Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 151
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Mol Cell ; 65(2): 296-309, 2017 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-28065600

RESUMO

In mammalian cells, histone deacetylase (HDAC) and Sirtuin (SIRT) are two families responsible for removing acetyl groups from acetylated proteins. Here, we describe protein deacetylation coupled with deacetylimination as a function of lysyl oxidase (LOX) family members. LOX-like 3 (Loxl3) associates with Stat3 in the nucleus to deacetylate and deacetyliminate Stat3 on multiple acetyl-lysine sites. Surprisingly, Loxl3 N-terminal scavenger receptor cysteine-rich (SRCR) repeats, rather than the C-terminal oxidase catalytic domain, represent the major deacetylase/deacetyliminase activity. Loxl3-mediated deacetylation/deacetylimination disrupts Stat3 dimerization, abolishes Stat3 transcription activity, and restricts cell proliferation. In Loxl3-/- mice, Stat3 is constitutively acetylated and naive CD4+ T cells are potentiated in Th17/Treg cell differentiation. When overexpressed, the SRCR repeats from other LOX family members can catalyze protein deacetylation/deacetylimination. Thus, our findings delineate a hitherto-unknown mechanism of protein deacetylation and deacetylimination catalyzed by lysyl oxidases.


Assuntos
Aminoácido Oxirredutases/metabolismo , Linfócitos T CD4-Positivos/enzimologia , Colite/enzimologia , Processamento de Proteína Pós-Traducional , Fator de Transcrição STAT3/metabolismo , Acetilação , Aminoácido Oxirredutases/deficiência , Aminoácido Oxirredutases/genética , Animais , Linfócitos T CD4-Positivos/imunologia , Catálise , Diferenciação Celular , Núcleo Celular/enzimologia , Proliferação de Células , Colite/genética , Colite/imunologia , Modelos Animais de Doenças , Genótipo , Células HEK293 , Células HeLa , Humanos , Células MCF-7 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Domínios Proteicos , Multimerização Proteica , Interferência de RNA , Fator de Transcrição STAT3/genética , Linfócitos T Reguladores/enzimologia , Linfócitos T Reguladores/imunologia , Células Th17/enzimologia , Células Th17/imunologia , Transcrição Gênica , Transfecção
2.
J Biol Chem ; 298(7): 102102, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35667440

RESUMO

Oxidative DNA damage contributes to aging and the pathogenesis of numerous human diseases including cancer. 8-hydroxyguanine (8-oxoG) is the major product of oxidative DNA lesions. Although OGG1-mediated base excision repair is the primary mechanism for 8-oxoG removal, DNA mismatch repair has also been implicated in processing oxidative DNA damage. However, the mechanism of the latter is not fully understood. Here, we treated human cells defective in various 8-oxoG repair factors with H2O2 and performed biochemical, live cell imaging, and chromatin immunoprecipitation sequencing analyses to determine their response to the treatment. We show that the mismatch repair processing of oxidative DNA damage involves cohesive interactions between mismatch recognition protein MutSα, histone mark H3K36me3, and H3K36 trimethyltransferase SETD2, which activates the ATM DNA damage signaling pathway. We found that cells depleted of MutSα or SETD2 accumulate 8-oxoG adducts and fail to trigger H2O2-induced ATM activation. Furthermore, we show that SETD2 physically interacts with both MutSα and ATM, which suggests a role for SETD2 in transducing DNA damage signals from lesion-bound MutSα to ATM. Consistently, MutSα and SETD2 are highly coenriched at oxidative damage sites. The data presented here support a model wherein MutSα, SETD2, ATM, and H3K36me3 constitute a positive feedback loop to help cells cope with oxidative DNA damage.


Assuntos
Reparo de Erro de Pareamento de DNA , Histona-Lisina N-Metiltransferase , Proteínas MutS , Estresse Oxidativo , Dano ao DNA , Código das Histonas , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/genética , Humanos , Peróxido de Hidrogênio/farmacologia , Proteínas MutS/genética , Proteínas MutS/metabolismo
3.
EMBO Rep ; 22(4): e51298, 2021 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-33594776

RESUMO

Notch signaling and epigenetic factors are known to play critical roles in regulating tissue homeostasis in most multicellular organisms, but how Notch signaling coordinates with epigenetic modulators to control differentiation remains poorly understood. Here, we identify heterochromatin protein 1c (HP1c) as an essential epigenetic regulator of gut homeostasis in Drosophila. Specifically, we observe that HP1c loss-of-function phenotypes resemble those observed after Notch signaling perturbation and that HP1c interacts genetically with components of the Notch pathway. HP1c represses the transcription of Notch target genes by directly interacting with Suppressor of Hairless (Su(H)), the key transcription factor of Notch signaling. Moreover, phenotypes caused by depletion of HP1c in Drosophila can be rescued by expressing human HP1γ, suggesting that HP1γ functions similar to HP1c in Drosophila. Taken together, our findings reveal an essential role of HP1c in normal development and gut homeostasis by suppressing Notch signaling.


Assuntos
Proteínas de Drosophila , Animais , Proteínas Cromossômicas não Histona/genética , Drosophila/genética , Proteínas de Drosophila/genética , Heterocromatina , Homeostase , Humanos , Receptores Notch/genética
4.
J Biol Chem ; 296: 100466, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33640455

RESUMO

DNA mismatch repair (MMR) maintains genome stability primarily by correcting replication errors. MMR deficiency can lead to cancer development and bolsters cancer cell resistance to chemotherapy. However, recent studies have shown that checkpoint blockade therapy is effective in MMR-deficient cancers, thus the ability to identify cancer etiology would greatly benefit cancer treatment. MutS homolog 2 (MSH2) is an obligate subunit of mismatch recognition proteins MutSα (MSH2-MSH6) and MutSß (MSH2-MSH3). Precise regulation of MSH2 is critical, as either over- or underexpression of MSH2 results in an increased mutation frequency. The mechanism by which cells maintain MSH2 proteostasis is unknown. Using functional ubiquitination and deubiquitination assays, we show that the ovarian tumor (OTU) family deubiquitinase ubiquitin aldehyde binding 1 (OTUB1) inhibits MSH2 ubiquitination by blocking the E2 ligase ubiquitin transfer activity. Depleting OTUB1 in cells promotes the ubiquitination and subsequent degradation of MSH2, leading to greater mutation frequency and cellular resistance to genotoxic agents, including the common chemotherapy agents N-methyl-N'-nitro-N-nitrosoguanidine and cisplatin. Taken together, our data identify OTUB1 as an important regulator of MSH2 stability and provide evidence that OTUB1 is a potential biomarker for cancer etiology and therapy.


Assuntos
Reparo de Erro de Pareamento de DNA/fisiologia , Enzimas Desubiquitinantes/metabolismo , Proteína 2 Homóloga a MutS/metabolismo , DNA/metabolismo , Dano ao DNA , Reparo de Erro de Pareamento de DNA/genética , Reparo do DNA , Proteínas de Ligação a DNA/metabolismo , Enzimas Desubiquitinantes/genética , Instabilidade Genômica , Células HEK293 , Células HeLa , Humanos , Proteína 2 Homóloga a MutS/genética , Ubiquitinação/genética
5.
Mol Cell ; 53(5): 752-65, 2014 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-24530303

RESUMO

Impaired phosphatase activity contributes to the persistent activation of STAT3 in tumors. Given that STAT family members with various or even opposite functions are often phosphorylated or dephosphorylated by the same enzymes, the mechanism for STAT3-specific dephosphorylation in cells remains largely unknown. Here, we report that GdX (UBL4A) promotes STAT3 dephosphorylation via mediating the interaction between TC45 (the nuclear isoform of TC-PTP) and STAT3 specifically. GdX stabilizes the TC45-STAT3 complex to bestow upon STAT3 an efficient dephosphorylation by TC45. Inasmuch, GdX suppresses tumorigenesis and tumor development by reducing the level of phospho-STAT3 (p-STAT3), whereas deletion of GdX results in a high level of p-STAT3 and accelerated colorectal tumorigenesis induced by AOM/DSS. Thus, GdX converts TC45, a nonspecific phosphatase, into a STAT3-specific phosphatase by bridging an association between TC45 and STAT3.


Assuntos
Carcinogênese , Regulação Neoplásica da Expressão Gênica , Proteína Tirosina Fosfatase não Receptora Tipo 2/química , Fator de Transcrição STAT3/química , Ubiquitinas/química , Animais , Células COS , Transformação Celular Neoplásica , Chlorocebus aethiops , Citocinas/metabolismo , Fibroblastos/metabolismo , Deleção de Genes , Humanos , Células MCF-7 , Melanoma Experimental , Camundongos , Camundongos Endogâmicos BALB C , Metástase Neoplásica , Transplante de Neoplasias , Fosforilação , Ligação Proteica , Ubiquitinas/genética
6.
Br J Cancer ; 124(8): 1437-1448, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33531691

RESUMO

BACKGROUND: Signal transducer and activator of transcription 3 (STAT3) has been shown to upregulate gene transcription during tumorigenesis. However, how STAT3 initiates transcription remains to be exploited. This study is to reveal the role of CREPT (cell cycle-related and elevated-expression protein in tumours, or RPRD1B) in promoting STAT3 transcriptional activity. METHODS: BALB/c nude mice, CREPT overexpression or deletion cells were employed for the assay of tumour formation, chromatin immunoprecipitation, assay for transposase-accessible chromatin using sequencing. RESULTS: We demonstrate that CREPT, a recently identified oncoprotein, enhances STAT3 transcriptional activity to promote tumorigenesis. CREPT expression is positively correlated with activation of STAT3 signalling in tumours. Deletion of CREPT led to a decrease, but overexpression of CREPT resulted in an increase, in STAT3-initiated tumour cell proliferation, colony formation and tumour growth. Mechanistically, CREPT interacts with phosphorylated STAT3 (p-STAT3) and facilitates p-STAT3 to recruit p300 to occupy at the promoters of STAT3-targeted genes. Therefore, CREPT and STAT3 coordinately facilitate p300-mediated acetylation of histone 3 (H3K18ac and H3K27ac), further augmenting RNA polymerase II recruitment. Accordingly, depletion of p300 abolished CREPT-enhanced STAT3 transcriptional activity. CONCLUSIONS: We propose that CREPT is a co-activator of STAT3 for recruiting p300. Our study provides an alternative strategy for the therapy of cancers related to STAT3.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Transformação Celular Neoplásica/patologia , Proteína p300 Associada a E1A/metabolismo , Proteínas de Neoplasias/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Animais , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Feminino , Células HEK293 , Humanos , Células MCF-7 , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Células NIH 3T3 , Proteínas de Neoplasias/genética , Transplante de Neoplasias , Fosforilação , Transcrição Gênica
7.
Amino Acids ; 53(8): 1197-1209, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34223992

RESUMO

Riboflavin is an essential micronutrient for normal cellular growth and function. Lack of dietary riboflavin is associated with an increased risk for esophageal squamous cell carcinoma (ESCC). Previous studies have identified that the human riboflavin transporter SLC52A3a isoform (encoded by SLC52A3) plays a prominent role in esophageal cancer cell riboflavin transportation. Furthermore, SLC52A3 gene single nucleotide polymorphisms rs3746804 (T>C, L267P) and rs3746803 (C >T, T278M) are associated with ESCC risk. However, whether SLC52A3a (p.L267P) and (p.T278M) act in riboflavin transportation in esophageal cancer cell remains inconclusive. Here, we constructed the full-length SLC52A3a protein fused to green fluorescent protein (GFP-SLC52A3a-WT and mutants L267P, T278M, and L267P/T278M). It was confirmed by immunofluorescence-based confocal microscopy that SLC52A3a-WT, L267P, T278M, and L267P/T278M expressed in cell membrane, as well as in a variety of intracellular punctate structures. The live cell confocal imaging showed that SLC52A3a-L267P and L267P/T278M increased the intracellular trafficking of SLC52A3a in ESCC cells. Fluorescence recovery after photobleaching of GFP-tagged SLC52A3a meant that intracellular trafficking of SLC52A3a-L267P and L267P/T278M was rapid dynamics process, leading to its stronger ability to transport riboflavin. Taken together, the above results indicated that the rs3746804 (p.L267P) polymorphism promoted intracellular trafficking of SLC52A3a and riboflavin transportation in ESCC cells.


Assuntos
Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas do Esôfago/metabolismo , Proteínas de Membrana Transportadoras/genética , Polimorfismo de Nucleotídeo Único , Riboflavina/metabolismo , Transporte Biológico , Linhagem Celular Tumoral , Neoplasias Esofágicas/patologia , Carcinoma de Células Escamosas do Esôfago/patologia , Exoma , Proteínas de Fluorescência Verde/genética , Humanos , Reação em Cadeia da Polimerase/métodos
8.
J Biol Chem ; 294(17): 7037-7045, 2019 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-30858175

RESUMO

Proliferating cell nuclear antigen (PCNA) and its posttranslational modifications regulate DNA metabolic reactions, including DNA replication and repair, at replication forks. PCNA phosphorylation at Tyr-211 (PCNA-Y211p) inhibits DNA mismatch repair and induces misincorporation during DNA synthesis. Here, we describe an unexpected role of PCNA-Y211p in cancer promotion and development. Cells expressing phosphorylation-mimicking PCNA, PCNA-Y211D, show elevated hallmarks specific to the epithelial-mesenchymal transition (EMT), including the up-regulation of the EMT-promoting factor Snail and the down-regulation of EMT-inhibitory factors E-cadherin and GSK3ß. The PCNA-Y211D-expressing cells also exhibited active cell migration and underwent G2/M arrest. Interestingly, all of these EMT-associated activities required the activation of ATM and Akt kinases, as inactivating these protein kinases by gene knockdown or inhibitors blocked EMT-associated signaling and cell migration. We concluded that PCNA phosphorylation promotes cancer progression via the ATM/Akt/GSK3ß/Snail signaling pathway. In conclusion, this study identifies a novel PCNA function and reveals the molecular basis of phosphorylated PCNA-mediated cancer development and progression.


Assuntos
Neoplasias/patologia , Antígeno Nuclear de Célula em Proliferação/metabolismo , Transdução de Sinais , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Movimento Celular , Progressão da Doença , Transição Epitelial-Mesenquimal , Glicogênio Sintase Quinase 3 beta/metabolismo , Células HeLa , Humanos , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fatores de Transcrição da Família Snail/metabolismo
9.
J Cell Biochem ; 121(2): 1541-1551, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31535402

RESUMO

Mesenchymal stem cells (MSCs) have been proved to exert anti-inflammatory effects and regulate immune reactions. Traditional Chinese medicine (TCM), qi-fang-bi-min-tang, is effective for some patients with allergic diseases. However, it remains unclear whether MSCs combined with TCM could benefit the treatment of allergic rhinitis (AR). In this study, we reported an additional effect of TCM (qi-fang-bi-min-tang) on the therapy of AR under MSCs treatment. Intriguingly, we observed that TCM-treated MSCs significantly inhibited the symptoms of AR and reduced the pathological changes of nasal mucosa in ovalbumin (OVA)-induced rats. The expression levels of interferon Î³ (IFN-γ), interleukin-17 (IL-17), and IL-4 were significantly decreased in the plasma of AR rats after injection of TCM-treated MSCs. TCM-treated MSCs reduced the levels of histamine secreted by mast cells and immunoglobulin E (IgE) secreted by plasma cells. In addition, we found that MSCs combined with TCM had a better therapeutic effect than TCM alone on AR in an OVA-induced mouse model. After OVA induction, MSCs combined with TCM significantly reduced the ratio of T helper type 1 (Th1), Th2, and Th17, but increased the proportion of Treg in the spleen of mice. Consistently, the expression levels of IFN-γ, IL-4, and IL-17 were significantly decreased, but transforming growth factor-ß1 was significantly increased in the plasma of AR mice after treated with TCM and MSCs. Our results from both rats and mice indicated that the effects of TCM combined with MSCs on the AR might be through regulating the secretion of Th1, Th2, and Th17 cytokines. This study suggested that TCM (qi-fang-bi-min-tang)-treated MSCs could be used in the clinical therapy of AR.


Assuntos
Medicamentos de Ervas Chinesas/farmacologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/imunologia , Rinite Alérgica/terapia , Aloenxertos , Animais , Citocinas/imunologia , Masculino , Células-Tronco Mesenquimais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Ratos , Ratos Sprague-Dawley , Rinite Alérgica/imunologia , Rinite Alérgica/patologia , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/patologia
10.
J Hum Genet ; 65(11): 1039-1043, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32655147

RESUMO

Meckel-Gruber syndrome (MKS) is a clinically and genetically heterogeneous ciliopathy characterized by a triad of occipital encephalocele, polycystic kidneys, and postaxial polydactyly. Pathogenesis of MKS is related to dysfunction of primary cilia. However, reports on MKS caused by Tectonic2 (TCTN2) mutations are scanty whilst. There is no direct evidence of ciliogenesis in such MKS patients. Here, we identified two novel nonsense variants of TCTN2 (c.343G > T, p.E115*; c.1540C > T, p.Q514*) in a Chinese MKS fetus. Compared to reported TCTN2-causing MKS patients, our case represented an endocardial pad defect, which was not reported previously. We also found primary cilia protruded normally from the surface of epithelial cells in the affected fetal kidney tubules compared to controls, indicating TCTN2 is not necessary for ciliogenesis in the kidney. To our knowledge, this is the first case of MKS fetus caused by TCTN2 mutations from China.


Assuntos
Transtornos da Motilidade Ciliar/genética , Encefalocele/genética , Predisposição Genética para Doença , Rim/metabolismo , Proteínas de Membrana/genética , Doenças Renais Policísticas/genética , Retinose Pigmentar/genética , China , Transtornos da Motilidade Ciliar/patologia , Códon sem Sentido/genética , Encefalocele/patologia , Feminino , Feto/patologia , Dedos/anormalidades , Heterogeneidade Genética , Humanos , Rim/patologia , Masculino , Linhagem , Doenças Renais Policísticas/patologia , Polidactilia , Retinose Pigmentar/patologia , Dedos do Pé/anormalidades
11.
FASEB J ; 33(7): 8375-8385, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31002527

RESUMO

Rheumatoid arthritis (RA) is an autoimmune disease associated with synovial hyperplasia and bone and cartilage destruction. T cells, notably T helper (Th)-1 and Th17 cells, play a critical role in the pathologic process of RA. However, it remains unclear how Th1 and Th17 cells are regulated during RA. In this study, we report that the small ubiquitin-like protein X-linked gene in the G6PD cluster at Xq28 (GdX) regulates the balance of Th17 and regulatory T (Treg) cells during collagen-induced arthritis (CIA). We discovered that the splenocytes of GdX-knockout (KO) mice were insensitive to T-cell stimulants. Correspondingly, GdX-KO mice showed alleviative Th1-mediated delayed-type hypersensitivity and were resistant to CIA compared with wild-type mice. GdX-KO mice showed fewer swollen paws, lower serum proinflammatory cytokine and anti-collagen IgG levels, and decreased synovial hyperplasia. Mechanistically, we observed that deletion of GdX decreased the transcription of proinflammatory cytokines and impaired the Th1 and Th17 differentiation but increased the Treg cell proliferation. Consistently, deletion of GdX decreased the transcription level of T-cell-specific T-box transcription factor and RAR-related orphan receptor-γ transcription factor but increased that of forkhead box P3 after being challenged with type-II collagen. These findings suggested that GdX functions as an important regulator of Th1 or Th17 and Treg cell balance during the inflammatory responses. Therefore, GdX may be a potential target for the therapy of RA.-Fu, Y., Liu, S., Wang, Y., Ren, F., Fan, X., Liang, J., Liu, C., Li, J., Ju, Y., Chang, Z. GdX/UBL4A-knockout mice resist collagen-induced arthritis by balancing the population of Th1/Th17 and regulatory T cells.


Assuntos
Artrite Experimental/enzimologia , Linfócitos T Reguladores/enzimologia , Células Th1/enzimologia , Células Th17/enzimologia , Ubiquitinas/deficiência , Animais , Artrite Experimental/genética , Artrite Experimental/patologia , Citocinas/genética , Citocinas/metabolismo , Masculino , Camundongos , Camundongos Knockout , Células Th1/patologia , Células Th17/patologia , Transcrição Gênica , Ubiquitinas/metabolismo
12.
J Biol Chem ; 293(23): 8886-8899, 2018 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-29685889

RESUMO

The tumor suppressor phosphatase and tensin homolog (PTEN) plays a central role in regulating phosphatidylinositol 3-kinase (PI3K) signaling, and its gene is very frequently mutated in various human cancers. Numerous studies have revealed that PTEN levels are tightly regulated by both transcriptional and posttranslational modifications, with especially ubiquitylation significantly regulating PTEN protein levels. Although several ubiquitin ligases have been reported to mediate PTEN ubiquitylation in vitro, the ubiquitin ligase that promotes PTEN degradation in vivo has not been reported. Here we took advantage of specific knockout mouse models to demonstrate that WW domain-containing E3 ubiquitin protein ligase 2 (WWP2) promotes PTEN degradation under physiological conditions, whereas another ubiquitin ligase, carboxyl terminus of Hsp70-interacting protein (CHIP), had no such effect. WWP2 knockout mice exhibited reduced body size, elevated PTEN protein levels, and reduced phosphorylation levels of the serine/threonine kinase and PTEN target AKT. In contrast, we observed no elevation of PTEN protein levels in CHIP knockout tissues and mouse embryonic fibroblasts. Furthermore, PTEN protein levels in CHIP/WWP2 double knockout mice were very similar to those in WWP2 single knockout mice and significantly higher than in WT and CHIP knockout mice. Our results demonstrate that WWP2, rather than CHIP, is an ubiquitin ligase that promotes PTEN degradation in vivo Considering PTEN's significant role in tumor development, we propose that WWP2 may be a potential target for fine-tuning PTEN levels in anticancer therapies.


Assuntos
PTEN Fosfo-Hidrolase/metabolismo , Proteólise , Ubiquitina-Proteína Ligases/metabolismo , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , PTEN Fosfo-Hidrolase/genética , Fosfatidilinositol 3-Quinases/metabolismo , Estabilidade Proteica , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Ubiquitina-Proteína Ligases/genética
13.
J Biol Chem ; 293(20): 7618-7628, 2018 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-29618509

RESUMO

We previously demonstrated that p15RS, a newly discovered tumor suppressor, inhibits Wnt/ß-catenin signaling by interrupting the formation of ß-catenin·TCF4 complex. However, it remains unclear how p15RS helps exert such an inhibitory effect on Wnt signaling based on its molecular structure. In this study, we reported that dimerization of p15RS is required for its inhibition on the transcription regulation of Wnt-targeted genes. We found that p15RS forms a dimer through a highly conserved leucine zipper-like motif in the coiled-coil terminus domain. In particular, residues Leu-248 and Leu-255 were identified as being responsible for p15RS dimerization, as mutation of these two leucines into prolines disrupted the homodimer formation of p15RS and weakened its suppression of Wnt signaling. Functional studies further confirmed that mutations of p15RS at these residues results in diminishment of its inhibition on cell proliferation and tumor formation. We therefore concluded that dimerization of p15RS governed by the leucine zipper-like motif is critical for its inhibition of Wnt/ß-catenin signaling and tumorigenesis.


Assuntos
Regulação Neoplásica da Expressão Gênica , Zíper de Leucina , Melanoma/prevenção & controle , Proteínas Repressoras/química , Animais , Apoptose , Proliferação de Células , Feminino , Melanoma/metabolismo , Melanoma/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Multimerização Proteica , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Fator de Transcrição 4/antagonistas & inibidores , Fator de Transcrição 4/genética , Fator de Transcrição 4/metabolismo , Células Tumorais Cultivadas , Via de Sinalização Wnt , beta Catenina/antagonistas & inibidores , beta Catenina/metabolismo
14.
Dev Biol ; 424(1): 40-49, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28232075

RESUMO

Drosophila ovary is recognized as one of the best model systems to study stem cell biology in vivo. We had previously identified an autonomous role of the histone H1 in germline stem cell (GSC) maintenance. Here, we found that histone H1 depletion in escort cells (ECs) resulted in an increase of spectrosome-containing cells (SCCs), an ovary tumor-like phenotype. Further analysis showed that the Dpp pathway is excessively activated in these SCC cells, while the expression of bam is attenuated. In the H1-depleted ECs, both transposon activity and DNA damage had increased dramatically, followed by EC apoptosis, which is consistent with the role of H1 in other somatic cells. Surprisingly, H1-depleted ECs acquired cap cell characteristics including dpp expression, and the resulting abnormal Dpp level inhibits SCC further differentiation. Most interestingly, double knockdown of H1 and dpp in ECs can reduce the number of SCCs to the normal level, indicating that the additional Dpp secreted by ECs contributes to the germline tumor. Taken together, our findings indicate that histone H1 is an important epigenetic factor in controlling EC characteristics and a key suppressor of germline tumor.


Assuntos
Drosophila melanogaster/citologia , Drosophila melanogaster/metabolismo , Células Germinativas/metabolismo , Células Germinativas/patologia , Histonas/metabolismo , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Animais , Apoptose , Contagem de Células , Dano ao DNA , Elementos de DNA Transponíveis/genética , Feminino , Técnicas de Silenciamento de Genes , Modelos Biológicos , Fenótipo , Transdução de Sinais , Transcrição Gênica , Regulação para Cima
15.
J Cell Biochem ; 119(1): 1083-1092, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28695988

RESUMO

The CREPT (cell cycle-related and expression elevated protein in tumor, also known as RPRD1B) and p15RS (p15INK4b -related sequence, also known as RPRD1A) have been shown to regulate cell proliferation and alter the cell cycle through Wnt/ß-catenin pathway downstream genes in human. Although several studies have revealed the mechanism by which CREPT and p15RS regulate cell proliferation in human and mammals, it is still unclear how these genes function in poultry. In order to determine the function of CREPT and p15RS in chicken, we examined the expression of CREPT and p15RS in a variety of chicken tissues and DF-1 cells. Then, we determined the effect of overexpression or depletion of CREPT or p15RS, by transiently transfecting chicken DF-1 cells with overexpression and short hairpin RNA (shRNA) vectors respectively, on the regulation of cell proliferation. The results showed that CREPT and p15RS had different expression patterns and opposite effects on the cell cycling and proliferation. Knockdown of p15RS expression or overexpression of CREPT facilitated cell proliferation by promoting the cell-cycle transition from G0/G1 to S-phase and G2/M, whereas knockdown of CREPT or overexpression of p15RS inhibited cell proliferation. Mechanistically, CREPT and p15RS control DF-1 cell proliferation by regulating the expression of Wnt/ß-catenin pathway downstream regulatory genes, including ß-catenin, TCF4, and Cyclin D1. In conclusion, CREPT and p15RS regulate cell proliferation and the cell-cycle transition in chicken DF-1 cells by regulating the transcription of Wnt/ß-catenin pathway downstream regulatory genes.


Assuntos
Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Transcrição Gênica , Via de Sinalização Wnt , Animais , Ciclo Celular , Proteínas de Ciclo Celular/química , Linhagem Celular , Proliferação de Células , Galinhas , Humanos , Modelos Moleculares , Mapas de Interação de Proteínas , Distribuição Tecidual
16.
Mol Carcinog ; 57(10): 1408-1420, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29938829

RESUMO

CREPT (Cell-cycle-related and expression-elevated protein in tumor)/RPRD1B, a novel protein that enhances the transcription of Cyclin D1 to promote cell proliferation during tumorigenesis, was demonstrated highly expressed in most of tumors. However, it remains unclear how CREPT is regulated in colorectal cancers. In this study, we report that miR-383 negatively regulates CREPT expression. We observed that CREPT was up-regulated but the expression of miR-383 was down regulated in both colon cancer cell lines and colon tumor tissues. Intriguingly, we found that enforced expression of miR-383 inhibited the expression of CREPT at both the mRNA and protein level. Using a luciferase reporter, we showed that miR-383 targeted the 3'-UTR of CREPT mRNA directly. Consistently we observed that over expression of miR-383 shortened the half-life of CREPT mRNA in varieties of colorectal cancer cells. Furthermore, restoration of miR-383 inhibited cell growth and colony formation of colon cancer cells accompanied by inhibition of expression of CREPT and related downstream genes. Finally, we demonstrated that stable over expression of miR-383 in colon cancer cells decreased the growth of the tumors. Our results revealed that the abundant expression of CREPT in colorectal cancers is attributed to the decreased level of miR-383. This study shed a new light on the potential therapeutic therapy strategy for colorectal cancers using introduced miRNA.


Assuntos
Proteínas de Ciclo Celular/genética , Neoplasias Colorretais/genética , Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor , MicroRNAs/genética , Proteínas de Neoplasias/genética , Regiões 3' não Traduzidas/genética , Idoso , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/genética , Neoplasias Colorretais/metabolismo , Feminino , Células HCT116 , Células HEK293 , Células HT29 , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas de Neoplasias/metabolismo , Estabilidade de RNA/genética
18.
J Biol Chem ; 290(2): 861-71, 2015 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-25378394

RESUMO

TNF receptor 2 (TNFR2) exerts diverse roles in the pathogenesis of inflammatory and autoimmune diseases. Here, we report that TNFR2 but not TNFR1 forms a heteromer with interleukin-17 receptor D (IL-17RD), also named Sef, to activate NF-κB signaling. TNFR2 associates with IL-17RD, leading to mutual receptor aggregation and TRAF2 recruitment, which further activate the downstream cascade of NF-κB signaling. Depletion of IL-17RD impaired TNFR2-mediated activation of NF-κB signaling. Importantly, IL-17RD was markedly increased in renal tubular epithelial cells in nephritis rats, and a strong interaction of TNFR2 and IL-17RD was observed in the renal epithelia. The IL-17RD·TNFR2 complex in activation of NF-κB may explain the role of TNFR2 in inflammatory diseases including nephritis.


Assuntos
Inflamação/metabolismo , NF-kappa B/metabolismo , Nefrite/metabolismo , Receptores de Interleucina-17/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Animais , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Humanos , Inflamação/etiologia , Inflamação/patologia , Túbulos Renais Distais/metabolismo , Túbulos Renais Distais/patologia , NF-kappa B/genética , Nefrite/etiologia , Nefrite/patologia , Multimerização Proteica , Ratos , Receptores de Interleucina-17/química , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/química , Transdução de Sinais/genética , Ativação Transcricional/genética
19.
J Biol Chem ; 290(15): 9701-13, 2015 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-25697359

RESUMO

We previously reported that p15RS (p15INK4b-related sequence), a regulation of nuclear pre-mRNA domain containing protein, inhibited Wnt signaling by interrupting the formation of the ß-catenin·TCF4 complex. However, how p15RS functions as an intrinsic repressor to repress transcription remains unclear. In this study, we show that p15RS, through a specific interaction with HDAC2 (histone deacetylase 2), a deacetylase that regulates gene transcription, maintains histone H3 in a deacetylated state in the promoter region of Wnt-targeted genes where ß-catenin·TCF4 is bound. We observed that histone deacetylase inhibitors impair the ability of p15RS in inhibiting Wnt/ß-catenin signaling. Depletion of HDAC2 markedly disabled p15RS inhibition of Wnt/ß-catenin-mediated transcription. Interestingly, overexpression of p15RS decreases the level of acetylated histone H3 in the c-MYC promoter. Finally, we demonstrate that p15RS significantly enhances the association of HDAC2 and TCF4 and enhances the occupancy of HDAC2 to DNA, resulting in the deacetylation of histone H3 and the failure of ß-catenin interaction. We propose that p15RS acts as an intrinsic transcriptional repressor for Wnt/ß-catenin-mediated gene transcription at least partially through recruiting HDAC2 to occupy the promoter and maintaining deacetylated histone H3.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Histona Desacetilase 2/metabolismo , Proteínas Repressoras/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo , Acetilação , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Western Blotting , Proteínas de Ciclo Celular/genética , Proliferação de Células/genética , Ciclina D1/genética , Ciclina D1/metabolismo , Expressão Gênica , Células HEK293 , Histona Desacetilase 2/genética , Histonas/metabolismo , Humanos , Células MCF-7 , Microscopia Confocal , Regiões Promotoras Genéticas/genética , Ligação Proteica , Proteínas Proto-Oncogênicas c-myc/genética , Interferência de RNA , Proteínas Repressoras/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição 4 , Fatores de Transcrição/genética , beta Catenina/genética
20.
J Cell Sci ; 127(Pt 12): 2761-70, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24727614

RESUMO

Protein tyrosine phosphatases (PTPs) are involved in hematopoiesis, but the function of many PTPs is not well characterized in vivo. Here, we have identified Ptpn9a, an ortholog of human PTPN9, as a crucial regulator of erythroid cell development in zebrafish embryos. ptpn9a, but not ptpn9b, was expressed in the posterior lateral plate mesoderm and intermediate cell mass - two primitive hematopoietic sites during zebrafish embryogenesis. Morpholino-mediated knockdown of ptpn9a caused erythrocytes to be depleted by inhibiting erythroid cell maturation without affecting erythroid proliferation and apoptosis. Consistently, both dominant-negative PTPN9 (with mutation C515S) and siRNA against PTPN9 inhibited erythroid differentiation in human K562 cells. Mechanistically, depletion of ptpn9 in zebrafish embryos in vivo or in K562 cells in vitro increased phosphorylated STAT3, and the hyper-phosphorylated STAT3 entrapped and prevented the transcription factors GATA1 and ZBP-89 (also known as ZNF148) from regulating erythroid gene expression. These findings imply that PTPN9 plays an important role in erythropoiesis by disrupting an inhibitory complex of phosphorylated STAT3, GATA1 and ZBP-89, providing new cellular and molecular insights into the role of ptpn9a in developmental hematopoiesis.


Assuntos
Células Eritroides/enzimologia , Processamento de Proteína Pós-Traducional , Proteínas Tirosina Fosfatases não Receptoras/fisiologia , Fator de Transcrição STAT3/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra/fisiologia , Animais , Embrião não Mamífero/citologia , Embrião não Mamífero/enzimologia , Eritropoese , Fator de Transcrição GATA1/metabolismo , Gastrulação , Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Células K562 , Fosforilação , Fatores de Transcrição/metabolismo , Peixe-Zebra/embriologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA