Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cell Mol Life Sci ; 79(2): 93, 2022 Jan 24.
Article in English | MEDLINE | ID: mdl-35075545

ABSTRACT

Arterial hypertension causes left ventricular hypertrophy leading to dilated cardiomyopathy. Following compensatory cardiomyocyte hypertrophy, cardiac dysfunction develops due to loss of cardiomyocytes preceded or paralleled by cardiac fibrosis. Zyxin acts as a mechanotransducer in vascular cells that may promote cardiomyocyte survival. Here, we analyzed cardiac function during experimental hypertension in zyxin knockout (KO) mice. In zyxin KO mice, made hypertensive by way of deoxycorticosterone acetate (DOCA)-salt treatment telemetry recording showed an attenuated rise in systolic blood pressure. Echocardiography indicated a systolic dysfunction, and isolated working heart measurements showed a decrease in systolic elastance. Hearts from hypertensive zyxin KO mice revealed increased apoptosis, fibrosis and an upregulation of active focal adhesion kinase as well as of integrins α5 and ß1. Both interstitial and perivascular fibrosis were even more pronounced in zyxin KO mice exposed to angiotensin II instead of DOCA-salt. Stretched microvascular endothelial cells may release collagen 1α2 and TGF-ß, which is characteristic for the transition to an intermediate mesenchymal phenotype, and thus spur the transformation of cardiac fibroblasts to myofibroblasts resulting in excessive scar tissue formation in the heart of hypertensive zyxin KO mice. While zyxin KO mice per se do not reveal a cardiac phenotype, this is unmasked upon induction of hypertension and owing to enhanced cardiomyocyte apoptosis and excessive fibrosis causes cardiac dysfunction. Zyxin may thus be important for the maintenance of cardiac function in spite of hypertension.


Subject(s)
Angiotensin II/toxicity , Cardiomegaly/prevention & control , Fibrosis/prevention & control , Hypertension/complications , Myocytes, Cardiac/cytology , Zyxin/physiology , Animals , Apoptosis , Blood Pressure , Cardiomegaly/etiology , Cardiomegaly/pathology , Fibrosis/etiology , Fibrosis/pathology , Hypertension/chemically induced , Hypertension/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/metabolism
2.
Mol Biol Cell ; 32(13): 1221-1228, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33909446

ABSTRACT

Sensing physical forces is a critical first step in mechano-transduction of cells. Zyxin, a LIM domain-containing protein, is recruited to force-bearing actin filaments and is thought to repair and strengthen them. Yet, the precise force-induced protein interactions surrounding zyxin remain unclear. Using BioID analysis, we identified proximal proteins surrounding zyxin under normal and force-bearing conditions by label-free mass spectrometry analysis. Under force-bearing conditions, increased biotinylation of α-actinin 1, α-actinin 4, and AFAP1 were detected, and these proteins accumulated along force-bearing actin fibers independently from zyxin, albeit at a lower intensity than zyxin. VASP also accumulated along force-bearing actin fibers in a zyxin-dependent manner, but the biotinylation of VASP remained constant regardless of force, supporting the model of a free zyxin-VASP complex in the cytoplasm being corecruited to tensed actin fibers. In addition, ARHGAP42, a RhoA GAP, was also identified as a proximal protein of zyxin and colocalized with zyxin along contractile actin bundles. The overexpression of ARHGAP42 reduced the rate of small wound closure, a zyxin-dependent process. These results demonstrate that the application of proximal biotinylation can resolve the proximity and composition of protein complexes as a function of force, which had not been possible with traditional biochemical analysis.


Subject(s)
Biomechanical Phenomena/physiology , Zyxin/metabolism , Zyxin/physiology , Actin Cytoskeleton/metabolism , Actins/metabolism , Animals , Cell Adhesion Molecules/metabolism , Dogs , Focal Adhesions/metabolism , Madin Darby Canine Kidney Cells , Mechanical Phenomena , Microfilament Proteins/metabolism , Phosphoproteins/metabolism , Stress, Mechanical , Zyxin/chemistry
3.
Cell Rep ; 33(7): 108396, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33207197

ABSTRACT

Zyxin is a cytoskeletal LIM-domain protein that regulates actin cytoskeleton assembly and gene expression. In the present work, we find that zyxin downregulation in Xenopus laevis embryos reduces the expression of numerous genes that regulate cell differentiation, but it enhances that of several genes responsible for embryonic stem cell status, specifically klf4, pou5f3.1, pou5f3.2, pou5f3.3, and vent2.1/2. For pou5f3 family genes (mammalian POU5F1/OCT4 homologs), we show that this effect is the result of mRNA stabilization due to complex formation with the Y-box protein Ybx1. When bound to Ybx1, zyxin interferes with the formation of these complexes, thereby stimulating pou5f3 mRNA degradation. In addition, in zebrafish embryos and human HEK293 cells, zyxin downregulation increases mRNA levels of the pluripotency genes KLF4, NANOG, and POU5F1/OCT4. Our findings indicate that zyxin may play a role as a switch among morphogenetic cell movement, differentiation, and embryonic stem cell status.


Subject(s)
Embryonic Stem Cells/metabolism , Zyxin/metabolism , Zyxin/physiology , Animals , Body Patterning/genetics , Cell Differentiation/genetics , Cytoskeletal Proteins/metabolism , Cytoskeleton/metabolism , Embryonic Stem Cells/physiology , Gene Expression Regulation, Developmental/genetics , HEK293 Cells , Humans , Kruppel-Like Factor 4 , Morphogenesis , Neural Plate/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Xenopus laevis/metabolism , Zebrafish/metabolism
4.
Cell Death Dis ; 11(8): 674, 2020 08 21.
Article in English | MEDLINE | ID: mdl-32826880

ABSTRACT

Proteases modulate critical processes in cutaneous tissue repair to orchestrate inflammation, cell proliferation and tissue remodeling. However, the functional consequences and implications in healing impairments of most cleavage events are not understood. Using iTRAQ-based Terminal Amine Isotopic Labeling of Substrates (TAILS) we had characterized proteolytic signatures in a porcine wound healing model and identified two neo-N termini derived from proteolytic cleavage of the focal adhesion protein and mechanotransducer zyxin. Here, we assign these proteolytic events to the activity of either caspase-1 or serine protease HtrA1 and analyze the biological relevance of the resultant zyxin truncations. By cellular expression of full-length and truncated zyxin proteins, we demonstrate nuclear translocation of a C-terminal zyxin fragment that could also be generated in vitro by HtrA1 cleavage and provide evidence for its anti-apoptotic activities, potentially by regulating the expression of modulators of cell proliferation, protein synthesis and genome stability. Targeted degradomics correlated endogenous generation of the same zyxin fragment with increased cell density in human primary dermal fibroblasts. Hence, this newly identified HtrA1-zyxin protease signaling axis might present a novel mechanism to transiently enhance cell survival in environments of increased cell density like in wound granulation tissue.


Subject(s)
High-Temperature Requirement A Serine Peptidase 1/metabolism , Zyxin/metabolism , Cell Count , Cell Nucleus/metabolism , Cell Survival , HeLa Cells , High-Temperature Requirement A Serine Peptidase 1/physiology , Humans , Peptide Hydrolases/metabolism , Protein Processing, Post-Translational , Proteolysis , Proteome/metabolism , Proteomics , Skin/metabolism , THP-1 Cells , Zyxin/physiology
5.
Nat Commun ; 8: 15817, 2017 06 12.
Article in English | MEDLINE | ID: mdl-28604737

ABSTRACT

Cytoskeletal mechanics regulates cell morphodynamics and many physiological processes. While contractility is known to be largely RhoA-dependent, the process by which localized biochemical signals are translated into cell-level responses is poorly understood. Here we combine optogenetic control of RhoA, live-cell imaging and traction force microscopy to investigate the dynamics of actomyosin-based force generation. Local activation of RhoA not only stimulates local recruitment of actin and myosin but also increased traction forces that rapidly propagate across the cell via stress fibres and drive increased actin flow. Surprisingly, this flow reverses direction when local RhoA activation stops. We identify zyxin as a regulator of stress fibre mechanics, as stress fibres are fluid-like without flow reversal in its absence. Using a physical model, we demonstrate that stress fibres behave elastic-like, even at timescales exceeding turnover of constituent proteins. Such molecular control of actin mechanics likely plays critical roles in regulating morphodynamic events.


Subject(s)
Stress Fibers/physiology , Zyxin/physiology , rhoA GTP-Binding Protein/physiology , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/physiology , Animals , Mechanotransduction, Cellular , Mice , NIH 3T3 Cells , Optogenetics , Stress Fibers/metabolism , Zyxin/genetics , Zyxin/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
6.
Nat Commun ; 8: 14639, 2017 03 03.
Article in English | MEDLINE | ID: mdl-28256511

ABSTRACT

Endothelial exocytosis of Weibel-Palade body (WPB) is one of the first lines of defence against vascular injury. However, the mechanisms that control WPB exocytosis in the final stages (including the docking, priming and fusion of granules) are poorly understood. Here we show that the focal adhesion protein zyxin is crucial in this process. Zyxin downregulation inhibits the secretion of von Willebrand factor (VWF), the most abundant cargo in WPBs, from human primary endothelial cells (ECs) induced by cAMP agonists. Zyxin-deficient mice exhibit impaired epinephrine-stimulated VWF release, prolonged bleeding time and thrombosis, largely due to defective endothelial secretion of VWF. Using live-cell super-resolution microscopy, we visualize previously unappreciated reorganization of pre-existing actin filaments around WPBs before fusion, dependent on zyxin and an interaction with the actin crosslinker α-actinin. Our findings identify zyxin as a physiological regulator of endothelial exocytosis through reorganizing local actin network in the final stage of exocytosis.


Subject(s)
Actin Cytoskeleton/metabolism , Endothelial Cells/metabolism , Exocytosis/physiology , Zyxin/physiology , von Willebrand Factor/metabolism , Actinin/metabolism , Animals , Bleeding Time , Colforsin/pharmacology , Cyclic AMP/agonists , Endothelial Cells/drug effects , Epinephrine/pharmacology , Exocytosis/drug effects , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Intravital Microscopy , Mice , Mice, Inbred C57BL , Mice, Knockout , Primary Cell Culture , RNA, Small Interfering/metabolism , Thrombosis/pathology , Weibel-Palade Bodies/metabolism
7.
PLoS One ; 12(3): e0171728, 2017.
Article in English | MEDLINE | ID: mdl-28278518

ABSTRACT

Bronchospasm induced in non-asthmatic human subjects can be easily reversed by a deep inspiration (DI) whereas bronchospasm that occurs spontaneously in asthmatic subjects cannot. This physiological effect of a DI has been attributed to the manner in which a DI causes airway smooth muscle (ASM) cells to stretch, but underlying molecular mechanisms-and their failure in asthma-remain obscure. Using cells and tissues from wild type and zyxin-/- mice we report responses to a transient stretch of physiologic magnitude and duration. At the level of the cytoskeleton, zyxin facilitated repair at sites of stress fiber fragmentation. At the level of the isolated ASM cell, zyxin facilitated recovery of contractile force. Finally, at the level of the small airway embedded with a precision cut lung slice, zyxin slowed airway dilation. Thus, at each level zyxin stabilized ASM structure and contractile properties at current muscle length. Furthermore, when we examined tissue samples from humans who died as the result of an asthma attack, we found increased accumulation of zyxin compared with non-asthmatics and asthmatics who died of other causes. Together, these data suggest a biophysical role for zyxin in fatal asthma.


Subject(s)
Asthma/physiopathology , Lung/physiopathology , Muscle Contraction/physiology , Zyxin/physiology , Adolescent , Adult , Animals , Case-Control Studies , Cytoskeleton , Female , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Smooth Muscle , Prospective Studies , Severity of Illness Index , Stress Fibers , Young Adult
8.
Nat Commun ; 7: 11123, 2016 Mar 31.
Article in English | MEDLINE | ID: mdl-27030211

ABSTRACT

The evolutionarily conserved Hippo pathway is a regulator that controls organ size, cell growth and tissue homeostasis. Upstream signals of the Hippo pathway have been widely studied, but how microenvironmental factors coordinately regulate this pathway remains unclear. In this study, we identify LIM domain protein Zyxin, as a scaffold protein, that in response to hypoxia and TGF-ß stimuli, forms a ternary complex with Lats2 and Siah2 and stabilizes their interaction. This interaction facilitates Lats2 ubiquitination and degradation, Yap dephosphorylation and subsequently activation. We show that Zyxin is required for TGF-ß and hypoxia-induced Lats2 downregulation and deactivation of Hippo signalling in MDA-MB-231 cells. Depletion of Zyxin impairs the capability of cell migration, proliferation and tumourigenesis in a xenograft model. Zyxin is upregulated in human breast cancer and positively correlates with histological stages and metastasis. Our study demonstrates that Zyxin-Lats2-Siah2 axis may serve as a potential therapeutic target in cancer treatment.


Subject(s)
Nuclear Proteins/physiology , Protein Serine-Threonine Kinases/physiology , Transforming Growth Factor beta/physiology , Tumor Suppressor Proteins/physiology , Ubiquitin-Protein Ligases/physiology , Zyxin/physiology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Carcinogenesis/genetics , Cell Hypoxia , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cellular Microenvironment , Female , HEK293 Cells , Heterografts/metabolism , Heterografts/pathology , Hippo Signaling Pathway , Humans , Mice , Mice, Inbred BALB C , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proteolysis , Signal Transduction , Transcription Factors , Transforming Growth Factor beta/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , YAP-Signaling Proteins , Zyxin/genetics , Zyxin/metabolism
9.
J Am Heart Assoc ; 4(6): e001712, 2015 Jun 12.
Article in English | MEDLINE | ID: mdl-26071033

ABSTRACT

BACKGROUND: Exposure of vascular smooth muscle cells (VSMCs) to excessive cyclic stretch such as in hypertension causes a shift in their phenotype. The focal adhesion protein zyxin can transduce such biomechanical stimuli to the nucleus of both endothelial cells and VSMCs, albeit with different thresholds and kinetics. However, there is no distinct vascular phenotype in young zyxin-deficient mice, possibly due to functional redundancy among other gene products belonging to the zyxin family. Analyzing zyxin function in VSMCs at the cellular level might thus offer a better mechanistic insight. We aimed to characterize zyxin-dependent changes in gene expression in VSMCs exposed to biomechanical stretch and define the functional role of zyxin in controlling the resultant VSMC phenotype. METHODS AND RESULTS: DNA microarray analysis was used to identify genes and pathways that were zyxin regulated in static and stretched human umbilical artery-derived and mouse aortic VSMCs. Zyxin-null VSMCs showed a remarkable shift to a growth-promoting, less apoptotic, promigratory and poorly contractile phenotype with ≈90% of the stretch-responsive genes being zyxin dependent. Interestingly, zyxin-null cells already seemed primed for such a synthetic phenotype, with mechanical stretch further accentuating it. This could be accounted for by higher RhoA activity and myocardin-related transcription factor-A mainly localized to the nucleus of zyxin-null VSMCs, and a condensed and localized accumulation of F-actin upon stretch. CONCLUSIONS: At the cellular level, zyxin is a key regulator of stretch-induced gene expression. Loss of zyxin drives VSMCs toward a synthetic phenotype, a process further consolidated by exaggerated stretch.


Subject(s)
Mechanotransduction, Cellular/physiology , Muscle, Smooth, Vascular/physiology , Zyxin/physiology , Acetazolamide , Animals , Apoptosis/physiology , Biomechanical Phenomena/physiology , Blotting, Western , Cells, Cultured , Fluorescent Antibody Technique , Gene Expression/physiology , Mice , Mice, Knockout , Muscle, Smooth, Vascular/metabolism , Oligonucleotide Array Sequence Analysis , Phenotype , Real-Time Polymerase Chain Reaction , Umbilical Arteries/physiology
10.
Curr Biol ; 25(6): 679-689, 2015 Mar 16.
Article in English | MEDLINE | ID: mdl-25728696

ABSTRACT

BACKGROUND: Coordinated multicellular growth during development is achieved by the sensing of spatial and nutritional boundaries. The conserved Hippo (Hpo) signaling pathway has been proposed to restrict tissue growth by perceiving mechanical constraints through actin cytoskeleton networks. The actin-associated LIM proteins Zyxin (Zyx) and Ajuba (Jub) have been linked to the control of tissue growth via regulation of Hpo signaling, but the study of Zyx has been hampered by a lack of genetic tools. RESULTS: We generated a zyx mutant in Drosophila using TALEN endonucleases and used this to show that Zyx antagonizes the FERM-domain protein Expanded (Ex) to control tissue growth, eye differentiation, and F-actin accumulation. Zyx membrane targeting promotes the interaction between the transcriptional co-activator Yorkie (Yki) and the transcription factor Scalloped (Sd), leading to activation of Yki target gene expression and promoting tissue growth. Finally, we show that Zyx's growth-promoting function is dependent on its interaction with the actin-associated protein Enabled (Ena) via a conserved LPPPP motif and is antagonized by Capping Protein (CP). CONCLUSIONS: Our results show that Zyx is a functional antagonist of Ex in growth control and establish a link between actin filament polymerization and Yki activity.


Subject(s)
Actins/physiology , Drosophila Proteins/physiology , Drosophila melanogaster/growth & development , Drosophila melanogaster/physiology , Membrane Proteins/physiology , Nuclear Proteins/physiology , Trans-Activators/physiology , Zyxin/physiology , Amino Acid Sequence , Amino Acid Substitution , Animals , Animals, Genetically Modified , Base Sequence , DNA/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Eye/growth & development , Eye/metabolism , Female , Genes, Insect , Membrane Proteins/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutant Proteins/genetics , Mutant Proteins/physiology , Nuclear Proteins/genetics , Organ Size/genetics , Organ Size/physiology , Organogenesis/genetics , Organogenesis/physiology , Trans-Activators/genetics , Wings, Animal/growth & development , Wings, Animal/metabolism , YAP-Signaling Proteins , Zyxin/genetics
12.
Development ; 141(20): 3922-33, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25252943

ABSTRACT

We describe the identification of zyxin as a regulator of synapse maintenance in mechanosensory neurons in C. elegans. zyx-1 mutants lacked PLM mechanosensory synapses as adult animals. However, most PLM synapses initially formed during development but were subsequently lost as the animals developed. Vertebrate zyxin regulates cytoskeletal responses to mechanical stress in culture. Our work provides in vivo evidence in support of such a role for zyxin. In particular, zyx-1 mutant synaptogenesis phenotypes were suppressed by disrupting locomotion of the mutant animals, suggesting that zyx-1 protects mechanosensory synapses from locomotion-induced forces. In cultured cells, zyxin is recruited to focal adhesions and stress fibers via C-terminal LIM domains and modulates cytoskeletal organization via the N-terminal domain. The synapse-stabilizing activity was mediated by a short isoform of ZYX-1 containing only the LIM domains. Consistent with this notion, PLM synaptogenesis was independent of α-actinin and ENA-VASP, both of which bind to the N-terminal domain of zyxin. Our results demonstrate that the LIM domain moiety of zyxin functions autonomously to mediate responses to mechanical stress and provide in vivo evidence for a role of zyxin in neuronal development.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/metabolism , Gene Expression Regulation, Developmental , Synapses/physiology , Zyxin/physiology , Actinin/metabolism , Animals , Animals, Genetically Modified , Axons/metabolism , Carrier Proteins/chemistry , Cytoskeleton/metabolism , Focal Adhesions/metabolism , Movement , Mutation , Neurons/metabolism , Phenotype , Phosphoproteins/chemistry , Protein Isoforms/physiology , Protein Structure, Tertiary , Stress, Mechanical
13.
Mol Biol Cell ; 24(8): 1232-49, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23427270

ABSTRACT

In vertebrates, zyxin is a LIM-domain protein belonging to a family composed of seven members. We show that the nematode Caenorhabditis elegans has a unique zyxin-like protein, ZYX-1, which is the orthologue of the vertebrate zyxin subfamily composed of zyxin, migfilin, TRIP6, and LPP. The ZYX-1 protein is expressed in the striated body-wall muscles and localizes at dense bodies/Z-discs and M-lines, as well as in the nucleus. In yeast two-hybrid assays ZYX-1 interacts with several known dense body and M-line proteins, including DEB-1 (vinculin) and ATN-1 (α-actinin). ZYX-1 is mainly localized in the middle region of the dense body/Z-disk, overlapping the apical and basal regions containing, respectively, ATN-1 and DEB-1. The localization and dynamics of ZYX-1 at dense bodies depend on the presence of ATN-1. Fluorescence recovery after photobleaching experiments revealed a high mobility of the ZYX-1 protein within muscle cells, in particular at dense bodies and M-lines, indicating a peripheral and dynamic association of ZYX-1 at these muscle adhesion structures. A portion of the ZYX-1 protein shuttles from the cytoplasm into the nucleus, suggesting a role for ZYX-1 in signal transduction. We provide evidence that the zyx-1 gene encodes two different isoforms, ZYX-1a and ZYX-1b, which exhibit different roles in dystrophin-dependent muscle degeneration occurring in a C. elegans model of Duchenne muscular dystrophy.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/metabolism , Dystrophin/metabolism , Muscles/metabolism , Zyxin/physiology , Actinin/metabolism , Amino Acid Sequence , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans Proteins/chemistry , Gene Expression , Molecular Sequence Data , Muscles/cytology , Organ Specificity , Phylogeny , Protein Isoforms/chemistry , Protein Isoforms/physiology , Protein Transport , Sequence Homology, Amino Acid , Zyxin/chemistry
14.
Sci Signal ; 5(254): ra91, 2012 Dec 11.
Article in English | MEDLINE | ID: mdl-23233529

ABSTRACT

Vascular cells respond to supraphysiological amounts of stretch with a characteristic phenotypic change that results in dysfunctional remodeling of the affected arteries. Although the pathophysiological consequences of stretch-induced signaling are well characterized, the mechanism of mechanotransduction is unclear. We focused on the mechanotransducer zyxin, which translocates to the nucleus to drive gene expression in response to stretch. In cultured human endothelial cells and perfused femoral arteries isolated from wild-type and several knockout mouse strains, we characterized a multistep signaling pathway whereby stretch led to a transient receptor potential channel 3-mediated release of the endothelial vasoconstrictor peptide endothelin-1 (ET-1). ET-1, through autocrine activation of its B-type receptor, elicited the release of pro-atrial natriuretic peptide (ANP), which caused the autocrine activation of the ANP receptor guanylyl cyclase A (GC-A). Activation of GC-A, in turn, led to protein kinase G-mediated phosphorylation of zyxin at serine 142, thereby triggering the translocation of zyxin to the nucleus, where it was required for stretch-induced gene expression. Thus, we have identified a stretch-induced signaling pathway in vascular cells that leads to the activation of zyxin, a cytoskeletal protein specifically involved in transducing mechanical stimuli.


Subject(s)
Arteries/cytology , Cell Nucleus/metabolism , Endothelin-1/metabolism , Gene Expression Regulation/genetics , Mechanotransduction, Cellular/physiology , Zyxin/metabolism , Active Transport, Cell Nucleus/physiology , Animals , Atrial Natriuretic Factor/metabolism , Cells, Cultured , Endothelial Cells/metabolism , Guanylate Cyclase/metabolism , Humans , Mice , Mice, Knockout , Phosphorylation , Stress, Mechanical , Transient Receptor Potential Channels/metabolism , Zyxin/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...