Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
J BUON ; 26(1): 293, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33721478

RESUMO

The Editors of JBUON issue an Expression of Concern to 'Boswellic acid exerts potent anticancer effects in HCT-116 human colon cancer cells mediated via induction of apoptosis, cell cycle arrest, cell migration inhibition and inhibition of PI3K/AKT signalling pathway', by Dan Wang, Shuke Ge, Jichang Bai, Yongwei Song, JBUON 2018;23(2):340-345; PMID:29745074. Following the publication of the above article, readers drew to our attention that part of the data was possibly unreliable. We sent emails to the authors with a request to provide the raw data to prove the originality, but received no reply. Therefore, as we continue to work through the issues raised, we advise readers to interpret the information presented in the article with due caution. We thank the readers for bringing this matter to our attention. We apologize for any inconvenience it may cause.


Assuntos
Inibição de Migração Celular/genética , Neoplasias do Colo/tratamento farmacológico , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Triterpenos/uso terapêutico , Apoptose , Neoplasias do Colo/patologia , Células HCT116 , Humanos , Transdução de Sinais , Triterpenos/farmacologia
2.
Oncogene ; 40(8): 1516-1530, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33452462

RESUMO

The basic leucine zipper ATF-like transcription factor 2 (BATF2) has been implicated in inflammatory responses and anti-tumour effects. Little, however, is known regarding its extracellular role in maintaining a non-supportive cancer microenvironment. Here, we show that BATF2 inhibits glioma growth and myeloid-derived suppressor cells (MDSCs) recruitment. Interestingly, extracellular vesicles (EVs) from BATF2-overexpressing glioma cell lines (BATF2-EVs) inhibited MDSCs chemotaxis in vitro. Moreover, BATF2 inhibited intracellular SDF-1α and contributes to decreased SDF-1α in EVs. In addition, BATF2 downregulation-induced MDSCs recruitment were reversed by blocking SDF-1α/CXCR4 signalling upon AMD3100 treatment. Specifically, detection of EVs in 24 pairs of gliomas and healthy donors at different stages revealed that the abundance of BATF2-positive EVs in plasma (BATF2+ plEVs) can distinguish stage III-IV glioma from stage I-II glioma and healthy donors. Taken together, our study identified novel regulatory functions of BATF2 in regulating MDSCs recruitment, providing a prognostic value in terms of the number of BATF2+ plEVs in glioma stage.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/genética , Quimiocina CXCL12/genética , Glioblastoma/genética , Receptores CXCR4/genética , Proteínas Supressoras de Tumor/genética , Adulto , Idoso , Animais , Benzilaminas/farmacologia , Linhagem Celular Tumoral , Inibição de Migração Celular/genética , Ciclamos/farmacologia , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioblastoma/sangue , Glioblastoma/patologia , Xenoenxertos , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Células Supressoras Mieloides/metabolismo , Células Supressoras Mieloides/patologia , Estadiamento de Neoplasias , Transdução de Sinais/efeitos dos fármacos , Microambiente Tumoral/efeitos dos fármacos , Adulto Jovem
3.
Laryngoscope ; 130(11): E640-E645, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-31872875

RESUMO

OBJECTIVE: This study aimed to investigate the role of lncRNA miR143HG in laryngeal squamous cell carcinoma (LSCC). METHODS: Quantitative polymerase chain reaction (PCR) and paired t test were used to measure and compare expression levels of miR143HG and miR-21 in LSCC and nontumor tissues. To analyze the interactions between miR143HG and miR-21, UM-SCC-17A cells were transfected miR143HG expression vector or miR-21 mimic. The effects of miR143HG and miR-21 overexpression on UM-SCC-17A cell invasion and migration were analyzed by transwell assays. RESULTS: We found that miR143HG was downregulated in LSCC and inversely correlated with miR-21. In LSCC cells, miR143HG overexpression led to the downregulated expression of miR-21, whereas miR-21 overexpression failed to affect miR143HG. Methylation-specific PCR results showed that miR143HG overexpression led to increased methylation of miR-21. Low expression levels of miR143HG were correlated with poor survival. Overexpression of miR143HG led to decreased, whereas miR-21 overexpression resulted in increased rate of LSCC cell migration and invasion. In addition, miR-21 overexpression led to reduced effects of miR143HG on cell invasion and migration. CONCLUSION: Therefore, miR143HG suppresses miR-21 via methylation to regulate cell behaviors in LSCC. LEVEL OF EVIDENCE: NA Laryngoscope, 130:E640-E645, 2020.


Assuntos
Inibição de Migração Celular/genética , Neoplasias Laríngeas/genética , MicroRNAs/metabolismo , RNA Longo não Codificante/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Adulto , Idoso , Feminino , Humanos , Masculino , Metilação , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase em Tempo Real
4.
BMC Cancer ; 14: 181, 2014 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-24625091

RESUMO

BACKGROUND: SMAD4 is a gastrointestinal malignancy-specific tumor suppressor gene found mutated in one third of colorectal cancer specimens and half of pancreatic tumors. SMAD4 inactivation by allelic deletion or intragenic mutation mainly occurs in the late stage of human pancreatic ductal adenocarcinoma (PDAC). Various studies have proposed potential SMAD4-mediated anti-tumor effects in human malignancy; however, the relevance of SMAD4 in the PDAC molecular phenotype has not yet been fully characterized. METHODS: The AsPC-1, CFPAC-1 and PANC-1 human PDAC cell lines were used. The restoration or knockdown of SMAD4 expression in PDAC cells were confirmed by western blotting, luciferase reporter and immunofluorescence assays. In vitro cell proliferation, xenograft, wound healing, quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR), Western blotting, and immunohistochemistry analysis were conducted using PDAC cells in which SMAD4 was either overexpressed or knocked down. RESULTS: Here, we report that re-expression of SMAD4 in SMAD4-null PDAC cells does not affect tumor cell growth in vitro or in vivo, but significantly enhances cells migration in vitro. SMAD4 restoration transcriptionally activates the TGF-ß1/Nestin pathway and induces expression of several transcriptional factors. In contrast, SMAD4 loss in PDAC leads to increased expression of E-cadherin, vascular endothelial growth factor (VEGF), epidermal growth factor receptor (EGFR) and CD133. Furthermore, SMAD4 loss causes alterations to multiple kinase pathways (particularly the phosphorylated ERK/p38/Akt pathways), and increases chemoresistance in vitro. Finally, PDAC cells with intact SMAD4 are more sensitive to TGF-ß1 inhibitor treatment to reduced cell migration; PDAC cells lacking SMAD4 showed decreased cell motility in response to EGFR inhibitor treatment. CONCLUSIONS: This study revealed the molecular basis for SMAD4-dependent differences in PDAC with the aim of identifying the subset of patients likely to respond to therapies targeting the TGF-ß or EGFR signaling pathways and of identifying potential therapeutic interventions for PDAC patients with SMAD4 defects.


Assuntos
Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Fenótipo , Proteína Smad4/deficiência , Animais , Carcinoma Ductal Pancreático/metabolismo , Linhagem Celular Tumoral , Inibição de Migração Celular/genética , Movimento Celular/genética , Proliferação de Células , Feminino , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Neoplasias Pancreáticas/metabolismo , Proteína Smad4/genética , Proteína Smad4/metabolismo , Neoplasias Pancreáticas
5.
Mol Immunol ; 58(2): 214-22, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24389043

RESUMO

INTRODUCTION: Mixed lineage kinase 3 (MLK3) is part of the intracellular regulatory system that connects extracellular cytokine or mitogen signals received through G-protein coupled receptors to changes in gene expression. MLK3 activation stimulates motility of epithelial cells and epithelial-derived tumor cells, but its role in mediating the migration of other cell types remains unknown. Since neutrophils play a crucial role in innate immunity and contribute to the pathogenesis of several diseases, we therefore examined whether MLK3 might regulate the motility of mouse neutrophils responding to a chemotactic stimulus, the model bacterial chemoattractant fMLP. METHODS: The expression of Mlk3 in mouse neutrophils was determined by immunocytochemistry and by RT-PCR. In vitro chemotaxis in a gradient of fMLP, fMLP-stimulated random motility, fMLP-stimulated F-actin formation were measured by direct microscopic observation using neutrophils pre-treated with a novel small molecule inhibitor of MLK3 (URMC099) or neutrophils obtained from Mlk3-/- mice. In vivo effects of MLK3 inhibition were measured by counting the fMLP-induced accumulation of neutrophils in the peritoneum following pre-treatment with URMC099 in wild-type C57Bl/6 or mutant Mlk3-/- mice. RESULTS: The expression of Mlk3 mRNA and protein was observed in neutrophils purified from wild-type C57Bl/6 mice but not in neutrophils from mutant Mlk3-/- mice. Chemotaxis by wild-type neutrophils induced by a gradient of fMLP was reduced by pre-treatment with URMC099. Neutrophils from C57Bl/6 mice pretreated with URMC099 and neutrophils from Mlk3-/- mice moved far less upon fMLP-stimulation and did not form F-actin as readily as untreated neutrophils from C57Bl/6 controls. In vivo recruitment of neutrophils into the peritoneum by fMLP was significantly reduced in wild-type mice treated with URMC099, as well as in untreated Mlk3-/- mice-thereby confirming the role of MLK3 in neutrophil migration. CONCLUSIONS: Mlk3 mRNA is expressed in murine neutrophils. Genetic or pharmacologic inhibition of MLK3 blocks fMLP-mediated motility of neutrophils both in vitro and in vivo, suggesting that MLK3 may be a therapeutic target in human diseases characterized by exuberant neutrophil migration.


Assuntos
Fatores Quimiotáticos/farmacologia , Doenças do Sistema Imunitário/induzido quimicamente , Transtornos Leucocíticos/induzido quimicamente , MAP Quinase Quinase Quinases/fisiologia , N-Formilmetionina Leucil-Fenilalanina/farmacologia , Animais , Inibição de Migração Celular/efeitos dos fármacos , Inibição de Migração Celular/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Células Cultivadas , Doenças do Sistema Imunitário/genética , Transtornos Leucocíticos/genética , MAP Quinase Quinase Quinases/antagonistas & inibidores , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ativação de Neutrófilo/efeitos dos fármacos , Ativação de Neutrófilo/genética , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Pirróis/farmacologia , MAP Quinase Quinase Quinase 11 Ativada por Mitógeno
6.
J Immunol ; 191(1): 424-33, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23729444

RESUMO

Glucocorticoid-induced leucine zipper (GILZ) is an anti-inflammatory protein first identified in T lymphocytes. We recently observed that GILZ is highly expressed in synovial endothelial cells in rheumatoid arthritis. However, the function of GILZ in endothelial cells is unknown. To investigate the actions of GILZ in this cell type, we induced GILZ expression in HUVECs via transient transfection. GILZ overexpression significantly reduced the capacity of TNF-stimulated HUVECs to support leukocyte rolling, adhesion, and transmigration. These effects were associated with decreased expression of E-selectin, ICAM-1, CCL2, CXCL8, and IL-6. Experiments in a human microvascular endothelial cell line demonstrated that TNF-inducible NF-κB activity was significantly inhibited by overexpression of GILZ. Exogenous GILZ inhibited TNF-induced NF-κB p65 DNA binding, although this occurred in the absence of an effect on p65 nuclear translocation, indicating that the mechanism of action of exogenous GILZ in endothelial cells differs from that reported in other cell types. GILZ overexpression also inhibited TNF-induced activation of p38, ERK, and JNK MAPKs, as well as increased expression of the MAPK inhibitory phosphatase, MKP-1. In contrast, silencing endogenous GILZ in glucocorticoid-treated HUVECs did not alter their capacity to support leukocyte interactions. These data demonstrate that exogenous GILZ exerts inhibitory effects on endothelial cell adhesive function via a novel pathway involving modulation of NF-κB p65 DNA binding and MAPK activity. Induction of GILZ expression in endothelial cells may represent a novel therapeutic modality with the potential to inhibit inflammatory leukocyte recruitment.


Assuntos
Endotélio Vascular/imunologia , Endotélio Vascular/metabolismo , Sistema de Sinalização das MAP Quinases/imunologia , Fator de Transcrição RelA/metabolismo , Fatores de Transcrição/genética , Migração Transendotelial e Transepitelial/imunologia , Adesão Celular/genética , Adesão Celular/imunologia , Comunicação Celular/imunologia , Linhagem Celular , Inibição de Migração Celular/genética , Inibição de Migração Celular/imunologia , Endotélio Vascular/fisiologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Leucócitos/imunologia , Leucócitos/metabolismo , Sistema de Sinalização das MAP Quinases/genética , Microcirculação/genética , Microcirculação/imunologia , Cultura Primária de Células , Distribuição Aleatória , Fatores de Transcrição/biossíntese , Fatores de Transcrição/fisiologia , Migração Transendotelial e Transepitelial/genética
7.
J Immunol ; 191(1): 395-406, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23733875

RESUMO

Cyclooxygenase (Cox)-2 dependent PGs modulate several functions in many pathophysiological processes, including migration of immune cells. In this study, we addressed the role of Cox-2 in macrophage migration by using in vivo and in vitro models. Upon thioglycolate challenge, CD11b(+) F4/80(+) macrophages showed a diminished ability to migrate to the peritoneal cavity in cox-2(-/-) mice. In vivo migration of cox-2(-/-) macrophages from the peritoneal cavity to lymph nodes, as well as cell adhesion to the mesothelium, was reduced in response to LPS. In vitro migration of cox-2(-/-) macrophages toward MCP-1, RANTES, MIP-1α, or MIP-1ß, as well as cell adhesion to ICAM-1 or fibronectin, was impaired. Defects in cell migration were not due to changes in chemokine receptor expression. Remarkably, cox-2(-/-) macrophages showed a deficiency in focal adhesion formation, with reduced phosphorylation of paxillin (Tyr(188)). Interestingly, expression of the p110γ catalytic subunit of PI3K was severely reduced in the absence of Cox-2, leading to defective Akt phosphorylation, as well as cdc42 and Rac-1 activation. Our results indicate that the paxillin/p110γ-PI3K/Cdc42/Rac1 axis is defective in cox-2(-/-) macrophages, which results in impaired cell adhesion and migration.


Assuntos
Inibição de Migração Celular/imunologia , Classe Ib de Fosfatidilinositol 3-Quinase/deficiência , Ciclo-Oxigenase 2/deficiência , Macrófagos Peritoneais/enzimologia , Macrófagos Peritoneais/imunologia , Fosfatidilinositol 3-Quinases/deficiência , Transdução de Sinais/imunologia , Animais , Adesão Celular/genética , Adesão Celular/imunologia , Inibição de Migração Celular/genética , Células Cultivadas , Classe Ib de Fosfatidilinositol 3-Quinase/fisiologia , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/fisiologia , Macrófagos Peritoneais/patologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Imunológicos , Fosfatidilinositol 3-Quinases/fisiologia , Transdução de Sinais/genética , Proteína cdc42 de Ligação ao GTP/deficiência , Proteínas rac1 de Ligação ao GTP/deficiência
8.
Med Oncol ; 30(2): 542, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23529765

RESUMO

Accumulating evidence has shown that microRNAs are involved in multiple processes in cancer development and progression. Recently, miR-22 has been identified as a tumor-suppressing microRNA in many human cancers. However, the specific function of miR-22 in gastric cancer is unclear at this point. In this study, we first measured miR-22 expression level in 30 pairs of gastric cancer and matched normal tissues, two normal and six gastric cancer cell lines by real-time quantitative RT-PCR. We found that the expression of miR-22 in gastric cancer tissues and cell lines was much lower than that in normal control, respectively. Transfection of miR-22 expression plasmid could significantly inhibit the cell migration and invasion in SGC-7901 and NCL-N87 gastric cancer cell lines. Moreover, we also showed that Sp1 was negatively regulated by miR-22 at the posttranscriptional level, via a specific target site within the 3'UTR by luciferase reporter assay. The expression of Sp1 was inversely correlated with miR-22 expression in gastric cancer tissues, and knockdown of Sp1 by siRNA inhibited cell malignant behaviors. Thus, our findings suggest that miR-22 acts as tumor suppressor by targeting the Sp1 gene and inhibiting gastric cancer cell migration and invasion. The findings of this study contribute to current understanding of the functions of miR-22 in gastric cancer.


Assuntos
Movimento Celular/genética , Regulação para Baixo/genética , Regulação Neoplásica da Expressão Gênica , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Fator de Transcrição Sp1/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Linhagem Celular Tumoral , Inibição de Migração Celular/genética , Técnicas de Silenciamento de Genes/métodos , Marcação de Genes/métodos , Humanos , MicroRNAs/biossíntese , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Fator de Transcrição Sp1/biossíntese , Fator de Transcrição Sp1/genética , Neoplasias Gástricas/metabolismo
9.
J Immunol ; 189(12): 5612-21, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23136203

RESUMO

Vaccine adjuvant-induced inflammation augments vaccine immunity in part by recruiting APCs to vaccine draining lymph nodes (LNs). However, the role of one APC subtype, inflammatory monocytes, in regulating vaccine immunity in healthy animals has not been fully examined in detail. Therefore, vaccine-mediated monocyte recruitment and subsequent immune responses were investigated using murine vaccination models and in vitro assays. Recruitment of inflammatory monocytes to vaccine draining LNs was rapid and mediated primarily by local production of MCP-1, as revealed by studies in MCP-1(-/-) mice. Interrupting monocyte recruitment to LNs by either transient monocyte depletion or monocyte migration blockade led to marked amplification of both cellular and humoral immune responses to vaccination. These results were most consistent with the idea that rapidly mobilized inflammatory monocytes were actually suppressing vaccine responses. The suppressive nature of vaccine-elicited monocytes was confirmed using in vitro cocultures of murine monocytes and T cells. Furthermore, it was determined that inflammatory monocytes suppressed T cell responses by sequestering cysteine, as cysteine supplementation in vitro and in vivo appreciably augmented vaccine responses. These findings indicated, therefore, that vaccination-elicited inflammation, although necessary for effective immunity, also generated potent counter-regulatory immune responses that were mediated primarily by inflammatory monocytes. Therefore, interrupting monocyte-mediated vaccine counterregulatory responses may serve as an effective new strategy for broadly amplifying vaccine immunity.


Assuntos
Vacinas Anticâncer/antagonistas & inibidores , Vacinas Anticâncer/imunologia , Tolerância Imunológica/imunologia , Monócitos/imunologia , Monócitos/patologia , Vacinas de DNA/antagonistas & inibidores , Vacinas de DNA/imunologia , Animais , Vacinas Anticâncer/administração & dosagem , Cátions , Linhagem Celular Tumoral , Inibição de Migração Celular/genética , Inibição de Migração Celular/imunologia , Cisteína/administração & dosagem , Tolerância Imunológica/genética , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Lipossomos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Knockout , Monócitos/metabolismo , Receptores CCR2/antagonistas & inibidores , Receptores CCR2/deficiência , Receptores CCR2/genética , Vacinas de DNA/administração & dosagem
10.
J Immunol ; 189(2): 980-7, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-22706085

RESUMO

Various heterotrimeric G(i) proteins are considered to be involved in cell migration and effector function of immune cells. The underlying mechanisms, how they control the activation of myeloid effector cells, are not well understood. To elucidate isoform-redundant and -specific roles for Gα(i) proteins in these processes, we analyzed mice genetically deficient in Gα(i2) or Gα(i3). First, we show an altered distribution of tissue macrophages and blood monocytes in the absence of Gα(i2) but not Gα(i3). Gα(i2)-deficient but not wild-type or Gα(i3)-deficient mice exhibited reduced recruitment of macrophages in experimental models of thioglycollate-induced peritonitis and LPS-triggered lung injury. In contrast, genetic ablation of Gα(i2) had no effect on Gα(i)-dependent peritoneal cytokine production in vitro and the phagocytosis-promoting function of the Gα(i)-coupled C5a anaphylatoxin receptor by liver macrophages in vivo. Interestingly, actin rearrangement and CCL2- and C5a anaphylatoxin receptor-induced chemotaxis but not macrophage CCR2 and C5a anaphylatoxin receptor expression were reduced in the specific absence of Gα(i2). Furthermore, knockdown of Gα(i2) caused decreased cell migration and motility of RAW 264.7 cells, which was rescued by transfection of Gα(i2) but not Gα(i3). These results indicate that Gα(i2), albeit redundant to Gα(i3) in some macrophage activation processes, clearly exhibits a Gα(i) isoform-specific role in the regulation of macrophage migration.


Assuntos
Inibição de Migração Celular/imunologia , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/deficiência , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/deficiência , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/fisiologia , Macrófagos/imunologia , Macrófagos/patologia , Lesão Pulmonar Aguda/imunologia , Lesão Pulmonar Aguda/patologia , Animais , Inibição de Migração Celular/genética , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/genética , Lipopolissacarídeos/toxicidade , Macrófagos/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Monócitos/imunologia , Monócitos/patologia , Peritonite/induzido quimicamente , Peritonite/imunologia , Peritonite/patologia , Tioglicolatos/toxicidade
11.
J Virol ; 86(9): 5165-78, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22357270

RESUMO

Epstein-Barr virus (EBV) establishes a persistent latent infection in B lymphocytes and is associated with the development of numerous human tumors. Epstein-Barr nuclear antigen 3C (EBNA 3C) is essential for B-cell immortalization, has potent cell cycle deregulation capabilities, and functions as a regulator of both viral- and cellular-gene expression. We performed transcription profiling on EBNA 3C-expressing B cells and identified several chemokines and members of integrin receptor-signaling pathways, including CCL3, CCL4, CXCL10, CXCL11, ITGA4, ITGB1, ADAM28, and ADAMDEC1, as cellular target genes that could be repressed by the action of EBNA 3C alone. Chemotaxis assays demonstrated that downregulation of CXCL10 and -11 by EBNA 3C is sufficient to reduce the migration of cells expressing the CXCL10 and -11 receptor CXCR3. Gene repression by EBNA 3C was accompanied by decreased histone H3 lysine 9/14 acetylation and increased histone H3 lysine 27 trimethylation. In an EBV-positive cell line expressing all latent genes, we identified binding sites for EBNA 3C at ITGB1 and ITGA4 and in a distal regulatory region between ADAMDEC1 and ADAM28, providing the first demonstration of EBNA 3C association with cellular-gene control regions. Our data implicate indirect mechanisms in CXCL10 and CXCL11 repression by EBNA 3C. In summary, we have unveiled key cellular pathways repressed by EBNA 3C that are likely to contribute to the ability of EBV-immortalized cells to modulate immune responses, adhesion, and B-lymphocyte migration to facilitate persistence in the host.


Assuntos
Antígenos Virais/metabolismo , Regulação para Baixo/genética , Integrinas/genética , Regiões Promotoras Genéticas , Transdução de Sinais , Proteínas ADAM/genética , Animais , Sítios de Ligação , Adesão Celular/genética , Linhagem Celular , Inibição de Migração Celular/genética , Quimiocinas/genética , Quimiotaxia/genética , Antígenos Nucleares do Vírus Epstein-Barr , Regulação da Expressão Gênica , Humanos , Camundongos , Receptores CXCR3/metabolismo , Elementos Reguladores de Transcrição
12.
J Immunol ; 187(12): 6327-34, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22102721

RESUMO

Fascin, as a substrate of protein kinase C (PKC), is a well-known cytoskeletal regulatory protein required for cell migration, invasion, and adhesion in normal and cancer cells. In an effort to identify the role of fascin in PKC-mediated cellular signaling, its expression was suppressed by stable transfection of specific short hairpin RNAs (shRNAs) in mouse monocytic leukemia RAW264.7 cells. Suppression of fascin expression resulted in impaired cellular migration and invasion through extracellular matrix proteins. Unexpectedly, the specific shRNA transfectants exhibited a marked reduction in LPS-induced expression of TNF-α and IL-6 by blocking the translation of their mRNAs. Transient transfection assay using a luciferase expression construct containing the 3' untranslated region of TNF-α or IL-6 mRNA revealed a significant reduction in both LPS- and PMA- (the direct activator of PKC) induced reporter activity in cells transfected with fascin-specific shRNA, indicating that fascin-mediated translational regulation targeted 3' untranslated region. Furthermore, LPS-induced translational activation of reporter expression was blocked by a pharmacological inhibitor of PKC, and the dominant-negative form of PKCα attenuated LPS-induced translational activation. The same type of regulation was also observed in the human monocytic leukemia cell line THP-1 and in mouse peritoneal macrophages. These data demonstrate the involvement of fascin in the PKC-mediated translational regulation of TNF-α and IL-6 expression during the LPS response.


Assuntos
Proteínas de Transporte/fisiologia , Interleucina-6/genética , Lipopolissacarídeos/farmacologia , Proteínas dos Microfilamentos/fisiologia , Biossíntese de Proteínas/imunologia , Proteína Quinase C/fisiologia , Transdução de Sinais/imunologia , Fator de Necrose Tumoral alfa/genética , Animais , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/biossíntese , Linhagem Celular , Linhagem Celular Tumoral , Inibição de Migração Celular/genética , Inibição de Migração Celular/imunologia , Proteínas da Matriz Extracelular/antagonistas & inibidores , Proteínas da Matriz Extracelular/fisiologia , Humanos , Interleucina-6/antagonistas & inibidores , Interleucina-6/biossíntese , Ativação de Macrófagos/genética , Ativação de Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos ICR , Proteínas dos Microfilamentos/antagonistas & inibidores , Proteínas dos Microfilamentos/biossíntese , Transdução de Sinais/genética , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/biossíntese
13.
Prostate ; 71(11): 1239-50, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21656834

RESUMO

BACKGROUND: The Androgen Receptor (AR) plays a key role in controlling prostate gland homeostasis and contributes to prostate carcinogenesis. The identification of its target genes should provide new candidates that may be implicated in cancer initiation and progression. METHODS: Transcriptomic experiments and chromatin immunoprecipitation were combined to identify direct androgen regulated genes. Real-time quantitative PCR (RT-qPCR) analyses were performed to measure TM4SF1 mRNA levels in prostate cancer and benign prostatic hyperplasia (BPH) specimens. Immunohistochemical methods were used to compare TM4SF1 protein expression profiles in the same cohort. A targeted siRNAs knockdown strategy was used, prior to wound healing assays, to analyze the role of TM4SF1 in cell migration in vitro. RESULTS: We demonstrate for the first time that TM4SF1 is a direct target gene of the AR, a transcription factor of the steroid nuclear receptor family. A functional androgen response element was identified in the promoter region of the gene. In addition, TM4SF1 mRNA expression was higher in cancer samples compared to BPH tissues. The TM4SF1 protein mediates cell motility of prostate cancer cells where it is predominantly localized in the cytoplasm, in contrast to its apical membrane localization in normal prostate epithelial cells. CONCLUSIONS: Our results reveal a novel function for TM4SF1 in AR signaling. The TM4SF1 mRNA expression is higher in prostate cancer tissues as compared to BPH samples. Inhibition of cell migration after targeted knockdown of TM4SF1 protein expression suggests its contribution to prostate cancer cell metastasis.


Assuntos
Antígenos de Superfície/biossíntese , Biomarcadores Tumorais/biossíntese , Inibição de Migração Celular/genética , Regulação Neoplásica da Expressão Gênica , Proteínas de Neoplasias/biossíntese , Neoplasias da Próstata/metabolismo , Receptores Androgênicos/biossíntese , Antígenos de Superfície/genética , Antígenos de Superfície/fisiologia , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/fisiologia , Linhagem Celular Tumoral , Células HeLa , Humanos , Masculino , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiologia , Hiperplasia Prostática/genética , Hiperplasia Prostática/metabolismo , Neoplasias da Próstata/genética , Receptores Androgênicos/genética , Receptores Androgênicos/fisiologia
14.
Mol Pharmacol ; 80(2): 328-36, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21586597

RESUMO

Chemokines of the CC class are key mediators of monocyte recruitment and macrophage differentiation and have a well documented role in many inflammatory diseases. Blockade of chemokine activity is therefore an attractive target for anti-inflammatory therapy. 35K (vCCI) is a high-affinity chemokine binding protein expressed by poxviruses, which binds all human and murine CC chemokines, preventing their interaction with chemokine receptors. We developed an Fc-fusion protein of 35K with a modified human IgG1 Fc domain and expressed this construct in human embryonic kidney 293T cells. Purified 35K-Fc is capable of inhibiting CC chemokine-induced calcium flux, chemotaxis, and ß-arrestin recruitment in primary macrophages and transfected cells. To elucidate the residues involved in chemokine neutralization, we performed site-directed mutagenesis of six key amino acids in 35K and expressed the mutant Fc-fusion proteins in vitro. We screened the mutants for their ability to block chemokine-induced ß-arrestin recruitment in transfected cells and to inhibit primary macrophage signaling in an electric cell substrate impedance sensing assay. Using a sterile model of acute inflammation, zymosan-induced peritonitis, we confirmed that wild-type 35K-Fc can reduce monocyte recruitment, whereas one mutant (R89A) showed a more pronounced blockade of monocyte influx and another mutant (E143K) showed total loss of function. We believe that 35K-Fc will be a useful tool for exploring the role of CC chemokines in chronic inflammatory pathologies, and we have identified a higher potency form of the molecule that may have potential therapeutic applications in chronic inflammatory disease.


Assuntos
Quimiocinas CC/antagonistas & inibidores , Quimiocinas CC/genética , Quimiocinas/genética , Fragmentos Fc das Imunoglobulinas/genética , Mutagênese Sítio-Dirigida/métodos , Mutação/fisiologia , Animais , Arrestinas/antagonistas & inibidores , Arrestinas/metabolismo , Cálcio/antagonistas & inibidores , Cálcio/metabolismo , Inibição de Migração Celular/genética , Quimiocinas/metabolismo , Quimiocinas CC/metabolismo , Quimiocinas CXC , Humanos , Fragmentos Fc das Imunoglobulinas/metabolismo , Imunoglobulina G/química , Imunoglobulina G/genética , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica/genética , Proteínas Recombinantes de Fusão/síntese química , Proteínas Recombinantes de Fusão/genética , Transfecção , beta-Arrestinas
15.
Mol Cancer Res ; 9(5): 545-52, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21478267

RESUMO

Forkhead transcription factor, Foxd3, plays a critical role during development by controlling the lineage specification of neural crest cells. Notably, Foxd3 is highly expressed during the wave of neural crest cell migration that forms peripheral neurons and glial cells but is downregulated prior to migration of cells that give rise to the melanocytic lineage. Melanoma is the deadliest form of skin cancer and is derived from melanocytes. Recently, we showed that FOXD3 expression is elevated following the targeted inhibition of the B-RAF-MEK (MAP/ERK kinase)-ERK (extracellular signal-regulated kinase)1/2 pathway in mutant B-RAF melanoma cells. Because melanoma cells are highly migratory and invasive in a B-RAF-dependent manner, we explored the role of FOXD3 in these processes. In this study, we show that ectopic FOXD3 expression inhibits the migration, invasion, and spheroid outgrowth of mutant B-RAF melanoma cells. Upregulation of FOXD3 expression following inhibition of B-RAF and MEK correlates with the downregulation of Rnd3, a Rho GTPase and inhibitor of RhoA-ROCK signaling. Indeed, expression of FOXD3 alone was sufficient to downregulate Rnd3 expression at the mRNA and protein levels. Mechanistically, FOXD3 was found to be recruited to the Rnd3 promoter. Inhibition of ROCK partially restored migration in FOXD3-expressing cells. These data show that FOXD3 expression downregulates migration and invasion in melanoma cells and Rnd3, a target known to be involved in these properties.


Assuntos
Inibição de Migração Celular/genética , Fatores de Transcrição Forkhead/metabolismo , Melanócitos/metabolismo , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , Proteínas Proto-Oncogênicas B-raf/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Linhagem Celular Tumoral , Regulação para Baixo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fatores de Transcrição Forkhead/genética , Humanos , Melanoma/genética , Melanoma/metabolismo , Melanoma/patologia , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Mutação , Proteínas Proto-Oncogênicas B-raf/genética , Transdução de Sinais , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia
16.
J Neurosci Res ; 89(7): 1079-90, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21503949

RESUMO

Malignant gliomas have a tendency to invade diffusely into surrounding healthy brain tissues, thereby precluding their successful surgical removal. Intersectin1 (ITSN1) as a molecular linker in the central nervous system is well known as an important regulator of endocytosis and exocytosis. ITSN1 has two isoforms: ITSN1-l and ITSN1-s. In this study, we show that siRNA-mediated down regulation of ITSN1-s inhibited migration and invasion of glioma cells. In addition, we demonstrate the possible mechanisms by which ITSN1-s functions in migration and invasion. Several key proteins, including cofilin, LIMK, PAK, FAK, integrin ß1, and MMP-9, which are critical for cells migration and invasion, were probably involved in ITSN1-s signaling pathways. These results suggest that ITSN1-s contributes to glioma cells migration and invasion by regulating the formation of cytoskeleton, influencing adhesion and increasing expression of MMP-9. Our results indicate that ITSN1-s is a critical factor in gliomas invasion and identify that ITSN1-s is a new potentially antiinvasion target for therapeutic intervention in gliomas.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Movimento Celular , Glioblastoma/metabolismo , Glioblastoma/patologia , Invasividade Neoplásica , Proteínas Adaptadoras de Transporte Vesicular/genética , Animais , Neoplasias Encefálicas/genética , Linhagem Celular Tumoral , Inibição de Migração Celular/genética , Movimento Celular/genética , Movimento Celular/fisiologia , Modelos Animais de Doenças , Glioblastoma/genética , Humanos , Masculino , Camundongos , Camundongos Nus , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiologia , Interferência de RNA/fisiologia
17.
J Immunol ; 186(7): 4088-97, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21339367

RESUMO

B cell chemotaxis occurs in response to specific chemokine gradients and is critical for homeostasis and immune response. The molecular regulation of B cell membrane-actin interactions during migration is poorly understood. In this study, we report a role for ezrin, a member of the membrane-cytoskeleton cross-linking ezrin-radixin-moesin proteins, in the regulation of the earliest steps of B cell polarization and chemotaxis. We visualized chemokine-induced changes in murine B cell morphology using scanning electron microscopy and spatiotemporal dynamics of ezrin in B cells using epifluorescence and total internal reflection microscopy. Upon chemokine stimulation, ezrin is transiently dephosphorylated to assume an inactive conformation and localizes to the lamellipodia. B cells expressing a phosphomimetic conformationally active mutant of ezrin or those in which ezrin dephosphorylation was pharmacologically inhibited displayed impaired microvillar dynamics, morphological polarization, and chemotaxis. Our data suggest a 2-fold involvement of ezrin in B cell migration, whereby it first undergoes chemokine-induced dephosphorylation to facilitate membrane flexibility, followed by relocalization to the actin-rich lamellipodia for dynamic forward protrusion of the cells.


Assuntos
Subpopulações de Linfócitos B/citologia , Subpopulações de Linfócitos B/imunologia , Inibição de Migração Celular/imunologia , Quimiotaxia de Leucócito/imunologia , Proteínas do Citoesqueleto/química , Proteínas do Citoesqueleto/genética , Citoesqueleto/química , Citoesqueleto/imunologia , Actinas/metabolismo , Substituição de Aminoácidos/genética , Substituição de Aminoácidos/imunologia , Animais , Subpopulações de Linfócitos B/química , Linhagem Celular Tumoral , Inibição de Migração Celular/genética , Quimiotaxia de Leucócito/genética , Proteínas do Citoesqueleto/biossíntese , Citoesqueleto/genética , Camundongos , Camundongos Endogâmicos C57BL , Proteínas dos Microfilamentos/química , Proteínas dos Microfilamentos/genética , Mutagênese Sítio-Dirigida , Fosforilação/genética , Fosforilação/imunologia , Conformação Proteica , Pseudópodes/genética , Pseudópodes/imunologia , Pseudópodes/metabolismo , Fase de Repouso do Ciclo Celular/genética , Fase de Repouso do Ciclo Celular/imunologia , Treonina/química , Treonina/genética
18.
J Immunol ; 186(5): 3197-205, 2011 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21282514

RESUMO

CXCL5, a member of the CXC family of chemokines, contributes to neutrophil recruitment during lung inflammation, but its regulation is poorly understood. Because the T cell-derived cytokine IL-17A enhances host defense by triggering production of chemokines, particularly in combination with TNF-α, we hypothesized that IL-17A would enhance TNF-α-induced expression of CXCL5. Intratracheal coadministration of IL-17A and TNF-α in mice induced production of CXCL1, CXCL2, and CXCL5, which was associated with increased neutrophil influx in the lung at 8 and 24 h. The synergistic effects of TNF-α and IL17A were greatly attenuated in Cxcl5(-/-) mice at 24 h, but not 8 h, after exposure, a time when CXCL5 expression was at its peak in wild-type mice. Bone marrow chimeras produced using Cxcl5(-/-) donors and recipients demonstrated that lung-resident cells were the source of CXCL5. Using differentiated alveolar epithelial type II (ATII) cells derived from human fetal lung, we found that IL-17A enhanced TNF-α-induced CXCL5 transcription and stabilized TNF-α-induced CXCL5 transcripts. Whereas expression of CXCL5 required activation of NF-κB, IL-17A did not increase TNF-α-induced NF-κB activation. Apical costimulation of IL-17A and TNF-α provoked apical secretion of CXCL5 by human ATII cells in a transwell system, whereas basolateral costimulation led to both apical and basolateral secretion of CXCL5. The observation that human ATII cells secrete CXCL5 in a polarized fashion may represent a mechanism to recruit neutrophils in host defense in a fashion that discriminates the site of initial injury.


Assuntos
Quimiocina CXCL5/biossíntese , Interleucina-17/fisiologia , Alvéolos Pulmonares/imunologia , Alvéolos Pulmonares/metabolismo , Fator de Necrose Tumoral alfa/fisiologia , Lesão Pulmonar Aguda/genética , Lesão Pulmonar Aguda/imunologia , Lesão Pulmonar Aguda/patologia , Animais , Inibição de Migração Celular/genética , Inibição de Migração Celular/imunologia , Células Cultivadas , Quimiocina CXCL1/biossíntese , Quimiocina CXCL2/biossíntese , Quimiocina CXCL5/deficiência , Quimiocina CXCL5/metabolismo , Quimiotaxia de Leucócito/genética , Quimiotaxia de Leucócito/imunologia , Modelos Animais de Doenças , Quimioterapia Combinada , Humanos , Mediadores da Inflamação/metabolismo , Mediadores da Inflamação/fisiologia , Interleucina-17/administração & dosagem , Interleucina-17/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/imunologia , Neutrófilos/patologia , Pneumonia Bacteriana/imunologia , Pneumonia Bacteriana/metabolismo , Pneumonia Bacteriana/patologia , Alvéolos Pulmonares/patologia , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/biossíntese , Índice de Gravidade de Doença , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Fator de Necrose Tumoral alfa/administração & dosagem , Fator de Necrose Tumoral alfa/biossíntese
19.
Clin Cancer Res ; 17(9): 2702-11, 2011 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-21349996

RESUMO

PURPOSE: To elucidate the role of Semaphorin-3F (SEMA3F), originally described as an axon guiding chemorepulsant implicated in nerve development, in the progression of colorectal carcinoma. EXPERIMENTAL DESIGN: SEMA3F and its receptor NRP2 were examined in 72 cases of human colorectal carcinoma specimens and cell lines LoVo, SW480, and SW620 with immunohistochemistry and Western blotting. SEMA3F mRNA expression in the frozen tissue specimens and cell lines was examined with quantitative reverse transcriptase-PCR. Confocal laser scanning microscopy was used for detection of cellular localization of the proteins by immunofluorescent staining. MTT assay, flow cytometry, cell adhesion and migration, and xenografts were used to evaluate biological significance of SEMA3F. RESULTS: SEMA3F was significantly reduced in colorectal carcinoma tissues and cell lines. Overexpression of SEMA3F resulted in reduced proliferation, adhesion to fibronectin, and migratory capability as well as reduced S-phase population and integrin αvß3 expression of SW480 colon cancer cells. In addition, SEMA3F-overexpressing cells exhibited diminished tumorigenesis when transplanted orthotopically in nude mice and reduced liver metastases. Moreover, transfection of siRNA targeting SEMA3F in colon cancer cells increased their tumorigenicity in vivo. CONCLUSIONS: Endogenous SEMA3F acts as a suppressor of the growth and metastasis of human colorectal cancer cells.


Assuntos
Carcinoma/patologia , Proliferação de Células , Neoplasias Colorretais/patologia , Proteínas de Membrana/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Axônios/metabolismo , Axônios/fisiologia , Carcinoma/genética , Carcinoma/metabolismo , Carcinoma/prevenção & controle , Linhagem Celular Tumoral , Inibição de Migração Celular/genética , Proliferação de Células/efeitos dos fármacos , Fatores Quimiotáticos/antagonistas & inibidores , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/prevenção & controle , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Terapia Genética , Humanos , Masculino , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Metástase Neoplásica , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/genética , RNA Interferente Pequeno/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Cell Cycle ; 10(1): 73-81, 2011 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21200149

RESUMO

Cyclin D1 overexpression is a common feature of many human malignancies. Genomic deletion analysis has demonstrated a key role for cyclin D1 in cellular proliferation, angiogenesis, and cellular migration. To investigate the mechanisms contributing to cyclin D1 functions, we purified cyclin D1a-associated complexes by affinity chromatography and identified the PACSIN 2 (protein kinase C and casein kinase substrate in neurons 2) protein by mass spectrometry. The PACSIN 2, but not the related PACSIN 1 and 3, directly bound wild-type cyclin D1 (cyclin D1a) at the carboxyl terminus, and failed to bind cyclin D1b, the alternative splicing variant of cyclin D1. PACSIN 2 knockdown induced cellular migration and reduced cell spreading in LNCaP cells expressing cyclin D1a. In cyclin D1(-/-) mouse embryonic fibroblasts (MEFs), cyclin D1a, but not cyclin D1b, reduced the cell spreading to a polarized morphology. siPACSIN 2 had no effect on cellular migration of cyclin D1(-/-) MEFs. Cyclin D1a restored the migratory ability of cyclin D1(-/-) MEFs, which was further enhanced by knocking down PACSIN 2 with siRNA. The cyclin D1-associated protein, PACSIN 2, regulates cell spreading and migration, which are dependent on cyclin D1 expression.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Inibição de Migração Celular/genética , Ciclina D1/metabolismo , Sítios de Splice de RNA/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Ciclina D1/genética , Células HEK293 , Humanos , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Células NIH 3T3 , Ligação Proteica/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA