Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Biochem J ; 465(2): 259-70, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25360794

RESUMO

Gelatinase B/matrix metalloproteinase-9 (MMP-9) (EC 3.4.24.35) cleaves many substrates and is produced by most cell types as a zymogen, proMMP-9, in complex with the tissue inhibitor of metalloproteinases-1 (TIMP-1). Natural proMMP-9 occurs as monomers, homomultimers and heterocomplexes, but our knowledge about the overall structure of proMMP-9 monomers and multimers is limited. We investigated biochemical, biophysical and functional characteristics of zymogen and activated forms of MMP-9 monomers and multimers. In contrast with a conventional notion of a dimeric nature of MMP-9 homomultimers, we demonstrate that these are reduction-sensitive trimers. Based on the information from electrophoresis, AFM and TEM, we generated a 3D structure model of the proMMP-9 trimer. Remarkably, the proMMP-9 trimers possessed a 50-fold higher affinity for TIMP-1 than the monomers. In vivo, this finding was reflected in a higher extent of TIMP-1 inhibition of angiogenesis induced by trimers compared with monomers. Our results show that proMMP-9 trimers constitute a novel structural and functional entity that is differentially regulated by TIMP-1.


Assuntos
Precursores Enzimáticos/química , Metaloproteinase 9 da Matriz/química , Modelos Moleculares , Complexos Multiproteicos/química , Inibidor Tecidual de Metaloproteinase-1/química , Precursores Enzimáticos/genética , Precursores Enzimáticos/metabolismo , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Microscopia de Força Atômica , Microscopia Eletrônica de Transmissão , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Complexos Multiproteicos/ultraestrutura , Inibidor Tecidual de Metaloproteinase-1/genética , Inibidor Tecidual de Metaloproteinase-1/metabolismo
2.
Blood ; 122(25): 4054-67, 2013 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-24174628

RESUMO

A proangiogenic function of tissue-infiltrating monocytes/macrophages has long been attributed to their matrix metalloproteinase-9 zymogen (proMMP-9). Herein, we evaluated the capacity of human monocytes, mature M0 macrophages, and M1- and M2-polarized macrophages to induce proMMP-9-mediated angiogenesis. Only M2 macrophages induced angiogenesis at levels comparable with highly angiogenic neutrophils previously shown to release their proMMP-9 in a unique form, free of tissue inhibitor of metalloproteinases-1 (TIMP-1). Macrophage differentiation was accompanied by induction of low-angiogenic, TIMP-1-encumbered proMMP-9. However, polarization toward the M2, but not the M1 phenotype, caused a substantial downregulation of TIMP-1 expression, resulting in production of angiogenic, TIMP-deficient proMMP-9. Correspondingly, the angiogenic potency of M2 proMMP-9 was lost after its complexing with TIMP-1, whereas TIMP-1 silencing in M0/M1 macrophages rendered them both angiogenic. Similar to human cells, murine bone marrow-derived M2 macrophages also shut down their TIMP-1 expression and produced proMMP-9 unencumbered by TIMP-1. Providing proof that angiogenic capacity of murine M2 macrophages depended on their TIMP-free proMMP-9, Mmp9-null M2 macrophages were nonangiogenic, although their TIMP-1 was severely downregulated. Our study provides a unifying molecular mechanism for high angiogenic capacity of TIMP-free proMMP-9 that would be uniquely produced in a pathophysiological microenvironment by influxing neutrophils and/or M2 polarized macrophages.


Assuntos
Diferenciação Celular/fisiologia , Precursores Enzimáticos/metabolismo , Macrófagos/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Neovascularização Fisiológica/fisiologia , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Animais , Embrião de Galinha , Regulação para Baixo/fisiologia , Precursores Enzimáticos/genética , Humanos , Macrófagos/citologia , Metaloproteinase 9 da Matriz/genética , Camundongos , Camundongos Mutantes , Neutrófilos/citologia , Neutrófilos/metabolismo , Inibidor Tecidual de Metaloproteinase-1/genética
3.
Cell Commun Signal ; 12: 61, 2014 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-25248616

RESUMO

BACKGROUND: INPP4B and PTEN dual specificity phosphatases are frequently lost during progression of prostate cancer to metastatic disease. We and others have previously shown that loss of INPP4B expression correlates with poor prognosis in multiple malignancies and with metastatic spread in prostate cancer. RESULTS: We demonstrate that de novo expression of INPP4B in highly invasive human prostate carcinoma PC-3 cells suppresses their invasion both in vitro and in vivo. Using global gene expression analysis, we found that INPP4B regulates a number of genes associated with cell adhesion, the extracellular matrix, and the cytoskeleton. Importantly, de novo expressed INPP4B suppressed the proinflammatory chemokine IL-8 and induced PAK6. These genes were regulated in a reciprocal manner following downregulation of INPP4B in the independently derived INPP4B-positive LNCaP prostate cancer cell line. Inhibition of PI3K/Akt pathway, which is highly active in both PC-3 and LNCaP cells, did not reproduce INPP4B mediated suppression of IL-8 mRNA expression in either cell type. In contrast, inhibition of PKC signaling phenocopied INPP4B-mediated inhibitory effect on IL-8 in either prostate cancer cell line. In PC-3 cells, INPP4B overexpression caused a decline in the level of metastases associated BIRC5 protein, phosphorylation of PKC, and expression of the common PKC and IL-8 downstream target, COX-2. Reciprocally, COX-2 expression was increased in LNCaP cells following depletion of endogenous INPP4B. CONCLUSION: Taken together, we discovered that INPP4B is a novel suppressor of oncogenic PKC signaling, further emphasizing the role of INPP4B in maintaining normal physiology of the prostate epithelium and suppressing metastatic potential of prostate tumors.


Assuntos
Monoéster Fosfórico Hidrolases/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Proteína Quinase C/metabolismo , Animais , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Ciclo-Oxigenase 2/metabolismo , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Indóis/farmacologia , Proteínas Inibidoras de Apoptose/metabolismo , Interleucina-8/genética , Masculino , Maleimidas/farmacologia , Camundongos SCID , Invasividade Neoplásica , Monoéster Fosfórico Hidrolases/genética , Proteína Quinase C/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , RNA Interferente Pequeno/genética , Survivina , Quinases Ativadas por p21/genética
4.
Am J Pathol ; 179(3): 1455-70, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21741942

RESUMO

Tumor-associated neutrophils contribute to neovascularization by supplying matrix metalloproteinase-9 (MMP-9), a protease that has been genetically and biochemically linked to induction of angiogenesis. Specific roles of inflammatory neutrophils and their distinct proMMP-9 in the coordinate regulation of tumor angiogenesis and tumor cell dissemination, however, have not been addressed. We demonstrate that the primary tumors formed by highly disseminating variants of human fibrosarcoma and prostate carcinoma recruit elevated levels of infiltrating MMP-9-positive neutrophils and concomitantly exhibit enhanced levels of angiogenesis and intravasation. Specific inhibition of neutrophil influx by interleukin 8 (IL-8) neutralization resulted in the coordinated diminishment of tumor angiogenesis and intravasation, both of which were rescued by purified neutrophil proMMP-9. However, if neutrophil proMMP-9, naturally devoid of tissue inhibitor of metalloproteinases (TIMP), was delivered in complex with TIMP-1 or in a mixture with TIMP-2, the protease failed to rescue the inhibitory effects of anti-IL8 therapy, indicating that the TIMP-free status of proMMP-9 is critical for facilitating tumor angiogenesis and intravasation. Our findings directly link tumor-associated neutrophils and their TIMP-free proMMP-9 with the ability of aggressive tumor cells to induce the formation of new blood vessels that serve as conduits for tumor cell dissemination. Thus, treatment of cancers associated with neutrophil infiltration may benefit from specific targeting of neutrophil MMP-9 at early stages to prevent ensuing tumor angiogenesis and tumor metastasis.


Assuntos
Precursores Enzimáticos/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Neovascularização Patológica/enzimologia , Neutrófilos/metabolismo , Inibidores Teciduais de Metaloproteinases/fisiologia , Animais , Linhagem Celular Tumoral , Embrião de Galinha , Fibrossarcoma/irrigação sanguínea , Fibrossarcoma/metabolismo , Fibrossarcoma/secundário , Humanos , Interleucina-8/farmacologia , Masculino , Camundongos , Camundongos Nus , Camundongos SCID , Invasividade Neoplásica , Neoplasias da Próstata/irrigação sanguínea , Neoplasias da Próstata/metabolismo
5.
J Biomed Biotechnol ; 2012: 564259, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23097597

RESUMO

Plasmin, one of the most potent and reactive serine proteases, is involved in various physiological processes, including embryo development, thrombolysis, wound healing and cancer progression. The proteolytic activity of plasmin is tightly regulated through activation of its precursor, plasminogen, only at specific times and in defined locales as well as through inhibition of active plasmin by its abundant natural inhibitors. By exploiting the plasminogen activating system and overexpressing distinct components of the plasminogen activation cascade, such as pro-uPA, uPAR and plasminogen receptors, malignant cells can enhance the generation of plasmin which in turn, modifies the tumor microenvironment to sustain cancer progression. While plasmin-mediated degradation and modification of extracellular matrix proteins, release of growth factors and cytokines from the stroma as well as activation of several matrix metalloproteinase zymogens, all have been a focus of cancer research studies for decades, the ability of plasmin to cleave transmembrane molecules and thereby to generate functionally important cleaved products which induce outside-in signal transduction, has just begun to receive sufficient attention. Herein, we highlight this relatively understudied, but important function of the plasmin enzyme as it is generated de novo at the interface between cross-talking cancer and host cells.


Assuntos
Comunicação Celular/fisiologia , Membrana Celular/metabolismo , Fibrinolisina/metabolismo , Modelos Biológicos , Animais , Humanos
6.
Biochim Biophys Acta ; 1803(1): 103-20, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19800930

RESUMO

A number of extensive reviews are available discussing the roles of MMPs in various aspects of cancer progression from benign tumor formation to overt cancer present with deadly metastases. This review will focus specifically on the evidence functionally linking the MMPs and tumor-induced angiogenesis in various in vivo models. Emphasis has been placed on the cellular origin of the MMPs in tumor tissue, the requirement of proMMP activation and the resulting proteolytic activity for the induction and progression of tumor angiogenesis, and the pleiotropic roles for some of the MMPs. The functional mechanisms of the angiogenic MMPs are discussed as well as their catalytic detection in complex biological systems. In addition, the contribution of active MMPs to metastatic spread and establishment of secondary metastasis will be discussed in view of the findings indicating that MMPs are involved in the preparation of pre-metastatic niches. Finally, the most recent evidence, indicating the pro-metastatic consequences of anti-angiogenic therapies employing MMP inhibitors will be presented as examples highlighting possible outcomes of interfering with the pleiotropic nature of the MMP functionality.


Assuntos
Metaloproteinases da Matriz/metabolismo , Neoplasias/irrigação sanguínea , Neoplasias/enzimologia , Neovascularização Patológica/enzimologia , Animais , Ativação Enzimática , Humanos , Neoplasias/patologia , Neovascularização Patológica/terapia , Processamento de Proteína Pós-Traducional , Células-Tronco/citologia
7.
J Biol Chem ; 284(38): 25854-66, 2009 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-19608737

RESUMO

The structural and catalytic requirements for neutrophil MMP-9 proenzyme (proMMP-9) to induce angiogenesis were investigated using a quantitative angiogenesis model based on grafting of collagen onplants onto the chorioallantoic membrane of chick embryos. Both physiological activation of neutrophil proMMP-9 and proteolytic activity of the generated MMP-9 enzyme were critically dependent on the tissue inhibitor of metalloproteinase (TIMP)-free status of the zymogen. The presence of an intact active site and hemopexin domain were required for full angiogenesis-inducing activity of the MMP-9 enzyme. Timed additions of TIMP-1 to the onplants containing TIMP-free neutrophil proMMP-9 indicated that in vivo activation of the zymogen occurred during the first 24 h after grafting. Within the onplant tissue, MMP-9 activation was accompanied by proteolytic modifications of fibrillar collagen and an influx of host proteins, the rate of which depended on the TIMP-free status of the zymogen. By quantifying the levels of host angiogenic factors, we demonstrated that basic fibroblast growth factor (FGF-2) was a major cytokine becoming bioavailable in the onplant tissue undergoing a neutrophil proMMP-9-mediated angiogenic switch. Inhibition of angiogenesis with specific function-blocking antibodies further indicated an involvement of a FGF-2/FGFR-2 pathway in neutrophil proMMP-9-induced angiogenesis. The enhanced angiogenesis catalyzed by neutrophil MMP-9 appears to evoke also a localized, low threshold level vascular endothelial growth factor (VEGF)/VEGFR-2 pathway, likely functioning in the formation and/or stabilization of blood vessels. That neutrophil proMMP-9, unencumbered by TIMP-1, directly mediates FGF-2-dependent angiogenesis was also demonstrated in our quantitative mouse angiogenesis model employing subcutaneous collagen implants, thus implicating the novel TIMP-free MMP-9/FGF-2/FGFR-2 pathway in proMMP-9-induced angiogenesis in a mammalian setting.


Assuntos
Precursores Enzimáticos/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Neovascularização Fisiológica/fisiologia , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais/fisiologia , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Animais , Embrião de Galinha , Precursores Enzimáticos/genética , Precursores Enzimáticos/farmacologia , Fator 2 de Crescimento de Fibroblastos/genética , Humanos , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/farmacologia , Camundongos , Neovascularização Fisiológica/efeitos dos fármacos , Neutrófilos/enzimologia , Estrutura Terciária de Proteína/fisiologia , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Transdução de Sinais/efeitos dos fármacos , Inibidor Tecidual de Metaloproteinase-1/genética
8.
Am J Pathol ; 175(4): 1638-52, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19729488

RESUMO

To analyze the process of tumor cell intravasation, we used the human tumor-chick embryo spontaneous metastasis model to select in vivo high (PC-hi/diss) and low (PC-lo/diss) disseminating variants from the human PC-3 prostate carcinoma cell line. These variants dramatically differed in their intravasation and dissemination capacities in both chick embryo and mouse spontaneous metastasis models. Concomitant with enhanced intravasation, PC-hi/diss exhibited increased angiogenic potential in avian and murine models. PC-hi/diss angiogenesis and intravasation were dependent on increased secretion of vascular endothelial growth factor (VEGF), since treating developing tumors with a function-blocking anti-VEGF antibody simultaneously inhibited both processes without affecting primary tumor growth. PC-hi/diss cells were also more migratory and invasive, suggestive of heightened ability to escape from primary tumors due to matrix-degrading activity. Consistent with this suggestion, PC-hi/diss cells produced more of the serine protease urokinase-type plasminogen activator (uPA) as compared with PC-lo/diss. The functional role of uPA in PC-hi/diss dissemination was confirmed by inhibition of invasion, angiogenesis, and intravasation with specific function-blocking antibodies that prevented uPA activation and blocked uPA activity. These processes were similarly sensitive to aprotinin, a potent inhibitor of serine proteases, including uPA-generated plasmin. Thus, our comparison of the PC-3 intravasation variants points to key roles for the uPA-plasmin system in PC-hi/diss intravasation, possibly via (1) promoting tumor cell matrix invasion and (2) facilitating development of VEGF-dependent angiogenic blood vessels.


Assuntos
Neovascularização Patológica/patologia , Neoplasias da Próstata/irrigação sanguínea , Neoplasias da Próstata/patologia , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Adesão Celular , Linhagem Celular Tumoral , Movimento Celular , Embrião de Galinha , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Invasividade Neoplásica , Metástase Neoplásica , Fator A de Crescimento do Endotélio Vascular/biossíntese
9.
Proc Natl Acad Sci U S A ; 104(51): 20262-7, 2007 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-18077379

RESUMO

Several lines of evidence have implicated matrix metalloproteinase 9 (MMP-9) as a protease inducing an angiogenic switch critical for tumor progression. Among MMP-9-expressing cell types, including cancer cells and tumor-associated leukocytes, inflammatory neutrophils appear to provide an important source of MMP-9 for tumor angiogenesis. However, delivery of MMP-9 by neutrophils has not been mechanistically linked to its catalytic activity at the angiogenic site. By using a modified angiogenic model, allowing for a direct analysis of exogenously added cells and their products in collagen onplants grafted on the chorioallantoic membrane of the chicken embryo, we demonstrate that intact human neutrophils and their granule contents are highly angiogenic. Furthermore, purified neutrophil MMP-9, isolated from the released granules as a zymogen (proMMP-9), constitutes a distinctly potent proangiogenic moiety inducing angiogenesis at subnanogram levels. The angiogenic response induced by neutrophil proMMP-9 required activation of the tissue inhibitor of metalloproteinases (TIMP)-free zymogen and the catalytic activity of the activated enzyme. That the high angiogenic potency of neutrophil proMMP-9 is associated with its unique TIMP-free status was confirmed when a generated and purified stoichiometric complex of neutrophil proMMP-9 with TIMP-1 failed to induce angiogenesis. Recombinant human proMMP-9, operationally free of TIMP-1, also induced angiogenesis at subnanomolar levels, but lost its proangiogenic potential when stoichiometrically complexed with TIMP-1. Similar proMMP-9/TIMP-1 complexes, but naturally produced by human monocytic U937 cells and HT-1080 fibrosarcoma cells, did not stimulate angiogenesis. These findings provide biochemical evidence that infiltrating neutrophils, in contrast to other cell types, deliver a potent proangiogenic moiety, i.e., the unencumbered TIMP-free MMP-9.


Assuntos
Metaloproteinase 9 da Matriz/metabolismo , Neovascularização Patológica/enzimologia , Neutrófilos/enzimologia , Animais , Anticorpos Monoclonais/farmacologia , Linhagem Celular Tumoral , Embrião de Galinha , Precursores Enzimáticos/genética , Precursores Enzimáticos/metabolismo , Precursores Enzimáticos/farmacologia , Humanos , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/farmacologia , Inibidores de Metaloproteinases de Matriz , Neovascularização Fisiológica , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia , Inibidor Tecidual de Metaloproteinase-1/metabolismo
10.
Theranostics ; 10(9): 4116-4133, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32226543

RESUMO

Background: CUB domain-containing protein 1 (CDCP1) is a cell surface receptor regulating key signalling pathways in malignant cells. CDCP1 has been proposed as a molecular target to abrogate oncogenic signalling pathways and specifically deliver anti-cancer agents to tumors. However, the development of CDCP1-targeting agents has been questioned by its frequent proteolytic processing which was thought to result in shedding of the CDCP1 extracellular domain limiting its targetability. In this study, we investigated the relevance of targeting CDCP1 in the context of pancreatic ductal adenocarcinoma (PDAC) and assess the impact of CDCP1 proteolysis on the effectiveness of CDCP1 targeting agents. Methods: The involvement of CDCP1 in PDAC progression was assessed by association analysis in several PDAC cohorts and the proteolytic processing of CDCP1 was evaluated in PDAC cell lines and patient-derived cells. The consequences of CDCP1 proteolysis on its targetability in PDAC cells was assessed using immunoprecipitation, immunostaining and biochemical assays. The involvement of CDCP1 in PDAC progression was examined by loss-of-function in vitro and in vivo experiments employing PDAC cells expressing intact or cleaved CDCP1. Finally, we generated antibody-based imaging and therapeutic agents targeting CDCP1 to demonstrate the feasibility of targeting this receptor for detection and treatment of PDAC tumors. Results: High CDCP1 expression in PDAC is significantly associated with poorer patient survival. In PDAC cells proteolysis of CDCP1 does not always result in the shedding of CDCP1-extracellular domain which can interact with membrane-bound CDCP1 allowing signal transduction between the different CDCP1-fragments. Targeting CDCP1 impairs PDAC cell functions and PDAC tumor growth independently of CDCP1 cleavage status. A CDCP1-targeting antibody is highly effective at delivering imaging radionuclides and cytotoxins to PDAC cells allowing specific detection of tumors by PET/CT imaging and superior anti-tumor effects compared to gemcitabine in in vivo models. Conclusion: Independent of its cleavage status, CDCP1 exerts oncogenic functions in PDAC and has significant potential to be targeted for improved radiological staging and treatment of this cancer. Its elevated expression by most PDAC tumors and lack of expression by normal pancreas and other major organs, suggest that targeting CDCP1 could benefit a significant proportion of PDAC patients. These data support the further development of CDCP1-targeting agents as personalizable tools for effective imaging and treatment of PDAC.


Assuntos
Antígenos de Neoplasias/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Moléculas de Adesão Celular/metabolismo , Neoplasias Pancreáticas/metabolismo , Animais , Biomarcadores Tumorais/metabolismo , Carcinoma Ductal Pancreático/terapia , Linhagem Celular Tumoral , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Neoplasias Pancreáticas/terapia , Medicina de Precisão , Proteólise
11.
IUBMB Life ; 61(7): 723-30, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19514048

RESUMO

In the last few years dysregulated expression of the cell surface glycoprotein CUB domain-containing protein 1 (CDCP1) has been associated with several cancers and this cell surface molecule has been recognized both as a tumor marker and as a potential target to disrupt progression of cancer. Here we summarize what is known about CDCP1 including its structural features, expression in normal and cancerous tissues, and the in vitro experiments and studies in animal models that have provided the key insights into its potential role in tumor formation and metastasis in humans. We conclude by highlighting opportunities and challenges in targeting CDCP1 in cancer.


Assuntos
Antígenos CD/fisiologia , Biomarcadores Tumorais/fisiologia , Moléculas de Adesão Celular/fisiologia , Proteínas de Neoplasias/fisiologia , Neoplasias/fisiopatologia , Sequência de Aminoácidos , Antígenos de Neoplasias , Progressão da Doença , Células-Tronco Hematopoéticas/fisiologia , Dados de Sequência Molecular , Neoplasias/tratamento farmacológico , Fosforilação , Alinhamento de Sequência , Células-Tronco/fisiologia , Quinases da Família src/fisiologia
12.
J Cell Biol ; 160(2): 267-77, 2003 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-12527751

RESUMO

Invasive tumor dissemination in vitro and in vivo involves the proteolytic degradation of ECM barriers. This process, however, is only incompletely attenuated by protease inhibitor-based treatment, suggesting the existence of migratory compensation strategies. In three-dimensional collagen matrices, spindle-shaped proteolytically potent HT-1080 fibrosarcoma and MDA-MB-231 carcinoma cells exhibited a constitutive mesenchymal-type movement including the coclustering of beta 1 integrins and MT1-matrix metalloproteinase (MMP) at fiber bindings sites and the generation of tube-like proteolytic degradation tracks. Near-total inhibition of MMPs, serine proteases, cathepsins, and other proteases, however, induced a conversion toward spherical morphology at near undiminished migration rates. Sustained protease-independent migration resulted from a flexible amoeba-like shape change, i.e., propulsive squeezing through preexisting matrix gaps and formation of constriction rings in the absence of matrix degradation, concomitant loss of clustered beta 1 integrins and MT1-MMP from fiber binding sites, and a diffuse cortical distribution of the actin cytoskeleton. Acquisition of protease-independent amoeboid dissemination was confirmed for HT-1080 cells injected into the mouse dermis monitored by intravital multiphoton microscopy. In conclusion, the transition from proteolytic mesenchymal toward nonproteolytic amoeboid movement highlights a supramolecular plasticity mechanism in cell migration and further represents a putative escape mechanism in tumor cell dissemination after abrogation of pericellular proteolysis.


Assuntos
Movimento Celular/fisiologia , Proteínas da Matriz Extracelular/metabolismo , Mesoderma/metabolismo , Metástase Neoplásica/fisiopatologia , Neoplasias/metabolismo , Actinas/metabolismo , Amoeba/citologia , Amoeba/metabolismo , Animais , Tamanho Celular/fisiologia , Colágeno/metabolismo , Derme/citologia , Derme/metabolismo , Endopeptidases/efeitos dos fármacos , Endopeptidases/metabolismo , Feminino , Humanos , Integrina beta1/metabolismo , Metaloproteinase 14 da Matriz , Metaloproteinases da Matriz Associadas à Membrana , Mesoderma/citologia , Mesoderma/transplante , Metaloendopeptidases/metabolismo , Camundongos , Peptídeo Hidrolases/metabolismo , Inibidores de Proteases/farmacologia , Transplante de Tecidos , Células Tumorais Cultivadas
13.
Oncogene ; 37(14): 1815-1829, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29348457

RESUMO

Latent transforming growth factor ß (TGFß)-binding proteins (LTBPs) are important for the secretion, activation, and function of mature TGFß, especially so in cancer cell physiology. However, specific roles of the LTBPs remain understudied in the context of the primary tumor microenvironment. Herein, we investigated the role of LTBP3 in the distinct processes involved in cancer metastasis. By using three human tumor cell lines of different tissue origin (epidermoid HEp-3 and prostate PC-3 carcinomas and HT-1080 fibrosarcoma) and several metastasis models conducted in both mammalian and avian settings, we show that LTBP3 is involved in the early dissemination of primary cancer cells, namely in the intravasation step of the metastatic cascade. Knockdown of LTBP3 in all tested cell lines led to significant inhibition of tumor cell intravasation, but did not affect primary tumor growth. LTBP3 was dispensable in the late steps of carcinoma cell metastasis that follow tumor cell intravasation, including vascular arrest, extravasation, and tissue colonization. However, LTBP3 depletion diminished the angiogenesis-inducing potential of HEp-3 cells in vivo, which was restorable by exogenous delivery of LTBP3 protein. A similar compensatory approach rescued the dampened intravasation of LTBP3-deficient HEp-3 cells, suggesting that LTBP3 regulates the induction of the intravasation-supporting angiogenic vasculature within developing primary tumors. Using our recently developed microtumor model, we confirmed that LTBP3 loss resulted in the development of intratumoral vessels with an abnormal microarchitecture incompatible with efficient intravasation of HEp-3 carcinoma cells. Collectively, these findings demonstrate that LTBP3 represents a novel oncotarget that has distinctive functions in the regulation of angiogenesis-dependent tumor cell intravasation, a critical process during early cancer dissemination. Our experimental data are also consistent with the survival prognostic value of LTBP3 expression in early-stage head and neck squamous cell carcinomas, further indicating a specific role for LTBP3 in cancer progression toward metastatic disease.


Assuntos
Proteínas de Ligação a TGF-beta Latente/fisiologia , Neoplasias/genética , Neoplasias/patologia , Animais , Linhagem Celular Tumoral , Embrião de Galinha , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas de Ligação a TGF-beta Latente/antagonistas & inibidores , Proteínas de Ligação a TGF-beta Latente/genética , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Invasividade Neoplásica , Metástase Neoplásica , Neoplasias/mortalidade , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , RNA Interferente Pequeno/farmacologia , Análise de Sobrevida
15.
Cancer Res ; 65(23): 10959-69, 2005 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-16322244

RESUMO

The human tumor/chick embryo model involving grafting of human HT-1080 fibrosarcoma cells on the chorioallantoic membrane was used in conjunction with quantitative real-time Alu PCR to select in vivo a pair of isogenic cell lines (HT-hi/diss and HT-lo/diss), dramatically differing in their ability to disseminate from the primary tumor (i.e., intravasate into the chorioallantoic membrane vasculature and metastasize to the lungs). During an immunohistochemical time course study, HT-hi/diss cells were sequentially visualized having escaped from the primary tumors, engaged with the blood vessels, and eventually observed inside the chorioallantoic membrane capillaries, thus reflecting early intravasating events. In contrast, HT-lo/diss cells seemed restricted to their primary tumor. Importantly, after i.v. inoculation, both variants arrested, extravasated, and proliferated in host tissues with similar efficiencies, highlighting that the observed earlier events at the periphery of the primary tumor could account for their differential dissemination. In a mechanistic probing of these events, we determined that HT-hi/diss intravasation was sensitive to a broad-range matrix metalloproteinase (MMP) inhibitor. To analyze the possible role of individual MMPs, membrane-bound MMP-14 and secreted MMP-9 were individually down-regulated in HT-hi/diss cells with their corresponding small interfering RNAs. Despite efficient down-regulation of MMP-14, neither intravasation nor metastasis of HT-hi/diss cells was affected significantly. However, a substantial down-regulation of MMP-9 was accompanied by a surprising 3-fold increase in intravasation and metastasis. The results emphasize a rising awareness that targeting certain MMPs might result in an enhanced malignancy, exemplified herein at the intravasation level as this step of the metastatic cascade is dissected and quantified.


Assuntos
Fibrossarcoma/irrigação sanguínea , Fibrossarcoma/enzimologia , Metaloproteinase 9 da Matriz/metabolismo , Metaloproteinases da Matriz/metabolismo , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Linhagem Celular Tumoral , Embrião de Galinha , Membrana Corioalantoide/patologia , Regulação para Baixo , Fibrossarcoma/patologia , Humanos , Imuno-Histoquímica , Neoplasias Pulmonares/secundário , Metaloproteinase 9 da Matriz/biossíntese , Metaloproteinase 9 da Matriz/genética , Inibidores de Metaloproteinases de Matriz , Metaloproteinases da Matriz/biossíntese , Metaloproteinases da Matriz/imunologia , Metaloproteinases da Matriz Associadas à Membrana , Neovascularização Patológica/enzimologia , Neovascularização Patológica/patologia , RNA Interferente Pequeno/genética
16.
Cell Rep ; 19(3): 601-616, 2017 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-28423322

RESUMO

Intravasation, active entry of cancer cells into the circulation, is often considered to be a relatively late event in tumor development occurring after stromal invasion. Here, we provide evidence that intravasation can be initiated early during tumor development and proceed in parallel to or independent of tumor invasion into surrounding stroma. By applying direct and unbiased intravasation-scoring methods to two histologically distinct human cancer types in live-animal models, we demonstrate that intravasation takes place almost exclusively within the tumor core, involves intratumoral vasculature, and does not involve vasculotropic cancer cells invading tumor-adjacent stroma and migrating along tumor-converging blood vessels. Highlighting an additional role for EGFR in cancer, we find that EGFR is required for the development of an intravasation-sustaining intratumoral vasculature. Intratumoral localization of intravasation supports the notion that overt metastases in cancer patients could be initiated much earlier during cancer progression than appreciated within conventional clinical tumor staging systems.


Assuntos
Movimento Celular , Neoplasias/patologia , Animais , Linhagem Celular Tumoral , Embrião de Galinha , Modelos Animais de Doenças , Orelha/patologia , Receptores ErbB/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Humanos , Hipóxia/patologia , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Invasividade Neoplásica , Metástase Neoplásica , Neovascularização Patológica , Permeabilidade , Células Estromais/patologia
17.
Cancer Res ; 62(2): 580-8, 2002 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-11809713

RESUMO

Membrane-type (MT) 1 matrix metalloproteinase (MMP) is up-regulated in many tumor types and has been implicated in tumor progression and metastasis. MT1-MMP is critical for pericellular degradation of the extracellular matrix, thereby promoting tumor cell invasion and dissemination. To grow efficiently in vivo, tumor cells induce angiogenesis in both primary solid tumors and metastatic foci. The present study describes a functional link between the expression of MT1-MMP and vascular endothelial growth factor (VEGF) production in human glioma U251 xenografts in athymic mice. To investigate the effects of MT1-MMP on VEGF expression, U251 cells were stably transfected with MT1-MMP to generate the U-MT cell line overexpressing the enzyme. In vitro, the U-MT cells had an increased rate of proliferation and migration as well as the ability to activate the MMP-2 proenzyme and directionally remodel a three-dimensional collagen matrix. These findings suggested higher tumorigenicity of U-MT cells relative to the vector-control U-neo cells. In agreement with the in vitro data, U-MT xenografts in BALB/c nu/nu mice displayed markedly increased growth rates and elevated levels of angiogenesis. In contrast, U-neo cells formed small, minimally vascularized tumors. The elevated angiogenesis in U-MT xenografts was associated with an up-regulation of VEGF expression in tumor cells. In addition, U-MT cells in vitro secreted twice as much VEGF as the control cells. GM6001, a hydroxamate inhibitor of MMP activity, down-regulated the production of VEGF in U-MT cells to the levels observed in the U-neo control. Our results demonstrate that the enhanced tumorigenicity of glioma cells overexpressing MT1-MMP involves stimulation of angiogenesis through the up-regulation of VEGF production.


Assuntos
Fatores de Crescimento Endotelial/biossíntese , Glioma/metabolismo , Linfocinas/biossíntese , Metaloendopeptidases/fisiologia , Neovascularização Patológica/metabolismo , Animais , Divisão Celular/fisiologia , Movimento Celular/fisiologia , Colágeno/metabolismo , Fatores de Crescimento Endotelial/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Glioma/irrigação sanguínea , Glioma/genética , Glioma/patologia , Humanos , Linfocinas/genética , Metaloproteinase 14 da Matriz , Metaloproteinases da Matriz Associadas à Membrana , Metaloendopeptidases/biossíntese , Metaloendopeptidases/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Transplante Heterólogo , Células Tumorais Cultivadas , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
18.
Methods Mol Biol ; 1430: 283-98, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27172961

RESUMO

The mechanisms governing the development of angiogenic blood vessels, which not only deliver the nutrients to growing tumors but also provide the conduits for tumor cell dissemination, are still not fully resolved. The model systems based on the grafting of human tumor cells onto the chorioallantoic membrane (CAM) of the chick embryo offer several advantages to study complex processes underlying tumor angiogenesis and tumor cell dissemination. In particular, the CAM model described here allows for investigation of multiple microtumors as independent entities, thereby greatly facilitating quantification and statistical analyses of tumor neovascularization and cancer spreading. This CAM microtumor system was designed specifically to measure the level of tumor cell intravasation in combination with quantitative analyses of the microarchitecture and permeability of the intratumoral angiogenic blood vessels. By using this newly established microtumor model we have demonstrated the functional involvement of tumor matrix metalloproteinase-1 (MMP-1) and epidermal growth factor receptor (EGFR) in regulating the development of a distinct angiogenic vasculature capable of sustaining tumor cell intravasation and metastasis.


Assuntos
Membrana Corioalantoide/irrigação sanguínea , Membrana Corioalantoide/patologia , Neovascularização Patológica/metabolismo , Animais , Permeabilidade Capilar , Linhagem Celular Tumoral , Embrião de Galinha , Membrana Corioalantoide/metabolismo , Receptores ErbB/metabolismo , Humanos , Metaloproteinase 1 da Matriz/metabolismo , Invasividade Neoplásica , Transplante de Neoplasias
19.
Matrix Biol ; 44-46: 94-112, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25912949

RESUMO

Metastasis is a distinct stage of cancer progression that requires the development of angiogenic blood vessels serving as conduits for tumor cell dissemination. An accumulated body of evidence indicates that metastasis-supporting neovasculature should possess certain structural characteristics allowing for the process of tumor cell intravasation, an active entry of cancer cells into the vessel interior. It appears that the development of tumor vessels with lumens of a distinctive size and support of these vessels by a discontinuous pericyte coverage constitute critical microarchitectural requirements to: (a) provide accessible points for vessel wall penetration by primary tumor cells; (b) provide enough lumen space for a tumor cell or cell aggregate upon intravasation; and (c) allow for sufficient rate of blood flow to carry away intravasated cells from the primary tumor to the next, proximal or distal site. This review will primarily focus on the functional roles of matrix metalloproteinases (MMPs), which catalytically trigger the development of an intravasation-sustaining neovasculature at the early stages of tumor growth and are also required for the maintenance of a metastasis-supporting state of blood vessels at later stages of cancer progression.


Assuntos
Metaloproteinases da Matriz/metabolismo , Neoplasias/patologia , Neovascularização Patológica/enzimologia , Animais , Humanos , Metástase Neoplásica , Neoplasias/irrigação sanguínea , Neoplasias/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
20.
Neoplasia ; 17(8): 634-49, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26408256

RESUMO

Many malignant characteristics of cancer cells are regulated through pathways induced by the tyrosine kinase activity of the epidermal growth factor receptor (EGFR). Herein, we show that besides directly affecting the biology of cancer cells per se, EGFR also regulates the primary tumor microenvironment. Specifically, our findings demonstrate that both the expression and signaling activity of EGFR are required for the induction of a distinct intratumoral vasculature capable of sustaining tumor cell intravasation, a critical rate-limiting step in the metastatic cascade. An intravasation-sustaining mode of intratumoral angiogenic vessels depends on high levels of tumor cell EGFR and the interplay between EGFR-regulated production of interleukin 8 by tumor cells, interleukin-8-induced influx of tumor-infiltrating neutrophils delivering their unique matrix metalloproteinase-9, and neutrophil matrix metalloproteinase-9-dependent release of the vascular permeability and endothelial growth factor, VEGF. Our data indicate that through VEGF-mediated disruption of endothelial layer integrity and increase of intratumoral vasculature permeability, EGFR activity significantly facilitates active intravasation of cancer cells. Therefore, this study unraveled an important but overlooked function of EGFR in cancer, namely, its ability to create an intravasation-sustaining microenvironment within the developing primary tumor by orchestrating several interrelated processes required for the initial steps of cancer metastasis through vascular routes. Our findings also suggest that EGFR-targeted therapies might be more effective when implemented in cancer patients with early-staged primary tumors containing a VEGF-dependent angiogenic vasculature. Accordingly, early EGFR inhibition combined with various anti-VEGF approaches could synergistically suppress tumor cell intravasation through inhibiting the highly permeable angiogenic vasculature induced by EGFR-overexpressing aggressive cancer cells.


Assuntos
Membrana Corioalantoide/irrigação sanguínea , Receptores ErbB/metabolismo , Neoplasias/metabolismo , Neovascularização Patológica/metabolismo , Animais , Western Blotting , Linhagem Celular Tumoral , Embrião de Galinha , Membrana Corioalantoide/metabolismo , Membrana Corioalantoide/patologia , Receptores ErbB/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Metaloproteinase 9 da Matriz/metabolismo , Microscopia de Fluorescência , Invasividade Neoplásica , Metástase Neoplásica , Transplante de Neoplasias , Neoplasias/irrigação sanguínea , Neoplasias/genética , Neovascularização Patológica/genética , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Microambiente Tumoral/genética , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa