Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 511
Filtrar
1.
Oncol Rep ; 48(2)2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35775375

RESUMO

The clinical introduction of molecular imaging for the management of oropharyngeal squamous cell carcinoma (OPSCC) relies on the identification of relevant cancer­specific biomarkers. The application of three membrane­bound receptors, namely urokinase­type plasminogen activator receptor (uPAR), tissue factor (TF) and EGFR have been previously explored for targeted imaging and therapeutic strategies in a broad range of solid cancers. The present study aimed to investigate the expression patterns of uPAR, EGFR and TF by immunohistochemistry (IHC) to evaluate their potential for targeted imaging and prognostic value in OPSCC. In a retrospective cohort of 93 patients with primary OPSCC, who were balanced into the 45 human papillomavirus (HPV)­positive and 48 HPV­negative groups, the IHC­determined expression profiles of uPAR, TF and EGFR in large biopsy or tumor resection specimens were analyzed. Using the follow­up data, overall survival (OS) and recurrence­free survival were measured. Specifically, associations between survival outcome, biomarker expression and clinicopathological factors were examined using Cox proportional hazards model and log­rank test following Kaplan­Meier statistics. After comparing the expression pattern of biomarkers within the tumor compartment with that in the adjacent normal tissues, uPAR and TF exhibited a highly tumor­specific expression pattern, whereas EGFR showed a homogeneous expression within the tumor compartment as well as a consistent expression in the normal mucosal epithelium and salivary gland tissues. The positive expression rate of uPAR, TF and EGFR in the tumors was 98.9, 76.3 and 98.9%, respectively. No statistically significant association between biomarker expression and survival outcome could be detected. Higher uPAR expression levels had a trend towards reduced OS according to results from univariate analysis (P=0.07; hazard ratio=2.01; 95% CI=0.92­4.37). Taken together, these results suggest that uPAR, TF and EGFR may be suitable targets for molecular imaging and therapy in OPSCC. In particular, uPAR may be an attractive target owing to their high positive expression rates in tumors and a highly tumor­specific expression pattern.


Assuntos
Neoplasias Orofaríngeas , Infecções por Papillomavirus , Carcinoma de Células Escamosas de Cabeça e Pescoço , Biomarcadores Tumorais/biossíntese , Receptores ErbB/biossíntese , Humanos , Imagem Molecular , Terapia de Alvo Molecular , Neoplasias Orofaríngeas/diagnóstico por imagem , Neoplasias Orofaríngeas/tratamento farmacológico , Neoplasias Orofaríngeas/patologia , Neoplasias Orofaríngeas/virologia , Papillomaviridae , Infecções por Papillomavirus/diagnóstico por imagem , Infecções por Papillomavirus/metabolismo , Infecções por Papillomavirus/patologia , Prognóstico , Receptores de Ativador de Plasminogênio Tipo Uroquinase/biossíntese , Estudos Retrospectivos , Carcinoma de Células Escamosas de Cabeça e Pescoço/diagnóstico por imagem , Carcinoma de Células Escamosas de Cabeça e Pescoço/tratamento farmacológico , Tromboplastina/biossíntese
2.
Transfus Apher Sci ; 60(6): 103237, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34419356

RESUMO

SARS-CoV-2 attaches to the angiotensin-converting enzyme 2 (ACE-2) receptor on human cells. The virus causes hypercytokinemia, capillary leak, pulmonary edema, acute respiratory distress syndrome, acute cardiac injury, and leads to death. Mesenchymal stem cells (MSCs) are ACE-2 negative cells; therefore, can escape from SARS-CoV-2. MSCs prevent hypercytokinemia and help the resolution of the pulmonary edema and other damages occurred during the course of COVID-19. In addition, MSCs enhance the regeneration of the lung and other tissues affected by SARS-CoV-2. The case series reported beneficial effect of MSCs in COVID-19 treatment. However, there are some concerns about the safety of MSCs, particularly referring to the increased risk of disseminated intravascular coagulation, and thromboembolism due to the expression of TF/CD142. Prospective, randomized, large scale studies are needed to reveal the optimum dose, administration way, time, efficacy, and safety of MSCs in the COVID-19 treatment.


Assuntos
COVID-19 , Pulmão/fisiologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Regeneração , SARS-CoV-2/metabolismo , COVID-19/sangue , COVID-19/epidemiologia , COVID-19/terapia , Coagulação Intravascular Disseminada/sangue , Coagulação Intravascular Disseminada/etiologia , Humanos , Peptidil Dipeptidase A/metabolismo , Estudos Prospectivos , Fatores de Risco , Tromboembolia/sangue , Tromboembolia/etiologia , Tromboplastina/biossíntese
3.
J Cancer Res Clin Oncol ; 146(2): 467-475, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31734835

RESUMO

PURPOSE: The expression of active tissue factor (TF) on the surface of microvesicles (MVs) is essential for the activation of the coagulation system and transduction of the signaling pathways in cancer cells. In its use as a biomarker for cancer-associated venous thromboembolism (VTE), TF has shown high expression variability. As a contribution to this discussion, we present a study investigating plasma samples from patients with various progressive tumors at high risk for VTE. METHODS: Based on our previous study uncovering microvesicles (MVs), the larger ectosome-like extracellular vesicles (EV), as the major source of TF activity in EV preparations, we now determined TF activity on enriched MVs isolated from plasma of cancer patients and compared it with that on MVs from healthy individuals. RESULTS: We found considerably higher amounts of MVs as well as higher levels of MV-bound TF activities in the plasma of cancer patients. We also show that preparations from plasma of cancer patients have the potency to induce ERK phosphorylation in a human tumor cell line through proteinase-activated receptor two (PAR2) activation. CONCLUSION: We suggest that MVs instead of whole EV preparations, and TF activity rather than its antigenic quantification should be used in clinical studies for identifying patients with progressive tumors at high risk for VTE.


Assuntos
Tromboplastina/metabolismo , Idoso , Estudos de Casos e Controles , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/patologia , Feminino , Humanos , Sistema de Sinalização das MAP Quinases , Masculino , Neoplasias Pancreáticas/sangue , Neoplasias Pancreáticas/patologia , Fosforilação , Tromboplastina/biossíntese , Tromboembolia Venosa/sangue , Tromboembolia Venosa/patologia
4.
J Thromb Thrombolysis ; 49(2): 228-234, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31612355

RESUMO

Antiphospholipid antibodies (aPL) are heterogeneous and there is evidence that binding specificity determines which cellular effects they can trigger. We have therefore hypothesised that the induction of tissue factor (TF) in monocytes and endothelial cells by aPL depends on their binding specificity. To further investigate this, we have analyzed the ability of three human monoclonal aPL with distinctly different binding specificities to induce transcription and cell surface expression of TF in monocytes and endothelial cells. Results with human monoclonal aPL were validated with IgG-fractions obtained from patients with APS. We confirmed previous results that a lipid reactive human monoclonal aPL rapidly induced TF transcription and cell surface expression in monocytes and endothelial cells. A monoclonal aPL reactive against ß2 glycoprotein I (ß2GPI) induced TF with a delayed time course. This was fully dependent on the induction of tumor necrosis factor alpha (TNFα) secretion as capture of TNFα by adalimumab prevented TF induction. This pattern was confirmed with patient IgG fractions. Both lipid reactive and anti-ß2GPI induced TF transcription. Unexpectedly, this activity of anti-ß2GPI was mediated fully by TNFα which was secreted in response to incubation with anti-ß2GPI. The role of TNFα in mediating TF induction by anti-ß2GPI may have wider implications for APS pathogenesis.


Assuntos
Anticorpos Monoclonais/farmacologia , Tromboplastina/biossíntese , Fator de Necrose Tumoral alfa/biossíntese , beta 2-Glicoproteína I/farmacologia , Expressão Gênica , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Tromboplastina/genética , Fator de Necrose Tumoral alfa/genética
5.
Nano Lett ; 19(7): 4721-4730, 2019 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-31180684

RESUMO

Within tumors, the coagulation-inducing protein tissue factor (TF), a major initiator of blood coagulation, has been shown to play a critical role in the hematogenous metastasis of tumors, due to its effects on tumor hypercoagulability and on the mediation of interactions between platelets and tumor cells. Targeting tumor-associated TF has therefore great therapeutic potential for antimetastasis therapy and preventing thrombotic complication in cancer patients. Herein, we reported a novel peptide-based nanoparticle that targets delivery and release of small interfering RNA (siRNA) into the tumor site to silence the expression of tumor-associated TF. We showed that suppression of TF expression in tumor cells blocks platelet adhesion surrounding tumor cells in vitro. The downregulation of TF expression in intravenously administered tumor cells (i.e., simulated circulating tumor cells [CTCs]) prevented platelet adhesion around CTCs and decreased CTCs survival in the lung. In a breast cancer mouse model, siRNA-containing nanoparticles efficiently attenuated TF expression in the tumor microenvironment and remarkably reduced the amount of lung metastases in both an experimental lung metastasis model and tumor-bearing mice. What's more, this strategy reversed the hypercoagulable state of the tumor bearing mice by decreasing the generation of thrombin-antithrombin complexes (TAT) and activated platelets, both of which are downstream products of TF. Our study describes a promising approach to combat metastasis and prevent cancer-associated thrombosis, which advances TF as a therapeutic target toward clinic applications.


Assuntos
Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inativação Gênica , Neoplasias Pulmonares , Nanopartículas , Proteínas de Neoplasias , Neoplasias Experimentais , RNA Interferente Pequeno , Trombofilia , Tromboplastina , Trombose , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Camundongos Nus , Nanopartículas/química , Nanopartículas/uso terapêutico , Metástase Neoplásica , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Células Neoplásicas Circulantes/metabolismo , Células Neoplásicas Circulantes/patologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Trombofilia/genética , Trombofilia/metabolismo , Trombofilia/prevenção & controle , Tromboplastina/biossíntese , Tromboplastina/genética , Trombose/genética , Trombose/metabolismo , Trombose/patologia , Trombose/prevenção & controle
6.
Arterioscler Thromb Vasc Biol ; 39(3): 402-412, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30602303

RESUMO

Objective- Mutations in Krüppel like factor-11 ( KLF11), a gene also known as maturity-onset diabetes mellitus of the young type 7, contribute to the development of diabetes mellitus. KLF11 has anti-inflammatory effects in endothelial cells and beneficial effects on stroke. However, the function of KLF11 in the cardiovascular system is not fully unraveled. In this study, we investigated the role of KLF11 in vascular smooth muscle cell biology and arterial thrombosis. Approach and Results- Using a ferric chloride-induced thrombosis model, we found that the occlusion time was significantly reduced in conventional Klf11 knockout mice, whereas bone marrow transplantation could not rescue this phenotype, suggesting that vascular KLF11 is critical for inhibition of arterial thrombosis. We further demonstrated that vascular smooth muscle cell-specific Klf11 knockout mice also exhibited significantly reduced occlusion time. The expression of tissue factor (encoded by the F3 gene), a main initiator of the coagulation cascade, was increased in the artery of Klf11 knockout mice, as determined by real-time quantitative polymerase chain reaction and immunofluorescence. Furthermore, vascular smooth muscle cells isolated from Klf11 knockout mouse aortas showed increased tissue factor expression, which was rescued by KLF11 overexpression. In human aortic smooth muscle cells, small interfering RNA-mediated knockdown of KLF11 increased tissue factor expression. Consistent results were observed on adenovirus-mediated overexpression of KLF11. Mechanistically, KLF11 downregulates F3 at the transcriptional level as determined by reporter and chromatin immunoprecipitation assays. Conclusions- Our data demonstrate that KLF11 is a novel transcriptional suppressor of F3 in vascular smooth muscle cells, constituting a potential molecular target for inhibition of arterial thrombosis.


Assuntos
Proteínas Reguladoras de Apoptose/fisiologia , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Proteínas Repressoras/fisiologia , Tromboplastina/biossíntese , Trombose/prevenção & controle , Animais , Antitrombina III/análise , Proteínas Reguladoras de Apoptose/antagonistas & inibidores , Proteínas Reguladoras de Apoptose/deficiência , Proteínas Reguladoras de Apoptose/genética , Transplante de Medula Óssea , Células Cultivadas , Cloretos/toxicidade , Imunoprecipitação da Cromatina , Regulação para Baixo , Feminino , Compostos Férricos/toxicidade , Regulação da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peptídeo Hidrolases/análise , Agregação Plaquetária , Interferência de RNA , Proteínas Recombinantes/metabolismo , Proteínas Repressoras/antagonistas & inibidores , Proteínas Repressoras/deficiência , Proteínas Repressoras/genética , Tromboplastina/genética , Trombose/induzido quimicamente , Transcrição Gênica
7.
J Pak Med Assoc ; 68(11): 1644-11649, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30410143

RESUMO

OBJECTIVE: To investigate if lectin-like oxidised low density lipoprotein receptor is implicated in oxidised low density lipoprotein induced up regulation of tissue factor and whether recombinant domain V of beta (2)-Glycoprotein I expressed in Pichia pastoris inhibits the binding of oxidised and lectin-like low density lipoprotein. METHODS: The expression of tissue factor and lectin-like oxidised low density lipoprotein receptor was detected using Western blot methods. Small interference ribonucleic acid of lectin-like oxidised low density lipoprotein receptor was used to block lectin-like oxidised low density lipoprotein receptor expression. Flow cytometry was used to test the effect of beta (2)-Glycoprotein I expressed in Pichia pastoris on the binding of oxidised low density lipoprotein with lectin-like oxidised low density lipoprotein receptor by using the lectin-like oxidised low density lipoprotein receptor-expressing 293T cells. RESULTS: Oxidised low density lipoprotein at 5-10 g/mL increased tissue factor and lectin-like oxidised low density lipoprotein receptor expression, whereas 20-50 g/mL oxidised low density lipoprotein attenuated tissue factor expression. Inhibiting lectin-like oxidised low density lipoprotein receptor expression by small interference ribonucleic acid of lectin-like oxidised low density lipoprotein receptor impaired oxidised low density lipoprotein-induced tissue factor over expression in macrophages. Pretreatment with beta (2)-Glycoprotein I expressed in Pichia pastoris led to a strong inhibition of tissue factor and lectin-like oxidised low density lipoprotein receptor expression in a dose-dependent manner in macrophages. Flow cytometry analysis showed that beta (2)-Glycoprotein I expressed in Pichia pastoris attenuated the interaction of oxidised low density lipoprotein with lectin-like oxidised low density lipoprotein receptor in lectin-like oxidised low density lipoprotein receptor-expressing 293T cells. CONCLUSIONS: Lectin-like oxidised low density lipoprotein receptor was implicated in the expression of tissue factor induced by oxidised low density lipoprotein, and beta (2)-Glycoprotein I expressed in Pichia pastoris inhibited oxidised low density lipoprotein-induced tissue factor and lectin-like oxidised low density lipoprotein receptor expression, at least in part, via inhibition of the interaction between oxidised low density lipoprotein and lectin-like oxidised low density lipoprotein receptor.


Assuntos
Regulação da Expressão Gênica , Lipoproteínas LDL/metabolismo , Macrófagos/metabolismo , Tromboplastina/biossíntese , beta 2-Glicoproteína I/metabolismo , Animais , Western Blotting , Camundongos , Oxirredução , Coelhos
8.
J Thromb Haemost ; 16(9): 1800-1813, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29971917

RESUMO

Essentials Tumor-bearing mice were employed to follow oncogenic HRAS sequences in plasma, and blood cells. Cancer DNA accumulated in leukocytes above levels detected in exosomes, platelets and plasma. Extracellular vesicles and nucleosomes are required for uptake of tumor DNA by leukocytes. Uptake of tumor-derived extracellular vesicles by leukocytes triggers coagulant phenotype. SUMMARY: Background Tumor-derived extracellular vesicles (EVs) and free nucleosomes (NSs) carry into the circulation a wealth of cancer-specific, bioactive and poorly understood molecular cargoes, including genomic DNA (gDNA). Objective Here we investigated the distribution of extracellular oncogenic gDNA sequences (HRAS and HER2) in the circulation of tumor-bearing mice. Methods and Results Surprisingly, circulating leukocytes (WBCs), especially neutrophils, contained the highest levels of mutant gDNA, which exceeded the amount of this material recovered from soluble fractions of plasma, circulating EVs, platelets, red blood cells (RBCs) and peripheral organs, as quantified by digital droplet PCR (ddPCR). Tumor excision resulted in disappearance of the WBC-associated gDNA signal within 2-9 days, which is in line with the expected half-life of these cells. EVs and nucleosomes were essential for the uptake of tumor-derived extracellular DNA by neutrophil-like cells and impacted their phenotype. Indeed, the exposure of granulocytic HL-60 cells to EVs from HRAS-driven cancer cells resulted in a selective increase in tissue factor (TF) procoagulant activity and interleukin 8 (IL-8) production. The levels of circulating thrombin-antithrombin complexes (TAT) were markedly elevated in mice harboring HRAS-driven xenografts. Conclusions Myeloid cells may represent a hitherto unrecognized reservoir of cancer-derived, EV/NS-associated oncogenic gDNA in the circulation, and a possible novel platform for liquid biopsy in cancer. In addition, uptake of this material alters the phenotype of myeloid cells, induces procoagulant and proinflammatory activity and may contribute to systemic effects associated with cancer.


Assuntos
DNA de Neoplasias/sangue , Vesículas Extracelulares/química , Genes erbB-2 , Genes ras , Células Mieloides/química , Neutrófilos/química , Animais , Antitrombina III , Plaquetas/química , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Sobrevivência Celular , Transformação Celular Neoplásica , DNA de Neoplasias/farmacocinética , Exossomos/química , Feminino , Células HL-60 , Xenoenxertos , Humanos , Interleucina-8/biossíntese , Camundongos , Camundongos SCID , Células Mieloides/metabolismo , Transplante de Neoplasias , Neutrófilos/metabolismo , Nucleossomos/química , Peptídeo Hidrolases/sangue , Plasma/química , Ratos , Células THP-1 , Tromboplastina/biossíntese , Carga Tumoral
9.
J Cell Mol Med ; 22(9): 4545-4549, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29893509

RESUMO

Recently, platelet-derived growth factors present in lysates became an extreme interest in the field of regenerative medicine such as in wound healing and as substitutes to foetal bovine serum in xeno-free cell culture systems. However, the generation of such platelet lysates completely depends on the availability of platelet donors. In this study, the possibility to use in vitro-generated megakaryocytes derived from induced pluripotent stem cells (iPSCs) as a cell source for typical platelet growth factors was investigated. Therefore, the presence and levels of those factors were characterized in in vitro-produced megakaryocytes. In comparison with platelets, in vitro-generated megakaryocytes showed a multifold increased content in transcript and protein levels of typical platelet growth factors including platelet-derived growth factors (PDGFs), transforming growth factor (TGF)-1ß, vascular endothelial cell factor (VEGF)-A, epidermal growth factor (EGF), insulin-like growth factor (IGF)-1 and tissue factor (TF). Hence, iPSC-derived megakaryocytes may serve as an efficient cell source for a donor-independent generation of growth factor-rich lysates with a broad application potential in innovative cell culture systems and regenerative therapies.


Assuntos
Expressão Gênica , Células-Tronco Pluripotentes Induzidas/citologia , Megacariócitos/citologia , Plaquetas/citologia , Plaquetas/metabolismo , Diferenciação Celular , Extratos Celulares/química , Proliferação de Células , Fator de Crescimento Epidérmico/biossíntese , Fator de Crescimento Epidérmico/genética , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Fator de Crescimento Insulin-Like I/biossíntese , Fator de Crescimento Insulin-Like I/genética , Megacariócitos/metabolismo , Fator de Crescimento Derivado de Plaquetas/biossíntese , Fator de Crescimento Derivado de Plaquetas/genética , Cultura Primária de Células , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Medicina Regenerativa/métodos , Tromboplastina/biossíntese , Tromboplastina/genética , Fator de Crescimento Transformador beta1/biossíntese , Fator de Crescimento Transformador beta1/genética , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/genética
10.
Cancer Biol Ther ; 19(5): 416-426, 2018 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-29333924

RESUMO

Glomeruloid vascular proliferation (GVP) is a diagnostic hallmark and links to aggressive behavior, therapy resistance and poor prognosis in glioblastoma (GBM). It lacks clinical approaches to predict and monitor its formation and dynamic change. Yet the mechanism of GVPs also remains largely unknown. Using an in situ GBM xenograft mouse model, combined clinical MRI images of pre-surgery tumor and pathological investigation, we demonstrated that the inhibition of tissue factor (TF) decreased GVPs in Mouse GBM xenograft model. TF shRNA reduced microvascular area and diameter, other than bevacizumab. TF dominantly functions via PAR2/HB-EGF-dependent activation under hypoxia in endothelial cells (ECs), resulting in a reduction of GVPs and cancer cells invasion. TF expression strongly correlated to GVPs and microvascular area (MVA) in GBM specimens from 56 patients, which could be quantitatively evaluated in an advanced MRI images system in 33 GBM patients. This study presented an approach to assess GVPs that could be served as a MRI imaging biomarker in GBM and uncovered a molecular mechanism of GVPs.


Assuntos
Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/diagnóstico por imagem , Glioblastoma/irrigação sanguínea , Glioblastoma/diagnóstico por imagem , Imageamento por Ressonância Magnética/métodos , Tromboplastina/biossíntese , Adulto , Idoso , Animais , Biomarcadores Tumorais/biossíntese , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Glioblastoma/metabolismo , Xenoenxertos , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Neovascularização Patológica/diagnóstico por imagem , Neovascularização Patológica/metabolismo , Transdução de Sinais , Adulto Jovem
11.
Clin Respir J ; 12(3): 1125-1133, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28419722

RESUMO

BACKGROUND: To examine the expression of D-dimer, fibrinogen (FIB), leukocyte, C-reactive protein (CRP) and tissue factor (TF) released from monocyte in non-small cell lung cancer (NSCLC) patients with or without venous thromboembolism (VTE) and analyse the correlation, to explore the possible mechanisms. METHODS: Seventy-two patients confirmed the diagnosis of lung cancer, among whom 10 with VTE were enrolled into the study from November 2012 to January 2014 in the First Affiliated Hospital of Fujian Medical University and 30 healthy subjects were also enrolled as the control group. Ficoll and Percoll density gradient centrifugation separated of peripheral blood monocyte. Monocyte TF mRNA expression was detected using reverse transcriptase-polymerase chain reaction (RT-PCR). RESULTS: There were significant differences in different stages of the cancer (P < .05) and no significance among the histopathologic types (P > .05) for the expression of monocyte TF mRNA in NSCLC patients, its expression was significantly higher in cancer with lymph node metastasis than those without lymph node metastasis (P < .01). Meanwhile, in NSCLC patients with VTE, the expression of monocyte TF mRNA was significantly higher than that in patients without VTE (P < .01). Difference of the survival curves between the low monocyte TF mRNA expression and the high monocyte TF mRNA expression was significant (Log-rank x2 = 4.923, P < .05). CONCLUSIONS: Monocyte TF may be a relevant source of TF-mediated thrombogenicity in NSCLC patients and may be associated with prognosis in NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/genética , Estadiamento de Neoplasias , RNA Neoplásico/genética , Tromboplastina/genética , Adulto , Idoso , Biópsia , Broncoscopia , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/secundário , Feminino , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Metástase Linfática , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Monócitos/metabolismo , Prognóstico , Estudos Retrospectivos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Toracoscopia , Tromboplastina/biossíntese , Tomografia Computadorizada por Raios X
12.
Int J Oncol ; 51(6): 1747-1756, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29075790

RESUMO

Most cancer cells trigger thrombin generation (TG) to various extent. In the present study, we dissected the mechanisms responsible for the procoagulant activity of pancreatic adenocarcinoma cells (BXPC3), a highly thrombogenic cancer type, and breast cancer cells (MCF7), a less thombogenic tumor type. TG of normal plasma was assessed by the Thrombinoscope (CAT®) in the presence or absence of cancer cells. TG was also assessed in plasma depleted of clotting factors, in plasma spiked with tissue factor (TF) and/or procoagulant phospholipids, in plasma spiked with an anti-TF monoclonal antibody or with corn trypsin inhibitor (CTI). The presence of alternatively spliced TF (asTF), TF activity (TFa) and cancer procoagulant (CP) levels were determined. TFa and asTF were highly expressed by BXPC3 cells, compared to MCF7 cells, while CP levels were higher in MCF7 cells. BXPC3 cells had a stronger effect on TG than MCF7 cells. Accordingly, anti-TF had more inhibitory activity on TG triggered by BXPC3 cells while CTI had more pronounced inhibitory effect on TG triggered by MCF7 cells. TG enhancement by both BXPC3 and MCF7 cells was mediated by FVII and intrinsic tenase while FXII and FXI were also important for MCF7 cells. The induction of TG by BXPC3 cells was mainly driven by the TF pathway while TG generation triggered by MCF7 cells was also driven by FXII activation. Therefore, hypercoagulability results from a combination of the inherent procoagulant properties of cancer cell-associated TF as well as of procoagulant phospholipids in the plasma microenvironment.


Assuntos
Neoplasias da Mama/metabolismo , Fator XII/metabolismo , Neoplasias Pancreáticas/metabolismo , Trombina/biossíntese , Tromboplastina/metabolismo , Anticorpos Monoclonais/farmacologia , Linhagem Celular Tumoral , Neoplasias do Colo/metabolismo , Cisteína Endopeptidases/metabolismo , Feminino , Células HCT116 , Células HT29 , Células Endoteliais da Veia Umbilical Humana , Humanos , Células MCF-7 , Proteínas de Neoplasias/metabolismo , Neoplasias Ovarianas/metabolismo , Plasma Rico em Plaquetas , Tromboplastina/biossíntese , Tromboplastina/imunologia
13.
Gynecol Oncol ; 146(1): 146-152, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28501328

RESUMO

OBJECTIVE: Ovarian clear cell carcinoma (OCCC) and high grade serous ovarian cancer (HGSOC) are associated with the highest risk of VTE among patients with epithelial ovarian cancer (EOC). Tissue factor (TF) is a transmembrane glycoprotein which can trigger thrombosis. We sought to evaluate if there is an association between VTE and tumor expression of tissue factor (TF), plasma TF, and microvesicle TF (MV TF) activity in this high-risk population. METHODS: We performed a case-control study of OCCC and HGSOC patients with and without VTE. 105 patients who underwent surgery at a tertiary care center between January 1995 and October 2013 were included. Plasma TF was measured with an enzyme-linked immunosorbent assay. A TF-dependent Factor Xa generation assay was used to measure MV TF activity. Immunohistochemical (IHC) analysis was performed to evaluate tumor expression of TF. RESULTS: 35 women with OCCC or HGSOC diagnosed with VTE within 9months of surgery were included in the case group. Those with VTE had a worse OS, p<0.0001, with a greater than three-fold increase in risk of death, HR 3.33 (CI 1.75-6.35). There was no significant difference in median plasma TF level or MV TF activity level between patients with and without VTE. OCCC patients had greater expression of TF in their tumors than patients with HGSOC, p<0.0001. CONCLUSIONS: TFMV activity and plasma TF level were not predictive of VTE in this patient population. Given the extensive expression of TF in OCCC tumors, it is unlikely IHC expression will be useful in risk stratification for VTE in this population.


Assuntos
Neoplasias Epiteliais e Glandulares/sangue , Neoplasias Epiteliais e Glandulares/patologia , Neoplasias Ovarianas/sangue , Neoplasias Ovarianas/patologia , Tromboplastina/metabolismo , Trombose Venosa/sangue , Trombose Venosa/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma Epitelial do Ovário , Estudos de Casos e Controles , Técnica de Imunoensaio Enzimático de Multiplicação , Feminino , Humanos , Imuno-Histoquímica , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Análise de Sobrevida , Tromboplastina/biossíntese
14.
Int J Gynecol Cancer ; 27(1): 37-43, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27755234

RESUMO

OBJECTIVES: Our 2007 study of 32 patients with ovarian cancer reported the possible involvement of tissue factor (TF) in the development of venous thromboembolism (VTE) before treatment, especially in clear cell carcinoma (CCC). This follow-up study further investigated this possibility in a larger cohort. METHODS: We investigated the intensity of TF expression (ITFE) and other variables for associations with VTE using univariate and multivariate analyses in 128 patients with epithelial ovarian cancer initially treated between November 2004 and December 2010, none of whom had received neoadjuvant chemotherapy. Before starting treatment, all patients were ultrasonographically screened for VTE. The ITFE was graded based on immunostaining of surgical specimens. RESULTS: Histological types were serous carcinoma (n = 42), CCC (n = 12), endometrioid carcinoma (n = 15), mucinous carcinoma (n = 53), and undifferentiated carcinoma (n = 6). The prevalence of VTE was significantly higher in CCC (34%) than in non-CCC (17%, P = 0.03). As ITFE increased, the frequencies of CCC and VTE increased significantly (P < 0.001 and P = 0.014, respectively). Multivariate analysis identified TF expression and pretreatment dimerized plasmin fragment D level as significant independent risk factors for VTE development. These factors showed particularly strong impacts on advanced-stage disease (P = 0.021). CONCLUSIONS: The 2007 cohort was small, preventing multivariate analysis. This study of a larger cohort yielded stronger evidence that the development of VTE in epithelial ovarian cancer may involve TF expression in cancer tissues.


Assuntos
Neoplasias Epiteliais e Glandulares/metabolismo , Neoplasias Ovarianas/metabolismo , Tromboplastina/biossíntese , Tromboembolia Venosa/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Análise de Variância , Biomarcadores Tumorais/biossíntese , Carcinoma Epitelial do Ovário , Estudos de Coortes , Feminino , Humanos , Pessoa de Meia-Idade , Neoplasias Epiteliais e Glandulares/diagnóstico por imagem , Neoplasias Epiteliais e Glandulares/patologia , Neoplasias Ovarianas/diagnóstico por imagem , Neoplasias Ovarianas/patologia , Tromboembolia Venosa/diagnóstico por imagem , Tromboembolia Venosa/patologia
15.
Crit Rev Oncog ; 22(3-4): 219-248, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29604900

RESUMO

It is widely recognized that a strong correlation exists between cancer and aberrant hemostasis. Patients with various types of cancers often develop thrombosis, a phenomenon commonly referred to as Trousseau syndrome. Tissue factor (TF) is expressed by tumor cells and contributes to a variety of pathologic processes, such as thrombosis, tumor growth, tumor angiogenesis, and metastasis. Tissue factor is expressed in two naturally occurring protein isoforms: membrane-bound full-length TF (flTF) and soluble alternatively spliced TF (asTF). Tissue factor is the primary initiator of blood coagulation, and it triggers intracellular signaling through protease-activated receptors (PARs). PARs are activated either by TF/FVIIa complexes or by thrombin generated following coagulation activation. Furthermore, the noncoagulant asTF retains an integrin-binding site and stimulates angiogenesis by ligating endothelial integrins αvß3 and α6ß1. Lastly, the increased TF expression in tumors is associated with the release in blood of TF-positive procoagulant microparticles that favor thromboembolic complications. Therefore, the interruption of asTF and flTF signaling represents a potential antiangiogenic strategy. In this review, we summarize the current knowledge on the role of TF in cancer, and we explore therapeutic perspectives based on TF targeting.


Assuntos
Neoplasias/metabolismo , Neovascularização Patológica/metabolismo , Trombofilia/metabolismo , Tromboplastina/biossíntese , Carga Tumoral/fisiologia , Neoplasias Vasculares/metabolismo , Animais , Biomarcadores Tumorais/biossíntese , Previsões , Humanos , Neoplasias/patologia , Neovascularização Patológica/patologia , Trombofilia/patologia , Neoplasias Vasculares/patologia
16.
Cancer Res ; 76(14): 4270-82, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27221703

RESUMO

Epithelial-mesenchymal transition (EMT) is prominent in circulating tumor cells (CTC), but how it influences metastatic spread in this setting is obscure. Insofar as blood provides a specific microenvironment for tumor cells, we explored a potential link between EMT and coagulation that may provide EMT-positive CTCs with enhanced colonizing properties. Here we report that EMT induces tissue factor (TF), a major cell-associated initiator of coagulation and related procoagulant properties in the blood. TF blockade by antibody or shRNA diminished the procoagulant activity of EMT-positive cells, confirming a functional role for TF in these processes. Silencing the EMT transcription factor ZEB1 inhibited both EMT-associated TF expression and coagulant activity, further strengthening the link between EMT and coagulation. Accordingly, EMT-positive cells exhibited a higher persistance/survival in the lungs of mice colonized after intravenous injection, a feature diminished by TF or ZEB1 silencing. In tumor cells with limited metastatic capability, enforcing expression of the EMT transcription factor Snail increased TF, coagulant properties, and early metastasis. Clinically, we identified a subpopulation of CTC expressing vimentin and TF in the blood of metastatic breast cancer patients consistent with our observations. Overall, our findings define a novel EMT-TF regulatory axis that triggers local activation of coagulation pathways to support metastatic colonization of EMT-positive CTCs. Cancer Res; 76(14); 4270-82. ©2016 AACR.


Assuntos
Coagulação Sanguínea , Transição Epitelial-Mesenquimal , Células Neoplásicas Circulantes/patologia , Tromboplastina/biossíntese , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Endogâmicos BALB C , Homeobox 1 de Ligação a E-box em Dedo de Zinco/fisiologia
17.
Sci Rep ; 6: 25171, 2016 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-27142573

RESUMO

Tissue factor (TF) is the primary trigger of coagulation. Elevated levels of TF are found in atherosclerotic plaques, and TF leads to thrombus formation when released upon plaque rupture. Interleukin (IL)-33 was previously shown to induce angiogenesis and inflammatory activation of endothelial cells (ECs). Here, we investigated the impact of IL-33 on TF in human ECs, as a possible new link between inflammation and coagulation. IL-33 induced TF mRNA and protein in human umbilical vein ECs and coronary artery ECs. IL-33-induced TF expression was ST2- and NF-κB-dependent, but IL-1-independent. IL-33 also increased cell surface TF activity in ECs and TF activity in ECs-derived microparticles. IL-33-treated ECs reduced coagulation time of whole blood and plasma but not of factor VII-deficient plasma. In human carotid atherosclerotic plaques (n = 57), TF mRNA positively correlated with IL-33 mRNA expression (r = 0.691, p < 0.001). In this tissue, IL-33 and TF protein was detected in ECs and smooth muscle cells by immunofluorescence. Furthermore, IL-33 and TF protein co-localized at the site of clot formation within microvessels in plaques of patients with symptomatic carotid stenosis. Through induction of TF in ECs, IL-33 could enhance their thrombotic capacity and thereby might impact on thrombus formation in the setting of atherosclerosis.


Assuntos
Coagulação Sanguínea , Células Endoteliais/metabolismo , Inflamação/patologia , Interleucina-33/metabolismo , Tromboplastina/biossíntese , Células Cultivadas , Humanos , RNA Mensageiro/biossíntese
18.
Blood Cells Mol Dis ; 57: 85-90, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26852662

RESUMO

Granulocyte-macrophage colony stimulating factor (GM-CSF) induces procoagulant activity of macrophages. Tissue factor (TF) is a membrane-bound glycoprotein and substance P (SP) is a pro-inflammatory neuropeptide involved in the formation of membrane blebs. This study investigated the role of SP in TF release by GM-CSF-dependent macrophages. SP significantly decreased TF levels in whole-cell lysates of GM-CSF-dependent macrophages. TF was detected in the culture supernatant by enzyme-linked immunosorbent assay after stimulation of macrophages by SP. Aprepitant (an SP/neurokinin 1 receptor antagonist) reduced TF release from macrophages stimulated with SP. Pretreatment of macrophages with a radical scavenger(pyrrolidinedithiocarbamate) also limited the decrease of TF in whole-cell lysates after stimulation with SP. A protein kinase C inhibitor (rottlerin) partially blocked this macrophage response to SP, while it was significantly inhibited by a ROCK inhibitor (Y-27632) or a dynamin inhibitor (dinasore). An Akt inhibitor (perifosine) also partially blocked this response. Furthermore, siRNA targeting p22phox, ß-arrestin 2, or Rho A, blunted the release of TF from macrophages stimulated with SP. In other experiments, visceral adipocytes derived from cryopreserved preadipocytes were found to produce SP. In conclusion, SP enhances the release of TF from macrophages via the p22phox/ß-arrestin 2/Rho A signaling pathway.


Assuntos
Arrestinas/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Macrófagos/efeitos dos fármacos , NADPH Oxidases/genética , Substância P/farmacologia , Tromboplastina/metabolismo , Quinases Associadas a rho/genética , Acetofenonas/farmacologia , Adipócitos/citologia , Adipócitos/metabolismo , Amidas/farmacologia , Aprepitanto , Arrestinas/antagonistas & inibidores , Arrestinas/metabolismo , Benzopiranos/farmacologia , Dinaminas/antagonistas & inibidores , Dinaminas/genética , Dinaminas/metabolismo , Sequestradores de Radicais Livres/farmacologia , Regulação da Expressão Gênica , Humanos , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/citologia , Macrófagos/metabolismo , Morfolinas/farmacologia , NADPH Oxidases/antagonistas & inibidores , NADPH Oxidases/metabolismo , Antagonistas dos Receptores de Neurocinina-1/farmacologia , Fosforilcolina/análogos & derivados , Fosforilcolina/farmacologia , Cultura Primária de Células , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Pirrolidinas/farmacologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Substância P/biossíntese , Tiocarbamatos/farmacologia , Tromboplastina/biossíntese , beta-Arrestina 2 , beta-Arrestinas , Quinases Associadas a rho/antagonistas & inibidores , Quinases Associadas a rho/metabolismo
19.
J Appl Toxicol ; 36(1): 151-60, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25858758

RESUMO

Airborne exposure to particulate matter with diameter < 10 mcM (PM10) has been linked to an increased risk of thromboembolic events, but the mechanisms are not completely understood. The aim of this study was to evaluate the effect of PM10 phagocytosis on the release of procoagulant molecules in human differentiating macrophages, and that of PM10 inhalation in an experimental model in rats. Human monocytes were separated from the peripheral blood by the lymphoprep method, differentiated in vitro and treated with standard PM10 or vehicle. Sprague-Dawley rats were instilled intratracheally with PM10 or vehicle alone. The outcome was expression of proinflammatory genes and of tissue factor (TF). In human differentiating macrophages, PM10 exposure upregulated inflammatory genes, but most consistently induced TF mRNA and protein levels, but not TF protein inhibitor, resulting in increased TF membrane expression and a procoagulant phenotype. Differentiation towards the anti-inflammatory M2 phenotype inhibited PM10 -mediated TF expression. TF induction required phagocytosis of PM10 , whereas phagocytosis of inert particles was less effective. PM10 phagocytosis was associated with a gene expression profile consistent with intracellular retention of iron, inducing oxidative stress. Both PM10 and iron activated the stress kinases ERK1/2 pathway, involved in the induction of TF expression. In rats, alveolar exposure to PM10 was associated with pulmonary recruitment of inflammatory cells and resulted in local, but not systemic, induction of TF expression, which was sufficient to increase circulating TF levels. In conclusion, TF induction by differentiating lung macrophages, activated following phagocytosis, contributes to the increased risk of thromboembolic complications associated with PM10 exposure.


Assuntos
Macrófagos/efeitos dos fármacos , Material Particulado/toxicidade , Fagocitose/efeitos dos fármacos , Tromboplastina/biossíntese , Adulto , Animais , Diferenciação Celular/efeitos dos fármacos , Citocalasina D/farmacologia , Humanos , Ferro/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Macrófagos/fisiologia , Masculino , Ratos , Ratos Sprague-Dawley , Tromboplastina/genética
20.
Semin Thromb Hemost ; 41(7): 756-64, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26408922

RESUMO

Thrombosis is a major cause of morbidity and mortality in cancer patients. Many clinical factors contribute to the high thrombotic risk of this condition, including the type of malignancy, its disease stage, anticancer therapies, and comorbidities. However, the cancer cell-specific prothrombotic properties together with the host cell inflammatory response are important players in the pathogenesis of the cancer-associated hypercoagulability. Tissue factor (TF) is the most important procoagulant protein expressed by cancer cells, and with other cancer tissue procoagulant properties highly contributes to the procoagulant phenotype of malignant cells. Recent discoveries indicate that oncogenes determine the procoagulant protein expression, including TF, in cancer tissues. In addition, in malignancy, TF is also overexpressed by host normal blood cells triggered by cancer-derived inflammatory stimuli. As a consequence, a subclinical activation of blood coagulation is typically present in cancer patients, as demonstrated by abnormalities of circulating thrombotic biomarkers. The relevance of measuring these biomarkers to determine the patient thrombotic risk level is under active investigation. The goal is to identify the high-risk subgroups to establish more accurate and targeted anticoagulation strategies to prevent thrombosis in cancer patients. Ultimately, the clarification of specific molecular mechanisms triggering blood coagulation in specific cancer types may also indicate alternative ways to inhibit clotting activation in these conditions.


Assuntos
Regulação Neoplásica da Expressão Gênica , Proteínas de Neoplasias/biossíntese , Neoplasias/sangue , Trombofilia/sangue , Tromboplastina/biossíntese , Trombose/sangue , Humanos , Neoplasias/mortalidade , Neoplasias/patologia , Neoplasias/terapia , Trombofilia/mortalidade , Trombofilia/patologia , Trombofilia/prevenção & controle , Trombose/mortalidade , Trombose/patologia , Trombose/prevenção & controle
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA