Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cancer Lett ; 543: 215791, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35700821

RESUMEN

Multiple myeloma (MM) is a hematologic malignancy derived from clonal expansion of plasma cells within the bone marrow and it may progress to the extramedullary region in late stage of the disease course. c-Maf, an oncogenic zipper leucine transcription factor, is overexpressed in more than 50% MM cell lines and primary species in association with chromosomal translocation, aberrant signaling transduction and modulation of stability. By triggering the transcription of critical genes including CCND2, ITGB7, CCR1, ARK5, c-Maf promotes MM progress, proliferation, survival and chemoresistance. Notably, c-Maf is usually expressed at the embryonic stage to promote cell differentiation but less expressed in healthy adult cells. c-Maf has long been proposed as a promising therapeutic target of MM and a panel of small molecule compounds have been identified to downregulate c-Maf and display potent anti-myeloma activities. In the current article, we take a concise summary on the advances in c-Maf biology, pathophysiology, and targeted drug discovery in the potential treatment of MM.


Asunto(s)
Mieloma Múltiple , Médula Ósea/patología , Carcinogénesis/metabolismo , Humanos , Factor de Transcripción MafF/metabolismo , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/genética , Mieloma Múltiple/metabolismo , Células Plasmáticas
2.
BMC Cardiovasc Disord ; 21(1): 448, 2021 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-34535081

RESUMEN

BACKGROUND: Vascular endothelial cell apoptosis is the leading risk factor of atherosclerosis (AS). The purpose of our study was to use a new generation high-throughput transcription factor (TF) detection method to identify novel key TFs in vascular endothelial cell apoptosis induced by palmitic acid (PA). METHODS: Human umbilical vein endothelial cells (HUVECs) were treated with 0, 300, or 500 µM PA. Candidate TFs in the three groups were identified by differential expression, pathway enrichment, Western Blot (WB), and RT-qPCR analyses. Apoptosis was assessed by fluorescence-activated cell sorting (FACS) using FITC-annexin V and propidium iodide staining. RESULTS: We established a HUVEC apoptosis model to simulate the process of atherosclerosis onset and identified 51 significant TFs. of the 51 TFs, v-maf musculoaponeurotic fibrosarcoma oncogene family protein G (MAFG) and v-maf musculoaponeurotic fibrosarcoma oncogene family protein F (MAFF), were matched to known AS signalling pathways and were validated by WB and RT-qPCR analyses in our study. Overexpression of MAFG or MAFF in HUVECs significantly inhibited PA-induced early apoptosis. CONCLUSIONS: We identified MAFF and MAFG as novel key TFs in vascular endothelial cell apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Aterosclerosis/metabolismo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Factor de Transcripción MafF/metabolismo , Factor de Transcripción MafG/metabolismo , Proteínas Nucleares/metabolismo , Ácido Palmítico/toxicidad , Proteoma , Proteómica , Proteínas Represoras/metabolismo , Aterosclerosis/genética , Aterosclerosis/patología , Células Cultivadas , Cromatografía Liquida , Regulación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Factor de Transcripción MafF/genética , Factor de Transcripción MafG/genética , Proteínas Nucleares/genética , Mapas de Interacción de Proteínas , Proteínas Represoras/genética , Transducción de Señal , Espectrometría de Masas en Tándem , Transcripción Genética
3.
Nat Commun ; 12(1): 4308, 2021 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-34262028

RESUMEN

Hypoxia plays a critical role in tumor progression including invasion and metastasis. To determine critical genes regulated by hypoxia that promote invasion and metastasis, we screen fifty hypoxia inducible genes for their effects on invasion. In this study, we identify v-maf musculoaponeurotic fibrosarcoma oncogene homolog F (MAFF) as a potent regulator of tumor invasion without affecting cell viability. MAFF expression is elevated in metastatic breast cancer patients and is specifically correlated with hypoxic tumors. Combined ChIP- and RNA-sequencing identifies IL11 as a direct transcriptional target of the heterodimer between MAFF and BACH1, which leads to activation of STAT3 signaling. Inhibition of IL11 results in similar levels of metastatic suppression as inhibition of MAFF. This study demonstrates the oncogenic role of MAFF as an activator of the IL11/STAT3 pathways in breast cancer.


Asunto(s)
Neoplasias de la Mama/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Interleucina-11/metabolismo , Factor de Transcripción MafF/metabolismo , Proteínas Nucleares/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/mortalidad , Hipoxia de la Célula , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Factor de Transcripción MafF/genética , Ratones , Invasividad Neoplásica/patología , Metástasis de la Neoplasia/patología , Proteínas Nucleares/genética , Pronóstico , Transducción de Señal , Transcripción Genética
4.
J Virol ; 95(15): e0076721, 2021 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-33980595

RESUMEN

Hepatitis B virus (HBV) is a stealth virus that exhibits only minimal induction of the interferon system, which is required for both innate and adaptive immune responses. However, 90% of acutely infected adults can clear the virus, suggesting the presence of additional mechanisms that facilitate viral clearance. Here, we report that Maf bZIP transcription factor F (MafF) promotes host defense against infection with HBV. Using a small interfering RNA (siRNA) library and an HBV/NanoLuc (NL) reporter virus, we screened to identify anti-HBV host factors. Our data showed that silencing of MafF led to a 6-fold increase in luciferase activity after HBV/NL infection. Overexpression of MafF reduced HBV core promoter transcriptional activity, which was relieved upon mutation of the putative MafF binding region. Loss of MafF expression through CRISPR/Cas9 editing (in HepG2-hNTCP-C4 cells) or siRNA silencing (in primary hepatocytes [PXB cells]) induced HBV core RNA and HBV pregenomic RNA (pgRNA) levels, respectively, after HBV infection. MafF physically binds to the HBV core promoter and competitively inhibits HNF-4α binding to an overlapping sequence in the HBV enhancer II sequence (EnhII), as seen by chromatin immunoprecipitation (ChIP) analysis. MafF expression was induced by interleukin-1ß (IL-1ß) or tumor necrosis factor alpha (TNF-α) treatment in both HepG2 and PXB cells, in an NF-κB-dependent manner. Consistently, MafF expression levels were significantly enhanced and positively correlated with the levels of these cytokines in patients with chronic HBV infection, especially in the immune clearance phase. IMPORTANCE HBV is a leading cause of chronic liver diseases, infecting about 250 million people worldwide. HBV has developed strategies to escape interferon-dependent innate immune responses. Therefore, the identification of other anti-HBV mechanisms is important for understanding HBV pathogenesis and developing anti-HBV strategies. MafF was shown to suppress transcription from the HBV core promoter, leading to significant suppression of the HBV life cycle. Furthermore, MafF expression was induced in chronic HBV patients and in primary human hepatocytes (PXB cells). This induction correlated with the levels of inflammatory cytokines (IL-1ß and TNF-α). These data suggest that the induction of MafF contributes to the host's antiviral defense by suppressing transcription from selected viral promoters. Our data shed light on a novel role for MafF as an anti-HBV host restriction factor.


Asunto(s)
Hepatitis B Crónica/patología , Inmunidad Innata/inmunología , Factor de Transcripción MafF/metabolismo , Proteínas Nucleares/metabolismo , Transcripción Genética/genética , Sistemas CRISPR-Cas/genética , Línea Celular Tumoral , Células Hep G2 , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/fisiología , Humanos , Interleucina-1beta/inmunología , Factor de Transcripción MafF/genética , Proteínas Nucleares/genética , Regiones Promotoras Genéticas/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Factor de Necrosis Tumoral alfa/inmunología
5.
Circulation ; 143(18): 1809-1823, 2021 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-33626882

RESUMEN

BACKGROUND: Coronary artery disease (CAD) is a multifactorial condition with both genetic and exogenous causes. The contribution of tissue-specific functional networks to the development of atherosclerosis remains largely unclear. The aim of this study was to identify and characterize central regulators and networks leading to atherosclerosis. METHODS: Based on several hundred genes known to affect atherosclerosis risk in mouse (as demonstrated in knockout models) and human (as shown by genome-wide association studies), liver gene regulatory networks were modeled. The hierarchical order and regulatory directions of genes within the network were based on Bayesian prediction models, as well as experimental studies including chromatin immunoprecipitation DNA-sequencing, chromatin immunoprecipitation mass spectrometry, overexpression, small interfering RNA knockdown in mouse and human liver cells, and knockout mouse experiments. Bioinformatics and correlation analyses were used to clarify associations between central genes and CAD phenotypes in both human and mouse. RESULTS: The transcription factor MAFF (MAF basic leucine zipper transcription factor F) interacted as a key driver of a liver network with 3 human genes at CAD genome-wide association studies loci and 11 atherosclerotic murine genes. Most importantly, expression levels of the low-density lipoprotein receptor (LDLR) gene correlated with MAFF in 600 CAD patients undergoing bypass surgery (STARNET [Stockholm-Tartu Atherosclerosis Reverse Network Engineering Task]) and a hybrid mouse diversity panel involving 105 different inbred mouse strains. Molecular mechanisms of MAFF were tested in noninflammatory conditions and showed positive correlation between MAFF and LDLR in vitro and in vivo. Interestingly, after lipopolysaccharide stimulation (inflammatory conditions), an inverse correlation between MAFF and LDLR in vitro and in vivo was observed. Chromatin immunoprecipitation mass spectrometry revealed that the human CAD genome-wide association studies candidate BACH1 (BTB domain and CNC homolog 1) assists MAFF in the presence of lipopolysaccharide stimulation with respective heterodimers binding at the MAF recognition element of the LDLR promoter to transcriptionally downregulate LDLR expression. CONCLUSIONS: The transcription factor MAFF was identified as a novel central regulator of an atherosclerosis/CAD-relevant liver network. MAFF triggered context-specific expression of LDLR and other genes known to affect CAD risk. Our results suggest that MAFF is a missing link between inflammation, lipid and lipoprotein metabolism, and a possible treatment target.


Asunto(s)
Aterosclerosis/metabolismo , Colesterol/metabolismo , Proteínas de Unión al ADN/metabolismo , Inflamación/metabolismo , Factor de Transcripción MafF/metabolismo , Proteínas Nucleares/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Ratones Noqueados
6.
Oncol Res ; 28(3): 299-309, 2020 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-31969212

RESUMEN

MafF is a member of the basic leucine zipper (bZIP) transcription factor Maf family and is commonly downregulated in multiple cancers. But the expression and function of MafF in hepatocellular carcinoma (HCC) remain unclear. In this study, we investigated the relationship between endogenous MafF expression and HCC progression and explored the regulatory mechanism of MafF expression in HCC. We found that MafF decreased in HCC tissues and cells. Lentivirus-mediated MafF overexpression inhibited HCC cell proliferation and induced cell apoptosis. Bioinformatics analysis and luciferase assay identified MafF as a direct target of miR-224-5p. RNA pull-down assay demonstrated that circular RNA circ-ITCH could sponge miR-224-5p specifically in HCC. The rescue experiments further elucidated that the expression and antitumor effects of MafF could be regulated via the circ-ITCH/miR-224-5p axis. This study verified that MafF acted as a tumor suppressor in HCC and revealed the upstream regulation mechanism of MafF, which provided a new perspective for potential therapeutic targets of HCC.


Asunto(s)
Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Factor de Transcripción MafF/metabolismo , MicroARNs/genética , Proteínas Nucleares/metabolismo , ARN Circular , Proteínas Represoras/genética , Ubiquitina-Proteína Ligasas/genética , Apoptosis/genética , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Hepáticas/patología , Interferencia de ARN
7.
Dis Markers ; 2020: 8861358, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33488846

RESUMEN

The small musculoaponeurotic fibrosarcoma (sMaf) proteins MafF, MafG, and MafK are basic region leucine zipper- (bZIP-) type transcription factors and display tissue- or stimulus-specific expression patterns. As the oxidative stress reactive proteins, sMafs are implicated in various neurological disorders. In the present study, the expressions of sMafs were investigated across five databases gathering transcriptomic data from 74 Alzheimer's disease (AD) patients and 66 controls in the Gene Expression Omnibus (GEO) database. The expression of MafF was increased in the hippocampus of AD patients, which was negatively correlated with the expression of the glutamate cysteine ligase catalytic subunit (GCLC). Furthermore, MafF was significantly increased in patients with Braak stage V-VI, compared to those with Braak stage III-IV. ß-Amyloid (Aß), a strong inducer of oxidative stress, plays a crucial role in the pathogenesis of AD. The responsive expressions of sMafs to Aß-induced oxidative stress were studied in the APP/PS1 mouse model of AD, Aß intrahippocampal injection rats, and several human cell lines from different tissue origins. This study revealed that only the induction of MafF was accompanied with reduction of GCLC and glutathione (GSH). MafF knockdown suppressed the increase of GSH induced by Aß. Among sMafs, MafF is the most responsive to Aß-induced oxidative stress and might potentiate the inhibition of antioxidation. These results provide a better understanding of sMaf modulation in AD and highlight MafF as a potential therapeutic target in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Factor de Transcripción MafF/metabolismo , Proteínas Nucleares/metabolismo , Células A549 , Anciano , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/toxicidad , Animales , Células Hep G2 , Hipocampo/metabolismo , Humanos , Factor de Transcripción MafF/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Nucleares/genética , Estrés Oxidativo , Ratas , Ratas Sprague-Dawley
8.
J Cell Mol Med ; 23(4): 2517-2525, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30669188

RESUMEN

Cytokines play key roles in a variety of reproductive processes including normal parturition as well as preterm birth. Our previous data have shown that MAFF, a member of the MAF family of bZIP transcription factors, is rapidly induced by pro-inflammatory cytokines in PHM1-31 myometrial cells. We performed loss-of-function studies in PHM1-31 cells to identify MAFF dependent genes. We showed that knockdown of MAFF significantly decreased CXCL1 chemokine and CSF3 cytokine transcript and protein levels. Using chromatin immunoprecipitation analyzes, we confirmed CXCL1 and CSF3 genes as direct MAFF targets. We also demonstrated that MAFF function in PHM1-31 myometrial cells is able to control cytokine and matrix metalloproteinase gene expression in THP-1 monocytic cells in a paracrine fashion. Our studies provide valuable insights into the MAFF dependent transcriptional network governing myometrial cell function. The data suggest a role of MAFF in parturition and/or infection-induced preterm labour through modulation of inflammatory processes in the microenvironment.


Asunto(s)
Quimiocina CXCL1/genética , Factor Estimulante de Colonias de Granulocitos/genética , Factor de Transcripción MafF/genética , Metaloproteinasas de la Matriz/genética , Miocitos del Músculo Liso/metabolismo , Miometrio/metabolismo , Proteínas Nucleares/genética , Línea Celular , Quimiocina CXCL1/metabolismo , Femenino , Regulación de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos/metabolismo , Humanos , Factor de Transcripción MafF/antagonistas & inhibidores , Factor de Transcripción MafF/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Miocitos del Músculo Liso/citología , Miometrio/citología , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/metabolismo , Comunicación Paracrina , Embarazo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Células THP-1 , Transcripción Genética
9.
J Exp Med ; 215(8): 2019-2034, 2018 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-30042191

RESUMEN

Immunoglobulin A (IgA) promotes health by regulating the composition and function of gut microbiota, but the molecular requirements for such homeostatic IgA function remain unknown. We found that a heavily glycosylated monoclonal IgA recognizing ovalbumin coats Bacteroides thetaiotaomicron (B. theta), a prominent gut symbiont of the phylum Bacteroidetes. In vivo, IgA alters the expression of polysaccharide utilization loci (PUL), including a functionally uncharacterized molecular family provisionally named Mucus-Associated Functional Factor (MAFF). In both mice and humans, MAFF is detected predominantly in mucus-resident bacteria, and its expression requires the presence of complex microbiota. Expression of the MAFF system facilitates symbiosis with other members of the phylum Firmicutes and promotes protection from a chemically induced model of colitis. Our data reveal a novel mechanism by which IgA promotes symbiosis and colonic homeostasis.


Asunto(s)
Bacterias/metabolismo , Microbioma Gastrointestinal , Inmunoglobulina A/metabolismo , Simbiosis , Adulto , Anciano , Anciano de 80 o más Años , Animales , Anticuerpos Monoclonales/metabolismo , Bacterias/genética , Bacteroides/genética , Bacteroides/fisiología , Colon/metabolismo , Proteínas de Unión al ADN , Femenino , Regulación Bacteriana de la Expresión Génica , Glicosilación , Homeostasis , Humanos , Lipopolisacáridos/metabolismo , Factor de Transcripción MafF/metabolismo , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Modelos Biológicos , Moco/metabolismo , Proteínas Nucleares/metabolismo , Ovalbúmina/metabolismo , Fenotipo
10.
Int J Mol Sci ; 19(5)2018 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-29757260

RESUMEN

Retinoids exert antitumor effects through the retinoic acid receptor α (RARα). In the present study, we sought to identify the factors involved in the RARα-mediated transcriptional regulation of the tumor suppressor gene and the tissue factor pathway inhibitor 2 (TFPI2) in hepatocellular carcinoma (HCC). All-trans-retinoic acid (ATRA) was used in the in vitro experiments. Cell invasiveness was measured using trans-well invasion assay. ATRA significantly increased TFPI2 expression through RARα in a human HCC cell line known as HuH7. TFPI2 was vital in the ATRA-mediated suppression of HuH7 cell invasion. The musculo-aponeurotic fibrosarcoma oncogene homolog B (MAFB) significantly enhanced the activation of the TFPI2 promoter via RARα while MAFF inhibited it. The knockdown of RARα or MAFB counteracted the ATRA-mediated suppression of HuH7 cell invasion while the knockdown of MAFF inhibited the invasion. TFPI2 expression in HCC tissues was significantly downregulated possibly due to the decreased expression of RARβ and MAFB. Patients with HCC expressing low MAFB and high MAFF levels showed the shortest disease-free survival time. These results suggest that MAFB and MAFF play critical roles in the antitumor effects of retinoids by regulating the expression of retinoid target genes such as TFPI2 and can be promising for developing therapies to combat HCC invasion.


Asunto(s)
Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Factor de Transcripción MafB/genética , Factor de Transcripción MafF/genética , Proteínas Nucleares/genética , Retinoides/metabolismo , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/mortalidad , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Movimiento Celular/genética , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Estimación de Kaplan-Meier , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/mortalidad , Neoplasias Hepáticas/patología , Factor de Transcripción MafB/metabolismo , Factor de Transcripción MafF/metabolismo , Proteínas Nucleares/metabolismo , Pronóstico , Regiones Promotoras Genéticas , Receptor alfa de Ácido Retinoico/metabolismo , Retinoides/farmacología , Transcripción Genética , Células Tumorales Cultivadas
11.
BMC Syst Biol ; 11(Suppl 5): 91, 2017 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-28984203

RESUMEN

BACKGROUND: Molecular mechanisms of the functional alteration of hematopoietic stem cells (HSCs) in leukemic environment attract intensive research interests. As known in previous researches, Maff and Egr3 are two important genes having opposite functions on cell cycle; however, they are both highly expressed in HSCs under leukemia. Hence, exploring the molecular mechanisms of how the genes act on cell cycle will help revealing the functional alteration of HSCs. RESULTS: We herein utilize the bioinformatic resources to computationally model the acting mechanisms of Maff and Egr3 on cell cycle. Using the data of functional experiments as reference, molecular acting mechanisms are optimally enumerated through model selection. The results are consolidated by evidences from gene sequence analysis, thus having enhanced the confidence of our pilot findings, which suggest that HSCs possibly undergo a "adaptation - suppression" process in response to the malignant environment of leukemia. CONCLUSION: As a pilot research, our results may provide valuable insights for further experimental studies. Meanwhile, our research method combining computational modeling and data from functional experiments can be worthwhile for knowledge discovery; and it can be generalized and extended to other biological/biomedical studies.


Asunto(s)
Proteína 3 de la Respuesta de Crecimiento Precoz/metabolismo , Células Madre Hematopoyéticas/patología , Leucemia/metabolismo , Leucemia/patología , Factor de Transcripción MafF/metabolismo , Modelos Biológicos , Regulación Neoplásica de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Humanos , Leucemia/genética , Biología de Sistemas
12.
J Alzheimers Dis ; 56(4): 1525-1539, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28222515

RESUMEN

Many lines of evidence suggest that Parkinson's disease (PD) and Alzheimer's disease (AD) have common characteristics, such as mitochondrial dysfunction and oxidative stress. As the underlying molecular mechanisms are unclear, we perform a meta-analysis with 9 microarray datasets of PD studies and 7 of AD studies to explore it. Functional enrichment analysis revealed that PD and AD both showed dysfunction in the synaptic vesicle cycle, GABAergic synapses, phagosomes, oxidative phosphorylation, and TCA cycle pathways, and AD had more enriched genes. Comparing the differentially expressed genes between AD and PD, we identified 54 common genes shared by more than six tissues. Among them, 31 downregulated genes contained the antioxidant response element (ARE) consensus sequence bound by NRF2. NRF2 is a transcription factor, which protects cells against oxidative stress through coordinated upregulation of ARE-driven genes. To our surprise, although NRF2 was upregulated, its target genes were all downregulated. Further exploration found that MAFF was upregulated in all tissues and significantly negatively correlated with the 31 NRF2-dependent genes in diseased conditions. Previous studies have demonstrated over-expressed small MAFs can form homodimers and act as transcriptional repressors. Therefore, MAFF might play an important role in dysfunction of NRF2 regulatory network in PD and AD.


Asunto(s)
Enfermedad de Alzheimer/genética , Factor 2 Relacionado con NF-E2/genética , Enfermedad de Parkinson/genética , Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Humanos , Factor de Transcripción MafF/genética , Factor de Transcripción MafF/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Enfermedad de Parkinson/metabolismo , Transducción de Señal
13.
Oncogene ; 34(19): 2493-504, 2015 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-24998848

RESUMEN

The TP53 tumor-suppressor gene is frequently mutated in human cancer. Missense mutations can add novel functions (gain-of-function, GOF) that promote tumor malignancy. Here we report that mutant (mut) p53 promotes tumor malignancy by suppressing the expression of a natural occurring anti-inflammatory cytokine, the secreted interleukin-1 receptor antagonist (sIL-1Ra, IL1RN). We show that mutp53 but not wild-type (wt) p53 suppresses the sIL-1Ra production in conditioned media of cancer cells. Moreover, mutp53, but not wtp53, binds physically the sIL-1Ra promoter and the protein-protein interaction with the transcriptional co-repressor MAFF (v-MAF musculoaponeurotic fibrosarcoma oncogene family, protein F) is required for mutp53-induced sIL-1Ra suppression. Remarkably, when exposed to IL-1 beta (IL-1ß) inflammatory stimuli, mutp53 sustains a ready-to-be-activated in vitro and in vivo cancer cells' response through the sIL-1Ra repression. Taken together, these results identify sIL-1Ra as a novel mutp53 target gene, whose suppression might be required to generate a chronic pro-inflammatory tumor microenvironment through which mutp53 promotes tumor malignancy.


Asunto(s)
Proteínas de Unión al ADN/genética , Inflamación/genética , Proteína Antagonista del Receptor de Interleucina 1/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/genética , Línea Celular Tumoral , Células HT29 , Células Hep G2 , Humanos , Inflamación/inmunología , Proteína Antagonista del Receptor de Interleucina 1/biosíntesis , Proteína Antagonista del Receptor de Interleucina 1/genética , Interleucina-1beta/farmacología , Células MCF-7 , Factor de Transcripción MafF/metabolismo , Mutación , Neoplasias/genética , Neoplasias/mortalidad , Proteínas Nucleares/metabolismo , Pronóstico , Regiones Promotoras Genéticas/genética , Unión Proteica , Interferencia de ARN , ARN Interferente Pequeño , Microambiente Tumoral/inmunología
14.
Cutan Ocul Toxicol ; 29(3): 171-92, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20491607

RESUMEN

Allergic contact dermatitis (ACD) is a significant safety concern for developers of cosmetic, personal care, chemical, pharmaceutical, and medical device products. The guinea pig maximization test (GMPT) and the murine local lymph node assay (LLNA) are accepted methods for determining chemical sensitization. Recent legislative initiatives in Europe require the development of new in vitro alternatives to animal tests for chemical sensitization. The aim of this project was to develop an in vitro screening method that uses a human skin cell line (HaCaT), chemical reactivity, and gene expression profiling to identify positive and negative responses, to place chemicals into potency categories of extreme/strong (ES), moderate (M), weak (W), and nonsensitizers (N), and to provide an estimate of corresponding LLNA values. The method and processing algorithm were developed from a training set of 39 chemicals possessing a wide range of sensitization potencies. Three cationic metals, chromium (Cr), nickel (Ni), and silver (Ag), were also evaluated in this model. Chemical reactivity was determined by measuring glutathione (GSH) depletion in a cell free matrix. Three signaling pathways (Keap1/Nrf 2/ARE/EpRE, ARNT/AhR/XRE, and Nrf1/MTF/MRE) that are known to be activated by sensitizing agents were monitored by measuring the relative abundance of 11 genes whose expression is controlled by one of these 3 pathways. Final exposure concentrations were based on toxicity and solubility. A range-finding experiment was conducted with each compound to determine cytotoxicity and solubility. Six exposure concentrations (0.1 to 2,500 microM) and an exposure time of 24 hours were used in the final experiments. Glutathione depletion alone did not provide the accuracy necessary to differentiate potency categories. However, chemical reactivity combined with gene expression profiles significantly improved the in vitro predictions. A predicted toxicity index (PTI) was determined for each test chemical. A comparison of LLNA values with PTI values revealed an inverse relationship. The large variation in LLNA data for compounds in the same potency category makes direct extrapolation from PTI to LLNA difficult. To challenge the system, 58 additional compounds were submitted in a blinded manner. Compounds placed into ES and M categories were considered positive, whereas compounds classified as W or N were considered negative. Accuracy was approximately 84%, with a sensitivity of 81% and a specificity of 92%. The model correctly identified 2 of 3 cationic metals as positive. In conclusion, the method described here demonstrates a valuable in vitro method for identifying chemicals and metals that induce skin sensitization.


Asunto(s)
Dermatitis Alérgica por Contacto/etiología , Expresión Génica/efectos de los fármacos , Queratinocitos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Pruebas de Toxicidad/métodos , Animales , Línea Celular , Citocromo P-450 CYP1A1/metabolismo , Glutatión/química , Haptenos/toxicidad , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch , Queratinocitos/metabolismo , Ensayo del Nódulo Linfático Local , Factor de Transcripción MafF/metabolismo , Proteínas Nucleares/metabolismo , Elementos de Respuesta
15.
J Microbiol Methods ; 79(1): 96-100, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19723544

RESUMEN

The human gene MafF (hMafF) is a member of bZip transcription factor Maf family, but it alone cannot activate its target genes. In 2006, a novel hMafF interacting protein (MIP) was identified. Transient transfection assay in Hela cells suggested that co-expression of MIP and hMafF could activate US2-driven transcription. In this work, we constructed a series of plasmids and transformed YM4271 yeast strain to establish a recombinant yeast detection system. In this system, MIP's expression level could be regulated using glucose incubation or galactose-induced incubation. The expression level of reporter gene LacZ in obtained recombinant yeast strains was measured using quantitative liquid assay. By comparing and analyzing the beta-galactosidase activities of different yeast strains or the same yeast strain in different culture media, the effect of MIP on transactivation driven by nUS2-hMafF was finally determined. Only in the presence of both MIP and hMafF could the nUS2-pLacZi reporter in yeast genome be activated. More importantly, this work established a novel recombinant yeast detection system, which may serve as a powerful tool to study the regulatory mechanisms of transcription complex in the future.


Asunto(s)
Acuaporinas/metabolismo , Proteínas del Ojo/metabolismo , Factor de Transcripción MafF/metabolismo , Proteínas Nucleares/metabolismo , Mapeo de Interacción de Proteínas/métodos , Factores de Transcripción/metabolismo , Activación Transcripcional , Acuaporinas/genética , Fusión Artificial Génica , Proteínas del Ojo/genética , Genes Reporteros , Humanos , Factor de Transcripción MafF/genética , Proteínas Nucleares/genética , Saccharomyces cerevisiae/genética , Transcripción Genética , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo
16.
Arch Biochem Biophys ; 449(1-2): 87-93, 2006 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-16549056

RESUMEN

The human transcription factor MafF (hMafF) lacks a transactivation domain, it contains a heptad-leucine repeat motif (viz., Leu-zipper) that may mediate protein-protein interactions to regulate transcriptional activities. A protein with a coiled-coil domain encoded by a novel human gene (GenBank Accession No. AF289559) was found to interact with hMafF in vitro and in vivo and is designated as a MafF interacting protein (MIP). Here, we provide evidence that the coiled-coil domain of MIP is essential for binding to the Leu-zipper of hMafF and that the interaction between MIP and hMafF causes the translocalization of MIP from cytoplasm to nucleolus in HELA cells. We used a promoter-reporter system containing six tandem repeats of the US2 element, located in the promoter of the human oxytocin receptor gene and reported to bind specifically to hMafF, to understand effects on transcriptional activation of hMafF, and its interaction with MIP. Expression of hMafF or MIP alone did not alter basal reporter transcription activity, whereas co-expression of hMafF and MIP activated transcription efficiently. Moreover, truncated MIP, still containing the coiled-coil domain, transactivated as well as the full-length MIP did, and highlighting that MIP acts as a co-activator of hMafF.


Asunto(s)
Acuaporinas/química , Acuaporinas/metabolismo , Proteínas del Ojo/química , Proteínas del Ojo/metabolismo , Factor de Transcripción MafF/química , Factor de Transcripción MafF/metabolismo , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Activación Transcripcional/fisiología , Secuencia de Aminoácidos , Sitios de Unión , Regulación de la Expresión Génica/fisiología , Células HeLa , Humanos , Datos de Secuencia Molecular , Unión Proteica
17.
Antioxid Redox Signal ; 8(1-2): 53-9, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16487037

RESUMEN

Exposure to inorganic arsenic has been associated with various forms of cancer, nervous system pathogenesis, and vascular diseases, as well as reproductive and developmental toxicity. Here, the effect of inorganic arsenic on placental JAR choriocarcinoma cells was assessed. The nuclear protein levels of the CNC transcription factor Nrf2 were strongly induced in the presence of arsenic. Dosage response experiments showed that 0.5 microM of arsenic is sufficient to augment Nrf2 levels. The expression of the Nrf2 dimerization partners MafG and MafK appeared not to be modulated by arsenic, whereas MafF protein levels were slightly increased. Arsenic also induced the binding of endogenous Nrf2/small Maf DNA-binding complexes to a stress response element (StRE) recognition site. In addition, arsenic caused oxidative stress in the choriocarcinoma cell model as evidenced by an increase in intracellular H2O2 levels. Expression of the enzyme heme oxygenase-1 (HO-1), a known Nrf2 target gene, was upregulated by exposure of JAR cells to arsenic. These results suggest that Nrf2/small Maf heterodimers may play an important role in the response to arsenic-mediated stress in placental cells.


Asunto(s)
Arsénico/toxicidad , Coriocarcinoma/fisiopatología , Factor 2 Relacionado con NF-E2/biosíntesis , Estrés Oxidativo/efectos de los fármacos , Placenta/fisiopatología , Proteínas Proto-Oncogénicas c-maf/metabolismo , Neoplasias Uterinas/fisiopatología , Línea Celular , Línea Celular Tumoral , Cartilla de ADN , Dimerización , Embrión de Mamíferos , Femenino , Humanos , Riñón , Cinética , Factor de Transcripción MafF/metabolismo , Factor 2 Relacionado con NF-E2/efectos de los fármacos , Proteínas Nucleares/metabolismo , Placenta/efectos de los fármacos , Plásmidos , Embarazo , Proteínas Proto-Oncogénicas c-maf/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
Biol Reprod ; 74(4): 699-705, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16371591

RESUMEN

The MAF (proto-)oncogene family of basic-leucine zipper transcription factors plays crucial roles in the control of mammalian gene expression and development. Here we analyzed the regulation of the human MAFF gene, coding for a small MAF transcription factor, in uterine smooth muscle cells. We found that MAFF transcript levels are induced by proinflammatory cytokines in PHM1-31 myometrial cells. We observed an important induction by interleukin 1 beta (IL1B) and a weaker upregulation by tumor necrosis factor (TNF), whereas interleukin 6 (IL6) treatment had no effect. Time course experiments revealed a rapid induction of MAFF transcripts within 30 min following IL1B treatment. The presence of actinomycin D inhibited the upregulation, suggesting that regulation of MAFF mRNA levels occurs at the transcriptional level. We generated a MAFF-specific antiserum and determined that MAFF protein was also induced by TNF and IL1B in PHM1-31 cells. In contrast, it was particularly interesting that the transcript and protein levels of the highly homologous MAFG and MAFK genes are not modulated by these cytokines. Our results suggest a possible specific role for MAFF in proinflammatory cytokine-mediated control of myometrial gene expression and provide the first link between a small MAF transcription factor and the inflammatory response.


Asunto(s)
Citocinas/fisiología , Factor de Transcripción MafF/metabolismo , Miometrio/metabolismo , Células Cultivadas , Femenino , Humanos , Interleucina-1/fisiología , Interleucina-6/fisiología , Factores de Transcripción Maf Pequeños/metabolismo , Proto-Oncogenes Mas , Transcripción Genética , Factores de Necrosis Tumoral/fisiología , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...