Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Int J Mol Sci ; 20(9)2019 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-31075975

RESUMO

PDCD4 is a novel tumor suppressor to show multi-functions inhibiting cell growth, tumor invasion, metastasis, and inducing apoptosis. PDCD4 protein binds to the translation initiation factor eIF4A, some transcription factors, and many other factors and modulates the function of the binding partners. PDCD4 downregulation stimulates and PDCD4 upregulation inhibits the TPA-induced transformation of cells. However, PDCD4 gene mutations have not been found in tumor cells but gene expression was post transcriptionally downregulated by micro environmental factors such as growth factors and interleukins. In this review, we focus on the suppression mechanisms of PDCD4 protein that is induced by the tumor promotors EGF and TPA, and in the inflammatory conditions. PDCD4-protein is phosphorylated at 2 serines in the SCFßTRCP ubiquitin ligase binding sequences via EGF and/or TPA induced signaling pathway, ubiquitinated, by the ubiquitin ligase and degraded in the proteasome system. The PDCD4 protein synthesis is inhibited by microRNAs including miR21.


Assuntos
Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Sequência de Aminoácidos , Animais , Apoptose/genética , Carcinogênese/genética , Carcinogênese/patologia , Regulação Neoplásica da Expressão Gênica , Humanos , Fosforilação , Transcrição Gênica , Proteínas Supressoras de Tumor/química
2.
Int J Mol Sci ; 20(5)2019 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-30813635

RESUMO

Hypoxia-inducible factor 1 (HIF-1) plays important roles in cancer cell biology. HIF-1α is reportedly activated by several factors, including protein kinase C (PKC), in addition to hypoxia. We investigated the role of PKC isoforms and the effects of vitamin K2 (VK2) in the activation process of HIF-1α. Human hepatocellular carcinoma (HCC)-derived Huh7 cells were cultured under normoxic and hypoxic (1% O2) conditions with or without the PKC stimulator TPA. The expression, transcriptional activity and nuclear translocation of HIF-1α were examined under treatment with PKC inhibitors, siRNAs against each PKC isoform and VK2. Hypoxia increased the expression and activity of HIF-1α. TPA increased the HIF-1α activity several times under both normoxic and hypoxic conditions. PKC-δ siRNA-mediated knockdown, PKC-δ inhibitor (rottlerin) and pan-PKC inhibitor (Ro-31-8425) suppressed the expression and transcriptional activity of HIF-1α. VK2 significantly inhibited the TPA-induced HIF-1α transcriptional activity and suppressed the expression and nuclear translocation of HIF-1α induced by TPA without altering the HIF-1α mRNA levels. These data indicate that PKC-δ enhances the HIF-1α transcriptional activity by increasing the nuclear translocation, and that VK2 might suppress the HIF-1α activation through the inhibition of PKC in HCC cells.


Assuntos
Carcinoma Hepatocelular/patologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Neoplasias Hepáticas/patologia , Proteína Quinase C/metabolismo , Vitamina K 2/farmacologia , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Isoenzimas/metabolismo , Regiões Promotoras Genéticas/genética , Transporte Proteico , Acetato de Tetradecanoilforbol/farmacologia , Fator A de Crescimento do Endotélio Vascular/genética
3.
Luminescence ; 32(6): 1009-1016, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28378893

RESUMO

Certain marine organisms have been known to cause allergic reactions among occupational fishermen. We have previously reported that bronchial asthma among the workers engaged in spiny lobster fishing in Japan was caused by octocorals such as Dendronephthya sp. and Scleronephthya gracillima (previously named Alcyonium gracillimum). Now we have found another octocoral, Scleronephthya gracillima (Kuekenthal), which causes the allergic disease in fishermen. The octocoral was characterized as a new green fluorescent protein (GFP)-like family. The new allergen has a molecular mass of 27 kDa in 1D and 2D SDS-PAGE under reduced conditions. The 27 kDa component was determined to be an allergen by western blotting, ECL immune staining method and absorption of patient sera with the antigen. Furthermore, the combination of analysis with LC-ESI-MS/MS and MASCOT search in the NCBInr database concluded the 27 kDa component had the sequence YPADI/LPDYFK, and that the 22 kDa component had the sequence QSFPEGFSWER, which both matched a GFP-like protein in Acropora aculeus and in Montastraea annularis. Further analysis by MALDI-TOF/MS/MS and MASCOT search in the NCBInr database of all 27 kDa eight spot components from 2D SDS-PAGE indicated that the sequence QSFPEGFSWER also matched as GFP-like protein in Lobophyllia hemprichii and Scleractinia sp. To our knowledge, this is the first report of the new allergenic protein that corresponds to a new GFP-like protein named Akane, and which has fluorescent emissions in the red and green part of the spectra at 628 nm and 508 nm, respectively.


Assuntos
Alérgenos/química , Antozoários/imunologia , Proteínas de Fluorescência Verde/química , Alérgenos/imunologia , Motivos de Aminoácidos , Animais , Antozoários/química , Mapeamento de Epitopos , Fluorescência , Proteínas de Fluorescência Verde/imunologia , Peso Molecular , Espectrometria de Massas em Tandem
4.
Tumour Biol ; 34(6): 3879-85, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23838800

RESUMO

Nowadays, digestive tract cancers become the commonest neoplasia and one of the leading causes of cancer deaths worldwide. The development of diagnosis and therapy is urgently required. Programmed cell death 4 (PDCD4), a new tumor suppressor, has been documented to be a potential diagnostic tool and treatment target for neoplasia due to the inhabitation of tumor promotion/progression and metastasis. However, its role in human digestive tract cancers is few available up to now. In this study, we examined the expression of PDCD4 in human digestive tract cancers (61 gastric cancer, 65 colorectal cancer, and 69 pancreatic cancer patients) by Western blot analysis, reverse transcription (RT)-PCR, and immunohistochemistry. Western blot, RT-PCR, and immunohistochemistry examination showed that expressions of PDCD4 were significantly lower in cancers specimens than in noncancerous tissues. Among the different differentiated cancer tissues, PDCD4 expression was significantly lower in moderately or poorly differentiated cancers than in well-differentiated cancers (p < 0.05). Our findings suggested that PDCD4 might be a potentially valuable molecular target in diagnosis and therapy for human digestive tract cancers.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Transformação Celular Neoplásica/metabolismo , Regulação para Baixo , Neoplasias Gastrointestinais/metabolismo , Proteínas de Ligação a RNA/metabolismo , Idoso , Proteínas Reguladoras de Apoptose/genética , Western Blotting , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Feminino , Mucosa Gástrica/metabolismo , Neoplasias Gastrointestinais/genética , Neoplasias Gastrointestinais/patologia , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Proteínas de Ligação a RNA/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estômago/patologia
5.
Hum Cell ; 36(2): 583-601, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36522523

RESUMO

Although programmed cell death 4 (PDCD4) was initially reported as a tumor suppressor and has been shown to inhibit cancer cell growth and metastasis, recent studies have demonstrated that loss of PDCD4 expression also induces growth inhibition by inducing apoptosis and/or cellular senescence. At present, the roles of PDCD4 in the activation and profibrogenic properties of myofibroblasts, which are critically involved in organ fibrosis, such as that in the liver, are unclear. We, therefore, investigated the roles of PDCD4 in myofibroblasts using human hepatic stellate cell line Lieming Xu-2 (LX-2). PDCD4 knockdown inhibited LX-2 proliferation and induced a senescent phenotype with increased ß-galactosidase staining and p21 expression in a p53-independent manner together with downregulation of the notch signaling mediator RBJ-κ/CSL. During PDCD4 knockdown, alpha smooth muscle actin (α-SMA; an activation marker of myofibroblasts), matrix metalloproteinases MMP-1 and MMP-9, and collagen IV were upregulated, but the expression of collagen1α1 and collagen III was markedly downregulated without any marked change in the expression of tissue inhibitor of metalloproteinase-1 (TIMP-1). These results demonstrated that knockdown of PDCD4 induced the cellular senescence phenotype and activated myofibroblasts while suppressing the profibrogenic phenotype, suggesting roles of PDCD4 in cellular senescence and fibrogenesis in the liver.


Assuntos
Proteínas Reguladoras de Apoptose , Células Estreladas do Fígado , Proteínas de Ligação a RNA , Humanos , Apoptose , Proteínas Reguladoras de Apoptose/genética , Colágeno/metabolismo , Fenótipo , Proteínas de Ligação a RNA/genética , Inibidor Tecidual de Metaloproteinase-1/genética , Inibidor Tecidual de Metaloproteinase-1/metabolismo
6.
Biochim Biophys Acta ; 1803(9): 1020-7, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20471435

RESUMO

Transforming growth factor-beta1 (TGF-beta1) induces apoptosis in normal hepatocytes and hepatoma cells. PDCD4 is involved in TGF-beta1-induced apoptosis via the Smad pathway. The tumor promoter 12-O-tetradecanoylphorbor-13-acetate (TPA), a protein kinase C stimulator, inhibits TGF-beta1-induced apoptosis. However, the mechanisms of TPA action on PDCD4 expression remain to be elucidated. Therefore. the regulatory mechanism of PDCD4 expression by PKC was investigated. The treatment of the human hepatoma cell line, Huh7 with TPA suppressed PDCD4 protein expression and TGF-beta1 failed to increase the PDCD4 protein expression. PKC inhibitors Ro-31-8425 or bisindolylmaleimide-1-hydrocholoride (pan-PKC inhibitors) and rottlerin (PKCdelta inhibitor), but not Go6976 (PKCalpha inhibitor), enhanced the induction of PDCD4 protein by TGF-beta1. Furthermore, siRNA-mediated knockdown of PKCdelta and epsilon, but not PKCalpha, augmented the TGF-beta1-stimulated PDCD4 protein expression. However, TPA or pan-PKC inhibitor did not alter the PDCD4 mRNA expression either under basal- and TGF-beta1-treated conditions. The down-regulation of PDCD4 by TPA was restored by treatment with the proteasome inhibitor MG132. These data suggest that two isoforms of PKCs are involved in the regulation of the PDCD4 protein expression related to the proteasomal degradation pathway.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Proteína Quinase C/fisiologia , Proteínas de Ligação a RNA/metabolismo , Proteínas Reguladoras de Apoptose/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes Supressores de Tumor/fisiologia , Humanos , Isoenzimas/metabolismo , Isoenzimas/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatidilinositol 3-Quinases/fisiologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Complexo de Endopeptidases do Proteassoma/fisiologia , Inibidores de Proteassoma , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-akt/fisiologia , RNA Interferente Pequeno/farmacologia , Proteínas de Ligação a RNA/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Acetato de Tetradecanoilforbol/farmacologia , Fator de Crescimento Transformador beta1/farmacologia
7.
Cells ; 9(1)2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31952347

RESUMO

PDCD4 (programmed cell death 4) is a tumor suppressor that plays a crucial role in multiple cellular functions, such as the control of protein synthesis and transcriptional control of some genes, the inhibition of cancer invasion and metastasis. The expression of this protein is controlled by synthesis, such as via transcription and translation, and degradation by the ubiquitin-proteasome system. The mitogens, known as tumor promotors, EGF (epidermal growth factor) and TPA (12-O-tetradecanoylphorbol-13-acetate) stimulate the degradation of PDCD4 protein. However, the whole picture of PDCD4 degradation mechanisms is still unclear, we therefore investigated the relationship between PDCD4 and autophagy. The proteasome inhibitor MG132 and the autophagy inhibitor bafilomycin A1 were found to upregulate the PDCD4 levels. PDCD4 protein levels increased synergistically in the presence of both inhibitors. Knockdown of p62/SQSTM1 (sequestosome-1), a polyubiquitin binding partner, also upregulated the PDCD4 levels. P62 and LC3 (microtubule-associated protein 1A/1B-light chain 3)-II were co-immunoprecipitated by an anti-PDCD4 antibody. Colocalization particles of PDCD4, p62 and the autophagosome marker LC3 were observed and the colocalization areas increased in the presence of autophagy and/or proteasome inhibitor(s) in Huh7 cells. In ATG (autophagy related) 5-deficient Huh7 cells in which autophagy was impaired, the PDCD4 levels were increased at the basal levels and upregulated in the presence of autophagy inhibitors. Based on the above findings, we concluded that after phosphorylation in the degron and ubiquitination, PDCD4 is degraded by both the proteasome and autophagy systems.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Autofagia , Proteínas de Ligação a RNA/metabolismo , Proteínas Reguladoras de Apoptose/genética , Humanos , Proteínas de Ligação a RNA/genética , Células Tumorais Cultivadas
8.
Front Oncol ; 8: 661, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30687637

RESUMO

While the over-expression of tumor suppressor programmed cell death 4 (PDCD4) induces apoptosis, it was recently shown that PDCD4 knockdown also induced apoptosis. In this study, we examined the cell cycle regulators whose activation is affected by PDCD4 knockdown to investigate the contribution of PDCD4 to cell cycle regulation in three types of hepatoma cells: HepG2, Huh7 (mutant p53 and p16-deficient), and Hep3B (p53- and Rb-deficient). PDCD4 knockdown suppressed cell growth in all three cell lines by inhibiting Rb phosphorylation via down-regulating the expression of Rb itself and CDKs, which phosphorylate Rb, and up-regulating the expression of the CDK inhibitor p21 through a p53-independent pathway. We also found that apoptosis was induced in a p53-dependent manner in PDCD4 knockdown HepG2 cells (p53+), although the mechanism of cell death in PDCD4 knockdown Hep3B cells (p53-) was different. Furthermore, PDCD4 knockdown induced cellular senescence characterized by ß-galactosidase staining, and p21 knockdown rescued the senescence and cell death as well as the inhibition of Rb phosphorylation induced by PDCD4 knockdown. Thus, PDCD4 is an important cell cycle regulator of hepatoma cells and may be a promising therapeutic target for the treatment of hepatocellular carcinoma.

9.
Int J Mol Med ; 42(2): 957-965, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29786754

RESUMO

Although interferon (IFN)­based treatment of patients with chronic hepatitis C virus (HCV) infection is widely applied, treatment resistance is often observed in patients with advanced liver fibrosis. Given that the molecular mechanisms of IFN resistance in liver fibrosis remain elusive, the present study investigated the effects of extracellular matrix (ECM) on IFN signaling in hepatic cells. The native HuH­7 human hepatoma cell line and HuH­7 cells were stably transfected with full­length HCV­RNA fused with Renilla luciferase (OR6 cells) were cultured on ECM­coated dishes or non­coated plastic dishes (NDs), and treated with human IFN­α. In Huh­7 cells cultured on coated dishes, the IFN­stimulated response element (ISRE) luciferase activity was measured following ISRE plasmid transfection and the expression of IFN­stimulated genes (ISG) were significantly lower than those in cells cultured on NDs. In addition, after IFN­α treatment, the amount of HCV­RNA and viral protein produced by OR6 cells cultured on coated dishes was higher than that produced by cells cultured on NDs. When cells were treated with ß1­integrin­blocking antibody to disrupt the cell­matrix interaction, the ISRE luciferase activity was restored, and the protein expression of ISG was increased, while that of HCV proteins was suppressed. Treatment of cells with integrin­linked kinase (ILK) inhibitor or focal adhesion kinase (FAK) inhibitor restored the ISRE luciferase activity and expression of ISG proteins. These results suggested that ß1­integrin­mediated signals affected the IFN signaling and promoted HCV replication. Therefore, the accumulation of ECM in liver fibrosis may impair IFN signaling through ß1­integrin­mediated signaling involving ILK and FAK.


Assuntos
Matriz Extracelular/imunologia , Hepacivirus/fisiologia , Hepatite C/imunologia , Interferon-alfa/imunologia , Transdução de Sinais , Linhagem Celular Tumoral , Matriz Extracelular/virologia , Hepacivirus/genética , Hepacivirus/imunologia , Hepatite C/virologia , Hepatócitos/imunologia , Hepatócitos/virologia , Humanos , Proteínas Serina-Treonina Quinases/imunologia , RNA Viral/genética , Replicação Viral
10.
Oncogene ; 23(49): 8135-45, 2004 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-15361828

RESUMO

The growth of human breast tumor cells is regulated through signaling involving cell surface growth factor receptors and nuclear receptors of the steroid/thyroid/retinoid receptor gene family. Retinoic acid receptors (RARs), members of the steroid/thyroid hormone receptor gene family, are ligand-dependent transcription factors, which have in vitro and in vivo growth inhibitory activity against breast cancer cells. RAR-agonists inhibit the proliferation of many human breast cancer cell lines, particularly those whose growth is stimulated by estradiol (E2) or growth factors. Additionally, RAR-agonists and synthetic retinoids such as Ferentinide have been shown to induce apoptosis in malignant breast cells but not normal breast cells. To better define the genes involved in RAR-mediated growth inhibition of breast cancer cells, we used oligonucleotide microarray analysis to create a database of genes that are potentially regulated by RAR-agonists in breast cancer cells. We found that PDCD4 (programmed cell death 4), a tumor suppressor gene presently being evaluated as a target for chemoprevention, was induced about three-fold by the RARalpha-selective agonist Am580, in T-47D breast cancer cells. RAR pan-agonists and Am580, but not retinoid X receptors (RXR)-agonists, stimulate the expression of PDCD4 in a wide variety of retinoid-inhibited breast cancer cell lines. RAR-agonists did not induce PDCD4 expression in breast cancer cell lines, which were not growth inhibited by retinoids. We also observed that antiestrogen and the HER-2/neu antagonist, Herceptin (Trastuzumab), also induced PDCD4 expression in T-47D cells, suggesting that PDCD4 may play a central role in growth inhibition in breast cancer cells. Transient overexpression of PDCD4 in T-47D (ER+, RAR+) and MDA-MB-231 (ER-, RAR-) cells resulted in apoptotic death, suggesting a role for PDCD4 in mediating apoptosis in breast cancer cells. PDCD4 protein expression has previously been reported in small ductal epithelium of normal breast. To date, there has been no report of induction of PDCD4 expression by RAR-agonists, antiestrogen or HER2/neu antagonist in breast cancer cells and its potential role in apoptosis in these cells.


Assuntos
Apoptose , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Antagonistas de Estrogênios/farmacologia , Regulação Neoplásica da Expressão Gênica , Proteínas de Ligação a RNA/genética , Receptor ErbB-2/fisiologia , Receptores do Ácido Retinoico/fisiologia , Proteínas Reguladoras de Apoptose , Ciclo Celular , Linhagem Celular Tumoral , Humanos , Proteínas de Ligação a RNA/fisiologia , Receptor ErbB-2/antagonistas & inibidores , Receptores do Ácido Retinoico/agonistas , Receptores X de Retinoides , Fatores de Transcrição/fisiologia
11.
Mech Dev ; 119 Suppl 1: S11-9, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14516654

RESUMO

We have identified a secreted glycoprotein, neural epidermal growth factor-like like 2 (NELL2), in a screen designed to isolate molecules regulating sensory neuron genesis and differentiation in the dorsal root ganglia (DRG). In investigating NELL2 expression during embryogenesis, we demonstrate here that NELL2 is highly regulated spatially and temporally, being only transiently expressed in discrete regions of the central (CNS) and peripheral nervous systems (PNS) and in a subset of mesoderm derived structures during their peak periods of development. In the CNS and PNS, NELL2 is maximally expressed as motor and sensory neurons differentiate. Interestingly, its expression is restricted to sublineages of the neural crest, being strongly expressed throughout the immature DRG, but excluded from sympathetic ganglia. Similarly during muscle development, NELL2 is specifically expressed by hypaxial muscle precursor cells in the differentiating somite and derivatives in the forelimbs and body wall, but not by epaxial muscle precursors. Furthermore, NELL2 is differentially regulated in the CNS and PNS; in the CNS, NELL2 is only expressed by nascent, post-mitotic neurons as they commence their differentiation, yet in the PNS, NELL2 is expressed by subsets of progenitor cells in addition to nascent neurons. Based on this restricted spatial and temporal expression pattern, functional studies are in progress to determine NELL2's role during neuronal differentiation in both the PNS and CNS.


Assuntos
Gânglios Espinais , Neurônios , Animais , Diferenciação Celular , Família de Proteínas EGF , Gânglios Espinais/citologia , Proteínas do Tecido Nervoso/metabolismo , Crista Neural/metabolismo , Neurônios/metabolismo
12.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 27(5): 597-600, 2005 Oct.
Artigo em Zh | MEDLINE | ID: mdl-16274040

RESUMO

OBJECTIVE: To investigate the expression of programmed cell death 4 (PDCD4) protein and its clinicopathological significance in human pancreatic cancer. METHODS: Immunohistochemistry was used to examine the expression of PDCD4 protein in 69 specimens of pancreatic cancer and Western blot in 8 fresh specimens. RESULTS: The expression of PDCD4 protein was significantly lower in all 8 fresh pancreatic cancer tissues than that in non-cancerous tissues detected by Western blot. Compared with non-cancerous pancreatic tissue (> 80% of positive cells), low PDCD4 expression was shown in 69 pancreatic cancer tissues (< 30% of positive cells in 36 cases and 30%-80% of positive expression cells in 33 cases). In the 33 cases with 30% and 80% of positive expression cells, the expression rates of PDCD4 protein were 57.6%, 24.2%, and 18.2% in well, moderately, and poorly differentiated cancers, respectively. In the 36 cases less than 30% of positive expression cells, however, the expression rate of PDCD4 protein in well, moderately, and poorly differentiated cases were 19.4%, 41.7%, and 38.9%, respectively. 67.4% (15/23) of the moderately differentiated cases and 70% (14/20) of the poorly differentiated cases showed < 30% of positive expression cells. Only 26.9% (7/26) of the well differentiated cases, however, showed < 30% of positive expression cells, indicating that low PDCD4 expression was associated with histological grade (P < 0.01). There was no relationship between PDCD4 expression and other clinicopathological parameters including patients' sex, age, and TNM stage. CONCLUSIONS: Expression of PDCD4 protein is low in human pancreatic cancer and is correlated with the differentiation levels of human pancreatic cancer. PDCD4 may play an important role in the occurrence and development of pancreatic carcinomas.


Assuntos
Proteínas Reguladoras de Apoptose/biossíntese , Biomarcadores Tumorais/análise , Neoplasias Pancreáticas/metabolismo , Proteínas de Ligação a RNA/biossíntese , Adulto , Idoso , Western Blotting , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Neoplasias Pancreáticas/patologia
13.
Gene Expr Patterns ; 2(1-2): 7-15, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12617830

RESUMO

We have identified a secreted glycoprotein, neural epidermal growth factor-like like 2 (NELL2), in a screen designed to isolate molecules regulating sensory neuron genesis and differentiation in the dorsal root ganglia (DRG). In investigating NELL2 expression during embryogenesis, we demonstrate here that NELL2 is highly regulated spatially and temporally, being only transiently expressed in discrete regions of the central (CNS) and peripheral nervous systems (PNS) and in a subset of mesoderm derived structures during their peak periods of development. In the CNS and PNS, NELL2 is maximally expressed as motor and sensory neurons differentiate. Interestingly, its expression is restricted to sublineages of the neural crest, being strongly expressed throughout the immature DRG, but excluded from sympathetic ganglia. Similarly during muscle development, NELL2 is specifically expressed by hypaxial muscle precursor cells in the differentiating somite and derivatives in the forelimbs and body wall, but not by epaxial muscle precursors. Furthermore, NELL2 is differentially regulated in the CNS and PNS; in the CNS, NELL2 is only expressed by nascent, post-mitotic neurons as they commence their differentiation, yet in the PNS, NELL2 is expressed by subsets of progenitor cells in addition to nascent neurons. Based on this restricted spatial and temporal expression pattern, functional studies are in progress to determine NELL2's role during neuronal differentiation in both the PNS and CNS.


Assuntos
Diferenciação Celular/genética , Células Musculares/metabolismo , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Animais , Sistema Nervoso Central/embriologia , Sistema Nervoso Central/metabolismo , Embrião de Galinha , Células Musculares/citologia , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/metabolismo , Crista Neural/metabolismo , Neurônios/citologia , Especificidade de Órgãos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Somitos/metabolismo
14.
Cell Signal ; 26(3): 603-10, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24334270

RESUMO

In this study, we demonstrate that EGF inhibits the TGF-ß1-induced apoptosis of Huh7 cells. TGF-ß1 up-regulates the expression of PDCD4 causing apoptosis, by stimulating the synthesis of PDCD4 mRNA via the Smad signaling pathway. TGF-ß1 also inhibits the activation of S6 kinase 1 which phosphorylates the serine 67 residue of PDCD4 and leads to the phosphorylation of serine 71 and serine 76 in the ß-TRCP binding sequence. This phosphorylation sequence causes the protein to be degraded in the ubiquitin-proteasome system. EGF activates S6 kinase 1 via the PI3K-Akt-mTOR signaling pathway and stimulates the degradation of PDCD4. EGF also suppresses PDCD4 mRNA levels. As the mTOR inhibitor rapamycin up-regulated PDCD4 mRNA levels, the PI3K-Akt-mTOR signaling pathway may control the transcription of the PDCD4 gene as well as the degradation of the protein. TPA also inhibited the TGF-ß1-induced apoptosis of Huh7 cells, stimulating the degradation of the PDCD4-protein. Analyses using PDCD4 mutants with changes of serines 67, 71 and 76 to alanine revealed that the phosphorylation of serine 67 is not essential for the TPA-induced suppression of the protein. The mitogens could not suppress the PDCD4-mutant proteins with changes of serine 71 and/or serine 76 to alanine, however, indicating that phosphorylations at these residues are necessary for the proteasome-mediated degradation of PDCD4. The phosphor-mimic S71/D and S76/D mutants were able to be degraded in the ubiquitin-proteasome system unlike the mutants with changes of serine to alanine. The expression of S71/D mutant was suppressed with EGF but that of S76/D mutant was not indicating that at least partly the phosphorylation of both sites was mediated by different enzymes.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Apoptose/genética , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas Reguladoras de Apoptose/biossíntese , Proteínas Reguladoras de Apoptose/genética , Linhagem Celular Tumoral , Fator de Crescimento Epidérmico/farmacologia , Humanos , Mutação , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , RNA Mensageiro/biossíntese , Proteínas de Ligação a RNA/biossíntese , Proteínas de Ligação a RNA/genética , Proteínas Quinases S6 Ribossômicas 70-kDa , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Sirolimo/farmacologia , Proteínas Smad/genética , Serina-Treonina Quinases TOR/antagonistas & inibidores , Transcrição Gênica , Ativação Transcricional , Fator de Crescimento Transformador beta1/farmacologia
15.
J Nutr Biochem ; 23(12): 1668-75, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22475810

RESUMO

Vitamin K (VK) has diverse protective effects against osteoporosis, atherosclerosis and carcinogenesis. We recently reported that menatetrenone, a VK2 analogue, suppressed nuclear factor (NF)-κB activation in human hepatoma cells. Although NF-κB is regulated by isoforms of protein kinase C (PKC), the involvement of PKCs in VK2-mediated NF-κB inhibition remains unknown. Therefore, the effects of VK2 on the activation and the kinase activity of each PKC isoform were investigated. The human hepatoma Huh7 cells were treated with PKC isoform-specific inhibitors and/or siRNAs against each PKC isoform with or without 12-O-tetradecanoylphorbol-13-acetate (TPA). VK2 inhibited the TPA-induced NF-κB activation in Huh7 cells. NF-κB activity was inhibited by the pan-PKC inhibitor Ro-31-8425, but not by the PKCα-specific inhibitor Gö6976. The knockdown of individual PKC isoforms including PKCα, δ and ɛ showed only marginal effects on the NF-κB activity. However, the knockdown of both PKCδ and PKCɛ, together with treatment with a PKCα-specific inhibitor, depressed the NF-κB activity. VK2 suppressed the PKCα kinase activity and the phosphorylation of PKCɛ after TPA treatment, but neither the activation nor the enzyme activity of PKCδ was affected. The knockdown of PKCɛ abolished the TPA-induced phosphorylation of PKD1, and the effects of PKD1 knockdown on NF-κB activation were similar to those of PKCɛ knockdown. Collectively, all of the PKCs, including α, δ and ɛ, and PKD1 are involved in the TPA-mediated activation of NF-κB. VK2 inhibited the NF-κB activation through the inhibition of PKCα and ɛ kinase activities, as well as subsequent inhibition of PKD1 activation.


Assuntos
Carcinoma Hepatocelular/tratamento farmacológico , Neoplasias Hepáticas/tratamento farmacológico , NF-kappa B/metabolismo , Proteína Quinase C/metabolismo , Vitamina K 2/farmacologia , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Inibidores Enzimáticos/farmacologia , Técnicas de Silenciamento de Genes , Humanos , Indóis/farmacologia , Neoplasias Hepáticas/metabolismo , Maleimidas/farmacologia , NF-kappa B/genética , Fosforilação , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/genética , Proteína Quinase C-delta/genética , Proteína Quinase C-delta/metabolismo , Proteína Quinase C-épsilon/genética , Proteína Quinase C-épsilon/metabolismo , RNA Interferente Pequeno , Acetato de Tetradecanoilforbol/farmacologia
16.
Front Physiol ; 2: 78, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22028694

RESUMO

Hepatocellular carcinoma (HCC) often arises from chronically diseased livers. Persistent liver inflammation causes the accumulation of excessive extracellular matrix (ECM) proteins and impairs the liver function, finally leading to the development of HCC. A pleiotropic cytokine, transforming growth factor (TGF)-ß1, plays critical roles throughout the process of fibrogenesis and hepatocarcinogenesis. In the liver, TGF-ß1 inhibits the proliferation of hepatocytes and stimulates the production of ECM from hepatic stellate cells (HSCs) to maintain tissue homeostasis. During disease progression, both growth factors/cytokines and the ECM alter the TGF-ß1 signals by modifying the phosphorylation of Smad proteins at their C-terminal and linker regions. TGF-ß1 stimulates the expression of integrins, cellular receptors for ECM, along with an increase in ECM accumulation. The activation of integrins by the ECM modulates the response to TGF-ß1 in hepatic cells, resulting in their resistance to TGF-ß1-induced growth suppression in hepatocytes and the sustained production of ECM proteins in activated HSCs/myofibroblasts. Both growth factor receptors and integrins modify the expression and/or functions of the downstream effectors of TGF-ß1, resulting in the escape of hepatocytes from TGF-ß1-induced apoptosis. Recent studies have revealed that the alterations of Smad phosphorylation that occur as the results of the crosstalk between TGF-ß1, growth factors and integrins could change the nature of TGF-ß1 signals from tumor suppression to promotion. Therefore, the modification of Smad phosphorylation could be an attractive target for the prevention and/or treatment of HCC.

17.
Oncol Rep ; 26(6): 1385-92, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21894439

RESUMO

The expression patterns of PDCD4, a tumor suppressor, and ß-catenin were immunohistologically investigated in gastric carcinoma tissues. In normal gastric tissues, PDCD4 was strongly expressed in the cell nuclei, but weakly expressed in the cytoplasm. In gastric adenocarcinoma tissues, nuclear PDCD4 expression was decreased, while cytoplasmic PDCD4 expression was unchanged or somewhat increased. In gastric signet ring cell carcinoma tissues, PDCD4 expression patterns were different from the expression patterns of the adenocarcinoma tissues, and PDCD4 was localized in the nuclei of the carcinoma cells as a belt in the middle of the epithelial layer. The nuclear localization of PDCD4 in the adenocarcinoma tissues was correlated with the membrane localization of ß-catenin, the activation of which stimulates invasion of colon cancer cells. PDCD4 expression was correlated with ß-catenin expression in gastric carcinoma cell lines, but not with E-cadherin, as the binding partner in the cell membrane.


Assuntos
Adenocarcinoma/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Expressão Gênica , Proteínas de Ligação a RNA/metabolismo , Neoplasias Gástricas/metabolismo , beta Catenina/metabolismo , Adenocarcinoma/patologia , Idoso , Idoso de 80 Anos ou mais , Proteínas Reguladoras de Apoptose/genética , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas de Ligação a RNA/genética , Neoplasias Gástricas/patologia , beta Catenina/genética
18.
Int J Oncol ; 35(6): 1441-7, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19885568

RESUMO

Integrins, heterodimeric receptors for the extracellular matrix, are known to modulate transforming growth factor-beta1 (TGF-beta1)-mediated cell behavior. However, the interplay between beta1-integrin and Smad signaling, regulated by TGF-beta1, is not clearly understood. This study focuses on the alterations of the regulatory Smads (R-Smads) by TGF-beta1 in beta1-integrin-transfected HepG2 cells. The phosphorylation at the C-terminal site of R-Smads by TGF-beta1 was impaired in the beta1-integrin-transfected cells. However, the R-Smads were constitutively phosphorylated at the linker region in a MAP kinase-dependent manner. Furthermore, the expression of a mutant Smad3, that lacks the phosphorylation sites in the linker region, restored the TGF-beta1-induced Smad transcriptional activity. These results suggest that beta1-integrin impairs the TGF-beta1-mediated signals through the altered phosphorylation of the R-Smads.


Assuntos
Integrina beta1/metabolismo , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Western Blotting , Matriz Extracelular/metabolismo , Humanos , Integrina beta1/genética , Fosforilação , Transdução de Sinais/fisiologia , Transfecção
19.
Exp Dermatol ; 16(3): 179-84, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17286809

RESUMO

Expression of a tumor suppressor gene, programmed cell death 4 (PDCD4), was investigated at the protein level in the human skin. Immunohistochemically, PDCD4 protein expressed mainly in suprabasal layers, while PDCD4-positive and -negative areas were observed discontinuously in the basal cell layer of the epidermis. In hair follicles, the suprabulbar area including the hair and inner root sheath was immunoreactive, while the bulbar area, containing germinative cells which were strongly proliferating cell nuclear antigen (PCNA)-positive, was not or less. PDCD4 therefore appears to be important in the differentiation of hair follicles. PDCD4-positive cells were localized in the inside layers while PCNA-positive cells were located in the basal layer in the outer root sheath of hair follicles. The cells of sebaceous glands and sweat glands also were PDCD4-positive. The PDCD4 protein was localized mostly in nuclei of cutaneous cells. PDCD4 expression was found to be suppressed in the epidermis overlying an adult T-cell lymphoma (ATL), possibly reflecting a paracrine effect of factors produced by ATL cells. PDCD4 expression was suppressed in the keratinocyte cell line HaCaT by exposure of cultures to epidermal growth factor, transforming growth factor-beta1 or hepatocyte growth factor. Immunohistochemically, various skin cancers tended to show less PDCD4 expression than normal skin. Promotion of expression might prove useful in preventing or treating certain skin cancers.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Proteínas de Ligação a RNA/metabolismo , Dermatopatias/metabolismo , Pele/citologia , Pele/metabolismo , Linhagem Celular , Núcleo Celular/metabolismo , Folículo Piloso/imunologia , Folículo Piloso/metabolismo , Humanos , Imuno-Histoquímica , Queratinócitos/citologia , Queratinócitos/metabolismo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Glândulas Sebáceas/citologia , Glândulas Sebáceas/metabolismo , Glândulas Sudoríparas/citologia , Glândulas Sudoríparas/metabolismo
20.
Biochem Biophys Res Commun ; 293(1): 617-21, 2002 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-12054647

RESUMO

Programmed cell death 4 (PDCD4) has a common MI domain sharing with death associated protein 5 (DAP5) and a component of eukaryotic translation initiation factor (eIF4G) complex and it might also work as a tumor suppressor. We could find that the message and product of Pdcd4 gene were up-regulated in senescent human diploid fibroblasts. In yeast two hybrid analysis, the C-terminal region of PDCD4 interacted with ribosomal protein S13 (RPS13), ribosomal protein L5 (RPL5), and TI-227H. In in vitro binding assay, RPS13, a component of 40S ribosome was stably bound to PDCD4. We also found that PDCD4 was localized to polysome fractions. We could pull out eIF4G with GST-PDCD4, but eIF4E did not interact with PDCD4. From these results, we could assume that PDCD4 might regulate the eIF4G-dependent translation through direct interactions with eIF4G and RPS13 in senescent fibroblasts.


Assuntos
Senescência Celular/fisiologia , Fibroblastos/fisiologia , Regulação da Expressão Gênica/fisiologia , Proteínas/genética , Proteínas de Ligação a RNA , Animais , Apoptose , Proteínas Reguladoras de Apoptose , Células Cultivadas , Cicloeximida/farmacologia , Fator de Iniciação Eucariótico 4G , Células HeLa , Humanos , Recém-Nascido , Masculino , Fatores de Iniciação de Peptídeos/metabolismo , Polirribossomos/metabolismo , Proteínas/metabolismo , Coelhos , Proteínas Recombinantes/metabolismo , Proteínas Ribossômicas/metabolismo , Fenômenos Fisiológicos da Pele
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA