Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Redox Biol ; 62: 102670, 2023 06.
Article in English | MEDLINE | ID: mdl-36958249

ABSTRACT

Keratinocytes of the mammalian skin provide not only mechanical protection for the tissues, but also transmit mechanical, chemical, and thermal stimuli from the external environment to the sensory nerve terminals. Sensory nerve fibers penetrate the epidermal basement membrane and function in the tight intercellular space among keratinocytes. Here we show that epidermal keratinocytes produce hydrogen peroxide upon the activation of the NADPH oxidase dual oxidase 1 (DUOX1). This enzyme can be activated by increasing cytosolic calcium levels. Using DUOX1 knockout animals as a model system we found an increased sensitivity towards certain noxious stimuli in DUOX1-deficient animals, which is not due to structural changes in the skin as evidenced by detailed immunohistochemical and electron-microscopic analysis of epidermal tissue. We show that DUOX1 is expressed in keratinocytes but not in the neural sensory pathway. The release of hydrogen peroxide by activated DUOX1 alters both the activity of neuronal TRPA1 and redox-sensitive potassium channels expressed in dorsal root ganglia primary sensory neurons. We describe hydrogen peroxide, produced by DUOX1 as a paracrine mediator of nociceptive signal transmission. Our results indicate that a novel, hitherto unknown redox mechanism modulates noxious sensory signals.


Subject(s)
Hydrogen Peroxide , NADPH Oxidases , Animals , Dual Oxidases/genetics , Hydrogen Peroxide/metabolism , NADPH Oxidases/metabolism , Peroxides , Nociception , NADPH Oxidase 1 , Mammals/metabolism
2.
Redox Biol ; 54: 102385, 2022 08.
Article in English | MEDLINE | ID: mdl-35803124

ABSTRACT

Peroxidasin (PXDN) is involved in the crosslinking of collagen IV, a major constituent of basement membranes. Disruption of basement membrane integrity as observed in genetic alterations of collagen IV or PXDN can result in developmental defects and diverse pathologies. Hence, the study of PXDN activity in (patho)physiological contexts is highly relevant. So far, measurements of PXDN activity have been reported from purified proteins, cell lysates and de-cellularized extracellular matrix. Here, for the first time we report the measurement of PXDN activity in live cells using the Amplex Red assay with a signal amplifying modification. We observe that bromide addition enhances the obtained signal, most likely due to formation of HOBr. Abrogation of signal amplification by the HOBr scavenger carnosine supports this hypothesis. Both, pharmacological inhibition as well as complementary genetic approaches confirm that the obtained signal is indeed related to PXDN activity. We validate the modified assay by investigating the effect of Brefeldin A, to inhibit the secretory pathway and thus the access of PXDN to the extracellular Amplex Red dye. Our method opens up new possibilities to investigate the activity of PXDN in (patho)physiological contexts.


Subject(s)
Bromides , Extracellular Matrix Proteins , Collagen Type IV/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Peroxidase/metabolism , Peroxidasin
3.
Antioxidants (Basel) ; 10(10)2021 Sep 30.
Article in English | MEDLINE | ID: mdl-34679700

ABSTRACT

Peroxidasin (PXDN) and peroxidasin-like protein (PXDNL) are members of the peroxidase-cyclooxygenase superfamily. PXDN functions in basement membrane synthesis by forming collagen IV crosslinks, while the function of PXDNL remains practically unknown. In this work, we characterized the post-translational proteolytic processing of PXDN and PXDNL. Using a novel knock-in mouse model, we demonstrate that the proteolytic cleavage of PXDN occurs in vivo. With the help of furin-specific siRNA we also demonstrate that the proprotein-convertase, furin participates in the proteolytic processing of PXDN. Furthermore, we demonstrate that only the proteolytically processed PXDN integrates into the extracellular matrix, highlighting the importance of the proteolysis step in PXDN's collagen IV-crosslinking activity. We also provide multiple lines of evidence for the importance of peroxidase activity in the proteolytic processing of PXDN. Finally, we show that PXDNL does not undergo proteolytic processing, despite containing sequence elements efficiently recognized by proprotein convertases. Collectively, our observations suggest a previously unknown protein quality control during PXDN synthesis and the importance of the peroxidase activity of PXDN in this process.

5.
Front Immunol ; 10: 394, 2019.
Article in English | MEDLINE | ID: mdl-30891045

ABSTRACT

Peroxidase enzymes can oxidize a multitude of substrates in diverse biological processes. According to the latest phylogenetic analysis, there are four major heme peroxidase superfamilies. In this review, we focus on certain members of the cyclooxygenase-peroxidase superfamily (also labeled as animal heme peroxidases) and their connection to specific NADPH oxidase enzymes which provide H2O2 for the one- and two-electron oxidation of various peroxidase substrates. The family of NADPH oxidases is a group of enzymes dedicated to the production of superoxide and hydrogen peroxide. There is a handful of known and important physiological functions where one of the seven known human NADPH oxidases plays an essential role. In most of these functions NADPH oxidases provide H2O2 for specific heme peroxidases and the concerted action of the two enzymes is indispensable for the accomplishment of the biological function. We discuss human and other metazoan examples of such cooperation between oxidases and peroxidases and analyze the biological importance of their functional interaction. We also review those oxidases and peroxidases where this kind of partnership has not been identified yet.


Subject(s)
Hemeproteins/metabolism , NADPH Oxidases/metabolism , Peroxidase/metabolism , Animals , Eosinophils/metabolism , Humans , Hydrogen Peroxide/metabolism , Leukocytes/metabolism , Oxidation-Reduction , Prostaglandin-Endoperoxide Synthases/metabolism
6.
Sci Rep ; 8(1): 15255, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30323305

ABSTRACT

The processes involved in renewal of the epithelium that lines the mouse stomach remain unclear. Apart from the cells in the isthmus, several other populations located deeper in the gastric glands have been suggested to contribute to the maintenance of the gastric epithelium. Here, we reveal that Lrig1 is expressed in the basal layer of the forestomach and the lower part of glands in the corpus and pylorus. In the glandular epithelium of the stomach, Lrig1 marks a heterogeneous population comprising mainly non-proliferative cells. Yet, fate-mapping experiments using a knock-in mouse line expressing Cre specifically in Lrig1+ cells demonstrate that these cells are able to contribute to the long-term maintenance of the gastric epithelium. Moreover, when cultured in vitro, cells expressing high level of Lrig1 have much higher organoid forming potential than the corresponding cellular populations expressing lower levels of Lrig1. Taken together, these observations show that Lrig1 is expressed primarily by differentiated cells, but that these cells can be recruited to contribute to the maintenance of the gastric epithelium. This confirms previous observations that cells located in the lower segments of gastric glands can participate in tissue replenishment.


Subject(s)
Biomarkers , Cell Proliferation , Epithelial Cells/metabolism , Gastric Mucosa/metabolism , Membrane Glycoproteins/physiology , Nerve Tissue Proteins/physiology , Animals , Biomarkers/metabolism , Cell Dedifferentiation/genetics , Cell Differentiation/genetics , Cell Proliferation/genetics , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/physiology , Gastric Mucosa/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Stomach/cytology
7.
Redox Biol ; 16: 314-321, 2018 06.
Article in English | MEDLINE | ID: mdl-29573705

ABSTRACT

Collagen IV is a major component of the basement membrane in epithelial tissues. The NC1 domains of collagen IV protomers are covalently linked together through sulfilimine bonds, the formation of which is catalyzed by peroxidasin. Although hydrogen peroxide is essential for this reaction, the exact source of the oxidant remains elusive. Members of the NOX/DUOX NADPH oxidase family are specifically devoted to the production of superoxide and hydrogen peroxide. Our aim in this study was to find out if NADPH oxidases contribute in vivo to the formation of collagen IV sulfilimine crosslinks. We used multiple genetically modified in vivo model systems to provide a detailed assessment of this question. Our data indicate that in various peroxidasin-expressing tissues sulfilimine crosslinks between the NC1 domains of collagen IV can be readily detected in the absence of functioning NADPH oxidases. We also analyzed how subatmospheric oxygen levels influence the collagen IV network in collagen-producing cultured cells with rapid matrix turnover. We showed that collagen IV crosslinks remain intact even under strongly hypoxic conditions. Our hypothesis is that during collagen IV network formation PXDN cooperates with a NOX/DUOX-independent H2O2 source that is functional also at very low ambient oxygen levels.


Subject(s)
Collagen Type IV/metabolism , Epithelial Cells/metabolism , Extracellular Matrix Proteins/metabolism , NADPH Oxidases/metabolism , Peroxidase/metabolism , Basement Membrane/metabolism , Catalysis , Cell Line , Dual Oxidases/metabolism , Extracellular Matrix , Humans , Hydrogen Peroxide/metabolism , Oxygen/metabolism , Superoxides/metabolism , Peroxidasin
8.
Free Radic Biol Med ; 97: 204-211, 2016 08.
Article in English | MEDLINE | ID: mdl-27262981

ABSTRACT

Stimulation of mammalian cells by epidermal growth factor (EGF) elicits complex signaling events, including an increase in hydrogen peroxide (H2O2) production. Understanding the significance of this response is limited by the fact that the source of EGF-induced H2O2 production is unknown. Here we show that EGF-induced H2O2 production in epidermal cell lines is dependent on the agonist-induced calcium signal. We analyzed the expression of NADPH oxidase isoforms and found both A431 and HaCaT cells to express the calcium-sensitive NADPH oxidase, Dual oxidase 1 (Duox1) and its protein partner Duox activator 1 (DuoxA1). Inhibition of Duox1 expression by small interfering RNAs eliminated EGF-induced H2O2 production in both cell lines. We also demonstrate that H2O2 production by Duox1 leads to the oxidation of thioredoxin-1 and the cytosolic peroxiredoxins. Our observations provide evidence for a new signaling paradigm in which changes of intracellular calcium concentration are transformed into redox signals through the calcium-dependent activation of Duox1.


Subject(s)
Dual Oxidases/metabolism , ErbB Receptors/genetics , Hydrogen Peroxide/metabolism , NADPH Oxidases/metabolism , Animals , Calcium/metabolism , Calcium Signaling/genetics , Cytosol/metabolism , Dual Oxidases/genetics , ErbB Receptors/metabolism , Humans , NADPH Oxidases/genetics , Oxidation-Reduction , Protein Isoforms/metabolism , RNA, Small Interfering , Reactive Oxygen Species/metabolism , Signal Transduction , Thioredoxins/genetics , Thioredoxins/metabolism
9.
Trends Pharmacol Sci ; 37(4): 318-327, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26861575

ABSTRACT

Nox/Duox NADPH oxidases are now considered the primary, regulated sources of reactive oxygen species (ROS). These enzymes are expressed in diverse cells and tissues, and their products are essential in several physiological settings. Knockout mouse models are instrumental in identifying the physiological functions of Nox/Duox enzymes as well as in exploring the impact of their pharmacological targeting on disease progression. The currently available data from experiments on knockout animals suggest that the lack of non-phagocytic Nox/Duox enzymes often modifies the course and phenotype in many disease models. Nevertheless, as illustrated by studies on Nox4-deficient animals, the absence of Nox-derived ROS can also lead to aggravated disease manifestation, reinforcing the need for a more balanced view on the role of ROS in health and disease.


Subject(s)
NADH, NADPH Oxidoreductases/deficiency , NADH, NADPH Oxidoreductases/metabolism , Animals , Humans , Mice , Mice, Knockout , NADH, NADPH Oxidoreductases/genetics
10.
Free Radic Biol Med ; 83: 273-82, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25708780

ABSTRACT

Basement membranes provide structural support and convey regulatory signals to cells in diverse tissues. Assembly of collagen IV into a sheet-like network is a fundamental mechanism during the formation of basement membranes. Peroxidasin (PXDN) was recently described to catalyze crosslinking of collagen IV through the formation of sulfilimine bonds. Despite the significance of this pathway in tissue genesis, our understanding of PXDN function is far from complete. In this work we demonstrate that collagen IV crosslinking is a physiological function of mammalian PXDN. Moreover, we carried out structure-function analysis of PXDN to gain a better insight into its role in collagen IV synthesis. We identify conserved cysteines in PXDN that mediate the oligomerization of the protein into a trimeric complex. We also demonstrate that oligomerization is not an absolute requirement for enzymatic activity, but optimal collagen IV coupling is only catalyzed by the PXDN trimers. Localization experiments of different PXDN mutants in two different cell models revealed that PXDN oligomers, but not monomers, adhere on the cell surface in "hot spots," which represent previously unknown locations of collagen IV crosslinking.


Subject(s)
Antigens, Neoplasm/chemistry , Antigens, Neoplasm/metabolism , Basement Membrane/metabolism , Collagen Type IV/chemistry , Extracellular Matrix Proteins/physiology , Extracellular Matrix/metabolism , Peroxidase/physiology , Receptors, Interleukin-1/chemistry , Receptors, Interleukin-1/metabolism , Animals , Antigens, Neoplasm/genetics , Apoptosis , Blotting, Western , Catalysis , Cell Proliferation , Cells, Cultured , Collagen Type IV/metabolism , Cross-Linking Reagents/pharmacology , Female , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Immunoprecipitation , Mice , Mice, Knockout , Peroxidases , Protein Conformation , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Interleukin-1/genetics , Reverse Transcriptase Polymerase Chain Reaction , Structure-Activity Relationship , Peroxidasin
11.
Cardiovasc Res ; 101(3): 393-9, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24253521

ABSTRACT

AIMS: Peroxidases serve diverse biological functions including well-characterized activities in host defence and hormone biosynthesis. More recently, peroxidasin (PXDN) was found to be involved in collagen IV cross-linking in the extracellular matrix (ECM). The aim of this study was to characterize the expression and function of peroxidasin-like protein (PXDNL), a previously unknown peroxidase homologue. METHODS AND RESULTS: We cloned the PXDNL cDNA from the human heart and identified its expression pattern by northern blot, in situ hybridization, and immunohistochemistry. PXDNL is expressed exclusively in the heart and it has evolved to lose its peroxidase activity. The protein is produced by cardiomyocytes and localizes to cell-cell junctions. We also demonstrate that PXDNL can form a complex with PXDN and antagonizes its peroxidase activity. Furthermore, we show an increased expression of PXDNL in the failing myocardium. CONCLUSION: PXDNL is a unique component of the heart with a recently evolved inactivation of peroxidase function. The elevation of PXDNL levels in the failing heart may contribute to ECM dysregulation due to its antagonism of PXDN function.


Subject(s)
Extracellular Matrix Proteins/pharmacology , Gene Expression Regulation , Heart/drug effects , Peroxidase/pharmacology , Animals , Cells, Cultured , Extracellular Matrix/metabolism , Heart Failure/metabolism , Humans , In Situ Hybridization/methods , Oxidation-Reduction/drug effects , RNA, Messenger/metabolism , Peroxidasin
12.
Nat Cell Biol ; 14(7): 753-63, 2012 Jun 24.
Article in English | MEDLINE | ID: mdl-22729083

ABSTRACT

It is becoming clear that interconnected functional gene networks, rather than individual genes, govern stem cell self-renewal and differentiation. To identify epigenetic factors that impact on human epidermal stem cells we performed siRNA-based genetic screens for 332 chromatin modifiers. We developed a Bayesian mixture model to predict putative functional interactions between epigenetic modifiers that regulate differentiation. We discovered a network of genetic interactions involving EZH2, UHRF1 (both known to regulate epidermal self-renewal), ING5 (a MORF complex component), BPTF and SMARCA5 (NURF complex components). Genome-wide localization and global mRNA expression analysis revealed that these factors impact two distinct but functionally related gene sets, including integrin extracellular matrix receptors that mediate anchorage of epidermal stem cells to their niche. Using a competitive epidermal reconstitution assay we confirmed that ING5, BPTF, SMARCA5, EZH2 and UHRF1 control differentiation under physiological conditions. Thus, regulation of distinct gene expression programs through the interplay between diverse epigenetic strategies protects epidermal stem cells from differentiation.


Subject(s)
Cell Differentiation/genetics , Epidermal Cells , Epigenesis, Genetic , Gene Regulatory Networks , Keratinocytes/metabolism , Stem Cells/metabolism , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Antigens, Nuclear/genetics , Antigens, Nuclear/metabolism , Bayes Theorem , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Adhesion/genetics , Cells, Cultured , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Cluster Analysis , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Enhancer of Zeste Homolog 2 Protein , Gene Expression Regulation , Humans , Integrins/genetics , Integrins/metabolism , Models, Genetic , Multiprotein Complexes , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Polycomb Repressive Complex 2 , RNA Interference , RNA, Messenger/metabolism , Stem Cell Niche/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases
13.
Blood ; 119(2): 573-82, 2012 Jan 12.
Article in English | MEDLINE | ID: mdl-22096251

ABSTRACT

Members of the Rac/Rho family of small GTPases play an essential role in phagocytic cells in organization of the actin cytoskeleton and production of toxic oxygen compounds. GTPase-activating proteins (GAPs) decrease the amount of the GTP-bound active form of small GTPases, and contribute to the control of biologic signals. The number of potential Rac/RhoGAPs largely exceeds the number of Rac/Rho GTPases and the expression profile, and their specific role in different cell types is largely unknown. In this study, we report for the first time the properties of full-length ARHGAP25 protein, and show that it is specifically expressed in hematopoietic cells, and acts as a RacGAP both in vitro and in vivo. By silencing and overexpressing the protein in neutrophil model cell lines (PLB-985 and CosPhoxFcγR, respectively) and in primary macrophages, we demonstrate that ARHGAP25 is a negative regulator of phagocytosis acting probably via modulation of the actin cytoskeleton.


Subject(s)
GTPase-Activating Proteins/metabolism , Granulocytes/metabolism , Neutrophils/metabolism , Phagocytosis , cdc42 GTP-Binding Protein/metabolism , rac GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , Actins/metabolism , Animals , Blotting, Northern , COS Cells , Chlorocebus aethiops , Cytoskeleton/metabolism , GTPase-Activating Proteins/antagonists & inhibitors , GTPase-Activating Proteins/genetics , Humans , Immunoblotting , Macrophages/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Superoxides/metabolism
14.
PLoS One ; 6(8): e23653, 2011.
Article in English | MEDLINE | ID: mdl-21886807

ABSTRACT

Motility of normal and transformed cells within and across tissues requires specialized subcellular structures, e.g. membrane ruffles, lamellipodia and podosomes, which are generated by dynamic rearrangements of the actin cytoskeleton. Because the formation of these sub-cellular structures is complex and relatively poorly understood, we evaluated the role of the adapter protein SH3PXD2B [HOFI, fad49, Tks4], which plays a role in the development of the eye, skeleton and adipose tissue. Surprisingly, we find that SH3PXD2B is requisite for the development of EGF-induced membrane ruffles and lamellipodia, as well as for efficient cellular attachment and spreading of HeLa cells. Furthermore, SH3PXD2B is present in a complex with the non-receptor protein tyrosine kinase Src, phosphorylated by Src, which is consistent with SH3PXD2B accumulating in Src-induced podosomes. Furthermore, SH3PXD2B closely follows the subcellular relocalization of cortactin to Src-induced podosomes, EGF-induced membrane ruffles and lamellipodia. Because SH3PXD2B also forms a complex with the C-terminal region of cortactin, we propose that SH3PXD2B is a scaffold protein that plays a key role in regulating the actin cytoskeleton via Src and cortactin.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Movement , Pseudopodia/metabolism , Sequence Homology, Amino Acid , src Homology Domains , Actins/metabolism , Cortactin/metabolism , ErbB Receptors/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , HeLa Cells , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Macrophages/cytology , Macrophages/metabolism , Phosphatidylinositols/metabolism , Protein Binding , Protein Transport
15.
J Biol Chem ; 281(9): 6096-105, 2006 Mar 03.
Article in English | MEDLINE | ID: mdl-16380373

ABSTRACT

Sec14 protein was first identified in Saccharomyces cerevisiae, where it serves as a phosphatidylinositol transfer protein that is essential for the transport of secretory proteins from the Golgi complex. A protein domain homologous to Sec14 was identified in several mammalian proteins that regulates Rho GTPases, including exchange factors and GTPase activating proteins. P50RhoGAP, the first identified GTPase activating protein for Rho GTPases, is composed of a Sec14-like domain and a Rho-GTPase activating protein (GAP) domain. The biological function of its Sec14-like domain is still unknown. Here we show that p50RhoGAP is present on endosomal membranes, where it colocalizes with internalized transferrin receptor. We demonstrate that the Sec14-like domain of P50RhoGAP is responsible for the endosomal targeting of the protein. We also show that overexpression of p50RhoGAP or its Sec14-like domain inhibits transferrin uptake. Furthermore, both P50RhoGAP and its Sec14-like domain show colocalization with small GTPases Rab11 and Rab5. We measured bioluminescence resonance energy transfer between p50RhoGAP and Rab11, indicating that these proteins form molecular complex in vivo on endosomal membranes. The interaction was mediated by the Sec 14-like domain of p50RhoGAP. Our results indicate that Sec14-like domain, which was previously considered as a phospholipid binding module, may have a role in the mediation of protein-protein interactions. We suggest that p50RhoGAP provides a link between Rab and Rho GTPases in the regulation of receptor-mediated endocytosis.


Subject(s)
Endosomes/metabolism , GTPase-Activating Proteins/metabolism , rab GTP-Binding Proteins/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Endocytosis/physiology , GTPase-Activating Proteins/genetics , HeLa Cells , Humans , Intracellular Membranes/metabolism , Microtubules/metabolism , Phospholipid Transfer Proteins/genetics , Phospholipid Transfer Proteins/metabolism , Receptors, Transferrin/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Transferrin/metabolism , cdc42 GTP-Binding Protein/metabolism , rab GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/genetics
16.
Am J Physiol Cell Physiol ; 289(3): C673-85, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15857905

ABSTRACT

Myosin-based contractility plays important roles in the regulation of epithelial functions, particularly paracellular permeability. However, the triggering factors and the signaling pathways that control epithelial myosin light chain (MLC) phosphorylation have not been elucidated. Herein we show that plasma membrane depolarization provoked by distinct means, including high extracellular K(+), the lipophilic cation tetraphenylphosphonium, or the ionophore nystatin, induced strong diphosphorylation of MLC in kidney epithelial cells. In sharp contrast to smooth muscle, depolarization of epithelial cells did not provoke a Ca(2+) signal, and removal of external Ca(2+) promoted rather than inhibited MLC phosphorylation. Moreover, elevation of intracellular Ca(2+) did not induce significant MLC phosphorylation, and the myosin light chain kinase (MLCK) inhibitor ML-7 did not prevent the depolarization-induced MLC response, suggesting that MLCK is not a regulated element in this process. Instead, the Rho-Rho kinase (ROK) pathway is the key mediator because 1) depolarization stimulated Rho and induced its peripheral translocation, 2) inhibition of Rho by Clostridium difficile toxin B or C3 transferase abolished MLC phosphorylation, and 3) the ROK inhibitor Y-27632 suppressed the effect. Importantly, physiological depolarizing stimuli were able to activate the same pathway: L-alanine, the substrate of the electrogenic Na(+)-alanine cotransporter, stimulated Rho and induced Y-27632-sensitive MLC phosphorylation in a Na(+)-dependent manner. Together, our results define a novel mode of the regulation of MLC phosphorylation in epithelial cells, which is depolarization triggered and Rho-ROK-mediated but Ca(2+) signal independent. This pathway may be a central mechanism whereby electrogenic transmembrane transport processes control myosin phosphorylation and thereby regulate paracellular transport.


Subject(s)
Epithelial Cells/enzymology , Kidney Tubules, Distal/metabolism , Myosin Light Chains/metabolism , Protein Serine-Threonine Kinases/metabolism , rho GTP-Binding Proteins/metabolism , Amino Acid Transport Systems, Neutral/metabolism , Animals , Calcium/metabolism , Cell Membrane/metabolism , Dogs , Electric Stimulation , Intracellular Signaling Peptides and Proteins , Kidney Tubules, Distal/cytology , LLC-PK1 Cells , Membrane Potentials/physiology , Phosphorylation , Potassium/metabolism , Swine , Symporters/metabolism , rho-Associated Kinases
17.
Am J Physiol Cell Physiol ; 285(3): C555-66, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12748065

ABSTRACT

Hyperosmotic stress initiates adaptive responses, including phosphorylation of myosin light chain (MLC) and concomitant activation of Na+-K+-Cl- cotransporter (NKCC). Because the small GTPase Rho is a key regulator of MLC phosphorylation, we investigated 1) whether Rho is activated by hyperosmotic stress, and if so, what the triggering factors are, and 2) whether the Rho/Rho kinase (ROK) pathway is involved in MLC phosphorylation and NKCC activation. Rho activity was measured in tubular epithelial cells by affinity pulldown assay. Hyperosmolarity induced rapid (<1 min) and sustained (>20 min) Rho activation that was proportional to the osmotic concentration and reversed within minutes upon restoration of isotonicity. Both decreased cell volume at constant ionic strength and elevated total ionic strength at constant cell volume were capable of activating Rho. Changes in [Na+] and [K+] at normal total salinity failed to activate Rho, and Cl- depletion did not affect the hyperosmotic response. Thus alterations in cellular volume and ionic strength but not individual ion concentrations seem to be the critical triggering factors. Hyperosmolarity induced mono- and diphosphorylation of MLC, which was abrogated by the Rho-family blocker Clostridium toxin B. ROK inhibitor Y-27632 suppressed MLC phosphorylation under isotonic conditions and prevented its rise over isotonic levels in hypertonically stimulated cells. ML-7 had a smaller inhibitory effect. In contrast, it abolished the hypertonic activation of NKCC, whereas Y-27632 failed to inhibit this response. Thus hyperosmolarity activates Rho, and Rho/ROK pathway contributes to basal and hyperosmotic MLC phosphorylation. However, the hypertonic activation of NKCC is ROK independent, implying that the ROK-dependent component of MLC phosphorylation can be uncoupled from NKCC activation.


Subject(s)
Myosin Light Chains/metabolism , Protein Serine-Threonine Kinases/metabolism , Sodium-Potassium-Chloride Symporters/metabolism , Water-Electrolyte Balance/physiology , Animals , Hypertonic Solutions/pharmacology , Intracellular Signaling Peptides and Proteins , LLC-PK1 Cells , Osmotic Pressure , Phosphorylation , Solute Carrier Family 12, Member 2 , Swine , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases
18.
Am J Physiol Renal Physiol ; 284(5): F911-24, 2003 May.
Article in English | MEDLINE | ID: mdl-12505862

ABSTRACT

New research suggests that, during tubulointerstitial fibrosis, alpha-smooth muscle actin (SMA)-expressing mesenchymal cells might derive from the tubular epithelium via epithelial-mesenchymal transition (EMT). Although transforming growth factor-beta(1) (TGF-beta(1)) plays a key role in EMT, the underlying cellular mechanisms are not well understood. Here we characterized TGF-beta(1)-induced EMT in LLC-PK(1) cells and examined the role of the small GTPase Rho and its effector, Rho kinase, (ROK) in the ensuing cytoskeletal remodeling and SMA expression. TGF-beta(1) treatment caused delocalization and downregulation of cell contact proteins (ZO-1, E-cadherin, beta-catenin), cytoskeleton reorganization (stress fiber assembly, myosin light chain phosphorylation), and robust SMA synthesis. TGF-beta(1) induced a biphasic Rho activation. Stress fiber assembly was prevented by the Rho-inhibiting C3 transferase and by dominant negative (DN) ROK. The SMA promoter was activated strongly by constitutively active Rho but not ROK. Accordingly, TGF-beta(1)-induced SMA promoter activation was potently abrogated by two Rho-inhibiting constructs, C3 transferase and p190RhoGAP, but not by DN-ROK. Truncation analysis showed that the first CC(A/T)richGG (CArG B) serum response factor-binding cis element is essential for the Rho responsiveness of the SMA promoter. Thus Rho plays a dual role in TGF-beta(1)-induced EMT of renal epithelial cells. It is indispensable both for cytoskeleton remodeling and for the activation of the SMA promoter. The cytoskeletal effects are mediated via the Rho/ROK pathway, whereas the transcriptional effects are partially ROK independent.


Subject(s)
Actins/metabolism , Acute-Phase Proteins/physiology , Muscle, Smooth/metabolism , Transforming Growth Factor beta/physiology , Animals , Cell Differentiation/physiology , Cytoskeleton/physiology , Epithelial Cells/cytology , LLC-PK1 Cells , Mesoderm/cytology , Swine , Transforming Growth Factor beta1
SELECTION OF CITATIONS
SEARCH DETAIL
...