation , Cell Line , Cell Membrane/chemistry , Cell Membrane/genetics , Cell Nucleus/chemistry , Chlorocebus aethiops , Cloning, Molecular , Cytoskeleton/chemistry , Cytoskeleton/genetics , Green Fluorescent Proteins , Humans , Luminescent Proteins/metabolism , Mice , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Molecular Weight , Nerve Tissue Proteins , Nuclear Proteins , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Structure, Tertiary , Recombinant Proteins/metabolism , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL