Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 384
Filtrar
1.
Nature ; 618(7963): 169-179, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37225982

RESUMO

Target occupancy is often insufficient to elicit biological activity, particularly for RNA, compounded by the longstanding challenges surrounding the molecular recognition of RNA structures by small molecules. Here we studied molecular recognition patterns between a natural-product-inspired small-molecule collection and three-dimensionally folded RNA structures. Mapping these interaction landscapes across the human transcriptome defined structure-activity relationships. Although RNA-binding compounds that bind to functional sites were expected to elicit a biological response, most identified interactions were predicted to be biologically inert as they bind elsewhere. We reasoned that, for such cases, an alternative strategy to modulate RNA biology is to cleave the target through a ribonuclease-targeting chimera, where an RNA-binding molecule is appended to a heterocycle that binds to and locally activates RNase L1. Overlay of the substrate specificity for RNase L with the binding landscape of small molecules revealed many favourable candidate binders that might be bioactive when converted into degraders. We provide a proof of concept, designing selective degraders for the precursor to the disease-associated microRNA-155 (pre-miR-155), JUN mRNA and MYC mRNA. Thus, small-molecule RNA-targeted degradation can be leveraged to convert strong, yet inactive, binding interactions into potent and specific modulators of RNA function.


Assuntos
Endorribonucleases , MicroRNAs , RNA Mensageiro , Humanos , Genes jun/genética , Genes myc/genética , MicroRNAs/antagonistas & inibidores , MicroRNAs/química , MicroRNAs/genética , MicroRNAs/metabolismo , Conformação de Ácido Nucleico , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/química , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Relação Estrutura-Atividade , Especificidade por Substrato , Endorribonucleases/química , Endorribonucleases/metabolismo , Transcriptoma
2.
Environ Toxicol ; 37(9): 2291-2301, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35689653

RESUMO

Exposure to silica nanoparticles (SiNPs) is related to the dysregulation of pulmonary surfactant that maintains lung stability and function. Nevertheless, there are limited studies concerning the interaction and influence between SiNPs and pulmonary surfactant, and the damage and mechanism are still unclear. Herein, we used A549 cells to develop an in vitro model, with which we investigated the effect of SiNPs exposure on the expression of pulmonary surfactant and the potential regulatory mechanism. The results showed that SiNPs were of cytotoxicity in regarding of reduced cell viability and promoted the production of excessive reactive oxygen species (ROS). Additionally, the JNK/c-Jun signaling pathway was activated, and the expression of surfactant protein A (SP-A) and surfactant protein B (SP-B) was decreased. After the cells being treated with N-acetyl-L-cysteine (NAC), we found that the ROS content was effectively downregulated, and the expression of proteins related to JNK and c-Jun signaling pathways was suppressed. In contrast, the expression of SP-A and SP-B was enhanced. Furthermore, we treated the cells with JNK inhibitor and c-Jun-siRNA and found that the expression of protein related to JNK and c-Jun signaling pathways, as well as SP-A and SP-B, changed in line with that of NAC treatment. These findings suggest that SiNPs exposure can upregulate ROS and activate the JNK/c-Jun signaling pathway in A549 cells, thereby inhibiting the expression of SP-A and SP-B proteins.


Assuntos
Pulmão , Nanopartículas , Proteína A Associada a Surfactante Pulmonar , Proteína B Associada a Surfactante Pulmonar , Dióxido de Silício , Células A549 , Acetilcisteína/farmacologia , Apoptose , Genes jun/genética , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Pulmão/metabolismo , Nanopartículas/toxicidade , Proteína A Associada a Surfactante Pulmonar/metabolismo , Proteína B Associada a Surfactante Pulmonar/metabolismo , Surfactantes Pulmonares/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Dióxido de Silício/toxicidade
3.
Elife ; 112022 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-35076395

RESUMO

The class IIa histone deacetylases (HDACs) have pivotal roles in the development of different tissues. Of this family, Schwann cells express Hdac4, 5, and 7 but not Hdac9. Here, we show that a transcription factor regulated genetic compensatory mechanism within this family of proteins, blocks negative regulators of myelination ensuring peripheral nerve developmental myelination and remyelination after injury. Thus, when Hdac4 and 5 are knocked-out from Schwann cells in mice, a JUN-dependent mechanism induces the compensatory overexpression of Hdac7 permitting, although with a delay, the formation of the myelin sheath. When Hdac4, 5, and 7 are simultaneously removed, the myocyte-specific enhancer-factor d (MEF2D) binds to the promoter and induces the de novo expression of Hdac9, and although several melanocytic lineage genes are misexpressed and Remak bundle structure is disrupted, myelination proceeds after a long delay. Thus, our data unveil a finely tuned compensatory mechanism within the class IIa Hdac family, coordinated by distinct transcription factors, that guarantees the ability of Schwann cells to myelinate during development and remyelinate after nerve injury.


Assuntos
Regulação da Expressão Gênica/fisiologia , Genes jun/genética , Histona Desacetilases/genética , Nervos Periféricos/fisiologia , Remielinização , Células de Schwann/metabolismo , Animais , Feminino , Histona Desacetilases/metabolismo , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/metabolismo , Masculino , Camundongos
4.
FEBS J ; 289(3): 748-765, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34499807

RESUMO

Nucleoli are well defined for their function in ribosome biogenesis, but only a small fraction of the nucleolar proteome has been characterized. Here, we report that the proto-oncogene, c-Jun, is targeted to the nucleolus. Using live cell imaging in myogenic cells, we document that the c-Jun basic domain contains a unique, evolutionarily conserved motif that determines nucleolar targeting. Fos family Jun dimer partners, such as Fra2, while nuclear, do not co-localize with c-Jun in the nucleolus. A point mutation in c-Jun that mimics Fra2 (M260E) in its Nucleolar Localization sequence (NoLS) results in loss of c-Jun nucleolar targeting while still preserving nuclear localization. Fra2 can sequester c-Jun in the nucleoplasm, indicating that the stoichiometric ratio of heterodimeric partners regulates c-Jun nucleolar targeting. Finally, nucleolar localization of c-Jun modulates nucleolar architecture and ribosomal RNA accumulation. These studies highlight a novel role for Jun family proteins in the nucleolus, having potential implications for a diverse array of AP-1-regulated cellular processes.


Assuntos
Nucléolo Celular/genética , Antígeno 2 Relacionado a Fos/genética , Genes jun/genética , Ribossomos/genética , Sequência de Aminoácidos/genética , Linhagem Celular , Regulação da Expressão Gênica/genética , Humanos , Sinais de Localização Nuclear/genética , Proteínas Nucleares/genética , Transporte Proteico/genética , Proteoma/genética
5.
JCI Insight ; 6(13)2021 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-34236045

RESUMO

The AP-1 transcription factor c-Jun is required for Ras-driven tumorigenesis in many tissues and is considered as a classical proto-oncogene. To determine the requirement for c-Jun in a mouse model of K-RasG12D-induced lung adenocarcinoma, we inducibly deleted c-Jun in the adult lung. Surprisingly, we found that inactivation of c-Jun, or mutation of its JNK phosphorylation sites, actually increased lung tumor burden. Mechanistically, we found that protein levels of the Jun family member JunD were increased in the absence of c-Jun. In c-Jun-deficient cells, JunD phosphorylation was increased, and expression of a dominant-active JNKK2-JNK1 transgene further increased lung tumor formation. Strikingly, deletion of JunD completely abolished Ras-driven lung tumorigenesis. This work identifies JunD, not c-Jun, as the crucial substrate of JNK signaling and oncogene required for Ras-induced lung cancer.


Assuntos
Adenocarcinoma de Pulmão , Carcinogênese , Neoplasias Pulmonares , Proteínas Proto-Oncogênicas c-jun/metabolismo , Proteínas ras/metabolismo , Adenocarcinoma de Pulmão/genética , Adenocarcinoma de Pulmão/metabolismo , Animais , Carcinogênese/genética , Carcinogênese/metabolismo , Regulação Neoplásica da Expressão Gênica/genética , Inativação Gênica , Genes jun/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , MAP Quinase Quinase 7/genética , MAP Quinase Quinase 7/metabolismo , Sistema de Sinalização das MAP Quinases , Camundongos , Fosforilação , Proteínas Proto-Oncogênicas c-jun/genética , Fator de Transcrição AP-1/metabolismo
6.
Cell Death Dis ; 11(9): 811, 2020 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-32980857

RESUMO

Glaucoma is a neurodegenerative disease characterized by loss of retinal ganglion cells (RGCs), the output neurons of the retina. Multiple lines of evidence show the endothelin (EDN, also known as ET) system is important in glaucomatous neurodegeneration. To date, the molecular mechanisms within RGCs driving EDN-induced RGC death have not been clarified. The pro-apoptotic transcription factor JUN (the canonical target of JNK signaling) and the endoplasmic reticulum stress effector and transcription factor DNA damage inducible transcript 3 (DDIT3, also known as CHOP) have been shown to act downstream of EDN receptors. Previous studies demonstrated that JUN and DDIT3 were important regulators of RGC death after glaucoma-relevant injures. Here, we characterized EDN insult in vivo and investigated the role of JUN and DDIT3 in EDN-induced RGC death. To accomplish this, EDN1 ligand was intravitreally injected into the eyes of wildtype, Six3-cre+Junfl/fl (Jun-/-), Ddit3 null (Ddit3-/-), and Ddit3-/-Jun-/- mice. Intravitreal EDN1 was sufficient to drive RGC death in vivo. EDN1 insult caused JUN activation in RGCs, and deletion of Jun from the neural retina attenuated RGC death after EDN insult. However, deletion of Ddit3 did not confer significant protection to RGCs after EDN1 insult. These results indicate that EDN caused RGC death via a JUN-dependent mechanism. In addition, EDN signaling is known to elicit potent vasoconstriction. JUN signaling was shown to drive neuronal death after ischemic insult. Therefore, the effects of intravitreal EDN1 on retinal vessel diameter and hypoxia were explored. Intravitreal EDN1 caused transient retinal vasoconstriction and regions of RGC and Müller glia hypoxia. Thus, it remains a possibility that EDN elicits a hypoxic insult to RGCs, causing apoptosis via JNK-JUN signaling. The importance of EDN-induced vasoconstriction and hypoxia in causing RGC death after EDN insult and in models of glaucoma requires further investigation.


Assuntos
Endotelina-1/metabolismo , Genes jun/genética , Glaucoma/genética , Células Ganglionares da Retina/metabolismo , Animais , Morte Celular , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Endogâmicos C57BL
7.
Ann Clin Lab Sci ; 50(3): 354-360, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32581025

RESUMO

OBJECTIVE: Aim of this study is to assess effects of Evi-1 on regulation of c-Jun N-terminal kinase (JNK)/c-Jun pathway on colorectal cancer and the relationship of their expression with clinicopathological characteristics and prognosis. METHODS: The clinical data of 394 CRC patients and the mRNA expression of Evi-1 were downloaded from The Cancer Genome Atlas (TCGA). Immunohistochemical (IHC) method was used to detect the protein expression of Evi-1, JNK and c-Jun in CRC tissues and adjacent normal tissues. RESULTS: The TCGA dataset demonstrated that Evi-1 mRNA was upregulated in CRC. Evi-1 mRNA expression was significantly correlated with lymphatic metastasis, T stage, distant metastasis and TNM stage. IHC staining showed that Remmele immunoreactive Score (IRS) of Evi-1, JNK and c-Jun were highly expressed in CRC tissues compared with normal tissues adjacent to cancer. Evi-1 in CRC tissues was negatively correlated with JNK and c-Jun. Overexpression of Evi-1, JNK and c-Jun was significantly correlated with the degree of differentiation, T staging, lymphatic metastasis, distant metastasis and TNM classification. CONCLUSION: The present study shows that Evi-1 expression is negatively correlated with JNK and c-Jun expression, and closely related to some clinical data. Further, Evi-1 combined with JNK and c-Jun can be used for adverse prognosis of CRC.


Assuntos
Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Proteína do Locus do Complexo MDS1 e EVI1/metabolismo , Adulto , Idoso , Biomarcadores Tumorais/metabolismo , Diferenciação Celular/genética , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Neoplasias Colorretais/metabolismo , Bases de Dados Genéticas , Feminino , Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/genética , Genes jun/genética , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Metástase Linfática/genética , Proteína do Locus do Complexo MDS1 e EVI1/genética , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias/métodos , Prognóstico , Estudos Retrospectivos
8.
Cytokine ; 126: 154862, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31634687

RESUMO

Stimulation of H295R adrenocortical carcinoma cells with angiotensin II or cytokines induces the secretion of the chemokine interleukin-8 (IL-8). Here, we have analyzed the molecular mechanism of stimulus-induced IL-8 expression. IL-8 expression and IL-8 promoter activity increased in H295R cells expressing an activated Gαq-coupled designer receptor. H295R cells stimulated with either interleukin-1ß (IL-1ß) or phorbol ester also showed elevated IL-8 mRNA levels and higher IL-8 promoter activities. Deletion and point mutations of the IL-8 promoter revealed that the AP-1 binding site within the IL-8 promoter is essential to connect designer receptor stimulation with the transcriptional activation of the IL-8 gene. Expression of a constitutively active mutant of c-Jun, or expression of constitutively active mutants of the protein kinases MEKK1 and MKK6 confirmed that the IL-8 gene is a bona fide target of AP-1 in adrenocortical carcinoma cells. Upregulation of IL-8 expression in IL-1ß-treated H295R cells required NF-κB while the phorbol ester TPA used both the AP-1 and NF-κB sites of the IL-8 gene to stimulate IL-8 expression. These data were corroborated in experiments with chromatin-embedded AP-1 or NF-κB-responsive reporter genes. While stimulation of Gαq-coupled designer receptors increased the AP-1 activity in the cells, IL-1ß specifically stimulated NF-κB-regulated transcription. Stimulation of the cells with TPA increased both AP-1 and NF-κB activities. We conclude that stimulation of Gαq-coupled designer receptors or IL-1 receptors triggers distinct signaling pathways in H295R cells leading to the activation of either AP-1 or NF-κB. Nevertheless, both signaling cascades converge to the IL-8 gene, inducing IL-8 gene transcription.


Assuntos
Neoplasias do Córtex Suprarrenal/metabolismo , Carcinoma Adrenocortical/metabolismo , Interleucina-8/metabolismo , NF-kappa B/metabolismo , Fator de Transcrição AP-1/metabolismo , Neoplasias do Córtex Suprarrenal/genética , Carcinoma Adrenocortical/genética , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/genética , Genes jun/genética , Humanos , Interleucina-1beta/farmacologia , Interleucina-8/genética , MAP Quinase Quinase 6/genética , MAP Quinase Quinase 6/metabolismo , MAP Quinase Quinase Quinase 1/genética , MAP Quinase Quinase Quinase 1/metabolismo , Mutação Puntual , Regiões Promotoras Genéticas , Deleção de Sequência , Acetato de Tetradecanoilforbol/farmacologia , Ativação Transcricional/efeitos dos fármacos , Ativação Transcricional/genética , Regulação para Cima
9.
Gut ; 68(10): 1858-1871, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31118247

RESUMO

BACKGROUND AND AIMS: The unique expression pattern makes oncofetal proteins ideal diagnostic biomarkers and therapeutic targets in cancer. However, few oncofetal proteins have been identified and entered clinical practice. METHODS: Fetal liver, adult liver and hepatocellular carcinoma (HCC) tissues were employed to assess the expression of hepatic leukaemia factor (HLF). The impact of HLF on HCC onset and progression was investigated both in vivo and in vitro. The association between HLF and patient prognosis was determined in patient cohorts. The correlation between HLF expression and sorafenib benefits in HCC was further evaluated in patient cohorts and patient-derived xenografts (PDXs). RESULTS: HLF is a novel oncofetal protein which is reactivated in HCC by SOX2 and OCT4. Functional studies revealed that HLF transactivates c-Jun to promote tumour initiating cell (TIC) generation and enhances TIC-like properties of hepatoma cells, thus driving HCC initiation and progression. Consistently, our clinical investigations elucidated the association between HLF and patient prognosis and unravelled the close correlation between HLF levels and c-Jun expression in patient HCCs. Importantly, HLF/c-Jun axis determines the responses of hepatoma cells to sorafenib treatment, and interference of HLF abrogated c-Jun activation and enhanced sorafenib response. Analysis of patient cohorts and PDXs further suggests that HLF/c-Jun axis might serve as a biomarker for sorafenib benefits in HCC patients. CONCLUSIONS: Our findings uncovered HLF as a novel oncofetal protein and revealed the crucial role of the HLF/c-Jun axis in HCC development and sorafenib response, rendering HLF as an optimal target for the prevention and intervention of HCC.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/genética , Carcinoma Hepatocelular/genética , Resistencia a Medicamentos Antineoplásicos , Genes jun/genética , Neoplasias Hepáticas/genética , Sorafenibe/farmacologia , Adulto , Antineoplásicos/farmacologia , Apoptose , Fatores de Transcrição de Zíper de Leucina Básica/biossíntese , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , DNA de Neoplasias/genética , Progressão da Doença , Feminino , Humanos , Imunoprecipitação , Zíper de Leucina , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/patologia , Masculino , Prognóstico
10.
J Cell Physiol ; 234(5): 6688-6695, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30478830

RESUMO

Neuropathic pain (NP) may cause serious brain diseases, but the genes associated with the metabolic pathway and transcript factors of NP remain unclear. This study is aimed to identify the therapy target genes for NP and to investigate the metabolic pathways and transcript factors associated with NP. The differentially expressed genes of three brain tissues (nucleus accumbens, periaqueductal gray, and prefrontal cortex) dealt with NP stimulation were analyzed. Besides, The Database for Annotation, Visualization, and Integrated Discovery and Tfacts datasets were used in the analysis of the genes related to the metabolic pathway and transcript factors of the brain. Eight genes were found to coexpress in all three tissues. A functional enrichment analysis showed that the upregulated genes were mostly enriched in pathways as inflammatory response, calcium-mediated signaling, cytokine-cytokine receptor interaction, and extracellular matrix (ECM)-receptor interaction, whereas the downregulated genes were mostly enriched in pathways as phospholipid metabolic processes, positive regulation of protein kinase B signaling, and metabolism of xenobiotics by cytochrome P450. Finally, 135 and 98 transcript factors genes were upregulated and downregulated, among which SP1, MYC, CTNNB1, CREB1, JUN were identified as the most critical genes because the number of up- and downregulated gene ranked at the top. In conclusion, the pathways of immune response and cytokine-cytokine receptor interaction were determined as the main metabolic pathways of NP affecting the brain, and SP1, MYC, CTNNB1, CREB1, JUN genes were recognized as the most enriched genes in this process, which may provide evidence for the diagnosis and treatment research of neuropathic pain.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Genes jun/genética , Genes myc/genética , Imunoglobulinas/genética , beta Catenina/genética , Animais , Encéfalo/metabolismo , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica/genética , Redes Reguladoras de Genes/genética , Masculino , Camundongos , Mapas de Interação de Proteínas/genética , Receptores de Superfície Celular/genética
11.
Fish Shellfish Immunol ; 84: 639-647, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30366093

RESUMO

Hemocyanin (HMC) is a respiratory glycoprotein, which also plays multifunctional non-specific innate immune defense functions in shrimp. However, the transcriptional regulatory mechanisms of the hemocyanin gene expression have not been reported. In the present study, we cloned a 4324 bp fragment of small subunit hemocyanin (HMCs) gene of Litopenaeus vannamei including the 5'-flanking region, from upstream 2475 bp to downstream 1849 bp (exon 1-intron 1-exon 2) by genome walking method. Four deletion constructs were then generated and their promoter activity assessed using the luciferase reporter system. Interestingly, we identified an alternative promoter (+1516/+1849 bp) located in exon 2, which has stronger promoter activity than the full-length or the other constructs. Bioinformatics analyses revealed that the alternative promoter region contains two conserved binding sites of the transcription factor c-Jun. Mutational analysis and electrophoretic mobility shift assay showed that Litopenaeus vannamei c-Jun (Lvc-Jun) binds to the region +1582/+1589 bp and +1831/+1837 bp of the alternative promoter. Furthermore, overexpression of Lvc-Jun significantly increased the alternative promoter activity, while co-transfection with dsRNA-Lvc-Jun significantly reduced the alternative promoter activity of HMCs. Taken together, our present data indicate that the transcription factor Lvc-Jun is essential for the transcriptional regulation of the HMCs gene expression.


Assuntos
Proteínas de Artrópodes/genética , Regulação da Expressão Gênica , Hemocianinas/genética , Penaeidae/genética , Regiões Promotoras Genéticas/genética , Fatores de Transcrição/genética , Região 5'-Flanqueadora , Animais , Proteínas de Artrópodes/metabolismo , Sequência de Bases , Sítios de Ligação , Clonagem Molecular , Genes jun/genética , Hemocianinas/metabolismo , Penaeidae/imunologia , Penaeidae/metabolismo , Fatores de Transcrição/metabolismo
12.
Cell Death Dis ; 9(10): 977, 2018 09 24.
Artigo em Inglês | MEDLINE | ID: mdl-30250020

RESUMO

Gastric cancer (GC) ranks among the top five malignant tumors worldwide by the incidence and mortality rate. However, the mechanisms underlying its progression are poorly understood. In this study, we investigated the role of SIRT1, a class III deacetylase, in the invasion and metastasis of GC. Here, we found that knockdown of SIRT1 promoted GC cell migration and invasion in vitro and metastasis in vivo. Forced expression of SIRT1 in GC cells had the opposite effects. Then, we used mRNA microarray to identify the target genes that are regulated by SIRT1 and found that ARHGAP5 was downregulated by SIRT1. The results of the mRNA microarray were confirmed in several GC cell lines. Furthermore, SIRT1 inhibited the expression of ARHGAP5 by physically associating with transcription factor c-JUN and deacetylating and inhibiting the transcriptional activity of c-JUN. Then the expression dynamics and clinical significance of ARHGAP5 were analyzed using clinical samples and database. The expression of ARHGAP5 was increased in GC, and positively correlated with tumor size, tumor infiltration, lymph node metastasis, and clinical stage. And multivariate analyses indicated that ARHGAP5 served as an independent prognostic marker of GC. In addition, the biological effects of ARHGAP5 in SIRT1-mediated inhibition of GC migration and invasion were investigated using both in vitro and in vivo models. Silencing of ARHGAP5 considerably inhibited the migration and invasion of GC, and ARHGAP5 was found to be involved in the SIRT1-mediated inhibition of GC migration and invasion. Our results indicate that SIRT1 suppresses migration and invasion of GC by downregulating ARHGAP5 through an interaction with c-JUN, and these phenomena represent a novel mechanism of the antitumor action of SIRT1.


Assuntos
Proteínas Ativadoras de GTPase/metabolismo , Sirtuína 1/metabolismo , Neoplasias Gástricas/patologia , Animais , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Progressão da Doença , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Inativação Gênica , Genes jun/genética , Humanos , Masculino , Camundongos , Camundongos Nus , Análise Multivariada , Invasividade Neoplásica , Metástase Neoplásica , Sirtuína 1/genética , Transfecção
13.
Metab Brain Dis ; 33(5): 1501-1508, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29916183

RESUMO

Chronic exposure to morphine can produce tolerance, dependence and addiction, but the underlying neurobiological basis is still incompletely understood. c-Jun, as an important component of the activator protein-1 transcription factor, is supposed to take part in regulating gene expression in AC/cAMP/PKA signaling. MicroRNA (miRNA) has emerged as a critical regulator of neuronal functions. Although a number of miRNAs have been reported to regulate the µ-opioid receptor expression, there has been no report about miRNAs to regulate chronic morphine-induced, naloxone-precipitated cAMP overshoot. Our results showed that chronic morphine pretreatment induced naloxone-precipitated cAMP overshoot in concentration- and time-dependent manners in HEK 293/µ cells. Chronic morphine pretreatment alone elevated both c-Jun protein and miR-139-5p expression levels, while dramatically artificial elevation of miR-139-5p inhibited c-Jun at the translational level. Furthermore, dramatically artificial upregulation of intracellular miR-139-5p limited chronic morphine-induced, naloxone-precipitated cAMP overshoot. These findings suggested that miR-139-5p was involved in regulating chronic morphine-induced, naloxone-precipitated cAMP overshoot in a negative feedback manner through its target c-Jun, which extends our understanding of neurobiological mechanisms underlying morphine dependence and addiction.


Assuntos
Adenilil Ciclases/metabolismo , Analgésicos Opioides/farmacologia , AMP Cíclico/análogos & derivados , MicroRNAs/fisiologia , Morfina/farmacologia , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Proteínas Proto-Oncogênicas c-jun/metabolismo , Tionucleotídeos/metabolismo , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Retroalimentação Fisiológica , Regulação da Expressão Gênica , Genes jun/genética , Células HEK293 , Humanos , MicroRNAs/genética , Mimetismo Molecular , Dependência de Morfina/genética , Dependência de Morfina/metabolismo , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição AP-1/genética , Regulação para Cima/efeitos dos fármacos
14.
JAMA Dermatol ; 154(7): 819-823, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29898214

RESUMO

Importance: Molecular alterations in lichen sclerosus-associated vulvar squamous cell carcinoma (LS-VSCC) are largely unknown. Objective: To determine whether the retinoic acid receptor ß (RARß) tumor-suppressor gene is involved in the onset and/or progression of LS-VSCC. Design, Setting, and Participants: The case-control study, conducted at University-Hospital of Ferrara, Italy, included 20 LS-VSCC (mean [SD] age, 75 [3] years) and 20 cancer-associated vulvar LS (caVLS; mean [SD] age, 62 [11] years) formalin-fixed embedded tissue specimens, 20 cancer-free vulvar LS (cfVLS), and 20 normal skin fresh specimens from diagnostic biopsies and women surgically treated for nonmalignant skin lesions, respectively. RARß gene expression and promoter methylation were investigated in LS-VSCC and caVLS adjacent to VSCC specimens, and in cfVLS and normal skin specimens, as controls, by RT-Q real-time polymerase chain reaction (PCR) analysis, and sequencing of PCR-amplified bisulfite-treated DNA. c-Jun expression, an RARß pathway-related gene, was also investigated. Main Outcomes and Measures: RARß expression, correlation with its promoter methylation and c-Jun expression, and association with onset or progression of LS-VSCC. Results: In LS-VSCC, RARß messenger RNA was 3.4-, 3.6-, and 4.8-fold lower than in caVLS (P = .001), cfVLS (P = .005), and normal skin (P < .001), respectively. The RARß mRNA levels were similar in caVLS, cfVLS, and normal skin. The RARß promoter was hypermethylated in 18 (90%) of 20 LS-VSCC, 11 (55%) of 20 cfVLS, 10 (50%) of 20 caVLS, and 5 (25%) of 20 in the normal skin group. The degree of methylation of RARß promoter was higher in LS-VSCC, ranging from 5 to 9 (full promoter methylation) CpGs methylated, than in caVLS (P = .02), cfVLS (P = .03), or normal skin (P < .001), which was up to 5 CpGs methylated. Importantly, 0 of 8 LS-VSCC with 5 to 6 CpGs methylated and 5 (63%) of 8 LS-VSCC with 7 to 8 CpGs methylated were from patients with lymph node metastasis at diagnosis, respectively, whereas there were 2 of 2 (100%) LS-VSCC samples with 9 CpG methylated from patients with lymph node metastasis at diagnosis and subsequent recurrence. In LS-VSCC c-Jun mRNA was 4.3-, 1.4-, and 2.6-fold higher than in caVLS (P < .001), cfVLS (P = .001), and normal skin (P < .001), respectively. The expression of c-Jun was similar in caVLS, cfVLS, and normal skin. Conclusions and Relevance: Hypermethylation-induced RARß down-expression was associated with LS-VSCC and correlates with the upregulation of c-Jun. The degree of methylation of RARß promoter increased with the malignancy of LS-VSCC. Therefore, RARß gene dysregulation may play a role in progression of LS-VSCC, and RARß promoter methylation status may be used as a prognostic marker in clinical treatment of patients with LS-VSCC.


Assuntos
Carcinoma de Células Escamosas/genética , Receptores do Ácido Retinoico/genética , Neoplasias Vulvares/genética , Idoso , Carcinoma de Células Escamosas/etiologia , Carcinoma de Células Escamosas/secundário , Estudos de Casos e Controles , Ilhas de CpG/genética , Metilação de DNA , Progressão da Doença , Feminino , Expressão Gênica , Genes jun/genética , Humanos , Metástase Linfática/genética , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Líquen Escleroso Vulvar/complicações , Líquen Escleroso Vulvar/patologia , Neoplasias Vulvares/complicações , Neoplasias Vulvares/patologia
15.
Nature ; 554(7693): 533-537, 2018 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-29443959

RESUMO

Chronic inflammation increases the risk of developing one of several types of cancer. Inflammatory responses are currently thought to be controlled by mechanisms that rely on transcriptional networks that are distinct from those involved in cell differentiation. The orphan nuclear receptor NR5A2 participates in a wide variety of processes, including cholesterol and glucose metabolism in the liver, resolution of endoplasmic reticulum stress, intestinal glucocorticoid production, pancreatic development and acinar differentiation. In genome-wide association studies, single nucleotide polymorphisms in the vicinity of NR5A2 have previously been associated with the risk of pancreatic adenocarcinoma. In mice, Nr5a2 heterozygosity sensitizes the pancreas to damage, impairs regeneration and cooperates with mutant Kras in tumour progression. Here, using a global transcriptomic analysis, we describe an epithelial-cell-autonomous basal pre-inflammatory state in the pancreas of Nr5a2+/- mice that is reminiscent of the early stages of pancreatitis-induced inflammation and is conserved in histologically normal human pancreases with reduced expression of NR5A2 mRNA. In Nr5a2+/-mice, NR5A2 undergoes a marked transcriptional switch, relocating from differentiation-specific to inflammatory genes and thereby promoting gene transcription that is dependent on the AP-1 transcription factor. Pancreatic deletion of Jun rescues the pre-inflammatory phenotype, as well as binding of NR5A2 to inflammatory gene promoters and the defective regenerative response to damage. These findings support the notion that, in the pancreas, the transcriptional networks involved in differentiation-specific functions also suppress inflammatory programmes. Under conditions of genetic or environmental constraint, these networks can be subverted to foster inflammation.


Assuntos
Diferenciação Celular/genética , Regulação da Expressão Gênica , Inflamação/genética , Pâncreas/metabolismo , Pâncreas/patologia , Receptores Citoplasmáticos e Nucleares/metabolismo , Transcriptoma , Células Acinares/metabolismo , Células Acinares/patologia , Animais , Cromatina/genética , Cromatina/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Redes Reguladoras de Genes/genética , Genes jun/genética , Heterozigoto , Humanos , Camundongos , Especificidade de Órgãos/genética , Pancreatite/genética , Regiões Promotoras Genéticas/genética , Receptores Citoplasmáticos e Nucleares/deficiência , Receptores Citoplasmáticos e Nucleares/genética , Fator de Transcrição AP-1/metabolismo
16.
Mol Med Rep ; 16(5): 6570-6579, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28901441

RESUMO

Multiple symmetric lipomatosis (MSL) is a rare disorder characterized by aberrant multiple and symmetric subcutaneous adipose tissue accumulation in the face, neck, shoulders, back, chest and abdomen, severely affecting the quality of life of patients. At present, precise MSL etiology and pathogenesis remain to be elucidated. The present study first utilized a digital gene expression technique with a next­generation sequencing platform to profile differentially expressed genes in three cases of MSL vs. normal control tissue. cDNA libraries from these tissue specimens were constructed and DNA sequenced for identification of differentially expressed genes, which underwent bioinformatic analysis using the Gene Ontology (GO) enrichment, Kyoto Encyclopedia of Genes and Genomes (KEGG) and protein­protein interaction (PPI) network analyses. As a result, a total of 859 differentially expressed genes were identified, including 308 upregulated genes (C19orf80, Apelin, C21orf33, FAM166B and HSD11B2 were mostly upregulated 6.984­, 4.670­, 4.412­, 3.693­ and 3.561­fold, respectively) and 551 downregulated genes [FosB proto­oncogene, AP­1 transcription factor subunit (FOSB), selectin (SEL) E, RAR related orphan receptor (ROR) B, salt inducible kinase (SIK)1 and epidermal growth factor­like protein (EGFL)6 were mostly downregulated ­9.845, ­8.243, ­8.123, ­7.702 and ­7.664 fold, respectively). The GO functional enrichment analysis demonstrated these differentially expressed genes were predominantly involved in biological processes and cellular components, while the KEGG pathway enrichment analysis demonstrated that ribosome, non­alcoholic fatty liver disease, human T­lymphotropic virus type 1 (HTLV­I) infection and Alzheimer's disease pathways were altered in MSL. The PPI network data demonstrated ubiquitin C (UBC), translocator protein (TSPO), Jun Proto­Oncogene, AP­1 Transcription Factor (JUN) and FOS were among these differentially expressed genes that participated in regulation of adipocyte differentiation, although no previous study has linked them to MSL. In conclusion, the present study profiled differentially expressed genes in MSL and identified gene pathways that may be associated with MSL development and progression.


Assuntos
Tecido Adiposo/metabolismo , Redes Reguladoras de Genes/genética , Lipomatose Simétrica Múltipla/genética , Diferenciação Celular/genética , Biologia Computacional/métodos , Regulação para Baixo/genética , Perfilação da Expressão Gênica/métodos , Biblioteca Gênica , Genes jun/genética , Humanos , Masculino , Pessoa de Meia-Idade , Mapas de Interação de Proteínas/genética , Receptores de GABA/genética , Regulação para Cima/genética
17.
Mol Med Rep ; 16(2): 1635-1642, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28627654

RESUMO

Solid pseudopapillary neoplasm (SPN) of the pancreas is a low-grade malignant neoplasm that accounts for ~5% of cystic pancreatic tumors and ~0.9­2.7% of exocrine pancreatic tumors. The transcription profiling data (GSE43795) of 14 SPN and 6 control samples were downloaded from the Gene Expression Omnibus (GEO) database. Using the Limma package, Student's t­tests were performed to identify differentially expressed genes (DEGs) between SPN and control samples [with the following criterion: False discovery rate (FDR)<0.01 and log2 fold­change (FC)≥3]. Pathway and functional enrichment analyses were performed to investigate the biological processes that the DEGs were involved in. Protein­protein interaction (PPI) network and sub­network analyses were conducted to comprehensively understand the interactions between DEGs. The screened DEGs were further annotated according to information relating to transcription factors and tumor associated genes (TAGs). A total of 710 upregulated and 710 downregulated DEGs were observed, including 74 transcriptional factors and 124 TAGs. Membrane metallo­endopeptidase (MME), matrix metalloproteinase (MMP)-2 and MMP­9 were also identified as key TAGs. Following PPI network analysis, hub nodes of epidermal growth factor receptor (EGFR), proto­oncogene tyrosine protein kinase Fyn (FYN), c­JUN (JUN), glucagon (GCG), c­Myc (MYC) and CD44 were identified, the majority of which participate in the epidermal growth factor receptor (ErbB) and gonadotropin-releasing hormone (GnRH) signaling pathways. A sub­network involving 70 gene nodes was also identified, with EGFR as the central gene. MME, MMP­2 and MMP­9 contribute to proliferative diabetic retinopathy and also involved in SPN. The genes EGFR, FYN, JUN, GCG, MYC and CD44 may therefore be key genes in SPN, and the ErbB and GnRH signaling pathways may be an important contributor to SPN progression.


Assuntos
Biologia Computacional , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , Genes Neoplásicos , Neoplasias Epiteliais e Glandulares/genética , Pâncreas/metabolismo , Neoplasias Pancreáticas/genética , Regulação para Baixo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Genes jun/genética , Glucagon/genética , Glucagon/metabolismo , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Humanos , Receptores de Hialuronatos/genética , Receptores de Hialuronatos/metabolismo , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Neoplasias Epiteliais e Glandulares/metabolismo , Neprilisina/genética , Neprilisina/metabolismo , Neoplasias Pancreáticas/metabolismo , Mapas de Interação de Proteínas , Proteínas Proto-Oncogênicas c-fyn/genética , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proto-Oncogenes/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Regulação para Cima
18.
Prenat Diagn ; 37(2): 133-143, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27885689

RESUMO

OBJECTIVES: The objective of the study is to investigate the role of methylation levels at promoter regions of placental vascularization genes (VEGF, EGFR, and c-jun) in pathogenesis and diagnosis of placental disorders. METHODS: We analyzed DNA and histone methylation at promoters of VEGF, EGFR, and c-jun via methylation-sensitive high-resolution melting and chromatin immunoprecipitation assay in pregnant women with normal pregnancy in first, second, and third trimesters (n = 30 in each group) and pregnant women with pregnancy complicated with preeclampsia (n = 30) and hydatidiform mole (n = 15). RESULTS: The higher expression of VEGF, EGFR, and c-jun in early pregnancy was observed to be independent of DNA methylation, while it was associated with H3 K9/K27 trimethylations. Also, abnormally higher expression of c-jun in GTDs was associated with lower H3K9me3 level at its promoter. Under preeclampsia conditions, we observed dysregulation of both DNA methylation and H3 trimethylation and subsequent low expression of VEGF, EGFR, and c-jun. Importantly, our promoter methylation data indicated that VEGF may act as novel fetal DNA diagnostic marker for preeclampsia and molar pregnancies in maternal plasma. CONCLUSION: These findings emphasize the importance of dysregulated epigenetic phenomenon behind the pathologies of placental disorders and use of promoter region DNA methylation as an epigenetic marker for these pathological pregnancies. © 2016 John Wiley & Sons, Ltd.


Assuntos
Metilação de DNA/fisiologia , Doenças Placentárias/diagnóstico , Doenças Placentárias/genética , Células-Tronco/metabolismo , Trofoblastos/metabolismo , Adulto , Receptores ErbB/genética , Feminino , Genes jun/genética , Humanos , Mola Hidatiforme/diagnóstico , Mola Hidatiforme/genética , Doenças Placentárias/metabolismo , Doenças Placentárias/patologia , Pré-Eclâmpsia/diagnóstico , Pré-Eclâmpsia/genética , Valor Preditivo dos Testes , Gravidez , Diagnóstico Pré-Natal/métodos , Regiões Promotoras Genéticas , Células-Tronco/citologia , Trofoblastos/citologia , Células Tumorais Cultivadas , Neoplasias Uterinas/diagnóstico , Neoplasias Uterinas/genética , Fator A de Crescimento do Endotélio Vascular/genética , Adulto Jovem
19.
Medicine (Baltimore) ; 95(31): e4473, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27495086

RESUMO

Heroin addiction is a complex psychiatric disorder with a chronic course and a high relapse rate, which results from the interaction between genetic and environmental factors. Heroin addiction has a substantial heritability in its etiology; hence, identification of individuals with a high genetic propensity to heroin addiction may help prevent the occurrence and relapse of heroin addiction and its complications. The study aimed to identify a small set of genetic signatures that may reliably predict the individuals with a high genetic propensity to heroin addiction. We first measured the transcript level of 13 genes (RASA1, PRKCB, PDK1, JUN, CEBPG, CD74, CEBPB, AUTS2, ENO2, IMPDH2, HAT1, MBD1, and RGS3) in lymphoblastoid cell lines in a sample of 124 male heroin addicts and 124 male control subjects using real-time quantitative PCR. Seven genes (PRKCB, PDK1, JUN, CEBPG, CEBPB, ENO2, and HAT1) showed significant differential expression between the 2 groups. Further analysis using 3 statistical methods including logistic regression analysis, support vector machine learning analysis, and a computer software BIASLESS revealed that a set of 4 genes (JUN, CEBPB, PRKCB, ENO2, or CEBPG) could predict the diagnosis of heroin addiction with the accuracy rate around 85% in our dataset. Our findings support the idea that it is possible to identify genetic signatures of heroin addiction using a small set of expressed genes. However, the study can only be considered as a proof-of-concept study. As the establishment of lymphoblastoid cell line is a laborious and lengthy process, it would be more practical in clinical settings to identify genetic signatures for heroin addiction directly from peripheral blood cells in the future study.


Assuntos
Predisposição Genética para Doença , Dependência de Heroína/genética , Adulto , Proteína beta Intensificadora de Ligação a CCAAT/genética , Proteínas Estimuladoras de Ligação a CCAAT/genética , Estudos de Casos e Controles , Perfilação da Expressão Gênica , Genes jun/genética , Humanos , Modelos Logísticos , Linfócitos/citologia , Masculino , Fosfopiruvato Hidratase/genética , Proteína Quinase C beta/genética , RNA Ribossômico 18S/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Software , Máquina de Vetores de Suporte
20.
Nature ; 536(7614): 96-9, 2016 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-27462815

RESUMO

Eukaryotic mRNAs contain a 5' cap structure that is crucial for recruitment of the translation machinery and initiation of protein synthesis. mRNA recognition is thought to require direct interactions between eukaryotic initiation factor 4E (eIF4E) and the mRNA cap. However, translation of numerous capped mRNAs remains robust during cellular stress, early development, and cell cycle progression despite inactivation of eIF4E. Here we describe a cap-dependent pathway of translation initiation in human cells that relies on a previously unknown cap-binding activity of eIF3d, a subunit of the 800-kilodalton eIF3 complex. A 1.4 Å crystal structure of the eIF3d cap-binding domain reveals unexpected homology to endonucleases involved in RNA turnover, and allows modelling of cap recognition by eIF3d. eIF3d makes specific contacts with the cap, as exemplified by cap analogue competition, and these interactions are essential for assembly of translation initiation complexes on eIF3-specialized mRNAs such as the cell proliferation regulator c-Jun (also known as JUN). The c-Jun mRNA further encodes an inhibitory RNA element that blocks eIF4E recruitment, thus enforcing alternative cap recognition by eIF3d. Our results reveal a mechanism of cap-dependent translation that is independent of eIF4E, and illustrate how modular RNA elements work together to direct specialized forms of translation initiation.


Assuntos
Fator de Iniciação 3 em Eucariotos/metabolismo , Iniciação Traducional da Cadeia Peptídica , Capuzes de RNA/metabolismo , Proteínas de Ligação a RNA/metabolismo , Ligação Competitiva , Cristalografia por Raios X , Fator de Iniciação 3 em Eucariotos/química , Fator de Iniciação 4E em Eucariotos/antagonistas & inibidores , Fator de Iniciação 4E em Eucariotos/metabolismo , Fator de Iniciação 4F em Eucariotos/antagonistas & inibidores , Fator de Iniciação 4F em Eucariotos/metabolismo , Genes jun/genética , Humanos , Modelos Moleculares , Filogenia , Ligação Proteica , Estrutura Terciária de Proteína , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Capuzes de RNA/química , Capuzes de RNA/genética , Proteínas de Ligação a RNA/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...