Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 26
1.
Thromb Haemost ; 116(6): 1122-1130, 2016 Nov 30.
Article En | MEDLINE | ID: mdl-27580629

Our previous studies revealed that the interaction of fibrin with the very low density lipoprotein receptor (VLDLR) promotes transendothelial migration of leukocytes and thereby inflammation, and localised the fibrin-binding site to CR-domains 2-4 of this receptor. In the present study, we tested interaction of three anti-VLDLR monoclonal antibodies, mAb 1H10, 1H5, and 5F3, with recombinant fragments of VLDLR containing various combinations of its CR-domains and found that the epitopes for mAb 1H10 and mAb 1H5 overlap with the fibrin-binding site of VLDLR. Based on these findings, we hypothesised that mAb 1H10 and mAb 1H5 should inhibit fibrin-VLDLR interaction and modulate leukocyte transmigration. To test this hypothesis, we first demonstrated that these monoclonal antibodies both have high affinity to the fibrin-binding fragments of the VLDL receptor and efficiently inhibit interaction between the VLDLR-binding fragment of fibrin and the fibrin-binding fragments of VLDLR. Next, in the in vitro experiments using leukocyte transendothelial migration assay we found that both monoclonal antibodies efficiently inhibit leukocyte transmigration induced by fibrin mimetic NDSK-II. Finally, in vivo experiments using mouse model of peritonitis revealed that mAb 1H10 and mAb 1H5 both significantly reduce infiltration of leukocytes into the peritoneum. Furthermore, our experiments using mouse model of myocardial ischemia-reperfusion injury revealed that both monoclonal antibodies significantly reduce myocardial injury induced by ischaemia-reperfusion. Thus, the results obtained indicate that monoclonal antibodies 1H10 and 1H5 are novel specific inhibitors of fibrin-VLDLR-dependent leukocyte transmigration pathway. They may represent potential therapeutics for treatment of fibrin-dependent inflammation including myocardial ischaemia-reperfusion injury.


Antibodies, Monoclonal/immunology , Fibrin/metabolism , Leukocytes/cytology , Receptors, LDL/immunology , Transendothelial and Transepithelial Migration , Animals , Epitope Mapping , Human Umbilical Vein Endothelial Cells , Humans , Mice, Inbred C57BL , Receptors, LDL/metabolism
2.
Exp Dermatol ; 24(6): 455-61, 2015 Jun.
Article En | MEDLINE | ID: mdl-25808322

Expression of the metastasis suppressor NME1 in melanoma is associated with reduced cellular motility and invasion in vitro and metastasis in vivo, but the underlying molecular mechanisms are not completely understood. Herein, we report a novel mechanism through which NME1 controls melanoma cell morphology via upregulation of the extracellular matrix (ECM) protein fibronectin. Expression of NME1 strongly suppressed cell motility in melanoma cell lines 1205LU and M14. The resulting sedentary phenotype was associated with a more flattened appearance and marked increases in actin stress fibre and focal adhesion formation. NME1-induced focal adhesions were colocalized with dense deposits of fibronectin, which were absent or minimal in the corresponding NME1-deficient parental lines. NME1 was a strong inducer of fibronectin mRNA and protein expression, shown with reciprocal approaches of forced NME1 expression and shRNA-mediated knock-down. Increased synthesis and ECM deposition of fibronectin was necessary for NME1-induced cell spreading, as knock-down of fibronectin opposed the effects of NME1 on cell morphology. Fibronectin knock-down also reversed the ability of NME1 to promote aggregation when cells were plated on a non-adherent substratum. Similarly, inhibiting activation of the fibronectin receptor integrin α4ß1 with an anti-α4 antibody reversed the motility-suppressing effect of NME1. A positive correlation was observed between NME1 and fibronectin mRNA in clinical biopsies of normal skin, benign nevi and primary melanomas, but not in metastatic forms, suggesting the NME1/fibronectin axis represents a barrier to melanoma progression. In summary, these findings indicate fibronectin is an important effector of the motility-suppressing function of NME1 in melanoma cells.


Cell Adhesion/physiology , Cell Movement/physiology , Fibronectins/physiology , Melanoma/pathology , NM23 Nucleoside Diphosphate Kinases/physiology , Skin Neoplasms/pathology , Cell Line, Tumor , Disease Progression , Extracellular Matrix/pathology , Extracellular Matrix/physiology , Fibronectins/genetics , Gene Expression Regulation, Neoplastic/physiology , Humans , In Vitro Techniques , Melanoma/physiopathology , NM23 Nucleoside Diphosphate Kinases/genetics , Neoplasm Metastasis/pathology , Neoplasm Metastasis/physiopathology , RNA, Messenger/genetics , RNA, Messenger/physiology , Signal Transduction/physiology , Skin Neoplasms/physiopathology
3.
Naunyn Schmiedebergs Arch Pharmacol ; 388(2): 199-206, 2015 Feb.
Article En | MEDLINE | ID: mdl-25017017

The NME1 gene represents the prototypical metastasis suppressor, whose expression inhibits cell motility and metastasis without impact on primary tumor growth in a number of different human cancers. This report outlines our recent efforts to define the molecular mechanisms through which NME1 both suppresses cell motility and promotes genomic integrity in the setting of human melanoma. Forced NME1 expression in a variety of melanoma-derived cell lines was shown to induce dynamic changes in cell morphology and reorganization of the actin cytoskeleton, with formation of a network of thick stress fibers and assembly of fibronectin fibrils at large focal adhesions. Moreover, NME1 expression results in adhesion reprogramming through an impact on integrin repertoire and focal adhesion dynamics. Having previously demonstrated that NME1 expression promotes repair of DNA damage induced by ultraviolet radiation (UVR) in both yeast and mammalian cells, probably via the nucleotide excision repair pathway, we have more recently demonstrated that NME1 is rapidly recruited to double-strand breaks. This preliminary result represents the first evidence of direct interactions between NME1 and DNA in the context of DNA repair and has set the stage for current efforts to probe its functional interactions with double-strand break repair pathways. Discussed herein are molecular models to explain the interactions of NME1 with such diverse cellular functions as cell motility and DNA repair, potentially through its nucleoside diphosphate kinase and 3'-5' exonuclease activities.


Melanoma , NM23 Nucleoside Diphosphate Kinases/metabolism , Skin Neoplasms , Actins/metabolism , Animals , Cell Movement , DNA Repair , Focal Adhesions , Genomic Instability , Humans , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
4.
J Cell Sci ; 127(Pt 16): 3578-92, 2014 Aug 15.
Article En | MEDLINE | ID: mdl-24963132

Filamin C (FLNc) and Xin actin-binding repeat-containing proteins (XIRPs) are multi-adaptor proteins that are mainly expressed in cardiac and skeletal muscles and which play important roles in the assembly and repair of myofibrils and their attachment to the membrane. We identified the dystrophin-binding protein aciculin (also known as phosphoglucomutase-like protein 5, PGM5) as a new interaction partner of FLNc and Xin. All three proteins colocalized at intercalated discs of cardiac muscle and myotendinous junctions of skeletal muscle, whereas FLNc and aciculin also colocalized in mature Z-discs. Bimolecular fluorescence complementation experiments in developing cultured mammalian skeletal muscle cells demonstrated that Xin and aciculin also interact in FLNc-containing immature myofibrils and areas of myofibrillar remodeling and repair induced by electrical pulse stimulation (EPS). Fluorescence recovery after photobleaching (FRAP) experiments showed that aciculin is a highly dynamic and mobile protein. Aciculin knockdown in myotubes led to failure in myofibril assembly, alignment and membrane attachment, and a massive reduction in myofibril number. A highly similar phenotype was found upon depletion of aciculin in zebrafish embryos. Our results point to a thus far unappreciated, but essential, function of aciculin in myofibril formation, maintenance and remodeling.


Cytoskeletal Proteins/metabolism , DNA-Binding Proteins/metabolism , Filamins/metabolism , Myofibrils/metabolism , Nuclear Proteins/metabolism , Phosphoglucomutase/metabolism , Animals , Cell Line , Cells, Cultured , Cytoskeletal Proteins/genetics , DNA-Binding Proteins/genetics , Filamins/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Myoblasts/metabolism , Myofibrils/genetics , Nuclear Proteins/genetics , Phosphoglucomutase/genetics , Protein Binding
5.
J Am Heart Assoc ; 3(2): e000599, 2014 Apr 10.
Article En | MEDLINE | ID: mdl-24721796

BACKGROUND: Vascular aging is closely associated with increased vascular stiffness. It has recently been demonstrated that decreased nitric oxide (NO)-induced S-nitrosylation of tissue transglutaminase (TG2) contributes to age-related vascular stiffness. In the current study, we tested the hypothesis that exercise restores NO signaling and attenuates vascular stiffness by decreasing TG2 activity and cross-linking in an aging rat model. METHODS AND RESULTS: Rats were subjected to 12 weeks of moderate aerobic exercise. Aging was associated with diminished phosphorylated endothelial nitric oxide synthase and phosphorylated vasodilator-stimulated phosphoprotein abundance, suggesting reduced NO signaling. TG2 cross-linking activity was significantly increased in old animals, whereas TG2 abundance remained unchanged. These alterations were attenuated in the exercise cohort. Simultaneous measurement of blood pressure and pulse wave velocity (PWV) demonstrated increased aortic stiffness in old rats, compared to young, at all values of mean arterial pressure (MAP). The PWV-MAP correlation in the old sedentary and old exercise cohorts was similar. Tensile testing of the vessels showed increased stiffness of the aorta in the old phenotype with a modest restoration of mechanical properties toward the young phenotype with exercise. CONCLUSIONS: Increased vascular stiffness during aging is associated with decreased TG2 S-nitrosylation, increased TG2 cross-linking activity, and increased vascular stiffness likely the result of decreased NO bioavailability. In this study, a brief period of moderate aerobic exercise enhanced NO signaling, attenuated TG cross-linking activity, and reduced ex vivo tensile properties, but failed to reverse functional vascular stiffness in vivo, as measured by PWV.


Aging/metabolism , Aorta/enzymology , Aorta/physiopathology , GTP-Binding Proteins/metabolism , Physical Exertion , Transglutaminases/metabolism , Vascular Stiffness , Age Factors , Animals , Arterial Pressure , Cell Adhesion Molecules/metabolism , Male , Microfilament Proteins/metabolism , Nitric Oxide Synthase Type III/metabolism , Phosphoproteins/metabolism , Phosphorylation , Protein Glutamine gamma Glutamyltransferase 2 , Pulse Wave Analysis , Rats , Rats, Inbred F344 , Running , Signal Transduction , Tensile Strength
6.
Physiol Rev ; 94(2): 383-417, 2014 Apr.
Article En | MEDLINE | ID: mdl-24692352

Transglutaminases (TGs) are multifunctional proteins having enzymatic and scaffolding functions that participate in regulation of cell fate in a wide range of cellular systems and are implicated to have roles in development of disease. This review highlights the mechanism of action of these proteins with respect to their structure, impact on cell differentiation and survival, role in cancer development and progression, and function in signal transduction. We also discuss the mechanisms whereby TG level is controlled and how TGs control downstream targets. The studies described herein begin to clarify the physiological roles of TGs in both normal biology and disease states.


Signal Transduction , Transglutaminases/metabolism , Animals , Cell Differentiation , Gene Expression Regulation, Enzymologic , Humans , Neoplasms/enzymology , Neoplasms/pathology , Transcription, Genetic , Transglutaminases/genetics
7.
PLoS One ; 9(2): e89285, 2014.
Article En | MEDLINE | ID: mdl-24586660

Tissue transglutaminase (TG2) mediates protein crosslinking through generation of ε-(γ-glutamyl) lysine isopeptide bonds and promotes cell adhesion through interaction with fibronectin (FN) and integrins. Cell adhesion to the peritoneal matrix regulated by TG2 facilitates ovarian cancer dissemination. Therefore, disruption of the TG2-FN complex by small molecules may inhibit cell adhesion and metastasis. A novel high throughput screening (HTS) assay based on AlphaLISA™ technology was developed to measure the formation of a complex between His-TG2 and the biotinylated FN fragment that binds TG2 and to discover small molecules that inhibit this protein-protein interaction. Several hits were identified from 10,000 compounds screened. The top candidates selected based on >70% inhibition of the TG2/FN complex formation were confirmed by using ELISA and bioassays measuring cell adhesion, migration, invasion, and proliferation. In conclusion, the AlphaLISA bead format assay measuring the TG2-FN interaction is robust and suitable for HTS of small molecules. One compound identified from the screen (TG53) potently inhibited ovarian cancer cell adhesion to FN, cell migration, and invasion and could be further developed as a potential inhibitor for ovarian cancer dissemination.


Fibronectins/antagonists & inhibitors , GTP-Binding Proteins/antagonists & inhibitors , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Protein Interaction Maps/drug effects , Small Molecule Libraries/pharmacology , Transglutaminases/antagonists & inhibitors , Blotting, Western , Cell Adhesion/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Enzyme-Linked Immunosorbent Assay , Female , Fibronectins/metabolism , GTP-Binding Proteins/metabolism , High-Throughput Screening Assays , Humans , Ovarian Neoplasms/pathology , Protein Glutamine gamma Glutamyltransferase 2 , Transglutaminases/metabolism , Tumor Cells, Cultured , Wound Healing/drug effects
8.
J Vasc Res ; 51(6): 418-28, 2014.
Article En | MEDLINE | ID: mdl-25612735

BACKGROUND: Phenotypic switch of vascular smooth muscle cells (VSMCs) accompanies neointima formation and associates with vascular diseases. Platelet-derived growth factor (PDGF)-induced activation of PDGFR/Akt1 and ß-catenin signaling pathways in VSMCs has been implicated in vessel occlusion. Transglutaminase 2 (TG2) regulates these pathways and its levels are increased in the neointima. OBJECTIVE: The aim of this study was to evaluate the role of TG2 in PDGF/ß-catenin signaling cross-talk and assess its contribution to neointima. METHODS: Aortic VSMCs from wild-type and TG2 knockout mice were tested in vitro for levels of VSMC markers, proliferation, migration and PDGF-induced activation of PDGFR/Akt1 and ß-catenin pathways. Neointima in these mice was studied ex vivo in coronary vessels using a heart slice model and in vivo using a carotid artery ligation model. RESULTS: Genetic deletion of TG2 attenuated the PDGF-induced phenotypic switch of aortic VSMCs, reduced their proliferation and migration rates, and inhibited PDGF-induced activation of PDGFR/Akt1 and ß-catenin pathways in both ex vivo and in vivo neointima models. Importantly, genetic deletion of TG2 also markedly attenuated vessel occlusion. CONCLUSIONS: TG2 promotes neointima formation by mediating the PDGF-induced activation of the PDGFR/Akt1 and ß-catenin pathways in VSMCs. This study identifies TG2 as a potential therapeutic target for blocking neointima in blood vessels.


Carotid Stenosis/enzymology , Coronary Stenosis/enzymology , GTP-Binding Proteins/metabolism , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Neointima , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-sis/pharmacology , Receptors, Platelet-Derived Growth Factor/agonists , Signal Transduction/drug effects , Transglutaminases/metabolism , beta Catenin/metabolism , Animals , Becaplermin , Carotid Stenosis/genetics , Carotid Stenosis/pathology , Carotid Stenosis/prevention & control , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Coronary Stenosis/pathology , Coronary Stenosis/prevention & control , Coronary Vessels/drug effects , Coronary Vessels/enzymology , Coronary Vessels/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , GTP-Binding Proteins/deficiency , GTP-Binding Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/pathology , Phenotype , Protein Glutamine gamma Glutamyltransferase 2 , Receptors, Platelet-Derived Growth Factor/metabolism , Time Factors , Transglutaminases/deficiency , Transglutaminases/genetics
9.
J Vasc Res ; 50(5): 383-95, 2013.
Article En | MEDLINE | ID: mdl-23988702

Inward remodeling of small arteries occurs after prolonged vasoconstriction, low blood flow, and in several models of hypertension. The cross-linking enzyme, transglutaminases 2 (TG2), is able to induce inward remodeling and stiffening of arteries. The activity of TG2 is dependent on its conformation, which can be open or closed, and on its redox state. Several factors have been shown to be involved in modulating TG2 activity, including Ca(2+) and GTP/GDP concentrations, as well as the redox state of the environment. This review introduces the hypothesis that mechanical force could be involved in regulating the activity of TG2 during inward remodeling by promoting its open and reduced active state. Several aspects of TG2, such as its structure and localization, are assessed in order to provide arguments that support the hypothesis. We conclude that a direct activation of TG2 by mechanical force exerted by smooth muscle cells may explain the link between smooth muscle activation and inward remodeling, as observed in several physiological and pathological conditions.


Arteries/enzymology , GTP-Binding Proteins/metabolism , Transglutaminases/metabolism , Animals , Arteries/physiology , Calcium/metabolism , Cell Adhesion/physiology , Disulfides/metabolism , Enzyme Activation , Fibronectins/metabolism , GTP-Binding Proteins/antagonists & inhibitors , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , Humans , Integrin alpha5beta1/metabolism , Integrins/metabolism , Mice , Muscle, Smooth/cytology , Muscle, Smooth/physiology , Protein Conformation/drug effects , Protein Glutamine gamma Glutamyltransferase 2 , Stress, Mechanical , Talin/metabolism , Transglutaminases/antagonists & inhibitors , Vasoconstriction/physiology , Vasodilation/physiology , Vinculin/metabolism
10.
Amino Acids ; 44(1): 261-9, 2013 Jan.
Article En | MEDLINE | ID: mdl-21984378

The multifunctional enzyme tissue transglutaminase (TG2) contributes to the development and progression of several cardiovascular diseases. Extracellular rather than intracellular TG2 is enzymatically active, however, the mechanism by which it is exported out of the cell remains unknown. Nitric oxide (NO) is shown to constrain TG2 externalization in endothelial and fibroblast cells. Here, we examined the role of both exogenous and endogenous (endothelial cell-derived) NO in regulating TG2 localization in vascular cells and tissue. NO synthase inhibition in endothelial cells (ECs) using N-nitro L-arginine methyl ester (L-NAME) led to a time-dependent decrease in S-nitrosation and increase in externalization of TG2. Laminar shear stress led to decreased extracellular TG2 in ECs. S-nitrosoglutathione treatment led to decreased activity and externalization of TG2 in human aortic smooth muscle and fibroblast (IMR90) cells. Co-culture of these cells with ECs resulted in increased S-nitrosation and decreased externalization and activity of TG2, which was reversed by L-NAME. Aged Fischer 344 rats had higher tissue scaffold-associated TG2 compared to young. NO regulates intracellular versus extracellular TG2 localization in vascular cells and tissue, likely via S-nitrosation. This in part, explains increased TG2 externalization and activity in aging aorta.


Endothelial Cells/enzymology , Myocytes, Smooth Muscle/enzymology , Nitric Oxide/physiology , Transglutaminases/metabolism , Age Factors , Animals , Aorta/cytology , Cell Line , Coculture Techniques , Endothelium, Vascular/cytology , Extracellular Matrix/enzymology , Fibroblasts/enzymology , GTP-Binding Proteins , Humans , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/physiology , Nitrosation , Protein Glutamine gamma Glutamyltransferase 2 , Protein Transport , Rats , Rats, Inbred F344
12.
Int Rev Cell Mol Biol ; 294: 1-97, 2012.
Article En | MEDLINE | ID: mdl-22364871

Transglutaminase 2 (TG2 or tissue transglutaminase) is a highly complex multifunctional protein that acts as transglutaminase, GTPase/ATPase, protein disulfide isomerase, and protein kinase. Moreover, TG2 has many well-documented nonenzymatic functions that are based on its noncovalent interactions with multiple cellular proteins. A vast array of biochemical activities of TG2 accounts for its involvement in a variety of cellular processes, including adhesion, migration, growth, survival, apoptosis, differentiation, and extracellular matrix organization. In turn, the impact of TG2 on these processes implicates this protein in various physiological responses and pathological states, contributing to wound healing, inflammation, autoimmunity, neurodegeneration, vascular remodeling, tumor growth and metastasis, and tissue fibrosis. TG2 is ubiquitously expressed and is particularly abundant in endothelial cells, fibroblasts, osteoblasts, monocytes/macrophages, and smooth muscle cells. The protein is localized in multiple cellular compartments, including the nucleus, cytosol, mitochondria, endolysosomes, plasma membrane, and cell surface and extracellular matrix, where Ca(2+), nucleotides, nitric oxide, reactive oxygen species, membrane lipids, and distinct protein-protein interactions in the local microenvironment jointly regulate its activities. In this review, we discuss the complex biochemical activities and molecular interactions of TG2 in the context of diverse subcellular compartments and evaluate its wide ranging and cell type-specific biological functions and their regulation.


Gene Expression Regulation, Enzymologic/physiology , Transglutaminases/genetics , Transglutaminases/metabolism , Animals , Apoptosis/physiology , Cell Adhesion/physiology , Cell Compartmentation/physiology , Cell Differentiation/physiology , Cell Enlargement , Cell Movement/physiology , Cell Survival/physiology , Extracellular Matrix/enzymology , GTP-Binding Proteins , Humans , Protein Glutamine gamma Glutamyltransferase 2
13.
J Cell Physiol ; 227(5): 2089-96, 2012 May.
Article En | MEDLINE | ID: mdl-21769866

Although the pivotal role of platelet derived growth factor (PDGF)-mediated signaling in vascular diseases was demonstrated, the pathophysiological mechanisms driving its over-activation remain incompletely understood. Tissue transglutaminase (tTG) is a multifunctional protein expressed in the vasculature, including smooth muscle cells (SMCs), and implicated in several vascular pathologies. The goal of this study is to define the regulation of PDGF-BB/PDGFRß-induced signaling pathways and cell responses by tTG in vascular SMCs. We find that in human aortic SMCs, shRNA-mediated depletion and over-expression of tTG reveals its ability to down-regulate PDGFRß levels and induce receptor clustering. In these cells, tTG specifically amplifies the activation of PDGFRß and its multiple downstream signaling targets in response to PDGF-BB. Furthermore, tTG promotes dedifferentiation and increases survival, proliferation, and migration of human aortic SMCs mediated by this growth factor. Finally, PDGF-BB stimulates tTG expression in human aortic SMCs in culture and in the blood vessels in response to injury. Together, our results show that tTG in vascular SMCs acts as a principal enhancer within the PDGF-BB/PDGFRß signaling axis involved in phenotypic modulation of these cells, thereby suggesting a novel role for this protein in the progression of vascular diseases.


Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Proto-Oncogene Proteins c-sis/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Signal Transduction/physiology , Transglutaminases/metabolism , Animals , Becaplermin , Cell Movement , Cell Proliferation , Cell Survival , Humans , Male , Mice , Mice, Inbred C57BL , Myocytes, Smooth Muscle/cytology , Proto-Oncogene Proteins c-sis/genetics , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Receptor, Platelet-Derived Growth Factor beta/genetics
14.
Commun Integr Biol ; 4(5): 584-6, 2011 Sep.
Article En | MEDLINE | ID: mdl-22046470

Nitric oxide (NO) is an endogenous second messenger which acts as a potent vasodilator, anti-inflammatory, anti-thrombotic and pro-angiogenic agent in the vasculature. Recent studies revealed that the effects of NO on blood vessels are mediated in part by its ability to regulate protein trafficking machinery and vesicle-based exocytosis in vascular cells. Specifically, NO-dependent S-nitrosylation of N-ethylmaleimide sensitive factor (NSF), an ATPase that enables membrane fusion, was shown to inhibit exocytosis of vesicular secretory compartments such as endothelial Weibel-Palade bodies, platelet alpha granules and cytolytic granules from activated lymphocytes. Tissue transglutaminase (tTG or TG2) is a multifunctional protein synthesized and secreted by various cell types in the vasculature, which is involved in multiple vascular diseases, including atherosclerosis, vascular calcification and age-dependent aortic stiffening. Our recent findings indicate that tTG is delivered to the cell surface and the extracellular matrix (ECM) via a non-classical ER/Golgi-independent secretion pathway, which depends on the recycling endosomes and the NSF function. Here we report that NO attenuates the unconventional secretion of tTG in human aortic endothelial cells. NO-dependent downregulation of extracellular tTG levels via inhibition of its secretion might be a part of general physiological mechanism which limits externalization of adhesive, pro-inflammatory and thrombogenic proteins in the vasculature.

15.
FEBS J ; 278(24): 4704-16, 2011 Dec.
Article En | MEDLINE | ID: mdl-21902810

Tissue transglutaminase (TG2) is a ubiquitously expressed member of the transglutaminase family of Ca(2+)-dependent crosslinking enzymes. Unlike other family members, TG2 is a multifunctional protein, which has several other well documented enzymatic and non-enzymatic functions. A significant body of evidence accumulated over the last decade reveals multiple and complex activities of this protein on the cell surface and in the extracellular matrix (ECM), including its role in the regulation of cell-ECM interactions and outside-in signaling by several types of transmembrane receptors. Moreover, recent findings indicate a dynamic regulation of the levels and functions of extracellular TG2 by several complementary mechanisms. This review summarizes and assesses recent research into the emerging functions and regulation of extracellular TG2.


Extracellular Matrix/metabolism , GTP-Binding Proteins/physiology , Transglutaminases/physiology , Cell Adhesion/physiology , Cross-Linking Reagents/metabolism , Cysteine/metabolism , Enzyme Activation , Fibronectins/metabolism , GTP-Binding Proteins/metabolism , Humans , Integrins/metabolism , Low Density Lipoprotein Receptor-Related Protein-5/physiology , Low Density Lipoprotein Receptor-Related Protein-6/physiology , Macrophages/physiology , Matrix Metalloproteinases, Membrane-Associated/metabolism , Mesenchymal Stem Cell Transplantation , Protein Conformation/drug effects , Protein Glutamine gamma Glutamyltransferase 2 , Protein Transport/physiology , Receptors, Growth Factor/physiology , Signal Transduction/physiology , Syndecan-4/physiology , Transglutaminases/metabolism , beta Catenin/physiology
16.
PLoS One ; 6(4): e19414, 2011 Apr 27.
Article En | MEDLINE | ID: mdl-21556374

Although endosomal compartments have been suggested to play a role in unconventional protein secretion, there is scarce experimental evidence for such involvement. Here we report that recycling endosomes are essential for externalization of cytoplasmic secretory protein tissue transglutaminase (tTG). The de novo synthesized cytoplasmic tTG does not follow the classical ER/Golgi-dependent secretion pathway, but is targeted to perinuclear recycling endosomes, and is delivered inside these vesicles prior to externalization. On its route to the cell surface tTG interacts with internalized ß1 integrins inside the recycling endosomes and is secreted as a complex with recycled ß1 integrins. Inactivation of recycling endosomes, blocking endosome fusion with the plasma membrane, or downregulation of Rab11 GTPase that controls outbound trafficking of perinuclear recycling endosomes, all abrogate tTG secretion. The initial recruitment of cytoplasmic tTG to recycling endosomes and subsequent externalization depend on its binding to phosphoinositides on endosomal membranes. These findings begin to unravel the unconventional mechanism of tTG secretion which utilizes the long loop of endosomal recycling pathway and indicate involvement of endosomal trafficking in non-classical protein secretion.


Endosomes/metabolism , Phospholipids/metabolism , Transglutaminases/metabolism , Animals , Mice , NIH 3T3 Cells
17.
Circ Res ; 107(1): 117-25, 2010 Jul 09.
Article En | MEDLINE | ID: mdl-20489165

RATIONALE: Although an age-related decrease in NO bioavailability contributes to vascular stiffness, the underlying molecular mechanisms remain incompletely understood. We hypothesize that NO constrains the activity of the matrix crosslinking enzyme tissue transglutaminase (TG2) via S-nitrosylation in young vessels, a process that is reversed in aging. OBJECTIVE: We sought to determine whether endothelium-dependent NO regulates TG2 activity by S-nitrosylation and whether this contributes to age-related vascular stiffness. METHODS AND RESULTS: We first demonstrate that NO suppresses activity and increases S-nitrosylation of TG2 in cellular models. Next, we show that nitric oxide synthase (NOS) inhibition leads to increased surface and extracellular matrix-associated TG2. We then demonstrate that endothelium-derived bioactive NO primarily mediates its effects through TG2, using TG2(-/-) mice chronically treated with the NOS inhibitor l-N(G)-nitroarginine methyl ester (L-NAME). We confirm that TG2 activity is modulated by endothelium-derived bioactive NO in young rat aorta. In aging rat aorta, although TG2 expression remains unaltered, its activity increases and S-nitrosylation decreases. Furthermore, TG2 inhibition decreases vascular stiffness in aging rats. Finally, TG2 activity and matrix crosslinks are augmented with age in human aorta, whereas abundance remains unchanged. CONCLUSIONS: Decreased S-nitrosylation of TG2 and increased TG activity lead to enhanced matrix crosslinking and contribute to vascular stiffening in aging. TG2 appears to be the member of the transglutaminase family primarily contributing to this phenotype. Inhibition of TG2 could thus represent a therapeutic target for age-associated vascular stiffness and isolated systolic hypertension.


Aging/metabolism , Endothelium, Vascular/enzymology , GTP-Binding Proteins/metabolism , Nitric Oxide/physiology , Transglutaminases/metabolism , Adult , Age Factors , Aged , Aged, 80 and over , Aging/pathology , Animals , Cells, Cultured , Endothelium, Vascular/pathology , GTP-Binding Proteins/antagonists & inhibitors , Humans , Male , Mice , Mice, Transgenic , Middle Aged , NIH 3T3 Cells , Nitric Oxide/antagonists & inhibitors , Protein Glutamine gamma Glutamyltransferase 2 , Rats , Rats, Inbred F344 , Transglutaminases/antagonists & inhibitors
18.
J Biol Chem ; 284(24): 16693-16703, 2009 Jun 12.
Article En | MEDLINE | ID: mdl-19386600

A functional collaboration between growth factor receptors such as platelet derived growth factor receptor (PDGFR) and integrins is required for effective signal transduction in response to soluble growth factors. However, the mechanisms of synergistic PDGFR/integrin signaling remain poorly understood. Our previous work showed that cell surface tissue transglutaminase (tTG) induces clustering of integrins and amplifies integrin signaling by acting as an integrin binding adhesion co-receptor for fibronectin. Here we report that in fibroblasts tTG enhances PDGFR-integrin association by interacting with PDGFR and bridging the two receptors on the cell surface. The interaction between tTG and PDGFR reduces cellular levels of the receptor by accelerating its turnover. Moreover, the association of PDGFR with tTG causes receptor clustering, increases PDGF binding, promotes adhesion-mediated and growth factor-induced PDGFR activation, and up-regulates downstream signaling. Importantly, tTG is required for efficient PDGF-dependent proliferation and migration of fibroblasts. These results reveal a previously unrecognized role for cell surface tTG in the regulation of the joint PDGFR/integrin signaling and PDGFR-dependent cell responses.


Fibroblasts/cytology , Fibroblasts/enzymology , Integrins/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Transglutaminases/metabolism , Animals , Cell Adhesion/physiology , Cell Division/physiology , Cell Movement/physiology , Cells, Cultured , Dermis/cytology , Down-Regulation/physiology , Humans , Mice , NIH 3T3 Cells , Receptor Aggregation/physiology , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptors, Cell Surface/metabolism , Signal Transduction/physiology , Transglutaminases/genetics , Up-Regulation/physiology , Wound Healing/physiology
19.
J Cell Sci ; 120(Pt 18): 3188-99, 2007 Sep 15.
Article En | MEDLINE | ID: mdl-17711877

Tissue transglutaminase functions as a protein crosslinking enzyme and an integrin-binding adhesion co-receptor for fibronectin on the cell surface. These activities of transglutaminase and the involvement of this protein in cell-matrix adhesion, integrin-mediated signaling, cell migration and matrix organization suggest a precise and efficient control of its cell-surface expression. We report a novel mechanism of regulation of surface transglutaminase through internalization and subsequent lysosomal degradation. Constitutive endocytosis of cell-surface transglutaminase depends on plasma membrane cholesterol and the activity of dynamin-2, and involves both clathrin-coated pits and lipid rafts or caveolae. Furthermore, the key matrix ligands of transglutaminase, fibronectin and platelet-derived growth factor, promote its endocytosis from the cell surface. Our results also indicate that transglutaminase interacts in vitro and on the cell surface with the major endocytic receptor, low-density lipoprotein receptor-related protein 1, and demonstrate the requirement for this receptor in the endocytosis of transglutaminase. Finally, a deficiency of this endocytic receptor or blockade of endo-lysosomal function upregulate transglutaminase expression on the cell surface, leading to increased cell adhesion and matrix crosslinking. These findings characterize a previously unknown pathway of transglutaminase internalization and degradation that might be crucial for regulation of its adhesive and signaling functions on the cell surface and reveal a novel functional link between cell-matrix adhesion and endocytosis.


Endocytosis/physiology , Lysosomes/enzymology , Membrane Microdomains/enzymology , Receptors, LDL/metabolism , Signal Transduction/physiology , Transglutaminases/metabolism , Tumor Suppressor Proteins/metabolism , Animals , CHO Cells , Cell Adhesion/physiology , Cholesterol/metabolism , Cricetinae , Cricetulus , Dynamin II/metabolism , Fibronectins/metabolism , GTP-Binding Proteins , Humans , Integrins/metabolism , Low Density Lipoprotein Receptor-Related Protein-1 , Mice , NIH 3T3 Cells , Protein Glutamine gamma Glutamyltransferase 2
20.
Mol Biol Cell ; 17(4): 1606-19, 2006 Apr.
Article En | MEDLINE | ID: mdl-16452636

Tissue transglutaminase (tTG) is a multifunctional protein that serves as cross-linking enzyme and integrin-binding adhesion coreceptor for fibronectin on the cell surface. Previous work showed activation of small GTPase RhoA via enzymatic transamidation by cytoplasmic tTG. Here, we report an alternative nonenzymatic mechanism of RhoA activation by cell surface tTG. Direct engagement of surface tTG with specific antibody or the fibronectin fragment containing modules I(6)II(1,2)I(7-9) increases RhoA-GTP levels. Integrin-dependent signaling to RhoA and its downstream target Rho-associated coiled-coil containing serine/threonine protein kinase (ROCK) is amplified by surface tTG. tTG expression on the cell surface elevates RhoA-GTP levels in nonadherent and adherent cells, delays maximal RhoA activation upon cell adhesion to fibronectin and accelerates a rise in RhoA activity after binding soluble integrin ligands. These data indicate that surface tTG induces integrin clustering regardless of integrin-ligand interactions. This notion is supported by visualization of integrin clusters, increased susceptibility of integrins to chemical cross-linking, and biochemical detection of large integrin complexes in cells expressing tTG. In turn, integrin aggregation by surface tTG inhibits Src kinase activity and decreases activation of the Src substrate p190RhoGAP. Moreover, pharmacological inhibition of Src kinase reveals inactivation of Src signaling as the primary cause of elevated RhoA activity in cells expressing tTG. Together, these findings show that surface tTG amplifies integrin-mediated signaling to RhoA/ROCK via integrin clustering and down-regulation of the Src-p190RhoGAP regulatory pathway.


Carrier Proteins/metabolism , GTP-Binding Proteins/physiology , Integrin beta1/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , Transglutaminases/physiology , rhoA GTP-Binding Protein/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Cell Adhesion , Cell Membrane/enzymology , Cell Polarity , DNA-Binding Proteins , Down-Regulation , Enzyme Activation , Fibronectins/chemistry , GTP-Binding Proteins/genetics , GTPase-Activating Proteins , Integrin beta1/immunology , Ligands , Mice , NIH 3T3 Cells , Phosphorylation , Protein Glutamine gamma Glutamyltransferase 2 , Repressor Proteins , Signal Transduction , Transglutaminases/genetics , Up-Regulation
...