Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biomolecules ; 12(4)2022 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-35454092

RESUMO

The plasminogen activation system regulates the activity of the serine protease, plasmin. The role of plasminogen receptors in cancer progression is being increasingly appreciated as key players in modulation of the tumor microenvironment. The interaction of plasminogen with cells to promote plasminogen activation requires the presence of proteins exposing C-terminal lysines on the cell surface. Plg-RKT is a structurally unique plasminogen receptor because it is an integral membrane protein that is synthesized with and binds plasminogen via a C-terminal lysine exposed on the cell surface. Here, we have investigated the expression of Plg-RKT in human breast tumors and human breast cancer cell lines. Breast cancer progression tissue microarrays were probed with anti-Plg-RKT mAB and we found that Plg-RKT is widely expressed in human breast tumors, that its expression is increased in tumors that have spread to draining lymph nodes and distant organs, and that Plg-RKT expression is most pronounced in hormone receptor (HR)-positive tumors. Plg-RKT was detected by Western blotting in human breast cancer cell lines. By flow cytometry, Plg-RKT cell surface expression was highest on the most aggressive tumor cell line. Future studies are warranted to address the functions of Plg-RKT in breast cancer.


Assuntos
Neoplasias da Mama , Receptores de Superfície Celular , Neoplasias da Mama/genética , Membrana Celular/metabolismo , Feminino , Humanos , Plasminogênio/metabolismo , Receptores de Superfície Celular/genética , Serina Proteases , Microambiente Tumoral
2.
Blood ; 131(6): 674-685, 2018 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-29246902

RESUMO

The tissue factor (TF) pathway serves both hemostasis and cell signaling, but how cells control these divergent functions of TF remains incompletely understood. TF is the receptor and scaffold of coagulation proteases cleaving protease-activated receptor 2 (PAR2) that plays pivotal roles in angiogenesis and tumor development. Here we demonstrate that coagulation factor VIIa (FVIIa) elicits TF cytoplasmic domain-dependent proangiogenic cell signaling independent of the alternative PAR2 activator matriptase. We identify a Lys-Gly-Glu (KGE) integrin-binding motif in the FVIIa protease domain that is required for association of the TF-FVIIa complex with the active conformer of integrin ß1. A point mutation in this motif markedly reduces TF-FVIIa association with integrins, attenuates integrin translocation into early endosomes, and reduces delayed mitogen-activated protein kinase phosphorylation required for the induction of proangiogenic cytokines. Pharmacologic or genetic blockade of the small GTPase ADP-ribosylation factor 6 (arf6) that regulates integrin trafficking increases availability of TF-FVIIa with procoagulant activity on the cell surface, while inhibiting TF-FVIIa signaling that leads to proangiogenic cytokine expression and tumor cell migration. These experiments delineate the structural basis for the crosstalk of the TF-FVIIa complex with integrin trafficking and suggest a crucial role for endosomal PAR2 signaling in pathways of tissue repair and tumor biology.


Assuntos
Fator VIIa/química , Fator VIIa/metabolismo , Integrina beta1/metabolismo , Domínios e Motivos de Interação entre Proteínas , Receptor PAR-2/metabolismo , Fator 6 de Ribosilação do ADP , Animais , Sítios de Ligação/genética , Células Cultivadas , Fator VIIa/genética , Humanos , Integrina beta1/química , Camundongos , Células NIH 3T3 , Neovascularização Fisiológica/genética , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas/genética , Mapas de Interação de Proteínas , Receptor PAR-2/genética , Transdução de Sinais/genética , Tromboplastina/química , Tromboplastina/metabolismo
3.
Amino Acids ; 49(11): 1867-1883, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28894966

RESUMO

The transformation from normal to malignant phenotype in human cancers is associated with aberrant cell-surface glycosylation. Thus, targeting glycosylation changes in cancer is likely to provide not only better insight into the roles of carbohydrates in biological systems, but also facilitate the development of new molecular probes for bioanalytical and biomedical applications. In the reported study, we have synthesized lectinomimics based on odorranalectin 1; the smallest lectin-like cyclic peptide isolated from the frog Odorrana grahami skin, and assessed the ability of these peptides to bind specific carbohydrates on molecular and cellular levels. In addition, we have shown that the disulfide bond found in 1 can be replaced with a lactam bridge. However, the orientation of the lactam bridge, peptides 2 and 3, influenced cyclic peptide's conformation and thus these peptides' ability to bind carbohydrates. Naturally occurring 1 and its analog 3 that adopt similar conformation in water bind preferentially L-fucose, and to a lesser degree D-galactose and N-acetyl-D-galactosamine, typically found within the mucin O-glycan core structures. In cell-based assays, peptides 1 and 3 showed a similar binding profile to Aleuria aurantia lectin and these two peptides inhibited the migration of metastatic breast cancer cell lines in a Transwell assay. Altogether, the reported data demonstrate the feasibility of designing lectinomimics based on cyclic peptides.


Assuntos
Sistemas de Liberação de Medicamentos , Lectinas , Neoplasias/metabolismo , Peptídeos Cíclicos/síntese química , Peptidomiméticos/síntese química , Polissacarídeos/metabolismo , Ligação Competitiva , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Fucose/agonistas , Fucose/metabolismo , Células Hep G2 , Humanos , Concentração Inibidora 50 , Lactamas/química , Lectinas/química , Lectinas/metabolismo , Células MCF-7 , Simulação de Acoplamento Molecular , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Peptídeos Cíclicos/química , Peptídeos Cíclicos/metabolismo , Peptídeos Cíclicos/farmacologia , Peptidomiméticos/química , Peptidomiméticos/metabolismo , Peptidomiméticos/farmacologia , Polissacarídeos/química , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Relação Estrutura-Atividade
4.
Int Rev Cell Mol Biol ; 309: 259-302, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24529725

RESUMO

Plasminogen (PLG) is the zymogen of plasmin, the major enzyme that degrades fibrin clots. In addition to its binding and activation on fibrin clots, PLG also specifically interacts with cell surfaces where it is more efficiently activated by PLG activators, compared with the reaction in solution. This results in association of the broad-spectrum proteolytic activity of plasmin with cell surfaces that functions to promote cell migration. Here, we review emerging data establishing a role for PLG, plasminogen receptors and the newly discovered plasminogen receptor, Plg-RKT, in macrophage recruitment in the inflammatory response, and we address mechanisms by which the interplay between PLG and its receptors regulates inflammation.


Assuntos
Macrófagos/metabolismo , Plasminogênio/metabolismo , Receptores de Superfície Celular/metabolismo , Sequência de Aminoácidos , Animais , Humanos , Macrófagos/patologia , Dados de Sequência Molecular , Peritonite/patologia , Plasminogênio/química , Proteômica , Receptores de Superfície Celular/química
5.
Oncoimmunology ; 2(10): e25989, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-24244896

RESUMO

New treatments are needed for brain metastasis, which is associated with high morbidity and mortality. Two novel cellular and gene therapy modalities were evaluated in xenograft models for human breast cancer. The individual and especially the combined treatments with alloreactive cytotoxic T lymphocytes and replicating retroviral vectors coding for prodrug activating enzymes followed later with nontoxic prodrug demonstrated efficacy without off-target effects.

6.
Adipocyte ; 2(3): 165-9, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23991363

RESUMO

The importance of the microenvironment in breast cancer growth and progression is becoming increasingly clear. Adipocytes are abundant in the mammary microenvironment, and recent studies show that adipocytes produce endocrine, inflammatory, and angiogenic factors that have tremendous potential to affect adjacent breast cancer cells. Yet, the extent to which local adipocyte function contributes to the pathogenesis of breast cancer is largely unexplored. Here we describe a unique animal model to study interactions between adipocytes and breast cancer cells in the tumor microenvironment. Our results suggest that local interactions between adipocytes and tumor cells are sufficient to promote the growth of hormone-dependent breast cancer. We also demonstrate that leptin signaling in adipocytes induces aromatase expression, expected to result in higher estrogen in the microenvironment thus enabling mammary tumorigenesis.

7.
Clin Cancer Res ; 19(15): 4137-48, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23780889

RESUMO

PURPOSE: Individual or combined strategies of cellular therapy with alloreactive CTLs (alloCTL) and gene therapy using retroviral replicating vectors (RRV) encoding a suicide prodrug activating gene were explored for the treatment of breast tumors metastatic to the brain. EXPERIMENTAL DESIGN: AlloCTL, sensitized to the HLA of MDA-MB-231 breast cancer cells, were examined in vitro for antitumor functionality toward breast cancer targets. RRV encoding the yeast cytosine deaminase (CD) gene was tested in vivo for virus spread, ability to infect, and kill breast cancer targets when exposed to 5-fluorocytosine (5-FC). Individual and combination treatments were tested in subcutaneous and intracranial xenograft models with 231BR, a brain tropic variant. RESULTS: AlloCTL preparations were cytotoxic, proliferated, and produced IFN-γ when coincubated with target cells displaying relevant HLA. In vivo, intratumorally placed alloCTL trafficked through one established intracranial 231BR focus to another in contralateral brain and induced tumor cell apoptosis. RRV-CD efficiently spread in vivo, infected 231BR and induced their apoptosis upon 5-FC exposure. Subcutaneous tumor volumes were significantly reduced in alloCTL and/or gene therapy-treated groups compared to control groups. Mice with established intracranial 231BR tumors treated with combined alloCTL and RRV-CD had a median survival of 97.5 days compared with single modalities (50-83 days); all experimental treatment groups survived significantly longer than sham-treated groups (median survivals 31.5 or 40 days) and exhibited good safety/toxicity profiles. CONCLUSION: The results indicate combining cellular and suicide gene therapies is a viable strategy for the treatment of established breast tumors in the brain.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Terapia Genética , Linfócitos T Citotóxicos , Adenoviridae , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/secundário , Neoplasias da Mama/patologia , Terapia Combinada , Citosina Desaminase/genética , Citosina Desaminase/uso terapêutico , Feminino , Flucitosina/administração & dosagem , Genes Transgênicos Suicidas/genética , Vetores Genéticos , Humanos , Camundongos , Pró-Fármacos/administração & dosagem
8.
PLoS One ; 8(4): e61071, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23593394

RESUMO

Several markers identify cancer stem cell-like populations, but little is known about the functional roles of stem cell surface receptors in tumor progression. Here, we show that the endothelial protein C receptor (EPCR), a stem cell marker in hematopoietic, neuronal and epithelial cells, is crucial for breast cancer growth in the orthotopic microenvironment of the mammary gland. Mice with a hypomorphic allele of EPCR show reduced tumor growth in the PyMT-model of spontaneous breast cancer development and deletion of EPCR in established PyMT tumor cells significantly attenuates transplanted tumor take and growth. We find expansion of EPCR(+) cancer stem cell-like populations in aggressive, mammary fat pad-enhanced human triple negative breast cancer cells. In this model, EPCR-expressing cells have markedly increased mammosphere- and tumor-cell initiating activity compared to another stable progenitor-like subpopulation present at comparable frequency. We show that receptor blocking antibodies to EPCR specifically attenuate in vivo tumor growth initiated by either EPCR(+) cells or the heterogenous mixture of EPCR(+) and EPCR(-) cells. Furthermore, we have identified tumor associated macrophages as a major source for recognized ligands of EPCR, suggesting a novel mechanism by which cancer stem cell-like populations are regulated by innate immune cells in the tumor microenvironment.


Assuntos
Antígenos CD/metabolismo , Neoplasias da Mama/metabolismo , Transformação Celular Neoplásica/metabolismo , Glicoproteínas/metabolismo , Receptores de Superfície Celular/metabolismo , Tecido Adiposo/metabolismo , Animais , Antígenos CD/genética , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica/genética , Análise por Conglomerados , Modelos Animais de Doenças , Receptor de Proteína C Endotelial , Feminino , Perfilação da Expressão Gênica , Glicoproteínas/antagonistas & inibidores , Glicoproteínas/genética , Humanos , Macrófagos/metabolismo , Macrófagos/patologia , Glândulas Mamárias Animais/metabolismo , Camundongos , Células-Tronco Neoplásicas/metabolismo , Receptores de Superfície Celular/antagonistas & inibidores , Receptores de Superfície Celular/genética , Transplante Heterólogo , Carga Tumoral/genética
10.
Blood ; 116(26): 6106-13, 2010 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-20861457

RESUMO

Constitutive expression of tissue factor (TF) by cancer cells triggers local activation of the coagulation cascade and promotes breast cancer progression through cell signaling involving protease activated receptor (PAR)2. In human breast cancer, TF and PAR2 are up-regulated and TF cytoplasmic domain phosphorylation is correlated with relapse. Here we show that cancer cell PAR2 signaling promotes angiogenesis independent of PAR2 phosphorylation at the recognized ß-arrestin recruitment site. Similar to PAR2(-/-) mice, TF cytoplasmic domain-deleted (TF(ΔCT)) mice have delayed spontaneous breast cancer development in the polyoma middle T model. Simultaneous deletion of PAR2 in TF(ΔCT) mice did not further delay tumor appearance, consistent with overlapping roles of TF and PAR2 in promoting the angiogenic switch in early stages of breast cancer. In advanced carcinomas, tumor-associated macrophages were reduced in TF(ΔCT) and TF(ΔCT)/PAR2(-/-) mice, and increased tumor vessel diameters of TF(ΔCT) mice were partially reversed by PAR2-deficiency, indicating that the TF cytoplasmic domain has additional roles that are interdependent with PAR2 signaling in regulating host angiogenic responses. These experiments demonstrate a crosstalk of tumor cell TF cytoplasmic domain and PAR2 signaling and provide a possible mechanism for the close correlation between TF phosphorylation and cancer recurrence of TF and PAR2-positive clinical breast cancer.


Assuntos
Neoplasias Mamárias Experimentais/irrigação sanguínea , Neoplasias Mamárias Experimentais/patologia , Neovascularização Patológica , Receptor PAR-2/fisiologia , Tromboplastina/fisiologia , Animais , Feminino , Humanos , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais
11.
Mol Cancer Ther ; 9(11): 3024-32, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20833754

RESUMO

Bone marrow hypoplasia and pancytopenia are among the most undesirable sequelae of chemotherapy for the treatment of cancer. We recently showed that hyaluronan (HA) facilitates hematopoietic recovery in tumor-free animals receiving chemotherapeutic agents. However, following a chemotherapeutic regimen in tumor-bearing animals, it is possible that residual tumor cells might respond to systemic injections of HA. Thus, in this study, we investigated the effect of HA on the regrowth of residual tumor cells following chemotherapy. As a model, we used the HCT-8 human colon carcinoma cell line, which expresses the HA receptor CD44, binds exogenous HA, and is susceptible to a chemotherapy protocol containing irinotecan and 5-fluorouracil in a human/mouse xenograft model. HCT-8 cells were implanted in severe combined immunodeficient mice, followed by irinotecan/5-fluorouracil treatment. After three rounds of chemotherapy, residual tumors were allowed to regrow in the presence or absence of HA. The dynamics of tumor regrowth in the group treated with HA was slower compared with the control group. By week 5 after tumor implantation, the difference in the size of regrown tumors was statistically significant and correlated with lower proliferation and higher apoptosis in HA-treated tumors as compared with controls. This finding provides evidence that HA treatment does not stimulate but delays the growth of residual cancer cells, which is an important parameter in establishing whether the use of HA can enhance current chemotherapeutic strategies.


Assuntos
Carcinoma/tratamento farmacológico , Neoplasias do Colo/tratamento farmacológico , Ácido Hialurônico/farmacologia , Recidiva Local de Neoplasia/prevenção & controle , Animais , Carcinoma/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Quimioterapia Adjuvante , Neoplasias do Colo/patologia , Citostáticos/administração & dosagem , Citostáticos/farmacologia , Esquema de Medicação , Feminino , Humanos , Ácido Hialurônico/administração & dosagem , Camundongos , Camundongos SCID , Modelos Biológicos , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Biochem Biophys Res Commun ; 383(2): 167-71, 2009 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-19341712

RESUMO

The gene for phosphatidylinositol-4-phosphate adaptor-2 (FAPP2) encodes a cytoplasmic lipid transferase with a plekstrin homology domain that has been implicated in vesicle maturation and transport from trans-Golgi to the plasma membrane. The introduction of ribozymes targeting the FAPP2 gene in colon carcinoma cells induced their apoptosis in the presence of Fas agonistic antibody. Furthermore, by quantitative PCR we showed that a siRNA specific to FAPP2, but not a randomized siRNA control, reduced FAPP2 gene expression in tumor cells. Transfection of FAPP2 siRNA into human tumor cells then incubated with FasL resulted in reduction of viable cell numbers. Also, FAPP2 siRNA transfected glioma and breast tumor cells showed significant increases in apoptosis upon incubation with soluble FasL, but the apoptosis did not necessarily correlate with increased Fas expression. These data demonstrate a previously unknown role for FAPP2 in conferring resistance to apoptosis and indicate that FAPP2 may be a target for cancer therapy.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Apoptose , Proteína Ligante Fas/agonistas , Neoplasias/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Anticorpos/imunologia , Apoptose/genética , Linhagem Celular Tumoral , Regulação para Baixo , Proteína Ligante Fas/imunologia , Proteína Ligante Fas/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias/patologia , RNA Interferente Pequeno/genética
13.
Cancer Ther ; 6(B): 865-876, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19081762

RESUMO

The PATZ1 gene encodes a transcription factor that belongs to the BTB/POZ group of transcriptional regulators and has been implicated as a transcriptional repressor. We cloned cDNA from glioma cell lines and found they expressed transcript variant 2 of PATZ1. We designed a specific siRNA against PATZ1 and showed that this siRNA, but not a control randomized siRNA, reduced PATZ1 expression in glioma cells as determined by quantitative PCR. In a panel of human glioma cell lines incubated with proapoptotic FasL, those transfected with PATZ1 siRNA displayed reduced cell numbers by the MTT colorimetric assay, relative to those transfected with randomized siRNA. Further studies showed that in 10-08-MG, U-251MG, U-87MG, and T98G cells PATZ1 siRNA significantly increased apoptosis in response to incubation with soluble FasL, as shown by a morphologic acridine orange/ethidium bromide apoptotic assay. Using an apoptosis specific cDNA microarray we further demonstrated that down-regulation of PATZ1 by siRNA resulted in the upregulation of death receptor pro-apoptotic genes including caspase 8 and Death Receptor 5 (DR5) in U-373MG cells. Since DR5 is the receptor for TRAIL we tested whether PATZ1 downregulation also sensitized cells to TRAIL-induced apoptosis and found that PATZ1 siRNA, but not control siRNA, sensitized U-251MG and T98G glioma cells to TRAIL-induced apoptosis. Altogether, these data demonstrate a previously unknown role for the transcription factor PATZ1 in conferring resistance to apoptosis and indicate that modulation of PATZ1 expression may be a therapeutic strategy for gliomas.

14.
Cancer Res ; 68(17): 7219-27, 2008 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-18757438

RESUMO

The G protein-coupled protease-activated receptors (PAR) are key signaling components for proteases in vascular biology and tumor progression. To address the contributions of PAR1 and PAR2 to breast cancer development, we established cohorts of mouse mammary tumor virus-polyoma middle T (PyMT) PAR1(-/-) and PAR2(-/-) mice, considering that the PyMT model recapitulates aspects of human disease. Appearance of palpable tumors, tumor expansion, and metastasis was indistinguishable between wild-type and PAR1(-/-) mice. PAR1(-/-) breast cancer cells were no longer responsive to thrombin in vitro, excluding compensatory up-regulation of alternative thrombin receptors and indicating that thrombin-PAR1 signaling is dispensable in breast tumor microenvironments. In contrast, palpable tumors and multifocal disease developed slower in PAR2(-/-) mice, and as a consequence of delayed tumor onset, metastasis was reduced. Analysis of early tumors showed persistence of adenomas with delayed appearance of vascularized adenocarcinomas in PAR2(-/-) mice. Furthermore, CXCL1 production by early PAR2(-/-) tumors was reduced. These results are consistent with previous xenograft data that implicated breast cancer PAR2 signaling in the induction of proangiogenic growth factors and chemokines. This study establishes that protease signaling contributes to mammary tumor development and that PAR2, rather than the thrombin receptor PAR1, plays a crucial role in the angiogenic switch.


Assuntos
Adenocarcinoma/fisiopatologia , Antígenos Transformantes de Poliomavirus/genética , Neoplasias Mamárias Experimentais/fisiopatologia , Receptor PAR-1/metabolismo , Receptor PAR-2/metabolismo , Transdução de Sinais , Adenocarcinoma/irrigação sanguínea , Animais , Western Blotting , Ensaio de Imunoadsorção Enzimática , Humanos , Imuno-Histoquímica , Neoplasias Mamárias Experimentais/irrigação sanguínea , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Patológica/genética , Reação em Cadeia da Polimerase
15.
Blood ; 111(1): 190-9, 2008 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17901245

RESUMO

Coagulation activation by tissue factor (TF) is implicated in cancer progression, cancer-associated thrombosis and metastasis. The role of direct TF signaling pathways in cancer, however, remains incompletely understood. Here we address how TF contributes to primary tumor growth by using a unique pair of isotype-matched antibodies that inhibit either coagulation (monoclonal antibody [Mab]-5G9) or direct signaling (Mab-10H10). We demonstrate that the inhibitory antibody of direct TF-VIIa signaling not only blocks TF-VIIa mediated activation of PAR2, but also disrupts the interaction of TF with integrins. In epithelial and TF-expressing endothelial cells, association of TF with beta1 integrins is regulated by TF extracellular ligand binding and independent of PAR2 signaling or proteolytic activity of VIIa. In contrast, alpha3beta1 integrin association of TF is constitutive in breast cancer cells and blocked by Mab-10H10 but not by Mab-5G9. Mab-5G9 has antitumor activity in vivo, but we show here that Mab-10H10 is at least as effective in suppressing human xenograft tumors in 2 different models. Breast tumor growth was also attenuated by blocking PAR2 signaling. These results show that tumor cell TF-PAR2 signaling is crucial for tumor growth and suggest that anti-TF strategies can be applied in cancer therapy with minor impairment of TF-dependent hemostatic pathways.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/secundário , Transdução de Sinais/fisiologia , Tromboplastina/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Divisão Celular , Linhagem Celular Transformada , Endotélio Vascular/citologia , Fator VIIa/metabolismo , Fator VIIa/farmacologia , Humanos , Integrina beta1/metabolismo , Queratinócitos/citologia , Queratinócitos/metabolismo , Camundongos , Camundongos SCID , Receptor PAR-1/metabolismo , Receptor PAR-2/metabolismo , Transdução de Sinais/imunologia , Tromboplastina/imunologia , Veias Umbilicais/citologia
16.
Breast Cancer Res Treat ; 110(2): 211-22, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17687643

RESUMO

The chemokine receptor CXCR4 is functionally expressed on the cell surface of various cancer cells, and plays a role in cell proliferation and migration of these cells. Specifically, in breast cancer cells the CXCR4/CXCL12 axis has been implicated in cell migration in vitro and in metastasis in vivo, but the underlying signaling mechanisms are incompletely understood. The xenograft-derived MDA-MB-231 breast cancer cell line (231mfp), which was shown previously to grow more aggressively than the parent cells, showed increased CXCR4 expression at the mRNA, total protein and cell surface expression level. This correlated with an enhanced response to CXCL12, specifically in augmented and prolonged Akt activation in a G(i), Src family kinase and PI-3 kinase dependent fashion. 231mfp cells migrated towards CXCL12--in contrast to the parent cell line--and this chemotaxis was blocked by inhibition of G(i), Src family kinases, PI-3 kinase and interestingly, Akt itself, as could be shown with two pharmacological inhibitors, a dominant negative Akt construct and with Akt shRNA. Collectively, we have demonstrated that prolonged Akt activation is an important signaling pathway for breast cancer cells expressing CXCR4 and is necessary for CXCL12-dependent cell migration.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Quimiocina CXCL12/metabolismo , Quimiotaxia , Ativação Enzimática , Humanos , Metaloproteinases da Matriz/metabolismo , Modelos Biológicos , Toxina Pertussis/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Receptores CXCR4/metabolismo , Transdução de Sinais
17.
Arterioscler Thromb Vasc Biol ; 27(6): 1456-62, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17363687

RESUMO

OBJECTIVE: Tissue factor (TF) initiates coagulation and indirectly triggers thrombin-dependent protease activated receptor (PAR) signaling. The TF-VIIa complex also directly cleaves PAR2 and promotes angiogenesis in vitro in TF cytoplasmic domain-deleted (TF(deltaCT)) mice. Here we address the effect of PAR1 and PAR2 deficiency on angiogenesis in vivo. METHODS AND RESULTS: In hypoxia-driven angiogenesis of oxygen induced retinopathy (OIR), wild-type, PAR1-/-, PAR2-/-, and TF(deltaCT) mice showed a comparable regression of the superficial vascular plexus during the initial exposure of mice to hyperoxia. However, TF(deltaCT) mice revascularized areas of central vaso-obliteration significantly faster than wild-type animals. Pharmacological inhibition of the TF-VIIa complex, but not of Xa, and blockade of tyrosine kinase receptor pathways with Gleevec reversed accelerated angiogenesis of TF(deltaCT) mice to revascularization rates observed in wild-type mice. Genetic deletion of PAR2, but not of PAR1, abolished enhanced revascularization of TF(deltaCT) mice. PAR1 knock-out animals were indistinguishable from wild-type mice in the model of retinal neoangiogenesis and angiogenesis-dependent subcutaneous tumor growth was unaltered in PAR1- and PAR2-deficient animals. CONCLUSION: Loss of the TF cytoplasmic domain results in accelerated hypoxia-induced angiogenesis mediated by TF-VIIa signaling. PAR2 signaling is sufficient for this proangiogenic effect without apparent contributions of mouse host cell PAR1.


Assuntos
Hiperóxia/metabolismo , Hipóxia/complicações , Receptor PAR-1/metabolismo , Receptor PAR-2/metabolismo , Neovascularização Retiniana/etiologia , Vasos Retinianos/metabolismo , Transdução de Sinais , Tromboplastina/metabolismo , Animais , Benzamidas , Inibidores dos Fatores de Coagulação Sanguínea/farmacologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Fator VIIa/metabolismo , Hiperóxia/induzido quimicamente , Hiperóxia/patologia , Hipóxia/metabolismo , Hipóxia/patologia , Mesilato de Imatinib , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Neoplasias Experimentais/irrigação sanguínea , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Oxigênio , Piperazinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/farmacologia , Receptor PAR-1/deficiência , Receptor PAR-1/genética , Receptor PAR-2/deficiência , Receptor PAR-2/genética , Neovascularização Retiniana/metabolismo , Neovascularização Retiniana/patologia , Vasos Retinianos/patologia , Transdução de Sinais/efeitos dos fármacos , Tromboplastina/genética , Fatores de Tempo
18.
Proc Natl Acad Sci U S A ; 103(38): 13932-7, 2006 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-16959886

RESUMO

Cell-surface tissue factor (TF) binds the serine protease factor VIIa to activate coagulation or, alternatively, to trigger signaling through the G protein-coupled, protease-activated receptor 2 (PAR2) relevant to inflammation and angiogenesis. Here we demonstrate that TF.VIIa-mediated coagulation and cell signaling involve distinct cellular pools of TF. The surface-accessible, extracellular Cys186-Cys209 disulfide bond of TF is critical for coagulation, and protein disulfide isomerase (PDI) disables coagulation by targeting this disulfide. A TF mutant (TF C209A) with an unpaired Cys186 retains TF.VIIa signaling activity, and it has reduced affinity for VIIa, a characteristic of signaling TF on cells with constitutive TF expression. We further show that PDI suppresses TF coagulant activity in a nitric oxide-dependent pathway, linking the regulation of TF thrombogenicity to oxidative stress in the vasculature. Furthermore, a unique monoclonal antibody recognizes only the noncoagulant, cryptic conformation of TF. This antibody inhibits formation of the TF.PAR2 complex and TF.VIIa signaling, but it does not prevent coagulation activation. These experiments delineate an upstream regulatory mechanism that controls TF function, and they provide initial evidence that TF.VIIa signaling can be specifically inhibited with minimal effects on coagulation.


Assuntos
Comunicação Celular/fisiologia , Dissulfetos/química , Fator VIIa/metabolismo , Tromboplastina/metabolismo , Coagulação Sanguínea/fisiologia , Linhagem Celular , Cisteína/metabolismo , Dissulfetos/metabolismo , Humanos , Isomerismo , Complexos Multiproteicos , Óxido Nítrico/metabolismo , Oxirredução , Estresse Oxidativo , Isomerases de Dissulfetos de Proteínas/metabolismo , Receptor PAR-2/metabolismo , Tromboplastina/química
19.
Proc Natl Acad Sci U S A ; 103(29): 11009-14, 2006 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-16822849

RESUMO

Studies at the interface of chemistry and biology have allowed us to develop an immunotherapeutic approach called chemically programmed antibodies (cpAbs), which combines the merits of traditional small-molecule drug design with immunotherapy. In this approach, a catalytic antibody catalyzes the covalent conjugation of a small molecule or peptide to the active site of the antibody, effectively recruiting the binding specificity of the conjugated molecule to the antibody. In essence, this technology provides the tools for breaking the "one antibody-one target axiom" of immunochemistry. Our studies in this area have focused on using the chemistry of the well studied aldolase catalytic antibodies of which mAb 38C2 is a member. Previously, we explored reversible assembly of cpAbs available through diketone chemistry. In this article, we explore a unique proadapter assembly strategy wherein an antibody 38C2-catalyzed transformation unveils a reactive tag that then reacts to form a stable covalent bond with the antibody. An integrin alpha(v)beta 3 antagonist was synthesized with the designed proadapter and studied using human breast cancer cell lines MDA-MB-231 and MDA-MB-435. We demonstrate that this approach allows for (i) the effective assembly of cpAbs in vitro and in vivo, (ii) selective retargeting of 38C2 to integrin alpha(v)beta 3 expressing breast cancer cell lines, (iii) intracellular delivery of cpAbs into cells, (iv) dramatically increased circulatory half-life, and (v) substantial enhancement of the therapeutic effect over the peptidomimetic itself in animal models of breast cancer metastasis. We believe that this technology possesses potential for the treatment and diagnosis of disease.


Assuntos
Anticorpos/imunologia , Antígenos/imunologia , Animais , Antígenos/química , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Linhagem Celular Tumoral , Feminino , Humanos , Técnicas Imunológicas , Integrina alfaVbeta3/antagonistas & inibidores , Integrina alfaVbeta3/imunologia , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/prevenção & controle , Neoplasias Pulmonares/secundário , Camundongos , Camundongos SCID , Estrutura Molecular
20.
Semin Thromb Hemost ; 32 Suppl 1: 61-8, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16673267

RESUMO

Advanced cancer is associated with a hypercoagulable state that is triggered by tissue factor (TF). TF-initiated thrombin generation is crucial for metastasis through fibrin and platelet deposition, as well as thrombin-dependent protease-activated receptor (PAR) 1 signaling. Surprisingly, PAR2, which is not cleaved by thrombin, appears to cosignal with PAR1 to elicit thrombin effects in metastatic tumor cells. In contrast to TF-driven thrombin pathways in metastasis, direct TF signaling plays a role in angiogenesis-dependent tumor growth. In TF cytoplasmic-domain-deleted mice, PAR2-dependent angiogenesis and tumor growth is enhanced, demonstrating a role for host cell TF signaling. In tumor cells, TF-factor VIIa (FVIIa) activates PAR2 and thereby regulates proangiogenic growth factor expression as well as integrins involving crosstalk with the TF cytoplasmic domain. In addition to thrombin-PAR signaling in metastasis and TF-FVIIa-PAR2 signaling in tumor growth, it is likely that additional protease pathways will prove to be crucial activators of PARs in cancer. Transmembrane serine proteases as well as matrix metalloproteinase are prime candidates for accessory pathways to regulate metastasis, tumor expansion, and angiogenesis dependent on specific features of the local tumor microenvironment.


Assuntos
Neoplasias/etiologia , Trombina/metabolismo , Animais , Humanos , Metástase Neoplásica , Neoplasias/sangue , Neoplasias/patologia , Neovascularização Patológica , Receptores Ativados por Proteinase/metabolismo , Tromboplastina/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...