Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neoplasia ; 16(10): 771-88, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25379015

RESUMO

According to established notion, one of the major angiogenesis-inducing factors, pro-matrix metalloproteinase-9 (proMMP-9), is supplied to the tumor microenvironment by tumor-associated macrophages (TAMs). Accumulated evidence, however, indicates that tumor-associated neutrophils (TANs) are also critically important for proMMP-9 delivery, especially at early stages of tumor development. To clarify how much angiogenic proMMP-9 is actually contributed by TAMs and TANs, we quantitatively evaluated TAMs and TANs from different tumor types, including human xenografts and syngeneic murine tumors grown in wild-type and Mmp9-knockout mice. Whereas host MMP-9 competence was required for full angiogenic potential of both normal and tumor-associated leukocytes, direct comparisons of neutrophils versus macrophages and TANs versus TAMs demonstrated that macrophages and TAMs secrete 40- to 50-fold less proMMP-9 than the same numbers of neutrophils or TANs. Correspondingly, the levels of MMP-9-mediated in vivo angiogenesis induced by neutrophils and TANs substantially exceeded those induced by macrophages and TAMs. MMP-9-delivering TANs were also required for development of metastasis-supporting intratumoral vasculature, characterized by ≥ 11-µm size lumens and partial coverage with stabilizing pericytes. Importantly, MMP-9-producing TAMs exhibit M2-skewed phenotype but do not express tissue inhibitor of metalloproteinases-1 (TIMP-1), a novel characteristic allowing them to secrete TIMP-1-free, neutrophil-like MMP-9 zymogen unencumbered by its natural inhibitor. Together, our findings support the notion whereby TANs, capable of immediate release of their pre-stored cargo, are the major contributors of highly angiogenic MMP-9, whereas tumor-influxing precursors of macrophages require time to differentiate, polarize into M2-skewed TAMs, shut down their TIMP-1 expression, and only then, initiate relatively low-level production of TIMP-free MMP-9 zymogen.


Assuntos
Indutores da Angiogênese/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Neutrófilos/patologia , Microambiente Tumoral/fisiologia , Animais , Linhagem Celular Tumoral , Humanos , Macrófagos/enzimologia , Macrófagos/patologia , Masculino , Metaloproteinase 9 da Matriz/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Patológica , Neutrófilos/metabolismo , Neoplasias da Próstata/irrigação sanguínea , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/patologia , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
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.
Cancer Res ; 73(14): 4196-211, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23687338

RESUMO

Intravasation, the active entry of primary tumor cells into the vasculature, remains the least studied step in the metastatic cascade. Protease-mediated escape and stromal invasion of tumor cells represent widely accepted processes leading up to the intravasation step. However, molecular factors that contribute directly to tumor cell vascular penetration have not been identified. In this study, the in vivo role of the collagenolytic protease, MMP-1, in cancer cell intravasation and metastasis was analyzed by using a highly disseminating variant of human HEp3 epidermoid carcinoma, HEp3-hi/diss. Although naturally acquired or experimentally induced MMP-1 deficiency substantially suppressed HEp3-hi/diss intravasation, supplementation of recombinant MMP-1 to MMP-1-silenced primary tumors restored their impaired vascular dissemination. Surprisingly, abrogation of MMP-1 production and activity did not significantly affect HEp3-hi/diss migration or matrix invasion, suggesting noncollagenolytic mechanisms underlying MMP-1-dependent cell intravasation. In support of such noncollagenolytic mechanisms, MMP-1 silencing in HEp3-hi/diss cells modulated the microarchitecture and integrity of the angiogenic vasculature in a novel microtumor model. Concomitantly, MMP-1 deficiency led to decreased levels of intratumoral vascular permeability, tumor cell intravasation, and metastatic dissemination. Taking advantage of PAR1 deficiency of HEp3-hi/diss cells, we further show that endothelial PAR1 is a putative nontumor-cell/nonmatrix target, activation of which by carcinoma-produced MMP-1 regulates endothelial permeability and transendothelial migration. The inhibitory effects of specific PAR1 antagonists in live animals have also indicated that the mechanisms of MMP-1-dependent vascular permeability in tumors involve endothelial PAR1 activation. Together, our findings mechanistically underscore the contribution of a tumor MMP-1/endothelial PAR1 axis to actual intravasation events manifested by aggressive carcinoma cells.


Assuntos
Carcinoma de Células Escamosas/enzimologia , Metaloproteinase 1 da Matriz/metabolismo , Receptor PAR-1/metabolismo , Animais , Permeabilidade Capilar , Carcinoma de Células Escamosas/irrigação sanguínea , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Embrião de Galinha , Endotélio/irrigação sanguínea , Endotélio/enzimologia , Endotélio/metabolismo , Endotélio/patologia , Humanos , Metaloproteinase 1 da Matriz/deficiência , Metaloproteinase 1 da Matriz/genética , Invasividade Neoplásica , Metástase Neoplásica , Neovascularização Patológica/enzimologia , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Receptor PAR-1/deficiência , Receptor PAR-1/genética
4.
Neoplasia ; 13(9): 806-21, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21969814

RESUMO

Urokinase-type plasminogen activator (uPA) and plasmin have long been implicated in cancer progression. However, the precise contributions of the uPA/plasmin system to specific steps involved in cancer cell dissemination have not been fully established. Herein, we have used a highly disseminating variant of the human PC-3 prostate carcinoma cell line, PC-hi/diss, as a prototype of aggressive carcinomas to investigate the mechanisms whereby pro-uPA activation and uPA-generated plasmin functionally contribute to specific stages of metastasis. The PC-hi/diss cells secrete and activate significant amounts of pro-uPA, leading to efficient generation of plasmin in solution and at the cell surface. In a mouse orthotopic xenograft model, treatment with the specific pro-uPA activation-blocking antibody mAb-112 significantly inhibited local invasion and distant metastasis of the PC-hi/diss cells. To mechanistically examine the uPA/plasmin-mediated aspects of tumor cell dissemination, the anti-pro-uPA mAb-112 and the potent serine protease inhibitor, aprotinin, were used in parallel in a number of in vivo assays modeling various rate-limiting steps in early metastatic spread. Our findings demonstrate that, by generating plasmin, activated tumor-derived uPA facilitates early stages of PC-hi/diss dissemination, specifically the escape from the primary tumor and tumor cell intravasation. Moreover, through a series of in vitro and in vivo analyses, we suggest that PC-hi/diss-invasive escape and dissemination may be enhanced by cleavage of stromal fibronectin by uPA-generated plasmin. Together, our findings point to inhibition of pro-uPA activation at the apex of the uPA/plasmin cascade as a therapy-valid approach to control onset of tumor escape and ensuing metastatic spread.


Assuntos
Fibrinolisina/metabolismo , Metástase Neoplásica , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Animais , Anticorpos Monoclonais/imunologia , Aprotinina/farmacologia , Linhagem Celular Tumoral , Movimento Celular , Embrião de Galinha , Ativação Enzimática , Fibronectinas/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Invasividade Neoplásica , Inibidores de Serina Proteinase/farmacologia , Transplante Heterólogo , Evasão Tumoral , Ativador de Plasminogênio Tipo Uroquinase/imunologia
5.
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
6.
Clin Exp Metastasis ; 26(8): 1033-47, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19842048

RESUMO

Increased metastatic and angiogenic potentials of aggressive human colon carcinoma cells were verified in independent chick embryo models by comparing in vivo highly metastatic SW620 colon carcinoma cell line with its isogenic, non-metastatic SW480 cell variant. In the experimental metastasis model, both cell types rapidly arrested in the chorioallantoic membrane (CAM) vasculature as demonstrated by quantitative PCR and immunohistochemistry. Live cell imaging also indicated that both SW620 and SW480 cells efficiently extravasated from the CAM capillary system. However, only few SW480 cells were present in the CAM tissue after 24-48 h. In contrast, the numbers of SW620 cells increased exponentially, indicating proliferative and survival advantages of metastatic colon carcinoma cells in vivo. Multicellular SW620 foci were identified in close proximity to CAM blood vessels. A positive correlation between increased metastatic ability and VEGF-expression of colon carcinoma SW620 cells was demonstrated by the substantial inhibitory effects of anti-VEGF treatment on the levels of metastatic colonization and density of blood vessels adjacent to tumor cell foci. Furthermore, the chick embryo angiogenesis model confirmed high levels of VEGF-dependent angiogenesis induced by SW620 cells, but not SW480 cells. Thus, chick embryo experimental metastasis and CAM angiogenesis models appear to coordinately reflect critical features of advanced colon carcinomas, i.e., the acquisition of enhanced survival and increased angiogenic potentials, both constituting critical determinants of colon cancer progression. The use of rapid and quantitative chick embryo models might provide alternative approaches to conventional mammalian model systems for screening anti-cancer agents.


Assuntos
Neoplasias do Colo/irrigação sanguínea , Neoplasias do Colo/patologia , Modelos Animais de Doenças , Metástase Neoplásica , Neovascularização Patológica , Animais , Linhagem Celular Tumoral , Embrião de Galinha , Membrana Corioalantoide/irrigação sanguínea , Humanos , Fator A de Crescimento do Endotélio Vascular/metabolismo
7.
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
8.
Mol Cancer Res ; 7(8): 1197-211, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19671673

RESUMO

The function of CUB domain-containing protein 1 (CDCP1), a recently described transmembrane protein expressed on the surface of hematopoietic stem cells and normal and malignant cells of different tissue origin, is not well defined. The contribution of CDCP1 to tumor metastasis was analyzed by using HeLa carcinoma cells overexpressing CDCP1 (HeLa-CDCP1) and a high-disseminating variant of prostate carcinoma PC-3 naturally expressing high levels of CDCP1 (PC3-hi/diss). CDCP1 expression rendered HeLa cells more aggressive in experimental metastasis in immunodeficient mice. Metastatic colonization by HeLa-CDCP1 was effectively inhibited with subtractive immunization-generated, CDCP1-specific monoclonal antibody (mAb) 41-2, suggesting that CDCP1 facilitates relatively late stages of the metastatic cascade. In the chick embryo model, time- and dose-dependent inhibition of HeLa-CDCP1 colonization by mAb 41-2 was analyzed quantitatively to determine when and where CDCP1 functions during metastasis. Quantitative PCR and immunohistochemical analyses indicated that CDCP1 facilitated tumor cell survival soon after vascular arrest. Live cell imaging showed that the function-blocking mechanism of mAb 41-2 involved enhancement of tumor cell apoptosis, confirmed by attenuation of mAb 41-2-mediated effects with the caspase inhibitor z-VAD-fmk. Under proapoptotic conditions in vitro, CDCP1 expression conferred HeLa-CDCP1 cells with resistance to doxorubicin-induced apoptosis, whereas ligation of CDCP1 with mAb 41-2 caused additional enhancement of the apoptotic response. The functional role of naturally expressed CDCP1 was shown by mAb 41-2-mediated inhibition of both experimental and spontaneous metastasis of PC3-hi/diss. These findings confirm that CDCP1 functions as an antiapoptotic molecule and indicate that during metastasis CDCP1 facilitates tumor cell survival likely during or soon after extravasation.


Assuntos
Antígenos CD/metabolismo , Moléculas de Adesão Celular/metabolismo , Membrana Celular/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Animais , Anticorpos Monoclonais/farmacologia , Antígenos de Neoplasias , Apoptose/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Embrião de Galinha , Modelos Animais de Doenças , Doxorrubicina/farmacologia , Células HeLa , Humanos , Cinética , Masculino , Camundongos , Metástase Neoplásica/patologia , Fosfotirosina/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Ensaio Tumoral de Célula-Tronco
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.
J Biol Chem ; 282(49): 35964-77, 2007 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-17895241

RESUMO

The role of tumor-derived matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinase (TIMPs) in cancer cell dissemination was analyzed by employing two variants of human HT-1080 fibrosarcoma, HT-hi/diss and HT-lo/diss, which differ by 50-100-fold in their ability to intravasate and metastasize in the chick embryo. HT-hi/diss and HT-lo/diss were compared by quantitative reverse transcription-PCR and Western blot analyses for mRNA and protein expression of nine MMPs (MMP-1, -2, -3, -7, -8, -9, -10, -13, and -14) and three TIMPs (TIMP-1, -2, and -3) in cultured cells in vitro and in primary tumors in vivo. MMP-1 and MMP-9 were more abundant in the HT-hi/diss variant, both in cultures and in tumors, whereas the HT-lo/diss variant consistently expressed higher levels of MMP-2, TIMP-1, and TIMP-2. Small interfering RNA-mediated down-regulation of MMP-2 and TIMP-2 increased intravasation of HT-lo/diss cells. Coordinately, treatment of the developing HT-hi/diss tumors with recombinant TIMP-1 and TIMP-2 significantly reduced HT-hi/diss cell intravasation. However, a substantial increase of HT-hi/diss dissemination was observed upon small interfering RNA-mediated down-regulation of three secreted MMPs, including the interstitial collagenase MMP-1 and the two gelatinases, MMP-2 and MMP-9, but not the membrane-tethered MMP-14. The addition of recombinant pro-MMP-9 protein to the HT-hi/diss tumors reversed the increased intravasation of HT-hi/diss cells, in which MMP-9 was stably down-regulated by short hairpin RNA interference. This rescue did not occur if the pro-MMP-9 was stoichiometrically complexed with TIMP-1, pointing to a direct role of the MMP-9 enzyme in regulation of HT-hi/diss intravasation. Collectively, these findings demonstrate that tumor-derived MMPs may have protective functions in cancer cell intravasation, i.e. not promoting but rather catalytically interfering with the early stages of cancer dissemination.


Assuntos
Fibrossarcoma/metabolismo , Regulação Neoplásica da Expressão Gênica , Metaloproteinases da Matriz/biossíntese , Proteínas de Neoplasias/biossíntese , Inibidores Teciduais de Metaloproteinases/biossíntese , Animais , Linhagem Celular Tumoral , Embrião de Galinha , Fibrossarcoma/tratamento farmacológico , Fibrossarcoma/patologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Inibidores de Metaloproteinases de Matriz , Invasividade Neoplásica , Metástase Neoplásica , Proteínas de Neoplasias/antagonistas & inibidores , Transplante de Neoplasias , RNA Interferente Pequeno/farmacologia , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/farmacologia , Inibidores Teciduais de Metaloproteinases/antagonistas & inibidores , Inibidores Teciduais de Metaloproteinases/farmacologia
11.
Blood ; 107(1): 317-27, 2006 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-16174763

RESUMO

A quantitative in vivo angiogenesis model employing collagen onplants placed on the chick embryo chorioallantoic membrane (CAM) has been used in this study to assess the spatial and temporal associations between neutrophil-like inflammatory cells, namely chicken heterophils, and the development of new blood vessels. Previously we have demonstrated that monocytes/macrophages infiltrating the onplants were associated with extracellular matrix remodeling and angiogenesis, in particular by delivering MMP-13 collagenase. By introducing chicken gelatinase B (chMMP-9) as a specific marker for heterophils, we now show that the onset and extent of angiogenesis induced by purified growth factors or by human HT-1080 fibrosarcoma cells correlated with the initial influx of chMMP-9-positive heterophils. This early heterophil arrival was followed by the infiltration of monocytes/macrophages and appeared to sustain further blood vessel formation. The disruption of inflammatory cell influx by 2 mechanistically distinct anti-inflammatory drugs, cortisone and ibuprofen, significantly inhibited angiogenesis, indicating a functional involvement of these inflammatory cells in new blood vessel development. A direct addition of isolated heterophils or purified chMMP-9 into the HT-1080 onplants engrafted into cortisone- or ibuprofen-treated embryos reversed the antiangiogenic effects of the drugs. The exogenously added heterophils induced in vivo a further infiltration of endogenous heterophils and monocytes and dramatically rescued the impaired angiogenesis, highlighting the importance of early inflammatory leukocytes in tumor-induced angiogenesis. Moreover, purified heterophils incorporated into onplants lacking growth factors or tumor cells induced angiogenesis in nontreated embryos, further indicating a direct proangiogenic role for neutrophil-like leukocytes.


Assuntos
Substâncias de Crescimento/farmacologia , Neovascularização Patológica/etiologia , Neutrófilos/fisiologia , Animais , Linhagem Celular Tumoral , Movimento Celular , Embrião de Galinha , Membrana Corioalantoide , Técnicas de Cocultura , Colágeno , Fibrossarcoma/patologia , Humanos , Inflamação/patologia , Leucócitos/citologia , Modelos Animais , Monócitos/citologia
12.
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
13.
J Biol Chem ; 277(11): 9133-8, 2002 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-11782457

RESUMO

In rat adipose cells, intracellular Glut4 resides in two distinct vesicular populations one of which contains cellugyrin whereas another lacks this protein (Kupriyanova, T. A., and Kandror, K. V. (2000) J. Biol. Chem. 275, 36263--36268). Cell surface biotinylated MPR and (125)I-labeled transferrin are accumulated in cellugyrin-positive vesicles and to a lesser extent in cellugyrin-negative vesicles. An average cellugyrin-positive vesicle carries not more than one molecule of either Glut4, insulin-responsive aminopeptidase (IRAP), or transferrin receptor (TfR), whereas cellugyrin-negative vesicles contain five to six molecules of Glut4, more than 10 molecules of IRAP, and still one molecule of TfR per vesicle. Cellugyrin-negative vesicles are translocated to the cell surface after insulin stimulation, whereas cellugyrin-positive vesicles maintain intracellular localization both in the absence and in the presence of insulin and, therefore, may be involved in interendosomal protein transport. Both cellugyrin-positive and cellugyrin-negative vesicles are present in extracts of non-homogenized cells and therefore may represent the major form of Glut4 storage in vivo.


Assuntos
Adipócitos/química , Proteínas de Transporte de Monossacarídeos/análise , Proteínas Musculares , Vesículas Transportadoras/química , Adipócitos/ultraestrutura , Animais , Membrana Celular/química , Transportador de Glucose Tipo 4 , Insulina/farmacologia , Masculino , Proteínas de Membrana/análise , Proteínas do Tecido Nervoso/análise , Fosfatidilinositol 3-Quinases/fisiologia , Ratos , Ratos Sprague-Dawley , Receptor IGF Tipo 2/análise , Receptores da Transferrina/análise , Sinaptogirinas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...