Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
J BUON ; 20(6): 1426-31, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26854437

RESUMO

PURPOSE: Erythropoiesis-stimulating agents (ESAs) are recommended for treating chemotherapy-induced anemia in breast cancer patients. Reduced survival rates in ESAs-treated patients have been reported, possibly due to thromboembolic complications, however the exact mechanism remains obscure. The principal activator of blood coagulation in cancer is tissue factor (TF). There are data that erythropoietin receptor (EPO-R) is expressed in tumor cells. The purpose of this study was to evaluate the expression of EPO-R and TF in loco in breast cancer. METHODS: The expression of EPO-R and TF was investigated in 24 invasive breast carcinoma specimens. Immunohistochemical (IHC) methodologies according to ABC technique and double-staining IHC procedure were employed utilizing antibodies against EPO-R and TF. RESULTS: Expression of EPO-R and TF was demonstrated in the tumor cells in all breast cancer specimens. No staining for EPO-R and TF was visualized in normal breast tissue. Double staining studies revealed co-expression of both EPO-R and TF in breast cancer cells and endothelial cells. CONCLUSIONS: EPO-R and TF expression and their coexpression in breast cancer cells suggest a possibility that EPO-R might be responsible for some adverse effects and reduced survival observed in ESAs-treated breast cancer patients with anemia, possibly due to the interaction with TF. Further experimental studies are warranted to determine the role of both EPO-R and TF in the treatment with ESAs of breast cancer patients with chemotherapy-induced anemia.


Assuntos
Neoplasias da Mama/química , Receptores da Eritropoetina/análise , Tromboplastina/análise , Anemia/tratamento farmacológico , Neoplasias da Mama/mortalidade , Feminino , Humanos , Imuno-Histoquímica , Tromboembolia/etiologia
2.
Ann Hematol ; 93(5): 779-84, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24158387

RESUMO

In gastric cancer, hemostatic system components contribute to cancer progression, as activation of factor X (FX) was observed. The protein Z (PZ)/protein Z-dependent protease inhibitor (ZPI) complex inhibits factor Xa proteolytic activity. The purpose of this study was to determine the distribution of ZPI and PZ in relation to FX, and prothrombin fragment (F1 + 2), a standard marker for blood coagulation activation, in human gastric cancer tissue. ABC procedures and a double staining method employed polyclonal antibodies against PZ, FX, and F1 + 2 and a monoclonal antibody against ZPI. In situ hybridization (ISH) methods employed biotin-labeled 25-nucleotide single-stranded DNA probes directed to either PZ or ZPI mRNAs. FX and components of PZ/ZPI coagulation inhibitory system were observed in cancer cells. F1 + 2 was observed in gastric cancer cells as well. Double staining studies revealed FX/PZ, FX/ZPI, and PZ/ZPI co-localization on gastric cancer cells. ISH studies demonstrated the presence of PZ mRNA and ZPI mRNA in gastric cancer cells indicating induced synthesis of these proteins. The co-localization of PZ/ZPI and FX in gastric cancer cells indicates in loco that these proteins may play a role in anticoagulant events at the tumor tissue.


Assuntos
Adenocarcinoma/genética , Proteínas Sanguíneas/genética , Fator Xa/genética , Regulação Neoplásica da Expressão Gênica , RNA Mensageiro/genética , Serpinas/genética , Neoplasias Gástricas/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Coagulação Sanguínea , Proteínas Sanguíneas/metabolismo , Progressão da Doença , Fator Xa/metabolismo , Humanos , Imuno-Histoquímica , Hibridização In Situ , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Protrombina/genética , Protrombina/metabolismo , RNA Mensageiro/metabolismo , Serpinas/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia
3.
J Pharmacol Exp Ther ; 331(3): 940-5, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19776384

RESUMO

Bovine aprotinin, a reversible inhibitor of plasmin and kallikrein, has been clinically approved for over two decades to prevent perioperative blood loss during cardiac surgery. However, because of postoperative renal dysfunction in thousands of these patients, aprotinin was voluntarily withdrawn from the market. Our earlier studies indicated that a R24K mutant of the first Kunitz-type domain of human tissue factor pathway inhibitor-2 (R24K KD1) exhibited plasmin inhibitory activity equivalent to aprotinin in vitro. In this study, we compared the effects on renal function after infusion of aprotinin and recombinant R24K KD1 in chronically instrumented, conscious rats. Aprotinin-infused rats exhibited statistically significant decreases in glomerular filtration rate and effective renal plasma flow relative to rats infused with phosphate-buffered saline (PBS) or R24K KD1 dissolved in PBS. In addition, aprotinin-treated rats exhibited marked increases in serum creatinine, blood urea nitrogen, urinary protein, and effective renal vascular resistance, whereas these renal parameters remained essentially unchanged in vehicle and R24K KD1-treated rats for a one-week period. Moreover, with use of a highly sensitive apoptosis detection assay, a significant increase in the rate of early and late apoptotic events in renal tubule cells occurred in aprotinin-treated rats relative to R24K KD1-treated rats. In addition, histological examination of the rat kidney revealed markedly higher levels of protein reabsorption droplets in the aprotinin-infused rats. Our data collectively provide suggestive evidence that R24K KD1 does not induce the renal dysfunction associated with aprotinin, and may be an effective clinical alternative to aprotinin as an antifibrinolytic agent in cardiac surgery.


Assuntos
Aprotinina/efeitos adversos , Glicoproteínas/efeitos adversos , Rim/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Aprotinina/administração & dosagem , Aprotinina/genética , Bovinos , Escherichia coli/genética , Feminino , Glicoproteínas/administração & dosagem , Glicoproteínas/genética , Humanos , Rim/patologia , Testes de Função Renal , Microscopia Confocal , Mutação , Ratos , Ratos Long-Evans , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/efeitos adversos , Proteínas Recombinantes/genética , Transfecção
4.
Arch Biochem Biophys ; 482(1-2): 58-65, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19103149

RESUMO

Tissue factor pathway inhibitor-2 (TFPI-2) is a serine proteinase inhibitor that induces caspase-mediated apoptosis when offered to a variety of tumor cells. In order to investigate the mechanism of TFPI-2-induced apoptosis, we initially studied the uptake and trafficking of TFPI-2 by HT-1080 cells. Exogenously offered TFPI-2 was rapidly internalized and distributed in both the cytosolic and nuclear fractions. Nuclear localization of TFPI-2 was also detected in a variety of endothelial cells constitutively expressing TFPI-2. Nuclear localization of TFPI-2 required a NLS sequence located in its Lys/Arg-rich C-terminal tail comprising residues 191-211, as a TFPI-2 construct lacking the C-terminal tail failed to localize to the nucleus. Complexes of TFPI-2 and importin-alpha were co-immunoprecipitated from cell lysates of HT-1080 cells either offered or overexpressing this protein, providing evidence that TFPI-2 was shuttled to the nucleus by the importin system. Our results provide the initial description of TFPI-2 internalization and translocation to the nucleus in a number of cells.


Assuntos
Núcleo Celular/metabolismo , Glicoproteínas/metabolismo , Carioferinas/metabolismo , Aorta , Apoptose , Capilares/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Endotélio Vascular/metabolismo , Fibrossarcoma , Glicoproteínas/genética , Humanos , Rim/embriologia , Cinética , Microscopia Confocal , Plasmídeos , Reação em Cadeia da Polimerase , Proteínas Recombinantes/metabolismo , Pele/irrigação sanguínea , Transfecção
5.
Apoptosis ; 13(5): 702-15, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18401718

RESUMO

Human TFPI-2 is an extracellular matrix-associated Kunitz-type serine proteinase inhibitor. We previously demonstrated that a human fibrosarcoma cell line, HT-1080, does not express TFPI-2, but genetic restoration of TFPI-2 expression in these cells markedly inhibited their growth and metastasis in vivo. In the present study, either full-length recombinant TFPI-2, or its mutated first Kunitz-type domain (R24K KD1), were offered to HT-1080 cells, and the degree of apoptosis assessed by nuclear fragmentation, ethidium bromide and acridine orange staining, fluorescence activated cell sorting, immunoblotting and gene expression profiling. R24K KD1 induced apoptosis in 69% of HT-1080 cells in a 48 h period compared to 39% for TFPI-2, while a KD1 preparation lacking a reactive site arginine/lysine residue (R24Q KD1) produced only an 18% apoptosis rate, suggesting that the observed apoptosis was related to proteinase inhibition. Immunoblotting experiments indicated increased caspase 3 and 9 activation, up-regulation of pro-apoptotic Bax and suppression of anti-apoptotic Bcl-2 protein. Finally, microarray analyses of R24K KD1-treated cells indicated elevated expression of several pro-apoptotic genes and under-expression of anti-apoptotic genes. Collectively, our results demonstrate that treatment of HT-1080 cells exogenously with either TFPI-2 or R24K KD1 activates caspase-mediated, pro-apoptotic signaling pathways resulting in apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Caspases/fisiologia , Glicoproteínas/fisiologia , Proteínas Reguladoras de Apoptose/biossíntese , Linhagem Celular Tumoral , Neoplasias do Colo/patologia , Regulação para Baixo , Fibrossarcoma/patologia , Citometria de Fluxo , Glicoproteínas/genética , Humanos , Proteínas Proto-Oncogênicas c-bcl-2/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Mensageiro/metabolismo , Proteínas Recombinantes/farmacologia , Regulação para Cima , Neoplasias da Bexiga Urinária/patologia , Proteína X Associada a bcl-2/efeitos dos fármacos , Proteína X Associada a bcl-2/metabolismo
6.
J Drug Target ; 16(3): 185-97, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18365880

RESUMO

Tissue factor (TF) is aberrantly expressed on tumor vascular endothelial cells (VECs) and on cancer cells in many malignant tumors, but not on normal VECs, making it a promising target for cancer therapy. As a transmembrane receptor for coagulation factor VIIa (fVIIa), TF forms a high-affinity complex with its cognate ligand, which is subsequently internalized through receptor-mediated endocytosis. Accordingly, we developed a method for selectively delivering EF24, a potent synthetic curcumin analog, to TF-expressing tumor vasculature and tumors using fVIIa as a drug carrier. EF24 was chemically conjugated to fVIIa through a tripeptide-chloromethyl ketone. After binding to TF-expressing targets by fVIIa, EF24 will be endocytosed along with the drug carrier and will exert its cytotoxicity. Our results showed that the conjugate inhibits vascular endothelial growth factor-induced angiogenesis in a rabbit cornea model and in a Matrigel model in athymic nude mice. The conjugate-induced apoptosis in tumor cells and significantly reduced tumor size in human breast cancer xenografts in athymic nude mice as compared with the unconjugated EF24. By conjugating potent drugs to fVIIa, this targeted drug delivery system has the potential to enhance therapeutic efficacy, while reducing toxic side effects. It may also prove to be useful for treating drug-resistant tumors and micro-metastases in addition to primary tumors.


Assuntos
Antineoplásicos/administração & dosagem , Compostos de Benzilideno/administração & dosagem , Fator VIIa/administração & dosagem , Neoplasias Experimentais/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Piperidonas/administração & dosagem , Tromboplastina/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Endocitose , Humanos , Camundongos , Neoplasias Experimentais/irrigação sanguínea , Coelhos , Tromboplastina/análise , Tromboplastina/fisiologia , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Mol Cancer ; 6: 20, 2007 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-17352822

RESUMO

BACKGROUND: Previous studies have shown that the expression of tissue factor pathway inhibitor-2 (TFPI-2), a matrix-associated Kunitz-type serine proteinase inhibitor, is markedly down-regulated in several tumor cells through hypermethylation of the TFPI-2 gene promoter. In the present study, RT-PCR analysis of total RNA from both human normal and tumor cells revealed a novel 289 nucleotide splice variant of the TFPI-2 transcript designated as aberrantly-spliced TFPI-2 (asTFPI-2). RESULTS: Nucleotide sequence analyses indicated that asTFPI-2 consists of complete exons II and V, fused with several nucleotides derived from exons III and IV, as well as six nucleotides derived from intron C. 5'- and 3'-RACE analyses of total RNA amplified exclusively the wild-type TFPI-2 transcript, indicating that asTFPI-2 lacks either a 5'-untranslated region (UTR) or a 3'-poly (A)+ tail. Quantitative real-time RT-PCR analyses revealed that several human tumor cells contain 4 to 50-fold more copies of asTFPI-2 in comparison to normal cells. In spite of the absence of a 5'-UTR or poly (A)+ tail, the asTFPI-2 variant exhibited a half-life of ~16 h in tumor cells. CONCLUSION: Our studies reveal the existence of a novel, aberrantly-spliced TFPI-2 transcript predominantly expressed in tumor cells and provides suggestive evidence for an additional mechanism for tumor cells to down-regulate TFPI-2 protein expression enhancing their ability to degrade the extracellular matrix.


Assuntos
Glicoproteínas/genética , Proteínas de Neoplasias/genética , Splicing de RNA , Regiões 5' não Traduzidas/genética , Linhagem Celular Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/metabolismo , Dactinomicina/farmacologia , Éxons/genética , Glicoproteínas/biossíntese , Glicoproteínas/química , Meia-Vida , Humanos , Íntrons/genética , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/química , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Inibidores da Síntese de Ácido Nucleico/farmacologia , Especificidade de Órgãos , Poli A/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Neoplásico/biossíntese , RNA Neoplásico/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcrição Gênica/efeitos dos fármacos , Regulação para Cima , Vísceras/metabolismo
8.
Arterioscler Thromb Vasc Biol ; 26(12): 2819-25, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17023682

RESUMO

OBJECTIVE: The purpose of this study is to investigate the expression and regulation of type-2 tissue factor pathway inhibitor (TFPI-2) in endothelial cells, as well as the regulation of human endothelial cell (EC) function by TFPI-2. METHODS AND RESULTS: Real-time polymerase chain reaction (PCR) and Western blot analysis revealed that vascular endothelial growth factor (VEGF) induced both time- and dose-dependent increase in TFPI-2 mRNA and protein expression in endothelial cells. TFPI-2 mRNA expression was also significantly upregulated by IL-1beta, and modestly increased by both tumor necrosis factor (TNF)-alpha and fibroblast growth factor (FGF)-2, but not placental growth factor (PlGF). VEGF upregulation of TFPI-2 was dramatically reduced by inhibition of the MEK pathway. Administration of TFPI-2 protein suppressed both VEGF and FGF-2 stimulation of EC proliferation in a dose-dependent manner. A recombinant preparation of the first Kunitz-type domain of TFPI-2 (KD1) did not suppress growth factor stimulation of EC proliferation, suggesting a mechanism distinct from the proteinase inhibitory activity of TFPI-2. Exogenously added TFPI-2 protein suppressed VEGF-induced EC migration in 2 different assays. Recombinant wt-KD1 or the R24K mutant of KD1, but not the R24Q mutant, dramatically suppressed VEGF-induced EC migration. TFPI-2 protein, but not recombinant KD1, blocked VEGF-induced activation of both Akt and ERK1/2 in ECs. At higher doses, TFPI-2 protein blocked VEGFR2 activation. CONCLUSIONS: Our data suggest that VEGF-upregulation of TFPI-2 expression in endothelial cells may represent a mechanism for negative feedback regulation and modulation of its pro-angiogenic action on endothelial cells. TFPI-2, or derivatives of TFPI-2, may be novel therapeutics for treatment of angiogenic disease processes.


Assuntos
Proliferação de Células , Endotélio Vascular/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Glicoproteínas/metabolismo , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/fisiologia , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Endotélio Vascular/citologia , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fator 2 de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica , Glicoproteínas/genética , Humanos , Interleucina-1beta/farmacologia , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo
9.
Thromb Haemost ; 95(2): 243-52, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16493485

RESUMO

The cornerstone of hemostasis is the ability of the organism to limit the enzymatic processes involved, thereby avoiding thrombosis. For this, anticoagulant systems in place involve serpins, such as PAI-1 and antithrombin III, which bind to their targeted serine proteases and limit their period of activity. We have previously identified the serine protease furin as a platelet-derived enzyme with an intrinsic role in platelet functions. We now report that furin enzymatic activity decreased rapidly following platelet activation, corresponding with the increase in formation of a high 180 M(r) SDS-stable complex composed of furin and the PI8 serpin. PI8 is shown to be a platelet-derived constituent, synthesized by megakaryocytes and stored in platelets prior to its release. Immunoprecipitation and purification of the PI8-furin complex confirmed their direct interaction and indicates that one of the roles of PI8 is to inhibit furin enzymatic activity. Furthermore, our findings demonstrate the inhibitory capacity of exogenous PI8 in platelet aggregation assays. The finding that PI8 is released by platelets and controls functional responses suggests a role for this serpin in platelet-regulated pathophysiological responses.


Assuntos
Plaquetas/enzimologia , Furina/metabolismo , Serpinas/metabolismo , Linhagem Celular , Furina/antagonistas & inibidores , Humanos , Megacariócitos/enzimologia , Ativação Plaquetária , Agregação Plaquetária , Receptores Fc
10.
Virchows Arch ; 448(1): 46-51, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16193294

RESUMO

Thrombin, acting via protease-activated receptors (PARs), and tissue factor (TF) are involved in inflammation, tissue repair and tumorigenesis. Hepatocellular carcinomas (HCCs) usually complicate chronic liver diseases characterised by inflammation and fibrosis. The aim of this study was to describe the expression of PARs and TF in normal liver, cirrhosis and HCCs. We performed an immunohistochemical detection of PAR-1, PAR-3, PAR-4 and human TF in human tissue samples from 19 subnormal livers, 33 cirrhosis and 30 HCCs. PAR-1 was found on endothelial cells of sinusoids and larger vessels. In cirrhosis, spindle-shaped cells within septa and T lymphocytes were PAR-1 positive. A few PAR-1-positive tumour cells were found in 10% of HCCs. PAR-4 expression was restricted to macrophages, B lymphocytes and nerves. PAR-3 expression was rare. Unexpectedly, TF was expressed in 95% of normal livers and in 94% of cirrhosis but only in 50% of HCCs (p<0.001). Staining was mostly hepatocellular. No association existed between TF labelling and clinicopathological characteristics of HCCs. In conclusion, the pattern of expression of PARs is compatible with its role in chronic liver disease by promoting inflammation via immune cells and neurogenic stimulation. However, our data do not support a role for PARs or TF in HCC progression.


Assuntos
Carcinoma Hepatocelular/metabolismo , Cirrose Hepática/metabolismo , Neoplasias Hepáticas/metabolismo , Fígado/metabolismo , Receptores Ativados por Proteinase/biossíntese , Tromboplastina/biossíntese , Carcinoma Hepatocelular/patologia , Feminino , Humanos , Imuno-Histoquímica , Fígado/patologia , Cirrose Hepática/patologia , Neoplasias Hepáticas/patologia , Masculino , Pessoa de Meia-Idade
11.
Oncogene ; 22(29): 4509-16, 2003 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-12881707

RESUMO

We have shown previously that the tissue factor pathway inhibitor-2 (TFPI-2), a broad range proteinase inhibitor, is highly expressed in low-grade gliomas, but, minimally expressed or undetectable in glioblastomas, and that enforced expression of this gene reduces the invasive properties of brain tumor cells. Here, we examined the role of promoter methylation as a mechanism of TFPI-2 gene silencing. In SNB19 glioblastoma cells, which have no detectable TFPI-2 expression, 5-aza-2'-deoxycytidine (5aC), an inhibitor of DNA methyltransferase, induced TFPI-2 mRNA in a dose-dependent manner. Trichostatin A (TSA), the histone deacetylase (HDAC) inhibitor, by itself, was more efficient than 5aC in inducing TFPI-2 transcripts, and the 5aC+TSA combination resulted in highly synergistic reactivation of the gene, both at the transcript and protein levels. In Hs683 glioma cells, which express the TFPI-2 gene at high levels, transfection of the in vitro methylated TFPI-2 promoter constructs resulted in a drastic decrease of promoter activity compared to the unmethylated promoter. Further, the methylation-specific PCR in SNB19 and Hs683 cells showed that TFPI-2 gene repression was closely linked with methylation of the CpG islands in the promoter. Finally, the chromatin immunoprecipitation assays in SNB19 cells showed that the methylated and repressed TFPI-2 promoter was associated with the methyl-CpG binding protein 2 (MeCP2), and that gene reactivation resulted in the loss of MeCP2 from this site. These studies establish that TFPI-2 is transcriptionally silenced through promoter methylation in SNB19 cells.


Assuntos
Neoplasias Encefálicas/metabolismo , Proteínas Cromossômicas não Histona , Metilação de DNA , Inativação Gênica , Glioblastoma/metabolismo , Glioma/metabolismo , Glicoproteínas/genética , Regiões Promotoras Genéticas , Proteínas Repressoras , Azacitidina/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Cromatina/metabolismo , Ilhas de CpG , DNA (Citosina-5-)-Metiltransferases/antagonistas & inibidores , DNA (Citosina-5-)-Metiltransferases/metabolismo , Proteínas de Ligação a DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Inibidores Enzimáticos/farmacologia , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Glioma/tratamento farmacológico , Glioma/genética , Glicoproteínas/metabolismo , Inibidores de Histona Desacetilases , Histona Desacetilases/metabolismo , Humanos , Ácidos Hidroxâmicos/farmacologia , Proteína 2 de Ligação a Metil-CpG , Reação em Cadeia da Polimerase/métodos
12.
Oncogene ; 21(6): 921-8, 2002 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-11840337

RESUMO

Tissue factor pathway inhibitor-2 (TFPI-2), a serine protease inhibitor abundant in the extra cellular matrix, is highly expressed in non-invasive cells but undetectable levels in highly invasive human glioma cells. The mechanisms responsible for its transcriptional regulation are not well elucidated. In this study, we made several deletion constructs from a 3.6 kb genomic fragment from Hs683 cells containing the 5'-flanking region of the TFPI-2 gene, transiently transfected with these constructs into non-invasive (Hs683) and highly invasive (SNB19) human glioma cells, and assessed their expression by using a luciferase reporter gene. Three constructs showed high promoter activity (pTF5, -670 to +1; pTF6, -312 to +1; pTF2, -1511 to +1). Another construct, pTF8 (-81 to +1), showed no activity. PTF9, a variant of pTF5 in which a further 231 bp fragment (-312 to -81) was deleted, from the [-670 to +1] pTF5 region, also showed no promoter activity. Hence, (-312 to -81) this region is essential for the transcription of TFPI-2 in glioma cells. Sequencing of this promoter region revealed that it has a high G+C content, contains potential SP1 and AP1 binding motifs, and lacks canonical TATA and CAAT boxes immediately upstream of the major transcriptional initiation site, although CAAT boxes were found about -3000 bp upstream of the transcription start site. We also found a strong repressor in the region between -927 to -1181, upstream of the major transcriptional initiation site, followed by positive elements or enhancers between -1511 to -1181. These positive elements masked the silencer effect. Finally TFPI-2 was induced in Hs683 cells transfected with the pTF6 construct (-312 to +1) and stimulated with phorbol-12-myristate-13-acetate (PMA). We conclude that the -312 to +1 region is critical for the minimal and inducible regulation of TFPI-2 in non-invasive (Hs683) and highly invasive (SNB19) human glioma cell lines.


Assuntos
Neoplasias Encefálicas/patologia , Regulação Neoplásica da Expressão Gênica , Glioma/patologia , Glicoproteínas/biossíntese , Invasividade Neoplásica/genética , Proteínas de Neoplasias/biossíntese , Regiões Promotoras Genéticas/genética , Sequência de Bases , Sítios de Ligação , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Elementos Facilitadores Genéticos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes Reporter , Glioma/genética , Glioma/metabolismo , Glicoproteínas/genética , Glicoproteínas/fisiologia , Humanos , Luciferases/biossíntese , Luciferases/genética , Dados de Sequência Molecular , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiologia , Proteínas Recombinantes de Fusão/biossíntese , Deleção de Sequência , Acetato de Tetradecanoilforbol/farmacologia , Fatores de Transcrição/metabolismo , Transcrição Gênica/efeitos dos fármacos , Transfecção , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/metabolismo
13.
Biochim Biophys Acta ; 1621(3): 242-5, 2003 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-12787920

RESUMO

Tissue factor pathway inhibitor-2 (TFPI-2) is a Kunitz-type serine proteinase inhibitor associated with the extracellular matrices of vascular cells. A recent report provided in vitro evidence that TFPI-2 may be a novel inhibitor of the matrix metalloproteinases MMP-1, MMP-13, MMP-2 and MMP-9. In studies aimed at identifying the structural elements of TFPI-2 mediating the putative inhibition of the above MMPs, we re-examined the ability of native TFPI-2 to form complexes with MMP-2, MMP-9 and MMP-1, as well as assess its ability to inhibit the proteolytic activity of the interstitial collagenase, activated MMP-1. We report here that TFPI-2 failed to form complexes with MMP-2, MMP-9 and MMP-1 as revealed in immunoprecipitation and ligand blotting studies. In addition, TFPI-2 had no influence on the proteolytic activity of activated MMP-1 towards triple-helical collagen. These data provide presumptive evidence that TFPI-2 does not bind to MMP-2, MMP-9 and MMP-1, or regulate MMP-1, in the extracellular matrix.


Assuntos
Glicoproteínas/metabolismo , Metaloproteinase 1 da Matriz/metabolismo , Inibidores de Serina Proteinase/metabolismo , Animais , Células Cultivadas , Colágeno/metabolismo , Ativação Enzimática , Matriz Extracelular/metabolismo , Glicoproteínas/química , Humanos , Substâncias Macromoleculares , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Ligação Proteica , Inibidores de Serina Proteinase/química
14.
Thromb Haemost ; 94(6): 1122-30, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16411383

RESUMO

Tissue factor pathway inhibitor-2 (TFPI-2) is a 32 kDa matrix-associated Kunitz-type serine proteinase inhibitor consisting of a short amino-terminal region,three tandem Kunitz-type domains and a positively charged carboxy-terminal tail. Human TFPI-2, previously designated as placental protein 5, inhibits a broad spectrum of serine proteinases almost exclusively through its first Kunitz-type domain, and is thought to play an important role in the regulation of extracellular matrix digestion and remodeling. In this context, reduced synthesis of TFPI-2 has been related to numerous pathophysiological processes such as inflammation, angiogenesis, atherosclerosis, retinal degeneration and tumor growth/metastasis. In this review, we document current information regarding the expression of TFPI-2 by various tissues, its inhibitory activity and proteinase specificity in-vitro, and discuss possible physiological roles for this inhibitor based on in-vivo studies.


Assuntos
Glicoproteínas/metabolismo , Inibidores de Serina Proteinase/metabolismo , Sequência de Aminoácidos , Animais , Adesão Celular/fisiologia , Movimento Celular/fisiologia , Matriz Extracelular/metabolismo , Regulação da Expressão Gênica , Glicoproteínas/química , Glicoproteínas/genética , Humanos , Dados de Sequência Molecular , Conformação Proteica , Serina Endopeptidases/metabolismo , Inibidores de Serina Proteinase/química , Inibidores de Serina Proteinase/genética , Transdução de Sinais/fisiologia
15.
J Drug Target ; 23(3): 257-65, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25510254

RESUMO

We have developed a specific technique for imaging cancer in vivo using Cy5.5-labeled factor VIIa (fVIIa), clotting-deficient FFRck-fVIIa, paclitaxel-FFRck-fVIIa, and anti-tissue factor (TF) antibody. FVIIa is the natural ligand for TF. We took advantage of the fact that vascular endothelial cells (VECs) in cancer, but not normal tissue, aberrantly express TF due to its induction by vascular endothelial growth factor (VEGF). Under physiological conditions, TF is expressed by stromal cells and outer blood vessel layers (smooth muscle and adventitia), but not by VECs. We hypothesized that labeled fVIIa or anti-TF antibodies could be used to image the tumor vasculature in vivo. To test this, Cy5.5-labeled fVIIa, FFRck-fVIIa, paclitaxel-FFRck-fVIIa, and anti-TF antibody were developed and administered to athymic nude mice carrying xenografts including glioma U87EGFRviii, pancreatic cancer ASPC-1 and Mia PaCa-2, and squamous cell carcinoma KB-V1. Cy5.5 labeled with these targeting proteins specifically localized to the tumor xenografts for at least 14 days but unconjugated Cy5.5 did not localize to any xenografts or organs. This method of imaging TF in the tumor VECs may be useful in detecting primary tumors and metastases as well as monitoring in vivo therapeutic responses.


Assuntos
Carbocianinas/análise , Fator VIIa/análise , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Imagem Óptica/métodos , Tromboplastina/imunologia , Clorometilcetonas de Aminoácidos/química , Animais , Carbocianinas/química , Células Cultivadas , Fator VIIa/química , Xenoenxertos/imunologia , Humanos , Camundongos , Neoplasias/imunologia , Neoplasias/patologia , Paclitaxel/química
16.
J Histochem Cytochem ; 50(11): 1443-54, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12417609

RESUMO

Ovalbumin-like serine protease inhibitors are mainly localized intracellularly and their in vivo functions are largely unknown. To elucidate their physiological role(s), we studied the expression of one of these inhibitors, protease inhibitor 8 (PI-8), in normal human tissues by immunohistochemistry using a PI-8-specific monoclonal antibody. PI-8 was strongly expressed in the nuclei of squamous epithelium of mouth, pharynx, esophagus, and epidermis, and by the epithelial layer of skin appendages, particularly by more differentiated epithelial cells. PI-8 was also expressed by monocytes and by neuroendocrine cells in the pituitary gland, pancreas, and digestive tract. Monocytes showed nuclear and cytoplasmic localization of PI-8, whereas neuroendocrine cells showed only cytoplasmic staining. In vitro nuclear localization of PI-8 was confirmed by confocal analysis using serpin-transfected HeLa cells. Furthermore, mutation of the P(1) residue did not affect the subcellular distribution pattern of PI-8, indicating that its nuclear localization is independent of the interaction with its target protease. We conclude that PI-8 has a unique distribution pattern in human tissues compared to the distribution patterns of other intracellular serpins. Additional studies must be performed to elucidate its physiological role.


Assuntos
Serpinas/metabolismo , Animais , Anticorpos Monoclonais , Células COS , Núcleo Celular/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/ultraestrutura , Humanos , Imuno-Histoquímica , Líquido Intracelular/metabolismo , Monócitos/metabolismo , Sistemas Neurossecretores/citologia , Sistemas Neurossecretores/metabolismo , Especificidade de Órgãos , Serpinas/imunologia , Frações Subcelulares/metabolismo
17.
Thromb Haemost ; 90(1): 140-6, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12876637

RESUMO

The progression of neoplasms is frequently associated with thromboembolic complications. Coagulation proteins, particularly tissue factor (TF), have been shown to play a role in tumor growth and metastatic dissemination. TFPI is the principal inhibitor of TF-dependent pathway of blood coagulation, but previous studies failed to detect antigenic TFPI in cancer tissue. Recently, a second inhibitor of tissue factor dependent pathway, TFPI-2 (also known as placental protein 5 [PP5] or matrix-associated serine protease inhibitor [MSPI]), has been described. Information on the presence of TFPI-2 within the malignant tumor tissue still remains obscure, and thus the aim of this study was to evaluate the expression of TFPI-2 in loco in several different neoplasms. TFPI-2 expression was demonstrated by immunohistochemical procedures in neoplastic cells of laryngeal, breast, gastric, colon, pancreatic, renal and endometrial cancer, as well as glial neoplasms. The intensity of staining was not uniform, with higher intensity in more differentiated tumors. G3 breast, gastric, endometrial and colon cancer cells revealed populations of cells that were either TFPI-2 positive or negative. Gastric and renal cancer tissue exhibited the presence of TFPI-2 in tumor infiltrating macrophages. TFPI-2 was also observed in normal tissue of the breast, stomach, colon and pancreas. These data demonstrate that the expression of TFPI-2 diminishes with an increasing degree of malignancy, which may suggest a role for TFPI-2 in the maintenance of tumor stability and inhibition of the growth of neoplasms.


Assuntos
Glicoproteínas/análise , Proteínas de Neoplasias/análise , Neoplasias/química , Carcinoma/química , Carcinoma/patologia , Diferenciação Celular , Progressão da Doença , Feminino , Glioma/química , Glioma/patologia , Glicoproteínas/fisiologia , Humanos , Técnicas Imunoenzimáticas , Macrófagos/química , Masculino , Proteínas de Neoplasias/fisiologia , Neoplasias/patologia , Especificidade de Órgãos
18.
Thromb Haemost ; 88(1): 98-103, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12152685

RESUMO

A modified form of the activated whole blood clotting time was used to evaluate response of blood from hemophilia patients to factor VIIa. Repeated assays of individuals over a one-year period showed consistency for each individual and significant difference between individuals. Four hemophilia patients with inhibitors gave low response to factor VIIa in the assay and were characterized as low or moderate clinical responders to factor VIIa therapy. Another four patients with moderate to good clinical responsiveness to factor VIIa therapy showed a strong response to factor VIIa in the assay. A factor VIIa mutant with enhanced membrane affinity showed 10- to 13-fold higher activity than wild type factor VIIa. The results may justify larger studies to determine the utility of this assay for evaluation of patient responsiveness and to set factor VIIa therapy levels.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Fator VIIa/farmacologia , Hemofilia A/sangue , Hemofilia B/sangue , Mutação , Adolescente , Adulto , Coagulação Sanguínea/genética , Testes de Coagulação Sanguínea , Membrana Celular/metabolismo , Relação Dose-Resposta a Droga , Fator VIIa/genética , Fator VIIa/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
19.
Thromb Haemost ; 91(3): 569-75, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14983234

RESUMO

Tissue factor pathway inhibitor-2 (TFPI-2) is a recently described serine proteinase inhibitor. Human and murine TFPI-2 share about 50% homology. The aim of this study was to investigate the cellular localization of human and murine TFPI-2 in the liver and the regulation of their expression during acute inflammation. Northern blot, in situ hybridization and studies on isolated hepatocytes demonstrated a high-level expression of TFPI-2 in murine hepatocytes. On the other hand, very little TFPI-2 mRNA expression could be detected in human liver. Studies with isolated human liver cells suggested that TFPI-2 expression in human liver was mainly observed in liver sinusoidal endothelial cells rather than hepatocytes. Liver murine TFPI-2 expression was greatly increased after lipopolysaccharide administration with a delayed kinetics as compared to alpha1-acid glycoprotein, a classical acute-phase reactant. Accordingly, studies with isolated cells showed that the increase in TFPI-2 transcripts occurred in non-hepatocytic cells. Moreover, the LPS response was abolished in mice with a hepatocyte-specific KO for the gp130 receptor, thus indicating that a mediator from hepatocytes is involved in the up-regulation of TFPI-2 in non-parenchymal cells. In conclusion, murine TFPI-2 is highly expressed in hepatocytes in the normal murine liver and is upregulated in non-parenchymal cells in the context of inflammation. The large difference in the level of liver expression of human and murine TFPI-2 suggests that despite significant sequence similarities, these proteins presumably have different functions in the two species.


Assuntos
Glicoproteínas/biossíntese , Inflamação/metabolismo , Fígado/metabolismo , Reação de Fase Aguda , Animais , Antígenos CD/metabolismo , Northern Blotting , Receptor gp130 de Citocina , Endopeptidases/metabolismo , Hepatócitos/metabolismo , Humanos , Hibridização In Situ , Interleucina-6/metabolismo , Cinética , Lipopolissacarídeos/metabolismo , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Especificidade da Espécie , Fatores de Tempo , Regulação para Cima
20.
Int J Oncol ; 22(6): 1277-83, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12738994

RESUMO

Tissue factor pathway inhibitor-2 (TFPI-2), a serine protease inhibitor abundant in the extracellular matrix, is expressed in high amounts in low-grade, non-invasive glioma cells but in low amounts in high-grade, highly invasive glioma cells. Overexpression of TFPI-2 by highly invasive glioma cells reduces their invasiveness and thus may be useful in cancer therapy. The mechanisms underlying the transcriptional regulation of TFPI-2 are not well elucidated. We previously reported that the -312 to +1 region of TFPI-2 was critical for the minimal, inducible regulation of TFPI-2 in gliomas. This region harbors sites for several transcription factors, including SP1 (-192 to -183 and -135 to -128), AP-1 (-310 to -300, -213 to -204, and -163 to -154), NF-kappaB (-229 to -221), an NF-kappaB-like site (-291 to -281), and Lyf-1 (-260 to -252). Here we transiently transfected low-grade Hs683 glioma cells with mutant constructs to clarify the role of these transcription factors in TFPI-2 regulation. Addition of phorbol 12-myristate 13-acetate, 1,2-diacyl-sn-glycerol, IFN-gamma, or IFN-alpha induced the expression of TFPI-2 wild-type promoter construct as well as TFPI-2 protein and mRNA in Hs683 cells. Mutations at either of two AP-1 sites (-310 to -300 and -163 to -154) or either of two SP1 sites (-192 to -183 and -135 to -128) resulted in reduced TFPI-2 activity, regardless of the presence of stimulator compounds, and reduction in DNA-protein binding (by electrophoretic mobility shift assay).


Assuntos
Regulação da Expressão Gênica/genética , Glicoproteínas/genética , Sequência de Bases , Ciclo Celular/efeitos dos fármacos , Primers do DNA , Regulação da Expressão Gênica/efeitos dos fármacos , Glioma , Glicoproteínas/metabolismo , Humanos , Interferon-alfa/farmacologia , Interferon gama/farmacologia , Luciferases/genética , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas/efeitos dos fármacos , RNA Mensageiro/genética , Proteínas Recombinantes/biossíntese , Acetato de Tetradecanoilforbol/farmacologia , Fatores de Transcrição/metabolismo , Transcrição Gênica/efeitos dos fármacos , Transfecção , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA