Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Physiol Rev ; 94(2): 383-417, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24692352

RESUMO

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.


Assuntos
Transdução de Sinais , Transglutaminases/metabolismo , Animais , Diferenciação Celular , Regulação Enzimológica da Expressão Gênica , Humanos , Neoplasias/enzimologia , Neoplasias/patologia , Transcrição Gênica , Transglutaminases/genética
2.
J Cell Sci ; 127(Pt 16): 3578-92, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-24963132

RESUMO

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.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Proteínas de Ligação a DNA/metabolismo , Filaminas/metabolismo , Miofibrilas/metabolismo , Proteínas Nucleares/metabolismo , Fosfoglucomutase/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Proteínas do Citoesqueleto/genética , Proteínas de Ligação a DNA/genética , Filaminas/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mioblastos/metabolismo , Miofibrilas/genética , Proteínas Nucleares/genética , Fosfoglucomutase/genética , Ligação Proteica
3.
Exp Dermatol ; 24(6): 455-61, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25808322

RESUMO

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.


Assuntos
Adesão Celular/fisiologia , Movimento Celular/fisiologia , Fibronectinas/fisiologia , Melanoma/patologia , Nucleosídeo NM23 Difosfato Quinases/fisiologia , Neoplasias Cutâneas/patologia , Linhagem Celular Tumoral , Progressão da Doença , Matriz Extracelular/patologia , Matriz Extracelular/fisiologia , Fibronectinas/genética , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Técnicas In Vitro , Melanoma/fisiopatologia , Nucleosídeo NM23 Difosfato Quinases/genética , Metástase Neoplásica/patologia , Metástase Neoplásica/fisiopatologia , RNA Mensageiro/genética , RNA Mensageiro/fisiologia , Transdução de Sinais/fisiologia , Neoplasias Cutâneas/fisiopatologia
4.
J Vasc Res ; 51(6): 418-28, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25612735

RESUMO

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.


Assuntos
Estenose das Carótidas/enzimologia , Estenose Coronária/enzimologia , Proteínas de Ligação ao GTP/metabolismo , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Neointima , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-sis/farmacologia , Receptores do Fator de Crescimento Derivado de Plaquetas/agonistas , Transdução de Sinais/efeitos dos fármacos , Transglutaminases/metabolismo , beta Catenina/metabolismo , Animais , Becaplermina , Estenose das Carótidas/genética , Estenose das Carótidas/patologia , Estenose das Carótidas/prevenção & controle , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Estenose Coronária/patologia , Estenose Coronária/prevenção & controle , Vasos Coronários/efeitos dos fármacos , Vasos Coronários/enzimologia , Vasos Coronários/patologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Proteínas de Ligação ao GTP/deficiência , Proteínas de Ligação ao GTP/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/enzimologia , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/enzimologia , Miócitos de Músculo Liso/patologia , Fenótipo , Proteína 2 Glutamina gama-Glutamiltransferase , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Fatores de Tempo , Transglutaminases/deficiência , Transglutaminases/genética
5.
Am J Physiol Heart Circ Physiol ; 305(6): H803-10, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-23873798

RESUMO

Nitric oxide (NO) can modulate arterial stiffness by regulating both functional and structural changes in the arterial wall. Tissue transglutaminase (TG2) has been shown to contribute to increased central aortic stiffness by catalyzing the cross-linking of matrix proteins. NO S-nitrosylates and constrains TG2 to the cytosolic compartment and thereby holds its cross-linking function latent. In the present study, the role of endothelial NO synthase (eNOS)-derived NO in regulating TG2 function was studied using eNOS knockout mice. Matrix-associated TG2 and TG2 cross-linking function were higher, whereas TG2 S-nitrosylation was lower in the eNOS(-/-) compared with wild-type (WT) mice. Pulse-wave velocity (PWV) and blood pressure measured noninvasively were elevated in the eNOS(-/-) compared with WT mice. Intact aortas and decellularized aortic tissue scaffolds of eNOS(-/-) mice were significantly stiffer, as determined by tensile testing. The carotid arteries of the eNOS(-/-) mice were also stiffer, as determined by pressure-dimension analysis. Invasive methods to determine the PWV-mean arterial pressure relationship showed that PWV in eNOS(-/-) and WT diverge at higher mean arterial pressure. Thus eNOS-derived NO regulates TG2 localization and function and contributes to vascular stiffness.


Assuntos
Aorta/fisiologia , Pressão Sanguínea/fisiologia , Endotélio Vascular/fisiologia , Proteínas de Ligação ao GTP/biossíntese , Óxido Nítrico Sintase Tipo III/metabolismo , Transglutaminases/biossíntese , Rigidez Vascular/fisiologia , Animais , Ativação Enzimática , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico Sintase Tipo III/genética , Proteína 2 Glutamina gama-Glutamiltransferase , Estresse Mecânico , Resistência à Tração/fisiologia
6.
J Vasc Res ; 50(5): 383-95, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23988702

RESUMO

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.


Assuntos
Artérias/enzimologia , Proteínas de Ligação ao GTP/metabolismo , Transglutaminases/metabolismo , Animais , Artérias/fisiologia , Cálcio/metabolismo , Adesão Celular/fisiologia , Dissulfetos/metabolismo , Ativação Enzimática , Fibronectinas/metabolismo , Proteínas de Ligação ao GTP/antagonistas & inibidores , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/metabolismo , Humanos , Integrina alfa5beta1/metabolismo , Integrinas/metabolismo , Camundongos , Músculo Liso/citologia , Músculo Liso/fisiologia , Conformação Proteica/efeitos dos fármacos , Proteína 2 Glutamina gama-Glutamiltransferase , Estresse Mecânico , Talina/metabolismo , Transglutaminases/antagonistas & inibidores , Vasoconstrição/fisiologia , Vasodilatação/fisiologia , Vinculina/metabolismo
7.
Amino Acids ; 44(1): 261-9, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21984378

RESUMO

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.


Assuntos
Células Endoteliais/enzimologia , Miócitos de Músculo Liso/enzimologia , Óxido Nítrico/fisiologia , Transglutaminases/metabolismo , Fatores Etários , Animais , Aorta/citologia , Linhagem Celular , Técnicas de Cocultura , Endotélio Vascular/citologia , Matriz Extracelular/enzimologia , Fibroblastos/enzimologia , Proteínas de Ligação ao GTP , Humanos , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase Tipo III/antagonistas & inibidores , Óxido Nítrico Sintase Tipo III/fisiologia , Nitrosação , Proteína 2 Glutamina gama-Glutamiltransferase , Transporte Proteico , Ratos , Ratos Endogâmicos F344
8.
J Cell Physiol ; 227(5): 2089-96, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-21769866

RESUMO

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.


Assuntos
Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/metabolismo , Proteínas Proto-Oncogênicas c-sis/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais/fisiologia , Transglutaminases/metabolismo , Animais , Becaplermina , Movimento Celular , Proliferação de Células , Sobrevivência Celular , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miócitos de Músculo Liso/citologia , Proteínas Proto-Oncogênicas c-sis/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética
9.
Circ Res ; 107(1): 117-25, 2010 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-20489165

RESUMO

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.


Assuntos
Envelhecimento/metabolismo , Endotélio Vascular/enzimologia , Proteínas de Ligação ao GTP/metabolismo , Óxido Nítrico/fisiologia , Transglutaminases/metabolismo , Adulto , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Envelhecimento/patologia , Animais , Células Cultivadas , Endotélio Vascular/patologia , Proteínas de Ligação ao GTP/antagonistas & inibidores , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Células NIH 3T3 , Óxido Nítrico/antagonistas & inibidores , Proteína 2 Glutamina gama-Glutamiltransferase , Ratos , Ratos Endogâmicos F344 , Transglutaminases/antagonistas & inibidores
10.
Mol Biol Cell ; 17(4): 1606-19, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16452636

RESUMO

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.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Ligação ao GTP/fisiologia , Integrina beta1/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Transglutaminases/fisiologia , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Adesão Celular , Membrana Celular/enzimologia , Polaridade Celular , Proteínas de Ligação a DNA , Regulação para Baixo , Ativação Enzimática , Fibronectinas/química , Proteínas de Ligação ao GTP/genética , Proteínas Ativadoras de GTPase , Integrina beta1/imunologia , Ligantes , Camundongos , Células NIH 3T3 , Fosforilação , Proteína 2 Glutamina gama-Glutamiltransferase , Proteínas Repressoras , Transdução de Sinais , Transglutaminases/genética , Regulação para Cima
11.
Front Biosci ; 11: 1057-76, 2006 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-16146797

RESUMO

Numerous studies over the last two decades revealed a complexity and multiple functions of tissue transglutaminase (tTG or TG2, EC 2.3.2.13). Besides the ability to catalyze Ca2+-dependent transamidation of proteins and formation of protein polymers via protease-resistant covalent isopeptide bonds, tTG also possesses GTPase enzymatic activity which links this protein to certain intracellular signaling pathways. Moreover, in addition to cytoplasmic and nuclear localization, a significant part of the protein pool is present on the cell surface. A number of recent findings indicate that surface tTG is involved in the interactions of cells with the surrounding extracellular matrix (ECM). In this review we will focus on the newly defined non-enzymatic adhesive function of tTG in cell-matrix interactions and discuss contributions of previously characterized enzymatic activities of tTG to cell-matrix adhesion and adhesion-dependent processes. Understanding molecular interactions and enzymatic activities of tTG will gain further insights into the role of this protein in normal human physiology and various pathological conditions.


Assuntos
Matriz Extracelular/metabolismo , Transglutaminases/fisiologia , Animais , Cálcio/metabolismo , Catálise , Adesão Celular , Comunicação Celular , Linhagem Celular , Membrana Celular/metabolismo , Movimento Celular , Reagentes de Ligações Cruzadas/farmacologia , Fibronectinas/química , GTP Fosfo-Hidrolases/metabolismo , Proteínas de Ligação ao GTP , Humanos , Integrinas/metabolismo , Ligantes , Lisina/química , Microscopia de Fluorescência , Modelos Biológicos , Ligação Proteica , Conformação Proteica , Proteína 2 Glutamina gama-Glutamiltransferase , Proteínas/química , Serina/química , Transdução de Sinais , Transglutaminases/metabolismo
12.
Thromb Haemost ; 116(6): 1122-1130, 2016 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-27580629

RESUMO

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.


Assuntos
Anticorpos Monoclonais/imunologia , Fibrina/metabolismo , Leucócitos/citologia , Receptores de LDL/imunologia , Migração Transendotelial e Transepitelial , Animais , Mapeamento de Epitopos , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos Endogâmicos C57BL , Receptores de LDL/metabolismo
13.
Naunyn Schmiedebergs Arch Pharmacol ; 388(2): 199-206, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25017017

RESUMO

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.


Assuntos
Melanoma , Nucleosídeo NM23 Difosfato Quinases/metabolismo , Neoplasias Cutâneas , Actinas/metabolismo , Animais , Movimento Celular , Reparo do DNA , Adesões Focais , Instabilidade Genômica , Humanos , Melanoma/genética , Melanoma/metabolismo , Melanoma/patologia , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia
14.
Biophys Chem ; 112(2-3): 257-66, 2004 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-15572257

RESUMO

Previous electron microscopy (EM) studies revealed that the proteolytically prepared, truncated, bovine fibrinogen alphaC-domain (Aalpha223-539 fragment) upon transfer from acidic to neutral pH formed ordered oligomers which could mimic alpha polymers of cross-linked fibrin. In this study, we demonstrated that although its recombinant analog, bAalpha224-538, as well as the full-length version of the alphaC-domain (bAalpha224-568), upon similar treatment also formed oligomers with ordered structure, both were monomeric when kept in neutral pH buffer. To search further for conditions for their oligomerization, we treated bAalpha224-568 with factor XIIIa, purified the cross-linked soluble fraction, and confirmed that it consisted of oligomers. Similar cross-linked oligomers were obtained with the recombinant human alphaC-domain (residues Aalpha221-610). In a cell adhesion assay, the adhesion of human umbilical vein endothelial cells (HUVEC) to the alphaC-domains substantially increased upon oligomerization. These results demonstrate that the recombinant alphaC-domains can form stable oligomers which may mimic properties of the alphaC-domains in cross-linked fibrin.


Assuntos
Fibrinogênio/química , Fragmentos de Peptídeos/química , Animais , Bovinos , Adesão Celular , Dimerização , Endotélio Vascular/citologia , Fator XIIIa/química , Fibrinogênio/metabolismo , Fibrinogênio/ultraestrutura , Humanos , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/ultraestrutura , Ligação Proteica , Estrutura Terciária de Proteína , Veias Umbilicais/citologia
15.
PLoS One ; 9(2): e89285, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24586660

RESUMO

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.


Assuntos
Fibronectinas/antagonistas & inibidores , Proteínas de Ligação ao GTP/antagonistas & inibidores , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/metabolismo , Mapas de Interação de Proteínas/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Transglutaminases/antagonistas & inibidores , Western Blotting , Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Feminino , Fibronectinas/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Ensaios de Triagem em Larga Escala , Humanos , Neoplasias Ovarianas/patologia , Proteína 2 Glutamina gama-Glutamiltransferase , Transglutaminases/metabolismo , Células Tumorais Cultivadas , Cicatrização/efeitos dos fármacos
16.
J Am Heart Assoc ; 3(2): e000599, 2014 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-24721796

RESUMO

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.


Assuntos
Envelhecimento/metabolismo , Aorta/enzimologia , Aorta/fisiopatologia , Proteínas de Ligação ao GTP/metabolismo , Esforço Físico , Transglutaminases/metabolismo , Rigidez Vascular , Fatores Etários , Animais , Pressão Arterial , Moléculas de Adesão Celular/metabolismo , Masculino , Proteínas dos Microfilamentos/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Fosfoproteínas/metabolismo , Fosforilação , Proteína 2 Glutamina gama-Glutamiltransferase , Análise de Onda de Pulso , Ratos , Ratos Endogâmicos F344 , Corrida , Transdução de Sinais , Resistência à Tração
17.
Sci Rep ; 3: 1460, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23492872

RESUMO

Controlling chirality in growth of single-walled carbon nanotubes (SWNTs) is important for exploiting their practical applications. For long it has been conceptually conceived that the structural control of SWNTs is potentially achievable by fabricating nanoparticle catalysts with proper structures on crystalline substrates via epitaxial growth techniques. Here, we have accomplished epitaxial formation of monometallic Co nanoparticles with well-defined crystal structure, and its use as a catalyst in the selective growth of SWNTs. Dynamics of Co nanoparticles formation and SWNT growth inside an atomic-resolution environmental transmission electron microscope at a low CO pressure was recorded. We achieved highly preferential growth of semiconducting SWNTs (~90%) with an exceptionally large population of (6, 5) tubes (53%) in an ambient CO atmosphere. Particularly, we also demonstrated high enrichment in (7, 6) and (9, 4) at a low growth temperature. These findings open new perspectives both for structural control of SWNTs and for elucidating the growth mechanisms.


Assuntos
Cobalto/química , Nanopartículas Metálicas/química , Nanotubos de Carbono/química , Catálise , Cristalografia por Raios X , Cinética , Nanopartículas Metálicas/ultraestrutura , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão
18.
Int Rev Cell Mol Biol ; 294: 1-97, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22364871

RESUMO

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.


Assuntos
Regulação Enzimológica da Expressão Gênica/fisiologia , Transglutaminases/genética , Transglutaminases/metabolismo , Animais , Apoptose/fisiologia , Adesão Celular/fisiologia , Compartimento Celular/fisiologia , Diferenciação Celular/fisiologia , Crescimento Celular , Movimento Celular/fisiologia , Sobrevivência Celular/fisiologia , Matriz Extracelular/enzimologia , Proteínas de Ligação ao GTP , Humanos , Proteína 2 Glutamina gama-Glutamiltransferase
19.
FEBS J ; 278(24): 4704-16, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21902810

RESUMO

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.


Assuntos
Matriz Extracelular/metabolismo , Proteínas de Ligação ao GTP/fisiologia , Transglutaminases/fisiologia , Adesão Celular/fisiologia , Reagentes de Ligações Cruzadas/metabolismo , Cisteína/metabolismo , Ativação Enzimática , Fibronectinas/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Humanos , Integrinas/metabolismo , Proteína-5 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Macrófagos/fisiologia , Metaloproteinases da Matriz Associadas à Membrana/metabolismo , Transplante de Células-Tronco Mesenquimais , Conformação Proteica/efeitos dos fármacos , Proteína 2 Glutamina gama-Glutamiltransferase , Transporte Proteico/fisiologia , Receptores de Fatores de Crescimento/fisiologia , Transdução de Sinais/fisiologia , Sindecana-4/fisiologia , Transglutaminases/metabolismo , beta Catenina/fisiologia
20.
Commun Integr Biol ; 4(5): 584-6, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22046470

RESUMO

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.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA