Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Biochem Biophys Res Commun ; 522(1): 53-60, 2020 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-31735331

RESUMO

Esophageal squamous cell carcinoma (ESCC) is one of the most common cancers worldwide. Protein tyrosine phosphatase 1B (PTP1B) is a member of protein tyrosine phosphatases (PTPs) family. In our previous work, PTP1B was found to be overexpressed in ESCC tissues and made contributions to the the cell migration and invasion as well as lung metastasis of ESCC. In this study, we explored the underlying molecular mechanisms. PTP1B enhanced cell migration and invasion by promoting epidermal growth factor receptor (EGFR) expression in ESCC, which was relied on phosphatase activity of PTP1B. Using GST-pulldown combined with LC/MS/MS, we found that nonmuscle myosin IIA (MYH9) was a novel substrate of PTP1B in ESCC cells. PTP1B dephosphorylated MYH9 at Y1408, by which PTP1B up-regulated EGFR expression and enhanced cell migration and invasion in ESCC. In conclusion, our study first reported that PTP1B was the positive regulator of EGFR by dephosphorylating MYH9 at Y1408 to promote cell migration and invasion, which revealed the regulatory mechanism of PTP1B-MYH9-EGFR axis in ESCC.


Assuntos
Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas do Esôfago/metabolismo , Regulação Neoplásica da Expressão Gênica , Cadeias Pesadas de Miosina/química , Proteína Tirosina Fosfatase não Receptora Tipo 1/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Receptores ErbB/metabolismo , Humanos , Invasividade Neoplásica , Fosforilação , Regulação para Cima
2.
Yi Chuan ; 37(4): 360-366, 2015 Apr.
Artigo em Zh | MEDLINE | ID: mdl-25881701

RESUMO

Immunoglobulin mu binding protein 2 (IGHMBP2) is located in 11q13.2, which is frequently amplified in esophageal squamous cell carcinoma (ESCC). IGHMBP2 encodes a helicase involved in DNA replication and repair. IGHMBP2 protein also regulates gene transcription. The present study aims to explore the amplification of IGHMBP2 and its potential role in ESCC. A further analysis of our previously reported array-CGH data showed that IGHMBP2 was amplified in 28.9% of primary ESCC tumors. Fluorescence in situ hybridization (FISH) and Western blot showed that IGHMBP2 was amplified and overexpressed in KYSE30, KYSE180, KYSE510 and KYSE150 esophageal cancer cell lines. Transwell assays demonstrated that knockdown of IGHMBP2 in KYSE30 and KYSE150 inhibited cell invasion and migration, and increased the expression levels of E-cadherin. When rescue plasmids expressing IGHMBP2 were introduced, the abilities of cell invasion and migration were restored. These data suggest that IGHMBP2 overexpression may promote invasion and migration of ESCC cells through down-regulation of E-cadherin.


Assuntos
Carcinoma de Células Escamosas/genética , Movimento Celular , Proteínas de Ligação a DNA/genética , Neoplasias Esofágicas/genética , Fatores de Transcrição/genética , Caderinas/genética , Caderinas/metabolismo , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas/fisiopatologia , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/metabolismo , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patologia , Neoplasias Esofágicas/fisiopatologia , Carcinoma de Células Escamosas do Esôfago , Humanos , Invasividade Neoplásica , Transdução de Sinais , Fatores de Transcrição/metabolismo
3.
Oncogene ; 37(24): 3260-3274, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29551772

RESUMO

Esophageal squamous cell carcinoma (ESCC) is one of the malignancies in digestive system, with a low 5-year survival rate. We previously revealed that Sequestosome 1 (SQSTM1/p62) protein levels were upregulated in ESCC tissues. However, it is unclear about the function of p62 and the underlying mechanism. Here, we used immunofluorescence and immunohistochemistry to investigate the expression of p62 in ESCC. Western blotting, quantitative RT-PCR, colony formation assay, flow cytometry, immunoprecipitation and xenograft tumor assay were used to analyze the role of p62 in vitro and vivo. Here, we showed that p62 serves as a regulator of cell apoptosis under serum starvation condition in ESCC cells. Through activating the protein kinase C iota (PKCiota)-S-phase kinase-associated protein 2 (SKP2) signaling pathway, p62 enhances cell apoptosis resistance and colony formation in vitro and tumor growth in mouse models. Through interaction with the domains PB1, p62 upregulated the expression of PKCiota and then depressed the ubiquitin-mediated proteasomal degradation of SKP2. p62-silencing combined with a PKCiota inhibitor ATM significantly enhanced cell apoptosis and inhibited cell survival. Immunohistochemical analysis revealed a positive association between the expression of p62 and SKP2 in primary ESCC tissues. And importantly, p62 presented a markedly cytoplasmic translocation in cancerous cells, including in 16 (30.76%) tumors at stage T1, as compared with its nuclear location in normal esophageal epithelial cells. In summary, p62 plays an anti-apoptotic role in ESCC cells via stabilizing SKP2 under serum starvation condition. These data suggest that p62 might be an early biomarker and a candidate therapeutic target of ESCC.


Assuntos
Apoptose/genética , Neoplasias Esofágicas/genética , Carcinoma de Células Escamosas do Esôfago/genética , Proteínas Quinases Associadas a Fase S/metabolismo , Proteína Sequestossoma-1/genética , Animais , Linhagem Celular Tumoral , Meios de Cultura , Neoplasias Esofágicas/patologia , Carcinoma de Células Escamosas do Esôfago/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Camundongos Endogâmicos BALB C , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Estabilidade Proteica , Proteínas Quinases Associadas a Fase S/genética , Proteína Sequestossoma-1/metabolismo , Ubiquitinação , Ensaios Antitumorais Modelo de Xenoenxerto
4.
J Mol Med (Berl) ; 96(8): 807-817, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29959473

RESUMO

Both polo-like kinase 1 (PLK1) and mammalian/mechanistic target of rapamycin (mTOR) are attractive therapeutic targets for cancer therapy. However, the efficacy of the combined inhibition of both pathways for treating esophageal squamous cell carcinoma (ESCC), an aggressive malignancy with poor prognosis, remains unknown. In this study, we found that suppression of PLK1 by specific siRNA or inhibitor attenuated mTOR activity in ESCC cells. Phosphorylated S6, a downstream effector of mTOR signaling, was significantly correlated with overexpression of PLK1 in a subset of ESCC. These data suggest that PLK1 activates mTOR signaling in vitro and in vivo. More importantly, the mTOR inhibitor rapamycin synergized with PLK1 inhibitor BI 2536 to inhibit ESCC cell proliferation in culture and in mice. Notably, combined treatment with BI 2536 and rapamycin produced more potent inhibitory effects on the activation of S6 and AKT than either alone. Further analysis reveals that PLK1 modulates both mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2) cascades. Therefore, dual inhibition of PLK1 and mTOR yields stronger antitumor effects, at least partially due to synergistic abrogated the activation of S6, eukaryotic initiation factor 4E-binding protein 1 (4E-BP1), and AKT by cooperatively blocking mTORC1 and mTORC2 cascades. These results provide evidence that the mTOR inhibitor rapamycin synergistically enhances the antitumor effect of PLK1 inhibitor BI 2536 in ESCC cells. Simultaneous targeting of PLK1 and mTOR may thus be a novel and promising therapeutic strategy for ESCC. KEY MESSAGES: PLK1 potentiates both mTORC1 and mTORC2 activities in ESCC cells. PLK1 expression positively correlated with mTOR activity in a subset of ESCC. Co-targeting of PLK1 and mTOR produced stronger antitumor effects partially due to synergistic inhibition of AKT, 4E-BP1 and S6 through cooperatively blocking mTORC2 and mTORC1 cascades. Combination targeting of PLK1 and mTOR may be a novel and promising therapeutic strategy for ESCC treatment.


Assuntos
Proteínas de Ciclo Celular/antagonistas & inibidores , Carcinoma de Células Escamosas do Esôfago/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Serina-Treonina Quinases TOR/antagonistas & inibidores , Animais , Apoptose/efeitos dos fármacos , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Carcinoma de Células Escamosas do Esôfago/tratamento farmacológico , Carcinoma de Células Escamosas do Esôfago/patologia , Feminino , Expressão Gênica , Humanos , Imuno-Histoquímica , Camundongos , Terapia de Alvo Molecular , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Quinase 1 Polo-Like
5.
Bone ; 39(6): 1226-35, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16926124

RESUMO

Fragility fractures, including neck of femur fractures, result from reductions in the amount, quality and architecture of bone. The aim of this study was to compare the cancellous bone structure, and static indices of bone turnover, in female patients, who had sustained fragility fracture at the femoral neck, with age-matched females without fragility fracture. Bone samples were taken from the intertrochanteric region of the proximal femur of female patients undergoing hip arthroplasty surgery for a subcapital fragility fracture of the femoral neck (#NOF) or from age-matched female control individuals at routine autopsy. Contiguous bone samples were analyzed for undecalcified histomorphometry and for mRNA expression. The histomorphometric data, which were normally distributed, indicated no difference between the mean values for any of the structural parameters in control and fracture samples. In particular, the bone volume (BV/TV) values were not different and did not change significantly with age in these cohorts of individuals aged >65 years. The static indices of bone turnover, eroded surface (ES/BS) and osteoid surface (OS/BS), were positively correlated with age in the >65-year-old control group (p < 0.055 and p < 0.03, respectively). The median values for these indices were not different between the fracture and control groups. However, both the median and the range of OS/BS values were increased for >65-year-old controls compared with a group of younger females aged <65 years, suggesting an increase in bone formation surface in older females in the proximal femur after 65 years of age. When the data were further interrogated, a reduction in the percentage osteoid surface to eroded surface quotient (OS/ES) was found for the fracture group compared with the age-matched control group suggesting a reduced adaptive modeling drift capability in the fracture group. In contiguous bone samples, increased median values for receptor activator of nuclear factor kappa beta (RANK) and interleukin-6 (IL-6) mRNA expression were observed in the fracture group. Study of cultured human osteoblasts showed that recombinant human IL-6 (rhIL-6) inhibited osteoblast differentiation, as measured by an increase in the immature osteoblast marker, STRO-1 and concomitantly decreased expression of the osteoblast maturation marker, alkaline phosphatase. Importantly, cells cultured in the presence of IL-6 showed significantly less mineral deposition in vitro compared with control cultures. These data suggest that perturbations in bone formation surface, relative to resorption surface, are potentially important in producing bone in the proximal femur with increased propensity to fracture.


Assuntos
Reabsorção Óssea/patologia , Fraturas do Colo Femoral/patologia , Idoso , Idoso de 80 Anos ou mais , Remodelação Óssea , Reabsorção Óssea/genética , Estudos de Casos e Controles , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Feminino , Fraturas do Colo Femoral/genética , Colo do Fêmur/metabolismo , Colo do Fêmur/patologia , Expressão Gênica , Humanos , Interleucina-6/farmacologia , Pessoa de Meia-Idade , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteogênese , Proteínas Recombinantes/farmacologia
6.
Cancer Res ; 63(17): 5438-45, 2003 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-14500379

RESUMO

Multiple myeloma (MM) is an incurable B-cell malignancy able to mediate massive destruction of the axial skeleton. The aim of this study was to examine the involvement of the tumor necrosis factor-ligand family member, receptor activator of nuclear factor-kappaB ligand (RANKL), and its naturally occurring antagonist, osteoprotegerin (OPG), in MM biology. Using flow cytometry and two independent anti-RANKL antibodies, we demonstrate RANKL expression in CD38(+++)CD45(+) and CD38(+++)CD45(-) myeloma plasma cell (MPC) subpopulations derived from patients with osteolytic MM. In addition, highly purified subpopulations of MPC express mRNA for both transmembrane and soluble RANKL isoforms but lack expression of OPG mRNA and protein. We also show that RANKL expressed by MPC is functional as in vitro coculture of CD38(+++)CD45(+) and CD38(+++)CD45(-) MPC subpopulations with peripheral blood mononuclear cells resulted in the formation of multinucleate, tartrate-resistant acid phosphatase-positive osteoclasts-like cells capable of forming typical resorption pits. Furthermore, high expression of membrane-associated RANKL by CD38(+++) MPC correlated with the presence of multiple radiological bone lesions in individuals with MM. Together, our data strongly suggest that RANKL expression by MPC confers on them the ability to participate directly in the formation of osteoclast in vivo and extends our knowledge of the involvement of RANKL and OPG in the osteolysis characteristic of this disease.


Assuntos
Proteínas de Transporte/biossíntese , Glicoproteínas de Membrana/biossíntese , Mieloma Múltiplo/metabolismo , Osteoclastos/patologia , Osteólise/metabolismo , Proteínas de Transporte/genética , Citometria de Fluxo , Expressão Gênica , Humanos , Glicoproteínas de Membrana/genética , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia , Osteólise/diagnóstico por imagem , Osteólise/genética , Osteólise/patologia , Plasmócitos/metabolismo , Ligante RANK , Radiografia , Receptor Ativador de Fator Nuclear kappa-B , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
J Bone Miner Res ; 18(6): 1088-98, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12817763

RESUMO

Human osteoblast phenotypes that support osteoclast differentiation and bone formation are not well characterized. Osteoblast differentiation markers were examined in relation to RANKL expression. RANKL expression was induced preferentially in immature cells. These results support an important link between diverse osteoblast functions. Cells of the osteoblast lineage support two apparently distinct functions: bone formation and promotion of osteoclast formation. The aim of this study was to examine the relationship between these phenotypes in human osteoblasts (NHBC), in terms of the pre-osteoblast marker, STRO-1, and the mature osteoblast marker, alkaline phosphatase (AP), and the expression of genes involved in osteoclast formation, RANKL and OPG. The osteotropic stimuli, 1alpha,25(OH)2vitamin D3 (vitD3) and dexamethasone, were found to have profound proliferative and phenotypic effects on NHBCs. VitD3 inhibited NHBC proliferation and increased the percentage of cells expressing STRO-1 over an extended culture period, implying that vitD3 promotes and maintains an immature osteogenic phenotype. Concomitantly, RANKL mRNA expression was upregulated and maintained in NHBC in response to vitD3. Dexamethasone progressively promoted the proliferation of AP-expressing cells, resulting in the overall maturation of the cultures. Dexamethasone had little effect on RANKL mRNA expression and downregulated OPG mRNA expression in a donor-dependent manner. Regression analysis showed that RANKL mRNA expression was associated negatively with the percentage of cells expressing AP (p < 0.01) in vitD3- and dexamethasone-treated NHBCs. In contrast, RANKL mRNA expression was associated positively with the percentage of STRO-1+ cells (p < 0.01). In NHBCs sorted by FACS based on STRO-1 expression (STRO-1bright and STRO-1dim populations), it was found that vitD3 upregulated the expression of RANKL mRNA preferentially in STRO-1bright cells. The results suggest that immature osteoblasts respond to osteotropic factors in a potentially pro-osteoclastogenic manner. Additionally, the dual roles of osteoblasts, in supporting osteoclastogenesis or forming bone, may be performed by the same lineage of cells at different stages of their maturation.


Assuntos
Proteínas de Transporte/genética , Diferenciação Celular/fisiologia , Regulação da Expressão Gênica/fisiologia , Glicoproteínas de Membrana/genética , Osteoblastos/citologia , Proteínas de Transporte/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Dexametasona/farmacologia , Fluoresceínas/farmacologia , Corantes Fluorescentes , Gliceraldeído-3-Fosfato Desidrogenases/efeitos dos fármacos , Gliceraldeído-3-Fosfato Desidrogenases/genética , Humanos , Glicoproteínas de Membrana/efeitos dos fármacos , Osteoblastos/efeitos dos fármacos , Fenótipo , Ligante RANK , RNA Mensageiro/genética , Receptor Ativador de Fator Nuclear kappa-B , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Succinimidas/farmacologia , Transcrição Gênica/efeitos dos fármacos , Vitamina D/farmacologia
9.
J Bone Miner Res ; 19(1): 147-54, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14753746

RESUMO

UNLABELLED: Bisphosphonates are used to prevent osteoclast-mediated bone loss. Zoledronic acid inhibits osteoclast maturation indirectly by increasing OPG protein secretion and decreasing transmembrane RANKL expression in human osteoblasts. The decreased transmembrane RANKL expression seems to be related to the upregulation of the RANKL sheddase, TACE. INTRODUCTION: Bisphosphonates (BPs) exhibit high affinity for hydroxyapatite mineral in bone and are used extensively to treat malignancy-associated bone disease and postmenopausal bone loss by inhibiting osteoclast (OC)-mediated bone resorption. MATERIALS AND METHODS: We examined the effect of the most potent nitrogen-containing BP available, zoledronic acid (ZOL), on the expression of RANKL and osteoprotegerin (OPG), critical factors in the regulation of OC formation and activation, in primary osteoblast (OB)-like cells derived from human bone, using flow cytometry, ELISA, semiquantitative reverse transcriptase-polymerase chain reaction (RT-PCR), in situ immunofluorescence staining, and Western blotting. RESULTS: Our studies show that ZOL, while not significantly affecting RANKL or OPG gene expression, markedly increased OPG protein secretion and reduced transmembrane RANKL protein expression in OB-like cells. The reduction in transmembrane RANKL expression was preceded by a marked increase in the expression of the metalloprotease-disintegrin, TNF-alpha converting enzyme (TACE). In addition, the decreased transmembrane expression of RANKL could be partially reversed by a TACE inhibitor, TAPI-2. CONCLUSIONS: Our studies indicate that ZOL, in addition to its direct effects on mature OCs, may inhibit the recruitment and differentiation of OCs by cleavage of transmembrane RANKL in OB-like cells by upregulating the sheddase, TACE.


Assuntos
Proteínas de Transporte/metabolismo , Difosfonatos/farmacologia , Imidazóis/farmacologia , Glicoproteínas de Membrana/metabolismo , Metaloendopeptidases/metabolismo , Osteoblastos/efeitos dos fármacos , Proteínas ADAM , Proteína ADAM17 , Western Blotting , Proteínas de Transporte/genética , Células Cultivadas , Citometria de Fluxo , Expressão Gênica/efeitos dos fármacos , Humanos , Ácidos Hidroxâmicos/farmacologia , Imuno-Histoquímica , Glicoproteínas de Membrana/genética , Metaloendopeptidases/antagonistas & inibidores , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteoprotegerina , Ligante RANK , Receptor Ativador de Fator Nuclear kappa-B , Receptores do Fator de Necrose Tumoral/metabolismo , Ácido Zoledrônico
10.
Bone ; 34(1): 112-23, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14751568

RESUMO

Previous studies have attributed the increase in bone mass observed following bisphophonate (BP) therapy to their effects on bone-resorbing osteoclasts (OCs). However, recent evidence suggests that BPs can also act directly on bone forming osteoblasts (OBs) to increase their anabolic activity. Using an established model of in vitro OB differentiation, we found that the potent nitrogen-containing BP, zoledronic acid (ZOL), may enhance the bone forming potential of human adult OB-like cells in vitro by inducing their differentiation. ZOL dose dependently induced both cytostasis and cell death in OB-like cells at concentrations of 0.5 microM or greater. Cells expressing high levels of the osteoprogenitor antigen, STRO-1, exhibited a greater proliferative potential than STRO-1negative/dim cells, and were more susceptible to the cytostatic and apoptotic effects of ZOL. ZOL was also found to promote bone cell differentiation, as evidenced by an increase in the number of cells exhibiting a more differentiated (STRO-1(-)/AP+ and STRO-1(-)/AP-) phenotype. Analysis of gene expression, using semi-quantitative RT-PCR, demonstrated that ZOL treatment resulted in a significant upregulation of osteocalcin (OCN) and bone morphogenetic protein-2 (BMP-2) gene expression. Furthermore, in vitro mineralisation studies revealed that ZOL enhanced mineralised matrix formation at concentrations between 5 and 25 microM. These results show that, in addition to its direct effects on OCs, ZOL also directly affects the proliferation and differentiation of human OB-like cells in vitro and may enhance bone formation in vivo.


Assuntos
Calcificação Fisiológica/efeitos dos fármacos , Difosfonatos/química , Difosfonatos/farmacologia , Imidazóis/química , Imidazóis/farmacologia , Nitrogênio/química , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Contagem de Células , Morte Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Citometria de Fluxo , Humanos , Regulação para Cima/efeitos dos fármacos , Ácido Zoledrônico
12.
Clin Cancer Res ; 20(23): 6153-62, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25231404

RESUMO

PURPOSE: We previously revealed that the calreticulin (CRT) gene is a candidate oncogene promoting cell migration and invasion and that neuropilin-1 (NRP1) is a possible effector downstream of CRT in esophageal squamous carcinoma cells. This study aims to explore the mechanisms underlying the migration and invasion of esophageal cancer cells regulated by CRT through NRP1. EXPERIMENTAL DESIGN: Quantitative reverse-transcription polymerase chain reaction, Western blot analysis, chromatin immunoprecipitation, and reporter gene assays were used to investigate the relationship between CRT and NRP1. In vitro and in vivo assays were carried out to evaluate the effects of NRP1 on malignant phenotypes of ESCC cells and tumor metastasis in NOD/SCID mice. Immunohistochemistry was performed to analyze the expression of CRT and NRP1 in esophageal squamous cell carcinomas (ESCC). RESULTS: Knockdown of CRT decreased the expression of NRP1. Inhibition of NRP1 reduced ESCC cell motility in vitro and experimental metastasis in vivo. Ectopic expression of NRP1 rescued the defects of cell migration and invasion in CRT-shRNA cells. CRT depletion inhibited STAT5A phosphorylation at the Y694 site via a CaMKII-independent pathway. Moreover, STAT5A directly regulated NRP1 transcription. Knockdown of CRT or NRP1 led to a downregulation of MMP2, MMP9, and FAK. Notably, positive correlation was found between CRT and NRP1 expression in ESCC tissues (P = 5.87 × 10(-5)). CRT and NRP1 coexpression was significantly associated with lymph node metastasis (P = 0.025). CONCLUSIONS: Our findings suggest that NRP1 is a critical downstream effector of CRT in promoting cell migration and invasion, which might contribute to the metastasis of ESCC.


Assuntos
Calreticulina/genética , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/metabolismo , Regulação Neoplásica da Expressão Gênica , Neuropilina-1/genética , Fator de Transcrição STAT5/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Linhagem Celular Tumoral , Movimento Celular/genética , Modelos Animais de Doenças , Neoplasias Esofágicas/patologia , Feminino , Técnicas de Silenciamento de Genes , Xenoenxertos , Humanos , Metástase Linfática , Camundongos , Transcrição Gênica
13.
Clin Cancer Res ; 19(1): 22-7, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23155186

RESUMO

Brentuximab vedotin (SGN-35), an intravenously administered CD30-specific antibody-drug conjugate, has recently been approved by the U.S. Food and Drug Administration for two indications, including (i) patients with Hodgkin lymphoma relapsing after autologous stem-cell transplantation (ASCT), or after two multidrug regimens in patients with Hodgkin lymphoma who are not candidates for ASCT; and (ii) patients with systemic anaplastic large cell lymphoma (ALCL) who failed at least one prior multidrug chemotherapy regimen. Patients with Hodgkin lymphoma and ALCL treated with brentuximab vedotin showed markedly high response rates for a single agent, exceeding 70% and 80% for Hodgkin lymphoma and ALCL, respectively. The complete response rate was equally as impressive, at 34% and 57% for Hodgkin lymphoma and ALCL, respectively. Results like these and from many other upcoming clinical trials, in which brentuximab vedotin is being investigated in the frontline setting, promise to profoundly change how we manage the CD30-positive lymphoproliferative malignancies. The mechanism of action, preclinical antitumor activity, and clinical activity of brentuximab vedotin against Hodgkin lymphoma, ALCL, and other CD30-expressing lymphomas are reviewed.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Doença de Hodgkin/tratamento farmacológico , Imunoconjugados/uso terapêutico , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/efeitos adversos , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Brentuximab Vedotin , Ensaios Clínicos Fase II como Assunto , Avaliação Pré-Clínica de Medicamentos , Transplante de Células-Tronco Hematopoéticas , Doença de Hodgkin/terapia , Humanos , Imunoconjugados/administração & dosagem , Imunoconjugados/efeitos adversos , Antígeno Ki-1/imunologia , Recidiva , Transplante Autólogo , Resultado do Tratamento
14.
Pharmacol Ther ; 136(1): 56-68, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22796518

RESUMO

Immunomodulatory drugs (IMiDs) have been used in hematologic malignancies for the last decade. However, the mechanism of action of IMiDs is largely unknown. Here we provide a comprehensive overview of pivotal studies, recent advances in the application of IMiDs in cancer as well as their effects on hematopoietic stem cells including the risk of secondary malignancies. IMiDs have a well-established role as first-line therapy for patients with newly diagnosed and relapsed/refractory multiple myeloma (MM). Variant combinations of IMiDs with other chemotherapy reagents show promising outcomes in MM. Recent concerns on increased rate of secondary cancer in MM patients treated with maintenance lenalidomide were raised. But analysis of maintenance studies showed that the benefit of maintenance outweighs the risk of secondary cancers in MM. IMiDs also show efficacy in myelodysplastic syndrome (MDS)/acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), Non-Hodgkin's lymphoma (NHL) and myelofibrosis (MF), but not in solid tumors. The major adverse effects are venous thromboembolism, neuropathy and cytopenias. IMiDs induce expansion and self-renewal of CD34+ hematopoietic progenitors and inhibit lineage maturation/differentiation by affecting critical transcription factors which might contribute to myelosuppression effect of IMiDs.


Assuntos
Fatores Imunológicos/uso terapêutico , Neoplasias/tratamento farmacológico , Humanos , Fatores Imunológicos/efeitos adversos , Fatores Imunológicos/farmacologia , Lenalidomida , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Leucemia Mieloide Aguda/tratamento farmacológico , Linfoma/tratamento farmacológico , Mieloma Múltiplo/tratamento farmacológico , Síndromes Mielodisplásicas/tratamento farmacológico , Segunda Neoplasia Primária/induzido quimicamente , Talidomida/análogos & derivados , Talidomida/uso terapêutico
15.
Anticancer Res ; 32(4): 1449-52, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22493384

RESUMO

Metastatic carcinoma of the pancreas from another primary site is uncommon and it accounts for 2-5% of all pancreatic cancer cases. We reported the case of one patient with pancreatic metastasis from colon carcinoma in the past and would like to add another six cases of pancreatic metastases from different types of cancer. The diagnosis of cancer metastatic to the pancreas should be suspected when patients have a history of malignancy, especially of kidney, skin, lung, colon and breast cancer. Besides imaging studies, such as computed tomography (CT) scan, bone scan and positron emission tomography (PET)/CT scan, endoscopic ultrasound (EUS)-guided biopsy has most value in ruling out second primary pancreatic cancer. The prognosis of pancreatic metastases is essentially determined by the underlying primary cancer and the potential treatment options.


Assuntos
Neoplasias Pancreáticas/secundário , Adulto , Idoso , Neoplasias do Colo/patologia , Feminino , Humanos , Masculino
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA