Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
1.
J Med Virol ; 96(4): e29580, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38566572

RESUMO

A persistent infection with human papillomavirus (HPV) can induce precancerous lesions of the cervix that may ultimately develop into cancer. Cervical cancer development has been linked to altered microRNA (miRNA) expression, with miRNAs regulating anchorage-independent growth being particularly important for the progression of precancerous lesions to cancer. In this study, we set out to identify and validate targets of miR-129-5p, a previously identified tumor suppressive miRNA involved in anchorage-independent growth and HPV-induced carcinogenesis. We predicted 26 potential miR-129-5p targets using online databases, followed by KEGG pathway enrichment analysis. RT-qPCR and luciferase assays confirmed that 3'UTR regions of six genes (ACTN1, BMPR2, CAMK4, ELK4, EP300, and GNAQ) were targeted by miR-129-5p. Expressions of ACTN1, CAMK4, and ELK4 were inversely correlated to miR-129-5p expression in HPV-transformed keratinocytes, and their silencing reduced anchorage-independent growth. Concordantly, miR-129-5p overexpression decreased protein levels of ACTN1, BMPR2, CAMK4 and ELK4 in anchorage-independent conditions. Additionally, c-FOS, a downstream target of ELK4, was downregulated upon miR-129-5p overexpression, suggesting regulation through the ELK4/c-FOS axis. ACTN1 and ELK4 expression was also upregulated in high-grade precancerous lesions and cervical cancers, supporting their clinical relevance. In conclusion, we identified six targets of miR-129-5p involved in the regulation of anchorage-independent growth, with ACTN1, BMPR2, ELK4, EP300, and GNAQ representing novel targets for miR-129-5p. For both ACTN1 and ELK4 functional and clinical relevance was confirmed, indicating that miR-129-5p-regulated ACTN1 and ELK4 expression contributes to HPV-induced carcinogenesis.


Assuntos
MicroRNAs , Infecções por Papillomavirus , Lesões Pré-Cancerosas , Neoplasias do Colo do Útero , Feminino , Humanos , Papillomavirus Humano , Infecções por Papillomavirus/genética , Infecções por Papillomavirus/patologia , Linhagem Celular Tumoral , MicroRNAs/genética , MicroRNAs/metabolismo , Queratinócitos/metabolismo , Queratinócitos/patologia , Carcinogênese/genética , Carcinogênese/patologia , Lesões Pré-Cancerosas/patologia , Proliferação de Células/genética , Proteínas Elk-4 do Domínio ets , Actinina/genética
2.
Adv Sci (Weinh) ; 10(32): e2303378, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37786278

RESUMO

Although the MAPK/MEK/ERK pathway is prevalently activated in colorectal cancer (CRC), MEK/ERK inhibitors show limited efficiency in clinic. As a downstream target of MAPK, ELK4 is thought to work primarily by forming a complex with SRF. Whether ELK4 can serve as a potential therapeutic target is unclear and the transcriptional regulatory mechanism has not been systemically analyzed. Here, it is shown that ELK4 promotes CRC tumorigenesis. Integrated genomics- and proteomics-based approaches identified SP1 and SP3, instead of SRF, as cooperative functional partners of ELK4 at genome-wide level in CRC. Serum-induced phosphorylation of ELK4 by MAPKs facilitated its interaction with SP1/SP3. The pathological neoangiogenic factor LRG1 is identified as a direct target of the ELK4-SP1/SP3 complex. Furthermore, targeting the ELK4-SP1/SP3 complex by combination treatment with MEK/ERK inhibitor and the relatively specific SP1 inhibitor mithramycin A (MMA) elicited a synergistic antitumor effect on CRC. Clinically, ELK4 is a marker of poor prognosis in CRC. A 9-gene prognostic model based on the ELK4-SP1/3 complex-regulated gene set showed robust prognostic accuracy. The results demonstrate that ELK4 cooperates with SP1 and SP3 to transcriptionally regulate LRG1 to promote CRC tumorigenesis in an SRF-independent manner, identifying the ELK4-SP1/SP3 complex as a potential target for rational combination therapy.


Assuntos
Neoplasias Colorretais , Regulação da Expressão Gênica , Humanos , Regiões Promotoras Genéticas , Neoplasias Colorretais/genética , Carcinogênese/genética , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Proteínas Elk-4 do Domínio ets/genética , Glicoproteínas
3.
Nat Commun ; 14(1): 6117, 2023 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-37777542

RESUMO

The proline synthesis is importantly involved in tumor growth under hypoxia, while the underlying mechanism remains to be further investigated. Here we show that pyrroline-5-carpoxylate reductase-1 (PYCR1), displaying a constant nuclear localization, is phosphorylated by nuclear IGF1R at Tyrosine 135 under hypoxia; this phosphorylation promotes the binding of PYCR1 to ELK4 and thus PYCR1 recruitment to ELK4-targeted genes promoter. Under hypoxia, ELK4-binding ability and enzymatic activity of PYCR1 are both required for ELK4-Sirt7-mediated transcriptional repression and cell growth maintenance, in which PYCR1-catalyzed NAD+ production stimulates the deacetylation activity of Sirt7 on H3K18ac that restrains genes transcription. Functionally, PYCR1 Tyr-135 phosphorylation exerts supportive effect on tumor growth under hypoxia, and the level of PYCR1 Tyr-135 phosphorylation is associated with malignancy of colorectal cancer (CRC). These data uncover the relationship between the compartmentally metabolic activity of PYCR1 and genes transcription regulation, and highlight the oncogenic role of PYCR1 during CRC development.


Assuntos
Neoplasias , Oxirredutases , Humanos , Linhagem Celular Tumoral , Proteínas Elk-4 do Domínio ets/metabolismo , Hipóxia/genética , Neoplasias/genética , Receptor IGF Tipo 1/genética , delta-1-Pirrolina-5-Carboxilato Redutase
4.
Balkan Med J ; 40(6): 409-414, 2023 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-37519006

RESUMO

Background: Cervical cancer (CC) is a prevalent gynecological carcinoma, and patients infected with human papillomavirus (HPV) have a higher morbidity rate. Aims: To explore the effects of ETS-like transcription factor 4 (ELK4) in patients with HPV+ CC. Study design: In vitro cell lines and human-sample study. Methods: The ELK4 levels in human tissue (65 HPV+ CC tissue and 25 HPV− normal cervical tissue) and cell lines (human cervical epithelial immortalized cell line H8 and CC cell lines HeLa [HPV18], CaSki [HPV16], and SiHa [HPV−]) were quantified using qRT-PCR and western blot assay. ELK4 knockdown transfection was effective and confirmed by western blotting. The MTT and EDU assays were used to evaluate cell viability and proliferation, respectively. Flow cytometry was used to detect the CC cell cycle stage. Stem cell markers, such as cluster of differentiation 133 (CD133), CD44, and aldehyde dehydrogenase 1, and the cervicospheres formed were measured. ChIP-qPCR and luciferase activity experiments were used to assess the bond between ELK4 and F-box protein 22 (FBXO22). Results: ELK4 was highly expressed in the HPV+ CC tissue. CC cells with ELK4 knockdown had lower viability and proliferation than the control cells. ELK4 knockdown blocked the progression of the cell cycle from G1 to S phase. ELK4 knockdown suppressed the stem cell-like characteristics of the HPV+ CC cells. ELK4 bonded with the FBXO22 promoter, inhibiting the levels of phosphatase and tensin homolog (PTEN). Conclusion: ELK4 facilitated cell cycle progression and stem cell-like characteristics by regulating the FBXO22/PTEN axis. Thus, ELK4 could be a potential therapeutic target to arrest the progress of HPV-associated CC.


Assuntos
Proteínas F-Box , Infecções por Papillomavirus , Neoplasias do Colo do Útero , Feminino , Humanos , Neoplasias do Colo do Útero/metabolismo , Papillomavirus Humano , Linhagem Celular Tumoral , Proliferação de Células , Células-Tronco/metabolismo , Células-Tronco/patologia , Ciclo Celular , PTEN Fosfo-Hidrolase/metabolismo , PTEN Fosfo-Hidrolase/farmacologia , Proteínas Elk-4 do Domínio ets/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores Citoplasmáticos e Nucleares/farmacologia , Proteínas F-Box/farmacologia
5.
Mol Biotechnol ; 65(10): 1608-1618, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36715861

RESUMO

Increasing evidences indicate the crucial role of circRNAs in tumorigenesis, but little is understood about their molecular mechanism in retinoblastoma (RB). This paper was designed for exploring the circ_0119412 function in cases with RB and the potential mechanism. RT-qPCR was utilized to ascertain circ_0119412 and miR-186-5p levels in RB tissues and cells, and western blotting was used to quantify ELK4 in RB cells. In addition, CCK-8 and scratch assays were carried out for evaluation of cell proliferation and migration, respectively. Apoptosis-related proteins levels (Bax and Bcl-2) were measure by western blotting. Tumor growth in vivo was detected utilizing xenograft tumor experiment. The targeting relationship between circ_0119412, miR-186-5p, and ELK4 was validated using a dual-luciferase reporter assay and an RNA immunoprecipitation (RIP) assay. In RB tissues and cells, Circ_0119412 and ELK4 expression were upregulated, while miR-186-5p expression was downregulated. In vitro assay revealed that downregulating circ_0119412 accelerated the cell apoptosis of RB cells and slowed down their migration and proliferation, and the in vivo assay indicated that circ_0119412 downregulation reduced the weight and volume of tumor in nude mice. In addition, miR-186-5p interference promoted the malignant behavior of RB cells, while ELK4 silencing showed an opposite trend. Mechanically, circ_0119412 can promote RB malignant phenotypes via miR-186-5p/ELK4 axis. Circ_0119412 was found to be upregulated in RB, and could accelerate the progression of RB via the miR-186-5p/ELK4 axis, indicating circ_0119412 may serve a promising clinical therapeutic target of RB.


Assuntos
MicroRNAs , Neoplasias da Retina , Retinoblastoma , Humanos , Animais , Camundongos , Retinoblastoma/genética , Camundongos Nus , Apoptose/genética , Proliferação de Células/genética , Neoplasias da Retina/genética , MicroRNAs/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proteínas Elk-4 do Domínio ets
6.
J Hematol Oncol ; 15(1): 53, 2022 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-35526007

RESUMO

BACKGROUND: Transfer RNA-derived fragments (tRFs) are a new class of small non-coding RNAs. Recent studies suggest that tRFs participate in some pathological processes. However, the biological functions and mechanisms of tRFs in non-small cell lung cancer (NSCLC) are largely unknown. METHODS: Differentially expressed tRFs were identified by tRF and tiRNA sequencing using 9 pairs of pre- and post-operation plasma from patients with NSCLC. Quantitative real-time PCR (qRT-PCR) and fluorescence in situ hybridization (FISH) were used to determine the levels of tRF in tissues, plasma, and cells. Gain- and loss-of-function experiments were implemented to investigate the oncogenic effects of tRF on NSCLC cells in vitro and in vivo. Chromatin immunoprecipitation (ChIP), luciferase reporter, RNA pulldown, mass spectrum, RNA immunoprecipitation (RIP), Western blot, co-immunoprecipitation (Co-IP) assays, and rescue experiments were performed to explore the regulatory mechanisms of tRF in NSCLC. RESULTS: AS-tDR-007333 was an uncharacterized tRF and significantly up-regulated in NSCLC tissues, plasma, and cells. Clinically, AS-tDR-007333 overexpression could distinguish NSCLC patients from healthy controls and associated with poorer prognosis of NSCLC patients. Functionally, overexpression of AS-tDR-007333 enhanced proliferation and migration of NSCLC cells, whereas knockdown of AS-tDR-007333 resulted in opposite effects. Mechanistically, AS-tDR-007333 promoted the malignancy of NSCLC cells by activating MED29 through two distinct mechanisms. First, AS-tDR-007333 bound to and interacted with HSPB1, which activated MED29 expression by enhancing H3K4me1 and H3K27ac in MED29 promoter. Second, AS-tDR-007333 stimulated the expression of transcription factor ELK4, which bound to MED29 promoter and increased its transcription. Therapeutically, inhibition of AS-tDR-007333 suppressed NSCLC cell growth in vivo. CONCLUSIONS: Our study identifies a new oncogenic tRF and uncovers a novel mechanism that AS-tDR-007333 promotes NSCLC malignancy through the HSPB1-MED29 and ELK4-MED29 axes. AS-tDR-007333 is a potential diagnostic or prognostic marker and therapeutic target for NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Carcinoma Pulmonar de Células não Pequenas/genética , Regulação da Expressão Gênica , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Hibridização in Situ Fluorescente , Neoplasias Pulmonares/genética , Chaperonas Moleculares , RNA de Transferência/genética , RNA de Transferência/metabolismo , Proteínas Elk-4 do Domínio ets/genética , Proteínas Elk-4 do Domínio ets/metabolismo
7.
Cell Death Dis ; 12(11): 957, 2021 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-34663788

RESUMO

Long non-coding RNAs (lncRNAs) play important regulatory roles in the initiation and progression of various cancers. However, the biological roles and the potential mechanisms of lncRNAs in gastric cancers remain unclear. Here, we report that the expression of lncRNA SNHG22 (small nucleolar RNA host gene 22) was significantly increased in GC (Gastric Cancer) tissues and cells, which confers poor prognosis of patients. Knockdown of SNHG22 inhibited the proliferation and invasion ability of GC cells. Moreover, we identified that the transcriptional factor, ELK4 (ETS transcription factor ELK4), could promote SNHG22 expression in GC cells. In addition, using RNA pull-down followed MS assay, we found that SNHG22 directly bound to EZH2 (enhancer of zeste 2 polycomb repressive complex 2 subunit) to suppress the expression of tumor suppressor genes. At the same time, SNHG22 sponged miR-200c-3p to increase Notch1 (notch receptor 1) expression. Taken together, our findings demonstrated the role of SNHG22 on promoting proliferation and invasion of GC cells. And we revealed a new regulatory mechanism of SNHG22 in GC cells. SNHG22 is a promising lncRNA biomarker for diagnosis and prognosis and a potential target for GC treatment.


Assuntos
Progressão da Doença , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , MicroRNAs/metabolismo , RNA Longo não Codificante/metabolismo , Receptor Notch1/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Proteínas Elk-4 do Domínio ets/metabolismo , Animais , Sequência de Bases , Sítios de Ligação , Linhagem Celular Tumoral , Proliferação de Células/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Genes Supressores de Tumor , Histonas/metabolismo , Humanos , Lisina/metabolismo , Metilação , Camundongos Endogâmicos BALB C , Camundongos Nus , MicroRNAs/genética , Invasividade Neoplásica , Regiões Promotoras Genéticas/genética , Ligação Proteica , RNA Longo não Codificante/genética , Regulação para Cima/genética
8.
J Transl Med ; 19(1): 342, 2021 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-34372882

RESUMO

BACKGROUND: We tried to elaborate the molecular mechanism of ETS-like transcription factor 4 (ELK4) affecting gastric cancer (GC) progression through M2 polarization of macrophages mediated by lysine-specific demethylase 5A (KDM5A)-Praja2 (PJA2)-kinase suppressor of ras 1 (KSR1) axis. METHODS: GC expression dataset was obtained from GEO database, and the downstream regulatory mechanism of ELK4 was predicted. Tumor-associated macrophages (TAMs) were isolated from GC tissues. The interaction among ELK4, KDM5A, PJA2 and KSR1 was analyzed by dual luciferase reporter gene, ChIP and Co-IP assays. The stability of KSR1 protein was detected by cycloheximide (CHX) treatment. After TAMs were co-cultured with HGC-27 cells, HGC-27 cell biological processes were assessed through gain- and loss-of function assays. Tumorigenicity was detected by tumorigenicity test in nude mice. RESULTS: In GC and TAMs, ELK4, KDM5A and KSR1 were highly expressed, while PJA2 was lowly expressed. M2 polarization of macrophages promoted the development of GC. ELK4 activated KDM5A by transcription and promoted macrophage M2 polarization. KDM5A inhibited the expression of PJA2 by removing H3K4me3 of PJA2 promoter, which promoted M2 polarization of macrophages. PJA2 reduced KSR1 by ubiquitination. ELK4 promoted the proliferative, migrative and invasive potentials of GC cells as well as the growth of GC xenografts by regulating KSR1. CONCLUSION: ELK4 may reduce the PJA2-dependent inhibition of KSR1 by transcriptional activation of KDM5A to promote M2 polarization of macrophages, thus promoting the development of GC.


Assuntos
Neoplasias Gástricas , Animais , Linhagem Celular Tumoral , Humanos , Ativação de Macrófagos , Macrófagos , Camundongos , Camundongos Nus , Proteína 2 de Ligação ao Retinoblastoma , Neoplasias Gástricas/genética , Ativação Transcricional , Ubiquitina-Proteína Ligases , Proteínas Elk-4 do Domínio ets
9.
Inflammation ; 44(5): 2106-2119, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34091806

RESUMO

Systemic lupus erythematosus (SLE) is a prototypic autoimmune disease and a common complication of SLE is lupus nephritis (LN) during which lupus autoantibodies and proinflammatory cytokines attack the kidney and cause renal dysfunction. The current treatments to LN are limited due to a poor understanding of the pathogenesis. Here, we studied the molecular mechanisms of LN by investigating the function of circELK4/miR-27b-3p axis. MRL/lpr mice and LPS-treated HK-2 cells were used as the mouse model and cell model of LN, respectively. Blood samples were collected from LN patients. qRT-PCR and western blot were used to measure expression levels of circELK4, miR-27b-3p, apoptosis-related proteins, cytokines, and STING/IRF-3/IFN-I signaling. ELISA was performed to examine levels of cytokines including IL-6 and TNF-α. H&E staining was used to examine kidney morphology. TUNEL staining and flow cytometry were used to determine cell apoptosis. Dual luciferase activity assay and RNA pull down were employed to validate the interactions of circELK4/miR-27b-3p and miR-27b-3p/STING. CircELK4 was elevated in LN mice, patients, and LPS-treated HK-2 cells. Knockdown of circELK4 attenuated renal injury in LN mice and LPS-induced HK-2 cell injury. CircELK4 directly bound to miR-27b-3p while miR-27b-3p targeted STING. Moreover, overexpression of circELK4 could partially reverse the effects of miR-27b-3p mimics on cell apoptosis and inflammation. Furthermore, circELK4/miR-27b-3p regulated renal cell damage via modulating STING/IRF3/IFN-I signaling. CircELK4 contributes to renal injury by promoting inflammation and cell apoptosis via acting as a miR-27b-3p sponge to modulate STING/IRF3/IFN-I signaling in LN.


Assuntos
Fator Regulador 3 de Interferon/metabolismo , Interferon Tipo I/metabolismo , Nefrite Lúpica/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Elk-4 do Domínio ets/metabolismo , Adulto , Animais , Linhagem Celular , Feminino , Células HEK293 , Humanos , Nefrite Lúpica/genética , Nefrite Lúpica/patologia , Masculino , Camundongos , Camundongos Endogâmicos MRL lpr , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Transdução de Sinais/fisiologia
10.
Oncogene ; 39(24): 4666-4680, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32398865

RESUMO

Aberrant expression of laminin-332 promotes tumour growth and metastasis in multiple cancers. However, the dysregulated expression and mechanism of action of LAMB3, which encodes the ß3 subunit of laminin-332, and the mechanism underlying dysregulated LAMB3 expression in CRC remain obscure. Here, we show that LAMB3 is overexpressed in CRC and that this overexpression is correlated with tumour metastasis and poor prognosis. Overexpression of LAMB3 promoted cell proliferation and cell migration in vitro and tumour growth and metastasis in vivo, while knockdown of LAMB3 elicited opposing effects. LAMB3 inhibited the tumour suppressive function of FOXO3/4 by activating AKT in CRC. Both the BET inhibitor JQ1 and the MEK inhibitor U0126 decreased the mRNA level of LAMB3 in multiple CRC cells. Mechanistically, ELK4 cooperated with BRD2 to regulate the transcription of LAMB3 in CRC by directly binding to the ETS binding motifs in the LAMB3 promoter. ELK4 was as acetylated at K125, which enhanced the interaction between ELK4 and BRD2. JQ1 disrupted the interaction between ELK4 and BRD2, resulting in decreased binding of BRD2 to the LAMB3 promoter and downregulation of LAMB3 transcription. Both ELK4 and BRD2 expression was associated with LAMB3 expression in CRC. LAMB3 expression was also negatively correlated with FOXO3/4 in CRC. Our study reveals the pro-tumorigenic role of LAMB3 through the AKT-FOXO3/4 axis and the transcriptional mechanism of LAMB3 in CRC, demonstrating that LAMB3 is a potential therapeutic target that can be targeted by BET inhibitors and MEK inhibitors.


Assuntos
Moléculas de Adesão Celular/metabolismo , Proteínas de Ciclo Celular/metabolismo , Neoplasias Colorretais/metabolismo , Proteína Forkhead Box O3/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Proteínas Elk-4 do Domínio ets/metabolismo , Acetilação , Animais , Moléculas de Adesão Celular/genética , Proteínas de Ciclo Celular/genética , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Feminino , Proteína Forkhead Box O3/genética , Fatores de Transcrição Forkhead/genética , Humanos , Masculino , Camundongos , Proteínas Proto-Oncogênicas c-akt/genética , Fatores de Transcrição/genética , Proteínas Elk-4 do Domínio ets/genética , Calinina
11.
RNA Biol ; 17(9): 1293-1308, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32372707

RESUMO

The blood-brain barrier (BBB) plays a pivotal role in the maintenance and regulation of the neural microenvironment. The BBB breakdown is a pathological change in early Alzheimer's disease (AD). RNA-binding proteins (RBPs) and long non-coding RNAs (lncRNAs) are involved in the regulation of BBB permeability. Our study demonstrates the role of TRA2A/LINC00662/ELK4 axis in regulating BBB permeability in AD microenvironment. In Aß1-42-incubated microvascular endothelial cells (ECs) of the BBB model in vitro, TRA2A and LINC00662 were enriched. TRA2A increased the stability of LINC00662 by binding with it. The knockdown of either TRA2A or LINC00662 decreased BBB permeability due to increased expression of tight junction-related proteins. ELK4 was less expressed in the BBB model in AD microenvironment in vitro. LINC00662 mediated the degradation of ELK4 mRNA by SMD pathway. Downregulation of ELK4 increased BBB permeability by increasing the tight junction-related protein expression.TRA2A/LINC00662/ELK4 axis plays a crucial role in the regulation of BBB permeability in AD microenvironment, which may provide a novel target for the therapy of AD.


Assuntos
Barreira Hematoencefálica/metabolismo , Microambiente Celular/genética , Regulação da Expressão Gênica , RNA Longo não Codificante/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas Elk-4 do Domínio ets/genética , Doença de Alzheimer/etiologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Biomarcadores , Células Endoteliais/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Fragmentos de Peptídeos/metabolismo , Permeabilidade , Regiões Promotoras Genéticas , Ligação Proteica , Interferência de RNA , Estabilidade de RNA , RNA Longo não Codificante/genética , Proteínas de Junções Íntimas/genética , Proteínas de Junções Íntimas/metabolismo
12.
Prostate ; 80(2): 198-208, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31794091

RESUMO

BACKGROUND: Both hormone-sensitive and castration- and enzalutamide-resistant prostate cancers (PCa) depend on the ternary complex factor (TCF) protein ELK1 to serve as a tethering protein for the androgen receptor (AR) to activate a critical set of growth genes. The two sites in ELK1 required for AR binding are conserved in other members of the TCF subfamily, ELK3 and ELK4. Here we examine the potential utility of the three proteins as prognosticators of disease recurrence in PCa. METHODS: Transcriptional activity assays; Retrospective analysis of PCa recurrence using data on 501 patients in The Cancer Genome Atlas (TCGA) database; Unpaired Wilcoxon rank-sum test and multiple comparison correction using the Holm's method; Spearman's correlations; Kaplan-Meier methods; Univariable and multivariable Cox regression analyses; LASSO-based penalized Cox regression models; Time-dependent area under the receiver operating characteristic (ROC) curve. RESULTS: ELK4 but not ELK3 was coactivated by AR similar to ELK1. Tumor expression of neither ELK3 nor ELK4 was associated with disease-free survival (DFS). ELK1 was associated with higher clinical T-stage, pathology T-stage, Gleason score, prognostic grade, and positive lymph node status. ELK1 was a negative prognosticator of DFS, independent of ELK3, ELK4, clinical T-stage, pathology T-stage, prognostic grade, lymph node status, age, and race. Inclusion of ELK1 increased the abilities of the Oncotype DX and Prolaris gene panels to predict disease recurrence, correctly predicting disease recurrence in a unique subset of patients. CONCLUSIONS: ELK1 is a strong, independent prognosticator of disease recurrence in PCa, underscoring its unique role in PCa growth. Inclusion of ELK1 may enhance the utility of currently used prognosticators for clinical decision making in prostate cancer.


Assuntos
Recidiva Local de Neoplasia/genética , Neoplasias da Próstata/genética , Proteínas Elk-1 do Domínio ets/genética , Adulto , Idoso , Análise por Conglomerados , Intervalo Livre de Doença , Células HeLa , Humanos , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/patologia , Prognóstico , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-ets/genética , Receptores Androgênicos/genética , Estudos Retrospectivos , Ativação Transcricional , Proteínas Elk-4 do Domínio ets/genética
13.
J Immunol ; 201(6): 1681-1691, 2018 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-30068599

RESUMO

In mouse thymocyte development, signaling by the TCR through the ERK pathway is required for positive selection of conventional naive T cells. The Ets transcription factor ELK4 (SAP-1), an ERK-regulated cofactor of the SRF transcription factor, plays an important role in positive selection by activating immediate-early genes such as the Egr transcription factor family. The role of ELK4-SRF signaling in development of other T cell types dependent on ERK signaling has been unclear. In this article, we show that ELK4, and its close relative ELK1, act cell autonomously in the thymus to control the generation of innate-like αß CD8+ T cells with memory-like characteristics. Mice lacking ELK4 and ELK1 develop increased numbers of innate-like αß CD8+ T cells, which populate the periphery. These cells develop cell autonomously rather than through expansion of PLZF+ thymocytes and concomitantly increased IL-4 signaling. Their development is associated with reduced TCR-mediated activation of ELK4-SRF target genes and can be partially suppressed by overexpression of the ELK4-SRF target gene EGR2. Consistent with this, partial inhibition of ERK signaling in peripheral CD8+T cells promotes the generation of cells with innate-like characteristics. These data establish that low-level ERK signaling through ELK4 (and ELK1) promotes innate-like αß CD8+ T cell differentiation, tuning conventional versus innate-like development.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Diferenciação Celular/imunologia , Sistema de Sinalização das MAP Quinases/imunologia , Timo/imunologia , Proteínas Elk-1 do Domínio ets/imunologia , Proteínas Elk-4 do Domínio ets/imunologia , Animais , Linfócitos T CD8-Positivos/citologia , Diferenciação Celular/genética , Imunidade Inata , Sistema de Sinalização das MAP Quinases/genética , Camundongos , Camundongos Knockout , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Timo/citologia , Proteínas Elk-1 do Domínio ets/genética , Proteínas Elk-4 do Domínio ets/genética
14.
Cancer Lett ; 404: 53-61, 2017 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-28716526

RESUMO

Gene fusions in cancer typically lead to the expression of a fusion protein or disrupt the expression of one of the parental genes. Here we report a new phenomenon whereby a fusion transcript functions as a long non-coding chimeric RNA (lnccRNA). This fusion RNA, SLC45A3-ELK4, generated by cis-splicing between neighboring genes, was found in prostate cancer. The fusion RNA encodes the same protein as ELK4. Intriguingly, we found that the fusion RNA level is less than 1% of wild type ELK4, unlikely to perturb the general pool of ELK4 protein. Nonetheless, when the fusion RNA, but not ELK4 is silenced, cell proliferation is inhibited in both androgen-dependent and castration-resistant prostate cancer cells. This growth arrest can be rescued by exogenous expression of the fusion and a mutant designed to prevent translation of the ELK4 protein. In the same setting, the mutant could also suppress CDKN1A and several other targets of SLC45A3-ELK4. In addition, similar to many long non-coding RNAs, the fusion RNA is enriched in the nuclear fraction. Altogether, these results indicate that SLC45A3-ELK4 regulates cancer cell proliferation by its transcript, not translated protein.


Assuntos
Proliferação de Células , Proteínas de Membrana Transportadoras/fisiologia , Proteínas de Fusão Oncogênica/fisiologia , Neoplasias da Próstata/metabolismo , RNA Longo não Codificante/fisiologia , Proteínas Elk-4 do Domínio ets/fisiologia , Núcleo Celular/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Fusão Gênica , Humanos , Masculino , Proteínas de Transporte de Monossacarídeos , Proteínas de Fusão Oncogênica/metabolismo , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , RNA Longo não Codificante/metabolismo , Células Tumorais Cultivadas
15.
J Biol Chem ; 292(32): 13296-13311, 2017 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-28655758

RESUMO

Sirtuin 7 (SIRT7), a member of the NAD+-dependent class III histone deacetylases, is involved in the regulation of various cellular processes and in resisting various stresses, such as hypoxia, low glucose levels, and DNA damage. Interestingly, SIRT7 is linked to the control of glycolysis, suggesting a role in glucose metabolism. Given the important roles of SIRT7, it is critical to clarify how SIRT7 activity is potentially regulated. It has been reported that some transcriptional and post-transcriptional regulatory mechanisms are involved. However, little is known how SIRT7 is regulated by the post-translational modifications. Here, we identified ubiquitin-specific peptidase 7 (USP7), a deubiquitinase, as a negative regulator of SIRT7. We showed that USP7 interacts with SIRT7 both in vitro and in vivo, and we further demonstrated that SIRT7 undergoes endogenous Lys-63-linked polyubiquitination, which is removed by USP7. Although the USP7-mediated deubiquitination of SIRT7 had no effect on its stability, the deubiquitination repressed its enzymatic activity. We also showed that USP7 coordinates with SIRT7 to regulate the expression of glucose-6-phosphatase catalytic subunit (G6PC), a gluconeogenic gene. USP7 depletion by RNA interference increased both G6PC expression and SIRT7 enzymatic activity. Moreover, SIRT7 targeted the G6PC promoter through the transcription factor ELK4 but not through forkhead box O1 (FoxO1). In summary, SIRT7 is a USP7 substrate and has a novel role as a regulator of gluconeogenesis. Our study may provide the basis for new clinical approaches to treat metabolic disorders related to glucose metabolism.


Assuntos
Regulação Enzimológica da Expressão Gênica , Glucose-6-Fosfatase/metabolismo , Regiões Promotoras Genéticas , Processamento de Proteína Pós-Traducional , Sirtuínas/metabolismo , Ubiquitina Tiolesterase/metabolismo , Proteínas Elk-4 do Domínio ets/metabolismo , Substituição de Aminoácidos , Linhagem Celular Tumoral , Deleção de Genes , Gluconeogênese , Glucose-6-Fosfatase/antagonistas & inibidores , Glucose-6-Fosfatase/genética , Células HEK293 , Humanos , Hidrólise , Lisina/metabolismo , Mutação , Fragmentos de Peptídeos/antagonistas & inibidores , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Domínios e Motivos de Interação entre Proteínas , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Interferência de RNA , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Sirtuínas/antagonistas & inibidores , Sirtuínas/genética , Especificidade por Substrato , Ubiquitina Tiolesterase/antagonistas & inibidores , Ubiquitina Tiolesterase/genética , Peptidase 7 Específica de Ubiquitina , Ubiquitinação , Proteínas Elk-4 do Domínio ets/genética
16.
Biochem J ; 474(9): 1509-1528, 2017 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-28275114

RESUMO

The adapter protein Dok-4 (downstream of kinase-4) has been reported as both an activator and inhibitor of Erk and Elk-1, but lack of knowledge about the identity of its partner molecules has precluded any mechanistic insight into these seemingly conflicting properties. We report that Dok-4 interacts with the transactivation domain of Elk-4 through an atypical phosphotyrosine-binding domain-mediated interaction. Dok-4 possesses a nuclear export signal and can relocalize Elk-4 from nucleus to cytosol, whereas Elk-4 possesses two nuclear localization signals that restrict interaction with Dok-4. The Elk-4 protein, unlike Elk-1, is highly unstable in the presence of Dok-4, through both an interaction-dependent mechanism and a pleckstrin homology domain-dependent but interaction-independent mechanism. This is reversed by proteasome inhibition, depletion of endogenous Dok-4 or lysine-to-arginine mutation of putative Elk-4 ubiquitination sites. Finally, Elk-4 transactivation is potently inhibited by Dok-4 overexpression but enhanced by Dok-4 knockdown in MDCK renal tubular cells, which correlates with increased basal and EGF-induced expression of Egr-1, Fos and cylcinD1 mRNA, and cell proliferation despite reduced Erk activation. Thus, Dok-4 can target Elk-4 activity through multiple mechanisms, including binding of the transactivation domain, nuclear exclusion and protein destabilization, without a requirement for inhibition of Erk.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Sinais de Exportação Nuclear/genética , Sinais de Localização Nuclear/genética , Proteínas Elk-4 do Domínio ets/genética , Transporte Ativo do Núcleo Celular/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Sequência de Aminoácidos , Animais , Células COS , Proliferação de Células/genética , Cães , Regulação da Expressão Gênica , Células HEK293 , Humanos , Immunoblotting , Células Madin Darby de Rim Canino , Camundongos , Microscopia Confocal , Ligação Proteica , Interferência de RNA , Homologia de Sequência de Aminoácidos , Técnicas do Sistema de Duplo-Híbrido , Proteínas Elk-4 do Domínio ets/metabolismo
17.
Genomics ; 109(2): 75-82, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28189763

RESUMO

DNA adenine methyltransferase identification (DamID) is an enzymatic technology for detecting DNA regions targeted by chromatin-associated proteins. Proteins are fused to bacterial DNA adenine methyltransferase (Dam) and expressed in cultured cells or whole organisms. Here, we used DamID to detect DNA regions bound by the cardiac-restricted transcription factors (TFs) NKX2-5 and SRF, and ubiquitously-expressed co-factors ELK1 and ELK4. We compared targets bound by these TFs as N- and C-terminal fusions with Dam, for both wild type (WT) NKX2-5 and mutant proteins mimicking those found in congenital heart disease. Overall, DamID is highly robust: while the orientation of WT Dam fusions can affect the size of the target sets, their signatures remained largely reproducible. Furthermore, a severe NKX2-5 mutant lacking the homeodomain showed strong steric effects negatively impacting target discovery. The extent of steric effect is likely to be dependent on the protein in question and the orientation of Dam fusion.


Assuntos
Cromatina/metabolismo , Regulação da Expressão Gênica , Técnicas Genéticas , Cardiopatias Congênitas/metabolismo , DNA Metiltransferases Sítio Específica (Adenina-Específica) , Animais , DNA/metabolismo , Cardiopatias Congênitas/genética , Proteína Homeobox Nkx-2.5/metabolismo , Humanos , Camundongos , Fator de Resposta Sérica/metabolismo , Proteínas Elk-1 do Domínio ets/metabolismo , Proteínas Elk-4 do Domínio ets/metabolismo
18.
Pharmazie ; 72(11): 687-693, 2017 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-29442044

RESUMO

We aimed to elucidate the roles and regulatory mechanism of miR-3188 in oxidized low-density lipoprotein (ox-LDL)-induced cell injury in THP-1 derived macrophages, thus providing a new insight for the treatment of atherosclerosis (AS). A total of 85 AS patients and 45 healthy controls were enrolled. The levels of miR-3188 and lipoprotein-associated phospholipase A2 (Lp-PLA2) in AS patients and healthy controls were detected. Then ox-LDL was used to treat human THP-1 derived macrophages. The effects of overexpression and suppression of miR-3188 on regulating ox-LDL-induced cell injury in THP-1 derived macrophages were investigated. Additionally, the potential target of miR-3188 was identified, which was verified by luciferase reporter assay. Besides, the relationship between miR-3188 and RhoA/ROCK pathway was explored. miR-3188 was downregulated in AS patients, while the levels of Lp-PLA2 in AS patients were increased. Ox-LDL significantly induced cell injury by decreasing cell viability, inducing cell apoptosis and increasing the production of inflammatory cytokines, including IL-1ß, IL-6, MCP-1 and TNF-α. In addition, miR-3188 was significantly downregulated after ox-LDL treatment. Overexpression of miR-3188 alleviated ox-LDL-induced cell injury, while inhibition of miR-3188 had opposite effects. ETS-domain protein 4 (ELK4) was a target of miR-3188. The effects of miR-3188 inhibition on ox-LDL-induced cell injury were markedly reversed by knockdown of ELK4. Besides, inhibition of miR-3188 enhanced ox-LDL-activated RhoA/ROCK pathway, while knockdown of ELK4 suppressed this pathway. Downregulation of miR-3188 may contribute to AS development via negatively regulating Lp-PLA2, targeting ELK4 and activating RhoA/ROCK pathway. miR-3188 may serve as a target for AS treatment.


Assuntos
Aterosclerose/patologia , MicroRNAs/fisiologia , Proteínas Elk-4 do Domínio ets/metabolismo , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , 1-Alquil-2-acetilglicerofosfocolina Esterase/metabolismo , Apoptose/fisiologia , Aterosclerose/genética , Estudos de Casos e Controles , Sobrevivência Celular/fisiologia , Citocinas/metabolismo , Regulação para Baixo , Feminino , Técnicas de Silenciamento de Genes , Humanos , Lipoproteínas LDL/metabolismo , Macrófagos/patologia , Masculino , MicroRNAs/genética , Pessoa de Meia-Idade , Proteínas Elk-4 do Domínio ets/genética
19.
PLoS One ; 11(3): e0150382, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26938874

RESUMO

The chimeric RNA, SLC45A3-ELK4, was found to be a product of cis-splicing between the two adjacent genes (cis-SAGe). Despite the biological and clinical significance of SLC45A3-ELK4, its generating mechanism has not been elucidated. It was shown in one cell line that the binding of transcription factor CTCF to the insulators located at or near the gene boundaries, inversely correlates with the level of the chimera. To investigate the mechanism of such cis-SAGe events, we sequenced potential regions that may play a role in such transcriptional read-through. We could not detect mutations at the transcription termination site, insulator sites, splicing sites, or within CTCF itself in LNCaP cells, thus suggesting a "soft-wired" mechanism in regulating the cis-SAGe event. To investigate the role CTCF plays in regulating the chimeric RNA expression, we compared the levels of CTCF binding to the insulators in different cell lines, as well as clinical samples. Surprisingly, we did not find an inverse correlation between CTCF level, or its bindings to the insulators and SLC45A3-ELK4 expression among different samples. However, in three prostate cancer cell lines, different environmental factors can cause the expression levels of the chimeric RNA to change, and these changes do inversely correlate with CTCF level, and/or its bindings to the insulators. We thus conclude that CTCF and its bindings to the insulators are not the primary reasons for differential SLC45A3-ELK4 expression in different cell lines, or clinical cases. However, they are the likely mechanism for the same cells to respond to different environmental cues, in order to regulate the expression of SLC45A3-ELK4 chimeric RNA. This response to different environmental cues is not general to other cis-SAGe events, as we only found one out of 16 newly identified chimeric RNAs showing a pattern similar to SLC45A3-ELK4.


Assuntos
Regulação Neoplásica da Expressão Gênica , Proteínas de Membrana Transportadoras/metabolismo , RNA/genética , Proteínas Repressoras/metabolismo , Proteínas Elk-4 do Domínio ets/metabolismo , Sequência de Bases , Sítios de Ligação , Fator de Ligação a CCCTC , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Células HEK293 , Humanos , Masculino , Dados de Sequência Molecular , Proteínas de Transporte de Monossacarídeos , Mutação , Mutação Puntual , Neoplasias da Próstata/metabolismo , Proteínas Repressoras/genética , Homologia de Sequência do Ácido Nucleico
20.
Oncogene ; 35(9): 1170-9, 2016 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-26028036

RESUMO

Cyclin-dependent kinase 2 (CDK2) is a known regulator in the cell cycle control of the G1/S and S/G2 transitions. However, the role of CDK2 in tumorigenesis is controversial. Evidence from knockout mice as well as colon cancer cell lines indicated that CDK2 is dispensable for cell proliferation. In this study, we found that ectopic CDK2 enhances Ras (G12V)-induced foci formation and knocking down CDK2 expression markedly decreases epidermal growth factor (EGF)-induced cell transformation mediated through the downregulation of c-fos expression. Interestingly, CDK2 directly phosphorylates ELK4 at Thr194 and Ser387 and regulates the ELK4 transcriptional activity, which serves as a mechanism to regulate c-fos expression. In addition, ELK4 is overexpressed in melanoma and knocking down the ELK4 or CDK2 expression significantly attenuated the malignant phenotype of melanoma cells. Taken together, our study reveals a novel function of CDK2 in EGF-induced cell transformation and the associated signal transduction pathways. This indicates that CDK2 is a useful molecular target for the chemoprevention and therapy against skin cancer.


Assuntos
Quinase 2 Dependente de Ciclina/genética , Melanoma/genética , Proteínas Proto-Oncogênicas c-fos/biossíntese , Proteínas Elk-4 do Domínio ets/biossíntese , Animais , Ciclo Celular/genética , Linhagem Celular , Proliferação de Células , Transformação Celular Neoplásica/genética , Quinase 2 Dependente de Ciclina/biossíntese , Fator de Crescimento Epidérmico/genética , Fator de Crescimento Epidérmico/metabolismo , Humanos , Melanoma/patologia , Camundongos , Fosforilação , Ativação Transcricional/genética , Proteínas Elk-4 do Domínio ets/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA