Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
J Biol Chem ; 292(50): 20769-20784, 2017 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-29046355

RESUMO

Matrix metalloproteinase-7 (MMP-7) plays important roles in tumor progression and metastasis. Our previous studies have demonstrated that MMP-7 binds to colon cancer cells via cell surface-bound cholesterol sulfate and induces significant cell aggregation by cleaving cell-surface protein(s). These aggregated cells exhibit a dramatically enhanced metastatic potential. However, the molecular mechanism inducing this cell-cell adhesion through the proteolytic action of MMP-7 remained to be clarified. Here, we explored MMP-7 substrates on the cell surface; the proteins on the cell surface were first biotinylated, and a labeled protein fragment specifically released from the cells after MMP-7 treatment was analyzed using LC-MS/MS. We found that hepatocyte growth factor activator inhibitor type 1 (HAI-1), a membrane-bound Kunitz-type serine protease inhibitor, is an MMP-7 substrate. We also found that the cell-bound MMP-7 cleaves HAI-1 mainly between Gly451 and Leu452 and thereby releases the extracellular region as soluble HAI-1 (sHAI-1). We further demonstrated that this sHAI-1 can induce cancer cell aggregation and determined that the HAI-1 region corresponding to amino acids 141-249, which does not include the serine protease inhibitor domain, has the cell aggregation-inducing activity. Interestingly, a cell-surface cholesterol sulfate-independent proteolytic action of MMP-7 is critical for the sHAI-1-mediated induction of cell aggregation, whereas cholesterol sulfate is needed for the MMP-7-catalyzed generation of sHAI-1. Considering that MMP-7-induced cancer cell aggregation is an important mechanism in cancer metastasis, we propose that sHAI-1 is an essential component of MMP-7-induced stimulation of cancer metastasis and may therefore represent a suitable target for antimetastatic therapeutic strategies.


Assuntos
Carcinoma/enzimologia , Neoplasias do Colo/enzimologia , Metaloproteinase 7 da Matriz/metabolismo , Proteínas de Neoplasias/metabolismo , Fragmentos de Peptídeos/metabolismo , Proteínas Secretadas Inibidoras de Proteinases/metabolismo , Substituição de Aminoácidos , Animais , Células CHO , Carcinoma/metabolismo , Carcinoma/patologia , Agregação Celular , Linhagem Celular Tumoral , Ésteres do Colesterol/química , Ésteres do Colesterol/metabolismo , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Cricetulus , Humanos , Metaloproteinase 7 da Matriz/química , Metaloproteinase 7 da Matriz/genética , Mutação , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Fragmentos de Peptídeos/antagonistas & inibidores , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Domínios e Motivos de Interação entre Proteínas , Proteínas Secretadas Inibidoras de Proteinases/antagonistas & inibidores , Proteínas Secretadas Inibidoras de Proteinases/química , Proteínas Secretadas Inibidoras de Proteinases/genética , Proteólise , Interferência de RNA , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Solubilidade , Especificidade por Substrato
2.
Plant J ; 84(4): 827-37, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26402640

RESUMO

Auxin is essential for plant growth and development, this makes it difficult to study the biological function of auxin using auxin-deficient mutants. Chemical genetics have the potential to overcome this difficulty by temporally reducing the auxin function using inhibitors. Recently, the indole-3-pyruvate (IPyA) pathway was suggested to be a major biosynthesis pathway in Arabidopsis thaliana L. for indole-3-acetic acid (IAA), the most common member of the auxin family. In this pathway, YUCCA, a flavin-containing monooxygenase (YUC), catalyzes the last step of conversion from IPyA to IAA. In this study, we screened effective inhibitors, 4-biphenylboronic acid (BBo) and 4-phenoxyphenylboronic acid (PPBo), which target YUC. These compounds inhibited the activity of recombinant YUC in vitro, reduced endogenous IAA content, and inhibited primary root elongation and lateral root formation in wild-type Arabidopsis seedlings. Co-treatment with IAA reduced the inhibitory effects. Kinetic studies of BBo and PPBo showed that they are competitive inhibitors of the substrate IPyA. Inhibition constants (Ki ) of BBo and PPBo were 67 and 56 nm, respectively. In addition, PPBo did not interfere with the auxin response of auxin-marker genes when it was co-treated with IAA, suggesting that PPBo is not an inhibitor of auxin sensing or signaling. We propose that these compounds are a class of auxin biosynthesis inhibitors that target YUC. These small molecules are powerful tools for the chemical genetic analysis of auxin function.


Assuntos
Proteínas de Arabidopsis/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Oxigenases/antagonistas & inibidores , Bibliotecas de Moléculas Pequenas/farmacologia , Arabidopsis/genética , Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Vias Biossintéticas/efeitos dos fármacos , Vias Biossintéticas/genética , Ácidos Borônicos/química , Ácidos Borônicos/farmacologia , Inibidores Enzimáticos/química , Regulação da Expressão Gênica de Plantas/efeitos dos fármacos , Ácidos Indolacéticos/metabolismo , Indóis/metabolismo , Indóis/farmacologia , Estrutura Molecular , Mutação , Oxigenases/genética , Oxigenases/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Plântula/genética , Plântula/metabolismo , Bibliotecas de Moléculas Pequenas/química
3.
Exp Cell Res ; 326(2): 267-79, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-24780821

RESUMO

Interaction between tumor cells and stromal fibroblasts plays essential roles in tumor progression. However, its detailed molecular mechanism remains unclear. To understand the mechanism, we investigated molecules mediating this interaction using the three-dimensional (3D) co-culture system of Panc-1 pancreatic carcinoma cells with normal fibroblasts. When the two kinds of cells were placed on the top of collagen gel, the tumor cells scattered into the fibroblast layer, apparently undergoing epithelial-mesenchymal transition. When fibroblasts were placed within collagen gel, Panc-1 cells actively invaded into the collagen gel, extending a microtubule-based long protrusion. Although transforming growth factor-ß (TGF-ß) and hepatocyte growth factor (HGF) individually stimulated the tumor cell invasion into collagen gel without fibroblasts, TGF-ß signaling inhibitors (SB431542 and LY2157299) significantly enhanced the Panc-1 cell invasion in the 3D co-culture with fibroblasts. Experiments with HGF/Met signaling inhibitors or with the fibroblast conditioned medium revealed that HGF was a major invasion-promoting factor secreted from fibroblasts and SB431542 increased the HGF secretion by blocking the HGF-suppressing activity of cancer cell-derived TGF-ß. These results indicate that HGF and TGF-ß are critical regulators for both tumor-stroma interaction and tumor invasion. The results also suggest that TGF-ß signaling inhibitors may promote tumor progression under some pathological conditions.


Assuntos
Fator de Crescimento de Hepatócito/metabolismo , Invasividade Neoplásica/fisiopatologia , Neoplasias Pancreáticas/metabolismo , Fator de Crescimento Transformador beta/antagonistas & inibidores , Benzamidas/farmacologia , Linhagem Celular , Linhagem Celular Tumoral , Técnicas de Cocultura , Colágeno/metabolismo , Dioxóis/farmacologia , Transição Epitelial-Mesenquimal , Fibroblastos/metabolismo , Fator de Crescimento de Hepatócito/antagonistas & inibidores , Humanos , Modelos Estatísticos , Invasividade Neoplásica/patologia , Neoplasias Pancreáticas/patologia , Transdução de Sinais , Células Estromais/metabolismo , Fator de Crescimento Transformador beta/metabolismo
4.
J Biol Chem ; 288(13): 9066-76, 2013 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-23395821

RESUMO

Synthetic inhibitors of matrix metalloproteinases (MMPs), designed previously, as well as tissue inhibitors of metalloproteinases (TIMPs) lack enzyme selectivity, which has been a major obstacle for developing inhibitors into safe and effective MMP-targeted drugs. Here we designed a fusion protein named APP-IP-TIMP-2, in which the ten amino acid residue sequence of APP-derived MMP-2 selective inhibitory peptide (APP-IP) is added to the N terminus of TIMP-2. The APP-IP and TIMP-2 regions of the fusion protein are designed to interact with the active site and the hemopexin-like domain of MMP-2, respectively. The reactive site of the TIMP-2 region, which has broad specificity against MMPs, is blocked by the APP-IP adduct. The recombinant APP-IP-TIMP-2 showed strong inhibitory activity toward MMP-2 (Ki(app) = 0.68 pm), whereas its inhibitory activity toward MMP-1, MMP-3, MMP-7, MMP-8, MMP-9, or MT1-MMP was six orders of magnitude or more weaker (IC50 > 1 µm). The fusion protein inhibited the activation of pro-MMP-2 in the concanavalin A-stimulated HT1080 cells, degradation of type IV collagen by the cells, and the migration of stimulated cells. Compared with the decapeptide APP-IP (t½ = 30 min), APP-IP-TIMP-2 (t½ ≫ 96 h) showed a much longer half-life in cultured tumor cells. Therefore, the fusion protein may be a useful tool to evaluate contributions of proteolytic activity of MMP-2 in various pathophysiological processes. It may also be developed as an effective anti-tumor drug with restricted side effects.


Assuntos
Inibidores Enzimáticos/farmacologia , Metaloproteinase 2 da Matriz/metabolismo , Inibidores de Metaloproteinases de Matriz/farmacologia , Amiloide/química , Sítios de Ligação , Linhagem Celular Tumoral , Colágeno Tipo IV/metabolismo , Concanavalina A/farmacologia , Desenho de Fármacos , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Fluoresceína-5-Isotiocianato/farmacologia , Regulação Neoplásica da Expressão Gênica , Vetores Genéticos , Humanos , Concentração Inibidora 50 , Cinética , Inibidores de Metaloproteinases de Matriz/química , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes/química , Fatores de Tempo
5.
Cancer Sci ; 105(2): 168-75, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24238220

RESUMO

Laminin γ2 (Lmγ2) chain, a subunit of laminin-332, is a typical molecular marker of invading cancer cells, and its expression correlates with poor prognosis of cancer patients. It was previously found that forced expression of Lmγ2 in cancer cells promotes their invasive growth in nude mice. However, the mechanism of the tumor-promoting activity of Lmγ2 remains unknown. Here we investigated the interaction between Lmγ2 and vascular endothelial cells. When treated with an N-terminal proteolytic fragment of γ2 (γ2pf), HUVECs became markedly retracted or shrunken. The overexpression of Lmγ2 or treatment with γ2pf stimulated T-24 bladder carcinoma cells to invade into the HUVEC monolayer and enhanced their transendothelial migration in vitro. Moreover, γ2pf increased endothelial permeability in vitro and in vivo. As the possible mechanisms, γ2pf activated ERK and p38 MAPK but inactivated Akt in HUVECs. Such effects of γ2pf led to prominent actin stress fiber formation in HUVECs, which was blocked by a ROCK inhibitor. In addition, γ2pf induced delocalization of VE-cadherin and ß-catenin from the intercellular junction. As possible receptors, γ2pf interacted with heparan sulfate proteoglycans on the surface of HUVECs. Moreover, we localized the active site of γ2pf to the N-terminal epidermal growth factor-like repeat. These data suggest that the interaction between γ2pf and heparan sulfate proteoglycans induces cytoskeletal changes of endothelial cells, leading to the loss of endothelial barrier function and the enhanced transendothelial migration of cancer cells. These activities of Lmγ2 seem to support the aberrant growth of cancer cells.


Assuntos
Permeabilidade Capilar/fisiologia , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Laminina/metabolismo , Animais , Antígenos CD/metabolismo , Caderinas/metabolismo , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Citoesqueleto/metabolismo , Citoesqueleto/patologia , Citoesqueleto/fisiologia , Células Endoteliais/fisiologia , Endotélio Vascular/patologia , Endotélio Vascular/fisiologia , Fator de Crescimento Epidérmico/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Junções Intercelulares/metabolismo , Junções Intercelulares/patologia , Junções Intercelulares/fisiologia , Laminina/farmacologia , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Recombinantes/farmacologia , Neoplasias da Bexiga Urinária/metabolismo , Neoplasias da Bexiga Urinária/patologia , beta Catenina/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
6.
J Biol Chem ; 286(38): 33236-43, 2011 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-21813640

RESUMO

Unlike other synthetic or physiological inhibitors for matrix metalloproteinases (MMPs), the ß-amyloid precursor protein-derived inhibitory peptide (APP-IP) having an ISYGNDALMP sequence has a high selectivity toward MMP-2. Our previous study identified amino acid residues of MMP-2 essential for its selective inhibition by APP-IP and demonstrated that the N to C direction of the decapeptide inhibitor relative to the substrate-binding cleft of MMP-2 is opposite that of substrate. However, detailed interactions between the two molecules remained to be clarified. Here, we determined the crystal structure of the catalytic domain of MMP-2 in complex with APP-IP. We found that APP-IP in the complex is indeed embedded into the substrate-binding cleft of the catalytic domain in the N to C direction opposite that of substrate. With the crystal structure, it was first clarified that the aromatic side chain of Tyr(3) of the inhibitor is accommodated into the S1' pocket of the protease, and the carboxylate group of Asp(6) of APP-IP coordinates bidentately to the catalytic zinc of the enzyme. The Ala(7) to Pro(10) and Tyr(3) to Ile(1) strands of the inhibitor extend into the nonprime and the prime sides of the cleft, respectively. Therefore, the decapeptide inhibitor has long range contact with the substrate-binding cleft of the protease. This mode of interaction is probably essential for the high MMP-2 selectivity of the inhibitor because MMPs share a common architecture in the vicinity of the catalytic center, but whole structures of their substrate-binding clefts have sufficient variety for the inhibitor to distinguish MMP-2 from other MMPs.


Assuntos
Metaloproteinase 2 da Matriz/química , Inibidores de Metaloproteinases de Matriz , Proteínas/metabolismo , Sequência de Aminoácidos , Domínio Catalítico , Cristalografia por Raios X , Humanos , Metaloproteinase 2 da Matriz/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Ligação Proteica , Estrutura Secundária de Proteína , Proteínas/química
7.
Cancer Sci ; 103(4): 691-9, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22321149

RESUMO

Angiomodulin (AGM/IGFBP-rP1), a glycoprotein of about 30 kDa, is overexpressed in tumor vasculature as well as some human cancer cell lines, but it has been suggested to be a tumor suppressor. To elucidate roles of angiomodulin (AGM) in tumor progression, we here examined distribution of AGM in three types of human cancer tissues by immunohistochemistry. The results showed that AGM was overexpressed in the stroma as well as the vasculature surrounding tumor cells in the human cancer tissues. AGM and α-smooth muscle actin (α-SMA) as an activated fibroblast marker were often colocalized in cancer-associated fibroblasts (CAFs). In vitro analysis indicated that transforming growth factor (TGF)-ß1 might be an important inducer of AGM in normal human fibroblasts. AGM strongly stimulated the expression of fibronectin and weakly that of α-SMA in normal fibroblasts. AGM significantly stimulated the proliferation and migration of fibroblasts. The AGM-induced expression of fibronectin and α-SMA was blocked by a TGF-ß signal inhibitor but neither the stimulation of cell growth nor migration. These results imply that AGM activates normal fibroblasts by TGF-ß-dependent and independent mechanisms. These findings also suggest that AGM and TGF-ß1 cooperatively or complementarily contribute to the stromal activation and connective tissue formation in human cancer tissues, contributing to tumor progression.


Assuntos
Fibroblastos/fisiologia , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/metabolismo , Neoplasias/metabolismo , Células Estromais/metabolismo , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Progressão da Doença , Humanos , Neoplasias/irrigação sanguínea , Fator de Crescimento Transformador beta/farmacologia
8.
J Biol Chem ; 285(37): 28862-73, 2010 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-20605794

RESUMO

Localization of secreted matrix metalloproteinases (MMPs) on the cell surface is required not only for processing of cell surface proteins, but also for controlled degradation of the extracellular matrix (ECM). Our previous study demonstrated that binding of MMP-7 (matrilysin) to cell surface cholesterol sulfate (CS) is essential for the cell membrane-associated proteolytic action of this MMP. In this study, we investigated the role of CS in the MMP-7-catalyzed degradation of protein components of ECM. We found that the degradation of laminin-332 (laminin-5) catalyzed by MMP-7 was accelerated dramatically in the presence of CS, whereas the sulfated lipid inhibited the degradation of casein catalyzed by the protease. The MMP-7-catalyzed degradation of fibronectin was partially inhibited in the presence of low concentrations of CS, whereas it was accelerated significantly at high concentrations of the lipid. Therefore, it is likely that CS alters the substrate preference of MMP-7. We also found that the proteins of which MMP-7-catalyzed degradation were accelerated by CS also had affinities for CS, suggesting that CS facilitates the proteolyses by cross-linking MMP-7 to its substrates. Moreover, MMP-7 tethered to cancer cell surface via CS degraded fibronectin and laminin-332 coated on a culture plate. The degradations of the adhesive proteins led to significant detachment of the cells from the plate. Taken together, our findings provide a novel mechanism in which cell surface CS promotes the proteolytic activities of MMP-7 toward selective substrates in the pericellular ECM, thereby contributing to cancer cell migration and metastasis.


Assuntos
Moléculas de Adesão Celular/metabolismo , Ésteres do Colesterol/farmacologia , Fibronectinas/metabolismo , Metaloproteinase 7 da Matriz/metabolismo , Inibidores de Serina Proteinase/farmacologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Metástase Neoplásica , Neoplasias/metabolismo , Neoplasias/patologia , Especificidade por Substrato/efeitos dos fármacos , Calinina
9.
J Biol Chem ; 285(45): 35068-78, 2010 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-20805229

RESUMO

The basement membrane (BM) proteins laminins, which consist of α, ß, and γ chains, support tissue structures and cellular functions. To date only α4 and α5 types of laminins have been identified in the BMs of blood vessels. Our recent study suggested the presence of novel α3B-containing laminins in vascular BMs. Here we identified and characterized the third member of vascular laminins, laminin-3B11 (Lm3B11). RT-PCR analysis showed that microvascular endothelial (MVE) cells and umbilical vein endothelial cells expressed the messages for the α3B, ß1, ß2, and γ1 chains. In the culture of MVE cells, α3B was associated with ß1 and γ1, producing Lm3B11. Recombinant Lm3B11 was overexpressed by introducing the cDNAs of the three chains into HEK-293 cells and purified to homogeneity. Purified Lm3B11 exhibited relatively weak cell adhesion activity through both α3ß1 and α6ß1 integrins. Most characteristically, Lm3B11 strongly stimulated MVE cells to extend many lamellipodial protrusions. This pseudopodial branching was blocked by an inhibitor for Src or phosphatidylinositol 3-kinase. Consistently, Lm3B11 stimulated the phosphorylation of Src and Akt more strongly than other laminins, suggesting that the integrin-derived signaling is mediated by these factors. The unique activity of Lm3B11 appears to be favorable to the branching of capillaries and venules.


Assuntos
Moléculas de Adesão Celular/metabolismo , Células Endoteliais/metabolismo , Microvasos/metabolismo , Pseudópodes/metabolismo , Moléculas de Adesão Celular/genética , Linhagem Celular , Células Endoteliais/citologia , Humanos , Microvasos/citologia , Proteína Oncogênica pp60(v-src)/genética , Proteína Oncogênica pp60(v-src)/metabolismo , Fosforilação/fisiologia , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Pseudópodes/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Calinina
10.
Cancer Sci ; 102(5): 1095-100, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21276136

RESUMO

Laminins present in the basement membranes (BM) of blood vessels are involved in angiogenesis and other vascular functions that are critical for tumor growth and metastasis. Two major vascular laminins, the α4 (laminin-411/421) and α5 (laminin-511/521) types, have been well characterized. We recently found a third type of vascular laminin, laminin-3B11, consisting of the α3B, ß1 and γ1 chains, and revealed its biological activity. Laminin-3B11 potently stimulates vascular endothelial cells to extend lamellipodial protrusions. To understand the roles of laminin-3B11 in blood vessel functions and tumor growth, we examined localization of the laminin α3B chain in normal mammary glands and breast cancers, in comparison with the α4 and α5 laminins. In the immunohistochemical analysis, the α3B laminin was co-localized with the α4 and α5 laminins in the BM of venules and capillaries of normal breast tissues, but α3B was scarcely detected in vessels near invasive breast carcinoma cells. In contrast, the α4 laminin was overexpressed in capillaries of invasive carcinomas, where a large number of macrophages were found. The α5 laminin appeared to be weakly downregulated in cancer tissues, especially in capillary vessels. Furthermore, our in vitro analysis indicated that TNF-α significantly suppressed the laminin α3B expression in vascular endothelial cells, while it, as well as IL-1ß and TGF-α, upregulated the α4 expression. These results suggest that Lm3B11/3B21 may be required for normal mature vessels and interfere with tumor angiogenesis.


Assuntos
Membrana Basal/metabolismo , Neoplasias da Mama/metabolismo , Regulação para Baixo , Células Endoteliais/metabolismo , Laminina/metabolismo , Neoplasias da Mama/patologia , Feminino , Humanos , Imuno-Histoquímica , Reação em Cadeia da Polimerase Via Transcriptase Reversa
11.
J Cell Biochem ; 106(4): 693-702, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19173304

RESUMO

Matrilysin (MMP-7) plays important roles in tumor progression. Previous studies have suggested that MMP-7 binds to tumor cell surface and promotes their metastatic potential. In this study, we identified C-type lectin domain family 3 member A (CLEC3A) as a membrane-bound substrate of MMP-7. Although this protein is known to be expressed specifically in cartilage, its message was found in normal breast and breast cancer tissues as well as breast and colon cancer cell lines. Because few studies have been done on CLEC3A, we overexpressed its recombinant protein in human cancer cells. CLEC3A was found in the cell membrane, extracellular matrix (ECM), and culture medium of the CLEC3A-expressing cells. CLEC3A has a basic sequence in the NH(2)-terminal domain and showed a strong heparin-binding activity. MMP-7 cleaved the 20-kDa CLEC3A protein, dividing it to a 15-kDa COOH-terminal fragment and an NH(2)-terminal fragment with the basic sequence. The 15-kDa fragment no longer had heparin-binding activity. Treatment of the CLEC3A-expressing cells with MMP-7 released the 15-kDa CLEC3A into the culture supernatant. Furthermore, the 20-kDa CLEC3A promoted cell adhesion to laminin-332 and fibronectin substrates, but this activity was abrogated by the cleavage by MMP-7. These results suggest that CLEC3A binds to heparan sulfate proteoglycans on cell surface, leading to the enhancement of cell adhesion to integrin ligands on ECM. It can be speculated that the cleavage of CLEC3A by MMP-7 weakens the stable adhesion of tumor cells to the matrix and promotes their migration in tumor microenvironments.


Assuntos
Neoplasias da Mama/patologia , Adesão Celular , Neoplasias do Colo/patologia , Lectinas Tipo C/metabolismo , Metaloproteinase 7 da Matriz/metabolismo , Sequência de Aminoácidos , Linhagem Celular Tumoral , Membrana Celular/química , Matriz Extracelular/química , Feminino , Humanos , Metástase Neoplásica , Fragmentos de Peptídeos , Ligação Proteica , Proteoglicanas/metabolismo , Especificidade por Substrato
12.
FEBS J ; 275(19): 4810-23, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18721140

RESUMO

Matrilysin (matrix metalloproteinase-7) plays important roles in tumor progression. It was previously found that matrilysin binds to the surface of colon cancer cells to promote their metastatic potential. In this study, we identified annexin II as a novel membrane-bound substrate of matrilysin. Treatment of human colon cancer cell lines with active matrilysin released a 35 k Da annexin II form, which lacked its N-terminal region, into the culture supernatant. The release of the 35 k Da annexin II by matrilysin was significantly enhanced in the presence of serotonin or heparin. Matrilysin hydrolyzed annexin II at the Lys9-Leu10 bond, thus dividing the protein into an N-terminal nonapeptide and the C-terminal 35 k Da fragment. Annexin II is known to serve as a cell surface receptor for tissue-type plasminogen activator (tPA). Although the matrilysin treatment liberated the 35 k Da fragment of annexin II from the cell surface, it significantly increased tPA binding to the cell membrane. A synthetic N-terminal nonapeptide of annexin II bound to tPA more efficiently than intact annexin II. This peptide formed a heterodimer with intact annexin II in test tubes and on cancer cell surfaces. These and other results suggested that the nonapeptide generated by matrilysin treatment might be anchored to the cell membrane, possibly by binding to intact annexin II, and interact with tPA via its C-terminal lysine. It is supposed that the cleavage of cell surface annexin II by matrilysin contributes to tumor invasion and metastasis by enhancing tPA-mediated pericellular proteolysis by cancer cells.


Assuntos
Anexina A2/metabolismo , Neoplasias do Colo/metabolismo , Metaloproteinase 7 da Matriz/metabolismo , Ativador de Plasminogênio Tecidual/metabolismo , Linhagem Celular Tumoral , Humanos , Fragmentos de Peptídeos/metabolismo
13.
FEBS J ; 273(3): 615-27, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16420484

RESUMO

Insulin-like growth factor (IGF) binding protein-related protein-1 (IGFBP-rP1) modulates cellular adhesion and growth in an IGF/insulin-dependent or independent manner. It also shows tumor-suppressive activity in vivo. We recently found that a single-chain IGFB-rP1 is proteolytically cleaved to a two-chain form by a trypsin-like, endogenous serine proteinase, changing its biological activities. In this study, we attempted to identify the IGFBP-rP1-processing enzyme. Of nine human cell lines tested, seven cell lines secreted IGFBP-rP1 at high levels, and two of them, ovarian clear cell adenocarcinoma (OVISE) and gastric carcinoma (MKN-45), highly produced the cleaved IGFBP-rP1. Serine proteinase inhibitors effectively blocked the IGFBP-rP1 cleavage in the OVISE cell culture. The conditioned medium of OVISE cells did not cleave purified IGFBP-rP1, but their membrane fraction had an IGFBP-rP1-cleaving activity. The membrane fraction contained an 80-kDa gelatinolytic enzyme, which was identified as the membrane-type serine proteinase matriptase (MT-SP1) by immunoblotting. When the membrane fraction was separated by SDS/PAGE, the IGFBP-rP1-cleaving activity comigrated with matriptase. A soluble form of matriptase purified in an inhibitor-free form efficiently cleaved IGFBP-rP1 at the same site as that found in a naturally cleaved IGFBP-rP1. Furthermore, small interfering RNAs for matriptase efficiently blocked both the matriptase expression and the cleavage of IGBP-rP1 in OVISE cells. These results demonstrate that IGFBP-rP1 is processed to the two-chain form by matriptase on the cell surface.


Assuntos
Membrana Celular/metabolismo , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/metabolismo , Serina Endopeptidases/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Membrana Celular/química , Eletroforese em Gel de Poliacrilamida , Humanos , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/efeitos dos fármacos , RNA Interferente Pequeno/farmacologia , Serina Endopeptidases/efeitos dos fármacos , Relação Estrutura-Atividade , Células Tumorais Cultivadas
14.
J Leukoc Biol ; 100(6): 1405-1412, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27365529

RESUMO

Hospital-acquired infections as a result of Acinetobacter baumannii have become problematic because of high rates of drug resistance. Although neutrophils play a critical role in early protection against bacterial infection, their interactions with A. baumannii remain largely unknown. To elucidate the interactions between A. baumannii and human neutrophils, we cocultured these cells and analyzed them by microscopy and flow cytometry. We found that A. baumannii adhered to neutrophils. We next examined neutrophil and A. baumannii infiltration into Matrigel basement membranes by an in vitro transmigration assay. Neutrophils were activated by A. baumannii, and invasion was enhanced. More interestingly, A. baumannii was transported together by infiltrating neutrophils. Furthermore, we observed by live cell imaging that A. baumannii and neutrophils moved together. In addition, A. baumannii-activated neutrophils showed increased IL-8 production. The transport of A. baumannii was suppressed by inhibiting neutrophil infiltration by blocking the effect of IL-8. A. baumannii appears to use neutrophils for transport by activating these cells via IL-8. In this study, we revealed a novel bacterial transport mechanism that A. baumannii exploits human neutrophils by adhering to and inducing IL-8 release for bacterial portage. This mechanism might be a new treatment target.


Assuntos
Infecções por Acinetobacter/microbiologia , Acinetobacter baumannii/fisiologia , Interleucina-8/fisiologia , Neutrófilos/microbiologia , Acinetobacter/fisiologia , Infecções por Acinetobacter/sangue , Infecções por Acinetobacter/imunologia , Acinetobacter baumannii/patogenicidade , Animais , Aderência Bacteriana , Translocação Bacteriana , Células Cultivadas , Colágeno Tipo I , Escherichia coli/fisiologia , Feminino , Géis , Humanos , Interleucina-8/biossíntese , Interleucina-8/sangue , Membranas Artificiais , Camundongos , Camundongos Endogâmicos C3H , Fagocitose , Pseudomonas aeruginosa/fisiologia , Explosão Respiratória , Especificidade da Espécie , Imagem com Lapso de Tempo , Virulência
15.
Oncogene ; 22(54): 8662-70, 2003 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-14647460

RESUMO

Matrilysin (MMP-7) is thought to contribute to invasive growth and metastasis of colon carcinoma and many other human cancers. The present study demonstrates that treatment of human colon carcinoma cells with active matrilysin induces cell aggregation in vitro and promotes liver metastasis in nude mice. When two kinds of colon carcinoma cell lines were incubated with active matrilysin, this enzyme efficiently bound to the cell surface and induced loose cell aggregation, which led to E-cadherin-mediated tight cell aggregation. Synthetic MMP inhibitors inhibited both the membrane binding of matrilysin and matrilysin-induced cell aggregation, while TIMP-2 inhibited only the cell aggregation. Two other active MMPs, stromelysin and gelatinase A, neither bound to cell membrane nor induced cell aggregation. Tumor cells in loose cell aggregates could reaggregate even after they were freed from matrilysin and dispersed. When injected into the spleen of nude mice, the tumor cells in the stable aggregates produced much larger metastatic nodules in the livers than control cells and those in the loose aggregates. These results suggest that matrilysin may enhance metastatic potential of tumor cells by processing a cell surface protein(s) and thereby inducing loose and then tight aggregation of tumor cells.


Assuntos
Adesão Celular/efeitos dos fármacos , Neoplasias do Colo/patologia , Metaloproteinase 7 da Matriz/farmacologia , Animais , Caderinas/fisiologia , Agregação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Metaloproteinase 7 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Metástase Neoplásica
16.
Clin Exp Metastasis ; 32(5): 405-15, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25990436

RESUMO

Laminin γ2 (Lmγ2) chain, a subunit of the basement membrane protein laminin-332, is regarded as a typical cancer invasion marker. The overexpression of Lmγ2 chain by invasive cancer cells correlates with poor prognosis of cancer patients, and its forced expression in human cancer cells promotes their invasive growth in a nude mouse model. However, its actual roles in cancer progression, as well as the mechanism of its proinvasive effect, remain unclear. CD44 is known to be an important cancer stem cell marker and support cancer progression and stem cell functions. Here we demonstrate that amino-terminal fragments of Lmγ2 interact with CD44 on the membrane of breast cancer cells. Lmγ2 highly bound to the metastatic cell line MDA-MB-231 but poorly to the benign cell line MCF-7. The membrane receptor for Lmγ2 on MDA-MB-231 cells was identified to be the standard form of CD44 (CD44s) by co-immunoprecipitation, affinity chromatography and direct protein interaction assay. Lmγ2 interacted with CD44s through EGF-like repeat 2/3 in the Lmγ2 amino-terminus. Amino-terminal fragments of Lmγ2 induced the phosphorylation of CD44 cytoplasmic domain and stimulated migration of the cancer cells in a CD44-dependent manner. This migration was blocked by inhibitors of TGF-ß receptor I (TGF-ßRI) kinase. These results suggest that two important tumor markers, Lmγ2 and CD44, cooperate for cancer progression and possibly for cancer stem cell functions. TGF-ßRI may be involved in the Lmγ2/CD44 interaction.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/secundário , Movimento Celular , Receptores de Hialuronatos/metabolismo , Laminina/metabolismo , Células-Tronco Neoplásicas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Animais , Western Blotting , Adesão Celular , Proliferação de Células , Cromatografia de Afinidade , Feminino , Humanos , Imunoprecipitação , Camundongos , Invasividade Neoplásica , Células-Tronco Neoplásicas/patologia , Fosforilação , Receptor do Fator de Crescimento Transformador beta Tipo I , Transdução de Sinais , Células Tumorais Cultivadas
17.
FEBS J ; 281(15): 3346-56, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24903600

RESUMO

Matrix metalloproteinase (MMP)-7 binds to cell surface cholesterol sulfate (CS) and acts as a membrane-associated protease. We have previously found that CS modulates the substrate preference of MMP-7, thereby regulating its pericellular proteolytic action. MMP-7 potentially associates with the cell surface via sulfatide (SM4) and cardiolipin (CL) when they are overexpressed on the cell surface. Here, we investigated the molecular interaction between these acidic lipids and MMP-7 or its substrates, and their effects on the activity of MMP-7. Studies using MMP-7 variants with low CS-binding ability suggested that these lipids interact with a similar site on MMP-7. The hydroxamate-based MMP inhibitor TAPI-1 markedly reduced the affinity of MMP-7 for CS and CL, whereas that for SM4 was not affected by TAPI-1. These three acidic lipids also had different effects on the hydrolytic activity of MMP-7 towards a small peptide substrate: SM4, CL and CS reduced the activity to 80%, 92%, and 20%, respectively. Nevertheless, SM4 and CS similarly accelerated the MMP-7-catalyzed degradation of fibronectin and laminin-332, whereas CL did not. The increased proteolysis of substrate was observed only when both substrate and enzyme had affinity for the lipid, suggesting that the lipids probably bring the reactants into closer proximity. Furthermore, MMP-7 bound to cell surface SM4 or CS cleaved specific cell surface proteins and released similar fragments, whereas the cleavage was not stimulated by cell surface CL-bound MMP-7. This study provides a novel mechanism by which acidic lipids differentially regulate pericellular proteolysis by MMP-7 through allosteric alteration of the substrate-binding site and their inherent affinities for MMP-7 substrates.


Assuntos
Cardiolipinas/metabolismo , Ésteres do Colesterol/metabolismo , Metaloproteinase 7 da Matriz/metabolismo , Sulfoglicoesfingolipídeos/metabolismo , Ligação Competitiva , Cardiolipinas/química , Moléculas de Adesão Celular/química , Linhagem Celular Tumoral , Membrana Celular/enzimologia , Ésteres do Colesterol/química , Dipeptídeos/química , Dipeptídeos/farmacologia , Fibronectinas/química , Humanos , Ácidos Hidroxâmicos/química , Ácidos Hidroxâmicos/farmacologia , Cinética , Metaloproteinase 7 da Matriz/química , Inibidores de Metaloproteinases de Matriz/química , Inibidores de Metaloproteinases de Matriz/farmacologia , Ligação Proteica , Proteólise , Sulfoglicoesfingolipídeos/química , Calinina
18.
Clin Exp Metastasis ; 31(3): 285-91, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24292405

RESUMO

Macrophages infiltrating tumor tissues (tumor-associated macrophages, TAM) affect the malignant behaviors of tumor cells. We previously reported that monocytes were differentiated into TAM-like cells secreting matrix metalloproteinase (MMP)-9 by co-culture with tumor cells, and that cell adhesion to extracellular matrix (ECM) proteins played a critical role in the differentiation. In this study, we found that the monocyte differentiation was promoted by laminin-332 (laminin-5), a major epithelial ECM component. We also demonstrated that the proteolytic processing of the γ2 chain of laminin-332 was essential for its activity but that the N-terminal short arm of the γ2 chain inhibited MMP-9 secretion. These results indicate that the activity of laminin-332 for monocyte differentiation is dynamically regulated by the proteolytic processing of the γ2 chain.


Assuntos
Moléculas de Adesão Celular/metabolismo , Diferenciação Celular/genética , Metaloproteinase 9 da Matriz/metabolismo , Monócitos/citologia , Adesão Celular , Moléculas de Adesão Celular/química , Moléculas de Adesão Celular/genética , Linhagem Celular Tumoral , Técnicas de Cocultura , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Humanos , Macrófagos/metabolismo , Metaloproteinase 9 da Matriz/genética , Neoplasias/genética , Neoplasias/patologia , Calinina
19.
Cancer Med ; 3(3): 537-49, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24737780

RESUMO

Angiomodulin (AGM) is a member of insulin-like growth factor binding protein (IGFBP) superfamily and often called IGFBP-rP1 or IGFBP-7. AGM was originally identified as a tumor-derived cell adhesion factor, which was highly accumulated in blood vessels of human cancer tissues. AGM is also overexpressed in cancer-associated fibroblasts (CAFs) and activates fibroblasts. However, some studies have shown tumor-suppressing activity of AGM. To understand the roles of AGM in cancer progression, we here investigated the expression of AGM in benign and invasive breast cancers and its functions in cancer vasculature. Immunohistochemical analysis showed that AGM was highly expressed in cancer vasculature even in ductal carcinoma in situ (DCIS) as compared to normal vasculature, while its expression in CAFs was more prominent in invasive carcinomas than DCIS. In vitro analyses showed that AGM was strongly induced by vascular endothelial cell growth factor (VEGF) in vascular endothelial cells. Although AGM stimulated neither the growth nor migration of endothelial cells, it supported efficient adhesion of endothelial cells. Integrin αvß3 was identified as a novel major receptor for AGM in vascular endothelial cells. AGM retracted endothelial cells by inducing actin stress fibers and loosened their VE-cadherin-mediated intercellular junction. Consequently, AGM increased vascular permeability both in vitro and in vivo. Furthermore, AGM and integrin αvß3 were highly expressed and colocalized in cancer vasculature. These results suggest that AGM cooperates with VEGF to induce the aberrant functions of cancer vasculature as a ligand of integrin αvß3.


Assuntos
Neoplasias da Mama/genética , Integrina alfaVbeta3/metabolismo , Proteínas de Neoplasias/biossíntese , Neovascularização Patológica/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Biomarcadores Tumorais/genética , Neoplasias da Mama/patologia , Permeabilidade Capilar/genética , Carcinoma Intraductal não Infiltrante/genética , Carcinoma Intraductal não Infiltrante/patologia , Moléculas de Adesão Celular/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana , Humanos , Integrina alfaVbeta3/genética , Ligantes , Proteínas de Neoplasias/metabolismo , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Fator A de Crescimento do Endotélio Vascular/genética
20.
PLoS One ; 7(12): e53209, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23300891

RESUMO

Epithelial-mesenchymal transition (EMT) is a crucial event in tumor invasion and metastasis. However, most of past EMT studies have been conducted in the conventional two-dimensional (2D) monolayer culture. Therefore, it remains unclear what invasive phenotypes are acquired by EMT-induced cancer cells. To address this point, we attempted to characterize EMT cells in more physiological, three-dimensional (3D) collagen gel culture. EMT was induced by treating three human carcinoma cell lines (A549, Panc-1 and MKN-1) with TGF-ß. The TGF-ß treatment stimulated these cells to overexpress the invasion markers laminin γ2 and MT1-MMP in 2D culture, in addition to the induction of well-known morphological change and EMT marker expression. EMT induction enhanced cell motility and adhesiveness to fibronectin and collagen in 2D culture. Although EMT cells showed comparable cell growth to control cells in 2D culture, their growth rates were extremely suppressed in soft agar and collagen gel cultures. Most characteristically, EMT-induced cancer cells commonly and markedly extended invasive protrusions in collagen gel. These protrusions were mainly supported by microtubules rather than actin cytoskeleton. Snail-introduced, stable EMT cells showed similar protrusions in 3D conditions without TGF-ß. Moreover, these protrusions were suppressed by colchicine or inhibitors of heat shock protein 90 (HSP-90) and protein phosphatase 2A. However, MMP inhibitors did not suppress the protrusion formation. These data suggest that EMT enhances tumor cell infiltration into interstitial stroma by extending microtubule-based protrusions and suppressing cell growth. The elevated cell adhesion to fibronectin and collagen and high cell motility also seem important for the tumor invasion.


Assuntos
Movimento Celular/fisiologia , Colágeno/metabolismo , Transição Epitelial-Mesenquimal/fisiologia , Microtúbulos/metabolismo , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Técnicas de Cultura de Células , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Humanos , Laminina/metabolismo , Microtúbulos/patologia , Fator de Crescimento Transformador beta/farmacologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa