Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancers (Basel) ; 14(9)2022 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-35565443

RESUMO

CXCR4 is a G-Protein coupled receptor that is expressed nearly ubiquitously and is known to control cell migration via its interaction with CXCL12, the most ancient chemokine. The functions of CXCR4/CXCL12 extend beyond cell migration and involve the recognition and disposal of unhealthy or tumor cells. The CXCR4/CXCL12 axis plays a relevant role in shaping the tumor microenvironment (TME), mainly towards dampening immune responses. Notably, CXCR4/CXCL12 cross-signal via the T and B cell receptors (TCR and BCR) and co-internalize with CD47, promoting tumor cell phagocytosis by macrophages in an anti-tumor immune process called ImmunoGenic Surrender (IGS). These specific activities in shaping the immune response might be exploited to improve current immunotherapies.

2.
Eur Radiol Exp ; 6(1): 7, 2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-35132475

RESUMO

BACKGROUND: Malignant mesothelioma (MM) is an aggressive tumor, with a poor prognosis, usually unresectable due to late diagnosis, mainly treated with chemotherapy. BoxA, a truncated form of "high mobility group box 1" (HMGB1), acting as an HMGB1 antagonist, might exert a defensive action against MM. We investigated the potential of BoxA for MM treatment using experimental 40-MHz ultrasound and optical imaging (OI) in a murine model. METHODS: Murine MM cells infected with a lentiviral vector expressing the luciferase gene were injected into the peritoneum of 14 BALB/c mice (7 × 104 AB1-B/c-LUC cells). These mice were randomized to treatment with BoxA (n = 7) or phosphate-buffered saline (controls, n = 7). The experiment was repeated with 40 mice divided into two groups (n = 20 + 20) and treated as above to confirm the result and achieve greater statistical power. Tumor presence was investigated by experimental ultrasound and OI; suspected peritoneal masses underwent histopathology and immunohistochemistry examination. RESULTS: In the first experiment, none of the 7 controls survived beyond day 27, whereas 4/7 BoxA-treated mice (57.1%) survived up to day 70. In the second experiment, 6/20 controls (30.0%) and 16/20 BoxA-treated mice (80.0%) were still alive at day 34 (p = 0.004). In both experiments, histology confirmed the malignant nature of masses detected using experimental ultrasound and OI. CONCLUSION: In our preclinical experience on a murine model, BoxA seems to exert a protective role toward MM. Both experimental ultrasound and OI proved to be reliable techniques for detecting MM peritoneal masses.


Assuntos
Proteína HMGB1 , Mesotelioma Maligno , Animais , Modelos Animais de Doenças , Camundongos , Imagem Óptica , Ultrassonografia
4.
EMBO Mol Med ; 13(6): e12344, 2021 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-33956406

RESUMO

Boosting antitumor immunity has emerged as a powerful strategy in cancer treatment. While releasing T-cell brakes has received most attention, tumor recognition by T cells is a pre-requisite. Radiotherapy and certain cytotoxic drugs induce the release of damage-associated molecular patterns, which promote tumor antigen cross-presentation and T-cell priming. Antibodies against the "do not eat me" signal CD47 cause macrophage phagocytosis of live tumor cells and drive the emergence of antitumor T cells. Here we show that CXCR4 activation, so far associated only with tumor progression and metastasis, also flags tumor cells to immune recognition. Both CXCL12, the natural CXCR4 ligand, and BoxA, a fragment of HMGB1, promote the release of DAMPs and the internalization of CD47, leading to protective antitumor immunity. We designate as Immunogenic Surrender the process by which CXCR4 turns in tumor cells to macrophages, thereby subjecting a rapidly growing tissue to immunological scrutiny. Importantly, while CXCL12 promotes tumor cell proliferation, BoxA reduces it, and might be exploited for the treatment of malignant mesothelioma and a variety of other tumors.


Assuntos
Antígeno CD47 , Mesotelioma , Animais , Linhagem Celular Tumoral , Imunização , Macrófagos , Mesotelioma/imunologia , Mesotelioma/metabolismo , Mesotelioma/terapia , Camundongos , Fagocitose
5.
Immunol Rev ; 280(1): 74-82, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-29027228

RESUMO

A single protein, HMGB1, directs the triggering of inflammation, innate and adaptive immune responses, and tissue healing after damage. HMGB1 is the best characterized damage-associated molecular pattern (DAMP), proteins that are normally inside the cell but are released after cell death, and allow the immune system to distinguish between antigens that are dangerous or not. Notably, cells undergoing severe stress actively secrete HMGB1 via a dedicated secretion pathway: HMGB1 is relocated from the nucleus to the cytoplasm and then to secretory lysosomes or directly to the extracellular space. Extracellular HMGB1 (either released or secreted) triggers inflammation and adaptive immunological responses by switching among multiple oxidation states, which direct the mutually exclusive choices of different binding partners and receptors. Immune cells are first recruited to the damaged tissue and then activated; thereafter, HMGB1 supports tissue repair and healing, by coordinating the switch of macrophages to a tissue-healing phenotype, activation and proliferation of stem cells, and neoangiogenesis. Inevitably, HMGB1 also orchestrates the support of stressed but illegitimate tissues: tumors. Concomitantly, HMGB1 enhances the immunogenicity of mutated proteins in the tumor (neoantigens), promoting anti-tumor responses and immunological memory. Tweaking the activities of HMGB1 in inflammation, immune responses and tissue repair could bring large rewards in the therapy of multiple medical conditions, including cancer.


Assuntos
Imunidade Adaptativa , Morte Celular , Dano ao DNA/imunologia , Proteína HMGB1/imunologia , Imunidade Inata , Inflamação/imunologia , Animais , Humanos , Oxirredução , Cicatrização
6.
Sci Rep ; 6: 22850, 2016 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-26961782

RESUMO

Malignant Mesothelioma is a highly aggressive cancer, which is difficult to diagnose and treat. Here we describe the molecular, cellular and morphological characterization of a syngeneic system consisting of murine AB1, AB12 and AB22 mesothelioma cells injected in immunocompetent BALB/c mice, which allows the study of the interplay of tumor cells with the immune system. Murine mesothelioma cells, like human ones, respond to exogenous High Mobility Group Box 1 protein, a Damage-Associated Molecular Pattern that acts as a chemoattractant for leukocytes and as a proinflammatory mediator. The tumors derived from AB cells are morphologically and histologically similar to human MM tumors, and respond to treatments used for MM patients. Our system largely recapitulates human mesothelioma, and we advocate its use for the study of MM development and treatment.


Assuntos
Neoplasias Pulmonares , Mesotelioma , Animais , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Cisplatino/uso terapêutico , Desoxicitidina/análogos & derivados , Desoxicitidina/uso terapêutico , Feminino , Proteína HMGB1/metabolismo , Humanos , Imunocompetência , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Mesotelioma/irrigação sanguínea , Mesotelioma/tratamento farmacológico , Mesotelioma/imunologia , Mesotelioma/patologia , Mesotelioma Maligno , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Pemetrexede/uso terapêutico , Análise de Sobrevida , Gencitabina
7.
Nucleic Acids Res ; 38(11): 3595-604, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20110257

RESUMO

PREP1 (PKNOX1) maps in the Down syndrome (DS) critical region of chromosome 21, is overexpressed in some DS tissues and might be involved in the DS phenotype. By using fibroblasts from DS patients and by overexpressing Prep1 in F9 teratocarcinoma and Prep1(i/i) MEF to single out the role of the protein, we report that excess Prep1 increases the sensitivity of cells to genotoxic stress and the extent of the apoptosis directly correlates with the level of Prep1. The apoptotic response of Prep1-overexpressing cells is mediated by the pro-apoptotic p53 protein that we show is a direct target of Prep1, as its depletion reverts the apoptotic phenotype. The induction of p53 overcomes the anti-apoptotic role of Bcl-X(L), previously shown to be also a Prep1 target, the levels of which are increased in Prep1-overexpressing cells as well. Our results provide a rationale for the involvement of PREP1 in the apoptotic phenotype of DS tissues and indicate that differences in Prep1 level can have drastic effects.


Assuntos
Apoptose , Síndrome de Down/metabolismo , Proteínas de Homeodomínio/metabolismo , Animais , Células Cultivadas , Síndrome de Down/patologia , Células-Tronco de Carcinoma Embrionário , Etoposídeo/toxicidade , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/efeitos da radiação , Vetores Genéticos , Proteínas de Homeodomínio/genética , Humanos , Camundongos , Fenótipo , Transcrição Gênica , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Raios Ultravioleta , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
8.
PLoS Biol ; 8(1): e1000270, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20052287

RESUMO

The position of genes in the interphase nucleus and their association with functional landmarks correlate with active and/or silent states of expression. Gene activation can induce chromatin looping from chromosome territories (CTs) and is thought to require de novo association with transcription factories. We identify two types of factory: "poised transcription factories," containing RNA polymerase II phosphorylated on Ser5, but not Ser2, residues, which differ from "active factories" associated with phosphorylation on both residues. Using the urokinase-type plasminogen activator (uPA) gene as a model system, we find that this inducible gene is predominantly associated with poised (S5p(+)S2p(-)) factories prior to activation and localized at the CT interior. Shortly after induction, the uPA locus is found associated with active (S5p(+)S2p(+)) factories and loops out from its CT. However, the levels of gene association with poised or active transcription factories, before and after activation, are independent of locus positioning relative to its CT. RNA-FISH analyses show that, after activation, the uPA gene is transcribed with the same frequency at each CT position. Unexpectedly, prior to activation, the uPA loci internal to the CT are seldom transcriptionally active, while the smaller number of uPA loci found outside their CT are transcribed as frequently as after induction. The association of inducible genes with poised transcription factories prior to activation is likely to contribute to the rapid and robust induction of gene expression in response to external stimuli, whereas gene positioning at the CT interior may be important to reinforce silencing mechanisms prior to induction.


Assuntos
Inativação Gênica/fisiologia , Ativação Transcricional/fisiologia , Ativador de Plasminogênio Tipo Uroquinase/genética , Anticorpos/imunologia , Montagem e Desmontagem da Cromatina/fisiologia , Efeitos da Posição Cromossômica/genética , Efeitos da Posição Cromossômica/fisiologia , Regulação Enzimológica da Expressão Gênica/genética , Regulação Enzimológica da Expressão Gênica/fisiologia , Genes/genética , Loci Gênicos/genética , Loci Gênicos/fisiologia , Células Hep G2 , Humanos , Metaloendopeptidases/fisiologia , RNA Polimerase II/fisiologia , Ativação Transcricional/genética , Ativador de Plasminogênio Tipo Uroquinase/imunologia
9.
Mol Biol Cell ; 20(15): 3543-51, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19477923

RESUMO

We have analyzed the role of actin polymerization in retinoic acid (RA)-induced HoxB transcription, which is mediated by the HoxB regulator Prep1. RA induction of the HoxB genes can be prevented by the inhibition of actin polymerization. Importantly, inhibition of actin polymerization specifically affects the transcription of inducible Hox genes, but not that of their transcriptional regulators, the RARs, nor of constitutively expressed, nor of actively transcribed Hox genes. RA treatment induces the recruitment to the HoxB2 gene enhancer of a complex composed of "elongating" RNAPII, Prep1, beta-actin, and N-WASP as well as the accessory splicing components p54Nrb and PSF. We show that inhibition of actin polymerization prevents such recruitment. We conclude that inducible Hox genes are selectively sensitive to the inhibition of actin polymerization and that actin polymerization is required for the assembly of a transcription complex on the regulatory region of the Hox genes.


Assuntos
Actinas/metabolismo , Proteínas de Homeodomínio/genética , Fatores de Transcrição/genética , Transcrição Gênica/efeitos dos fármacos , Tretinoína/farmacologia , Actinas/genética , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Citocalasina D/farmacologia , Proteínas de Ligação a DNA , Regulação para Baixo , Proteínas de Homeodomínio/metabolismo , Humanos , Immunoblotting , Mutação , Proteínas Associadas à Matriz Nuclear/genética , Proteínas Associadas à Matriz Nuclear/metabolismo , Inibidores da Síntese de Ácido Nucleico/farmacologia , Fatores de Transcrição de Octâmero/genética , Fatores de Transcrição de Octâmero/metabolismo , Fator de Processamento Associado a PTB , Polímeros/metabolismo , Interferência de RNA , RNA Polimerase II/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Receptores do Ácido Retinoico/genética , Receptores do Ácido Retinoico/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Neuronal da Síndrome de Wiskott-Aldrich/genética , Proteína Neuronal da Síndrome de Wiskott-Aldrich/metabolismo
10.
Mol Cell Biol ; 29(5): 1143-51, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19103748

RESUMO

The Prep1 homeodomain transcription factor is essential in embryonic development. Prep1 hypomorphic mutant mouse (Prep1(i/i)) embryos (embryonic day 9.5) display an increased terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end labeling reaction compared to wild-type (WT) littermates. Prep1(i/i) mouse embryo fibroblasts (MEFs) show an increased basal level of annexin V binding activity, reduction of the mitochondrial-membrane potential, and increased caspase 9 and 3 activation, indicating increased apoptosis. Prep1(i/i) MEFs also respond faster than WT MEFs to genotoxic stress, indicating increased activation of the intrinsic apoptotic pathways. We did not observe an increase in p53 or an abnormal p53 response to apoptotic stimuli. However, hypomorphic MEFs have decreased endogenous levels of antiapoptotic Bcl-X(L) mRNA and protein, and Bcl-x overexpression rescues the defect of Prep1(i/i) MEFs. Using transient transfections and chromatin immunoprecipitation, we identified the Bcl-x promoter as a novel target of Prep1. Thus, Prep1 directly controls mitochondrial homeostasis (and the apoptotic potential) by modulating Bcl-x gene expression.


Assuntos
Apoptose , Regulação da Expressão Gênica , Proteínas de Homeodomínio/fisiologia , Proteína bcl-X/genética , Animais , Células Cultivadas , Embrião de Mamíferos , Fibroblastos/citologia , Camundongos , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Regiões Promotoras Genéticas , Transdução de Sinais
11.
J Biol Chem ; 282(17): 12537-46, 2007 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-17331942

RESUMO

We show the interaction between the enhancer and the minimal promoter of urokinase-type plasminogen activator gene during active transcription by coupling micrococcal nuclease digestion of cross-linked, sonicated chromatin, and chromatin immunoprecipitation. This approach allowed the precise identification of the interacting genomic fragments, one of which is resistant to micrococcal nuclease cleavage. The interacting fragments form a single transcriptional control unit, as indicated by their common protein content. Furthermore, we show that the enhancer-MP interaction persists during the early stages of transcription and is lost upon alpha-amanitin treatment, indicating the requirement for active transcription. Our results support a looping model of interaction between the enhancer and the MP of the urokinase-type plasminogen activator gene.


Assuntos
Elementos Facilitadores Genéticos , Modelos Genéticos , Regiões Promotoras Genéticas , Transcrição Gênica , Ativador de Plasminogênio Tipo Uroquinase/biossíntese , Animais , Cromatina/química , Cromatina/genética , Cromatina/metabolismo , Imunoprecipitação da Cromatina , Nuclease do Micrococo/química , Ratos , Ativador de Plasminogênio Tipo Uroquinase/genética
12.
Int J Biochem Cell Biol ; 39(4): 690-4, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17118695

RESUMO

Urokinase-type plasminogen activator (uPA) is a serine protease involved in tissue remodeling and cell migration. At the gene level, the interplay between a complex enhancer, required for induced and basal transcription, and the minimal promoter finely tunes uPA expression. The active form of uPA is bound to its high affinity receptor on the cell surface, where specific inhibitors modulate its enzymatic activity. Such inhibitors also regulate the cell surface levels of uPA by triggering the internalization of the uPA-receptor-inhibitor complex. The role of uPA is not only linked to its action as an enzyme. In fact, the mere binding of uPA on the cell surface also brings about two events that broaden the spectrum of its biological functions: (1) a conformational change of the receptor, which, in turn, affects its interaction with other proteins; (2) a signal transduction which modulates the expression of apoptosis-related genes. Besides its applications as a thrombolytic agent and as a prognostic marker for tumors, uPA may provide the basis for other therapies, as the structure of the receptor-binding domain of uPA has become a model for the design of anti-cancer molecules.


Assuntos
Plasminogênio/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Animais , Humanos , Modelos Biológicos , Ligação Proteica , Conformação Proteica , Receptores de Superfície Celular/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase , Sequências Reguladoras de Ácido Nucleico , Ativador de Plasminogênio Tipo Uroquinase/química , Ativador de Plasminogênio Tipo Uroquinase/genética
13.
Mol Cell ; 23(5): 749-55, 2006 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-16949370

RESUMO

Myosin VI is the only myosin that moves toward the minus end of actin filaments, suggesting a unique biological function. Here, we show that myosin VI is present in the nucleus of mammalian cells where it colocalizes with newly transcribed mRNA and with RNA polymerase II (RNAPII) and is detected in the RNAPII complex. The colocalization and interaction of myosin VI with RNAPII require transcriptional activity. Chromatin immunoprecipitation (ChIP) demonstrates that myosin VI is recruited to the promoter and intragenic regions of active genes, encoding urokinase plasminogen activator (uPA), eukaryotic initiation factor 6 (p27/eIF6), and low-density lipoprotein receptor (LDLR), but not to noncoding, nonregulatory intergenic regions. Downregulation of myosin VI reduces steady-state mRNA levels of these genes in vivo, and antibodies to myosin VI reduce transcription in vitro. We suggest that myosin VI modulates RNAPII-dependent transcription of active genes, implicating the possibility of an actin-myosin based mechanism of transcription.


Assuntos
Núcleo Celular/metabolismo , Cadeias Pesadas de Miosina/metabolismo , RNA Polimerase II/metabolismo , Transcrição Gênica , Células Cultivadas , Células HT29 , Células HeLa , Humanos , Regiões Promotoras Genéticas/genética , Ligação Proteica , Transporte Proteico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
14.
J Immunol ; 176(2): 999-1006, 2006 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-16393986

RESUMO

Pertussis toxin B-oligomer (PTX-B) inhibits HIV replication in T lymphocytes and monocyte-derived macrophages by interfering with multiple steps of the HIV life cycle. PTX-B prevents CCR5-dependent (R5) virus entry in a noncompetitive manner, and it also exerts suppressive effects on both R5- and CXCR4-dependent HIV expression at a less-characterized postentry level. We demonstrate in this study that PTX-B profoundly inhibits HIV expression in chronically infected promonocytic U1 cells stimulated with several cytokines and, particularly, the IL-6-mediated effect, a cytokine that triggers viral production in these cells independently of NF-kappaB activation. From U1 cells we have subcloned a cell line, named U1-CR1, with increased responsiveness to IL-6. In these cells, PTX-B neither down-regulated the IL-6R nor prevented IL-6 induced signaling in terms of STAT3 phosphorylation and DNA binding. In contrast, PTX-B inhibited AP-1 binding to target DNA and modified its composition with a proportional increases in FosB, Fra2, and ATF2. PTX-B inhibited IL-6-induced HIV-1 long-terminal repeat-driven transcription from A, C, E, and F viral subtypes, which contain functional AP-1 binding sites, but failed to inhibit transcription from subtypes B and D LTR devoid of these sites. In addition, PTX-B inhibited the secretion of IL-6-induced, AP-1-dependent genes, including urokinase-type plasminogen activator, CXCL8/IL-8, and CCL2/monocyte chemotactic protein-1. Thus, PTX-B suppression of IL-6 induced expression of HIV and cellular genes in chronically infected promonocytic cells is strongly correlated to inhibition of AP-1.


Assuntos
Quimiocinas/biossíntese , HIV-1/efeitos dos fármacos , Interleucina-6/farmacologia , Toxina Pertussis/farmacologia , Fator de Transcrição AP-1/antagonistas & inibidores , Sequência de Bases , Sítios de Ligação/genética , Linhagem Celular , Células Clonais , DNA/genética , DNA/metabolismo , Expressão Gênica/efeitos dos fármacos , Repetição Terminal Longa de HIV/efeitos dos fármacos , HIV-1/genética , HIV-1/fisiologia , Humanos , Toxina Pertussis/química , Receptores de Interleucina-6/metabolismo , Fator de Transcrição STAT3/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/genética , Replicação Viral/efeitos dos fármacos
15.
Blood ; 104(1): 256-62, 2004 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-15031204

RESUMO

Two upstream regions of the human urokinase (uPA) gene regulate its transcription: the minimal promoter (MP) and the enhancer element. The activity of the minimal promoter is essential for basal uPA transcription in prostate adenocarcinoma PC3 cells. Binding of a phosphorylated Sp1 transcription factor is, in turn, essential for the activity of the MP. Here we report that the Jun kinase (JNK) pathway is required for the basal activity of the MP and for the expression of the endogenous uPA gene in PC3 cells and for activated transcription in LNCaP cells. On the other hand, the p42/p44 mitogen-activated protein kinase (MAPK) pathway activates uPA gene expression through Sp1 phosphorylation in HeLa, LNCaP, and CCL39-derivative cells that do not typically express uPA in basal conditions. In HeLa cells the dominant-negative form of JNK interferes with the p42/p44 MAPK activation of the uPA-MP. The results suggest that the stress-activated protein kinase (SAPK)/JNK pathway plays an important role in the phosphorylation of Sp1, which, in turn, leads to basal or activated transcription from the uPA-MP element.


Assuntos
Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fator de Transcrição Sp1/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/genética , Animais , Butadienos/farmacologia , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Relação Dose-Resposta a Droga , Ativação Enzimática/genética , Inibidores Enzimáticos/farmacologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Genes Reporter/genética , Células HeLa , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno , Sistema de Sinalização das MAP Quinases , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/farmacologia , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas Quinases Ativadas por Mitógeno/farmacologia , Nitrilas/farmacologia , Fosforilação , Regiões Promotoras Genéticas/genética , RNA Mensageiro/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Ativação Transcricional/efeitos dos fármacos , Ativador de Plasminogênio Tipo Uroquinase/biossíntese
16.
Blood ; 100(9): 3325-32, 2002 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-12384434

RESUMO

Activated transcription of the urokinase-type plasminogen activator (uPA) gene depends on the enhancer, located approximately 2 kb from the start of transcription. The proximal promoter, driving basal transcription, contains a GC-/GA-rich sequence immediately upstream of the TATA box. We have investigated the role played by this element in the transcription of the uPA gene in HeLa and PC3 cells, which do not express or constitutively express the gene, respectively. This region binds either Sp1 or Sp3, as monomers or multimers, but not a combination of the 2 proteins. The more efficient binding of Sp1 to the proximal promoter in PC3 cells is correlated to its phosphorylation state. Polymerase chain reaction (PCR)-coupled, chromatin immunoprecipitation experiments with anti-Sp1 antibodies indeed show an enrichment of proximal promoter sequences in PC3 cells and support the observed difference in transcription levels from proximal promoter constructs in HeLa versus PC3 cells. Furthermore, overexpression of Sp1 increases transcription from the reporter construct in HeLa cells, whereas in PC3 cells, overexpression of Sp3 does not reduce transcription from the same construct, indicating that the Sp1/Sp3 balance cannot be shifted. We conclude that the GC-/GA-rich element of the uPA regulatory region is an independent functional element, regulated by Sp family proteins. Phosphorylation of Sp1 determines the presence in vivo and the functionality of this element in PC3 cells. Thus, the cellular context determines the relevance of the GC-/GA-rich region in uPA gene transcription, which contributes to constitutive gene expression, related, in turn, to the invasive phenotype.


Assuntos
Adenocarcinoma/patologia , Elementos Facilitadores Genéticos/genética , Regulação Neoplásica da Expressão Gênica/genética , Invasividade Neoplásica/genética , Proteínas de Neoplasias/genética , Regiões Promotoras Genéticas/genética , Neoplasias da Próstata/patologia , Fator de Transcrição Sp1/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/genética , Adenocarcinoma/genética , Sequência de Bases , Proteínas de Ligação a DNA/metabolismo , Desoxirribonuclease I/farmacologia , Genes Reporter , Células HeLa/metabolismo , Humanos , Luciferases/biossíntese , Luciferases/genética , Masculino , Dados de Sequência Molecular , Família Multigênica , Proteínas de Neoplasias/biossíntese , Fosforilação , Reação em Cadeia da Polimerase , Neoplasias da Próstata/genética , Ligação Proteica , Processamento de Proteína Pós-Traducional , Proteínas Recombinantes de Fusão/biossíntese , Fator de Transcrição Sp3 , Fatores de Transcrição/metabolismo , Células Tumorais Cultivadas/patologia , Ativador de Plasminogênio Tipo Uroquinase/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...