Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Br J Cancer ; 124(12): 1921-1933, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33785878

RESUMO

BACKGROUND: Dynamic transitions of tumour cells along the epithelial-mesenchymal axis are important in tumorigenesis, metastasis and therapy resistance. METHODS: In this study, we have used cell lines, 3D spheroids and tumour samples in a variety of cell biological and transcriptome analyses to highlight the cellular and molecular dynamics of OSCC response to ionising radiation. RESULTS: Our study demonstrates a prominent hybrid epithelial-mesenchymal state in oral squamous cell carcinoma cells and tumour samples. We have further identified a key role for levels of E-cadherin in stratifying the hybrid cells to compartments with varying levels of radiation response and radiation-induced epithelial-mesenchymal transition. The response to radiation further entailed the generation of a new cell population with low expression levels of E-cadherin, and positive for Vimentin (ECADLow/Neg-VIMPos), a phenotypic signature that showed an enhanced capacity for radiation resistance and invasion. At the molecular level, transcriptome analysis of spheroids in response to radiation showed an initial burst of misregulation within the first 30 min that further declined, although still highlighting key alterations in gene signatures. Among others, pathway analysis showed an over-representation for the Wnt signalling pathway that was further confirmed to be functionally involved in the generation of ECADLow/Neg-VIMPos population, acting upstream of radiation resistance and tumour cell invasion. CONCLUSION: This study highlights the functional significance and complexity of tumour cell remodelling in response to ionising radiation with links to resistance and invasive capacity. An area of less focus in conventional radiotherapy, with the potential to improve treatment outcomes and relapse-free survival.


Assuntos
Carcinoma de Células Escamosas/patologia , Transição Epitelial-Mesenquimal , Neoplasias Bucais/patologia , Tolerância a Radiação/genética , Carcinoma de Células Escamosas/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Transição Epitelial-Mesenquimal/genética , Transição Epitelial-Mesenquimal/efeitos da radiação , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Genes de Troca/fisiologia , Genes de Troca/efeitos da radiação , Estudos de Associação Genética , Humanos , Neoplasias Bucais/genética , Invasividade Neoplásica , Fenótipo , Radiação Ionizante , Transcriptoma/efeitos da radiação , Via de Sinalização Wnt/genética , Via de Sinalização Wnt/efeitos da radiação
2.
Cells ; 8(5)2019 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-31137701

RESUMO

Hippo signaling controls cellular processes that ultimately impact organogenesis and homeostasis. Consequently, disease states including cancer can emerge when signaling is deregulated. The major pathway transducers Yap and Taz require cofactors to impart transcriptional control over target genes. Research into Yap/Taz-mediated epigenetic modifications has revealed their association with chromatin-remodeling complex proteins as a means of altering chromatin structure, therefore affecting accessibility and activity of target genes. Specifically, Yap/Taz have been found to associate with factors of the GAGA, Ncoa6, Mediator, Switch/sucrose nonfermentable (SWI/SNF), and Nucleosome Remodeling and Deacetylase (NuRD) chromatin-remodeling complexes to alter the accessibility of target genes. This review highlights the different mechanisms by which Yap/Taz collaborate with other factors to modify DNA packing at specific loci to either activate or repress target gene transcription.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Montagem e Desmontagem da Cromatina/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Fatores de Transcrição/metabolismo , Animais , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Genes de Troca/fisiologia , Via de Sinalização Hippo , Histona Metiltransferases/metabolismo , Humanos , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/metabolismo , Proteínas Nucleares/metabolismo , Transdução de Sinais , Transativadores/metabolismo , Proteínas com Motivo de Ligação a PDZ com Coativador Transcricional , Proteínas de Sinalização YAP
3.
Cell Rep ; 27(1): 226-237.e4, 2019 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-30943404

RESUMO

The mechanisms by which regulatory T cells (Tregs) migrate to and function within the hypoxic tumor microenvironment are unclear. Our studies indicate that specific ablation of hypoxia-inducible factor 1α (HIF-1α) in Tregs results in enhanced CD8+ T cell suppression versus wild-type Tregs under hypoxia, due to increased pyruvate import into the mitochondria. Importantly, HIF-1α-deficient Tregs are minimally affected by the inhibition of lipid oxidation, a fuel that is critical for Treg metabolism in tumors. Under hypoxia, HIF-1α directs glucose away from mitochondria, leaving Tregs dependent on fatty acids for mitochondrial metabolism within the hypoxic tumor. Indeed, inhibition of lipid oxidation enhances the survival of mice with glioma. Interestingly, HIF-1α-deficient-Treg mice exhibit significantly enhanced animal survival in a murine model of glioma, due to their stymied migratory capacity, explaining their reduced abundance in tumor-bearing mice. Thus HIF-1α acts as a metabolic switch for Tregs between glycolytic-driven migration and oxidative phosphorylation-driven immunosuppression.


Assuntos
Neoplasias Encefálicas , Movimento Celular/genética , Metabolismo Energético/genética , Glioblastoma , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Linfócitos T Reguladores/imunologia , Evasão Tumoral , Idoso , Idoso de 80 Anos ou mais , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Hipóxia Celular/genética , Hipóxia Celular/fisiologia , Células Cultivadas , Feminino , Genes de Troca/fisiologia , Glioblastoma/genética , Glioblastoma/imunologia , Glioblastoma/metabolismo , Glioblastoma/patologia , Glicólise/genética , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Terapia de Imunossupressão , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fosforilação Oxidativa , Linfócitos T Reguladores/metabolismo , Evasão Tumoral/genética , Evasão Tumoral/imunologia , Microambiente Tumoral/genética
5.
J Exp Med ; 214(10): 2889-2900, 2017 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-28827448

RESUMO

Epithelial carcinomas are well known to activate a prolonged wound-healing program that promotes malignant transformation. Wound closure requires the activation of keratinocyte migration via a dual-state molecular switch. This switch involves production of either the anti-migratory microRNA miR-198 or the pro-migratory follistatin-like 1 (FSTL1) protein from a single transcript; miR-198 expression in healthy skin is down-regulated in favor of FSTL1 upon wounding, which enhances keratinocyte migration and promotes re-epithelialization. Here, we reveal a defective molecular switch in head and neck squamous cell carcinoma (HNSCC). This defect shuts off miR-198 expression in favor of sustained FSTL1 translation, driving metastasis through dual parallel pathways involving DIAPH1 and FSTL1. DIAPH1, a miR-198 target, enhances directional migration through sequestration of Arpin, a competitive inhibitor of Arp2/3 complex. FSTL1 blocks Wnt7a-mediated repression of extracellular signal-regulated kinase phosphorylation, enabling production of MMP9, which degrades the extracellular matrix and facilitates metastasis. The prognostic significance of the FSTL1-DIAPH1 gene pair makes it an attractive target for therapeutic intervention.


Assuntos
Transformação Celular Neoplásica/metabolismo , Fator de Crescimento Epidérmico/fisiologia , Proteínas Relacionadas à Folistatina/fisiologia , MicroRNAs/fisiologia , Cicatrização/fisiologia , Animais , Western Blotting , Carcinoma de Células Escamosas/metabolismo , Proliferação de Células , Feminino , Genes de Troca/fisiologia , Neoplasias de Cabeça e Pescoço/metabolismo , Imunoprecipitação , Espectrometria de Massas , Camundongos Endogâmicos NOD
7.
Genes Dev ; 29(4): 426-39, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25691469

RESUMO

The initiation of chromosome morphogenesis marks the beginning of mitosis in all eukaryotic cells. Although many effectors of chromatin compaction have been reported, the nature and design of the essential trigger for global chromosome assembly remain unknown. Here we reveal the identity of the core mechanism responsible for chromosome morphogenesis in early mitosis. We show that the unique sensitivity of the chromosome condensation machinery for the kinase activity of Cdk1 acts as a major driving force for the compaction of chromatin at mitotic entry. This sensitivity is imparted by multisite phosphorylation of a conserved chromatin-binding sensor, the Smc4 protein. The multisite phosphorylation of this sensor integrates the activation state of Cdk1 with the dynamic binding of the condensation machinery to chromatin. Abrogation of this event leads to chromosome segregation defects and lethality, while moderate reduction reveals the existence of a novel chromatin transition state specific to mitosis, the intertwist configuration. Collectively, our results identify the mechanistic basis governing chromosome morphogenesis in early mitosis and how distinct chromatin compaction states can be established via specific thresholds of Cdk1 kinase activity.


Assuntos
Divisão Celular/genética , Cromossomos Fúngicos/genética , Quinases Ciclina-Dependentes/metabolismo , Saccharomyces cerevisiae/citologia , Saccharomyces cerevisiae/genética , Adenosina Trifosfatases/química , Adenosina Trifosfatases/metabolismo , Montagem e Desmontagem da Cromatina/genética , Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Genes de Troca/fisiologia , Mitose , Complexos Multiproteicos/química , Complexos Multiproteicos/metabolismo , Fosforilação , Ligação Proteica , Proteínas de Saccharomyces cerevisiae/metabolismo
8.
Biochim Biophys Acta ; 1849(4): 427-35, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25132386

RESUMO

Cancer cells and stem cells share many traits, including a tendency towards genomic instability. Human cancers exhibit tumor-specific genomic aberrations, which often affect their malignancy and drug response. During their culture propagation, human pluripotent stem cells (hPSCs) also acquire characteristic genomic aberrations, which may have significant impact on their molecular and cellular phenotypes. These aberrations vary in size from single nucleotide alterations to copy number alterations to whole chromosome gains. A prominent challenge in both cancer and stem cell research is to identify "driver aberrations" that confer a selection advantage, and "driver genes" that underlie the recurrence of these aberrations. Following principles that are already well-established in cancer research, candidate driver genes have also been suggested in hPSCs. Experimental validation of the functional role of such candidates can uncover whether these are bona fide driver genes. The identification of driver genes may bring us closer to a mechanistic understanding of the genomic instability of stem cells. Guided by terminologies and methodologies commonly applied in cancer research, such understanding may have important ramifications for both stem cell and cancer biology. This article is part of a Special Issue entitled: Stress as a fundamental theme in cell plasticity.


Assuntos
Genes de Troca/fisiologia , Instabilidade Genômica , Neoplasias/genética , Células-Tronco Pluripotentes/fisiologia , Seleção Genética , Aneuploidia , Animais , Dosagem de Genes , Humanos , Neoplasias/patologia
9.
Nat Cell Biol ; 16(7): 673-84, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24929361

RESUMO

Signal transduction requires the coordination of activities between different pathways. In mammalian cells, Raf-1 regulates the MST-LATS and MEK-ERK pathways. We found that a complex circuitry of competing protein interactions coordinates the crosstalk between the ERK and MST pathways. Combining mathematical modelling and experimental validation we show that competing protein interactions can cause steep signalling switches through phosphorylation-induced changes in binding affinities. These include Akt phosphorylation of MST2 and a feedback phosphorylation of Raf-1 Ser 259 by LATS1, which enables Raf-1 to suppress both MST2 and MEK signalling. Mutation of Raf-1 Ser 259 stimulates both pathways, simultaneously driving apoptosis and proliferation, whereas concomitant MST2 downregulation switches signalling to cell proliferation, transformation and survival. Thus, competing protein interactions provide a versatile regulatory mechanism for signal distribution through the dynamic integration of graded signals into switch-like responses.


Assuntos
Genes de Troca/fisiologia , Modelos Biológicos , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-raf/metabolismo , Transdução de Sinais , Animais , Apoptose/fisiologia , Proliferação de Células , Células Cultivadas , Embrião não Mamífero , Células HEK293 , Células HeLa , Via de Sinalização Hippo , Humanos , Sistema de Sinalização das MAP Quinases/fisiologia , Células MCF-7 , Camundongos , Células NIH 3T3 , Fosforilação , Ligação Proteica , Serina-Treonina Quinase 3 , Peixe-Zebra/embriologia
10.
Development ; 140(6): 1207-19, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23444353

RESUMO

Hematopoiesis is a classic system with which to study developmental potentials and to investigate gene regulatory networks that control choices among alternate lineages. T-cell progenitors seeding the thymus retain several lineage potentials. The transcription factor PU.1 is involved in the decision to become a T cell or a myeloid cell, and the developmental outcome of expressing PU.1 is dependent on exposure to Notch signaling. PU.1-expressing T-cell progenitors without Notch signaling often adopt a myeloid program, whereas those exposed to Notch signals remain in a T-lineage pathway. Here, we show that Notch signaling does not alter PU.1 transcriptional activity by degradation/alteration of PU.1 protein. Instead, Notch signaling protects against the downregulation of T-cell factors so that a T-cell transcriptional network is maintained. Using an early T-cell line, we describe two branches of this network. The first involves inhibition of E-proteins by PU.1 and the resulting inhibition of Notch signaling target genes. Effects of E-protein inhibition can be reversed by exposure to Notch signaling. The second network is dependent on the ability of PU.1 to inhibit important T-cell transcription factor genes such as Myb, Tcf7 and Gata3 in the absence of Notch signaling. We show that maintenance of Gata3 protein levels by Myb and Notch signaling is linked to the ability to retain T-cell identity in response to PU.1.


Assuntos
Fator de Transcrição GATA3/fisiologia , Genes de Troca , Hematopoese/genética , Células Progenitoras Mieloides/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Receptores Notch/fisiologia , Transativadores/fisiologia , Animais , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Linhagem da Célula/genética , Células Cultivadas , Embrião de Mamíferos , Fator de Transcrição GATA3/genética , Fator de Transcrição GATA3/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Genes de Troca/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Biológicos , Células Progenitoras Mieloides/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-myb/genética , Proteínas Proto-Oncogênicas c-myb/metabolismo , Proteínas Proto-Oncogênicas c-myb/fisiologia , Receptores Notch/genética , Receptores Notch/metabolismo , Transdução de Sinais/genética , Linfócitos T/metabolismo , Linfócitos T/fisiologia , Timócitos/metabolismo , Timócitos/fisiologia , Transativadores/genética , Transativadores/metabolismo
11.
Blood ; 121(13): 2553-62, 2013 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-23361909

RESUMO

B-cell lymphoma 11A (BCL11A) downregulation in human primary adult erythroid progenitors results in elevated expression of fetal γ-globin. Recent reports showed that BCL11A expression is activated by KLF1, leading to γ-globin repression. To study regulation of erythropoiesis and globin expression by KLF1 and BCL11A in an in vivo model, we used mice carrying a human ß-globin locus transgene with combinations of Klf1 knockout, Bcl11a floxed, and EpoR(Cre) knockin alleles. We found a higher percentage of reticulocytes in adult Klf1(wt/ko) mice and a mild compensated anemia in Bcl11a(cko/cko) mice. These phenotypes were more pronounced in compound Klf1(wt/ko)::Bcl11a(cko/cko) mice. Analysis of Klf1(wt/ko), Bcl11a(cko/cko), and Klf1(wt/ko)::Bcl11a(cko/cko) mutant embryos demonstrated increased expression of mouse embryonic globins during fetal development. Expression of human γ-globin remained high in Bcl11a(cko/cko) embryos during fetal development, and this was further augmented in Klf1(wt/ko)::Bcl11a(cko/cko) embryos. After birth, expression of human γ-globin and mouse embryonic globins decreased in Bcl11a(cko/cko) and Klf1(wt/ko)::Bcl11a(cko/cko) mice, but the levels remained much higher than those observed in control animals. Collectively, our data support an important role for the KLF1-BCL11A axis in erythroid maturation and developmental regulation of globin expression.


Assuntos
Proteínas de Transporte/genética , Eritropoese/genética , Genes de Troca/genética , Globinas/genética , Fatores de Transcrição Kruppel-Like/genética , Proteínas Nucleares/genética , Animais , Proteínas de Ligação a DNA , Embrião de Mamíferos , Eritropoese/fisiologia , Desenvolvimento Fetal/genética , Regulação da Expressão Gênica no Desenvolvimento , Rearranjo Gênico/genética , Rearranjo Gênico/fisiologia , Genes de Troca/fisiologia , Humanos , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Proteínas Repressoras , Reticulocitose/genética , Reticulocitose/fisiologia , Baço/citologia , Baço/embriologia , Baço/metabolismo
12.
Development ; 140(4): 897-905, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23325762

RESUMO

Development in multicellular organisms includes both small incremental changes and major switches of cell differentiation and proliferation status. During Drosophila oogenesis, the follicular epithelial cells undergo two major developmental switches that cause global changes in the cell-cycle program. One, the switch from the endoreplication cycle to a gene-amplification phase, during which special genomic regions undergo repeated site-specific replication, is attributed to Notch downregulation, ecdysone signaling activation and upregulation of the zinc-finger protein Tramtrack69 (Ttk69). Here, we report that the microRNA miR-7 exerts an additional layer of regulation in this developmental switch by regulating Ttk69 transcripts. miR-7 recognizes the 3' UTR of ttk69 transcripts and regulates Ttk69 expression in a dose-dependent manner. Overexpression of miR-7 effectively blocks the switch from the endocycle to gene amplification through its regulation of ttk69. miR-7 and Ttk69 also coordinate other cell differentiation events, such as vitelline membrane protein expression, that lead to the formation of the mature egg. Our studies reveal the important role miR-7 plays in developmental decision-making in association with signal-transduction pathways.


Assuntos
Ciclo Celular/fisiologia , Proteínas de Drosophila/metabolismo , Drosophila/embriologia , Células Epiteliais/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Genes de Troca/fisiologia , MicroRNAs/metabolismo , Folículo Ovariano/citologia , Proteínas Repressoras/metabolismo , Animais , Animais Geneticamente Modificados , Bromodesoxiuridina , Primers do DNA/genética , Drosophila/metabolismo , Proteínas de Drosophila/fisiologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Genes de Troca/genética , Imuno-Histoquímica , MicroRNAs/genética , Mutagênese Sítio-Dirigida , Proteínas Repressoras/fisiologia
14.
Horm Cancer ; 2(5): 272-85, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21887591

RESUMO

Calcitriol or 1,25-dihydroxyvitamin D3, the hormonally active form of vitamin D, as well as vitamin D analogs, has been shown to increase sensitivity to ionizing radiation in breast tumor cells. The current studies indicate that the combination of 1,25-dihydroxyvitamin D3 with radiation appears to kill p53 wild-type, estrogen receptor-positive ZR-75-1 breast tumor cells through autophagy. Minimal apoptosis was observed based on cell morphology by DAPI and TUNEL staining, annexin/PI analysis, caspase-3, and PARP cleavage as well as cell cycle analysis. Induction of autophagy was indicated by increased acridine orange staining, RFP-LC3 redistribution, and detection of autophagic vesicles by electron microscopy, while autophagic flux was monitored based on p62 degradation. The autophagy inhibitors, chloroquine and bafilomycin A1, as well as genetic suppression of the autophagic signaling proteins Atg5 or Atg 7 attenuated the impact of the combination treatment of 1,25 D3 with radiation. In contrast to autophagy mediating the effects of the combination treatment, the autophagy induced by radiation alone was apparently cytoprotective in that either pharmacological or genetic inhibition increased sensitivity to radiation. These studies support the potential utility of vitamin D for improving the impact of radiation for breast cancer therapy, support the feasibility of combining chloroquine with radiation for the treatment of breast cancer, and demonstrate the existence of an "autophagic switch" from cytoprotective autophagy with radiation alone to cytotoxic autophagy with the 1,25 D3-radiation combination.


Assuntos
Autofagia/efeitos dos fármacos , Neoplasias da Mama/patologia , Carcinoma/patologia , Cloroquina/farmacologia , Citoproteção/efeitos dos fármacos , Vitamina D/farmacologia , Autofagia/genética , Proteína 5 Relacionada à Autofagia , Proteína 7 Relacionada à Autofagia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/radioterapia , Carcinoma/tratamento farmacológico , Carcinoma/genética , Carcinoma/radioterapia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Sobrevivência Celular/efeitos da radiação , Senescência Celular/efeitos dos fármacos , Senescência Celular/genética , Senescência Celular/efeitos da radiação , Citoproteção/genética , Citotoxinas/farmacologia , Estudos de Viabilidade , Feminino , Genes de Troca/efeitos dos fármacos , Genes de Troca/fisiologia , Humanos , Proteínas Associadas aos Microtúbulos/antagonistas & inibidores , Proteínas Associadas aos Microtúbulos/genética , RNA Interferente Pequeno/farmacologia , Tolerância a Radiação/efeitos dos fármacos , Tolerância a Radiação/genética , Radiossensibilizantes/farmacologia , Células Tumorais Cultivadas , Enzimas Ativadoras de Ubiquitina/antagonistas & inibidores , Enzimas Ativadoras de Ubiquitina/genética
15.
PLoS Pathog ; 6(3): e1000806, 2010 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-20300604

RESUMO

To mate, the fungal pathogen Candida albicans must undergo homozygosis at the mating-type locus and then switch from the white to opaque phenotype. Paradoxically, opaque cells were found to be unstable at physiological temperature, suggesting that mating had little chance of occurring in the host, the main niche of C. albicans. Recently, however, it was demonstrated that high levels of CO(2), equivalent to those found in the host gastrointestinal tract and select tissues, induced the white to opaque switch at physiological temperature, providing a possible resolution to the paradox. Here, we demonstrate that a second signal, N-acetylglucosamine (GlcNAc), a monosaccharide produced primarily by gastrointestinal tract bacteria, also serves as a potent inducer of white to opaque switching and functions primarily through the Ras1/cAMP pathway and phosphorylated Wor1, the gene product of the master switch locus. Our results therefore suggest that signals produced by bacterial co-members of the gastrointestinal tract microbiota regulate switching and therefore mating of C. albicans.


Assuntos
Acetilglucosamina/metabolismo , Candida albicans/crescimento & desenvolvimento , Candida albicans/metabolismo , Genes Fúngicos Tipo Acasalamento/fisiologia , Transdução de Sinais/fisiologia , Adenilil Ciclases/genética , Adenilil Ciclases/metabolismo , Candida albicans/genética , Dióxido de Carbono/metabolismo , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 2/genética , Nucleotídeo Cíclico Fosfodiesterase do Tipo 2/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Trato Gastrointestinal/microbiologia , Genes de Troca/fisiologia , Fosforilação/fisiologia , Temperatura , Proteínas ras/metabolismo
16.
Pharmacol Res ; 58(5-6): 262-70, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18838122

RESUMO

In response to oxidative stress, the transcription factor NF-E2-related factor 2 (Nrf2) controls the fate of cells through transcriptional upregulation of antioxidant response element (ARE)-bearing genes, including those encoding endogenous antioxidants, phase II detoxifying enzymes, and transporters. Expression of the Nrf2-dependent proteins is critical for ameliorating or eliminating toxicants/carcinogens to maintain cellular redox homeostasis. As a result, activation of the Nrf2 pathway, by naturally-occurring compounds or synthetic chemicals at sub-toxic doses, confers protection against subsequent toxic/carcinogenic exposure. Thus, the use of dietary compounds or synthetic chemicals to boost the Nrf2-dependent adaptive response to counteract environmental insults has emerged to be a promising strategy for cancer prevention. Interestingly, recent emerging data has revealed the "dark" side of Nrf2. Nrf2 and its downstream genes are overexpressed in many cancer cell lines and human cancer tissues, giving cancer cells an advantage for survival and growth. Furthermore, Nrf2 is upregulated in resistant cancer cells and is thought to be responsible for acquired chemoresistance. Therefore, it may be necessary to inhibit the Nrf2 pathway during chemotherapy. This review is primarily focused on the role of Nrf2 in cancer, with emphasis on the recent findings indicating the cancer promoting function of Nrf2 and its role in acquired chemoresistance.


Assuntos
Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/fisiologia , Neoplasias/genética , Animais , Biotransformação/fisiologia , Genes de Troca/genética , Genes de Troca/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Proteína 1 Associada a ECH Semelhante a Kelch , Neoplasias/patologia , Neoplasias/prevenção & controle
17.
Mutat Res ; 647(1-2): 59-67, 2008 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-18786551

RESUMO

Development is a stepwise process in which multi-potent progenitor cells undergo lineage commitment, differentiation, proliferation and maturation to produce mature cells with restricted developmental potentials. This process is directed by spatiotemporally distinct gene expression programs that allow cells to stringently orchestrate intricate transcriptional activation or silencing events. In eukaryotes, chromatin structure contributes to developmental progression as a blueprint for coordinated gene expression by actively participating in the regulation of gene expression. Changes in higher order chromatin structure or covalent modification of its components are considered to be critical events in dictating lineage-specific gene expression during development. Mammalian cells utilize multi-subunit nuclear complexes to alter chromatin structure. Histone-modifying complex catalyzes covalent modifications of histone tails including acetylation, methylation, phosphorylation and ubiquitination. ATP-dependent chromatin remodeling complex, which disrupts histone-DNA contacts and induces nucleosome mobilization, requires energy from ATP hydrolysis for its catalytic activity. Here, we discuss the diverse functions of ATP-dependent chromatin remodeling complexes during mammalian development. In particular, the roles of these complexes during embryonic and hematopoietic development are reviewed in depth. In addition, pathological conditions such as tumor development that are induced by mutation of several key subunits of the chromatin remodeling complex are discussed, together with possible mechanisms that underlie tumor suppression by the complex.


Assuntos
Montagem e Desmontagem da Cromatina , Desenvolvimento Embrionário/genética , Crescimento e Desenvolvimento/genética , Neoplasias/genética , Animais , Proteínas Cromossômicas não Histona/fisiologia , DNA Helicases/fisiologia , Regulação da Expressão Gênica , Genes de Troca/fisiologia , Hematopoese/genética , Humanos , Camundongos , Proteínas Nucleares/fisiologia , Linfócitos T/fisiologia , Fatores de Transcrição/fisiologia
18.
Cell Biochem Biophys ; 44(3): 342-8, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16679521

RESUMO

The tumor necrosis factor-alpha-converting enzyme (TACE) is a member of the disintegrin family of metalloproteinases (ADAMs) that plays a central role in the regulated shedding of a host of cell surface proteins. TACE is biosynthesized as a precursor protein with latent proteolytic activity (zymogen). TACE's zymogen inhibition is mediated by its Pro domain, a 197-amino acid region that serves this function as well as aiding in the secretion of this enzyme through the secretory pathway. We have discovered that a conserved "cysteine switch" consensus motif within TACE's Pro domain is, contrary to expectations, not required for maintenance of the inactive precursor state or for the secretion of this metalloproteinase in its functional form. The only role for this motif seems to be in decreasing TACE's susceptibility to proteolytic degradation during its biogenesis and maturation within the secretory pathway. Interestingly, the Pro domain of TACE seems to carry both its inhibitory and secretory functions through the same mechanism: it seems to prevent the Catalytic domain from accessing its native, functional state, resembling the function of true molecular chaperones. Recent evidence suggests that TACE may also be switched out of the active conformation even by small, drug-like molecules such as the synthetic compound SB-3CT. These findings point at the possibility of developing, in the near future, a new generation of antiinflammatory, noncompetitive TACE inhibitors that would exert negative allosteric modulation over the activity of this key enzyme, mediating several inflammatory diseases and certain cancers.


Assuntos
Proteínas ADAM/metabolismo , Cisteína/metabolismo , Precursores Enzimáticos/metabolismo , Genes de Troca/fisiologia , Proteínas ADAM/biossíntese , Proteínas ADAM/química , Proteínas ADAM/genética , Proteína ADAM17 , Domínio Catalítico , Sequência Conservada , Ativação Enzimática , Precursores Enzimáticos/fisiologia , Regulação da Expressão Gênica/fisiologia , Metaloendopeptidases/antagonistas & inibidores , Metaloendopeptidases/química , Metaloendopeptidases/metabolismo , Chaperonas Moleculares/química , Mutação/genética , Estrutura Terciária de Proteína/fisiologia , Proteínas Recombinantes , Fator de Necrose Tumoral alfa/metabolismo
19.
Toxicol Sci ; 88(1): 82-94, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16081525

RESUMO

Previous research from our laboratory has shown a switch-like response to PCB 126 mediated CYP1A1 induction in primary rat hepatocytes and in H4IIE rat hepatoma cells. On a single cell level, cells appear to be either "on" or "off" for CYP1A1 induction at a given dose; some cells never respond to PCB 126. These cells represent a non-responding population. Cells that are switched "on" by PCB 126 display varying levels of induction, much like the dimmer on a light switch. The goal of the present research is to begin to uncover the mechanism for this switch-like response to CYP1A1 induction in H4IIE rat hepatoma cells. The AhR pathway is modulated by multiple co-activators and by phosphorylation. This research focuses on the phosphorylation cascades initiated by PCB 126 and the role they play in CYP1A1 induction. Our research reveals a likely role for protein kinase C (PKC) in this switch response. Inhibition of PKC by H-7 dramatically reduced the percent of cells that express CYP1A1 in response to PCB 126 treatment, as determined by flow cytometry. The effect of H-7 was concentration dependent, decreasing the number of cells expressing CYP1A1 rather than decreasing the level of CYP1A1 in all cells. This finding provides further evidence for the switch-like behavior of CYP1A1 induction and implicates PKC in this response to PCB126. The protein kinase inhibitor, HA-1004, had only a minor effect on CYP1A1 induction. A high-throughput immunoblot screen for 40 proteins revealed the regulation of several proteins/phosphoproteins by PCB 126. Most importantly, two proteins containing phosphoserine/phoshothreonine residues were increased by PCB126 treatment. However, PKC translocation studies and activity studies failed to verify that PCB126 activates PKC. It is possible that constitutive PKC activity is sufficient to maintain phosphorylation of critical components of the AhR pathway. Immunoblotting studies showed that MAP kinases ERK and JNK are not activated by PCB 126 in H4IIE cells and the ERK inhibitor U0126 did not impair CYP1A1 induction. Additional studies are planned to further investigate the role of PKC in the switch-like response to PCB 126.


Assuntos
Carcinoma Hepatocelular/metabolismo , Citocromo P-450 CYP1A1/biossíntese , Genes de Troca/fisiologia , Hepatócitos/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteína Quinase C/metabolismo , Animais , Western Blotting , Contagem de Células , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Indução Enzimática , Antagonistas de Estrogênios/toxicidade , Citometria de Fluxo , Genes de Troca/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Fosforilação , Bifenilos Policlorados/toxicidade , Proteína Quinase C/antagonistas & inibidores , Inibidores de Proteínas Quinases/toxicidade , Ratos , Transdução de Sinais
20.
Curr Biol ; 15(9): 869-73, 2005 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-15886107

RESUMO

The Raf-MEK-ERK MAP kinase cascade transmits signals from activated receptors into the cell to regulate proliferation and differentiation. The cascade is controlled by the Ras GTPase, which recruits Raf from the cytosol to the plasma membrane for activation. In turn, MEK, ERK, and scaffold proteins translocate to the plasma membrane for activation. Here, we examine the input-output properties of the Raf-MEK-ERK MAP kinase module in mammalian cells activated in different cellular contexts. We show that the MAP kinase module operates as a molecular switch in vivo but that the input sensitivity of the module is determined by subcellular location. Signal output from the module is sensitive to low-level input only when it is activated at the plasma membrane. This is because the threshold for activation is low at the plasma membrane, whereas the threshold for activation is high in the cytosol. Thus, the circuit configuration of the module at the plasma membrane generates maximal outputs from low-level analog inputs, allowing cells to process and respond appropriately to physiological stimuli. These results reveal the engineering logic behind the recruitment of elements of the module from the cytosol to the membrane for activation.


Assuntos
Membrana Celular/metabolismo , Citosol/metabolismo , Ativação Enzimática/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Animais , Western Blotting , Células Cultivadas , Cricetinae , Genes de Troca/fisiologia , Microscopia de Fluorescência , Monoéster Fosfórico Hidrolases/metabolismo , Transfecção , Quinases raf/metabolismo , Proteínas ras/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA