Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Publication year range
1.
Histol Histopathol ; 39(4): 511-523, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37728155

ABSTRACT

BACKGROUND: Nasopharyngeal carcinoma (NPC) is characterized by a highly suppressive microenvironment that protects tumor cells against immune attack and facilitates tumor progression. MELK is upregulated in various tumors, whereas its function in the immune escape remains largely unknown. In this study, we investigated the role of MELK during immune escape in NPC. METHODS: Differentially expressed genes were filtered using GEO datasets and PPI network analysis. NPC cell colony formation and motility were examined, and the impact of CD8⁺ T cells on NPC cells was evaluated. A xenograft model was constructed to detect the growth of tumor cells and the T-cell phenotype of tumor infiltration. ChIP-qPCR and dual-luciferase assays were used to verify the transcriptional regulation of MELK by EP300/E2F1. FINDINGS: MELK was overexpressed in NPC, and sh-MELK suppressed the clonogenic ability, migration, and invasion of NPC cells and promoted the killing effects of CD8⁺ T cells. These in vitro findings were reproduced in vivo. EP300 synergized E2F1 to regulate the transcription of MELK in NPC cells. Loss of EP300 or E2F1 reverted the malignant phenotype of NPC cells and promoted the immune effect of CD8⁺ T cells. MELK further suppressed the immune effect of CD8⁺ T cells in the presence of sh-E2F1. INTERPRETATION: EP300 coordinated with E2F1 to promote the transcription of MELK which promoted the growth of NPC cells and repressed the killing effect of CD8⁺ T cells. Blockage of MELK may be a potential way to suppress the immune escape of NPC cells.


Subject(s)
Nasopharyngeal Neoplasms , Humans , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Cell Movement , Tumor Microenvironment , Protein Serine-Threonine Kinases/genetics , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , E2F1 Transcription Factor/pharmacology , E1A-Associated p300 Protein/genetics , E1A-Associated p300 Protein/pharmacology
2.
Biomater Sci ; 10(21): 6258-6266, 2022 Oct 25.
Article in English | MEDLINE | ID: mdl-36111777

ABSTRACT

Apoptosis of cardiomyocytes is a critical outcome of myocardial ischemia-reperfusion injury (MIRI), which leads to the permanent impairment of cardiac function. Upregulated E2F1 is implicated in inducing cardiomyocyte apoptosis, and thus intervention of the E2F1 signaling pathway via RNA interference may hold promising potential for rescuing the myocardium from MIRI. To aid efficient E2F1 siRNA (siE2F1) delivery into cardiomyocytes that are normally hard to transfect, a spherical, α-helical polypeptide (SPP) with potent membrane activity was developed via dendrimer-initiated ring-opening polymerization of N-carboxyanhydride followed by side-chain functionalization with guanidines. Due to its multivalent structure, SPP outperformed its linear counterpart (LPP) to feature potent siRNA binding affinity and membrane activity. Thus, SPP effectively delivered siE2F1 into cardiomyocytes and suppressed E2F1 expression both in vitro and in vivo after intramyocardial injection. The E2F1-miR421-Pink1 signaling pathway was disrupted, thereby leading to the reduction of MIRI-induced mitochondrial damage, apoptosis, and inflammation of cardiomyocytes and ultimately recovering the systolic function of the myocardium. This study provides an example of membrane-penetrating nucleic acid delivery materials, and it also provides a promising approach for the genetic manipulation of cardiomyocyte apoptosis for the treatment of MIRI.


Subject(s)
Dendrimers , Myocardial Reperfusion Injury , Rats , Animals , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/drug therapy , RNA, Small Interfering/pharmacology , Dendrimers/metabolism , Rats, Sprague-Dawley , Apoptosis , Peptides/genetics , Peptides/pharmacology , Peptides/metabolism , Guanidines/pharmacology , Guanidines/therapeutic use , Protein Kinases/metabolism , Protein Kinases/pharmacology , Protein Kinases/therapeutic use , E2F1 Transcription Factor/metabolism , E2F1 Transcription Factor/pharmacology
3.
Commun Biol ; 5(1): 702, 2022 07 14.
Article in English | MEDLINE | ID: mdl-35835838

ABSTRACT

The multifaceted nature of senescent cell cycle arrest necessitates the targeting of multiple factors arresting or promoting the cell cycle. We report that co-inhibition of ATM and ROCK by KU-60019 and Y-27632, respectively, synergistically increases the proliferation of human diploid fibroblasts undergoing replicative senescence through activation of the transcription factors E2F1 and FOXM1. Time-course transcriptome analysis identified FOXM1 and E2F1 as crucial factors promoting proliferation. Co-inhibition of the kinases ATM and ROCK first promotes the G2/M transition via FOXM1 activation, leading to accumulation of cells undergoing the G1/S transition via E2F1 activation. The combination of both inhibitors increased this effect more significantly than either inhibitor alone, suggesting synergism. Our results demonstrate a FOXM1- and E2F1-mediated molecular pathway enhancing cell cycle progression in cells with proliferative potential under replicative senescence conditions, and treatment with the inhibitors can be tested for senomorphic effect in vivo.


Subject(s)
Cellular Senescence , E2F1 Transcription Factor , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle , Cell Cycle Proteins/metabolism , Cell Proliferation , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , E2F1 Transcription Factor/pharmacology , Forkhead Box Protein M1/genetics , Forkhead Box Protein M1/metabolism , Forkhead Box Protein M1/pharmacology , Humans
4.
Zhonghua Shao Shang Za Zhi ; 35(2): 104-109, 2019 Feb 20.
Article in Chinese | MEDLINE | ID: mdl-30798576

ABSTRACT

Objective: To explore the regulatory mechanism of E2F1 transcription factor on M2 macrophages in full-thickness skin defect wounds of mice. Methods: E2F1 gene knockout heterozygotes C57BL/6 mice and wild-type C57BL/6 mice were introduced and self-reproduced. Two weeks after birth, E2F1 gene knockout homozygotes mice and wild-type mice were identified by polymerase chain reaction (PCR). Twelve identified 6-8 weeks old male E2F1 gene knockout homozygotes C57BL/6 mice and wild-type C57BL/6 mice were selected respectively according to the random number table and set as E2F1 gene knockout group and wild-type group. A full-thickness skin defect wound was made on the back of each mouse. On post injury day (PID) 2 and 7, 6 mice in each group were selected according to the random number table and sacrificed, and the wound tissue was excised. The expression of CD68 and CD206 double positive M2 macrophages was observed by immunofluorescence method, and the percentage of CD206 positive cells was calculated. The protein expression of CD206 was detected by Western blotting. The mRNA expression of arginase 1 was detected by real-time fluorescent quantitative reverse transcription PCR (RT-PCR). Wound tissue specimens of the two groups on PID 7 were obtained, and the protein and mRNA expressions of peroxisome proliferator-activated receptor gamma (PPAR-γ) were detected by Western blotting and real-time fluorescent quantitative RT-PCR respectively. The above-mentioned experiments were repeated four times. Three specimens of wound tissue of mice in wild-type group on PID 7 were obtained to detect the relationship between E2F1 and PPAR-γ by co-immunoprecipitation and Western blotting, and this experiment was repeated two times. Data were processed with unpaired t test. Results: The size of PCR products of E2F1 gene knockout homozygotes C57BL/6 mice and wild-type C57BL/6 mice were 227 and 172 bp respectively, which were the same as those of the designed DNA fragments. On PID 2 and 7, the number of CD68 and CD206 double positive M2 macrophages in the wound tissue of mice in E2F1 gene knockout group was more than that of wild-type group, and the percentages of CD206 positive cells in the wound tissue of mice in E2F1 gene knockout group were (0.234±0.032)% and (0.584±0.023)% respectively, which were significantly higher than (0.129±0.017)% and (0.282±0.071)% of wild-type group (t=3.29, 3.54, P<0.05). On PID 2 and 7, the protein expression of CD206 in the wound tissue of mice in E2F1 gene knockout group were 1.00±0.23 and 1.63±0.26 respectively, which were significantly higher than 0.43±0.06 and 0.97±0.08 of wild-type group (t=2.41, 2.45, P<0.05). On PID 2 and 7, the mRNA expressions of arginase 1 in the wound tissue of mice in E2F1 gene knockout group were 0.482±0.105 and 0.195±0.031 respectively, which were significantly higher than 0.163±0.026 and 0.108±0.017 of wild-type group (t=3.04, 2.86, P<0.05). On PID 7, the protein and mRNA expressions of PPAR-γ in the wound tissue of mice in E2F1 gene knockout group were 0.61±0.12 and 0.51±0.13 respectively, which were significantly higher than 0.20±0.04 and 0.20±0.04 of wild-type group (t=3.36, 2.86, P<0.05). On PID 7, detection of the wound tissue of mice in wild-type group showed that PPAR-γ had unidirectional effect on E2F1. Conclusions: E2F1 transcription factor affects the polarization of M2 macrophages by inhibiting the expression of PPAR-γ, thereby inhibiting the healing process of full-thickness skin defect wounds in mice.


Subject(s)
E2F1 Transcription Factor/pharmacology , Macrophages/drug effects , Skin , Animals , Male , Mice , Mice, Inbred C57BL , Wound Healing
5.
J Virol ; 81(24): 13499-508, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17913805

ABSTRACT

E2F transcription factors play pivotal roles in controlling the expression of genes involved in cell viability as well as genes involved in cell death. E2F1 is an important constituent of this protein family, which thus far contains eight members. The interaction of E2F1 with its major regulator, retinoblastoma protein (Rb), has been studied extensively in the past two decades, concentrating on the role of E2F1 in transcriptional regulation and the role of Rb in cell replication and cancer formation. Additionally, the effect of viral infections on E2F1/Rb interactions has been analyzed for different viruses, concentrating on cell division, which is essential for viral replication. In the present study, we monitored E2F1-Rb interactions during human herpesvirus 6A (HHV-6A) and HHV-6B infections of SupT1 T cells. The results have shown the following dramatic alterations in E2F1-Rb pathways compared to the pathways of parallel mock-infected control cultures. (i) The E2F1 levels were elevated during viral infections. (ii) The cellular localization of E2F1 was dramatically altered, and it was found to accumulate both in the cytoplasmic and nuclear fractions, as opposed to the strict nuclear localization seen in the mock-infected cells. (iii) Although E2F1 expression was elevated, two exemplary target genes, cyclin E and MCM5, were not upregulated. (iv) The Rb protein was dephosphorylated early postinfection, a trait that also occurred with UV-inactivated virus. (v) Infection was associated with significant reduction of E2F1/Rb complexing. (vi) HHV-6 infections were accompanied by cell cycle arrest. The altered E2F1-Rb interactions and functions might contribute to the observed cell cycle arrest.


Subject(s)
Cell Cycle/drug effects , E2F1 Transcription Factor/metabolism , Gene Expression Regulation , Herpesvirus 6, Human/pathogenicity , Retinoblastoma Protein/metabolism , T-Lymphocytes/virology , Cell Line , Cell Nucleus/metabolism , Cytoplasm/metabolism , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/pharmacology , Herpesvirus 6, Human/radiation effects , Humans , Retinoblastoma Protein/genetics , Ultraviolet Rays
6.
Stem Cells ; 25(10): 2439-47, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17600109

ABSTRACT

E2F-6 is a dominant-negative transcriptional repressor against other members of the E2F family. In this study, we investigated the expression and function of E2F-6 in human hematopoietic progenitor cells to clarify its role in hematopoiesis. We found that among E2F subunits, E2F-1, E2F-2, E2F-4, and E2F-6 were expressed in CD34(+) human hematopoietic progenitor cells. The expression of E2F-6 increased along with proliferation and decreased during differentiation of hematopoietic progenitors, whereas the other three species were upregulated in CD34(-) bone marrow mononuclear cells. Overexpression of E2F-6 did not affect the growth of immature hematopoietic cell line K562 but suppressed E2F-1-induced apoptosis, whereas it failed to inhibit apoptosis induced by differentiation inducers and anticancer drugs. Among E2F-1-dependent apoptosis-related molecules, E2F-6 specifically inhibited upregulation of Apaf-1 by competing with E2F-1 for promoter binding. E2F-6 similarly suppressed apoptosis and Apaf-1 upregulation in primary hematopoietic progenitor cells during cytokine-induced proliferation but had no effect when they were differentiated. As a result, E2F-6 enhanced the clonogenic growth of colony-forming unit-granulocyte, erythroid, macrophage, and megakaryocyte. These results suggest that E2F-6 provides a failsafe mechanism against loss of hematopoietic progenitor cells during proliferation. Disclosure of potential conflicts of interest is found at the end of this article.


Subject(s)
Apoptosis/drug effects , E2F6 Transcription Factor/physiology , Hematopoietic Stem Cells/drug effects , Apoptotic Protease-Activating Factor 1/biosynthesis , Apoptotic Protease-Activating Factor 1/genetics , Binding, Competitive , Cell Differentiation/drug effects , Cells, Cultured/cytology , Cells, Cultured/drug effects , Colony-Forming Units Assay , Culture Media, Serum-Free/pharmacology , Cytokines/pharmacology , E2F Transcription Factors/biosynthesis , E2F Transcription Factors/genetics , E2F1 Transcription Factor/antagonists & inhibitors , E2F1 Transcription Factor/pharmacology , Hematopoietic Stem Cells/cytology , Humans , K562 Cells/cytology , K562 Cells/drug effects , Promoter Regions, Genetic/drug effects , Recombinant Fusion Proteins/physiology , Transduction, Genetic , Up-Regulation
7.
Cell Death Differ ; 13(2): 260-72, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16052233

ABSTRACT

E2F1 is a transcription factor that plays a well-documented role during S phase progression and apoptosis. We had previously postulated that the low level of E2F1 in primary lung adenocarcinoma contributes to their carcinogenesis. Here, we show that E2F1 triggers apoptosis in various lung adenocarcinoma cell lines by a mechanism involving the specific downregulation of the cellular FLICE-inhibitory protein short, leading to caspase-8 activation at the death-inducing signaling complex. Importantly, we also provide evidence that E2F1 sensitizes tumor as well as primary cells to apoptosis mediated by FAS ligand or tumor necrosis factor-related apoptosis-inducing ligand, and enhances the cytotoxic effect of T lymphocytes against tumor cells. Finally, we describe the specific overexpression of c-FLIP(S) in human lung adenocarcinomas with low level of E2F1. Overall, our data identify E2F1 as a critical determinant of the cellular response to death-receptor-mediated apoptosis, and suggest that its downregulation contributes to the immune escape of lung adenocarcinoma tumor cells.


Subject(s)
Adenocarcinoma/pathology , Apoptosis , Down-Regulation , E2F1 Transcription Factor/physiology , Intracellular Signaling Peptides and Proteins/genetics , Lung Neoplasms/pathology , Adenocarcinoma/chemistry , Adenocarcinoma/genetics , Apoptosis Regulatory Proteins/pharmacology , Blotting, Western , CASP8 and FADD-Like Apoptosis Regulating Protein , Caspase 8 , Caspases/metabolism , Cell Line, Tumor , Cytotoxicity, Immunologic , DNA/metabolism , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/pharmacology , Enzyme Activation , Fas Ligand Protein , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation, Neoplastic , Humans , Intracellular Signaling Peptides and Proteins/physiology , Lung Neoplasms/chemistry , Lung Neoplasms/genetics , Membrane Glycoproteins/pharmacology , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes, Cytotoxic/immunology , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factors/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...