Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Reprod Fertil Dev ; 28(7): 974-981, 2016 Jun.
Article in English | MEDLINE | ID: mdl-25557047

ABSTRACT

Use of the dietary supplement quercetin is on the rise. Because previous studies imply an inhibitory effect of quercetin on male fertility, we explored the effects of this flavonoid on fertility in female mice. Birth outcomes, and ovarian morphology in 4-week-old offspring, were assessed in mice receiving dietary quercetin (5mgkg-1day-1) for 9 months during two breeding periods: from 2 to 6 months (prime reproductive age) and 8 to11 months of age. Quercetin increased birth spacing, leading to a 60% reduction in the number of litters, but enhanced folliculogenesis in ovaries of female offspring. While in young females quercetin caused an almost 70% increase in litter size, in older animals this effect was reversed. Consistent with the inhibitory activity of quercetin on the enzyme transglutaminase 2 (TG2), genetic ablation of TG2 in mice mirrors the effects of quercetin on birth outcomes and follicular development. Further, TG2-null mice lack responsiveness to quercetin ingestion. Our study shows for the first time that dietary quercetin can cause reduced reproductive potential in female mice and implies that TG2 may regulate ovarian ageing.


Subject(s)
Diet/veterinary , Fertility , GTP-Binding Proteins/physiology , Quercetin/administration & dosage , Transglutaminases/physiology , Animals , Dietary Supplements , Female , Mice , Pregnancy , Protein Glutamine gamma Glutamyltransferase 2
2.
Arterioscler Thromb Vasc Biol ; 35(3): 573-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25614285

ABSTRACT

OBJECTIVE: Phenotypic plasticity of vascular smooth muscle cells (VSMCs) contributes to cardiovascular disease. Chondrocyte-like transformation of VSMCs associates with vascular calcification and underlies the formation of aortic cartilaginous metaplasia induced in mice by genetic loss of matrix Gla protein (MGP). Previous microarray analysis identified a dramatic downregulation of Wnt16 in calcified MGP-null aortae, suggesting an antagonistic role for Wnt16 in the chondrogenic transformation of VSMCs. APPROACH AND RESULTS: Wnt16 is significantly downregulated in MGP-null aortae, before the histological appearance of cartilaginous metaplasia, and in primary MGP-null VSMCs. In contrast, intrinsic TGFß is activated in MGP-null VSMCs and is necessary for spontaneous chondrogenesis of these cells in high-density micromass cultures. TGFß3-induced chondrogenic transformation in wild-type VSMCs associates with Smad2/3-dependent Wnt16 downregulation, but Wnt16 does not suppress TGFß3-induced Smad activation. In addition, TGFß3 inhibits Notch signaling in wild-type VSMCs, and this pathway is downregulated in MGP-null aortae. Exogenous Wnt16 stimulates Notch activity and attenuates TGFß3-induced downregulation of Notch in wild-type VSMCs, prevents chondrogenesis in MGP-null and TGFß3-treated wild-type VSMCs, and stabilizes expression of contractile markers of differentiated VSMCs. CONCLUSIONS: We describe a novel TGFß-Wnt16-Notch signaling conduit in the chondrocyte-like transformation of VSMCs and identify endogenous TGFß activity in MGP-null VSMCs as a critical mediator of chondrogenesis. Our proposed model suggests that the activated TGFß pathway inhibits expression of Wnt16, which is a positive regulator of Notch signaling and a stabilizer of VSMC phenotype. These data advance the comprehensive mechanistic understanding of VSMC transformation and may identify a novel potential therapeutic target in vascular calcification.


Subject(s)
Cell Transdifferentiation , Chondrocytes/metabolism , Chondrogenesis , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Transforming Growth Factor beta/metabolism , Vascular Calcification/metabolism , Wnt Proteins/metabolism , Animals , Aorta/metabolism , COS Cells , Calcium-Binding Proteins/deficiency , Calcium-Binding Proteins/genetics , Chlorocebus aethiops , Chondrocytes/pathology , Extracellular Matrix Proteins/deficiency , Extracellular Matrix Proteins/genetics , Metaplasia , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Phenotype , RNA Interference , Rats , Receptors, Notch/metabolism , Signal Transduction , Transfection , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta3/metabolism , Vascular Calcification/genetics , Vascular Calcification/pathology , Wnt Proteins/genetics , Matrix Gla Protein
3.
BMC Genomics ; 15: 988, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25407537

ABSTRACT

BACKGROUND: Chromatin compactness has been considered a major determinant of gene activity and has been associated with specific chromatin modifications in studies on a few individual genetic loci. At the same time, genome-wide patterns of open and closed chromatin have been understudied, and are at present largely predicted from chromatin modification and gene expression data. However the universal applicability of such predictions is not self-evident, and requires experimental verification. RESULTS: We developed and implemented a high-throughput analysis for general chromatin sensitivity to DNase I which provides a comprehensive epigenomic assessment in a single assay. Contiguous domains of open and closed chromatin were identified by computational analysis of the data, and correlated to other genome annotations including predicted chromatin "states", individual chromatin modifications, nuclear lamina interactions, and gene expression. While showing that the widely trusted predictions of chromatin structure are correct in the majority of cases, we detected diverse "exceptions" from the conventional rules. We found a profound paucity of chromatin modifications in a major fraction of closed chromatin, and identified a number of loci where chromatin configuration is opposite to that expected from modification and gene expression patterns. Further, we observed that chromatin of large introns tends to be closed even when the genes are expressed, and that a significant proportion of active genes including their promoters are located in closed chromatin. CONCLUSIONS: These findings reveal limitations of the existing predictive models, indicate novel mechanisms of epigenetic regulation, and provide important insights into genome organization and function.


Subject(s)
Chromatin Assembly and Disassembly/genetics , Chromatin/genetics , Chromosome Mapping , Drosophila/genetics , Genome, Insect , Animals , Binding Sites , Chromatin/metabolism , Computational Biology/methods , Deoxyribonuclease I/metabolism , Protein Binding
4.
Biotechnol Lett ; 36(9): 1901-7, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24930093

ABSTRACT

Transglutaminase 2 (TG2) was used to attach biologically-active BMP2 to collagen type I-coated poly-L-lactic acid (PLLA) nanofibrous scaffolds. Irreversibly cross-linked BMP2 retained its activity and induced Smad-dependent gene expression in cells seeded on PLLA-BMP2 scaffolds. These modified scaffolds promote osteogenic differentiation of human bone marrow-derived mesenchymal stem cells (hBMSCs) cultured in low-serum and growth factor free medium and support deposition of the calcified matrix and induction of the molecular osteogenic markers Runx2, osteopontin, osteonectin and bone sialoprotein. Importantly, the PLLA-BMP2 scaffolds did not support chondrogenic differentiation in hBMSCs as there was no expression of chondrogenic markers aggrecan, Sox 9, and collagen type II, and no deposition of cartilaginous glycosaminoglycan-rich matrix. Thus, TG2-mediated cross-linking of BMP2 to a scaffold is a novel approach to induce osteoblast-specific programming of hBMSCs in a spatially controlled manner.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Cell Differentiation , Collagen/metabolism , Mesenchymal Stem Cells/physiology , Osteoblasts/physiology , Polyesters/metabolism , Tissue Scaffolds , Cells, Cultured , GTP-Binding Proteins/metabolism , Humans , Osteogenesis , Protein Glutamine gamma Glutamyltransferase 2 , Transglutaminases/metabolism
5.
Physiol Rev ; 94(2): 383-417, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24692352

ABSTRACT

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.


Subject(s)
Signal Transduction , Transglutaminases/metabolism , Animals , Cell Differentiation , Gene Expression Regulation, Enzymologic , Humans , Neoplasms/enzymology , Neoplasms/pathology , Transcription, Genetic , Transglutaminases/genetics
6.
Curr Pharm Des ; 20(37): 5811-20, 2014.
Article in English | MEDLINE | ID: mdl-24533936

ABSTRACT

Arterial calcification (AC) is a hallmark of many serious diseases, including atherosclerosis, chronic kidney disease, and diabetes. AC may also develop as a side-effect of therapy with anticoagulants, such as warfarin which is widely used for prophylaxis of thrombosis. In our studies, we established the relation between warfarin-induced AC and activation of enzyme transglutaminase 2 (TG2) and ß-catenin signaling. We showed that TG2-specific inhibitor KCC-009 significantly attenuated the damaging effects of warfarin on arterial tissue. A similar protective effect was also achieved with a dietary bioflavonoid quercetin that inhibits TG2 and ß-catenin signaling. We have shown that quercetin intercepts the chondrogenic transformation of vascular smooth muscle and also drastically attenuates calcifying cartilaginous metaplasia in another model of AC caused by genetic loss of matrix gla protein (MGP). These findings suggest that quercetin may be considered as a promising anti-AC therapeutic in the clinical settings of warfarin supplementation and MGP dysfunction. Further studies are required to test the efficacy of quercetin on other types of AC.


Subject(s)
Enzyme Inhibitors/pharmacology , Isoxazoles/pharmacology , Signal Transduction/drug effects , Transglutaminases/antagonists & inhibitors , Vascular Calcification/drug therapy , beta Catenin/antagonists & inhibitors , Animals , Humans , Transglutaminases/metabolism , Vascular Calcification/enzymology , Vascular Calcification/metabolism , beta Catenin/metabolism
7.
J Vasc Res ; 51(6): 418-28, 2014.
Article in English | MEDLINE | ID: mdl-25612735

ABSTRACT

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.


Subject(s)
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
8.
PLoS One ; 8(9): e76210, 2013.
Article in English | MEDLINE | ID: mdl-24098781

ABSTRACT

RATIONALE: Cartilaginous metaplasia of vascular smooth muscle (VSM) is characteristic for arterial calcification in diabetes and uremia and in the background of genetic alterations in matrix Gla protein (MGP). A better understanding of the molecular details of this process is critical for the development of novel therapeutic approaches to VSM transformation and arterial calcification. OBJECTIVE: This study aimed to identify the effects of bioflavonoid quercetin on chondrogenic transformation and calcification of VSM in the MGP-null mouse model and upon TGF-ß3 stimulation in vitro, and to characterize the associated alterations in cell signaling. METHODS AND RESULTS: Molecular analysis revealed activation of ß-catenin signaling in cartilaginous metaplasia in Mgp-/- aortae in vivo and during chondrogenic transformation of VSMCs in vitro. Quercetin intercepted chondrogenic transformation of VSM and blocked activation of ß-catenin both in vivo and in vitro. Although dietary quercetin drastically attenuated calcifying cartilaginous metaplasia in Mgp-/- animals, approximately one-half of total vascular calcium mineral remained as depositions along elastic lamellae. CONCLUSION: Quercetin is potent in preventing VSM chondrogenic transformation caused by diverse stimuli. Combined with the demonstrated efficiency of dietary quercetin in preventing ectopic chondrogenesis in the MGP-null vasculature, these findings indicate a potentially broad therapeutic applicability of this safe for human consumption bioflavonoid in the therapy of cardiovascular conditions linked to cartilaginous metaplasia of VSM. Elastocalcinosis is a major component of MGP-null vascular disease and is controlled by a mechanism different from chondrogenic transformation of VSM and not sensitive to quercetin.


Subject(s)
Calcium-Binding Proteins/deficiency , Chondrogenesis/drug effects , Extracellular Matrix Proteins/deficiency , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Quercetin/pharmacology , Vascular Calcification/drug therapy , Animals , DNA Primers/genetics , Immunohistochemistry , Luciferases , Metaplasia/drug therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Real-Time Polymerase Chain Reaction , Signal Transduction/physiology , Statistics, Nonparametric , Vascular Calcification/physiopathology , beta Catenin/metabolism , Matrix Gla Protein
9.
J Biol Chem ; 288(43): 31400-8, 2013 Oct 25.
Article in English | MEDLINE | ID: mdl-24036114

ABSTRACT

Mutations in matrix Gla protein (MGP) have been correlated with vascular calcification. In the mouse model, MGP null vascular disease presents as calcifying cartilaginous lesions and mineral deposition along elastin lamellae (elastocalcinosis). Here we examined the mechanisms underlying both of these manifestations. Genetic ablation of enzyme transglutaminase 2 (TG2) in Mgp(-/-) mice dramatically reduced the size of cartilaginous lesions in the aortic media, attenuated calcium accrual more than 2-fold, and doubled longevity as compared with control Mgp(-/-) animals. Nonetheless, the Mgp(-/-);Tgm2(-/-) mice still died prematurely as compared with wild-type and retained the elastocalcinosis phenotype. This pathology in Mgp(-/-) animals was developmentally preceded by extensive fragmentation of elastic lamellae and associated with elevated serine elastase activity in aortic tissue and vascular smooth muscle cells. Systematic gene expression analysis followed by an immunoprecipitation study identified adipsin as the major elastase that is induced in the Mgp(-/-) vascular smooth muscle even in the TG2 null background. These results reveal a central role for TG2 in chondrogenic transformation of vascular smooth muscle and implicate adipsin in elastin fragmentation and ensuing elastocalcinosis. The importance of elastin calcification in MGP null vascular disease is highlighted by significant residual vascular calcification and mortality in Mgp(-/-);Tgm2(-/-) mice with reduced cartilaginous lesions. Our studies identify two potential therapeutic targets in vascular calcification associated with MGP dysfunction and emphasize the need for a comprehensive approach to this multifaceted disorder.


Subject(s)
Aortic Diseases/metabolism , Calcium-Binding Proteins/metabolism , Elastin/metabolism , Extracellular Matrix Proteins/metabolism , GTP-Binding Proteins/metabolism , Transglutaminases/metabolism , Vascular Calcification/metabolism , Animals , Aortic Diseases/genetics , Aortic Diseases/pathology , Calcium-Binding Proteins/genetics , Complement Factor D/genetics , Complement Factor D/metabolism , Elastin/genetics , Extracellular Matrix Proteins/genetics , GTP-Binding Proteins/genetics , Mice , Mice, Knockout , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Protein Glutamine gamma Glutamyltransferase 2 , Transglutaminases/genetics , Vascular Calcification/genetics , Vascular Calcification/pathology , Matrix Gla Protein
10.
J Biomater Sci Polym Ed ; 24(17): 2006-17, 2013.
Article in English | MEDLINE | ID: mdl-23905695

ABSTRACT

Architecture of the poly(l-lactic acid) (PLLA) scaffolds is known to affect protein affinity and binding strength. Here, we demonstrate that nanofibrous electrospun PLLA scaffolds reversibly absorb the pro-migratory serum factors that stimulate migration of vascular smooth muscle via an NFkB-dependent mechanism. Further, we demonstrate that mesenchymal stem cells seeded on the PLLA scaffolds do not enhance muscle migration but may maintain the ability of induced cells to migrate in an NFkB-independent manner. These findings further support the promising application of PLLA scaffolds for therapeutic angiogenesis and vascular graft engineering.


Subject(s)
Blood Proteins/chemistry , Cell Movement , Lactic Acid/chemistry , Nanofibers/chemistry , Nanotechnology/methods , Polymers/chemistry , Absorption , Cell Movement/drug effects , Humans , Lactic Acid/pharmacology , Muscle, Smooth, Vascular/cytology , Polyesters , Polymers/pharmacology , Signal Transduction/drug effects , Tissue Scaffolds/chemistry
11.
Biotechnol Lett ; 35(12): 2193-9, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23892982

ABSTRACT

Transglutaminase-mediated cross-linking has been employed to optimize the mechanical properties and stability of tissue scaffolds. We have characterized tissue transglutaminase (TG2)-mediated cross-linking as a useful tool to deliver biologically-active TGF to mesenchymal stem cells (MSCs) and direct their differentiation towards a chondrogenic lineage. TGF-ß3 is irreversibly cross-linked by TG2 to collagen type II-coated poly(L-lactic acid) nanofibrous scaffolds and activates Smad phosphorylation and Smad-dependent expression of a luciferase reporter. Human bone marrow-derived MSCs cultured on these scaffolds deposit cartilaginous matrix after 14 days of culture at 50 % efficiency compared to chondrogenesis in the presence of soluble TGF-ß3. These findings are significant because they suggest a novel approach for the programming of MSCs in a spatially controlled manner by immobilizing biologically active TGF-ß3 via cross-linking to a collagen-coated polymeric scaffold.


Subject(s)
Chondrogenesis/drug effects , Collagen/chemistry , GTP-Binding Proteins/metabolism , Lactic Acid/chemistry , Mesenchymal Stem Cells/cytology , Polymers/chemistry , Tissue Scaffolds/chemistry , Transforming Growth Factor beta , Transglutaminases/metabolism , Cell Adhesion/drug effects , Cell Differentiation , Humans , Immobilized Proteins/chemistry , Immobilized Proteins/metabolism , Immobilized Proteins/pharmacology , Polyesters , Protein Glutamine gamma Glutamyltransferase 2 , Tissue Engineering/methods , Transforming Growth Factor beta/chemistry , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology
12.
Amino Acids ; 44(1): 179-87, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22194042

ABSTRACT

Of the eight catalytic transglutaminases (TGs), transglutaminase 2 (TG2) has been the most comprehensively studied due to its ubiquitous expression in multiple cell types. Despite the observed critical role for this enzyme in multiple biological processes in vitro, TG2 knockout mouse models have shown no severe developmental phenotypes, suggesting compensation by other TGs. To begin characterization of the compensating mechanisms, we analyzed total transamidating activity and expression patterns of all catalytically active TGs in seven different tissues/organs from wild-type and TG2 knockout mice. Inhibitory analysis with TG2-specific inhibitor KCC-009 suggests that relative contribution of TG2 in total transamidating activity differs in various tissues. Accordingly, our data indicate tissue-specific mechanisms of compensation for the loss of TG2, including transcriptional compensation in heart and liver versus functional compensation in aorta, kidney and skeletal/cartiagenous tissues. On the contrary, no compensation has been detected in skeletal muscle, suggesting a limited role for the TG2-mediated transamidation in normal development of this tissue.


Subject(s)
GTP-Binding Proteins/genetics , Muscle, Skeletal/enzymology , Transglutaminases/genetics , Animals , Aorta/enzymology , Cartilage/enzymology , Factor XIIIa/genetics , Factor XIIIa/metabolism , GTP-Binding Proteins/deficiency , Gene Expression , Kidney/enzymology , Liver/enzymology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/enzymology , Organ Specificity , Phenotype , Protein Glutamine gamma Glutamyltransferase 2 , Transglutaminases/deficiency , Transglutaminases/metabolism
13.
Arterioscler Thromb Vasc Biol ; 33(1): 43-51, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23117658

ABSTRACT

OBJECTIVE: In vitro, transglutaminase-2 (TG2)-mediated activation of the ß-catenin signaling pathway is central in warfarin-induced calcification, warranting inquiry into the importance of this signaling axis as a target for preventive therapy of vascular calcification in vivo. METHODS AND RESULTS: The adverse effects of warfarin-induced elastocalcinosis in a rat model include calcification of the aortic media, loss of the cellular component in the vessel wall, and isolated systolic hypertension, associated with accumulation and activation of TG2 and activation of ß-catenin signaling. These effects of warfarin can be completely reversed by intraperitoneal administration of the TG2-specific inhibitor KCC-009 or dietary supplementation with the bioflavonoid quercetin, known to inhibit ß-catenin signaling. Our study also uncovers a previously uncharacterized ability of quercetin to inhibit TG2. Quercetin reversed the warfarin-induced increase in systolic pressure, underlying the functional consequence of this treatment. Molecular analysis shows that quercetin diet stabilizes the phenotype of smooth muscle and prevents its transformation into osteoblastic cells. CONCLUSIONS: Inhibition of the TG2/ß-catenin signaling axis seems to prevent warfarin-induced elastocalcinosis and to control isolated systolic hypertension.


Subject(s)
Aortic Diseases/prevention & control , Enzyme Inhibitors/pharmacology , GTP-Binding Proteins/antagonists & inhibitors , Isoxazoles/pharmacology , Muscle, Smooth, Vascular/drug effects , Quercetin/pharmacology , Transglutaminases/antagonists & inhibitors , Vascular Calcification/prevention & control , Animals , Aorta/drug effects , Aorta/enzymology , Aorta/pathology , Aortic Diseases/chemically induced , Aortic Diseases/enzymology , Aortic Diseases/genetics , Aortic Diseases/pathology , Aortic Diseases/physiopathology , Blood Pressure/drug effects , Cell Line , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Activation , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Gene Expression Regulation , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Osteogenesis/drug effects , Phosphorylation , Protein Glutamine gamma Glutamyltransferase 2 , Rats , Rats, Wistar , Signal Transduction/drug effects , Transglutaminases/genetics , Transglutaminases/metabolism , Vascular Calcification/chemically induced , Vascular Calcification/enzymology , Vascular Calcification/genetics , Vascular Calcification/pathology , Vascular Calcification/physiopathology , Warfarin , beta Catenin/metabolism
14.
J Biol Chem ; 288(4): 2632-40, 2013 Jan 25.
Article in English | MEDLINE | ID: mdl-23223575

ABSTRACT

Warfarin can stimulate vascular calcification in vitro via activation of ß-catenin signaling and/or inhibition of matrix Gla protein (MGP) carboxylation. Calcification was induced in vascular smooth muscle cells (VSMCs) with therapeutic levels of warfarin in normal calcium and clinically acceptable phosphate levels. Although TGF/BMP and PKA pathways are activated in calcifying VSMCs, pharmacologic analysis reveals that their activation is not contributory. However, ß-catenin activity is important because inhibition of ß-catenin with shRNA or bioflavonoid quercetin prevents calcification in primary human VSMCs, rodent aortic rings, and rat A10 VSMC line. In the presence of quercetin, reactivation of ß-catenin using the glycogen synthase kinase-3ß (GSK-3ß) inhibitor LiCl restores calcium accumulation, confirming that quercetin mechanism of action hinges on inhibition of the ß-catenin pathway. Calcification in VSMCs induced by 10 µm warfarin does not associate with reduced levels of carboxylated MGP, and inhibitory effects of quercetin do not involve induction of MGP carboxylation. Further, down-regulation of MGP by shRNA does not alter the effect of quercetin. These results suggest a new ß-catenin-targeting strategy to prevent vascular calcification induced by warfarin and identify quercetin as a potential therapeutic in this pathology.


Subject(s)
Calcium-Binding Proteins/metabolism , Extracellular Matrix Proteins/metabolism , Quercetin/pharmacology , Vascular Calcification/chemically induced , Warfarin/pharmacology , Animals , Antioxidants/pharmacology , Aorta/pathology , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Profiling , Genes, Reporter , In Vitro Techniques , Luciferases/metabolism , Muscle, Smooth, Vascular/cytology , RNA, Small Interfering/metabolism , Rats , Signal Transduction , beta Catenin/metabolism , Matrix Gla Protein
15.
J Heart Lung Transplant ; 31(9): 1009-17, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22694852

ABSTRACT

BACKGROUND: Cardiac allograft vasculopathy (CAV) remains the main cause of long-term transplant rejection. CAV is characterized by hyperproliferation of vascular smooth muscle cells (VSMCs). Canonical ß-catenin signaling is a critical regulator of VSMC proliferation in development; however, the role of this pathway and its regulation in CAV progression are obscure. We investigated the activity of ß-catenin signaling and the role for a putative activating ligand, transglutaminase 2 (TG2), in chronic cardiac rejection. METHODS: Hearts from Bm12 mice were transplanted into C57BL/6 mice (class II mismatch), and allografts were harvested 8 weeks after transplantation. Accumulation and sub-cellular distribution of ß-catenin protein and expression of several components of ß-catenin signaling were analyzed as hallmarks of pathway activation. In vitro, platelet-derived growth factor treatment was used to mimic the inflammatory milieu in VSMC and organotypic heart slice cultures. RESULTS: Activation of ß-catenin in allografts compared with isografts or naïve hearts was evidenced by the augmented expression of ß-catenin target genes, as well as the accumulation and nuclear localization of the ß-catenin protein in VSMCs of the occluded allograft vessels. Expression of TG2, an activator of ß-catenin signaling in VSMCs, was dramatically increased in allografts. Further, our ex vivo data demonstrate that TG2 is required for VSMC proliferation and for ß-catenin activation by platelet-derived growth factor in cardiac tissue. CONCLUSIONS: ß-Catenin signaling is activated in occluded vessels in murine cardiac allografts. TG2 is implicated as an endogenous activator of this signaling pathway and may therefore have a role in the pathogenesis of CAV during chronic allograft rejection.


Subject(s)
GTP-Binding Proteins/physiology , Graft Rejection/physiopathology , Heart Transplantation , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/physiology , Neointima/physiopathology , Transglutaminases/physiology , beta Catenin/physiology , Animals , Chronic Disease , Mice , Mice, Inbred C57BL , Protein Glutamine gamma Glutamyltransferase 2 , Signal Transduction
16.
Int Rev Cell Mol Biol ; 294: 1-97, 2012.
Article in English | MEDLINE | ID: mdl-22364871

ABSTRACT

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.


Subject(s)
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
17.
J Orthop Res ; 30(8): 1221-8, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22271086

ABSTRACT

This study investigated whether the synthetic peptide B2A (B2A2-K-NS) could induce in vitro chondrogenic differentiation and enhance the in vivo repair of damaged cartilage in an osteoarthritis model. In vitro, micromass cultures of murine and human stem cells with and without B2A were used as models of chondrogenic differentiation. Micromasses were evaluated for gene expression using microarray analysis and quantitative PCR; and for extracellular matrix production by Alcian blue staining for sulfated glycosaminoglycan and immunochemical detection of collagen type II. In vivo, osteoarthritis was chemically induced in knees of adult rats by an injection of mono-iodoacetate (MIA) into the synovial space. Treatment was administered at 7- and 14 days after the MIA by injection into the synovial space of B2A or saline and terminated at 21 days, after which knee cartilage damage was determined and scored by histological analysis. In murine C3H10T1/2 micromass culture, B2A induced the expression of more than 11 genes associated with growth factors/receptors, transcription, and the extracellular matrix, including PDGF-AA. B2A also significantly increased the sulfated glycosaminoglycan and collagen of murine- and human micromass cultures. In the knee osteoarthritis model, B2A treatment enhanced cartilage repair compared to untreated knees as determined histologically by a decrease in damage indicators. These findings suggest that B2A induces stem cells chondrogenic differentiation in vitro and enhances cartilage repair in vivo. The results suggest that B2A might be useful to promote cartilage repair.


Subject(s)
Cell Differentiation/drug effects , Chondrocytes/drug effects , Chondrogenesis/drug effects , Osteoarthritis, Knee/therapy , Peptides/pharmacology , Animals , Cartilage/drug effects , Cartilage/physiology , Cell Line , Chondrogenesis/genetics , Collagen Type II/biosynthesis , Extracellular Matrix/metabolism , Glycosaminoglycans/biosynthesis , Humans , Intercellular Signaling Peptides and Proteins , Iodoacetic Acid , Male , Mice , Osteoarthritis, Knee/chemically induced , Peptides/therapeutic use , Pilot Projects , Rats
18.
Amino Acids ; 42(2-3): 1045-53, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21830118

ABSTRACT

Tissue transglutaminase (tTG) is a multifunctional enzyme with a plethora of potential applications in regenerative medicine and tissue bioengineering. In this study, we examined the role of tTG as a regulator of chondrogenesis in human mesenchymal stem cells (MSC) using nanofibrous scaffolds coated with collagen type XI. Transient treatment of collagen type XI films and 3D scaffolds with tTG results in enhanced attachment of MSC and supports rounded cell morphology compared to the untreated matrices or those incubated in the continuous presence of tTG. Accordingly, enhanced cell aggregation and augmented chondrogenic differentiation have been observed on the collagen type XI-coated poly-(L-lactide) nanofibrous scaffolds treated with tTG prior to cell seeding. These changes implicate that MSC chondrogenesis is enhanced by the tTG-mediated modifications of the collagen matrix. For example, exogenous tTG increases resistance to collagenolysis in collagen type XI matrices by catalyzing intermolecular cross-linking, detected by a shift in the denaturation temperature. In addition, tTG auto-crosslinks to collagen type XI as detected by western blot and immunofluorescent analysis. This study identifies tTG as a novel regulator of MSC chondrogenesis further contributing to the expanding use of these cells in cartilage bioengineering.


Subject(s)
Chondrogenesis/physiology , Collagen Type XI/physiology , Mesenchymal Stem Cells/chemistry , Transglutaminases/physiology , Calorimetry, Differential Scanning , Cell Differentiation , Electrophoresis, Polyacrylamide Gel , Humans , Immunohistochemistry
19.
Amino Acids ; 42(2-3): 1065-75, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21809079

ABSTRACT

We have characterized the protein cross-linking enzyme transglutaminase (TGs) genes in zebrafish, Danio rerio, based on the analysis of their genomic organization and phylogenetics. Thirteen zebrafish TG genes (zTGs) have been identified, of which 11 show high homology to only 3 mammalian enzymes: TG1, TG2 and FXIIIa. No zebrafish homologues were identified for mammalian TGs 3-7. Real-time PCR analysis demonstrated distinct temporal expression profiles for zTGs in larvae and adult fish. Analysis by in situ hybridization revealed restricted expression of zTG2b and zFXIIIa in skeletal elements, resembling expression of their mammalian homologues in osteo-chondrogenic cells. Mammalian TG2 and FXIIIa have been implicated in promoting osteoblast differentiation and bone mineralization in vitro, however, mouse models lacking either gene have no skeletal phenotype likely due to a compensation effect. We show in this study that mineralization of the newly formed vertebrae is significantly reduced in fish grown for 5 days in the presence of TG inhibitor KCC-009 added at 3-5 days post fertilization. This treatment reduces average vertebrae mineralization by 30%, with complete inhibition in some fish, and no effect on the overall growth and vertebrae number. This is the first in vivo demonstration of the crucial requirement for the TG-catalyzed cross-linking activity in bone mineralization.


Subject(s)
Calcification, Physiologic , Multigene Family , Transglutaminases/genetics , Zebrafish/genetics , Animals , Base Sequence , DNA Primers , In Situ Hybridization , Real-Time Polymerase Chain Reaction , Transglutaminases/antagonists & inhibitors
20.
Arterioscler Thromb Vasc Biol ; 32(1): 123-30, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22034513

ABSTRACT

OBJECTIVE: Accumulating experimental evidence implicates ß-catenin signaling and enzyme transglutaminase 2 (TG2) in the progression of vascular calcification, and our previous studies have shown that TG2 can activate ß-catenin signaling in vascular smooth muscle cells (VSMCs). Here we investigated the role of the TG2/ß-catenin signaling axis in vascular calcification induced by warfarin. METHODS AND RESULTS: Warfarin-induced calcification in rat A10 VSMCs is associated with the activation of ß-catenin signaling and is independent of oxidative stress. The canonical ß-catenin inhibitor Dkk1, but not the Wnt antagonist Wif-1, prevents warfarin-induced activation of ß-catenin, calcification, and osteogenic transdifferentiation in VSMCs. TG2 expression and activity are increased in warfarin-treated cells, in contrast to canonical Wnt ligands. Vascular cells with genetically or pharmacologically reduced TG2 activity fail to activate ß-catenin in response to warfarin. Moreover, warfarin-induced calcification is significantly reduced on the background of attenuated TG2 both in vitro and in vivo. CONCLUSIONS: TG2 is a critical mediator of warfarin-induced vascular calcification that acts through the activation of ß-catenin signaling in VSMCs. Inhibition of canonical ß-catenin pathway or TG2 activity prevents warfarin-regulated calcification, identifying the TG2/ß-catenin axis as a novel therapeutic target in vascular calcification.


Subject(s)
GTP-Binding Proteins/metabolism , Transglutaminases/metabolism , Vascular Calcification/chemically induced , Vascular Calcification/metabolism , Warfarin/toxicity , beta Catenin/metabolism , Animals , Anticoagulants/toxicity , Cell Line , GTP-Binding Proteins/deficiency , GTP-Binding Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Cardiovascular , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Protein Glutamine gamma Glutamyltransferase 2 , Rats , Signal Transduction/drug effects , Transglutaminases/deficiency , Transglutaminases/genetics , Up-Regulation/drug effects , Vascular Calcification/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...