Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 307
Filtrar
1.
Oncogene ; 36(49): 6805-6814, 2017 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-28846111

RESUMO

Overexpression of heme oxygenase-1 (HO-1), an endoplasmic reticulum-anchored enzyme, is observed in many cancers. HO-1 nuclear translocation has been shown to correlate with progression of several cancers. We recently reported that HO-1 is susceptible to intramembrane proteolysis and translocates to the nucleus to promote cancer growth and invasiveness without depending on its enzymatic activity. In the present study, we show that the HO-1 lacking C-terminal transmembrane segment (t-HO-1) was susceptible to acetylation by p300 and CREB-binding protein (CBP) histone acetyltransferase in the nucleus. Mass spectrometry analysis of HO-1 isolated from human embryonic kidney cells 293T (HEK293T) cells overexpressing CBP and t-HO-1 revealed two acetylation sites located at K243 and K256. Mutation of both lysine residues to arginine (R) abolished t-HO-1-enhanced tumor cell growth, migration and invasion. However, mutation of the lysine residues to glutamine (Q), a mimic of acetylated lysine, had no significant effect on t-HO-1-mediated tumorigenicity. Mechanistic studies demonstrated that transcriptional factor JunD interacted with wild-type (WT) t-HO-1 and mutant carrying K243/256Q but not K243/256 R mutation. Moreover, JunD-induced AP-1 transcriptional activity was significantly enhanced by coexpression with WT and acetylation-mimic but not acetylation-defective t-HO-1. Consistent with the in vitro observations, the implication of K243/256 acetylation in t-HO-1-enhanced tumorigenicity was also demonstrated in xenograft models. Immunohistochemistry performed with a specific antibody against acetyl-HO-1 showed the positive acetyl-HO-1 nuclear staining in human lung cancer tissues but not in the corresponding non-tumor tissues, supporting its clinical significance. Collectively, our findings highlight the importance of nuclear HO-1 post-translational modification in the induction of cancer progression.


Assuntos
Núcleo Celular/enzimologia , Proliferação de Células , Heme Oxigenase-1/metabolismo , Neoplasias/enzimologia , Acetilação , Animais , Linhagem Celular Tumoral , Feminino , Células HEK293 , Células HeLa , Heme Oxigenase-1/genética , Humanos , Lisina/genética , Lisina/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Nus , Mutação , Invasividade Neoplásica , Neoplasias/genética , Neoplasias/patologia , Transplante Heterólogo , Carga Tumoral
2.
Oncogenesis ; 6(5): e338, 2017 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-28530703

RESUMO

TRPV4 belongs to the 'Transient Receptor Potential' (TRP) superfamily. It has been identified to profoundly affect a variety of physiological processes, including nociception, heat sensation and inflammation. Unlike other TRP superfamily channels, its role in cancers are unknown until recently when we reported TRPV4 to be required for cancer cell softness that may promote breast cancer cell extravasation and metastasis. Here, we elucidated the molecular mechanisms mediated by TRPV4 in the metastatic breast cancer cells. TRPV4-mediated signaling was demonstrated to involve Ca2+-dependent activation of AKT and downregulation of E-cadherin expression, which was abolished upon TRPV4 silencing. Functionally, TRPV4-enhanced breast caner cell transendothelial migration requires AKT activity while a combination of transcriptional and post-translational regulation contributed to the TRPV4-mediated E-cadherin downregulation. Finally, mass spectrometry analysis revealed that TRPV4 is required for the expression of a network of secreted proteins involved in extracellular matrix remodeling. In conclusion, TRPV4 may regulate breast cancer metastasis by regulating cell softness through the Ca2+-dependent AKT-E-cadherin signaling axis and regulation of the expression of extracellular proteins.

3.
Oncogene ; 36(17): 2457-2471, 2017 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-27819672

RESUMO

Tumor microenvironment (TME) plays an active role in promoting tumor progression. To further understand the communication between TME and tumor cells, this study aimed at investigating the involvement of CD44, a type I cell surface receptor, in the crosstalk between tumor cells and TME. We have previously shown that chondroitin sulfate proteoglycan serglycin (SRGN), a CD44-interacting factor, was preferentially secreted by cancer-associated fibroblasts (CAFs) for promoting tumor growth in breast cancer patients. In this study, we show that SRGN is overexpressed in primary non-small cell lung cancers (NSCLCs), by both carcinoma and stromal cells. Using gain-of-function and loss-of-function approaches, we show that SRGN promotes NSCLC cell migration and invasion as well as colonization in the lung and liver in a CD44-dependent manner. SRGN induces lung cancer cell stemness, as demonstrated by its ability to enhance NSCLC cell sphere formation via Nanog induction, accompanied with increased chemoresistance and anoikis-resistance. SRGN promotes epithelial-mesenchymal transition by enhancing vimentin expression via CD44/NF-κB/claudin-1 (CLDN1) axis. In support, CLDN1 and SRGN expression are tightly linked together in primary NSCLC. Most importantly, increased expression of SRGN and/or CLDN1 predicts poor prognosis in primary lung adenocarcinomas. In summary, we demonstrate that SRGN secreted by tumor cells and stromal components in the TME promotes malignant phenotypes through interacting with tumor cell receptor CD44, suggesting that a combined therapy targeting both CD44 and its ligands in the TME may be an attractive approach for cancer therapy.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/patologia , Receptores de Hialuronatos/metabolismo , Neoplasias Pulmonares/patologia , Proteoglicanas/metabolismo , Microambiente Tumoral , Proteínas de Transporte Vesicular/metabolismo , Linhagem Celular Tumoral , Claudina-1/metabolismo , Transição Epitelial-Mesenquimal , Regulação Neoplásica da Expressão Gênica , Humanos , NF-kappa B/metabolismo , Fenótipo , Análise de Sobrevida
4.
Oncogenesis ; 5(7): e239, 2016 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-27376780

RESUMO

Esophageal cancer is a lethal malignancy worldwide. Previously, low expression of metastasis suppressor Nm23H1 and tight junction (TJ) protein claudin-1 (CLDN1) have been known to correlate with poor prognosis in esophageal squamous cell carcinoma (ESCC). However, the molecular interaction between them has not been clarified. In the present study, we first examined the expression of Nm23H1 and CLDN1 in 74 surgical ESCC samples by immunohistochemistry (IHC) to verify their clinicopathologic significance. The biologic effects of Nm23H1 gene silencing or overexpression in ESCC cell lines were then studied by migration and invasion studies, and its regulation on CLDN1 expression was also investigated by western blot analysis. Moreover, the expression of Nm23H1 and CLDN1 at the same invasion front of ESCC tumors was verified by immunofluorescence. The results showed a significantly positive correlation between the expression of Nm23H1 and CLDN1 (γ=0.296, P=0.011) in surgical specimens, especially for the 34 tumors with lymph-node metastasis (γ=0.455, P=0.007). In ESCC cell lines, silencing of Nm23H1 expression markedly enhanced cell invasiveness, accompanied by increased Akt phosphorylation and decreased CLDN1 expression. Conversely, Nm23H1-expressed transfectants exhibited reduced invasiveness, decreased Akt phosphorylation and correspondingly increased CLDN1 expression. Regain of CLDN1 expression in ESCC cells significantly suppressed invasiveness, but did not influence the Akt phosphorylation. Moreover, treating Nm23H1-depleted cells with the AKT inhibitor MK2206 recovered CLDN1 expression, and diminished the invasiveness of ESCC cells. Finally, decreased expressions of both CLDN1 and E-cadherin were observed at the invasive front of the Nm23H1-negative tumors. Overall, our current study documented that reduced Nm23H1 expression activates the AKT signaling pathway, results in diminished CLDN1 expression and potentiates invasiveness of ESCC cells. Enhancement of Nm23H1 expression, inhibition of the AKT signaling pathway, or combined, might be a potential treatment strategy in selective ESCC patients.

6.
Oncogene ; 34(22): 2910-21, 2015 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-25065598

RESUMO

Hepatocellular carcinoma (HCC) has a poor prognosis owing to aggressive phenotype. Gα12 gep oncogene product couples to G-protein-coupled receptors, whose ligand levels are frequently increased in tumor microenvironments. Here, we report Gα12 overexpression in human HCC and the resultant induction of zinc-finger E-box-binding homeobox 1 (ZEB1) as mediated by microRNA deregulation. Gα12 expression was higher in HCC than surrounding non-tumorous tissue. Transfection of Huh7 cell with an activated mutant of Gα12 (Gα12QL) deregulated microRNA (miRNA or miR)-200b/a/429, -194-2/192 and -194-1/215 clusters in the miRNome. cDNA microarray analyses disclosed the targets affected by Gα12 gene knockout. An integrative network of miRNAs and mRNA changes enabled us to predict ZEB1 as a key molecule governed by Gα12. Decreases of miR-200a/b, -192 and -215 by Gα12 caused ZEB1 induction. The ability of Gα12 to decrease p53 levels, as a result of activating protein-1 (AP-1)/c-Jun-mediated mouse double minute 2 homolog induction, contributed to transcriptional deregulation of the miRNAs. Gα12QL induced ZEB1 and other epithelial-mesenchymal transition markers with fibroblastoid phenotype change. Consistently, transfection with miR-200b, -192 or -215 mimic prevented the ability of Gα12QL to increase tumor cell migration/invasion. In xenograft studies, sustained knockdown of Gα12 decreased the overall growth rate and average volume of tumors derived from SK-Hep1 cell (mesenchymal-typed). In HCC patients, miR-192, -215 and/or -200a were deregulated with microvascular invasion or growth advantage. In the HCC samples with higher Gα12 level, a correlation existed in the comparison of relative changes of Gα12 and ZEB1. In conclusion, Gα12 overexpressed in HCC causes ZEB1 induction by deregulating p53-responsive miRNAs, which may facilitate epithelial-mesenchymal transition and growth of liver tumor. These findings highlight the significance of Gα12 upregulation in liver tumor progression, implicating Gα12 as an attractive therapeutic target.


Assuntos
Carcinoma Hepatocelular/genética , Transição Epitelial-Mesenquimal/genética , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/fisiologia , Neoplasias Hepáticas/genética , MicroRNAs/genética , Proteína Supressora de Tumor p53/fisiologia , Animais , Carcinoma Hepatocelular/patologia , Embrião de Galinha , Progressão da Doença , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/genética , Regulação Neoplásica da Expressão Gênica , Células Hep G2 , Humanos , Neoplasias Hepáticas/patologia , Camundongos , Camundongos Nus , Oncogenes/genética , Oncogenes/fisiologia , Células Tumorais Cultivadas , Regulação para Cima/genética
7.
Oncogene ; 34(10): 1220-30, 2015 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-24662830

RESUMO

Hec1 (highly expressed in cancer 1) or Nek2 (NIMA-related kinase 2) is often overexpressed in cancers with poor prognosis. Both are critical mitotic regulators, and phosphorylation of Hec1 S165 by Nek2 is required for proper chromosome segregation. Therefore, inactivation of Hec1 and Nek2 by targeting their interaction with small molecules represents an ideal strategy for tackling these types of cancers. Here we showed that new derivatives of INH (inhibitor for Nek2 and Hec1 binding) bind to Hec1 at amino acids 394-408 on W395, L399 and K400 residues, effectively blocking Hec1 phosphorylation on S165 by Nek2, and killing cancer cells at the nanomolar range. Mechanistically, the D-box (destruction-box) region of Nek2 specifically binds to Hec1 at amino acids 408-422, immediately adjacent to the INH binding motif. Subsequent binding of Nek2 to INH-bound Hec1 triggered proteasome-mediated Nek2 degradation, whereas the Hec1 binding defective Nek2 mutant, Nek2 R361L, resisted INH-induced Nek2 degradation. This finding unveils a novel drug-action mechanism where the binding of INHs to Hec1 forms a virtual death-trap to trigger Nek2 degradation and eventually cell death. Furthermore, analysis of the gene expression profiles of breast cancer patient samples revealed that co-elevated expressions of Hec1 and Nek2 correlated with the shortest survival. Treatment of mice with this kind of tumor with INHs significantly suppressed tumor growth without obvious toxicity. Taken together, the new INH derivatives are suitable for translation into clinical application.


Assuntos
Neoplasias/metabolismo , Neoplasias/patologia , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Benzamidas/química , Benzamidas/farmacologia , Sítios de Ligação , Linhagem Celular Tumoral , Proteínas do Citoesqueleto , Modelos Animais de Doenças , Progressão da Doença , Expressão Gênica , Xenoenxertos , Humanos , Indóis , Concentração Inibidora 50 , Mitose/efeitos dos fármacos , Modelos Moleculares , Conformação Molecular , Quinases Relacionadas a NIMA , Neoplasias/genética , Neoplasias/mortalidade , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/química , Proteínas Nucleares/genética , Fosforilação/efeitos dos fármacos , Prognóstico , Ligação Proteica/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/genética , Proteólise/efeitos dos fármacos , Tiazóis/química , Tiazóis/farmacologia , Valeratos
8.
Oncogene ; 34(18): 2360-70, 2015 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-24931165

RESUMO

Heme oxygenase-1 (HO-1) is a heme-degrading enzyme anchored in the endoplasmic reticulum by a carboxyl-terminal transmembrane segment (TMS). HO-1 is highly expressed in various cancers and its nuclear localization is associated with the progression of some cancers. Nevertheless, the mechanism underlying HO-1 nuclear translocation and its pathological significance remain elusive. Here we show that the signal peptide peptidase (SPP) catalyzes the intramembrane cleavage of HO-1. Coexpression of HO-1 with wild-type SPP, but not a dominant-negative SPP, promoted the nuclear localization of HO-1 in cells. Mass spectrometry analysis of cytosolic HO-1 isolated from HeLa cells overexpressing HO-1 and SPP revealed two adjacent intramembrane cleavage sites located after S275 and F276 within the TMS. Mutations of S275F276 to A275L276 significantly hindered SPP-mediated HO-1 cleavage and nuclear localization. Nuclear HO-1 was detected in A549 and DU145 cancer cell lines expressing high levels of endogenous HO-1 and SPP. SPP knockdown or inhibition significantly reduced nuclear HO-1 localization in A549 and DU145 cells. The positive nuclear HO-1 stain was also evident in lung cancer tissues expressing high levels of HO-1 and SPP. Overexpression of a truncated HO-1 (t-HO-1) lacking the TMS in HeLa and H1299 cells promoted cell proliferation and migration/invasion. The effect of t-HO-1 was not affected by a mutation in the catalytic site. However, blockade of t-HO-1 nuclear localization abolished t-HO-1-mediated effect. The tumorigenic effect of t-HO-1 was also demonstrated in the mouse model. These findings disclose that SPP-mediated intramembrane cleavage of HO-1 promotes HO-1 nuclear localization and cancer progression independent of HO-1 enzymatic activity.


Assuntos
Ácido Aspártico Endopeptidases/metabolismo , Núcleo Celular/metabolismo , Heme Oxigenase-1/metabolismo , Neoplasias/metabolismo , Animais , Ácido Aspártico Endopeptidases/genética , Linhagem Celular Tumoral , Proliferação de Células , Células HeLa , Heme Oxigenase-1/genética , Humanos , Espectrometria de Massas , Camundongos , Invasividade Neoplásica , Neoplasias/patologia
9.
Allergy ; 69(2): 186-98, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24191851

RESUMO

BACKGROUND: Recent evidence indicates that TNF-α is a key mediator of the development of dsRNA-enhanced Th2 cell response to inhaled allergens. Natural killer T (NKT) cells may be a candidate source of Th2-polarizing cytokines. OBJECTIVE: The objective of this study was to evaluate the role of lung NKT cells on the development of TNF-α-mediated Th2 cell response. METHODS: A virus-associated asthma mouse model was generated by the administration of ovalbumin (OVA, 75 µg) and poly[I:C] (0.1 µg). Role of NKT and type I NKT cells was evaluated using CD1d- and Jα18-deficient mice. TNF-α receptors (TNFRs) were antagonized by using TNFR blocking peptides. RESULTS: The number of infiltrated NKT cells was increased in a virus-associated asthma mouse model. Increase in Th2 and Th17 cytokine levels in wild-type mice were abolished in both CD1d- and Jα18-deficient mice. In vitro co-culture experiments with alveolar macrophages and NKT cells showed that TNF-α produced by macrophages in the presence of poly[I:C] acts on NKT cells, inducing production of Th2-polarizing cytokines. Moreover, the induction of Th2-polarizing cytokines by poly[I:C] or recombinant TNF-α was impaired in both CD1d- and Jα18-deficient mice and that the above effect was reversed by a TNF-α receptor-2 (TNFR2) blocking peptide, but not by a TNFR1 blocker. CONCLUSIONS: These findings suggest that NKT cells play a key role in the development of Th2 cell response to inhaled allergens and that TNF-α produced by alveolar macrophages induces Th2 cell response, via TNFR2 on NKT cells.


Assuntos
Asma/imunologia , Células T Matadoras Naturais/imunologia , Receptores Tipo II do Fator de Necrose Tumoral/imunologia , Células Th2/imunologia , Fator de Necrose Tumoral alfa/imunologia , Alérgenos/imunologia , Animais , Hiper-Reatividade Brônquica/imunologia , Técnicas de Cocultura , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Hipersensibilidade/imunologia , Ativação Linfocitária/imunologia , Macrófagos Alveolares/imunologia , Camundongos , Pneumonia/imunologia , RNA de Cadeia Dupla/imunologia
10.
Oncogene ; 33(23): 2968-77, 2014 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-23851503

RESUMO

Gain of function of membrane receptor was a good strategy exploited by cancer cells to benefit own growth and survival. Overexpression of HER2 has been found to serve as a target for developing trastuzumab to treat 20-25% of breast cancer. However, little or none of the other membrane receptor was found to be useful as a potential target for breast cancer treatment since then. Here, we showed that amplified signaling of interleukin-17 receptor B (IL-17RB) and its ligand IL-17B promoted tumorigenicity in breast cancer cells and impeded acinus formation in immortalized normal mammary epithelial cells. External signal transmitted through IL-17RB activated nuclear factor-κB to upregulate antiapoptotic factor Bcl-2 and induced etoposide resistance. Elevated expression of IL-17RB had a stronger correlation with poor prognosis than HER2 in breast cancer patients. Interestingly, breast cancer patients with high expression of IL-17RB and HER2 had the shortest survival rate. Depletion of IL-17RB in trastuzumab-resistant breast cancer cells significantly reduced their tumorigenic activity, suggesting that IL-17RB and HER2 have an independent role in breast carcinogenesis. Furthermore, treatment with antibodies specifically against IL-17RB or IL-17B effectively attenuated tumorigenicity of breast cancer cells. These results suggest that the amplified IL-17RB/IL-17B signaling pathways may serve as a therapeutic target for developing treatment to manage IL-17RB-associated breast cancer.


Assuntos
Neoplasias da Mama/patologia , Carcinogênese , NF-kappa B/metabolismo , Receptores de Interleucina-17/antagonistas & inibidores , Receptores de Interleucina-17/metabolismo , Animais , Apoptose/efeitos dos fármacos , Comunicação Autócrina , Neoplasias da Mama/metabolismo , Carcinogênese/efeitos dos fármacos , Linhagem Celular Tumoral , Etoposídeo/farmacologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Interleucina-17/imunologia , Interleucina-17/metabolismo , Células MCF-7 , Neoplasias Mamárias Experimentais , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , NF-kappa B/antagonistas & inibidores , Comunicação Parácrina , Receptor ErbB-2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Clin Exp Allergy ; 43(4): 443-54, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23517040

RESUMO

BACKGROUND: Many bacterial components in indoor dust can evoke inflammatory pulmonary diseases. Bacteria secrete nanometre-sized vesicles into the extracellular milieu, but it remains to be determined whether bacteria-derived extracellular vesicles in indoor dust are pathophysiologically related to inflammatory pulmonary diseases. OBJECTIVE: To evaluate whether extracellular vesicles (EV) in indoor air are related to the pathogenesis of pulmonary inflammation and/or asthma. METHODS: Indoor dust was collected from a bed mattress in an apartment. EV were prepared by sequential ultrafiltration and ultracentrifugation. Innate and adaptive immune responses were evaluated after airway exposure of EV. RESULTS: Repeated intranasal application of indoor-dust-induced neutrophilic pulmonary inflammation accompanied by lung infiltration of both Th1 and Th17 cells. EV 50-200 nm in diameter were present (102.5 µg protein concentration/g dust) in indoor dust. These vesicles were internalized by airway epithelial cells and alveolar macrophages, and this process was blocked by treatment of polymyxin B (an antagonist of lipopolysaccharide, an outer-membrane component of Gram-negative bacteria). Intranasal application of 0.1 or 1 µg of these vesicles for 4 weeks elicited neutrophilic pulmonary inflammation. This phenotype was accompanied by lung infiltration of both Th1 and Th17 cells, which were reversed by treatment of polymyxin B. Serum dust EV-reactive IgG1 levels were significantly higher in atopic children with asthma than in atopic healthy children and those with rhinitis or dermatitis. CONCLUSION & CLINICAL RELEVANCE: Indoor dust EV, especially derived from Gram-negative bacteria, is a possible causative agent of neutrophilic airway diseases.


Assuntos
Micropartículas Derivadas de Células/metabolismo , Poeira/imunologia , Bactérias Gram-Negativas/metabolismo , Neutrófilos/imunologia , Pneumonia/etiologia , Células Th1/imunologia , Células Th17/imunologia , Imunidade Adaptativa , Animais , Linhagem Celular , Criança , Bactérias Gram-Negativas/imunologia , Humanos , Imunidade Inata , Imunoglobulina E/sangue , Imunoglobulina G/sangue , Mediadores da Inflamação/metabolismo , Receptores de Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/imunologia , Pulmão/imunologia , Pulmão/patologia , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos
12.
Oncogene ; 32(9): 1193-201, 2013 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-22562243

RESUMO

Mitochondrial dysfunction has been a hallmark of cancer. However, whether it has a causative role awaits to be elucidated. Here, using an animal model derived from inactivation of SUV3, a mitochondrial helicase, we demonstrated that mSuv3+/- mice harbored increased mitochondrial DNA (mtDNA) mutations and decreased mtDNA copy numbers, leading to tumor development in various sites and shortened lifespan. These phenotypes were transmitted maternally, indicating the etiological role of the mitochondria. Importantly, reduced SUV3 expression was observed in human breast tumor specimens compared with corresponding normal tissues in two independent cohorts. These results demonstrated for the first time that maintaining mtDNA integrity by SUV3 helicase is critical for cancer suppression.


Assuntos
Neoplasias da Mama/genética , Transformação Celular Neoplásica/genética , RNA Helicases DEAD-box/genética , Genoma Mitocondrial , Instabilidade Genômica , Haploinsuficiência , Animais , Linhagem Celular Tumoral , Variações do Número de Cópias de DNA/genética , Perda do Embrião/genética , Feminino , Heterozigoto , Humanos , Estilo de Vida , Longevidade/genética , Camundongos
13.
Oncogene ; 32(6): 777-87, 2013 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-22410782

RESUMO

Prohibitin (PHB) is indispensable for Ras-induced Raf-1 activation, cell migration and growth; however, the exact role of PHB in the molecular pathogenesis of cancer metastasis remains largely unexamined. Here, we found a positive correlation between plasma membrane-associated PHB and the clinical stages of cancer. The level of PHB phosphorylated at threonine 258 (T258) and tyrosine 259 (Y259) in human cancer-cell membranes correlated with the invasiveness of cancer cells. Overexpression of phosphorylated PHB (phospho-PHB) in the lipid-raft domain of the cell membrane enhanced cell migration/invasion through PI3K/Akt and Raf-1/ERK activation. It also enhanced epithelial-mesenchymal transition, matrix metalloproteinase-2 activity and invasiveness of cancer cells in vitro. Immunoprecipitation analysis demonstrated that phospho-PHB associated with Raf-1, Akt and Ras in the membrane and was essential for the activation of Raf-1 signaling by Ras. Mice implanted with cancer cells stably overexpressing PHB in the plasma membrane showed enlarged cervical tumors, enhanced metastasis and shorter survival time compared with mice implanted with cancer cells without PHB overexpression. Dephosphorylation of PHB at T258 by site-directed mutagenesis diminished the in vitro and in vivo effects of PHB. These results suggest that increase in phospho-PHB T258 in the raft domain of the plasma membrane has a role in the Ras-driven activation of PI3K/Akt and Raf-1/ERK-signaling cascades and results in the promotion of cancer metastasis.


Assuntos
Neoplasias do Colo do Útero/metabolismo , Animais , Membrana Celular/metabolismo , Transição Epitelial-Mesenquimal , Feminino , Genes ras , Humanos , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Invasividade Neoplásica , Metástase Neoplásica , Transplante de Neoplasias , Fosforilação , Proibitinas , Proteínas Proto-Oncogênicas c-raf/genética , Proteínas Repressoras
14.
Allergy ; 67(10): 1271-81, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22913540

RESUMO

BACKGROUND: Recent evidence indicates that Staphylococcus aureus, one of the most important human pathogens, secretes vesicles into the extracellular milieu. OBJECTIVE: To evaluate whether inhalation of S. aureus-derived extracellular vesicles (EV) is causally related to the pathogenesis of inflammatory pulmonary diseases. METHODS: Staphylococcus aureus EV were prepared by sequential ultrafiltration and ultracentrifugation. The innate immune response was evaluated in vitro after the application of EV to airway epithelial cells and alveolar macrophages. In vivo innate and adaptive immune responses were evaluated after airway exposure to EV. Adjuvant effects of EV on the development of hypersensitivity to inhaled allergens were also evaluated after airway sensitization with S. aureus EV and ovalbumin (OVA). RESULTS: Staphylococcus aureus and S. aureus EV were detected in house dust. Alveolar macrophages produced both tumor necrosis α (TNF-α) and interleukin 6 (IL-6) after in vitro stimulation with S. aureus EV, whereas airway epithelial cells produced only IL-6. Repeated airway exposure to S. aureus EV induced both Th1 and Th17 cell responses and neutrophilic pulmonary inflammation, mainly via a Toll-like receptor 2 (TLR2)-dependent mechanism. In terms of adjuvant effects, airway sensitization with S. aureus EV and OVA resulted in neutrophilic pulmonary inflammation after OVA challenge alone. This phenotype was partly reversed by the absence of interferon γ (IFN-γ) or IL-17. CONCLUSION: Staphylococcus aureus EV can induce Th1 and Th17 neutrophilic pulmonary inflammation, mainly in a TLR2-dependent manner. Additionally, S. aureus EV enhance the development of airway hypersensitivity to inhaled allergens.


Assuntos
Vesículas Citoplasmáticas/imunologia , Pneumonia , Staphylococcus aureus , Células Th1/imunologia , Células Th17/imunologia , Vesículas Citoplasmáticas/ultraestrutura , Humanos , Infiltração de Neutrófilos/imunologia , Neutrófilos/imunologia , Pneumonia/imunologia , Pneumonia/fisiopatologia , Staphylococcus aureus/imunologia , Staphylococcus aureus/ultraestrutura
16.
Clin Exp Immunol ; 167(3): 438-46, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22288587

RESUMO

The immune receptor expressed on myeloid cells 1 (IREM-1/CD300F) has been shown to inhibit various inflammatory processes in myeloid cells, such as macrophages and mast cells. IREM-1 exerts its inhibitory effect through its intracellular immunoreceptor tyrosine-based inhibition motifs (ITIMs). In order to generate immunomodulatory molecules that can regulate the inflammatory activation of macrophages, decapeptides representing each of the five ITIM-like sequences in the cytoplasmic tail of IREM-1 were synthesized in conjugation with human immunodeficiency virus-transactivator of transcription (HIV-TAT(48-57)), which was added to promote internalization of the peptides. Interestingly, all these TAT-ITIM fusion peptides inhibited Toll-like receptor (TLR)-mediated production of proinflammatory molecules, including matrix metalloproteinase (MMP)-9, tumour necrosis factor (TNF)-α, monocyte chemotactic protein-1 (MCP-1) and interleukin (IL)-8. When various TLR ligands were used to stimulate the human macrophage-like cell line human acute monocytic leukaemia cell line (THP)-1, the TAT-ITIM peptides blocked both myeloid differentiation factor 88 (MyD88) and Toll-interleukin 1 receptor (TIR)-domain-containing adapter-inducing interferon-ß (TRIF)-mediated TLR signalling pathways. Utilization of specific inhibitors and detection of the active form of signalling adaptors by Western blot analysis further demonstrated that the inhibitory effects of these TAT-ITIM peptides require activation of Src homology 2 (SH2)-containing tyrosine phosphatase (SHP) and/or phosphoinositide 3-kinase (PI3K). These data indicate that these synthetic peptides may be used to regulate immune responses that involve TLR-mediated inflammatory activation of macrophages.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/imunologia , Fator 88 de Diferenciação Mieloide/imunologia , Fosfatidilinositol 3-Quinases/metabolismo , Receptores Imunológicos/imunologia , Proteínas Tirosina Fosfatases Contendo o Domínio SH2/metabolismo , Receptores Toll-Like/imunologia , Sequência de Aminoácidos , Linhagem Celular , Humanos , Interleucina-8/biossíntese , Metaloproteinase 9 da Matriz/biossíntese , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/imunologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , Receptores Imunológicos/química , Receptores Imunológicos/genética , Transdução de Sinais/imunologia , Produtos do Gene tat do Vírus da Imunodeficiência Humana/química , Produtos do Gene tat do Vírus da Imunodeficiência Humana/genética , Produtos do Gene tat do Vírus da Imunodeficiência Humana/imunologia
18.
Oncogene ; 30(21): 2463-74, 2011 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-21258409

RESUMO

MicroRNAs (miRNAs) are involved in tumorigenecity by regulating specific oncogenes and tumor suppressor genes, and their roles in breast cancer stem cells (BCSCs) are becoming apparent. Distinct from the CD44(+)/CD24(-/low) sub-population, we have isolated a novel PROCR(+)/ESA(+) BCSC sub-population. To explore miRNA-regulatory mechanisms in this sub-population, we performed miRNA expression profiling and found miR-495 as the most highly upegulated miRNA in PROCR(+)/ESA(+) cells. Coincidently, high upregulation of miR-495 was also found in CD44(+)/CD24(-/low) BCSCs, reflecting its potential importance in maintaining common BCSC properties. Ectopic expression of miR-495 in breast cancer cells promoted their colony formation in vitro and tumorigenesis in mice. miR-495 directly suppressed E-cadherin expression to promote cell invasion and inhibited REDD1 expression to enhance cell proliferation in hypoxia through post-transcriptional mechanism. miR-495 expression was directly modulated by transcription factor E12/E47, which itself is highly expressed in BCSCs. These findings reveal a novel regulatory pathway centered on miR-495 that contributes to BCSC properties and hypoxia resistance.


Assuntos
Caderinas/genética , MicroRNAs/genética , Células-Tronco Neoplásicas/metabolismo , Fator 3 de Transcrição/genética , Fatores de Transcrição/genética , Animais , Sequência de Bases , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Caderinas/metabolismo , Hipóxia Celular , Linhagem Celular , Linhagem Celular Tumoral , Regulação para Baixo , Feminino , Perfilação da Expressão Gênica , Células HEK293 , Humanos , Immunoblotting , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Dados de Sequência Molecular , Regiões Promotoras Genéticas/genética , Ligação Proteica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator 3 de Transcrição/metabolismo , Fatores de Transcrição/metabolismo , Transplante Heterólogo
19.
Eur J Vasc Endovasc Surg ; 40(6): 736-8, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20926320

RESUMO

Endograft mal-deployment during endovascular aneurysm repair (EVAR) may cause renal artery (RA) occlusion. We report bilateral RA occlusion following insertion of an endograft with suprarenal barb fixation. Attempted salvage using the 'pull-down' technique caused right RA dissection. Despite bilateral RA stenting, the right kidney was atrophic on an ultrasound scan at 6 months. This potential hazard of devices with suprarenal barb fixation should be considered when repositioning is attempted.


Assuntos
Aneurisma da Aorta Abdominal/cirurgia , Dissecção Aórtica/etiologia , Implante de Prótese Vascular/instrumentação , Prótese Vascular , Procedimentos Endovasculares/instrumentação , Rim/irrigação sanguínea , Obstrução da Artéria Renal/etiologia , Stents , Idoso , Dissecção Aórtica/diagnóstico por imagem , Aneurisma da Aorta Abdominal/diagnóstico por imagem , Atrofia , Implante de Prótese Vascular/efeitos adversos , Procedimentos Endovasculares/efeitos adversos , Feminino , Humanos , Hipertrofia , Rim/diagnóstico por imagem , Desenho de Prótese , Radiografia , Obstrução da Artéria Renal/diagnóstico por imagem , Obstrução da Artéria Renal/cirurgia , Circulação Renal , Reoperação , Fatores de Tempo , Ultrassonografia
20.
Clin Exp Immunol ; 161(3): 504-11, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20646006

RESUMO

The immune receptor expressed on myeloid cells 1 (IREM-1) has been known to regulate the activities of myeloid cells through its immunoreceptor tyrosine-based inhibition motifs (ITIMs) in its intracellular region. In order to investigate its effect on macrophage activation, a human macrophage cell line (THP-1) was tested after stimulation of its membrane-bound form of B cell activation factor (BAFF), which has been shown to modulate inflammatory activities through induction of proinflammatory mediator expression and suppression of phagocytosis. IREM-1-specific monoclonal antibodies detected the expression of high levels of IREM-1 in THP-1 cells. Cross-linking of IREM-1 with these antibodies resulted in the blockage of the BAFF-mediated expression of interleukin (IL)-8 and matrix metalloproteinase (MMP)-9 through inhibition of the activation of extracellular regulated kinase (ERK) and phosphorylation/degradation of IκB. Furthermore, cross-linking of IREM-1 also reversed the BAFF-mediated inhibition of phagocytosis. In order to demonstrate the role of ITIM in the IREM-1-mediated suppression of BAFF signalling, a decapeptide containing YADL (an ITIM in IREM-1) was fused with HIV-TAT(48-57) which was required for the internalization of the synthetic polypeptide (TAT-YADL). TAT-YADL, but not control peptides, recapitulated the effect of the anti-IREM-1 monoclonal antibody. These observations indicate that IREM-1 exerted its inhibitory effect on BAFF-medicated signalling through ITIM-mediated regulation of ERK activities in THP-1 cells.


Assuntos
Fator Ativador de Células B/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Macrófagos/metabolismo , Receptores Imunológicos/metabolismo , Sequência de Aminoácidos , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais/farmacologia , Fator Ativador de Células B/genética , Fator Ativador de Células B/imunologia , Linhagem Celular , Endocitose/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Citometria de Fluxo , Células HEK293 , Humanos , Interleucina-8/metabolismo , Macrófagos/citologia , Macrófagos/imunologia , Metaloproteinase 9 da Matriz/metabolismo , Oligopeptídeos/síntese química , Oligopeptídeos/metabolismo , Oligopeptídeos/farmacologia , Fosforilação , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , Receptores Imunológicos/genética , Receptores Imunológicos/imunologia , Transdução de Sinais , Transfecção , Células U937
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA