Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Mol Biol Cell ; 33(11): ar100, 2022 09 15.
Article in English | MEDLINE | ID: mdl-35767320

ABSTRACT

The small heat shock protein HspB1, also known as Hsp25/27, is a ubiquitously expressed molecular chaperone that responds to mechanical cues. Uniaxial cyclic stretch activates the p38 mitogen-activated protein kinase (MAPK) signaling cascade and increases the phosphorylation of HspB1. Similar to the mechanosensitive cytoskeletal regulator zyxin, phospho-HspB1 is recruited to features of the stretch-stimulated actin cytoskeleton. To evaluate the role of HspB1 and its phosphoregulation in modulating cell function, we utilized CRISPR/Cas9-edited HspB1-null cells and determined they were altered in behaviors such as actin cytoskeletal remodeling, cell spreading, and cell motility. In our model system, expression of WT HspB1, but not nonphosphorylatable HspB1, rescued certain characteristics of the HspB1-null cells including the enhanced cell motility of HspB1-null cells and the deficient actin reinforcement of stretch-stimulated HspB1-null cells. The recruitment of HspB1 to high-tension structures in geometrically constrained cells, such as actin comet tails emanating from focal adhesions, also required a phosphorylatable HspB1. We show that mechanical signals activate posttranslational regulation of the molecular chaperone, HspB1, and are required for normal cell behaviors including actin cytoskeletal remodeling, cell spreading, and cell migration.


Subject(s)
Actins , Heat-Shock Proteins, Small , Actins/metabolism , Cell Movement/physiology , HSP27 Heat-Shock Proteins/metabolism , Heat-Shock Proteins, Small/metabolism , Molecular Chaperones/metabolism , Phosphorylation
2.
Heliyon ; 8(12): e12147, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36619427

ABSTRACT

Formation of robust actomyosin stress fibers (SF) in response to cell stretch plays a key role in the transfer of information from the cytoplasm into the nucleus. Actin/LINC/Lamin (ALL) nuclear lines provide mechanical linkage between the actin cytoskeleton and the lamin nucleoskeleton across the nuclear envelope. To understand the establishment of ALL lines, we used live cell imaging of cells exposed to cyclic stretch. We discovered that nuclear pore complexes (NPCs) concentrate along ALL lines that are generated in response to uniaxial cyclic stretch. The ALL-associated NPCs display increased fluorescence intensity of nucleoporins Pom121, TPR and Nup153 relative to nucleoporins that are distal to the ALL lines. Here we test the hypothesis that a LINC complex component of ALL lines, SUN1 is involved in the integration of NPCs with ALL lines. We generated CRISPR SUN1 knockdown and knockout cell lines and show that SUN1 is essential for normal integration of NPCs to ALL lines. Loss or elimination of SUN1 significantly diminishes NPC/ALL line integration, demonstrating a key role for SUN1 in the recruitment or stabilization of NPCs to a discrete subdomain of the nuclear envelope at ALL lines. This work provides new insight into the mechanism by which cells respond to mechanical force through nuclear envelope remodeling.

3.
Mol Biol Cell ; 31(16): 1774-1787, 2020 07 21.
Article in English | MEDLINE | ID: mdl-31967947

ABSTRACT

Mechanical stimulation of fibroblasts induces changes in the actin cytoskeleton including stress fiber (SF) reinforcement and realignment. Here we characterize the nuclear response to mechanical stimulation (uniaxial cyclic stretch). Using fluorescence microscopy and quantitative image analysis we find that stretch-induced nuclear elongation and alignment perpendicular to the stretch vector are dependent on formin-regulated actin polymerization. The mechanosensitive transcription factors Yes-associated protein/Transcriptional coactivator with PDZ domain (YAP/TAZ) and myocardin-related transcription factor (MRTF-A, also known as MKL1 and MAL1) accumulate in the nucleus and activate their target genes in response to uniaxial cyclic stretch. We show that transmembrane actin nuclear (TAN) lines are induced by stretch stimulation and nuclear envelope (NE) proteins including nesprins, SUN2, and lamins form Linkers of the Nucleoskeleton and Cytoskeleton (LINC) complexes aligned with actin SFs. These NE structures are altered by pharmacological treatments (Cytochalasin D and Jasplakinolide) or genetic disruption (zyxin gene deletion) that alter actin, and their persistence requires maintenance of stretch stimulation. Nuclear pore complexes (NPCs) accumulate at TAN lines providing a potential mechanism for linking mechanical cues to NPC function.


Subject(s)
Mechanoreceptors/metabolism , Nuclear Pore/metabolism , Actin Cytoskeleton/metabolism , Actins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Nucleus/metabolism , Cytoskeleton/metabolism , Fibroblasts/metabolism , Membrane Proteins/metabolism , Mice , Nuclear Proteins/metabolism , Primary Cell Culture , Stress Fibers/metabolism , Stress, Mechanical , Trans-Activators/metabolism , Transcription Factors/metabolism , YAP-Signaling Proteins
4.
Mol Syst Biol ; 12(3): 860, 2016 Mar 10.
Article in English | MEDLINE | ID: mdl-26969729

ABSTRACT

The signaling events that drive familial breast cancer (FBC) risk remain poorly understood. While the majority of genomic studies have focused on genetic risk variants, known risk variants account for at most 30% of FBC cases. Considering that multiple genes may influence FBC risk, we hypothesized that a pathway-based strategy examining different data types from multiple tissues could elucidate the biological basis for FBC. In this study, we performed integrated analyses of gene expression and exome-sequencing data from peripheral blood mononuclear cells and showed that cell adhesion pathways are significantly and consistently dysregulated in women who develop FBC. The dysregulation of cell adhesion pathways in high-risk women was also identified by pathway-based profiling applied to normal breast tissue data from two independent cohorts. The results of our genomic analyses were validated in normal primary mammary epithelial cells from high-risk and control women, using cell-based functional assays, drug-response assays, fluorescence microscopy, and Western blotting assays. Both genomic and cell-based experiments indicate that cell-cell and cell-extracellular matrix adhesion processes seem to be disrupted in non-malignant cells of women at high risk for FBC and suggest a potential role for these processes in FBC development.


Subject(s)
Breast Neoplasms/metabolism , Genetic Predisposition to Disease , Signal Transduction , Aged , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Adhesion , Cohort Studies , Female , Gene Expression Profiling , Genetic Variation , Humans , Leukocytes, Mononuclear/metabolism , Middle Aged
5.
Genes Cancer ; 6(3-4): 129-43, 2015 Mar.
Article in English | MEDLINE | ID: mdl-26000096

ABSTRACT

In Ewing sarcoma, NKX2-2 is a critical activated target of the oncogenic transcription factor EWS/FLI that is required for transformation. However, its biological function in this malignancy is unknown. Here we provide evidence that NKX2-2 mediates the EWS/FLI-controlled block of mesenchymal features. Transcriptome-wide RNA sequencing revealed that NKX2-2 represses cell adhesion and extracellular matrix organization genes. NKX2-2-depleted cells form more focal adhesions and organized actin stress fibers, and spread over a wider area-hallmarks of mesenchymally derived cells. Furthermore, NKX2-2 represses the actin-stabilizing protein zyxin, suggesting that these morphological changes are attributable to zyxin de-repression. In addition, NKX2-2-knockdown cells display marked increases in migration and substrate adhesion. However, only part of the EWS/FLI phenotype is NKX2-2-dependent; consequently, NKX2-2 is insufficient to rescue EWS/FLI repression of mesenchymalization. Strikingly, we found that EWS/FLI-and NKX22-repressed genes are activated by ZEB2, which was previously shown to block Ewing sarcoma epithelialization. Together, these data support an emerging theme wherein Ewing sarcoma cells highly express transcription factors that maintain an undifferentiated state. Importantly, co-opting epithelial and mesenchymal traits by Ewing sarcoma cells may explain how the primary tumor grows rapidly while also "passively" metastasizing, without the need for transitions toward differentiated states, as in carcinomas.

6.
Mol Biol Cell ; 25(18): 2695-709, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25057021

ABSTRACT

Ewing sarcoma is the second-most-common bone cancer in children. Driven by an oncogenic chromosomal translocation that results in the expression of an aberrant transcription factor, EWS/FLI, the disease is typically aggressive and micrometastatic upon presentation. Silencing of EWS/FLI in patient-derived tumor cells results in the altered expression of hundreds to thousands of genes and is accompanied by dramatic morphological changes in cytoarchitecture and adhesion. Genes encoding focal adhesion, extracellular matrix, and actin regulatory proteins are dominant targets of EWS/FLI-mediated transcriptional repression. Reexpression of genes encoding just two of these proteins, zyxin and α5 integrin, is sufficient to restore cell adhesion and actin cytoskeletal integrity comparable to what is observed when the EWS/FLI oncogene expression is compromised. Using an orthotopic xenograft model, we show that EWS/FLI-induced repression of α5 integrin and zyxin expression promotes tumor progression by supporting anchorage-independent cell growth. This selective advantage is paired with a tradeoff in which metastatic lung colonization is compromised.


Subject(s)
Bone Neoplasms/metabolism , Cell Adhesion , Cytoskeleton/metabolism , Lung Neoplasms/metabolism , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Protein c-fli-1/metabolism , RNA-Binding Protein EWS/metabolism , Sarcoma, Ewing/metabolism , Animals , Bone Neoplasms/pathology , Cell Line, Tumor , Gene Expression , Gene Expression Regulation, Neoplastic , Humans , Integrin alphaV/metabolism , Lung/pathology , Lung Neoplasms/secondary , Male , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Sarcoma, Ewing/pathology , Zyxin/metabolism
7.
Clin Cancer Res ; 20(17): 4584-97, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24963049

ABSTRACT

PURPOSE: Ewing sarcoma is a pediatric bone tumor that absolutely relies on the transcriptional activity of the EWS/ETS family of fusion oncoproteins. While the most common fusion, EWS/FLI, utilizes lysine-specific demethylase 1 (LSD1) to repress critical tumor suppressors, small-molecule blockade of LSD1 has not yet been thoroughly explored as a therapeutic approach for Ewing sarcoma. We therefore evaluated the translational potential of potent and specific LSD1 inhibition with HCI2509 on the transcriptional program of both EWS/FLI and EWS/ERG as well as the downstream oncogenic phenotypes driven by EWS/ETS fusions in both in vitro and in vivo models of Ewing sarcoma. EXPERIMENTAL DESIGN: RNA-seq was used to compare the transcriptional profiles of EWS/FLI, EWS/ERG, and treatment with HCI2509 in both EWS/FLI- and EWS/ERG-containing cell lines. We then evaluated morphologic phenotypes of treated cells with immunofluorescence. The induction of apoptosis was evaluated using caspase-3/7 activation and TUNEL staining. Colony forming assays were used to test oncogenic transformation and xenograft studies with patient-derived cell lines were used to evaluate the effects of HCI2509 on tumorigenesis. RESULTS: HCI2509 caused a dramatic reversal of both the up- and downregulated transcriptional profiles of EWS/FLI and EWS/ERG accompanied by the induction of apoptosis and disruption of morphologic and oncogenic phenotypes modulated by EWS/FLI. Importantly, HCI2509 displayed single-agent efficacy in multiple xenograft models. CONCLUSIONS: These data support epigenetic modulation with HCI2509 as a therapeutic strategy for Ewing sarcoma, and highlight a critical dual role for LSD1 in the oncogenic transcriptional activity of EWS/ETS proteins.


Subject(s)
Bone Neoplasms/genetics , Histone Demethylases/genetics , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Protein c-ets-1/genetics , RNA-Binding Protein EWS/genetics , Sarcoma, Ewing/genetics , Apoptosis/genetics , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Histone Demethylases/antagonists & inhibitors , Humans , Proto-Oncogene Protein c-fli-1/genetics , Sarcoma, Ewing/pathology , Trans-Activators/genetics , Transcriptional Regulator ERG
8.
PLoS One ; 8(8): e69378, 2013.
Article in English | MEDLINE | ID: mdl-23990882

ABSTRACT

Contractile actomyosin stress fibers are critical for maintaining the force balance between the interior of the cell and its environment. Consequently, the actin cytoskeleton undergoes dynamic mechanical loading. This results in spontaneous, stochastic, highly localized strain events, characterized by thinning and elongation within a discrete region of stress fiber. Previous work showed the LIM-domain adaptor protein, zyxin, is essential for repair and stabilization of these sites. Using live imaging, we show paxillin, another LIM-domain adaptor protein, is also recruited to stress fiber strain sites. Paxillin recruitment to stress fiber strain sites precedes zyxin recruitment. Zyxin and paxillin are each recruited independently of the other. In cells lacking paxillin, actin recovery is abrogated, resulting in slowed actin recovery and increased incidence of catastrophic stress fiber breaks. For both paxillin and zyxin, the LIM domains are necessary and sufficient for recruitment. This work provides further evidence of the critical role of LIM-domain proteins in responding to mechanical stress in the actin cytoskeleton.


Subject(s)
Actins/chemistry , Paxillin/chemistry , Stress Fibers/metabolism , Zyxin/chemistry , Actomyosin/metabolism , Animals , Cell Line , Cell Separation , Cytoskeleton/metabolism , Fibroblasts/metabolism , Flow Cytometry , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Green Fluorescent Proteins/metabolism , Homeostasis , Image Processing, Computer-Assisted , Mice , Mice, Transgenic , Microscopy, Fluorescence , Phosphotyrosine/chemistry , Protein Structure, Tertiary , RNA Interference , Signal Transduction , Stochastic Processes
9.
Genes Cancer ; 3(2): 102-16, 2012 Feb.
Article in English | MEDLINE | ID: mdl-23050043

ABSTRACT

Ewing sarcoma is a tumor of the bone and soft tissue caused by the expression of a translocation-derived oncogenic transcription factor, EWS/FLI. Overt metastases are associated with a poor prognosis in Ewing sarcoma, but patients without overt metastases frequently harbor micrometastatic disease at presentation. This suggests that the metastatic potential of Ewing sarcoma exists at an early stage during tumor development. We have therefore explored whether the inciting oncogenic event in Ewing sarcoma, EWS/FLI, directly modulates tumor cell features that support metastasis, such as cell adhesion, cell migration, and cytoarchitecture. We used an RNAi-based approach in patient-derived Ewing sarcoma cell lines. Although we hypothesized that EWS/FLI might induce classic metastatic features, such as increased cell adhesion, migration, and invasion (similar to the phenotypes observed when epithelial malignancies undergo an epithelial-to-mesenchymal transition during the process of metastasis), surprisingly, we found the opposite. Thus, EWS/FLI expression inhibited the adhesion of isolated cells in culture and prevented adhesion in an in vivo mouse lung assay. Cell migration was similarly inhibited by EWS/FLI expression. Furthermore, EWS/FLI expression caused a striking loss of organized actin stress fibers and focal adhesions and a concomitant loss of cell spreading, suggesting that EWS/FLI disrupts the mesenchymal phenotype of a putative tumor cell-of-origin. These data suggest a new paradigm for the dissemination and metastasis of mesenchymally derived tumors: these tumors may disseminate via a "passive/stochastic" model rather than via an "active" epithelial-to-mesenchymal type transition. In the case of Ewing sarcoma, it appears that the loss of cell adhesion needed to promote tumor cell dissemination might be induced by the EWS/FLI oncogene itself rather than via an accumulation of stepwise mutations.

10.
Mol Biol Cell ; 23(10): 1846-59, 2012 May.
Article in English | MEDLINE | ID: mdl-22456508

ABSTRACT

Reinforcement of actin stress fibers in response to mechanical stimulation depends on a posttranslational mechanism that requires the LIM protein zyxin. The C-terminal LIM region of zyxin directs the force-sensitive accumulation of zyxin on actin stress fibers. The N-terminal region of zyxin promotes actin reinforcement even when Rho kinase is inhibited. The mechanosensitive integrin effector p130Cas binds zyxin but is not required for mitogen-activated protein kinase-dependent zyxin phosphorylation or stress fiber remodeling in cells exposed to uniaxial cyclic stretch. α-Actinin and Ena/VASP proteins bind to the stress fiber reinforcement domain of zyxin. Mutation of their docking sites reveals that zyxin is required for recruitment of both groups of proteins to regions of stress fiber remodeling. Zyxin-null cells reconstituted with zyxin variants that lack either α-actinin or Ena/VASP-binding capacity display compromised response to mechanical stimulation. Our findings define a bipartite mechanism for stretch-induced actin remodeling that involves mechanosensitive targeting of zyxin to actin stress fibers and localized recruitment of actin regulatory machinery.


Subject(s)
Stress Fibers/metabolism , Stress, Physiological , Zyxin/metabolism , Actinin/metabolism , Amino Acid Sequence , Animals , Binding Sites , Biomechanical Phenomena , Cell Adhesion Molecules/metabolism , Crk-Associated Substrate Protein/genetics , Crk-Associated Substrate Protein/metabolism , Fibroblasts/metabolism , Fibroblasts/physiology , Focal Adhesions/metabolism , Gene Expression , Humans , MAP Kinase Signaling System , Mice , Microfilament Proteins/metabolism , Microscopy, Fluorescence , Phosphoproteins/metabolism , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Protein Processing, Post-Translational , Protein Transport , Sequence Deletion , Zyxin/genetics , rho-Associated Kinases/metabolism
11.
Genes Cancer ; 1(5): 506-515, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20852740

ABSTRACT

Zyxin is a dual-function LIM domain protein that regulates actin dynamics in response to mechanical stress and shuttles between focal adhesions and the cell nucleus. Here we show that zyxin contributes to UV-induced apoptosis. Exposure of wild-type fibroblasts to UV-C irradiation results in apoptotic cell death, whereas cells harboring a homozygous disruption of the zyxin gene display a statistically significant survival advantage. To gain insight into the molecular mechanism by which zyxin promotes apoptotic signaling, we expressed an affinity-tagged zyxin variant in zyxin-null cells and isolated zyxin-associated proteins from cell lysates under physiological conditions. A 130-kDa protein that was co-isolated with zyxin was identified by microsequence analysis as the Cell Cycle and Apoptosis Regulator Protein-1 (CARP-1). CARP-1 associates with the LIM region of zyxin. Zyxin lacking the CARP-1 binding region shows reduced proapoptotic activity in response to UV-C irradiation. We demonstrate that CARP-1 is a nuclear protein. Zyxin is modified by phosphorylation in cells exposed to UV-C irradiation, and nuclear accumulation of zyxin is induced by UV-C exposure. These findings highlight a novel mechanism for modulating the apoptotic response to UV irradiation.

12.
J Cell Biol ; 174(3): 447-58, 2006 Jul 31.
Article in English | MEDLINE | ID: mdl-16880273

ABSTRACT

Cell-substrate contacts, called focal adhesions (FAs), are dynamic in rapidly moving cells. We show that supervillin (SV)--a peripheral membrane protein that binds myosin II and F-actin in such cells--negatively regulates stress fibers, FAs, and cell-substrate adhesion. The major FA regulatory sequence within SV (SV342-571) binds to the LIM domains of two proteins in the zyxin family, thyroid receptor-interacting protein 6 (TRIP6) and lipoma-preferred partner (LPP), but not to zyxin itself. SV and TRIP6 colocalize within large FAs, where TRIP6 may help recruit SV. RNAi-mediated decreases in either protein increase cell adhesion to fibronectin. TRIP6 partially rescues SV effects on stress fibers and FAs, apparently by mislocating SV away from FAs. Thus, SV interactions with TRIP6 at FAs promote loss of FA structure and function. SV and TRIP6 binding partners suggest several specific mechanisms through which the SV-TRIP6 interaction may regulate FA maturation and/or disassembly.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Focal Adhesions/metabolism , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Transcription Factors/metabolism , ATPases Associated with Diverse Cellular Activities , Adaptor Proteins, Signal Transducing/chemistry , Animals , COS Cells , Cattle , Cells, Cultured , Chlorocebus aethiops , Down-Regulation/genetics , Green Fluorescent Proteins/metabolism , Humans , LIM Domain Proteins , Mice , Microtubule-Associated Proteins/metabolism , Myocytes, Smooth Muscle/cytology , Nuclear Proteins/metabolism , Proteasome Endopeptidase Complex , Protein Binding , Rats , Regulatory Sequences, Nucleic Acid/genetics , Transcription Factors/chemistry , t-Complex Genome Region
13.
J Cell Biol ; 172(5): 771-82, 2006 Feb 27.
Article in English | MEDLINE | ID: mdl-16505170

ABSTRACT

Focal adhesions are specialized regions of the cell surface where integrin receptors and associated proteins link the extracellular matrix to the actin cytoskeleton. To define the cellular role of the focal adhesion protein zyxin, we characterized the phenotype of fibroblasts in which the zyxin gene was deleted by homologous recombination. Zyxin-null fibroblasts display enhanced integrin-dependent adhesion and are more migratory than wild-type fibroblasts, displaying reduced dependence on extracellular matrix cues. We identified differences in the profiles of 75- and 80-kD tyrosine-phosphorylated proteins in the zyxin-null cells. Tandem array mass spectrometry identified both modified proteins as isoforms of the actomyosin regulator caldesmon, a protein known to influence contractility, stress fiber formation, and motility. Zyxin-null fibroblasts also show deficits in actin stress fiber remodeling and exhibit changes in the molecular composition of focal adhesions, most notably by severely reduced accumulation of Ena/VASP proteins. We postulate that zyxin cooperates with Ena/VASP proteins and caldesmon to influence integrin-dependent cell motility and actin stress fiber remodeling.


Subject(s)
Actins/metabolism , Cell Adhesion Molecules/metabolism , Cell Movement/genetics , Cytoskeletal Proteins/metabolism , Metalloproteins/deficiency , Metalloproteins/genetics , Microfilament Proteins/metabolism , Phosphoproteins/metabolism , Actins/deficiency , Animals , Calmodulin-Binding Proteins/metabolism , Cell Adhesion/genetics , Cell Line, Transformed , Cells, Cultured , Depsipeptides/pharmacology , Extracellular Matrix/physiology , Fibroblasts/metabolism , Integrins/biosynthesis , Integrins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitosis/physiology , Stress Fibers/drug effects , Zyxin
14.
J Cell Biol ; 171(2): 209-15, 2005 Oct 24.
Article in English | MEDLINE | ID: mdl-16247023

ABSTRACT

Organs and tissues adapt to acute or chronic mechanical stress by remodeling their actin cytoskeletons. Cells that are stimulated by cyclic stretch or shear stress in vitro undergo bimodal cytoskeletal responses that include rapid reinforcement and gradual reorientation of actin stress fibers; however, the mechanism by which cells respond to mechanical cues has been obscure. We report that the application of either unidirectional cyclic stretch or shear stress to cells results in robust mobilization of zyxin from focal adhesions to actin filaments, whereas many other focal adhesion proteins and zyxin family members remain at focal adhesions. Mechanical stress also induces the rapid zyxin-dependent mobilization of vasodilator-stimulated phosphoprotein from focal adhesions to actin filaments. Thickening of actin stress fibers reflects a cellular adaptation to mechanical stress; this cytoskeletal reinforcement coincides with zyxin mobilization and is abrogated in zyxin-null cells. Our findings identify zyxin as a mechanosensitive protein and provide mechanistic insight into how cells respond to mechanical cues.


Subject(s)
Actin Cytoskeleton/metabolism , Actins/metabolism , Cytoskeleton/metabolism , Focal Adhesions/physiology , Glycoproteins/metabolism , Metalloproteins/metabolism , Animals , Cytoskeletal Proteins , Endothelial Cells/metabolism , Fibroblasts/metabolism , Humans , Mice , Stress, Mechanical , Zyxin
15.
Mol Cell Biol ; 23(1): 70-9, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12482962

ABSTRACT

Zyxin is an evolutionarily conserved protein that is concentrated at sites of cell adhesion, where it associates with members of the Enabled (Ena)/vasodilator-stimulated phosphoprotein (VASP) family of cytoskeletal regulators and is postulated to play a role in cytoskeletal dynamics and signaling. Zyxin transcripts are detected throughout murine embryonic development, and the protein is widely expressed in adults. Here we used a reverse genetic approach to examine the consequences of loss of zyxin function in the mouse. Mice that lack zyxin function are viable and fertile and display no obvious histological abnormalities in any of the organs examined. Because zyxin contributes to the localization of Ena/VASP family members at certain subcellular locations, we carefully examined the zyxin(-/-) mice for evidence of defects that have been observed when Ena/VASP proteins are compromised in the mouse. Specifically, we evaluated blood platelet function, nervous system development, and skin architecture but did not detect any defects in these systems. Zyxin is the founding member of a family of proteins that also includes the lipoma preferred partner (LPP) and thyroid receptor-interacting protein 6 (TRIP6). These zyxin family members display patterns of expression that significantly overlap that of zyxin. Western blot analysis indicates that there is no detectable upregulation of either LPP or TRIP6 expression in tissues derived from zyxin-null mice. Because zyxin family members may have overlapping functions, a comprehensive understanding of the role of these proteins in the mouse will require the generation of compound mutations in which multiple zyxin family members are simultaneously compromised.


Subject(s)
Adaptor Proteins, Signal Transducing , Blood Platelets/metabolism , Brain/growth & development , Epidermis/physiology , Gene Expression Regulation, Developmental , Metalloproteins/genetics , ATPases Associated with Diverse Cellular Activities , Amino Acid Sequence , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Epidermis/ultrastructure , Fertility/genetics , LIM Domain Proteins , Lung/physiology , Metalloproteins/metabolism , Mice , Mice, Mutant Strains , Microfilament Proteins , Molecular Sequence Data , Phosphoproteins/genetics , Phosphoproteins/metabolism , Proteasome Endopeptidase Complex , Reproduction/genetics , Spleen/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , Up-Regulation , Zinc Fingers , Zyxin
SELECTION OF CITATIONS
SEARCH DETAIL
...