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











Base de dados
Intervalo de ano de publicação
1.
Cell Mol Life Sci ; 81(1): 253, 2024 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-38852108

RESUMO

Post-transcriptional regulation of cytokine/chemokine mRNA turnover is critical for immune processes and contributes to the mammalian cellular response to diverse inflammatory stimuli. The ubiquitous RNA-binding protein human antigen R (HuR) is an integral regulator of inflammation-associated mRNA fate. HuR function is regulated by various post-translational modifications that alter its subcellular localization and ability to stabilize target mRNAs. Both poly (ADP-ribose) polymerase 1 (PARP1) and p38 mitogen-activated protein kinases (MAPKs) have been reported to regulate the biological function of HuR, but their specific regulatory and crosstalk mechanisms remain unclear. In this study, we show that PARP1 acts via p38 to synergistically promote cytoplasmic accumulation of HuR and stabilization of inflammation-associated mRNAs in cells under inflammatory conditions. Specifically, p38 binds to auto-poly ADP-ribosylated (PARylated) PARP1 resulting in the covalent PARylation of p38 by PARP1, thereby promoting the retention and activity of p38 in the nucleus. In addition, PARylation of HuR facilitates the phosphorylation of HuR at the serine 197 site mediated by p38, which then increases the translocation of HuR to the cytoplasm, ultimately stabilizing the inflammation-associated mRNA expression at the post-transcriptional level.


Assuntos
Citoplasma , Proteína Semelhante a ELAV 1 , Inflamação , Poli(ADP-Ribose) Polimerase-1 , RNA Mensageiro , Proteínas Quinases p38 Ativadas por Mitógeno , Proteína Semelhante a ELAV 1/metabolismo , Proteína Semelhante a ELAV 1/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Humanos , Poli(ADP-Ribose) Polimerase-1/metabolismo , Poli(ADP-Ribose) Polimerase-1/genética , Citoplasma/metabolismo , Inflamação/metabolismo , Inflamação/genética , Inflamação/patologia , RNA Mensageiro/metabolismo , RNA Mensageiro/genética , Fosforilação , Regulação da Expressão Gênica , Animais , Poli ADP Ribosilação/genética , Células HEK293 , Núcleo Celular/metabolismo , Camundongos
2.
J Immunol ; 208(10): 2376-2389, 2022 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-35444028

RESUMO

Proinflammatory cytokines/chemokines are commonly regulated by RNA-binding proteins at posttranscriptional levels. Human Ag R (HuR)/embryonic lethal abnormal vision-like 1 (ELAVL1) is one of the well-characterized RNA-binding proteins that increases the stability of short-lived mRNAs, which encode proinflammatory mediators. HuR employs its nucleocytoplasmic shuttling sequence (HNS) domain, interacting with poly(ADP-ribose) polymerase 1 (PARP1), which accounts for the enhanced poly-ADP-ribosylation and cytoplasmic shuttling of HuR. Also by using its HNS domain, HuR undergoes dimerization/oligomerization, underlying the increased binding of HuR with proinflammatory cytokine/chemokine mRNAs and the disassociation of the miRNA-induced silencing complex from the targets. Therefore, competitively blocking the interactions of HuR with its partners may suppress proinflammatory mediator production. In this study, peptides derived from the sequence of the HuR-HNS domain were synthesized, and their effects on interfering HuR interacting with PARP1 and HuR itself were analyzed. Moreover, cell-penetrating TAT-HuR-HNS3 was delivered into human and mouse cells or administered into mouse lungs with or without exposure of TNF-α or LPS. mRNA levels of proinflammatory mediators as well as neutrophil infiltration were evaluated. We showed that TAT-HuR-HNS3 interrupts HuR-PARP1 interaction and therefore results in a lowered poly-ADP-ribosylation level and decreased cytoplasmic distribution of HuR. TAT-HuR-HNS3 also blocks HuR dimerization and promotes Argonaute 2-based miRNA-induced silencing complex binding to the targets. Moreover, TAT-HuR-HNS3 lowers mRNA stability of proinflammatory mediators in TNF-α-treated epithelial cells and macrophages, and it decreases TNF-α-induced inflammatory responses in lungs of experimental animals. Thus, TAT-HuR-HNS3 is a promising lead peptide for the development of inhibitors to treat inflammation-related diseases.


Assuntos
Peptídeos Penetradores de Células , Proteína Semelhante a ELAV 1/imunologia , MicroRNAs , Animais , Peptídeos Penetradores de Células/genética , Peptídeos Penetradores de Células/metabolismo , Peptídeos Penetradores de Células/farmacologia , Quimiocinas/genética , Citocinas/metabolismo , Proteínas ELAV/genética , Proteínas ELAV/metabolismo , Proteína Semelhante a ELAV 1/genética , Proteína Semelhante a ELAV 1/metabolismo , Expressão Gênica , Camundongos , MicroRNAs/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Fator de Necrose Tumoral alfa/metabolismo
3.
Cancer Lett ; 537: 215667, 2022 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-35398531

RESUMO

Formation of the premetastatic niche is triggered by primary tumors and contributes to cancer metastasis. Evidence indicating the roles of macrophages in metastatic niche formation and organ-specific metastatic tropism has been steadily accumulating. However, the role of tissue-resident macrophages in the establishment of the premetastatic niche is not clearly defined. Here, we report that alveolar macrophages (AMs), which are lung tissue-resident macrophages, play a critical role in initiating the recruitment of monocytic myeloid-derived suppressor cells (mo-MDSCs) and the subsequent premetastatic niche formation by increasing CCL12 expression. We found that CXCL10 can induce CCL12 expression by activating CXCR3 and TLR4 in AMs. CXCR3/TLR4 deficiency or inhibition of its activity reduces CCL12 expression in AMs and subsequent mo-MDSC recruitment to the premetastatic niche, thereby attenuating lung metastasis. In addition, Ube2o is a negative modulator of CXCL10-induced CCL12 expression. Downregulation of Ube2o in AMs under tumor conditions enhances TAK1-NF-κB/ERK/JNK signaling and CXCL10-induced CCL12 expression by promoting TRAF6 polyubiquitination and inhibiting DDX3X degradation. Targeting mo-MDSC recruitment via the CXCL10-CXCR3/TLR4-CCL12 axis in AMs may have therapeutic potential for suppressing lung metastasis.


Assuntos
Neoplasias Pulmonares , Células Supressoras Mieloides , Quimiocina CXCL10/metabolismo , Humanos , Pulmão/patologia , Neoplasias Pulmonares/metabolismo , Macrófagos Alveolares/patologia , Células Supressoras Mieloides/metabolismo , Metástase Neoplásica/patologia , Receptor 4 Toll-Like/metabolismo , Enzimas de Conjugação de Ubiquitina/metabolismo
4.
Cell Mol Life Sci ; 79(1): 60, 2022 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-35000037

RESUMO

Parthanatos is a form of regulated cell death involved in the pathogenesis of many diseases, particularly neurodegenerative disorders, such as Parkinson's disease, Alzheimer's disease, Huntington's disease, and amyotrophic lateral sclerosis. Parthanatos is a multistep cell death pathway cascade that involves poly (ADP-ribose) polymerase 1 (PARP-1) overactivation, PAR accumulation, PAR binding to apoptosis-inducing factor (AIF), AIF release from the mitochondria, nuclear translocation of the AIF/macrophage migration inhibitory factor (MIF) complex, and MIF-mediated large-scale DNA fragmentation. All the key players in the parthanatos pathway are pleiotropic proteins with diverse functions. An in-depth understanding of the structure-based activity of the key factors, and the biochemical mechanisms of parthanatos, is crucial for the development of drugs and therapeutic strategies. In this review, we delve into the key players of the parthanatos pathway and reveal the multiple levels of therapeutic opportunities for treating parthanatos-based pathogenesis.


Assuntos
Fragmentação do DNA , Oxirredutases Intramoleculares/metabolismo , Fatores Inibidores da Migração de Macrófagos/metabolismo , Doenças Neurodegenerativas/patologia , Parthanatos/fisiologia , Poli(ADP-Ribose) Polimerase-1/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Fator de Indução de Apoptose/metabolismo , Humanos , Mitocôndrias/metabolismo , Doenças Neurodegenerativas/tratamento farmacológico , Poli Adenosina Difosfato Ribose/metabolismo
5.
Cell Death Dis ; 13(1): 36, 2022 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-35013108

RESUMO

Tumors modify myeloid cell differentiation and induce an immunosuppressive microenvironment. Granulocytic myeloid-derived suppressor cells (G-MDSCs), the main subgroup of myeloid-derived suppressor cells (MDSCs), are immature myeloid cells (IMCs) with immunosuppressive activity and exist in tumor-bearing hosts. The reason why these cells diverge from a normal differentiation pathway and are shaped into immunosuppressive cells remains unclear. Here, we reported that the increase of granulocyte colony-stimulating factor (G-CSF) in mouse serum with tumor progression encouraged G-MDSCs to obtain immunosuppressive traits in peripheral blood through the PI3K-Akt/mTOR pathway. Importantly, we found that downregulation of type I interferon (IFN-I) signaling in G-MDSCs was a prerequisite for their immunosuppressive effects. Suppressor of cytokine signaling (SOCS1), the action of which is dependent on IFN-I signaling, inhibited the activation of the PI3K-Akt/mTOR pathway by directly interacting with Akt, indicating that the differentiation of immunosuppressive G-MDSCs involves a transition from immune activation to immune tolerance. Our study suggests that increasing IFN-I signaling in G-MDSCs may be a strategy for reprograming immunosuppressive myelopoiesis and slowing tumor progression.


Assuntos
Tolerância Imunológica , Interferon Tipo I/metabolismo , Células Supressoras Mieloides/imunologia , Neoplasias/imunologia , Animais , Progressão da Doença , Fator Estimulador de Colônias de Granulócitos/metabolismo , Camundongos , Células Supressoras Mieloides/metabolismo , Células Supressoras Mieloides/patologia , Mielopoese , Neoplasias/metabolismo , Neoplasias/patologia , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Proteína 1 Supressora da Sinalização de Citocina/metabolismo , Serina-Treonina Quinases TOR , Evasão Tumoral , Microambiente Tumoral/imunologia
6.
Mol Nutr Food Res ; 65(15): e2100096, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34061433

RESUMO

SCOPE: The beneficial effects of probiotics in reducing gastrointestinal inflammation and in preventing colorectal cancer have been reported, but the mechanism underlying the immunomodulatory effect of probiotics in inhibiting extra-intestinal tumor progression remains unclear. METHODS AND RESULTS: This study shows that probiotic supplementation attenuate lung metastasis of melanoma cells in mice. Feeding mice with VSL#3 probiotics change the composition and proportion of gut microbiota. The changes in gut bacteria composition, such as in the abundance of Lachnospiraceae, Streptococcus, and Lachnoclostridium, are associated with the production of short-chain fatty acids in the gut. The concentrations of propionate and butyrate are upregulated in gut and blood after feeding VSL#3, and the increase in propionate and butyrate levels promotes the expression of chemokine (C-C motif) ligand 20 (CCL20) in lung endothelial cells and the recruitment of T helper 17 (Th17) cells to the lungs via the CCL20/chemokine receptor 6 axis. The recruitment of Th17 cells decreases the number of tumor foci in lungs and attenuates the lung metastasis of melanoma cells in mice. CONCLUSIONS: The results provide new information on the role and mechanisms of action of probiotics in attenuating extra-intestinal tumor metastasis.


Assuntos
Butiratos/metabolismo , Neoplasias Pulmonares/secundário , Melanoma Experimental/patologia , Probióticos/farmacologia , Propionatos/metabolismo , Animais , Quimiocina CCL20/metabolismo , Suplementos Nutricionais , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Ácidos Graxos Voláteis/metabolismo , Microbioma Gastrointestinal/fisiologia , Neoplasias Pulmonares/dietoterapia , Neoplasias Pulmonares/patologia , Camundongos Endogâmicos C57BL , Células Th17
7.
Cell Mol Life Sci ; 78(4): 1817-1835, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32789690

RESUMO

Poly(ADP-ribosyl)ation (PARylation) is an important post-translational modification mainly catalyzed by poly-ADP-ribose polymerase 1 (PARP1). In addition to having important roles in DNA damage detection and repair, it functions in gene expression regulation, especially at the posttranscriptional level. Embryonic lethal abnormal vision-like 1/human antigen R (ELAVL/HuR), a canonical 3' untranslated region AU-rich element-binding protein, is a crucial mRNA-stabilizing protein that protects target mRNAs from RNA-destabilizing protein- or microRNA-induced silencing complex (miRISC)-mediated degradation. Additionally, in some cases, HuR itself either promotes or suppresses translation. Here, we demonstrated that in response to inflammatory stimuli, the PARylation of HuR, mostly at the conserved D226 site, by PARP1 increased the formation of the HuR oligomer/multimer, and HuR oligomerization promoted the disassociation of miRISC and stabilized the pro-inflammatory gene mRNAs. The prevention of PARP1 activation or HuR oligomerization attenuated lipopolysaccharide-induced inflammatory gene expression and the airway recruitment of neutrophils in mouse lungs. The present study verified a novel mechanism of PARP1 and HuR PARylation in the RNA stability regulation, increasing our understanding of how PARP1 regulates gene expression.


Assuntos
Proteína Semelhante a ELAV 1/genética , Inflamação/genética , Poli(ADP-Ribose) Polimerase-1/genética , Poli ADP Ribosilação/genética , Animais , Dano ao DNA/genética , Reparo do DNA/genética , Modelos Animais de Doenças , Regulação da Expressão Gênica/genética , Humanos , Inflamação/induzido quimicamente , Inflamação/patologia , Lipopolissacarídeos/toxicidade , Neutrófilos/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/genética , Estabilidade de RNA/genética , RNA Mensageiro/genética
8.
J Biosci ; 452020.
Artigo em Inglês | MEDLINE | ID: mdl-32098913

RESUMO

As a tumor suppressor, p53 preserves genomic integrity in eukaryotes. However, limited evidence is available for the p53 shuttling between the cytoplasm and nucleus. Previous studies have shown that ß-actin polymerization negatively regulates p53 nuclear import through its interaction with p53. In this study, we found that DNA damage induces both ß-actin and p53 accumulation in the nucleus. ß-actin knockdown impaired the nuclear transport of p53. Additionally, ß-actin could interact with p53 which was enhanced in response to genotoxic stress. Furthermore, N terminal deletion mutants of p53 shows reduced levels of association with ß-actin. We further identified Ser15, Thr18 and Ser20 of p53 are critical to the ß-actin: p53 interaction, which upon mutation into alanine abrogates the binding. Taken together, this study reveals that ß-actin regulates the nuclear import of p53 through protein-protein interaction.


Assuntos
Actinas/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Antineoplásicos Fitogênicos/farmacologia , Etoposídeo/farmacologia , Proteína Supressora de Tumor p53/metabolismo , Actinas/genética , Transporte Ativo do Núcleo Celular/fisiologia , Linhagem Celular Tumoral , Dano ao DNA , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Proteína Supressora de Tumor p53/genética
9.
Cell Prolif ; 53(3): e12780, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32031738

RESUMO

OBJECTIVES: RING finger protein 8 (RNF8) is an E3 ligase that plays an essential role in DSB repair. p53 is a well-established tumour suppressor and cellular gatekeeper of genome stability. This study aimed at investigating the functional correlations between RNF8 and p53 in DSB damage repair. MATERIALS AND METHODS: In this article, wild-type, knockout and shRNA-depleted HCT116 and U2OS cells were stressed, and the roles of RNF8 and p53 were examined. RT-PCR and Western blot were utilized to investigate the expression of related genes in damaged cells. Cell proliferation, apoptosis and neutral cell comet assays were applied to determine the effects of DSB damage on differently treated cells. DR-GFP, EJ5-GFP and LacI-LacO targeting systems, flow cytometry, mass spectrometry, IP, IF, GST pull-down assay were used to explore the molecular mechanism of RNF8 and p53 in DSB damage repair. RESULTS: We found that RNF8 knockdown increased cellular sensitivity to DSB damage and decreased cell proliferation, which was correlated with high expression of the p53 gene. RNF8 improved the efficiency of DSB repair by inhibiting the pro-apoptotic function of p53. We also found that RNF8 restrains cell apoptosis by inhibiting over-activation of ATM and subsequently reducing p53 acetylation at K120 through regulating Tip60. CONCLUSIONS: Taken together, these findings suggested that RNF8 promotes efficient DSB repair by inhibiting the pro-apoptotic activity of p53 through regulating the function of Tip60.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA , Proteínas de Ligação a DNA/metabolismo , Lisina Acetiltransferase 5/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Apoptose , Linhagem Celular Tumoral , Células HCT116 , Humanos
10.
Cells ; 8(9)2019 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-31500199

RESUMO

Poly(ADP-ribosyl)ation (PARylation) is an essential post-translational modification catalyzed by poly(ADP-ribose) polymerase (PARP) enzymes. Poly(ADP-ribose) polymerase 1 (PARP1) is a well-characterized member of the PARP family. PARP1 plays a crucial role in multiple biological processes and PARP1 activation contributes to the development of various inflammatory and malignant disorders, including lung inflammatory disorders, cardiovascular disease, ovarian cancer, breast cancer, and diabetes. In this review, we will focus on the role and molecular mechanisms of PARPs enzymes in inflammation- and metabolic-related diseases. Specifically, we discuss the molecular mechanisms and signaling pathways that PARP1 is associated with in the regulation of pathogenesis. Recently, increasing evidence suggests that PARP inhibition is a promising strategy for intervention of some diseases. Thus, our in-depth understanding of the mechanism of how PARPs are activated and how their signaling downstream effecters can provide more potential therapeutic targets for the treatment of the related diseases in the future is crucial.


Assuntos
Poli ADP Ribosilação/fisiologia , Poli(ADP-Ribose) Polimerases/metabolismo , Poli(ADP-Ribose) Polimerases/fisiologia , Animais , Humanos , Inflamação/metabolismo , Inflamação/fisiopatologia , Doenças Metabólicas/metabolismo , Doenças Metabólicas/fisiopatologia , Poli Adenosina Difosfato Ribose/metabolismo , Inibidores de Poli(ADP-Ribose) Polimerases , Poli(ADP-Ribose) Polimerases/genética , Processamento de Proteína Pós-Traducional , Transdução de Sinais
11.
J Immunol ; 203(6): 1521-1531, 2019 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-31399520

RESUMO

Poly(ADP-ribosyl)ation is a rapid and transient posttranslational protein modification mostly catalyzed by poly(ADP-ribose) polymerase-1 (PARP1). Fundamental roles of activated PARP1 in DNA damage repair and cellular response pathways are well established; however, the precise mechanisms by which PARP1 is activated independent of DNA damage, and thereby playing a role in expression of inflammatory genes, remain poorly understood. In this study, we show that, in response to LPS or TNF-α exposure, the nonreceptor tyrosine kinase c-Abl undergoes nuclear translocation and interacts with and phosphorylates PARP1 at the conserved Y829 site. Tyrosine-phosphorylated PARP1 is required for protein poly(ADP-ribosyl)ation of RelA/p65 and NF-κB-dependent expression of proinflammatory genes in murine RAW 264.7 macrophages, human monocytic THP1 cells, or mouse lungs. Furthermore, LPS-induced airway lung inflammation was reduced by inhibition of c-Abl activity. The present study elucidated a novel signaling pathway to activate PARP1 and regulate gene expression, suggesting that blocking the interaction of c-Abl with PARP1 or pharmaceutical inhibition of c-Abl may improve the outcomes of PARP1 activation-mediated inflammatory diseases.


Assuntos
Genes abl/genética , Inflamação/genética , Fosforilação/genética , Poli(ADP-Ribose) Polimerase-1/genética , Tirosina/genética , Animais , Linhagem Celular , Dano ao DNA/genética , Reparo do DNA/genética , Feminino , Expressão Gênica/genética , Humanos , Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/genética , Processamento de Proteína Pós-Traducional/genética , Células RAW 264.7 , Transdução de Sinais/genética , Células THP-1
12.
Cell Mol Life Sci ; 76(17): 3283-3299, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31055645

RESUMO

Poly(ADP-ribosyl)ation (PARylation) is an important post-translational modification in which an ADP-ribose group is transferred to the target protein by poly(ADP-riboses) polymerases (PARPs). Since the discovery of poly-ADP-ribose (PAR) 50 years ago, its roles in cellular processes have been extensively explored. Although research initially focused on the functions of PAR and PARPs in DNA damage detection and repair, our understanding of the roles of PARPs in various nuclear and cytoplasmic processes, particularly in gene expression, has increased significantly. In this review, we discuss the current advances in understanding the roles of PARylation with a particular emphasis in gene expression through RNA biogenesis and processing. In addition to updating PARP's significance in transcriptional regulation, we specifically focus on how PARPs and PARylation affect gene expression, especially inflammation-related genes, at the post-transcriptional levels by modulating RNA processing and degrading. Increasing evidence suggests that PARP inhibition is a promising treatment for inflammation-related diseases besides conventional chemotherapy for cancer.


Assuntos
Poli(ADP-Ribose) Polimerases/genética , RNA/metabolismo , Transporte Ativo do Núcleo Celular , Cromatina/metabolismo , Regulação da Expressão Gênica , Humanos , Inflamação/metabolismo , Inflamação/patologia , Poli(ADP-Ribose) Polimerases/metabolismo , Poliadenilação , RNA/genética , Splicing de RNA , Proteínas de Ligação a RNA/metabolismo
13.
J Cell Physiol ; 234(7): 11871-11881, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30478995

RESUMO

Acquired resistance to cytotoxic antineoplastic agents is a major clinical challenge in tumor therapy; however, the mechanisms involved are still poorly understood. In this study, we show that knockdown of CtIP, a corepressor of CtBP, promotes cell proliferation and alleviates G2/M phase arrest in etoposide (Eto)-treated HCT116 cells. Although the expression of p21 and growth arrest and DNA damage inducible α (GADD45a), which are important targets of p53, was downregulated in CtIP-deficient HCT116 cells, p53 deletion did not affect G2/M arrest after Eto treatment. In addition, the phosphorylation levels of Ser317 and Ser345 in Chk1 and of Ser216 in CDC25C were lower in CtIP-deficient HCT116 cells than in control cells after Eto treatment. Our results indicate that CtIP may enhance cell sensitivity to Eto by promoting G2/M phase arrest, mainly through the ATR-Chk1-CDC25C pathway rather than the p53-p21/GADD45a pathway. The expression of CtIP may be a useful biomarker for predicting the drug sensitivity of colorectal cancer cells.


Assuntos
Endodesoxirribonucleases/metabolismo , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Proteína Supressora de Tumor p53/efeitos dos fármacos , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Proteínas de Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p21/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Dano ao DNA/efeitos dos fármacos , Etoposídeo/farmacologia , Células HCT116 , Humanos , Pontos de Checagem da Fase M do Ciclo Celular/efeitos dos fármacos , Proteína Supressora de Tumor p53/genética
14.
Mol Cell Probes ; 39: 57-60, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29627626

RESUMO

Poly (ADP-ribose) polymerase 1 (PARP1) is a DNA damage sensor that catalyzes the poly (ADP-ribose) (PAR) onto a variety of target proteins, such as histones, DSB repair factors and PARP1 itself under consumption of NAD+. Besides, PARP1 can affect a variety of proteins in noncovalent modification manner to carry out specific cellular functions. Here, we established a method to generate non-radiolabeled free PAR by PARG moderately cleaving PAR from autoPARylated PARP1, and utilized dot-blot assay to determine the interaction between free PAR and interested proteins. The methods to generate free PAR and detect the noncovalent interactions between proteins and free PAR are nonradioactive and convenient, which will facilitate the studies to explore the significance of PAR reading in various biological processes.


Assuntos
Técnicas de Sonda Molecular , Poli Adenosina Difosfato Ribose/análise , Proteína Semelhante a ELAV 1/metabolismo , Humanos , Proteínas Recombinantes de Fusão/isolamento & purificação
15.
Nat Commun ; 8: 14632, 2017 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-28272405

RESUMO

Poly(ADP-ribosyl)ation (PARylation) is mainly catalysed by poly-ADP-ribose polymerase 1 (PARP1), whose role in gene transcription modulation has been well established. Here we show that, in response to LPS exposure, PARP1 interacts with the adenylateuridylate-rich element-binding protein embryonic lethal abnormal vision-like 1 (Elavl1)/human antigen R (HuR), resulting in its PARylation, primarily at site D226. PARP inhibition and the D226 mutation impair HuR's PARylation, nucleocytoplasmic shuttling and mRNA binding. Increases in mRNA level or stability of pro-inflammatory cytokines/chemokines are abolished by PARP1 ablation or inhibition, or blocked in D226A HuR-expressing cells. The present study demonstrates a mechanism to regulate gene expression at the post-transcriptional level, and suggests that blocking the interaction of PARP1 with HuR could be a strategy to treat inflammation-related diseases that involve increased mRNA stability.


Assuntos
Proteína Semelhante a ELAV 1/genética , Regulação da Expressão Gênica , Inflamação/genética , Macrófagos Peritoneais/imunologia , Poli(ADP-Ribose) Polimerase-1/genética , Processamento de Proteína Pós-Traducional/genética , RNA Mensageiro/metabolismo , Animais , Quimiocinas/imunologia , Citocinas/imunologia , Proteína Semelhante a ELAV 1/imunologia , Proteína Semelhante a ELAV 1/metabolismo , Células HEK293 , Humanos , Inflamação/imunologia , Lipopolissacarídeos/farmacologia , Macrófagos Peritoneais/efeitos dos fármacos , Camundongos , Mutação , Poli(ADP-Ribose) Polimerase-1/antagonistas & inibidores , Poli(ADP-Ribose) Polimerase-1/imunologia , Poli ADP Ribosilação , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Transporte Proteico , Células RAW 264.7 , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
Toxicol Lett ; 232(1): 1-9, 2015 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-25280774

RESUMO

Oxidative stress-induced DNA damage results in over-activation of poly(ADP-ribose) polymerase 1 (PARP1), leading to parthanatos, a newly discovered cell elimination pathway. Inhibition of PARP1-dependent cell death has shown to improve the outcome of diseases, including stroke, heart ischemia, and neurodegenerative diseases. In the present study we aimed to detect whether estrogen plays a protective role in inhibiting parthanatos. We utilized human mammary adenocarcinoma cells (MCF7) that abundantly express the estrogen receptor alpha and beta (ERα and ERß). Parthanatos was induced by challenging the cells with hydrogen peroxide (H2O2). Microscopic imaging and molecular biological techniques, such as Western blot analysis and RNA interference, were performed. The results showed 17ß estradiol (E2) protected MCF7 cells from PARP1-dependent cell death by decreasing protein PARylation, and AIF translocation into nuclei/nucleoli. Down-regulation of ERα expression by siRNA before E2 addition resulted in the failure of the E2-mediated inhibition of H2O2-induced protein PARylation and AIF nucleolar translocation. Together these data suggest that estrogen via its alpha-type receptor inhibits oxidative stress-induced, PARP1-dependent cell death. The present study provided us insight into how to apply hormone therapy in intervention of parthanatos-implicated ischemic and degenerative diseases.


Assuntos
Antioxidantes/farmacologia , Fator de Indução de Apoptose/metabolismo , Apoptose/efeitos dos fármacos , Neoplasias da Mama/metabolismo , Nucléolo Celular/efeitos dos fármacos , Estradiol/farmacologia , Receptor alfa de Estrogênio/agonistas , Estresse Oxidativo/efeitos dos fármacos , Poli(ADP-Ribose) Polimerase-1/metabolismo , Transporte Ativo do Núcleo Celular , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Nucléolo Celular/metabolismo , Nucléolo Celular/patologia , Citoproteção , Fragmentação do DNA , Relação Dose-Resposta a Droga , Ativação Enzimática , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Humanos , Peróxido de Hidrogênio/toxicidade , Células MCF-7 , Interferência de RNA , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Transfecção
17.
Acta Pharmacol Sin ; 35(4): 496-503, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24632845

RESUMO

AIM: Daidzein (4',7-dihydroxyisoflavone) is an isoflavone exiting in many herbs that has shown anti-inflammation activity. The aim of this study was to investigate the mechanism underlying its anti-inflammatory action in murine lung epithelial cells. METHODS: C57BL/6 mice were intranasally exposed to TNF-α to induce lung inflammation. The mice were injected with daidzein (400 mg/kg, ip) before TNF-α challenge, and sacrificed 12 h after TNF-α challenge, and lung tissues were collected for analyisis. In in vitro studies, murine MLE-12 epithelial cells were treated with TNF-α (20 ng/mL). The expression of pro-inflammatory chemokine Cxcl2 mRNA and NF-κB transcriptional activity were examined using real-time PCR and a dual reporter assay. Protein poly-adenosine diphosphate-ribosylation (PARylation) was detecyed using Western blotting and immunoprecipitation assays. RESULTS: Pretreatment of the mice with daidzein markedly attenuated TNF-α-induced lung inflammation, and inhibited Cxcl2 expression in lung tissues. Furthermore, daidzein (10 µmol/L) prevented TNF-α-induced increases in Cxcl2 expression and activity and NF-κB transcriptional activity, and markedly inhibited TNF-α-induced protein PARylation in MLE-12 cells in vitro. In MLE-12 cells co-transfected with the PARP-1 expression plasmid and NF-κB-luc (or Cxcl2-luc) reporter plasmid, TNF-α markedly increased NF-κB (or Cxcl2) activation, which were significantly attenuated in the presence of daidzein (or the protein PARylation inhibitor PJ 34). PARP-1 activity assay showed that daidzein (10 µmol/L) reduced the activity of PARP-1 by ∼75%. CONCLUSION: The anti-inflammatory action of daidzein in murine lung epithelial cells seems to be mediated via a direct interaction with PARP-1, which inhibits RelA/p65 protein PARylation required for the transcriptional modulation of pro-inflammatory chemokines such as Cxcl2.


Assuntos
Anti-Inflamatórios/farmacologia , Quimiocina CXCL2/metabolismo , Células Epiteliais/efeitos dos fármacos , Isoflavonas/farmacologia , Pulmão/efeitos dos fármacos , Pneumonia/prevenção & controle , Poli(ADP-Ribose) Polimerases/metabolismo , Transcrição Gênica/efeitos dos fármacos , Fator de Necrose Tumoral alfa , Animais , Linhagem Celular , Quimiocina CXCL2/genética , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Regulação para Baixo , Inibidores Enzimáticos/farmacologia , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Genes Reporter , Pulmão/imunologia , Pulmão/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Pneumonia/induzido quimicamente , Pneumonia/imunologia , Pneumonia/metabolismo , Poli(ADP-Ribose) Polimerase-1 , Inibidores de Poli(ADP-Ribose) Polimerases , Poli(ADP-Ribose) Polimerases/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo , Transfecção
18.
Hum Immunol ; 73(5): 439-47, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22391342

RESUMO

Poly(ADP-ribosyl)ation, like acetylation, methylation and phosphorylation, is one of the essential post-translational protein modifications. Poly(ADP-ribose) polymerase 1 (PARP-1), the best characterized member of the PARP family, catalyzes PAR formation, and has been implemented in the in vivo and in vitro inflammatory disease models. However, the exact signaling pathways leading to PARP-1 activation and the molecular mechanisms of activated PARP-1 signaling of inflammatory genes' expression remains to be further elucidated. In the present study, murine macrophages, in vitro stimulated with lipopolysaccharide (LPS), showed a profound activation of PARP-1, and PARP-1-dependent expression of mRNA for interleukin (IL)-1ß and IL-18 inflammatory cytokines. Immunoprecipitation assays showed that LPS stimulation enhanced the binding of PARP-1 with p65 (RelA) and poly(ADP-ribosyl)ation of p65, which might account for the upregulated transcription activity of nuclear factor (NF)-κB and the increased expression of proinflammatory genes. The application of various signal pathway inhibitors revealed that besides the canonical ROS-DNA damage signal, ERK pathway modulated the activation of PARP-1. ERK inhibitor blocked the interaction of PARP-1 with ERK1/2, phosphorylation of PARP-1, and poly(ADP-ribosyl)ation of p65, suggesting that ERK-dependent phosphorylation of PARP-1 regulates PARP-1 activity and NF-κB activation. Taken together, our results suggest that an ERK-PARP-1-RelA pathway in macrophages promote inflammatory progression in septic diseases.


Assuntos
Inflamação/metabolismo , Lipopolissacarídeos/farmacologia , Sistema de Sinalização das MAP Quinases/genética , Macrófagos/efeitos dos fármacos , Poli(ADP-Ribose) Polimerases/metabolismo , Animais , Linhagem Celular , Expressão Gênica/efeitos dos fármacos , Inflamação/genética , Inflamação/imunologia , Interleucina-18/biossíntese , Interleucina-18/imunologia , Interleucina-1beta/biossíntese , Interleucina-1beta/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosforilação , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/genética , Inibidores de Proteínas Quinases/farmacologia , Processamento de Proteína Pós-Traducional , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo , Ativação Transcricional/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA