Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Medicine (Baltimore) ; 102(48): e36433, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-38050242

ABSTRACT

The dedifferentiation process of tumorigenesis and somatic cell reprogramming has some commonness and differences, which is the key question to cancer therapeutic strategy and stem cell applications. To further explore the commonalities and variance between carcinogenesis and induced pluripotent stem cell reprogramming, we investigated the role of stemness factors OSKM (OCT4, SOX2, KLF4, and MYC) in the pan-cancer process using public clinical data. Expression of OSKM in human pan-cancer was analyzed via the Genotype Tissue Expression (GTEx) and The Cancer Genome Atlas (TCGA) database based on the RNA-seq data of tissues. The correlation of expression between OSKM genes was analyzed via the Tumor Immune Evaluation Resource (TIMER) database, while the STRING tool was used to construct the protein-protein interaction network for OSKM. Prognostic impact of OSKM in pan-cancer was analyzed by Cox proportional hazards regression model. The relationships between OSKM and tumor stemness, tumor microenvironment and immune checkpoint and were performed by Sangerbox platform using Pearson correlation analysis. Our results showed that OSKM were universally expressed and significantly altered in tumors compared with adjacent normal tissues in most tumor types. In addition, correlation analysis revealed the relevance of OSKM genes to patient prognosis, cancer cell stemness, tumor microenvironment or immune checkpoint. However, there is little similarity between these genes in terms of how they function in each cancer type. This study elucidates the different roles of core stemness factors OSKM in pan-cancer, offering potential therapeutic targets for novel anti-cancer strategies and knowledge to minimize the potential carcinogenic effects during stem cell transplantation.


Subject(s)
Induced Pluripotent Stem Cells , Kruppel-Like Factor 4 , Neoplasms , Octamer Transcription Factor-3 , Proto-Oncogene Proteins c-myc , SOXB1 Transcription Factors , Humans , Cellular Reprogramming , Kruppel-Like Factor 4/genetics , Neoplasms/genetics , Neoplasms/metabolism , SOXB1 Transcription Factors/genetics , Tumor Microenvironment/genetics , Octamer Transcription Factor-3/genetics , Proto-Oncogene Proteins c-myc/genetics
2.
Cell Mol Biol Lett ; 28(1): 105, 2023 Dec 18.
Article in English | MEDLINE | ID: mdl-38105235

ABSTRACT

BACKGROUND: Pulmonary fibrosis is a growing clinical problem that develops as a result of abnormal wound healing, leading to breathlessness, pulmonary dysfunction and ultimately death. However, therapeutic options for pulmonary fibrosis are limited because the underlying pathogenesis remains incompletely understood. Circular RNAs, as key regulators in various diseases, remain poorly understood in pulmonary fibrosis induced by silica. METHODS: We performed studies with fibroblast cell lines and silica-induced mouse pulmonary fibrosis models. The expression of circZNF609, miR-145-5p, and KLF4 was determined by quantitative real-time polymerase chain reaction (qRT-PCR) analysis. RNA immunoprecipitation (RIP) assays and m6A RNA immunoprecipitation assays (MeRIP), Western blotting, immunofluorescence assays, and CCK8 were performed to investigate the role of the circZNF609/miR-145-5p/KLF4 axis and circZNF609-encoded peptides in fibroblast activation. RESULTS: Our data showed that circZNF609 was downregulated in activated fibroblasts and silica-induced fibrotic mouse lung tissues. Overexpression of circZNF609 could inhibit fibroblast activation induced by transforming growth factor-ß1 (TGF-ß1). Mechanically, we revealed that circZNF609 regulates pulmonary fibrosis via miR-145-5p/KLF4 axis and circZNF609-encoded peptides. Furthermore, circZNF609 was highly methylated and its expression was controlled by N6-methyladenosine (m6A) modification. Lastly, in vivo studies revealed that overexpression of circZNF609 attenuates silica-induced lung fibrosis in mice. CONCLUSIONS: Our data indicate that circZNF609 is a critical regulator of fibroblast activation and silica-induced lung fibrosis. The circZNF609 and its derived peptides may represent novel promising targets for the treatment of pulmonary fibrosis.


Subject(s)
MicroRNAs , Pulmonary Fibrosis , RNA, Circular , Animals , Mice , Lung/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/metabolism , Silicon Dioxide/adverse effects , Transforming Growth Factor beta1/metabolism , Kruppel-Like Factor 4/genetics , Kruppel-Like Factor 4/metabolism , RNA, Circular/genetics
3.
Biomed Res Int ; 2023: 1317998, 2023.
Article in English | MEDLINE | ID: mdl-37206297

ABSTRACT

This study is aimed at investigating the potential molecular features of allergic rhinitis (AR) and identifying gene signatures and related transcription factors using transcriptome analysis and in silico datasets. Transcriptome profiles were obtained using three independent cohorts (GSE101720, GSE19190, and GSE46171) comprising healthy controls (HC) and patients with AR. The pooled dataset (n = 82) was used to identify the critical signatures of AR compared with HC. Subsequently, key transcription factors were identified by a combined analysis using transcriptome and in silico datasets. Gene ontology: bioprocess (GO: BP) analysis using differentially expressed genes (DEGs) revealed that immune response-related genes were significantly enriched in AR compared with HC. Among them, IL1RL1, CD274, and CD44 were significantly higher in AR patients. We also identified key transcription factors between HC and AR using the in silico dataset and found that AR samples frequently express KLF transcription factor 4 (KLF4), which regulates immune response-related genes including IL1RL1, CD274, and CD44 in human nasal epithelial cells. Our integrative analysis of transcriptomic regulation provides new insights into AR, which may help in developing precision management for patients with AR.


Subject(s)
Gene Expression Regulation , Immunity , Kruppel-Like Factor 4 , Rhinitis, Allergic , Rhinitis, Allergic/genetics , Rhinitis, Allergic/immunology , Transcription Factors/genetics , Transcription Factors/immunology , Immunity/genetics , Immunity/immunology , Kruppel-Like Factor 4/genetics , Kruppel-Like Factor 4/immunology , Humans , Gene Expression Regulation/immunology , Gene Expression Profiling , Cell Line
4.
J Am Heart Assoc ; 12(4): e024303, 2023 02 21.
Article in English | MEDLINE | ID: mdl-36789992

ABSTRACT

Background Proper function of endothelial cells is critical for vascular integrity and organismal survival. Studies over the past 2 decades have identified 2 members of the KLF (Krüppel-like factor) family of proteins, KLF2 and KLF4, as nodal regulators of endothelial function. Strikingly, inducible postnatal deletion of both KLF2 and KLF4 resulted in widespread vascular leak, coagulopathy, and rapid death. Importantly, while transcriptomic studies revealed profound alterations in gene expression, the molecular mechanisms underlying these changes remain poorly understood. Here, we seek to determine mechanisms of KLF2 and KLF4 transcriptional control in multiple vascular beds to further understand their roles as critical endothelial regulators. Methods and Results We integrate chromatin occupancy and transcription studies from multiple transgenic mouse models to demonstrate that KLF2 and KLF4 have overlapping yet distinct binding patterns and transcriptional targets in heart and lung endothelium. Mechanistically, KLFs use open chromatin regions in promoters and enhancers and bind in context-specific patterns that govern transcription in microvasculature. Importantly, this occurs during homeostasis in vivo without additional exogenous stimuli. Conclusions Together, this work provides mechanistic insight behind the well-described transcriptional and functional heterogeneity seen in vascular populations, while also establishing tools into exploring microvascular endothelial dynamics in vivo.


Subject(s)
Endothelium , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors , Animals , Mice , Chromatin/metabolism , Endothelial Cells/metabolism , Endothelium/metabolism , Gene Expression , Kruppel-Like Factor 4/genetics , Kruppel-Like Factor 4/metabolism , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism
5.
FEBS J ; 290(1): 209-224, 2023 01.
Article in English | MEDLINE | ID: mdl-35942612

ABSTRACT

Macrophages are very important immune cells and play critical roles in tumour immunity. Macrophage subtypes can be divided into classical polarization (M1 macrophages) and alternative polarization (M2 macrophages) under different microenvironments. Krüppel-like factor 4 (KLF4) is an essential transcription factor for macrophage polarization. Our previous study has shown that KLF4 SUMOylation plays an important role in macrophage M2 polarization. In the present study, small ubiquitin-like modifier (SUMO) specific peptidase (SENP)1 was identified as a specific protease for KLF4 de-SUMOylation, with the SENP1-KLF4 axis playing a vital role in M1 macrophage polarization by affecting the nuclear factor kappa B signalling pathway. Additionally, the activity of tumour cells was weakened by KLF4 SUMOylation deficient macrophages. Hence, the SENP1-KLF4 axis is considered to play a crucial role in regulating lipopolysaccharide-induced macrophage M1 polarization, thereby affecting the activity of tumour cells. Therefore, the SENP1-KLF4 axis has therapeutic potential as a target in cancer therapy.


Subject(s)
Cysteine Endopeptidases , Kruppel-Like Factor 4 , Macrophages , Neoplasms , Humans , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Kruppel-Like Factor 4/genetics , Kruppel-Like Factor 4/metabolism , Lipopolysaccharides , Macrophages/metabolism , Neoplasms/metabolism , Signal Transduction , Tumor Microenvironment
6.
Respir Res ; 23(1): 340, 2022 Dec 10.
Article in English | MEDLINE | ID: mdl-36496404

ABSTRACT

BACKGROUND: Premature infants, subjected to supplemental oxygen and mechanical ventilation, may develop bronchopulmonary dysplasia, a chronic lung disease characterized by alveolar dysplasia and impaired vascularization. We and others have shown that hyperoxia causes senescence in cultured lung epithelial cells and fibroblasts. Although miR-34a modulates senescence, it is unclear whether it contributes to hyperoxia-induced senescence. We hypothesized that hyperoxia increases miR-34a levels, leading to cellular senescence. METHODS: We exposed mouse lung epithelial (MLE-12) cells and primary human small airway epithelial cells to hyperoxia (95% O2/5% CO2) or air (21% O2/5% CO2) for 24 h. Newborn mice (< 12 h old) were exposed to hyperoxia (> 95% O2) for 3 days and allowed to recover in room air until postnatal day 7. Lung samples from premature human infants requiring mechanical ventilation and control subjects who were not mechanically ventilated were employed. RESULTS: Hyperoxia caused senescence as indicated by loss of nuclear lamin B1, increased p21 gene expression, and senescence-associated secretory phenotype factors. Expression of miR-34a-5p was increased in epithelial cells and newborn mice exposed to hyperoxia, and in premature infants requiring mechanical ventilation. Transfection with a miR-34a-5p inhibitor reduced hyperoxia-induced senescence in MLE-12 cells. Additionally, hyperoxia increased protein levels of the oncogene and tumor-suppressor Krüppel-like factor 4 (KLF4), which were inhibited by a miR-34a-5p inhibitor. Furthermore, KLF4 knockdown by siRNA transfection reduced hyperoxia-induced senescence. CONCLUSION: Hyperoxia increases miR-34a-5p, leading to senescence in lung epithelial cells. This is dictated in part by upregulation of KLF4 signaling. Therefore, inhibiting hyperoxia-induced senescence via miR-34a-5p or KLF4 suppression may provide a novel therapeutic strategy to mitigate the detrimental consequences of hyperoxia in the neonatal lung.


Subject(s)
Bronchopulmonary Dysplasia , Hyperoxia , Kruppel-Like Factor 4 , MicroRNAs , Animals , Humans , Mice , Animals, Newborn , Bronchopulmonary Dysplasia/genetics , Bronchopulmonary Dysplasia/drug therapy , Carbon Dioxide , Cellular Senescence , Epithelial Cells/metabolism , Hyperoxia/genetics , Hyperoxia/metabolism , Kruppel-Like Factor 4/genetics , Kruppel-Like Factor 4/metabolism , Lung/metabolism , MicroRNAs/metabolism
7.
Theranostics ; 12(16): 6865-6882, 2022.
Article in English | MEDLINE | ID: mdl-36276637

ABSTRACT

Rationale: Dysregulation of signaling that governs self-renewal and differentiation of intestinal stem cells (ISCs) is a major cause of colorectal cancer (CRC) initiation and progression. Methods: qRT-PCR, western blotting, in situ hybridization, immunohistochemistry and immunofluorescence assays were used to detect the expression levels of MEX3A, KLF4 and E2F3 in CRC tissues. The biological functions of MEX3A were studied using Mex3a knockout (KO) and intestinal epithelium specific conditional knockout (cKO) mice, AOM-DSS mouse colorectal tumor model, Apc floxed mouse tumor model and intestinal and tumor organoids. Transcriptomic RNA sequencing (RNA-seq), RNA crosslinking immunoprecipitation (CLIP) and luciferase reporter assays were performed to explore the molecular mechanisms of MEX3A. Results: RNA-binding protein MEX3A, a specific ISC marker gene, becomes ectopically upregulated upon CRC and its levels negatively correlate with patient survival prognosis. MEX3A functions as an oncoprotein that retains cancer cells in undifferentiated and proliferative status and it enhances their radioresistance to DNA damage. Mechanistically, a rate limiting factor of cellular proliferation E2F3 induces MEX3A, which in turn activates WNT pathway by directly suppressing expression of its pro-differentiation transcription factor KLF4. Knockdown of MEX3A with siRNA or addition of KLF4 agonist significantly suppressed tumor growth both by increasing differentiation status of cancer cells and by suppressing their proliferation. Conclusions: It identifies E2F3-MEX3A-KLF4 axis as an essential coordinator of cancer stem cell self-renewal and differentiation, representing a potent new druggable target for cancer differentiation therapy.


Subject(s)
Colorectal Neoplasms , E2F3 Transcription Factor , Kruppel-Like Factor 4 , RNA-Binding Proteins , Animals , Mice , Cell Line, Tumor , Cell Proliferation/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Disease Models, Animal , Gene Expression Regulation, Neoplastic , RNA, Small Interfering , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Wnt Signaling Pathway , Kruppel-Like Factor 4/genetics , Kruppel-Like Factor 4/metabolism , E2F3 Transcription Factor/genetics , E2F3 Transcription Factor/metabolism
8.
Int J Biol Sci ; 18(13): 4869-4883, 2022.
Article in English | MEDLINE | ID: mdl-35982899

ABSTRACT

Non-small cell lung cancer (NSCLC) is one of the deadliest cancers in the world. Metastasis is considered one of the leading causes of treatment failure and death in NSCLC patients. A crucial factor of promoting metastasis in epithelium-derived carcinoma has been considered as epithelial-mesenchymal transition (EMT). Rictor, one of the components of mTORC2, has been reportedly involved in EMT and metastasis of human malignancies. However, the regulatory mechanisms of Rictor, Rictor-mediated EMT and metastasis in cancers remain unknown. Our present study indicates that Rictor is highly expressed in human NSCLC cell lines and tissues and is regulated, at least partially, at the transcriptional level. Knockdown of Rictor expression causes phenotype alterations through EMT, which is accompanied by the impairment of migration and invasion ability in NSCLC cells. Additionally, we have cloned and identified the human Rictor core promoter for the first time and confirmed that transcription factor KLF4 directly binds to the Rictor promoter and transcriptionally upregulated Rictor expression. Knockdown of KLF4 results in Rictor's downregulation accompanied by a series of characteristic changes of mesenchymal-epithelial transition (MET) and significantly decreases migration, invasion as well as metastasis of NSCLC cells. Re-introducing Rictor in KLF4-knockdown NSCLC cells partially reverses the epithelial phenotype to the mesenchymal phenotype and attenuates the inhibition of cell migration and invasion caused by KLF4 knocking down. Knockdown of KLF4 prevents mTOR/Rictor interaction and metastasis of NSCLC in vivo. The understanding of the regulator upstream of Rictor may provide an opportunity for the development of new inhibitors and the rational design of combination regimens based on different metastasis-related molecular targets and finally prevents cancer metastasis.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Rapamycin-Insensitive Companion of mTOR Protein , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Movement , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Humans , Kruppel-Like Factor 4/genetics , Lung Neoplasms/pathology , Rapamycin-Insensitive Companion of mTOR Protein/genetics , Rapamycin-Insensitive Companion of mTOR Protein/metabolism , Transcription Factors/metabolism
9.
Exp Lung Res ; 48(7-8): 213-223, 2022.
Article in English | MEDLINE | ID: mdl-35950640

ABSTRACT

PURPOSE: Bronchopulmonary dysplasia (BPD) is a long-term respiratory condition. More than a quarter of extremely premature newborns are harmed by BPD. At present, there are no apparent effective drugs or treatments for the condition. In this study, we aimed to investigate the functional role and mechanism of lymphoid enhancer-binding factor 1 (Lef1) in BPD in vitro. MATERIALS AND METHODS: Blood samples from BPD patients and healthy volunteers were gathered, and an in vitro model of BPD was developed in alveolar epithelial cells (AECs) MLE-12 induced by hyperoxia. Then expression of krüppel-like factor 4 (KLF4/Klf4) and LEF1/Lef1 were evaluated. After Lef1 overexpressing plasmid and the vector were transfected into hyperoxia-induced MLE-12 cells, cell proliferation assays were carried out. Cell apoptosis was investigated by a flow cytometry assay, and apoptosis related proteins Bcl-2, cleaved-caspase 3 and 9 were analyzed by a western blot assay. The binding between Klf4 and Lef1 promoter predicted on the JASPAR website was verified using luciferase and ChIP assays. For further study of the mechanism of Klf4 and Lef1 in BPD, gain-of-function experiments were performed. RESULTS: The mRNA levels of KLF4/Klf4 and LEF1/Lef1 were diminished in clinical BPD serum samples and hyperoxia-induced MLE-12 cells. Overexpression of Lef1 stimulated AEC proliferation and suppressed AEC apoptosis induced by hyperoxia. Mechanically, Klf4 bound to Lef1's promoter region and aids transcription. Moreover, the results of gain-of-function experiments supported that Klf4 could impede AEC damage induced by hyperoxia via stimulating Lef1. CONCLUSION: Klf4 and Lef1 expression levels were declined in hyperoxia-induced AECs, and Lef1 could be transcriptionally activated by Klf4 and protect against hyperoxia-induced AEC injury in BPD. As a result, Lef1 might become a prospective therapeutic target for BPD.


Subject(s)
Cell Hypoxia , Lymphoid Enhancer-Binding Factor 1 , Alveolar Epithelial Cells/metabolism , Bronchopulmonary Dysplasia/metabolism , Humans , Infant, Newborn , Kruppel-Like Factor 4/genetics , Kruppel-Like Factor 4/metabolism , Lymphoid Enhancer-Binding Factor 1/genetics , Lymphoid Enhancer-Binding Factor 1/metabolism
10.
Mol Biol Rep ; 49(9): 8495-8505, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35802277

ABSTRACT

BACKGROUND: The present study aimed to investigate the mechanisms through which long non-coding RNA (lncRNA) maternally expressed 3 (MEG3) affected the endothelial differentiation of mouse derived adipose-derived stem cells (ADSCs). MATERIALS AND METHODS: ADSCs were isolated and identified by specific surface marker detection. The effects of lncRNA MEG3 on endothelial differentiation of ADSCs were also detected via quantitative PCR, western blotting, immunofluorescence and Matrigel angiogenesis assays. In addition, using target gene prediction tools and luciferase reporter assays, the downstream target gene was demonstrated. RESULTS: LncRNA MEG3 targeted and reduced the expression levels of microRNA-145-5p (miR-145-5p), which upregulated the expression levels of Krüppel like factor 4 (KLF4), promoting endothelial differentiation of ADSCs. CONCLUSION: LncRNA MEG3 induced endothelial differentiation of ADSCs by targeting miR-145-5p/KLF4, which may provide novel insights to illustrate the mechanism of endothelial differentiation of ADSCs.


Subject(s)
Endothelium , Kruppel-Like Factor 4 , MicroRNAs , RNA, Long Noncoding , Stem Cells , Adipose Tissue/cytology , Animals , Cell Differentiation/genetics , Endothelium/cytology , Kruppel-Like Factor 4/genetics , Mice , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Stem Cells/metabolism
11.
Am J Pathol ; 192(9): 1250-1258, 2022 09.
Article in English | MEDLINE | ID: mdl-35750256

ABSTRACT

Along with a recent remarkable decrease in Helicobacter pylori-infected individuals, reports of gastric neoplasms such as sporadic foveolar-type gastric adenoma (FGA) in H. pylori-naive patients have been increasing. This tumor, with its raspberry-like appearance, is common in H. pylori-naive gastric mucosa. The current study investigated the genomic features of sporadic FGA. Fresh-frozen sporadic FGA tissue samples from H. pylori-naive patients were subjected to whole genome analysis using a next-generation sequencer. Proliferation ability and apoptotic profiles of human gastric epithelial cells, along with plasmid transfection of candidate variants, were examined. A mean of 6.65 × 108 total reads were obtained for each sample. Common genetic abnormalities in well-known proliferation driver genes of conventional gastric dysplasia/cancer were not found. However, a common single-nucleotide variation (SNV) was noted within the DNA-binding domain of the tumor suppressor gene KLF4. This novel SNV was located in the zinc finger 2 region. Additional experiments showed that it significantly suppressed proliferation of gastric epithelial cells compared with wild-type KLF4 plasmid-transfected cells, although suppression was reduced in early apoptotic phase-related genes. A novel SNV in the KLF4 zinc finger 2 region was commonly found in sporadic FGA tissue samples, which may explain the slow-growing properties of this neoplasm.


Subject(s)
Adenoma , Stomach Neoplasms , Adenoma/genetics , Adenoma/pathology , Adenomatous Polyps , Gastric Mucosa/pathology , Helicobacter Infections , Helicobacter pylori , Humans , Kruppel-Like Factor 4/genetics , Mutation , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology
12.
Kidney Int ; 102(1): 14-16, 2022 07.
Article in English | MEDLINE | ID: mdl-35738827

ABSTRACT

Krüppel-like factors (KLFs) are transcription factors with important roles in tissue homeostasis. KLF4 possesses antithrombotic and anti-inflammatory properties. In this issue, Estrada et al. show that endothelial KLF4 prevents complement deposition in glomeruli and in its absence the cell-bound complement regulator CD55 was reduced. The study included endothelial-specific KLF4 knockdown mice that mimic thrombotic microangiopathy and thrombotic microangiopathy patient biopsies showing decreased KLF4 and CD55. The results suggest that KLF4 is involved in the regulation of glomerular complement deposition.


Subject(s)
Complement Activation , Kidney Diseases , Kruppel-Like Factor 4 , Thrombotic Microangiopathies , Animals , Humans , Kidney Diseases/etiology , Kidney Glomerulus , Kruppel-Like Factor 4/genetics , Mice , Transcription Factors
13.
Cell Cycle ; 21(19): 2065-2078, 2022 10.
Article in English | MEDLINE | ID: mdl-35670736

ABSTRACT

SAMHD1 was reported to be related with the development of tumors, while its function in gastric cancer (GC) has not been elucidated yet. Here, we investigated the role and mechanism of SAMHD1 in regulating the proliferation of GC, as well as the mechanism of its expression regulation. Our results revealed that SAMHD1 was downregulated in GC tissues and cell lines, which was correlated with tumor size, depth of invasion and TNM stage. Overexpression of SAMHD1 inhibited the proliferation, clone formation, DNA synthesis and cell cycle progression, while knockdown of SAMHD1 promoted the proliferation of GC cells in vitro and vivo. Meanwhile, SAMHD1 inhibited the activation of MAPK p38 signaling pathway. Moreover, SB203580, as a MAPK p38 inhibitor, could reverse the proliferation and activation of MAPK p38 signaling pathway caused by knockdown of SAMHD1 in GC cells. Additionally, transcription factor Krüppel-like factor 4 (KLF4) bound to the core promoter of SAMHD1, increasing its transcriptional expression in GC cells. In conclusion, SAMHD1 suppressed the proliferation of GC through negatively regulating the activation of MAPK p38 signaling pathway and was upregulated by KLF4 in GC cells.


Subject(s)
Kruppel-Like Factor 4 , SAM Domain and HD Domain-Containing Protein 1 , Stomach Neoplasms , Cell Line, Tumor , Cell Movement , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Humans , Kruppel-Like Factor 4/genetics , SAM Domain and HD Domain-Containing Protein 1/genetics , Signal Transduction , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology
14.
Dis Markers ; 2022: 4090346, 2022.
Article in English | MEDLINE | ID: mdl-35637651

ABSTRACT

Non-small-cell lung cancer (NSCLC) is the most common lung cancer and a major cause of cancer mortality worldwide. Deguelin plays a vital inhibitory role in NSCLC initiation and development. However, the downstream mechanism of deguelin-suppressed metastasis of NSCLC cells is still not completely understood. Interestingly, phosphatase and tensin homologue deleted on chromosome 10 (PTEN) and Krüppel-like factor 4 (KLF4) also contribute to inhibition of metastasis in NSCLC cells. Here, we demonstrated that deguelin significantly upregulated PTEN and KLF4 expressions and PTEN positively upregulated KLF4 expression in NSCLC cells including A549 and PC9 cells. Moreover, overexpressions of PTEN and KLF4 inhibited the migration and invasion of NSCLC cells, an effect similar to that of deguelin. Furthermore, overexpressions of PTEN and KLF4 could suppress the epithelial-mesenchymal transition (EMT), an effect also similar to that of deguelin. Additionally, deguelin displayed a significant antitumor ability by upregulating PTEN and KLF4 expressions in mice model with NSCLC cells. Together, these results indicated that deguelin could be a potential therapeutic agent through upregulating PTEN and KLF4 expressions for NSCLC therapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Kruppel-Like Factor 4 , Lung Neoplasms , PTEN Phosphohydrolase , Rotenone , A549 Cells , Animals , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Humans , Kruppel-Like Factor 4/genetics , Kruppel-Like Factor 4/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Neoplasm Metastasis/prevention & control , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Rotenone/analogs & derivatives , Rotenone/pharmacology , Signal Transduction
15.
Int J Biol Sci ; 18(8): 3324-3336, 2022.
Article in English | MEDLINE | ID: mdl-35637963

ABSTRACT

Background: Hypertrophic scars (HS) often occur after burns, surgery and extensive trauma. Krüppel-like factor 4 (KLF4) is a member of the Krüppel-like factor family, a group of conserved zinc finger transcription factors that regulate diverse cellular processes. KLF4 can participate in the regulation of fibrotic diseases in many organs, such as the lung, liver, and heart. However, the antifibrotic effect of KLF4 in skin HS remains elusive. Result: This study observed the inhibition of KLF4 on fibrosis in vivo and in vitro. Our results revealed that KLF4 expression was decreased in HS tissue and fibroblasts. The results of KLF4 transfection confirmed its ability to alleviate the transdifferentiation of fibroblasts into myofibroblasts both in vitro and in vivo, thereby inhibiting the development of fibrosis. In addition, ChIP assays showed that BMP4 was the target gene of KLF4 for inhibiting skin fibrosis. Conclusions: Collectively, this evidence indicates that KLF4 is associated with BMP4 and could play an important regulatory role in HS formation by downregulating myofibroblast transdifferentiation. Our study provides a new target for the prevention and treatment of hypertrophic scars.


Subject(s)
Bone Morphogenetic Protein 4 , Cicatrix, Hypertrophic , Kruppel-Like Factor 4 , Bone Morphogenetic Protein 4/genetics , Cicatrix, Hypertrophic/genetics , Cicatrix, Hypertrophic/pathology , Fibroblasts/metabolism , Fibrosis , Humans , Kruppel-Like Factor 4/genetics
16.
Pharmacol Res ; 180: 106244, 2022 06.
Article in English | MEDLINE | ID: mdl-35550167

ABSTRACT

Sorafenib, a multikinase inhibitor, has been widely used as a first-line anticancer drug for advanced hepatocellular carcinoma (HCC). However, the development of drug resistance to sorafenib is frequently observed in clinical applications. Potential nonkinase targets of sorafenib have not been well documented and may provide insights into reversing drug resistance and enhancing drug efficacy. Herein, we report that sorafenib exerts its anticancer effects by activating metallothionein 1 G (MT1G) expression. MT1G is a novel marker in HCC that correlates well with patient survival. MT1G overexpression suppressed the cellular proliferation, migration, invasion, and tumour formation of HCC and sensitised cells to sorafenib treatment. However, the disruption of MT1G attenuated the anticancer effects of sorafenib. Mechanistically, sorafenib upregulated MT1G expression via hypomethylation of its promoter region by binding and inhibiting DNA methyltransferase 1 (DNMT1) and increasing its promoter accessibility in HCC cells. Activation of MT1G also inhibited CA9 transcription through the suppression of HIF1A as mediated by KLF4. Our collective data revealed that sorafenib exerts its anticancer effects through epigenetic regulation of the DNMT1/MT1G/KLF4/CA9 axis in HCC and the activation of MT1G might constitute a strategy for enhancing the effect of sorafenib to suppress HCC cells.


Subject(s)
Antineoplastic Agents , Carcinoma, Hepatocellular , Liver Neoplasms , Sorafenib , Antigens, Neoplasm , Antineoplastic Agents/pharmacology , Carbonic Anhydrase IX/genetics , Carbonic Anhydrase IX/metabolism , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Drug Resistance, Neoplasm , Epigenesis, Genetic , Humans , Kruppel-Like Factor 4/genetics , Kruppel-Like Factor 4/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Metallothionein/genetics , Metallothionein/metabolism , Signal Transduction/drug effects , Sorafenib/pharmacology
17.
Stem Cell Reports ; 17(5): 1070-1080, 2022 05 10.
Article in English | MEDLINE | ID: mdl-35452597

ABSTRACT

The Argonaute proteins (AGOs) are well known for their role in post-transcriptional gene silencing in the microRNA (miRNA) pathway. Here we show that in mouse embryonic stem cells, AGO1&2 serve additional functions that go beyond the miRNA pathway. Through the combined deletion of both Agos, we identified a specific set of genes that are uniquely regulated by AGOs but not by the other miRNA biogenesis factors. Deletion of Ago2&1 caused a global reduction of the repressive histone mark H3K27me3 due to downregulation at protein levels of Polycomb repressive complex 2 components. By integrating chromatin accessibility, prediction of transcription factor binding sites, and chromatin immunoprecipitation sequencing data, we identified the pluripotency factor KLF4 as a key modulator of AGO1&2-regulated genes. Our findings revealed a novel axis of gene regulation that is mediated by noncanonical functions of AGO proteins that affect chromatin states and gene expression using mechanisms outside the miRNA pathway.


Subject(s)
Argonaute Proteins , MicroRNAs , Animals , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Chromatin/genetics , Kruppel-Like Factor 4/genetics , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Mouse Embryonic Stem Cells/metabolism , Polycomb Repressive Complex 2/genetics
18.
Int Arch Allergy Immunol ; 183(7): 785-795, 2022.
Article in English | MEDLINE | ID: mdl-35390783

ABSTRACT

INTRODUCTION: Childhood asthma is a common chronic inflammatory lung disease in children, among which airway inflammation is the main driving factor of asthma symptoms. Follistatin-like protein 1 (FSTL1) is involved in multiple inflammatory processes, but its role in airway inflammation has not been fully elucidated. METHODS: We used lipopolysaccharide (LPS) to stimulate human primary bronchial epithelial (BEAS-2B) cells to establish an in vitro airway inflammation model. The expression of FSTL1 was detected by qPCR. Cell Counting Kit-8 and Annexin V-PI double staining was used to analyze the viability and apoptosis of BEAS-2B. The content of IL-6, IL-8 and TNF-α was determined by ELISA kit. Western blot was used to detect the protein expression level of the bone morphogenetic protein 4 (BMP4) and KLF4. The levels of superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GSH-Px), and malondialdehyde were measured to assess oxidative stress. RESULTS: The mRNA expression of FSTL1 was significantly increased in LPS-treated BEAS-2B cells. Silencing of FSTL1 inhibited the release of IL-6, IL-8, TNF-α, and cell apoptosis as well as enhanced the activities of SOD, CAT, and GSH-Px. Silencing of FSTL1 reversed the inflammatory state of cells by upregulating BMP4 and increasing the expression level of KLF4. CONCLUSION: Silencing of FSTL1 reduced LPS-induced BEAS-2B cell damage by regulating the BMP4/KLF4 axis. FSTL1 may be a potential target for the treatment of asthma.


Subject(s)
Asthma , Bone Morphogenetic Protein 4 , Follistatin-Related Proteins , Asthma/genetics , Asthma/metabolism , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/metabolism , Child , Epithelial Cells/metabolism , Follistatin-Related Proteins/genetics , Follistatin-Related Proteins/metabolism , Gene Silencing , Humans , Inflammation/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Kruppel-Like Factor 4/genetics , Kruppel-Like Factor 4/metabolism , Lipopolysaccharides/pharmacology , Oxidative Stress , Superoxide Dismutase , Tumor Necrosis Factor-alpha/metabolism
19.
Life Sci ; 296: 120447, 2022 May 01.
Article in English | MEDLINE | ID: mdl-35247439

ABSTRACT

AIMS: Esophageal squamous cell carcinoma (ESCC) is one of the deadliest digestive tract cancer with poor prognosis. In our previous comprehensive genomics study, we identified that hotspot mutations in the solute carrier family 35 member E2 (SLC35E2) promoter region was significantly associated with worse prognosis in patients with ESCC. However, the biological function and molecular mechanism of SLC35E2 remains unclear. This study was to investigate the malignant function and mechanism of SLC35E2 in ESCC. MAIN METHODS: Western blotting and qRT-PCR were used to assess the expression of SLC35E2 in ESCC cell lines. Luciferase assay and chromatin immunoprecipitation (ChIP) assay were used to assess the transcriptional inhibition of KLF4. Incucyte cell proliferation assay, colony formation assay and subcutaneous tumor formation in nude mice were used to assess the malignant function of SLC35E2. KEY FINDINGS: SLC35E2 can promote ESCC cell proliferation in vitro and in vivo. Krüppel-like factor 4 (KLF4), a transcriptional repressor in ESCC, binds to the SLC35E2 promoter and represses the expression of SLC35E2. The transcriptional suppression of KLF4 can be blocked by the mutation at -118 site of the SLC35E2 promoter. Besides, the accumulation of SLC35E2 expression contributes to the malignant phenotype of ESCC. SIGNIFICANCE: These results indicate that SLC35E2 may be used as a biomarker for prognosis as well as a therapeutic target for patients with ESCC.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Mutation , Animals , Female , Humans , Male , Cell Line, Tumor , Cell Proliferation/genetics , Esophageal Neoplasms/genetics , Esophageal Neoplasms/mortality , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/mortality , Gene Expression Regulation, Neoplastic , Kruppel-Like Factor 4/genetics , Mice, Inbred BALB C , Oncogenes , Prognosis , Promoter Regions, Genetic , Xenograft Model Antitumor Assays
20.
Epigenomics ; 14(4): 199-217, 2022 02.
Article in English | MEDLINE | ID: mdl-35172608

ABSTRACT

Aims: The authors aim to investigate the function of circPlekha7 in renal fibrosis. Methods: Human renal tissues from chronic kidney disease patients, kidney cell line and primary cultured renal tubular epithelial cells were used. TGF-ß1-treated human kidney 2 cells/tubular epithelial cells and a unilateral ureteral obstruction mouse model were employed to study renal fibrosis. Results: circPlekha7 was diminished in renal tissues from chronic kidney disease patients and TGF-ß1-treated human kidney 2 cells and tubular epithelial cells, while miR-493-3p was upregulated. Overexpression of circPlekha7 or knockdown of miR-493-3p suppressed TGF-ß1 induced enhancements on epithelial to mesenchymal transition and fibrogenesis, as well as attenuated renal fibrosis and injury in mice subjected to unilateral ureteral obstruction. circPlekha7 bound with miR-493-3p, which directly targeted KLF4. Conclusion: circPlekha7 inhibits epithelial to mesenchymal transition of renal tubular epithelial cells and fibrosis via targeting miR-493-3p to de-repress KLF4/mitofusin2 expression.


Chronic kidney disease (CKD) ultimately leads to complete kidney dysfunction. The incidence of CKD continues to rise as a result of the increasingly aging population, and the treatment is very limited. In this study, the authors identified a novel molecule, circPlekha7, that plays a crucial role in CKD development and progression. The level of circPlekha7 is lower in the kidney tissues of CKD patients, and increasing its level could attenuate kidney injury and fibrosis. This work helps researchers understand the disease better and, more importantly, provides new avenues to develop therapy.


Subject(s)
MicroRNAs , RNA, Circular , Renal Insufficiency, Chronic , Animals , Epithelial-Mesenchymal Transition , Fibrosis , Humans , Kidney/pathology , Kruppel-Like Factor 4/genetics , Kruppel-Like Factor 4/metabolism , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Circular/genetics , RNA, Circular/metabolism , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/pathology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL