Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Respir Res ; 17: 17, 2016 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-26887531

RESUMO

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a chronic progressively fatal disease. Extracellular Matrix Metalloproteinase Inducer (EMMPRIN) is a glycosylated transmembrane protein that induces the expression of some matrix metalloproteinase (MMP) in neighboring stromal cells through direct epithelial-stromal interactions. EMMPRIN is highly expressed in type II alveolar epithelial cells at the edges of the fibrotic areas in IPF lung sections. However, the exact role of EMMPRIN in IPF is unknown. METHODS: To determine if EMMPRIN contributes to lung fibroblast proliferation, resistance to apoptosis, and differentiation to myofibroblasts, normal Human lung fibroblasts (NHLF) transiently transfected with either EMMPRIN/GFP or GFP were treated with TGF- ß1 from 0 to 10 ng/ml for 48 h and examined for cell proliferation (thymidine incorporation), apoptosis (FACS analysis and Cell Death Detection ELISA assay), cell migration (Modified Boyden chamber) and differentiation to myofibroblasts using Western blot for α-smooth actin of cell lysates. The effect of EMMPRIN inhibition on NHLF proliferation, apoptosis, migration and differentiation to myofibroblasts after TGF- ß1 treatment was examined using EMMPRIN blocking antibody. We examined the mechanism by which EMMPRIN induces its effects on fibroblasts by studying the ß-catenin/canonical Wnt signaling pathway using Wnt luciferase reporter assays and Western blot for total and phosphorylated ß-catenin. RESULTS: Human lung fibroblasts overexpressing EMMPRIN had a significant increase in cell proliferation and migration compared to control fibroblasts. Furthermore, EMMPRIN promoted lung fibroblasts resistance to apoptosis. Lung fibroblasts overexpressing EMMPRIN showed a significantly increased expression of α- smooth muscle actin, a marker of differentiation to myofibroblasts compared to control cells. TGF-ß1 increased the expression of EMMPRIN in lung fibroblasts in a dose-dependent manner. Attenuation of EMMPRIN expression with the use of an EMMPRIN blocking antibody markedly inhibited TGF-ß1 induced proliferation, migration, and differentiation of fibroblasts to myofibroblasts. EMMPRIN overexpression in lung fibroblasts was found to induce an increase in TOPFLASH luciferase reporter activity when compared with control fibroblasts. CONCLUSION: These findings indicate that TGF-ß1 induces the release of EMMPRIN that activates ß-catenin/canonical Wnt signaling pathway. EMMPRIN overexpression induces an anti-apoptotic and pro-fibrotic phenotype in lung fibroblasts that may contribute to the persistent fibro-proliferative state seen in IPF.


Assuntos
Basigina/metabolismo , Fibroblastos/citologia , Fibroblastos/fisiologia , Pulmão/citologia , Pulmão/fisiologia , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Sobrevivência Celular/fisiologia , Células Cultivadas , Humanos , Miofibroblastos/citologia , Miofibroblastos/fisiologia
2.
Mol Pharmacol ; 87(3): 501-12, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25552486

RESUMO

Because cancer cell invasion is a critical determinant of metastasis, targeting invasion is a viable approach to prevent metastasis. Utilizing a novel three-dimensional high-throughput invasion assay, we screened a National Cancer Institute compound library and discovered compounds demonstrating inhibitory effects on cancer cell invasion. One hit, trifluoperazine, suppresses invasion of human cancer cell lines while displaying a limited cytotoxicity profile. This inhibition is due to the interference with cancer cell migratory ability but not proteolytic activity. Treatment of cancer cells with trifluoperazine significantly reduces angiogenesis and prevents cancer cell invasion through a chorioallantoic basement membrane. Mechanistically, treatment results in decreased phosphorylated AKT (Ser(473) and Thr(308)) and ß-catenin (Ser(552)). Lack of phosphorylation of Ser(552) of ß-catenin prevents ß-catenin nuclear relocation, resulting in decreased expression of vascular endothelial growth factor, likely mediated through dopamine receptor D2. Taken together, we demonstrated that trifluoperazine is responsible for reducing the angiogenic and invasive potential of aggressive cancer cells through dopamine receptor D2 to modulate the ß-catenin pathway and propose that trifluoperazine may be used as an antimetastasis chemotherapeutic.


Assuntos
Antineoplásicos/farmacologia , Antipsicóticos/farmacologia , Invasividade Neoplásica/prevenção & controle , Trifluoperazina/farmacologia , Animais , Linhagem Celular Tumoral , Embrião de Galinha , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Camundongos , Células NIH 3T3 , Invasividade Neoplásica/patologia
3.
Blood Cells Mol Dis ; 55(3): 241-7, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26227854

RESUMO

JAK2(V617F) mutation can be detected in the majority of myeloproliferative neoplasm (MPN) patients. The JAK2 inhibitor Ruxolitinib is the first FDA-approved treatment for MPNs. However, its use is limited by various dose related toxicities. Here, we studied the metabolic state and glutamine metabolism of BaF3-hEPOR-JAK2V617F and BaF3-hEPOR-JAK2WT cells. We found that the JAK2(V617F)-mutant cells were associated with increased oxygen consumption rate and extracellular acidification rate than the JAK2(WT) cells and there was an increased glutamine metabolism in JAK2(V617F)-mutant cells compared to wild-type cells. Glutaminase (GLS), the key enzyme in glutamine metabolism, was upregulated in the JAK2(V617F)-mutant BaF3 cells compared to the JAK2(WT) BaF3 cells. In MPN patient peripheral blood CD34+ cells, GLS expression was increased in JAK2(V617F)-mutant progenitor cells compared to JAK2 wild-type progenitor cells from the same patients and GLS levels were increased at the time of disease progression compared to at earlier time points. Moreover, GLS inhibitor increased the growth inhibitory effect of Ruxolitinib in both JAK2(V617F)-mutant cell lines and peripheral blood CD34+ cells from MPN patients. Therefore, GLS inhibitor should be further explored to enhance the therapeutic effectiveness of JAK2 inhibitor and allow the administration of lower doses of the drug to avoid its toxicity.


Assuntos
Glutamina/metabolismo , Janus Quinase 2/metabolismo , Transtornos Mieloproliferativos/metabolismo , Linhagem Celular Tumoral , Humanos , Janus Quinase 2/antagonistas & inibidores , Janus Quinase 2/genética , Mutação , Nitrilas , Pirazóis/farmacologia , Pirimidinas , Regulação para Cima
4.
Gynecol Oncol ; 137(2): 229-38, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25769657

RESUMO

GOALS: Contemporary management of epithelial ovarian cancer (EOC) uses biomarkers to monitor response to therapy. This study evaluates the role of invasive circulating tumor cells (iCTCs) in monitoring EOC treatment in comparison with serum cancer antigen 125 (CA125). METHODS: Molecular and microscopic analyses were used to identify seprase and CD44 as tumor progenitor (TP) markers. The iCTC flow cytometry assay was optimized using blood donated by 64 healthy donors, 49 patients with benign abdominal diseases and 123 EOC patients. Serial changes in iCTCs and CA125 were measured in 129 blood and 169 serum samples, respectively, from 31 EOC patients to assess their concordance during therapy and their relationship with risk of progressive disease (PD). RESULTS: The assay had 97% specificity and 83% sensitivity for detecting iCTCs in blood of EOC patients. iCTCs were detected in each monitoring patient (31/31, 100%) and in 110 of the 129 blood samples (85.3%). The concordance between changes in iCTCs/CA125 levels and changes in the intervals associated with no evidence of disease (NED) were markedly stronger (specificity: CA125 93.8%; iCTCs 90.6%), whereas increases in iCTCs (79.5%) were more sensitive than increases in CA125 (67.6%) to predict PD or relapse. Among the six patients who had greater than 6 measurements, iCTCs but not CA125 antedated changes in clinical status from PD to NED during and after chemotherapy and predated relapse. CONCLUSION: Serial measurements of iCTCs could predict therapeutic responsiveness in 31 EOC patients who underwent standard taxol/carboplatin therapy.


Assuntos
Neoplasias Epiteliais e Glandulares/sangue , Neoplasias Epiteliais e Glandulares/tratamento farmacológico , Células Neoplásicas Circulantes/patologia , Neoplasias Ovarianas/sangue , Neoplasias Ovarianas/tratamento farmacológico , Biomarcadores Tumorais/sangue , Antígeno Ca-125/sangue , Carcinoma Epitelial do Ovário , Estudos de Casos e Controles , Estudos de Coortes , Monitoramento Ambiental/métodos , Feminino , Citometria de Fluxo , Humanos , Receptores de Hialuronatos/sangue , Proteínas de Membrana/sangue , Recidiva Local de Neoplasia/sangue , Recidiva Local de Neoplasia/patologia , Estadiamento de Neoplasias , Neoplasias Epiteliais e Glandulares/patologia , Células Neoplásicas Circulantes/metabolismo , Neoplasias Ovarianas/patologia , Serina Endopeptidases/sangue
5.
Gynecol Oncol ; 134(3): 581-90, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24972191

RESUMO

GOALS: Circulating tumor cells (CTCs) have been introduced as a biomarker in detecting advanced epithelial ovarian cancer (EOC). The goals are to examine the prevalence of the invasive subpopulation of CTCs (iCTCs) in patients at high risk of EOC and to compare this biomarker to serum CA125. METHODS: We used a unique cell adhesion matrix (CAM)-based, functional cell enrichment and identification platform to isolate iCTCs from 129 preoperative patients. We confirmed the identity of iCTCs using positive epithelial (Epi+) markers and negative hematopoietic lineage (HL-) markers. Sensitivity and specificity of the assays were examined and iCTCs/CA125 were correlated with overall survival (OS), progression-free survival (PFS) and clinical parameters. RESULTS: We found a 41.2% sensitivity, 95.1% specificity and 77.8% positive predictive value (PPV) of the iCTC assay in detecting patients with stage I and II EOC malignancy, and a 83% sensitivity and 97.3% PPV in detecting all stages of EOC malignancy. However, a positive CA125 test provided weak evidence to detect stage I and II malignancy (61.6% PPV) and all EOC (92.1% PPV), because of its 76.2% specificity. A significantly stronger concordance in OS and PFS of clinical factors (tumor stage, debulking and platinum sensitivity) was noted for elevated iCTCs than for serum CA125. CONCLUSION: The CAM-initiated CTC enrichment/identification method enabled the detection of early stage EOC. iCTCs were better correlated with worse OS and PFS, more specific and better PPV than CA125 in detecting EOC malignancy in patients at high risk of EOC.


Assuntos
Biomarcadores Tumorais/sangue , Antígeno Ca-125/sangue , Neoplasias Epiteliais e Glandulares/sangue , Neoplasias Epiteliais e Glandulares/patologia , Células Neoplásicas Circulantes/patologia , Neoplasias Ovarianas/sangue , Neoplasias Ovarianas/patologia , Carcinoma Epitelial do Ovário , Contagem de Células , Intervalo Livre de Doença , Feminino , Humanos , Pessoa de Meia-Idade , Neoplasias Epiteliais e Glandulares/mortalidade , Neoplasias Ovarianas/mortalidade , Valor Preditivo dos Testes , Prognóstico , Sensibilidade e Especificidade , Taxa de Sobrevida , Células Tumorais Cultivadas
6.
J Biol Chem ; 286(38): 33167-77, 2011 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-21795678

RESUMO

Matrix metalloproteinases (MMPs) have been shown to be key players in both extracellular matrix remodeling and cell migration during cancer metastasis. MMP-14, a membrane-anchored MMP, in particular, is closely associated with these processes. The hemopexin (PEX) domain of MMP-14 has been proposed as the modulating region involved in the molecular cross-talk that initiates cell migration through homodimerization of MMP-14 as well as heterodimerization with the cell surface adhesion molecule CD44. In this study, minimal regions required for function within the PEX domain were investigated through a series of substitution mutations. Blades I and IV were found to be involved in cell migration. We found that blade IV is necessary for MMP-14 homodimerization and that blade I is required for CD44 MMP-14 heterodimerization. Cross-talk between MMP-14 and CD44 results in phosphorylation of EGF receptor and downstream activation of the MAPK and PI3K signaling pathways involved in cell migration. Based on these mutagenesis analyses, peptides mimicking the essential outermost strand motifs within the PEX domain of MMP-14 were designed. These synthetic peptides inhibit MMP-14-enhanced cell migration in a dose-dependent manner but have no effect on the function of other MMPs. Furthermore, these peptides interfere with cancer metastasis without affecting primary tumor growth. Thus, targeting the MMP-14 hemopexin domain represents a novel approach to inhibit MMP-14-mediated cancer dissemination.


Assuntos
Neoplasias da Mama/enzimologia , Neoplasias da Mama/patologia , Movimento Celular , Inibidores de Metaloproteinases de Matriz , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/mortalidade , Células COS , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Galinhas , Chlorocebus aethiops , Ativação Enzimática/efeitos dos fármacos , Receptores ErbB/metabolismo , Feminino , Humanos , Receptores de Hialuronatos/metabolismo , Metaloproteinase 14 da Matriz/química , Metaloproteinase 14 da Matriz/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Dados de Sequência Molecular , Neovascularização Patológica/enzimologia , Peptídeos/química , Peptídeos/farmacologia , Estrutura Terciária de Proteína , Transdução de Sinais/efeitos dos fármacos , Taxa de Sobrevida
7.
J Biol Chem ; 285(46): 35944-56, 2010 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-20837483

RESUMO

Non-proteolytic activities of matrix metalloproteinases (MMPs) have recently been shown to impact cell migration, but the precise mechanism remains to be understood. We previously demonstrated that the hemopexin (PEX) domain of MMP-9 is a prerequisite for enhanced cell migration. Using a biochemical approach, we now report that dimerization of MMP-9 through the PEX domain appears necessary for MMP-9-enhanced cell migration. Following a series of substitution mutations within the MMP-9 PEX domain, blade IV was shown to be critical for homodimerization, whereas blade I was required for heterodimerization with CD44. Blade I and IV mutants showed diminished enhancement of cell migration compared with wild type MMP-9-transfected cells. Peptides mimicking motifs in the outermost strands of the first and fourth blades of the MMP-9 PEX domain were designed. These peptides efficiently blocked MMP-9 dimer formation and inhibited motility of COS-1 cells overexpressing MMP-9, HT-1080, and MDA-MB-435 cells. Using a shRNA approach, CD44 was found to be a critical molecule in MMP-9-mediated cell migration. Furthermore, an axis involving a MMP-9-CD44-EGFR signaling pathway in cell migration was identified using antibody array and specific receptor tyrosine kinase inhibitors. In conclusion, we dissected the mechanism of pro-MMP-9-enhanced cell migration and developed structure-based inhibitory peptides targeting MMP-9-mediated cell migration.


Assuntos
Movimento Celular/fisiologia , Metaloproteinase 9 da Matriz/química , Metaloproteinase 9 da Matriz/metabolismo , Multimerização Proteica/fisiologia , Sequência de Aminoácidos , Animais , Western Blotting , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Inibidores Enzimáticos/farmacologia , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Humanos , Receptores de Hialuronatos/química , Receptores de Hialuronatos/genética , Receptores de Hialuronatos/metabolismo , Metaloproteinase 9 da Matriz/genética , Modelos Moleculares , Mutação , Oligopeptídeos/química , Oligopeptídeos/farmacologia , Ligação Proteica , Conformação Proteica , Multimerização Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína , Quinazolinas , Interferência de RNA , Transdução de Sinais/efeitos dos fármacos , Inibidor Tecidual de Metaloproteinase-1/química , Inibidor Tecidual de Metaloproteinase-1/genética , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Transfecção , Tirfostinas/farmacologia
8.
Connect Tissue Res ; 50(4): 223-31, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19637058

RESUMO

Activation of MMPs in tissues is an important component of tissue injury. Based on earlier reports that (latent) proMMP-2 is incapable of forming a complex with TIMP-1, we reasoned that the identification of MMP-2:TIMP-1 complexes in blood might serve as a surrogate marker ("smoking gun") of MMP-2 activation in tissues. Using specific antibodies, we developed a sensitive and specific assay to detect MMP-2:TIMP-1 complexes. We were perplexed to find that approximate 40% of plasma specimens from healthy individuals had detectable levels of the MMP-2:TIMP-1 complexes. Employing recombinant TIMP-1 bound Sepharose beads and Western blots, we demonstrated binding between recombinant proMMP-2 and TIMP-1 proteins. Recombinant MMP-2 lacking the catalytic domain also bound to TIMP-1 coated beads. These data are consistent with TIMP-1 binding to the hemopexin or hinge domain of proMMP-2. The explanation for the presence of plasma proMMP-2:TIMP-1 complexes in selected healthy individuals remains to be determined. In contrast to our immunoassay and bead-binding experiments, proMMP-2 failed to bind to immobilized TIMP-1 employing surface plasmon resonance technology. Additional studies are needed to clarify these contrasting results.


Assuntos
Precursores Enzimáticos/sangue , Gelatinases/sangue , Metaloproteases/sangue , Inibidor Tecidual de Metaloproteinase-1/sangue , Adulto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Ligação Proteica/fisiologia , Proteínas Recombinantes/metabolismo
9.
J Cell Physiol ; 217(3): 643-51, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18636552

RESUMO

The biological functions of matrix metalloproteinases (MMPs) extend beyond extracellular matrix degradation. Non-proteolytic activities of MMPs are just beginning to be understood. Herein, we evaluated the role of proMMPs in cell migration. Employing a Transwell chamber migration assay, we demonstrated that transfection of COS-1 cells with various proMMP cDNAs resulted in enhancement of cell migration. Latent MMP-2 and MMP-9 enhanced cell migration to a greater extent than latent MMP-1, -3, -11 and -28. To examine if proteolytic activity is required for MMP-enhanced cell migration, three experimental approaches, including fluorogenic substrate degradation assay, transfection of cells with catalytically inactive mutant MMP cDNAs, and addition of hydroxamic acid-derived MMP inhibitors, were employed. We demonstrated that the proteolytic activities of MMPs are not required for MMP-induced cell migration. To explore the mechanism underlying MMP-enhanced cell migration, structure-function relationship of MMP-9 on cell migration was evaluated. By using a domain swapping approach, we demonstrated that the hemopexin domain of proMMP-9 plays an important role in cell migration when examined by a transwell chamber assay and by a phagokinetic migration assay. TIMP-1, which interacts with the hemopexin domain of proMMP-9, inhibited cell migration, whereas TIMP-2 had no effect. Employing small molecular inhibitors, MAPK and PI3K pathways were found to be involved in MMP-9-mediated cell migration. In conclusion, we demonstrated that MMPs utilize a non-proteolytic mechanism to enhance epithelial cell migration. We propose that hemopexin homodimer formation is required for the full cell migratory function of proMMP-9.


Assuntos
Movimento Celular , Hemopexina/química , Metaloproteinases da Matriz/química , Metaloproteinases da Matriz/metabolismo , Animais , Células COS , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Chlorocebus aethiops , Inibidores Enzimáticos/farmacologia , Precursores Enzimáticos/metabolismo , Humanos , Metaloproteinase 9 da Matriz/química , Metaloproteinase 9 da Matriz/metabolismo , Inibidores de Metaloproteinases de Matriz , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Estrutura Terciária de Proteína , Transdução de Sinais/efeitos dos fármacos , Relação Estrutura-Atividade , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Inibidor Tecidual de Metaloproteinase-2/metabolismo , Transfecção
10.
Cancer Res ; 66(9): 4652-61, 2006 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-16651416

RESUMO

Dipeptidyl peptidase IV (DPP4/CD26) and seprase/fibroblast activation protein alpha are homologous type II transmembrane, homodimeric glycoproteins that exhibit unique prolyl peptidase activities. Human DPP4 is ubiquitously expressed in epithelial and endothelial cells and serves multiple functions in cleaving the penultimate positioned prolyl bonds at the NH(2) terminus of a variety of physiologically important peptides in the circulation. Recent studies showed a linkage between DPP4 and down-regulation of certain chemokines and mitogenic growth factors, and degradation of denatured collagens (gelatin), suggesting a role of DPP4 in the cell invasive phenotype. Here, we found the existence of a novel protease complex consisting of DPP4 and seprase in human endothelial cells that were activated to migrate and invade in the extracellular matrix in vitro. DPP4 and seprase were coexpressed with the three major protease systems (matrix metalloproteinase, plasminogen activator, and type II transmembrane serine protease) at the cell surface and organize as a complex at invadopodia-like protrusions. Both proteases were colocalized at the endothelial cells of capillaries, but not large blood vessels, in invasive breast ductal carcinoma in vivo. Importantly, monoclonal antibodies against the gelatin-binding domain of DPP4 blocked the local gelatin degradation by endothelial cells in the presence of the major metallo- and serine protease systems that modified pericellular collagenous matrices and subsequent cell migration and invasion. Thus, we have identified a novel mechanism involving the DPP4 gelatin-binding domain of the DPP4-seprase complex that facilitates the local degradation of the extracellular matrix and the invasion of the endothelial cells into collagenous matrices.


Assuntos
Movimento Celular/fisiologia , Extensões da Superfície Celular/enzimologia , Colágeno/metabolismo , Dipeptidil Peptidase 4/metabolismo , Células Endoteliais/citologia , Células Endoteliais/enzimologia , Matriz Extracelular/metabolismo , Peptídeo Hidrolases/metabolismo , Serina Endopeptidases/metabolismo , Anticorpos Monoclonais/farmacologia , Dipeptidil Peptidase 4/biossíntese , Gelatina/metabolismo , Humanos , Peptídeo Hidrolases/biossíntese , Serina Endopeptidases/biossíntese , Regulação para Cima
11.
Biomedicines ; 6(2)2018 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-29882767

RESUMO

Colorectal carcinoma (CRC) is the second leading cause of cancer-related mortality. The goals of this study are to evaluate the association between levels of invasive circulating tumor cells (iCTCs) with CRC outcomes and to explore the molecular characteristics of iCTCs. Peripheral blood from 93 patients with Stage I⁻IV CRC was obtained and assessed for the detection and characterization of iCTCs using a functional collagen-based adhesion matrix (CAM) invasion assay. Patients were followed and assessed for overall survival. Tumor cells isolated by CAM were characterized using cell culture and microarray analyses. Of 93 patients, 88 (95%) had detectable iCTCs, ranging over 0⁻470 iCTCs/mL. Patients with Stage I⁻IV disease exhibited median counts of 0.0 iCTCs/mL (n = 6), 13.0 iCTCs/mL (n = 12), 41.0 iCTCs/mL (n = 12), and 133.0 iCTCs/mL (n = 58), respectively (p < 0.001). Kaplan⁻Meier curve analysis demonstrated a significant survival benefit in patients with low iCTC counts compared with in patients with high iCTC counts (log-rank p < 0.001). Multivariable Cox model analysis revealed that iCTC count was an independent prognostic factor of overall survival (p = 0.009). Disease stage (p = 0.01, hazard ratio 1.66; 95% confidence interval: 1.12⁻2.47) and surgical intervention (p = 0.03, HR 0.37; 95% CI: 0.15⁻0.92) were also independent prognostic factors. Gene expression analysis demonstrated the expression of both endothelial and tumor progenitor cell biomarkers in iCTCs. CAM-based invasion assay shows a high detection sensitivity of iCTCs that inversely correlated with overall survival in CRC patients. Functional and gene expression analyses showed the phenotypic mosaics of iCTCs, mimicking the survival capability of circulating endothelial cells in the blood stream.

12.
Chem Biol ; 13(4): 347-9, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16632246

RESUMO

MMP inhibitor-tethered resins provide a method for detection of activated MMPs in tissues. These measurements will help to elucidate in situ mechanisms of MMP activation, further our understanding of the tumor microenvironment, and facilitate development of MMP-inhibitor therapy.


Assuntos
Metaloproteinases da Matriz/análise , Ativação Enzimática , Humanos , Metaloproteinases da Matriz/metabolismo , Modelos Biológicos , Inibidores Teciduais de Metaloproteinases/metabolismo
13.
Cancer Res ; 65(10): 4162-71, 2005 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-15899807

RESUMO

Furin, a potent proprotein convertase involved in activation of several cancer-related substrates, is synthesized as an inactive zymogen, thus minimizing the occurrence of premature enzymatic activity that would lead to inappropriate protein activation or degradation. This natural inhibitory mechanism is based on the presence of an inactivating prosegment at the NH2 terminal of the zymogen. After initial autocatalytic cleavage, the prosegment remains tightly associated with the convertase until it reaches the trans-Golgi network where the dissociation of the prosegment and activation of furin occurs. We hypothesized that the inhibitory properties of the preprosegment of furin (ppFur) could be beneficial if ectopically expressed in tumor cells. Transfection of four human head and neck squamous cell carcinoma cell lines with the complete ppFur cDNA sequence (pIRES-EGFP-ppFur) or with the empty expression vector (pIRES-EGFP) was done. The inhibitory effect was evaluated using in vivo tumorigenicity, invasion, anchorage-independent growth in soft agar, and proliferation assays, as well as by investigating impairment of furin substrates processing. Following transfection of ppFur, a significant reduction in cell proliferation, tumorigenicity, and invasiveness was observed in vitro and in vivo. These biological changes are directly related to the inhibition of furin-mediated activation of crucial cancer-related substrates, such as membrane type 1 matrix metalloproteinase, transforming growth factor-beta, insulin-like growth factor-1 receptor, and vascular endothelial growth factor-C. PpFur expression in head and neck squamous cell carcinoma cell lines showed a mechanistic link between furin inhibition, decreased substrate processing, cell proliferation, and invasive ability. These findings suggest that furin inhibition is a feasible approach to ameliorate and even abolish the malignant phenotype of various malignancies.


Assuntos
Carcinoma de Células Escamosas/enzimologia , Carcinoma de Células Escamosas/patologia , Furina/genética , Neoplasias de Cabeça e Pescoço/enzimologia , Neoplasias de Cabeça e Pescoço/patologia , Carcinoma de Células Escamosas/irrigação sanguínea , Carcinoma de Células Escamosas/genética , Processos de Crescimento Celular , Linhagem Celular Tumoral , Furina/biossíntese , Neoplasias de Cabeça e Pescoço/irrigação sanguínea , Neoplasias de Cabeça e Pescoço/genética , Humanos , Invasividade Neoplásica , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Fragmentos de Peptídeos/biossíntese , Fragmentos de Peptídeos/genética , Precursores de Proteínas/biossíntese , Precursores de Proteínas/genética , Transfecção
14.
Clin Exp Metastasis ; 23(5-6): 259-68, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17086359

RESUMO

We have isolated a novel soluble factor(s), neutrophil activator of matrix metalloproteinases (NAM), secreted by unstimulated normal human peripheral blood neutrophils that causes the activation of cell secreted promatrix metalloproteinase-2 (proMMP-2). Partially purified preparations of NAM have been isolated from the conditioned media of neutrophils employing gelatin-Sepharose chromatography and differential membrane filter centrifugation. NAM activity, as assessed by exposing primary human umbilical vein endothelial cells (HUVEC) or HT1080 cells to NAM followed by gelatin zymography, was seen within one hour. Tissue inhibitor of metalloproteinase-2 (TIMP-2) and hydroxamic acid derived inhibitors of MMPs (CT1746 and BB94) abrogated the activation of proMMP-2 by NAM, while inhibitors of serine and cysteine proteases showed no effect. NAM also produced an increase in TIMP-2 binding to HUVEC and HT1080 cell surfaces that was inhibited by TIMP-2, CT1746, and BB94. Time-dependent increases in MT1-MMP protein and mRNA were seen following the addition of NAM to cells. These data support a role for NAM in cancer dissemination.


Assuntos
Metaloproteinase 2 da Matriz/metabolismo , Metástase Neoplásica/fisiopatologia , Neutrófilos/metabolismo , Proteínas/isolamento & purificação , Adulto , Amidas/farmacologia , Calcimicina/farmacologia , Catepsina G , Catepsinas/farmacologia , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/enzimologia , Meios de Cultivo Condicionados/química , Endotélio Vascular/citologia , Ativação Enzimática , Precursores Enzimáticos/metabolismo , Humanos , Inflamação , Ionomicina/farmacologia , Metaloproteinase 14 da Matriz/biossíntese , Metaloproteinase 14 da Matriz/genética , Invasividade Neoplásica , Oligopeptídeos/farmacologia , Elastase Pancreática/farmacologia , Fenilalanina/análogos & derivados , Fenilalanina/farmacologia , Inibidores de Proteases/farmacologia , Proteínas/farmacologia , Proteínas/fisiologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Serina Endopeptidases/farmacologia , Especificidade por Substrato , Acetato de Tetradecanoilforbol/farmacologia , Tiofenos/farmacologia , Inibidor Tecidual de Metaloproteinase-2/farmacologia , Veias Umbilicais
15.
Clin Breast Cancer ; 6(6): 525-9, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16595036

RESUMO

BACKGROUND: The purpose of this study was to evaluate the prognostic and predictive utility of measuring plasma levels of matrix metalloproteinase (MMP)-7 and MMP-9 in patients with metastatic breast cancer (MBC) treated with the oral MMP inhibitor (MMPI) marimastat or a placebo. PATIENTS AND METHODS: We measured plasma levels of MMP-7 and MMP-9 using an enzyme-linked immunoassay method in 140 evaluable patients with MBC enrolled on a multicenter clinical trial that compared the MMPI marimastat with placebo. Specimens were collected after completion of first-line chemotherapy in patients who had responding or stable disease and 3, 6, and 12 months after initiation of marimastat (10 mg orally twice daily) or placebo. RESULTS: Baseline plasma MMP-7 and MMP-9 levels did not correlate with each other or with progression-free or overall survival. In addition, serial evaluation of plasma MMP-7 and MMP-9 levels revealed no significant changes over time in the marimastat or placebo groups or any correlation with trough plasma marimastat level. CONCLUSION: We conclude that measurement of plasma MMP-7 or MMP-9 levels, as performed in our trial, was not a useful prognostic or predictive factor in patients with MBC or in patients treated with an MMPI.


Assuntos
Biomarcadores Tumorais/sangue , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/enzimologia , Inibidores Enzimáticos/uso terapêutico , Ácidos Hidroxâmicos/uso terapêutico , Metaloproteinase 7 da Matriz/sangue , Metaloproteinase 9 da Matriz/sangue , Administração Oral , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias da Mama/patologia , Inibidores Enzimáticos/administração & dosagem , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Ácidos Hidroxâmicos/administração & dosagem , Pessoa de Meia-Idade , Placebos , Valor Preditivo dos Testes , Prognóstico
16.
FASEB J ; 19(11): 1507-9, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16014803

RESUMO

Airway smooth muscle (ASM) proliferation and migration are major components of airway remodeling in asthma. Asthmatic airways are exposed to mechanical strain, which contributes to their remodeling. Matrix metalloproteinase (MMP) plays an important role in remodeling. In the present study, we examined if the mechanical strain of human ASM (HASM) cells contributes to their proliferation and migration and the role of MMPs in this process. HASM were exposed to mechanical strain using the FlexCell system. HASM cell proliferation, migration and MMP release, activation, and expression were assessed. Our results show that cyclic strain increased the proliferation and migration of HASM; cyclic strain increased release and activation of MMP-1, -2, and -3 and membrane type 1-MMP; MMP release was preceded by an increase in extracellular MMP inducer; Prinomastat [a MMP inhibitor (MMPI)] significantly decreased cyclic strain-induced proliferation and migration of HASM; and the strain-induced increase in the release of MMPs was accompanied by an increase in tenascin-C release. In conclusion, cyclic mechanical strain plays an important role in HASM cell proliferation and migration. This increase in proliferation and migration is through an increase in MMP release and activation. Pharmacological MMPIs should be considered in the pursuit of therapeutic options for airway remodeling in asthma.


Assuntos
Basigina/fisiologia , Pulmão/citologia , Metaloendopeptidases/fisiologia , Miócitos de Músculo Liso/citologia , Asma/tratamento farmacológico , Asma/patologia , Movimento Celular , Proliferação de Células , Células Cultivadas , Humanos , Metaloproteinase 1 da Matriz/fisiologia , Metaloproteinase 2 da Matriz/fisiologia , Metaloproteinase 3 da Matriz/fisiologia , Metaloproteinases da Matriz/fisiologia , Metaloproteinases da Matriz Associadas à Membrana , Compostos Orgânicos/farmacologia , Estresse Mecânico , Tenascina/metabolismo , Inibidor Tecidual de Metaloproteinase-2/fisiologia
17.
Cancer Res ; 64(6): 2083-9, 2004 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-15026347

RESUMO

Bone metastasis is the most frequent complication of prostate cancer (PC). Elucidation of the biological basis of this specificity is required for the development of approaches for metastatic inhibition. We investigated the possibility that the preferential attachment of PC cells to bone marrow endothelium (as opposed to endothelium from other organs) affects this specificity. We selected, from peptide phage-displayed libraries, peptide ligands to surfaces of PC cells (C4-2B) attenuated (30-40%) binding of C4-2B cells to bone marrow endothelial cells (BMECs). We then determined the molecules on the surface of C4-2B cells interacted with the selected peptides using column affinity chromatography and a cDNA expression phage-displayed library generated from C4-2B cells in T7 phage. We identified a phage from the cDNA library that specifically bound to one of the selected peptides-L11. This phage displayed the amino acid sequence homologous for the COOH-terminal portion of prostate-specific antigen (PSA). To examine the possible direct involvement of PSA in the interactions between PC and BMECs, we performed a cell-cell adhesion assay. Antibodies to PSA attenuated PC cells adhesion to BMECs. In addition, exogenous proteolytically active PSA modulated this adhesion. Finally, inactivation of mRNA coding PSA by a small interfering RNA (siRNA) diminished C4-2B cell adhesion to BMECs. These results indicate that PSA expressed as secreted and surface-associated molecules in C4-2B cells is involved in cell-cell interactions and/or digests components of bone marrow endothelium for preferential adhesion and penetration of PC cells. The suggested experimental approach is a promising strategy for identification of cell surface molecules involved in intercellular interactions.


Assuntos
Células da Medula Óssea/metabolismo , Endotélio Vascular/metabolismo , Neoplasias da Próstata/metabolismo , Apoptose , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Adesão Celular , Movimento Celular , Humanos , Masculino , Invasividade Neoplásica , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Biblioteca de Peptídeos , Antígeno Prostático Específico/antagonistas & inibidores , Antígeno Prostático Específico/metabolismo , Neoplasias da Próstata/patologia , Ligação Proteica , RNA Interferente Pequeno/farmacologia , Células Tumorais Cultivadas
18.
Cancer Res ; 64(4): 1229-32, 2004 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-14983875

RESUMO

Emmprin (CD147; basigin) is a plasma membrane glycoprotein, enriched on the surface of many cancer cells, which induces matrix metalloproteinase synthesis via cell-cell interactions. Elevated emmprin production causes increased growth in vivo of human mammary carcinoma cells. In this study, we show that elevation of emmprin expression in less aggressive human carcinoma cells, which normally express low emmprin levels, induces the ability to grow under anchorage-independent conditions. We also found that elevated emmprin expression stimulates hyaluronan production and that the effect of emmprin on anchorage-independent growth is dependent on hyaluronan. Furthermore, emmprin stimulates cell survival pathway signaling in a hyaluronan-dependent manner. From these and other studies we conclude that emmprin enhances several malignant properties of cancer cells, including anchorage-independent growth, invasiveness, and chemoresistance.


Assuntos
Antígenos CD , Antígenos de Neoplasias , Neoplasias da Mama/patologia , Ácido Hialurônico/biossíntese , Glicoproteínas de Membrana/fisiologia , Basigina , Neoplasias da Mama/metabolismo , Divisão Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Feminino , Humanos
19.
Curr Cancer Drug Targets ; 16(7): 618-30, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26881932

RESUMO

Membrane type 1-matrix metalloproteinase (MT1-MMP, MMP-14) is associated with cancer invasion and metastasis leading to poor patient prognosis. MT1-MMP mediates cancer cell invasion via degradation of basement membrane and extracellular matrix, and induction of cell migration. However, MT1-MMP expression in the cancer stroma can drive invasion of carcinoma cells in vivo, suggesting MT1-MMP may also promote cancer invasiveness via paracrinemediated mechanisms. A major step in cancer cell metastasis is thought to be an epithelial-mesenchymal transition (EMT), in which carcinoma cells evolve from a stationary epithelial phenotype to a more motile mesenchymal phenotype. We demonstrate here that EMT is triggered by MT1-MMP-mediated activation of TGF-. signaling, involving induction of CUTL1 and subsequently, of Wnt5a. Mesenchymal-like cancer cells expressing endogenous MT1-MMP reverted to an epithelial phenotype when MT1-MMP, SMAD4, CUTL1, or Wnt5a expression or TGF-. activity was inhibited. Wnt5a knockdown in MT1- MMP expressing LNCaP cells caused decreased cell migration and cell growth in soft agar. While MT1-MMP expression did not affect total TGF-. level, MT1-MMP catalytic activity increased the availability of active TGF-., enabling MT1-MMP-expressing cells to activate the EMT in nearby cells. MT1-MMP-expressing cells induced co-cultured non-MT1-MMP-expressing cells to undergo EMT by a TGF-.-dependent process. These results highlight a pathway by which tumor invasiveness may be expanded via MT1-MMP-mediated activation of TGF-. signaling, enabling autocrine and paracrine-mediated induction of EMT.


Assuntos
Transição Epitelial-Mesenquimal/fisiologia , Metaloproteinase 14 da Matriz/metabolismo , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Proteínas de Homeodomínio/metabolismo , Humanos , Invasividade Neoplásica/patologia , Proteínas Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição , Proteína Wnt-5a/metabolismo
20.
J Clin Oncol ; 22(23): 4683-90, 2004 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-15570070

RESUMO

PURPOSE: To determine whether a matrix metalloproteinase inhibitor improves progression-free survival (PFS) in patients with metastatic breast cancer who have responding or stable disease after first-line chemotherapy. PATIENTS AND METHODS: One hundred seventy-nine eligible patients were randomly assigned to receive oral marimastat (10 mg bid; n = 114) or a placebo (n = 65) within 3 to 6 weeks of completing six to eight cycles of first-line doxorubicin- and/or taxane-containing chemotherapy for metastatic disease. Patients were evaluated every 3 months until disease progression. RESULTS: When comparing placebo with marimastat, there was no significant difference in PFS (median, 3.1 months v 4.7 months, respectively; hazard ratio, 1.26; 95% CI, 0.91 to 1.74; P = .16) or overall survival (median, 26.6 months v 24.7 months, respectively; hazard ratio, 1.03; 95% CI, 0.73 to 1.46; P = .86). Patients treated with marimastat were more likely to develop grade 2 or 3 musculoskeletal toxicity (MST), a known complication of the drug indicative of achieving a biologic effect, compared with patients administered placebo (63% v 22%, respectively; P < .0001). Patients with grade 2 or 3 MST had significantly inferior survival compared with patients who had grade 0 or 1 MST (median, 22.5 months v 28.2 months; P = .04). In addition, patients who had a marimastat plasma concentration of at least 10 ng/mL at month 1 and/or 3 were significantly more likely to have grade 2 to 3 MST (P < .0001). CONCLUSION: Marimastat does not prolong PFS when used after first-line chemotherapy for metastatic breast cancer. Patients with higher marimastat levels exhibited MST, and MST was associated with inferior survival.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Ácidos Hidroxâmicos/administração & dosagem , Invasividade Neoplásica/patologia , Administração Oral , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias da Mama/mortalidade , Relação Dose-Resposta a Droga , Método Duplo-Cego , Esquema de Medicação , Feminino , Seguimentos , Humanos , Pessoa de Meia-Idade , Terapia Neoadjuvante , Estadiamento de Neoplasias , Probabilidade , Modelos de Riscos Proporcionais , Valores de Referência , Medição de Risco , Análise de Sobrevida , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa