Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cancer Sci ; 113(6): 2179-2193, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35332604

RESUMO

Hepatocyte growth factor (HGF) activator inhibitor type-1 (HAI-1), encoded by the SPINT1 gene, is a transmembrane protease inhibitor that regulates membrane-anchored serine proteases, particularly matriptase. Here, we explored the role of HAI-1 in tongue squamous cell carcinoma (TSCC) cells. An immunohistochemical study of HAI-1 in surgically resected TSCC revealed the cell surface immunoreactivity of HAI-1 in the main portion of the tumor. The immunoreactivity decreased in the infiltrative front, and this decrease correlated with enhanced lymphatic invasion as judged by podoplanin immunostaining. In vitro homozygous deletion of SPINT1 (HAI-1KO) in TSCC cell lines (HSC3 and SAS) suppressed the cell growth rate but significantly enhanced invasion in vitro. The loss of HAI-1 resulted in enhanced pericellular activities of proteases, such as matriptase and urokinase-type plasminogen activator, which induced activation of HGF/MET signaling in the co-culture with pro-HGF-expressing fibroblasts and plasminogen-dependent plasmin generation, respectively. The enhanced plasminogen-dependent plasmin generation was abrogated partly by matriptase silencing. Culture supernatants of HAI-1KO cells had enhanced potency for converting the proform of vascular endothelial growth factor-C (VEGF-C), a lymphangiogenesis factor, into the mature form in a plasminogen-dependent manner. Furthermore, HGF significantly stimulated VEGF-C expression in TSCC cells. Orthotopic xenotransplantation into nude mouse tongue revealed enhanced lymphatic invasion of HAI-1KO TSCC cells compared to control cells. Our results suggest that HAI-1 insufficiency leads to dysregulated pericellular protease activity, which eventually orchestrates robust activation of protease-dependent growth factors, such as HGF and VEGF-C, in a tumor microenvironment to contribute to TSCC progression.


Assuntos
Carcinoma de Células Escamosas , Proteínas Secretadas Inibidoras de Proteinases , Neoplasias da Língua , Animais , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Fibrinolisina/genética , Homozigoto , Humanos , Camundongos , Plasminogênio/genética , Proteínas Secretadas Inibidoras de Proteinases/genética , Proteínas Secretadas Inibidoras de Proteinases/metabolismo , Deleção de Sequência , Serina Endopeptidases , Neoplasias da Língua/genética , Neoplasias da Língua/patologia , Microambiente Tumoral , Fator C de Crescimento do Endotélio Vascular/genética
2.
Nat Commun ; 12(1): 7112, 2021 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-34876572

RESUMO

Cardiac ATTR amyloidosis, a serious but much under-diagnosed form of cardiomyopathy, is caused by deposition of amyloid fibrils derived from the plasma protein transthyretin (TTR), but its pathogenesis is poorly understood and informative in vivo models have proved elusive. Here we report the generation of a mouse model of cardiac ATTR amyloidosis with transgenic expression of human TTRS52P. The model is characterised by substantial ATTR amyloid deposits in the heart and tongue. The amyloid fibrils contain both full-length human TTR protomers and the residue 49-127 cleavage fragment which are present in ATTR amyloidosis patients. Urokinase-type plasminogen activator (uPA) and plasmin are abundant within the cardiac and lingual amyloid deposits, which contain marked serine protease activity; knockout of α2-antiplasmin, the physiological inhibitor of plasmin, enhances amyloid formation. Together, these findings indicate that cardiac ATTR amyloid deposition involves local uPA-mediated generation of plasmin and cleavage of TTR, consistent with the previously described mechano-enzymatic hypothesis for cardiac ATTR amyloid formation. This experimental model of ATTR cardiomyopathy has potential to allow further investigations of the factors that influence human ATTR amyloid deposition and the development of new treatments.


Assuntos
Neuropatias Amiloides Familiares/metabolismo , Amiloide/metabolismo , Fibrinolisina/genética , Fibrinolisina/metabolismo , Placa Amiloide/metabolismo , Animais , Cardiomiopatias , Humanos , Camundongos Transgênicos , Pré-Albumina/metabolismo , Dobramento de Proteína , Proteólise
3.
Iran J Med Sci ; 46(6): 454-467, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34840386

RESUMO

Background: Ocriplasmin has been developed for the induction of posterior vitreous detachment in patients with vitreomacular adhesion. At physiological pH, ocriplasmin is susceptible to autolytic and proteolytic degradation, limiting its activity duration. These undesirable properties of ocriplasmin can be reduced by site-directed mutagenesis, so that its enzymatic activities can be augmented. This study aimed to design ocriplasmin variants with improved biological/physicochemical characteristics via bioinformatics tools. Methods: This study was performed in Tabriz University of Medical Sciences, Tabriz, Iran, 2019. Through site-directed mutagenesis, three ocriplasmin variants were designed. Structural analysis was performed on the wild-type variant and the mutant variants using the Protein Interactions Calculator (PIC) server. The interactions between the S-2403 substrate and the ocriplasmin variants were studied by molecular docking simulations, and binding capability was evaluated by the calculation of free binding energy. The conformational features of protein-substrate complex systems for all the variants were evaluated using molecular dynamic simulations at 100 nanoseconds. Results: The structural analysis of ocriplasmin revealed that the substitution of threonine for alanine 59 significantly reduced proteolytic activity, while the substitution of glutamic acid for lysine 156 influenced autolytic function. The molecular docking simulation results indicated the appropriate binding of the substrate to the ocriplasmin variants with high-to-low affinities. The binding affinity of the wild-type variant for the substrate was higher than that between the mutant variants and the substrate. Simulation analyses, consisting of the root-mean-square deviation, the root-mean-square fluctuation, and the center-of-mass average distance showed a higher affinity of the substrate for the wild type than for the mutant variants. Conclusion: The mutational analysis of ocriplasmin revealed that A59T and K156E mutagenesis could be used for the development of a new variant with higher therapeutic efficacy.


Assuntos
Biologia Computacional , Oftalmopatias/tratamento farmacológico , Fibrinolisina/administração & dosagem , Fibrinolisina/efeitos adversos , Fibrinolisina/genética , Fragmentos de Peptídeos/genética , Descolamento do Vítreo/induzido quimicamente , Análise Mutacional de DNA , Humanos , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Mutagênese , Proteólise , Aderências Teciduais/tratamento farmacológico , Corpo Vítreo
4.
Blood ; 138(3): 259-272, 2021 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-33827130

RESUMO

Acetaminophen (APAP)-induced liver injury is associated with activation of coagulation and fibrinolysis. In mice, both tissue factor-dependent thrombin generation and plasmin activity have been shown to promote liver injury after APAP overdose. However, the contribution of the contact and intrinsic coagulation pathways has not been investigated in this model. Mice deficient in individual factors of the contact (factor XII [FXII] and prekallikrein) or intrinsic coagulation (FXI) pathway were administered a hepatotoxic dose of 400 mg/kg of APAP. Neither FXII, FXI, nor prekallikrein deficiency mitigated coagulation activation or hepatocellular injury. Interestingly, despite the lack of significant changes to APAP-induced coagulation activation, markers of liver injury and inflammation were significantly reduced in APAP-challenged high-molecular-weight kininogen-deficient (HK-/-) mice. Protective effects of HK deficiency were not reproduced by inhibition of bradykinin-mediated signaling, whereas reconstitution of circulating levels of HK in HK-/- mice restored hepatotoxicity. Fibrinolysis activation was observed in mice after APAP administration. Western blotting, enzyme-linked immunosorbent assay, and mass spectrometry analysis showed that plasmin efficiently cleaves HK into multiple fragments in buffer or plasma. Importantly, plasminogen deficiency attenuated APAP-induced liver injury and prevented HK cleavage in the injured liver. Finally, enhanced plasmin generation and HK cleavage, in the absence of contact pathway activation, were observed in plasma of patients with acute liver failure due to APAP overdose. In summary, extrinsic but not intrinsic pathway activation drives the thromboinflammatory pathology associated with APAP-induced liver injury in mice. Furthermore, plasmin-mediated cleavage of HK contributes to hepatotoxicity in APAP-challenged mice independently of thrombin generation or bradykinin signaling.


Assuntos
Acetaminofen/efeitos adversos , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Fibrinolisina/metabolismo , Fibrinólise/efeitos dos fármacos , Cininogênios/metabolismo , Proteólise/efeitos dos fármacos , Acetaminofen/farmacologia , Animais , Doença Hepática Induzida por Substâncias e Drogas/genética , Doença Hepática Induzida por Substâncias e Drogas/patologia , Fator XII/genética , Fator XII/metabolismo , Feminino , Fibrinolisina/genética , Humanos , Cininogênios/genética , Masculino , Camundongos , Camundongos Knockout , Pré-Calicreína/genética , Pré-Calicreína/metabolismo
5.
Blood ; 129(21): 2896-2907, 2017 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-28320709

RESUMO

Inflammation resolution is an active process that functions to restore tissue homeostasis. The participation of the plasminogen (Plg)/plasmin (Pla) system in the productive phase of inflammation is well known, but its involvement in the resolution phase remains unclear. Therefore, we aimed to investigate the potential role of Plg/Pla in key events during the resolution of acute inflammation and its underlying mechanisms. Plg/Pla injection into the pleural cavity of BALB/c mice induced a time-dependent influx of mononuclear cells that were primarily macrophages of anti-inflammatory (M2 [F4/80high Gr1- CD11bhigh]) and proresolving (Mres [F4/80med CD11blow]) phenotypes, without changing the number of macrophages with a proinflammatory profile (M1 [F4/80low Gr1+ CD11bmed]). Pleural injection of Plg/Pla also increased M2 markers (CD206 and arginase-1) and secretory products (transforming growth factor ß and interleukin-6) and decreased the expression of inducible nitric oxide synthase (M1 marker). During the resolving phase of lipopolysaccharide (LPS)-induced inflammation when resolving macrophages predominate, we found increased Plg expression and Pla activity, further supporting a link between the Plg/Pla system and key cellular events in resolution. Indeed, Plg or Pla given at the peak of inflammation promoted resolution by decreasing neutrophil numbers and increasing neutrophil apoptosis and efferocytosis in a serine-protease inhibitor-sensitive manner. Next, we confirmed the ability of Plg/Pla to both promote efferocytosis and override the prosurvival effect of LPS via annexin A1. These findings suggest that Plg and Pla regulate several key steps in inflammation resolution, namely, neutrophil apoptosis, macrophage reprogramming, and efferocytosis, which have a major impact on the establishment of an efficient resolution process.


Assuntos
Anexina A1/metabolismo , Reprogramação Celular , Fibrinolisina/metabolismo , Macrófagos/metabolismo , Plasminogênio/metabolismo , Doença Aguda , Animais , Anexina A1/genética , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/metabolismo , Apoptose/efeitos dos fármacos , Apoptose/genética , Fibrinolisina/genética , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Lipopolissacarídeos/toxicidade , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Neutrófilos/metabolismo , Neutrófilos/patologia , Plasminogênio/genética , Células RAW 264.7
6.
PLoS One ; 12(3): e0174827, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28358853

RESUMO

Cleavage activation of the hemagglutinin (HA) protein by host proteases is a crucial step in the infection process of influenza A viruses (IAV). However, IAV exists in eighteen different HA subtypes in nature and their cleavage sites vary considerably. There is uncertainty regarding which specific proteases activate a given HA in the human respiratory tract. Understanding the relationship between different HA subtypes and human-specific proteases will be valuable in assessing the pandemic potential of circulating viruses. Here we utilized fluorogenic peptides mimicking the HA cleavage motif of representative IAV strains causing disease in humans or of zoonotic/pandemic potential and tested them with a range of proteases known to be present in the human respiratory tract. Our results show that peptides from the H1, H2 and H3 subtypes are cleaved efficiently by a wide range of proteases including trypsin, matriptase, human airway tryptase (HAT), kallikrein-related peptidases 5 (KLK5) and 12 (KLK12) and plasmin. Regarding IAVs currently of concern for human adaptation, cleavage site peptides from H10 viruses showed very limited cleavage by respiratory tract proteases. Peptide mimics from H6 viruses showed broader cleavage by respiratory tract proteases, while H5, H7 and H9 subtypes showed variable cleavage; particularly matriptase appeared to be a key protease capable of activating IAVs. We also tested HA substrate specificity of Factor Xa, a protease required for HA cleavage in chicken embryos and relevant for influenza virus production in eggs. Overall our data provide novel tool allowing the assessment of human adaptation of IAV HA subtypes.


Assuntos
Hemaglutininas/metabolismo , Vírus da Influenza A/metabolismo , Peptídeos/metabolismo , Fator Xa/genética , Fator Xa/metabolismo , Fibrinolisina/genética , Fibrinolisina/metabolismo , Hemaglutininas/genética , Humanos , Vírus da Influenza A Subtipo H9N2/metabolismo , Vírus da Influenza A/genética , Calicreínas/genética , Calicreínas/metabolismo , Peptídeo Hidrolases/genética , Peptídeo Hidrolases/metabolismo , Peptídeos/genética , Proteólise , Sistema Respiratório/metabolismo , Tripsina/genética , Tripsina/metabolismo
7.
Endocrinology ; 157(11): 4487-4498, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27690691

RESUMO

Rupture of fetal membranes (ROM) can initiate parturition at both term and preterm birth. Apoptosis of the amnion epithelium plays a key role in structural remodeling of the membranes preceding ROM. However, the causative factors for apoptosis remain unidentified. Toward the end of gestation, a feed-forward regeneration of cortisol via 11ß-hydroxysteroid dehydrogenase 1 exists in the fetal membranes. Here, we have examined whether cortisol accumulation is a causative factor for amnion cells apoptosis. By using primary human amnion epithelial and fibroblast cells, we demonstrated cortisol induced apoptosis specifically in epithelial cells but not in fibroblasts via reciprocal regulation of tissue-type plasminogen activator (tPA)/plasmin system. Cortisol increased PLAT expression, the gene encoding tPA, via glucocorticoid receptor binding to a glucocorticoid response element in PLAT promoter, thereby increasing plasmin activity in epithelial cells. Further study revealed that a Fas-mediated extrinsic apoptotic pathway was involved in the induction of epithelial cells apoptosis by cortisol, which was blocked by inhibiting either tPA or plasmin. Consistently, cortisol increased cleaved-caspase-3 and tPA abundance in amnion tissue explants. Moreover, the abundance of cortisol, cleaved-caspase-3, and tPA was significantly increased in amnion tissue after labor-initiated spontaneous rupture of membranes. In conclusion, local accumulation of cortisol is a causative factor for amnion epithelial apoptosis via activation of tPA/plasmin system toward the end of gestation. This may contribute to the ROM at both term and preterm birth.


Assuntos
Âmnio/citologia , Células Epiteliais/citologia , Células Epiteliais/metabolismo , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/metabolismo , Âmnio/metabolismo , Apoptose/genética , Apoptose/fisiologia , Western Blotting , Caspase 3/metabolismo , Células Cultivadas , Imunoprecipitação da Cromatina , Membranas Extraembrionárias/metabolismo , Feminino , Fibrinolisina/genética , Fibrinolisina/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Hidrocortisona/metabolismo , Marcação In Situ das Extremidades Cortadas , Técnicas In Vitro , Gravidez , Regiões Promotoras Genéticas/genética , Ligação Proteica , RNA Interferente Pequeno , Ativador de Plasminogênio Tecidual/genética , Ativador de Plasminogênio Tecidual/metabolismo
8.
Oncotarget ; 7(30): 47720-47737, 2016 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-27351226

RESUMO

The link between oncogenic RAS expression and the acquisition of the invasive phenotype has been attributed to alterations in cellular activities that control degradation of the extracellular matrix. Oncogenic RAS-mediated upregulation of matrix metalloproteinase 2 (MMP-2), MMP-9 and urokinase-type plasminogen activator (uPA) is critical for invasion through the basement membrane and extracellular matrix. The uPA converts cell surface-bound plasminogen to plasmin, a process that is regulated by the binding of plasminogen to specific receptors on the cell surface, however, the identity of the plasminogen receptors that function in this capacity is unclear. We have observed that transformation of cancer cells with oncogenic forms of RAS increases plasmin proteolytic activity by 2- to 4-fold concomitant with a 3-fold increase in cell invasion. Plasminogen receptor profiling revealed RAS-dependent increases in both S100A10 and cytokeratin 8. Oncogenic RAS expression increased S100A10 gene expression which resulted in an increase in S100A10 protein levels. Analysis with the RAS effector-loop mutants that interact specifically with Raf, Ral GDS pathways highlighted the importance of the RalGDS pathways in the regulation of S100A10 gene expression. Depletion of S100A10 from RAS-transformed cells resulted in a loss of both cellular plasmin generation and invasiveness. These results strongly suggest that increases in cell surface levels of S100A10, by oncogenic RAS, plays a critical role in RAS-stimulated plasmin generation, and subsequently, in the invasiveness of oncogenic RAS expressing cancer cells.


Assuntos
Anexina A2/metabolismo , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Peptídeo Hidrolases/metabolismo , Proteínas S100/metabolismo , Proteínas ras/genética , Células A549 , Animais , Anexina A2/genética , Linhagem Celular Tumoral , Ativação Enzimática , Fibrinolisina/genética , Fibrinolisina/metabolismo , Genes ras , Células HCT116 , Células HEK293 , Humanos , Células MCF-7 , Camundongos , Células NIH 3T3 , Peptídeo Hidrolases/genética , Proteínas S100/genética , Transfecção , Proteínas ras/biossíntese
9.
PLoS One ; 8(5): e62826, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23675430

RESUMO

Matriptase, a membrane-associated serine protease, plays an essential role in epidermal barrier function through activation of the glycosylphosphatidylinositol (GPI)-anchored serine protease prostasin. The matriptase-prostasin proteolytic cascade is tightly regulated by hepatocyte growth factor activator inhibitor (HAI)-1 such that matriptase autoactivation and prostasin activation occur simultaneously and are followed immediately by the inhibition of both enzymes by HAI-1. However, the mechanisms whereby matriptase acts on extracellular substrates remain elusive. Here we report that some active matriptase can escape HAI-1 inhibition by being rapidly shed from the cell surface. In the pericellular environment, shed active matriptase is able to activate hepatocyte growth factor (HGF), accelerate plasminogen activation, and shed syndecan 1. The amount of active matriptase shed is inversely correlated with the amount of antithrombin (AT) bound to the surface of the keratinocytes. Binding of AT to the surface of keratinocytes is dependent on a functional heparin binding site, Lys-125, and that the N-glycosylation site Asn-135 be unglycosylated. This suggests that ß-AT, and not α-AT, is responsible for regulation of pericellular matriptase activity in keratinocytes. Keratinocytes appear to rely on AT to regulate the level of pericellular active matriptase much more than breast and prostate epithelial cells in which AT regulation of matriptase activity occurs at much lower levels than keratinocytes. These results suggest that keratinocytes employ two distinct serine protease inhibitors to control the activation and processing of two different sets of matriptase substrates leading to different biological events: 1) HAI-1 for prostasin activation/inhibition, and 2) AT for the pericellular proteolysis involved in HGF activation, accelerating plasminogen activation, and shedding of syndecans.


Assuntos
Antitrombinas/farmacologia , Fibrinolisina/metabolismo , Fator de Crescimento de Hepatócito/metabolismo , Queratinócitos/efeitos dos fármacos , Serina Endopeptidases/metabolismo , Sindecanas/metabolismo , Antitrombinas/metabolismo , Sítios de Ligação , Linhagem Celular , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Feminino , Fibrinolisina/genética , Regulação da Expressão Gênica , Heparina/química , Heparina/metabolismo , Fator de Crescimento de Hepatócito/genética , Humanos , Queratinócitos/citologia , Queratinócitos/metabolismo , Masculino , Glândulas Mamárias Humanas/citologia , Glândulas Mamárias Humanas/efeitos dos fármacos , Glândulas Mamárias Humanas/metabolismo , Especificidade de Órgãos , Próstata/citologia , Próstata/efeitos dos fármacos , Próstata/metabolismo , Ligação Proteica , Proteínas Secretadas Inibidoras de Proteinases/genética , Proteínas Secretadas Inibidoras de Proteinases/metabolismo , Serina Endopeptidases/genética , Sindecanas/genética
10.
Matrix Biol ; 32(5): 277-87, 2013 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-23369837

RESUMO

Chronic kidney disease (CKD) is characterised by the pathological accumulation of extracellular matrix (ECM) proteins leading to progressive kidney scarring via glomerular and tubular basement membrane expansion. Increased ECM synthesis and deposition, coupled with reduced ECM breakdown contribute to the elevated ECM level in CKD. Previous pre-clinical studies have demonstrated that increased plasmin activity has a beneficial effect in the protein overload model of CKD. As plasmin activation is downregulated by the action of the thrombin activated fibrinolytic inhibitor (TAFI), we tested the hypothesis that inhibition of TAFI might increase plasmin activity and reduce ECM accumulation in an in vitro model of glucose induced ECM expansion. Treatment of NRK52E tubular epithelial cells with increasing concentrations of glucose resulted in a 40% increase in TAFI activity, a 38% reduction in plasmin activity and a subsequent increase in ECM accumulation. In this model system, application of the previously reported TAFI inhibitor UK-396082 [(2S)-5-amino-2-[(1-n-propyl-1H-imidazol-4-yl)methyl]pentanoic acid] caused a reduction in TAFI activity, increased plasmin activity and induced a parallel decrease in ECM levels. In contrast, RNAi knockdown of plasmin resulted in an increase in ECM levels. The data presented here indicate that high glucose induces TAFI activity, inhibiting plasmin activation which results in elevated ECM levels in tubular epithelial cells. The results support the hypothesis that UK-396082 is able to reduce TAFI activity, normalising plasmin activity and preventing excess ECM accumulation suggesting that TAFI inhibition may have potential as an anti-scarring strategy in CKD.


Assuntos
Aminoácidos/farmacologia , Carboxipeptidase B2/genética , Inibidores Enzimáticos/farmacologia , Células Epiteliais/efeitos dos fármacos , Matriz Extracelular/efeitos dos fármacos , Imidazóis/farmacologia , Túbulos Renais Proximais/efeitos dos fármacos , Animais , Carboxipeptidase B2/antagonistas & inibidores , Carboxipeptidase B2/metabolismo , Linhagem Celular , Células Epiteliais/enzimologia , Células Epiteliais/patologia , Matriz Extracelular/enzimologia , Matriz Extracelular/patologia , Fibrinolisina/antagonistas & inibidores , Fibrinolisina/genética , Fibrinolisina/metabolismo , Fibrinólise/efeitos dos fármacos , Fibrinólise/genética , Expressão Gênica/efeitos dos fármacos , Glucose/efeitos adversos , Humanos , Túbulos Renais Proximais/enzimologia , Túbulos Renais Proximais/patologia , Modelos Biológicos , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Insuficiência Renal Crônica/tratamento farmacológico , Insuficiência Renal Crônica/enzimologia , Insuficiência Renal Crônica/genética , Insuficiência Renal Crônica/patologia , Trombina/genética , Trombina/metabolismo
11.
Mol Cancer ; 12: 15, 2013 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-23442884

RESUMO

BACKGROUND: Previously, using miRNA microarray, we have found that miR-29c is significantly downregulated in advanced gastric carcinoma. In the present study, we investigated whether miR-29c functions as a tumor-suppressor miRNA in gastric carcinoma cells. For this purpose, we verified the downregulation of miR-29c in gastric carcinoma tissues, and assessed the biological effect of miR-29c on gastric carcinoma cells. RESULTS: In miR-29c-transfected cells, both proliferation and colony formation ability on soft agar were significantly decreased. Although apoptosis was not induced, BrdU incorporation and the proportion of cells positive for phospho-histone H3 (S10) were significantly decreased in miR-29c-transfected cells, indicating that miR-29c may be involved in the regulation of cell proliferation. To explain the mechanism of growth suppression by miR-29c, we explored differentially expressed genes (>2-fold) in miR-29c-transfected cells in comparison with negative control transfected cells using microarray. RCC2, PPIC and CDK6 were commonly downregulated in miR-29c-transfected MKN45, MKN7 and MKN74 cells, and all of the genes harbored miR-29c target sequences in the 3'-UTR of their mRNA. RCC2 and PPIC were actually upregulated in gastric carcinoma tissues, and therefore both were identified as possible targets of miR-29c in gastric carcinoma. To ascertain whether downregulation of RCC2 and/or PPIC is involved in the growth suppression by miR-29c, we transfected siRNAs against RCC2 and PPIC into MKN45 and determined cell viability, the rate of BrdU incorporation, and caspase activity. We found that RCC2-knockdown decreased both cell viability and BrdU incorporation without any increase of caspase activity, while PPIC-knockdown did not, indicating that downregulation of RCC2 may be at least partly responsible for the growth suppression by miR-29c. CONCLUSIONS: Our findings indicate that miR-29c may have tumor-suppressive functions in gastric carcinoma cells, and that its decreased expression may confer a growth advantage on tumor cells via aberrant expression of RCC2.


Assuntos
Carcinoma/genética , Proteínas Cromossômicas não Histona/genética , Regulação Neoplásica da Expressão Gênica , Fatores de Troca do Nucleotídeo Guanina/genética , MicroRNAs/genética , Neoplasias Gástricas/genética , Apoptose/genética , Carcinoma/metabolismo , Proliferação de Células , Sobrevivência Celular , Proteínas Cromossômicas não Histona/metabolismo , Quinase 6 Dependente de Ciclina/genética , Quinase 6 Dependente de Ciclina/metabolismo , Epigênese Genética , Fibrinolisina/genética , Fibrinolisina/metabolismo , Técnicas de Silenciamento de Genes , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Neoplasias Gástricas/metabolismo , alfa 2-Antiplasmina/genética , alfa 2-Antiplasmina/metabolismo
12.
FASEB J ; 26(11): 4445-57, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22815383

RESUMO

Rearrangement of the skin during wound healing depends on plasmin and plasminogen, which serve to degrade fibrin depositions in the provisional matrix and thereby facilitate keratinocyte migration. In the current study, we investigated whether plasmin and plasminogen likewise played a role during the development of skin cancer. To test this, we set up a chemically induced skin tumor model in a cohort of mice and found that skin tumor growth in Plg(-/-) male mice was reduced by 52% compared with wild-type controls. Histological analyses suggested that the growth-restricting effect of plasminogen deficiency was due to thrombosis and lost patency of the tumor vasculature, resulting in tumor necrosis. The connection between plasmin-dependent fibrinolysis, vascular patency, and tumor growth was further substantiated as the effect of plasminogen deficiency on tumor growth could be reverted by superimposing heterozygous fibrinogen deficiency on Plg(-/-) mice. Tumors derived from these Fib(-/+);Plg(-/-) mice displayed a significantly decreased level of tumor thrombosis compared with Plg(-/-) mice. In summary, these data indicate that plasmin-driven fibrinolysis facilitates tumor growth by maintaining patency of the tumor vasculature.


Assuntos
Fibrinolisina/metabolismo , Fibrinólise/fisiologia , Neoplasias Cutâneas/patologia , Animais , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Feminino , Fibrina/genética , Fibrina/metabolismo , Fibrinogênio/genética , Fibrinogênio/metabolismo , Fibrinolisina/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Laminina/genética , Laminina/metabolismo , Masculino , Camundongos , Camundongos Knockout , Neoplasias Experimentais/induzido quimicamente , Neoplasias Experimentais/metabolismo , Ovariectomia , Fatores Sexuais , Neoplasias Cutâneas/metabolismo , Trombose/metabolismo
13.
J Biol Chem ; 287(11): 8641-51, 2012 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-22267728

RESUMO

Accumulation of amyloid-ß peptides (Aß) in the brain is a common pathological feature of Alzheimer disease (AD). Aggregates of Aß are neurotoxic and appear to be critically involved in the neurodegeneration during AD pathogenesis. Accumulation of Aß could be caused by increased production, as indicated by several mutations in the amyloid precursor protein or the γ-secretase components presenilin-1 and presenilin-2 that cause familial early-onset AD. However, recent data also indicate a decreased clearance rate of Aß in AD brains. We recently demonstrated that Aß undergoes phosphorylation by extracellular or cell surface-localized protein kinase A, leading to increased aggregation. Here, we provide evidence that phosphorylation of monomeric Aß at Ser-8 also decreases its clearance by microglial cells. By using mass spectrometry, we demonstrate that phosphorylation at Ser-8 inhibited the proteolytic degradation of monomeric Aß by the insulin-degrading enzyme, a major Aß-degrading enzyme released from microglial cells. Phosphorylation also decreased the degradation of Aß by the angiotensin-converting enzyme. In contrast, Aß degradation by plasmin was largely unaffected by phosphorylation. Thus, phosphorylation of Aß could play a dual role in Aß metabolism. It decreases its proteolytic clearance and also promotes its aggregation. The inhibition of extracellular Aß phosphorylation, stimulation of protease expression and/or their proteolytic activity could be explored to promote Aß degradation in AD therapy or prevention.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Insulisina/metabolismo , Microglia/metabolismo , Peptidil Dipeptidase A/metabolismo , Proteólise , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Doença de Alzheimer/prevenção & controle , Peptídeos beta-Amiloides/genética , Linhagem Celular , Proteínas Quinases Dependentes de AMP Cíclico/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Fibrinolisina/genética , Fibrinolisina/metabolismo , Humanos , Insulisina/genética , Microglia/patologia , Peptidil Dipeptidase A/genética , Serina/genética , Serina/metabolismo
14.
J Biol Chem ; 286(37): 32231-43, 2011 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-21795691

RESUMO

Collagen deposition by fibroblasts contributes to scarring in fibrotic diseases. Activation of protein kinase A (PKA) by cAMP represents a pivotal brake on fibroblast activation, and the lipid mediator prostaglandin E(2) (PGE(2)) exerts its well known anti-fibrotic actions through cAMP signaling. However, fibrotic fibroblasts from the lungs of patients with idiopathic pulmonary fibrosis, or of mice with bleomycin-induced fibrosis, are resistant to the normal collagen-inhibiting action of PGE(2). In this study, we demonstrate that plasminogen activation to plasmin restores PGE(2) sensitivity in fibrotic lung fibroblasts from human and mouse. This involves amplified PKA signaling resulting from the promotion of new interactions between AKAP9 and PKA regulatory subunit II in the perinuclear region as well as from the inhibition of protein phosphatase 2A. This is the first report to show that an extracellular mediator can dramatically reorganize and amplify the intracellular PKA-A-kinase anchoring protein signaling network and suggests a new strategy to control collagen deposition by fibrotic fibroblasts.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Dinoprostona/farmacologia , Fibrinolisina/metabolismo , Fibroblastos/metabolismo , Pulmão/metabolismo , Ocitócicos/farmacologia , Fibrose Pulmonar/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas de Ancoragem à Quinase A/genética , Proteínas de Ancoragem à Quinase A/metabolismo , Animais , Antibióticos Antineoplásicos/efeitos adversos , Antibióticos Antineoplásicos/farmacologia , Bleomicina/efeitos adversos , Bleomicina/farmacologia , Núcleo Celular/genética , Núcleo Celular/metabolismo , AMP Cíclico/genética , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/genética , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Resistência a Medicamentos/efeitos dos fármacos , Resistência a Medicamentos/genética , Fibrinolisina/genética , Fibroblastos/patologia , Humanos , Pulmão/patologia , Camundongos , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Proteína Fosfatase 2/genética , Proteína Fosfatase 2/metabolismo , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/genética , Fibrose Pulmonar/patologia , Transdução de Sinais/genética
15.
Protein Pept Lett ; 18(12): 1204-11, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21707521

RESUMO

Galanin is a neuropeptide that is widely distributed in the central and peripheral nervous systems. In a previous study, we showed that a small cell lung carcinoma (SCLC) cell line, SBC-3A, released progalanin but not galanin, and that progalanin was then converted to galanin(1-20), the active form. Because the galanin(1-20) had undergone hydrolysis at Arg and Lys residues, the protease concerned was surmised to have a trypsin-like activity. The present study was performed to identify the trypsin-like protease which had previously been found to activate progalanin in this tumor tissue. The protease was isolated using chromatography and electrophoresis, and identified in tumor extracts from SBC-3A tumor-bearing mice; the major protease was found to be plasmin. We next confirmed that extracellular processing of progalanin occurs in SCLC tumor tissue (tumors produced by the implantation of SBC-3A cells into mice), and in two types of breast tumor tissue (obtained by implantation into mice of BT-549 and MDA-MB-436 cells). In cell culture, processed forms of progalanin were undetectable in SBC-3A, BT-549 or MDA-MB-436 cells. Conversely, gel filtration chromatography analysis of tumor extracts from SBC-3A, BT-549 and MDA-MB-436-bearing mice, revealed that galanin-like immunoreactivity (galanin-LI) in these tumor extracts was due to the presence of progalanin (14 kDa) and galanin(1-20) (2 kDa). Moreover, trypsin-like protease activity was elevated, and plasmin was expressed abundantly in SBC-3A, BT-549 and MDA-MB-436 tumors in mice. In addition, tranexamic acid, a plasmin inhibitor, inhibited progalanin conversion to galanin(1-20). The present study revealed that plasmin was present in tumor tissue, and that it was responsible for processing progalanin to galanin(1-20) in the extracellular environment.


Assuntos
Fibrinolisina/metabolismo , Galanina/metabolismo , Animais , Western Blotting , Linhagem Celular Tumoral , Eletroforese em Gel Bidimensional , Fibrinolisina/genética , Galanina/genética , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Camundongos , Radioimunoensaio , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Carcinoma de Pequenas Células do Pulmão/genética , Carcinoma de Pequenas Células do Pulmão/metabolismo
16.
Circ Res ; 108(12): 1419-28, 2011 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-21546607

RESUMO

RATIONALE: The antiangiogenic activity of rPAI-1(23), a truncated plasminogen activator inhibitor-1 (PAI-1) protein, induces vasa vasorum collapse and significantly reduces plaque area and plaque cholesterol in hypercholesterolemic low-density lipoprotein receptor-deficient/apolipoprotein B48-deficient mice. OBJECTIVE: The objective of this study was to examine rPAI-1(23)-stimulated mechanisms that cause vasa vasorum collapse. METHODS AND RESULTS: The rPAI-1(23) protein opposed PAI-1 antiproteolytic function by stimulating a 1.6-fold increase in plasmin activity compared with the saline-treated counterpart. The increased proteolytic activity corresponded to increased activity of matrix metalloproteinase-3 and degradation of fibrin(ogen), nidogen, and perlecan in the adventitia of descending aortas. PAI-1 activity was reduced by 48% in response to rPAI-1(23); however, PAI-1 protein expression levels were similar in the rPAI-1(23)- and saline-treated hypercholesterolemic mice. Coimmunoprecipitation assays demonstrated a novel PAI-1-plasminogen complex in protein from the descending aorta of rPAI-1(23)- and saline-treated mice, but complexed PAI-1 was 1.6-fold greater in rPAI-1(23)-treated mice. Biochemical analyses demonstrated that rPAI-1(23) and PAI-1 binding interactions with plasminogen increased plasmin activity and reduced PAI-1 antiproteolytic activity. CONCLUSIONS: We conclude that rPAI-1(23) causes regression or collapse of adventitial vasa vasorum in hypercholesterolemic mice by stimulating an increase in plasmin activity. The rPAI-1(23)-enhanced plasmin activity was achieved through a novel mechanism by which rPAI-1(23) and PAI-1 bound plasminogen in a cooperative manner to increase plasmin activity and reduce PAI-1 activity.


Assuntos
Inibidores da Angiogênese/farmacologia , Fibrinolisina/metabolismo , Hipercolesterolemia/metabolismo , Plasminogênio/metabolismo , Serpina E2/farmacologia , Vasa Vasorum/metabolismo , Animais , Fibrinolisina/genética , Hipercolesterolemia/genética , Hipercolesterolemia/patologia , Camundongos , Camundongos Knockout , Plasminogênio/genética , Vasa Vasorum/patologia
17.
J Biol Chem ; 286(11): 8952-60, 2011 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-21239499

RESUMO

The contribution of plasminogen (Plg)/plasmin, which have claimed to be the main fibrinolytic regulators in the bone metabolism, remains unclear. This study evaluated how the absence of Plg affects the function of osteoblast (OB) and osteoclast (OC). There was a larger population of pre-OCs in bone marrow-derived cells from the Plg(-/-) mice than the population of that from the WT mice. In addition, the absence of Plg suppressed the expression of osteoprotegerin in OBs. Moreover, an exogenous plasmin clearly induced the osteoprotegerin expression in Plg(-/-) OBs. The osteoclastogenesis of RAW264.7 mouse monocyte/macrophage lineage cells in co-culture with OBs from the Plg(-/-) mice was significantly accelerated in comparison with that in co-culture with OBs from the WT mice. Intriguingly, the accelerated OC differentiation of RAW264.7 cells co-cultured with Plg(-/-) OBs was clearly suppressed by the treatment of an exogenous plasmin. Consequently, Plg(-/-) mice display decreased bone mineral density. These findings could eventually lead to the development of new clinical therapies for bone disease caused by a disorder of the fibrinolytic system.


Assuntos
Densidade Óssea/fisiologia , Células da Medula Óssea/metabolismo , Diferenciação Celular/fisiologia , Fibrinolisina/metabolismo , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Plasminogênio/metabolismo , Animais , Doenças Ósseas/genética , Doenças Ósseas/metabolismo , Células da Medula Óssea/citologia , Osso e Ossos/citologia , Osso e Ossos/metabolismo , Linhagem Celular , Fibrinolisina/genética , Camundongos , Camundongos Knockout , Osteoblastos/citologia , Osteoclastos/citologia , Plasminogênio/genética
18.
Zhongguo Zhong Yao Za Zhi ; 35(15): 1925-30, 2010 Aug.
Artigo em Chinês | MEDLINE | ID: mdl-20931838

RESUMO

OBJECTIVE: To obtain the cDNA sequence encoding fibrinolytic enzyme from Eupolyphaga sinensis and express it in prokaryotic and eukaryotic expression system. METHOD: The primers were designed according to the cDNA of other animals'fibrinolytic enzyme. The cDNA sequence was cloned by RT-PCR and 3 RACE. RESULT: Sequence analysis revealed that the length of the cDNA fragment was 672 bp and encoded a protein of 224 amino acid residues, the N end amino acid sequence residues was IVGG in accordance with other fibrinolytic enzyme. The cDNA sequence was expressed in E. coli, inactive protein was obtained. While expressed in Pichia pastoris, recombinant protein had fibrinolytic activity. CONCLUSION: The cDNA sequence of fibrinolytic enzyme from E. sinensis Walker was cloned and expressed for the first time and it proved a good basis for further functional study of the enzyme.


Assuntos
Clonagem Molecular , Baratas/enzimologia , Fibrinolisina/genética , Expressão Gênica , Proteínas de Insetos/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Baratas/química , Baratas/genética , DNA Complementar/química , DNA Complementar/genética , Fibrinolisina/química , Fibrinolisina/metabolismo , Proteínas de Insetos/química , Proteínas de Insetos/metabolismo , Dados de Sequência Molecular , Alinhamento de Sequência
19.
Exp Mol Pathol ; 88(2): 278-86, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20079732

RESUMO

Annexin II, an abundant phospholipids binding cell surface protein, binds tPA and functions as a regulator of fibrinolysis. Annexin II also mediates angiogenesis and enhances tumor growth and metastasis. However, the mechanism supporting this role is not known. Using human breast cancer model we show that invasive human breast cancer cells (MDA-MB231) synthesize annexin II and tissue plasminogen activator (tPA). In vitro both annexin II and tPA interacts which in turn convert zymogen plasminogen to reactive enzyme plasmin. Cell surface produced plasmin inhibited the migration of MDA-MB231 cells. Silencing of annexin II gene in MDA-MB231 cells abolished tPA binding therefore inhibited tPA dependent plasmin generation. These annexin II suppressed MDA-MB231 cells showed reduced motility. Immunohistochemical analysis of prediagnosed clinical specimens showed abundant secretion of tPA and expression of annexin II on the surface of invasive human breast cancer cells which correlates with neovascularization of the tumor. Taken together, these data indicate that annexin II may regulate localized plasmin generation in breast cancer. This may be an early event switching breast cancer from the prevascular phase to the vascular phase and thus contributing to aggressive cancer with the possibility of metastasis. The data provide a mechanism explaining the role of annexin II in breast cancer progression and suggest that annexin II may be an attractive target for therapeutic strategies aimed to inhibit angiogenesis and breast cancer.


Assuntos
Anexina A2/fisiologia , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/patologia , Fibrinolisina/genética , Anexina A2/metabolismo , Sequência de Bases , Western Blotting , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Movimento Celular , Feminino , Inativação Gênica , Humanos , Imuno-Histoquímica , Neovascularização Patológica/prevenção & controle , Proteínas Recombinantes/metabolismo , Ativador de Plasminogênio Tecidual/genética , Ativador de Plasminogênio Tecidual/metabolismo , Cicatrização
20.
Cell Mol Life Sci ; 67(9): 1505-18, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20112045

RESUMO

The human alpha(2)-plasmin inhibitor (A2PI) possesses unique N- and C-terminal extensions that significantly influence its biological activities. The C-terminal segment, A2PIC (Asn(398)-Lys(452)), contains six lysines thought to be involved in the binding to lysine-binding sites in the kringle domains of human plasminogen, of which four (Lys(422), Lys(429), Lys(436), Lys(452)) are completely and two (Lys(406), Lys(415)) are partially conserved. Multiple Lys to Ala mutants of A2PIC were expressed in Escherichia coli and used in intrinsic fluorescence titrations with kringle domains K1, K4, K4 + 5, and K1 + 2 + 3 of human plasminogen. We were able to identify the C-terminal Lys(452) as the main binding partner in recombinant A2PIC (rA2PIC) constructs with isolated kringles. We could show a cooperative, zipper-like enhancement of the interaction between C-terminal Lys(452) and internal Lys(436) of rA2PIC and isolated K1 + 2 + 3, whereas the other internal lysine residues contribute only to a minor extent to the binding process. Sulfated Tyr(445) in the unique C-terminal segment revealed no influence on the binding affinity to kringle domains.


Assuntos
Fibrinolisina/metabolismo , Plasminogênio/metabolismo , Conformação Proteica , alfa 2-Antiplasmina/química , alfa 2-Antiplasmina/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Fibrinolisina/química , Fibrinolisina/genética , Humanos , Kringles , Lisina/química , Lisina/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Peptídeos/química , Peptídeos/genética , Peptídeos/metabolismo , Plasminogênio/química , Plasminogênio/genética , Ligação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , alfa 2-Antiplasmina/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA