Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.240
1.
Arch Esp Urol ; 77(3): 292-302, 2024 Apr.
Article En | MEDLINE | ID: mdl-38715171

BACKGROUND: Renal cell carcinoma (RCC), a common and highly invasive malignant tumour, presents clinical challenges due to its propensity for easy metastasis. Inferior vena cava tumour thrombus is a common RCC complication significantly impacting patient prognosis. This study investigates C-X-C chemokine receptor type 2 (CXCR2)/Snail-1-induced epithelial-mesenchymal transition (EMT) in RCC with inferior vena cava tumour thrombus. METHODS: Tissues from 51 RCC patients were analysed for CXCR2 and Snail-1 Messenger Ribonucleic Acid (mRNA) levels using Quantitative Real-Time Polymerase Chain Reaction (qRT-PCR). Elevated levels of both were observed in tumour and inferior vena cava tumour thrombus tissues. Using Short Hairpin RNA (shRNA) technology, we inhibited CXCR2 and Snail-1 expression to investigate their impact on EMT, invasiveness, and metastatic potential in RCC cells. RESULTS: Compared with that in the Short Hairpin RNA-Negative Control (ShNC) group, inhibition of CXCR2 and Snail-1 suppressed the degree of EMT, invasiveness, and metastatic ability of RCC cells (p < 0.01). Further mechanistic studies showed that CXCR2/Snail-1 participated in the formation and progression of RCC by regulating the extracellular signal-regulated kinase 1/2 (ERK1/2) signalling pathways. Additionally, compared with that in the ShNC group, knockdown of CXCR2 and Snail-1 significantly inhibited the expression of vascular endothelial growth factor (VEGF) and matrix metalloproteinase-9 (MMP-9; p < 0.01), thereby regulating the metastasis of RCC. CONCLUSIONS: Our findings suggest that CXCR2/Snail-1-induced EMT plays an important role in the formation and progression of RCC with inferior vena cava tumour thrombus. CXCR2/Snail-1 participates in the invasion and metastasis of RCC by regulating the expression of multiple signalling pathways and related genes. These results provide new insights and directions for the treatment of RCC.


Carcinoma, Renal Cell , Disease Progression , Epithelial-Mesenchymal Transition , Kidney Neoplasms , Snail Family Transcription Factors , Vena Cava, Inferior , Aged , Female , Humans , Male , Middle Aged , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/secondary , Kidney Neoplasms/pathology , Kidney Neoplasms/metabolism , Neoplasm Invasiveness , Snail Family Transcription Factors/metabolism , Tumor Cells, Cultured , Vena Cava, Inferior/pathology
2.
J Exp Clin Cancer Res ; 43(1): 135, 2024 May 04.
Article En | MEDLINE | ID: mdl-38702792

BACKGROUND: Rhabdomyosarcoma (RMS) is a rare malignancy and the most common soft tissue sarcoma in children. Vasculogenic mimicry (VM) is a novel tumor microcirculation model different from traditional tumor angiogenesis, which does not rely on endothelial cells to provide sufficient blood supply for tumor growth. In recent years, VM has been confirmed to be closely associated with tumor progression. However, the ability of RMS to form VM has not yet been reported. METHODS: Immunohistochemistry, RT-qPCR and western blot were used to test the expression level of SNAI2 and its clinical significance. The biological function in regulating vasculogenic mimicry and malignant progression of SNAI2 was examined both in vitro and in vivo. Mass spectrometry, co-immunohistochemistry, immunofluorescence staining, and ubiquitin assays were performed to explore the regulatory mechanism of SNAI2. RESULTS: Our study indicated that SNAI2 was abnormally expressed in patients with RMS and RMS cell lines and promoted the proliferation and metastasis of RMS. Through cell tubule formation experiments, nude mice Matrigel plug experiments, and immunohistochemistry (IHC), we confirmed that RMS can form VM and that SNAI2 promotes the formation of VM. Due to SNAI2 is a transcription factor that is not easily drugged, we used Co-IP combined with mass spectrometry to screen for the SNAI2-binding protein USP7 and TRIM21. USP7 depletion inhibited RMS VM formation, proliferation and metastasis by promoting SNAI2 degradation. We further demonstrated that TRIM21 is expressed at low levels in human RMS tissues and inhibits VM in RMS cells. TRIM21 promotes SNAI2 protein degradation through ubiquitination in the RMS. The deubiquitinase USP7 and E3 ligase TRIM21 function in an antagonistic rather than competitive mode and play a key role in controlling the stability of SNAI2 to determine the VM formation and progression of RMS. CONCLUSION: Our findings reveal a previously unknown mechanism by which USP7 and TRIM21 balance the level of SNAI2 ubiquitination, determining RMS vasculogenic mimicry, proliferation, and migration. This new mechanism may provide new targeted therapies to inhibit the development of RMS by restoring TRIM21 expression or inhibiting USP7 expression in RMS patients with high SNAI2 protein levels.


Neovascularization, Pathologic , Rhabdomyosarcoma , Ribonucleoproteins , Snail Family Transcription Factors , Ubiquitin-Specific Peptidase 7 , Humans , Snail Family Transcription Factors/metabolism , Snail Family Transcription Factors/genetics , Animals , Mice , Ubiquitin-Specific Peptidase 7/metabolism , Ubiquitin-Specific Peptidase 7/genetics , Rhabdomyosarcoma/metabolism , Rhabdomyosarcoma/pathology , Rhabdomyosarcoma/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Female , Disease Progression , Cell Proliferation , Male , Homeostasis , Cell Line, Tumor , Mice, Nude , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination
3.
Anal Cell Pathol (Amst) ; 2024: 8972022, 2024.
Article En | MEDLINE | ID: mdl-38715918

Preeclampsia (PE) manifests as a pregnancy-specific complication arising from compromised placentation characterized by inadequate trophoblast invasion. A growing body of evidence underscores the pivotal involvement of pseudogenes, a subset of long noncoding RNAs, in the pathological processes of PE. This study presents a novel finding, demonstrating a significant downregulation of the pseudogene PDIA3P1 in PE placental tissues compared to normal tissues. In vitro functional assays revealed that suppressing PDIA3P1 hindered trophoblast proliferation, invasion, and migration, concurrently upregulating the expression of secreted frizzled-related protein 1 (SFRP1). Further exploration of the regulatory role of PDIA3P1 in PE, utilizing human trophoblasts, established that PDIA3P1 exerts its function by binding to HuR, thereby enhancing the stability of Snail expression in trophoblasts. Overall, our findings suggest a crucial role for PDIA3P1 in regulating trophoblast properties and contributing to the pathogenesis of PE, offering potential targets for prognosis and therapeutic intervention.


Down-Regulation , Pre-Eclampsia , RNA, Long Noncoding , Snail Family Transcription Factors , Trophoblasts , Adult , Female , Humans , Pregnancy , Cell Movement/genetics , Cell Proliferation/genetics , Down-Regulation/genetics , Phenotype , Pre-Eclampsia/genetics , Pre-Eclampsia/metabolism , Pre-Eclampsia/pathology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Snail Family Transcription Factors/metabolism , Snail Family Transcription Factors/genetics , Trophoblasts/metabolism , Trophoblasts/pathology
4.
Int Immunopharmacol ; 135: 112322, 2024 Jun 30.
Article En | MEDLINE | ID: mdl-38788452

Idiopathic pulmonary fibrosis (IPF) is a chronic and progressive respiratory disorder characterized by poor prognosis, often presenting with acute exacerbation. The primary cause of death associated with IPF is acute exacerbation of IPF (AE-IPF). However, the pathophysiology of acute exacerbation has not been clearly elucidated yet. This study aims to investigate the underlying pathophysiological molecular mechanism in a mouse AE-PF model. C57BL/6J mice were intratracheally administered bleomycin (BLM, 5 mg/kg) to induce pulmonary fibrosis. After 14 days, lipopolysaccharide (LPS, 2 mg/kg) was injected via the trachea route. Histological assessments, including H&E and Masson staining, as well as inflammatory indicators, were included to evaluate the induction of AE-PF by BLM and LPS in mice. Transcriptomic profiling of pulmonary tissues identified CSF3 as one of the top 10 upregulated DEGs in AE-PF mice. Indeed, administration of exogenous CSF3 protein exacerbated AE-PF in mice. Mechanistically, CSF3 disrupted alveolar epithelial barrier integrity and permeability by regulating specialized cell adhesion complexes such as tight junctions (TJs) and adherens junctions (AJs) via PI3K/p-Akt/Snail pathway, contributing to the aggravation of AE-PF in mice. Moreover, the discovery of elevated sera CSF3 indicated a notable increase in IPF patients during the exacerbation of the disease. Pearson correlation analysis in IPF patients revealed significant positive associations between CSF3 levels and KL-6 levels, LDH levels, CRP levels, respectively. These results provide mechanistic insights into the role of CSF3 in exacerbating of lung fibrotic disease and indicate monitoring CSF3 levels may aid in early clinical decisions for alternative therapy in the management of rapidly progressing IPF.


Bleomycin , Idiopathic Pulmonary Fibrosis , Mice, Inbred C57BL , Animals , Humans , Mice , Idiopathic Pulmonary Fibrosis/pathology , Idiopathic Pulmonary Fibrosis/chemically induced , Male , Disease Models, Animal , Disease Progression , Female , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Signal Transduction , Middle Aged , Tight Junctions/metabolism , Tight Junctions/drug effects , Tight Junctions/pathology , Snail Family Transcription Factors/metabolism , Snail Family Transcription Factors/genetics , Proto-Oncogene Proteins c-akt/metabolism
5.
Biomed Res Int ; 2024: 8544837, 2024.
Article En | MEDLINE | ID: mdl-38803515

The loss of RAB25 expression-RAS superfamily of GTPase characteristic of numerous breast cancers-corresponds with H-RAS point mutations, particularly in triple-negative breast cancers (TNBC), a subtype associated with a poor prognosis. To address the poorly understood factors dictating the progression of TNBC tumors, we examine the cooperative effects that loss of RAB25 expression in human mammary epithelial cell (HMEC) lines with H-RAS mutations confers in tumorigenesis. HMECs were immortalized by transduction with LXSN CDK4 R24C, a mutant form of cyclin-dependent kinase, followed by transduction with hTERT, a catalytic subunit of the telomerase enzyme. We found that with the loss of RAB25 and overexpression of mutant H-RAS61L, immortal HMECs transformed toward anchorage-independent growth and acquired an increased ability to migrate. Furthermore, cells express low CD24, high CD44, and low claudin levels, indicating stem-like properties upon transformation. Besides, loss of RAB25 and overexpression of H-RAS61L resulted in increased expression of transcription factors Snail and Slug that drive these cells to lose E-cadherin and undergo epithelial-mesenchymal transition (EMT). This study confirms that loss of RAB25 and overexpression of mutant H-RAS can drive HMECs toward a mesenchymal stem-like state. Our findings reveal that RAB25 functions as a tumor suppressor gene, and loss of RAB25 could serve as a novel biomarker of the claudin-low type of TNBC.


Cell Transformation, Neoplastic , Claudins , Epithelial Cells , Epithelial-Mesenchymal Transition , rab GTP-Binding Proteins , Humans , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/genetics , Claudins/genetics , Claudins/metabolism , Female , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Oncogenes/genetics , Snail Family Transcription Factors/metabolism , Snail Family Transcription Factors/genetics , Mutation/genetics
6.
Sci Rep ; 14(1): 9616, 2024 04 26.
Article En | MEDLINE | ID: mdl-38671227

In this study, we aimed to study the role of TCONS_00006091 in the pathogenesis of oral squamous cellular carcinoma (OSCC) transformed from oral lichen planus (OLP). This study recruited 108 OSCC patients which transformed from OLP as the OSCC group and 102 OLP patients with no sign of OSCC as the Control group. ROC curves were plotted to measure the diagnostic values of TCONS_00006091, miR-153, miR-370 and let-7g, and the changes in gene expressions were measured by RT-qPCR. Sequence analysis and luciferase assays were performed to analyze the molecular relationships among these genes. Cell proliferation and apoptosis were observed via MTT and FCM. TCONS_00006091 exhibited a better diagnosis value for OSCC transformed from OLP. OSCC group showed increased TCONS_00006091 expression and decreased expressions of miR-153, miR-370 and let-7g. The levels of SNAI1, IRS and HMGA2 was all significantly increased in OSCC patients. And TCONS_00006091 was found to sponge miR-153, miR-370 and let-7g, while these miRNAs were respectively found to targe SNAI1, IRS and HMGA2. The elevated TCONS_00006091 suppressed the expressions of miR-153, miR-370 and let-7g, leading to the increased expression of SNAI1, IRS and HMGA2. Also, promoted cell proliferation and suppressed apoptosis were observed upon the over-expression of TCONS_00006091. This study demonstrated that the expressions of miR-153, miR-370 and let-7g were down-regulated by the highly expressed TCONS_00006091 in OSCC patients, which accordingly up-regulated the expressions of SNAI1, IRS and HMGA2, resulting in the promoted cell proliferation and suppressed cell apoptosis.


Apoptosis , Carcinoma, Squamous Cell , Cell Proliferation , Gene Expression Regulation, Neoplastic , HMGA2 Protein , MicroRNAs , Mouth Neoplasms , Snail Family Transcription Factors , Humans , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism , HMGA2 Protein/genetics , HMGA2 Protein/metabolism , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Mouth Neoplasms/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Cell Proliferation/genetics , Female , Male , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/metabolism , Apoptosis/genetics , Middle Aged , Up-Regulation , Cell Line, Tumor , Lichen Planus, Oral/genetics , Lichen Planus, Oral/metabolism , Lichen Planus, Oral/pathology
7.
Int J Mol Sci ; 25(8)2024 Apr 16.
Article En | MEDLINE | ID: mdl-38673959

Ovarian cancer poses a significant threat to patients in its advanced stages, often with limited treatment options available. In such cases, palliative management becomes the primary approach to maintaining a reasonable quality of life. Therefore, the administration of any medication that can benefit patients without a curative option holds potential. Resveratrol, a natural compound known for its in vitro anticancer activities, has generated contrasting results in vivo and human studies. In this study, we aimed to assess the anticancer effects of resveratrol on ovarian cancer cells grown on the chorioallantoic membrane (CAM) of chicken embryos. Two ovarian cancer cell lines, OVCAR-8 and SKOV-3, were cultured in collagen scaffolds for four days before being implanted on the CAM of chicken embryos on day 7. Different doses of resveratrol were applied to the CAM every two days for six days. Subsequently, CAM tissues were excised, fixed, and subjected to histological analysis. Some CAM tumours were extracted to analyse proteins through Western blotting. Our findings indicate that specific doses of resveratrol significantly reduce angiogenic activities, pNF-κB levels, and SLUG protein levels by using immunohistochemistry. These results suggest that resveratrol may have the potential to impact the behaviour of ovarian cancer CAM tumours, thereby warranting further consideration as a complementary treatment option for women with incurable ovarian cancer.


Chorioallantoic Membrane , Ovarian Neoplasms , Resveratrol , Resveratrol/pharmacology , Chorioallantoic Membrane/drug effects , Animals , Female , Chick Embryo , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Humans , Cell Line, Tumor , Snail Family Transcription Factors/metabolism , Neovascularization, Pathologic/drug therapy , NF-kappa B/metabolism , Antineoplastic Agents, Phytogenic/pharmacology
8.
Oral Oncol ; 152: 106798, 2024 May.
Article En | MEDLINE | ID: mdl-38615583

Acquired radio-resistance is thought to be one of the main causes of recurrent metastasis after failure of nasopharyngeal carcinoma (NPC) radiotherapy, which may be related to X-ray-induced epithelial-mesenchymal transition (EMT) activation. The circadian clock gene, BMAL1, has been shown to correlate with the sensitivity of NPCs to radiotherapy, but the specific mechanism has not been reported. NPC cells were irradiated by conventional fractionation to generate radiotherapy-resistant cells. NPC cells with BMAL1 gene stabilization/overexpression and interference were obtained by lentiviral transfection. Western blotting, colony formation analysis, cell counting kit-8 assays, wound-healing tests, Transwell assays, flow cytometry, the EDU method, nuclear plasma separation experiments, HE staining, immunohistochemical staining and TUNEL staining were performed to explore the influence and molecular mechanism of the circadian clock gene, BMAL1, on NPC-acquired radio-resistance and EMT through in vitro and in vivo experiments. The results indicated that there was a gradual downregulation of BMAL1 gene protein expression during the routine dose induction of radio-resistance in NPC cells. EMT activation was present in the radiation-resistant cell line 5-8FR, and was accompanied by the significant enhancement of proliferation, migration and invasion. The BMAL1 gene significantly increased the radiosensitivity of the radiation-resistant cell line 5-8FR and reversed the acquired radio-resistance of NPCs, which was accomplished by inhibiting the TGF-ß1/Smads/Snail1 axis-mediated EMT.


ARNTL Transcription Factors , Epithelial-Mesenchymal Transition , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms , Radiation Tolerance , Snail Family Transcription Factors , Transforming Growth Factor beta1 , Humans , Snail Family Transcription Factors/metabolism , ARNTL Transcription Factors/metabolism , ARNTL Transcription Factors/genetics , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Carcinoma/radiotherapy , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Carcinoma/pathology , Transforming Growth Factor beta1/metabolism , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/radiotherapy , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/genetics , Cell Line, Tumor , Animals , Mice , Smad Proteins/metabolism , Mice, Nude , Circadian Clocks , Male
9.
Development ; 151(10)2024 May 15.
Article En | MEDLINE | ID: mdl-38646822

The precise assembly of tissues and organs relies on spatiotemporal regulation of gene expression to coordinate the collective behavior of cells. In Drosophila embryos, the midgut musculature is formed through collective migration of caudal visceral mesoderm (CVM) cells, but how gene expression changes as cells migrate is not well understood. Here, we have focused on ten genes expressed in the CVM and the cis-regulatory sequences controlling their expression. Although some genes are continuously expressed, others are expressed only early or late during migration. Late expression relates to cell cycle progression, as driving string/Cdc25 causes earlier division of CVM cells and accelerates the transition to late gene expression. In particular, we found that the cell cycle effector transcription factor E2F1 is a required input for the late gene CG5080. Furthermore, whereas late genes are broadly expressed in all CVM cells, early gene transcripts are polarized to the anterior or posterior ends of the migrating collective. We show this polarization requires transcription factors Snail, Zfh1 and Dorsocross. Collectively, these results identify two sequential gene expression programs bridged by cell division that support long-distance directional migration of CVM cells.


Cell Division , Cell Movement , Drosophila Proteins , Gene Expression Regulation, Developmental , Animals , Cell Movement/genetics , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Cell Division/genetics , Mesoderm/metabolism , Mesoderm/cytology , Transcription Factors/metabolism , Transcription Factors/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Drosophila melanogaster/embryology , E2F1 Transcription Factor/metabolism , E2F1 Transcription Factor/genetics , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/cytology , Drosophila/genetics , Drosophila/metabolism , Drosophila/embryology , Snail Family Transcription Factors/metabolism , Snail Family Transcription Factors/genetics
10.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167202, 2024 Jun.
Article En | MEDLINE | ID: mdl-38670440

BACKGROUND & AIMS: Hypertrophic scar (HS) is a skin fibroproliferative disorder occurring after burns, surgeries or traumatic injuries, and it has caused a tremendous economic and medical burden. Its molecular mechanism is associated with the abnormal proliferation and transition of fibroblasts and excessive deposition of extracellular matrix. Cartilage intermediate layer protein 2 (CILP2), highly homologous to cartilage intermediate layer protein 1 (CILP1), is mainly secreted predominantly from chondrocytes in the middle/deeper layers of articular cartilage. Recent reports indicate that CILP2 is involved in the development of fibrotic diseases. We investigated the role of CILP2 in the progression of HS. METHODS AND RESULTS: It was found in this study that CILP2 expression was significantly higher in HS than in normal skin, especially in myofibroblasts. In a clinical cohort, we discovered that CILP2 was more abundant in the serum of patients with HS, especially in the early stage of HS. In vitro studies indicated that knockdown of CILP2 suppressed proliferation, migration, myofibroblast activation and collagen synthesis of hypertrophic scar fibroblasts (HSFs). Further, we revealed that CILP2 interacts with ATP citrate lyase (ACLY), in which CILP2 stabilizes the expression of ACLY by reducing the ubiquitination of ACLY, therefore prompting Snail acetylation and avoiding reduced expression of Snail. In vivo studies indicated that knockdown of CILP2 or ACLY inhibitor, SB-204990, significantly alleviated HS formation. CONCLUSION: CILP2 exerts a vital role in hypertrophic scar formation and might be a detectable biomarker reflecting the progression of hypertrophic scar and a therapeutic target for hypertrophic scar.


Cicatrix, Hypertrophic , Snail Family Transcription Factors , Adult , Animals , Female , Humans , Male , Mice , Acetylation , Cell Movement , Cell Proliferation , Cicatrix, Hypertrophic/metabolism , Cicatrix, Hypertrophic/pathology , Cicatrix, Hypertrophic/genetics , Fibroblasts/metabolism , Fibroblasts/pathology , Myofibroblasts/metabolism , Myofibroblasts/pathology , Snail Family Transcription Factors/metabolism , Snail Family Transcription Factors/genetics , ATP Citrate (pro-S)-Lyase/metabolism
11.
Cells ; 13(8)2024 Apr 17.
Article En | MEDLINE | ID: mdl-38667311

Actin is a protein of central importance to many cellular functions. Its localization and activity are regulated by interactions with a high number of actin-binding proteins. In a yeast two-hybrid (Y2H) screening system, snail family transcriptional repressor 2 (SNAI2 or slug) was identified as a yet unknown potential actin-binding protein. We validated this interaction using immunoprecipitation and analyzed the functional relation between slug and actin. Since both proteins have been reported to be involved in DNA double-strand break (DSB) repair, we focused on their interaction during this process after treatment with doxorubicin or UV irradiation. Confocal microscopy elicits that the overexpression of actin fused to an NLS stabilizes complexes of slug and γH2AX, an early marker of DNA damage repair.


Actins , Protein Binding , Snail Family Transcription Factors , Snail Family Transcription Factors/metabolism , Snail Family Transcription Factors/genetics , Actins/metabolism , Humans , Cell Nucleus/metabolism , Histones/metabolism , Two-Hybrid System Techniques , DNA Repair , Doxorubicin/pharmacology , DNA Breaks, Double-Stranded , Ultraviolet Rays , Animals
12.
Ecotoxicol Environ Saf ; 274: 116124, 2024 Apr 01.
Article En | MEDLINE | ID: mdl-38503108

OBJECTIVE: The primary objective of this study was to investigate the toxicological impact of Dibutyl phthalate (DBP) on the process of liver fibrosis transitioning into cirrhosis and the subsequent development of portal hypertension (PHT) through the mechanism of epithelial-mesenchymal transition (EMT) mediated by the ROS/TGF-ß/Snail-1 signaling pathway. METHOD: Carbon tetrachloride (CCl4) (1 mg/kg) was introduced in adult rats by oral feeding in CCl4 and CCl4+DBP groups twice a week for 8 weeks, and twice for another 8 week in CCl4 group. DBP was introduced by oral feeding in the CCl4+DBP group twice over the following 8 weeks. We subsequently analyzed hemodynamics measurements and liver cirrhosis degree, hepatic inflammation and liver function in the different groups. EMT related genes expression in rats in the groups of Control, DBP, CCl4 and CCl4+DBP were measured by immunohistochemistry (IHC). Enzyme-linked immunosorbent Assay (ELISA), qRT-PCR, western blot were used to detect the EMT related proteins and mRNA gene expression levels in rats and primary hepatocytes (PHCs). Reactive oxygen species (ROS) were examined with a ROS detection kit. RESULTS: The results showed that the CCl4+DBP group had higher portal pressure (PP) and lower mean arterial pressure (MAP) than the other groups. Elevated collagen deposition, profibrotic factor, inflammation, EMT levels were detected in DBP and CCl4+DBP groups. ROS, TGF-ß1 and Snail-1 were highly expressed after DBP exposure in vitro. TGF-ß1 had the potential to regulate Snail-1, and both of them were subject to regulation by ROS. CONCLUSION: DBP could influence the progression of EMT through its toxicological effect by ROS/TGF-ß1/Snail-1 signalling pathway, causing cirrhosis and PHT in final. The findings of this research might contribute to a novel comprehension of the underlying toxicological mechanisms and animal model involved in the progression of cirrhosis and PHT, and potentially offered a promising therapeutic target for the treatment of the disease.


Dibutyl Phthalate , Epithelial-Mesenchymal Transition , Hypertension, Portal , Transforming Growth Factor beta1 , Animals , Rats , Dibutyl Phthalate/toxicity , Fibrosis , Hypertension, Portal/chemically induced , Inflammation , Liver Cirrhosis/chemically induced , Reactive Oxygen Species , Signal Transduction , Snail Family Transcription Factors/metabolism , Transforming Growth Factor beta1/metabolism
13.
Gene ; 914: 148405, 2024 Jul 01.
Article En | MEDLINE | ID: mdl-38521110

The trophoblast epithelial-to-mesenchymal transition (EMT) is a procedure related to embryo implantation, spiral artery establishment and fetal-maternal communication, which is a key event for successful pregnancy. Inadequate EMT is one of the pathological mechanisms of recurrent miscarriage (RM). Whole-exome sequencing revealed that the mutation of bromodomain PHD-finger transcription factor (BPTF) was strongly associated with RM. In the present study, the effects of BPTF on EMT and the underlying mechanism were investigated. We found that the expression of BPTF in the villi of RM patients was significantly downregulated. Gene Ontology (GO) analysis revealed that BPTF participated in cell adhesion. The knockdown of BPTF prevented EMT and attenuated trophoblast invasion in vitro. BPTF activated Slug transcription by binding directly to the promoter region of the Slug gene. Interestingly, the protein levels of both Slug and BPTF were decreased in the villous cytotrophoblasts (VCTs) of RM villi. In conclusion, BPTF participates in the regulation of trophoblast EMT by activating Slug expression, suggesting that BPTF defects are an important factor in RM pathogenesis.


Antigens, Nuclear , Bromodomain Containing Proteins , Epithelial-Mesenchymal Transition , Nerve Tissue Proteins , Snail Family Transcription Factors , Transcription Factors , Trophoblasts , Trophoblasts/metabolism , Humans , Female , Snail Family Transcription Factors/metabolism , Snail Family Transcription Factors/genetics , Pregnancy , Transcription Factors/genetics , Transcription Factors/metabolism , Abortion, Habitual/genetics , Abortion, Habitual/metabolism , Cell Adhesion , Promoter Regions, Genetic , Adult
14.
Biochim Biophys Acta Mol Cell Res ; 1871(5): 119705, 2024 Jun.
Article En | MEDLINE | ID: mdl-38513918

Snail transcription factors play essential roles in embryonic development and participate in many physiological processes. However, these genes have been implicated in the development and progression of various types of cancer. In epithelial ovarian cancer, high expression of these transcription factors is usually associated with the acquisition of a more aggressive phenotype and thus, considered to be a poor prognostic factor. Numerous molecular signals create a complex network of signaling pathways regulating the expression and stability of Snails, which in turn control genes involved in vital cellular functions of ovarian cancer cells, such as invasion, survival, proliferation and chemoresistance.


Gene Expression Regulation, Neoplastic , Ovarian Neoplasms , Snail Family Transcription Factors , Humans , Snail Family Transcription Factors/metabolism , Snail Family Transcription Factors/genetics , Female , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Signal Transduction , Cell Proliferation , Animals , Carcinoma, Ovarian Epithelial/genetics , Carcinoma, Ovarian Epithelial/metabolism , Carcinoma, Ovarian Epithelial/pathology , Drug Resistance, Neoplasm/genetics
15.
Arterioscler Thromb Vasc Biol ; 44(5): e145-e167, 2024 May.
Article En | MEDLINE | ID: mdl-38482696

BACKGROUND: New blood vessel formation requires endothelial cells to transition from a quiescent to an invasive phenotype. Transcriptional changes are vital for this switch, but a comprehensive genome-wide approach focused exclusively on endothelial cell sprout initiation has not been reported. METHODS: Using a model of human endothelial cell sprout initiation, we developed a protocol to physically separate cells that initiate the process of new blood vessel formation (invading cells) from noninvading cells. We used this model to perform multiple transcriptomics analyses from independent donors to monitor endothelial gene expression changes. RESULTS: Single-cell population analyses, single-cell cluster analyses, and bulk RNA sequencing revealed common transcriptomic changes associated with invading cells. We also found that collagenase digestion used to isolate single cells upregulated the Fos proto-oncogene transcription factor. Exclusion of Fos proto-oncogene expressing cells revealed a gene signature consistent with activation of signal transduction, morphogenesis, and immune responses. Many of the genes were previously shown to regulate angiogenesis and included multiple tip cell markers. Upregulation of SNAI1 (snail family transcriptional repressor 1), PTGS2 (prostaglandin synthase 2), and JUNB (JunB proto-oncogene) protein expression was confirmed in invading cells, and silencing JunB and SNAI1 significantly reduced invasion responses. Separate studies investigated rounding 3, also known as RhoE, which has not yet been implicated in angiogenesis. Silencing rounding 3 reduced endothelial invasion distance as well as filopodia length, fitting with a pathfinding role for rounding 3 via regulation of filopodial extensions. Analysis of in vivo retinal angiogenesis in Rnd3 heterozygous mice confirmed a decrease in filopodial length compared with wild-type littermates. CONCLUSIONS: Validation of multiple genes, including rounding 3, revealed a functional role for this gene signature early in the angiogenic process. This study expands the list of genes associated with the acquisition of a tip cell phenotype during endothelial cell sprout initiation.


Gene Expression Profiling , Human Umbilical Vein Endothelial Cells , Neovascularization, Physiologic , Proto-Oncogene Proteins c-fos , Transcriptome , rho GTP-Binding Proteins , Animals , Humans , Mice , Cells, Cultured , Cyclooxygenase 2/metabolism , Cyclooxygenase 2/genetics , Endothelial Cells/metabolism , Gene Expression Profiling/methods , Human Umbilical Vein Endothelial Cells/metabolism , Neovascularization, Physiologic/genetics , Phenotype , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , rho GTP-Binding Proteins/metabolism , rho GTP-Binding Proteins/genetics , Signal Transduction , Single-Cell Analysis , Snail Family Transcription Factors/metabolism , Snail Family Transcription Factors/genetics
16.
Cancer Biomark ; 39(3): 231-243, 2024.
Article En | MEDLINE | ID: mdl-38217587

BACKGROUND: Epithelial-mesenchymal transition (EMT) is an important biological process by which malignant tumor cells to acquire migration and invasion abilities. This study explored the role of KLF5 in the EMT process of in cervical cancer cell lines. OBJECTIVE: Krüpple-like factor 5 (KLF5) is a basic transcriptional factor that plays a key role in cell-cycle arrest and inhibition of apoptosis. However, the molecular mechanism by which KLF5 mediates the biological functions of cervical cancer cell lines has not been elucidated. Here, we focus on the potential function of ELF5 in regulating the EMT process in in vitro model of cervical cancer cell lines. METHOD: Western-blot and real-time quantitative PCR were used to detect the expression of EMT-related genes in HeLa cells. MTT assays, cell scratch and Transwell assays were used to assess HeLa cells proliferation and invasion capability. Using the bioinformatics tool JASPAR, we identified a high-scoring KLF5-like binding sequence in the SNAI1 gene promoter. Luciferase reporter assays was used to detect transcriptional activity for different SNAI1 promoter truncates. RESULT: After overexpressing the KLF5 gene in HeLa cells, KLF5 not only significantly inhibited the invasion and migration of HeLa cells, but also increased the expression of E-cadherin and decreased the expression of N-cadherin and MMP9. In addition, the mRNA expression of upstream regulators of E-cadherin, such as SNAI1, SLUG, ZEB1/2 and TWIST1 was also decreased. Furthermore, KLF5 inhibiting the expression of the SNAI1 gene via binding its promoter region, and the EMT of Hela cells was promoted after overexpression of the SNAI1 gene. CONCLUSION: These results indicate that KLF5 can downregulate the EMT process of HeLa cells by decreasing the expression of the SNAI1 gene, thereby inhibiting the migration and invasion of HeLa cervical cancer cells.


Uterine Cervical Neoplasms , Female , Humans , HeLa Cells , Uterine Cervical Neoplasms/pathology , Cell Line, Tumor , Factor V/genetics , Factor V/metabolism , Cadherins/genetics , Cadherins/metabolism , Epithelial-Mesenchymal Transition/genetics , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism
17.
Exp Cell Res ; 434(1): 113864, 2024 01 01.
Article En | MEDLINE | ID: mdl-38040050

Metastasis is the primary cause of cancer-related deaths and remains poorly understood. Deubiquitinase OTU domain containing 4 (OTUD4) has been reported to regulate antiviral immune responses and resistance to radio- or chemo-therapies in certain cancers. However, the role of OTUD4 in cancer metastasis remain unknown. Here, we demonstrate that the depletion of OTUD4 in triple-negative breast cancer (TNBC) cells markedly suppress cell clonogenic ability, migration, invasion and cancer stem cell population in vitro as well as metastasis in vivo. Mechanistically, the tumor promoting function of OTUD4 is mainly mediated by deuiquitinating and stabilizing Snail1, one key transcriptional factor in the epithelial-mesenchymal transition. The inhibitory effect of targeting OTUD4 could be largely reversed by the reconstitution of Snail1 in OTUD4-deficient cells. Overall, our study establishes the OTUD4-Snail1 axis as an important regulatory mechanism of breast cancer metastasis and provides a rationale for potential therapeutic interventions in the treatment of TNBC.


Snail Family Transcription Factors , Triple Negative Breast Neoplasms , Ubiquitin-Specific Proteases , Ubiquitin-Specific Proteases/metabolism , MDA-MB-231 Cells , HEK293 Cells , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/physiopathology , Neoplasm Metastasis/genetics , Snail Family Transcription Factors/metabolism , Humans , Female , Animals , Mice , Cell Movement/genetics , Neoplasm Invasiveness/genetics , Protein Stability
18.
J Biol Chem ; 300(1): 105580, 2024 Jan.
Article En | MEDLINE | ID: mdl-38141763

Cancer cells acquire malignant phenotypes through an epithelial-mesenchymal transition, which is induced by environmental factors or extracellular signaling molecules, including transforming growth factor-ß (TGF-ß). Among epithelial-mesenchymal transition-associated cell responses, cell morphological changes and cell motility are closely associated with remodeling of the actin stress fibers. Here, we examined the TGF-ß signaling pathways leading to these cell responses. Through knockdown experiments in A549 lung adenocarcinoma cells, we found that Smad3-mediated induction of Snail, but not that of Slug, is indispensable for morphological changes, stress fiber formation, and enhanced motility in cells stimulated with TGF-ß. Ectopic expression of Snail in SMAD3-knockout cells rescued the defect in morphological changes and stress fiber formation by TGF-ß, indicating that the role of Smad3 in these responses is to upregulate Snail expression. Mechanistically, Snail is required for TGF-ß-induced upregulation of Wnt5b, which in turn activates RhoA and subsequent stress fiber formation in cooperation with phosphoinositide 3-kinase. However, ectopic expression of Snail in SMAD3-knockout cells failed to rescue the defect in cell motility enhancement by TGF-ß, indicating that activation of the Smad3/Snail/Wnt5b axis is indispensable but not sufficient for enhancing cell motility; a Smad3-dependent but Snail-independent pathway to activate Rac1 is additionally required. Therefore, the Smad3-dependent pathway leading to enhanced cell motility has two branches: a Snail-dependent branch to activate RhoA and a Snail-independent branch to activate Rac1. Coordinated activation of these branches, together with activation of non-Smad signaling pathways, mediates enhanced cell motility induced by TGF-ß.


Signal Transduction , Smad3 Protein , Snail Family Transcription Factors , Stress Fibers , Transforming Growth Factor beta , rho GTP-Binding Proteins , Humans , A549 Cells , Cell Movement , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition , Phosphatidylinositol 3-Kinases/metabolism , rho GTP-Binding Proteins/metabolism , Smad3 Protein/deficiency , Smad3 Protein/genetics , Smad3 Protein/metabolism , Snail Family Transcription Factors/deficiency , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism , Stress Fibers/metabolism , Transforming Growth Factor beta/metabolism , Enzyme Activation , Actins/metabolism , Mesoderm/metabolism , Mesoderm/pathology
19.
J Transl Med ; 21(1): 882, 2023 Dec 06.
Article En | MEDLINE | ID: mdl-38057853

Pancreatic ductal adenocarcinoma (PDAC) is characterized by a hypoxic microenvironment, a high rate of heterogeneity as well as a high likelihood of recurrence. Mounting evidence has affirmed that long non-coding RNAs (lncRNAs) participate in the carcinogenesis of PDAC cells. In this study, we revealed significantly decreased expression of GATA6-AS1 in PDAC based on the GEO dataset and our cohorts, and showed that low GATA6-AS1 expression was linked to unfavorable clinicopathologic characteristics as well as a poor prognosis. Gain- and loss-of-function studies demonstrated that GATA6-AS1 suppressed the proliferation, invasion, migration, and epithelial-mesenchymal transition (EMT) process of PDAC cells under hypoxia. In vivo data confirm the suppressive roles of GATA6-AS1/SNAI1 in tumor growth and lung metastasis of PDAC. Mechanistically, hypoxia-driven E26 transformation-specific sequence-1 (ETS1), as an upstream modulatory mechanism, was essential for the downregulation of GATA6-AS1 in PDAC cells. GATA6-AS1 inhibited the expression of fat mass and obesity-associated protein (FTO), an N6-methyladenosine (m6A) eraser, and repressed SNAI1 mRNA stability in an m6A-dependent manner. Our data suggested that GATA6-AS1 can inhibit PDAC cell proliferation, invasion, migration, EMT process and metastasis under hypoxia, and disrupting the GATA6-AS1/FTO/SNAI1 axis might be a viable therapeutic approach for refractory hypoxic pancreatic cancers.


Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , RNA, Long Noncoding , Humans , Cell Line, Tumor , Pancreatic Neoplasms/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Gene Expression Regulation, Neoplastic , Cell Movement/genetics , Tumor Microenvironment , GATA6 Transcription Factor/genetics , GATA6 Transcription Factor/metabolism , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/genetics , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism
20.
Cell Cycle ; 22(21-22): 2436-2448, 2023 11.
Article En | MEDLINE | ID: mdl-38146657

Endometriosis is a benign high prevalent disease exhibiting malignant features. However, the underlying pathogenesis and key molecules of endometriosis remain unclear. By integrating and analysis of existing expression profile datasets, we identified coxsackie and adenovirus receptor (CXADR), as a novel key gene in endometriosis. Based on the results of immunohistochemistry (IHC), we confirmed significant down-regulation of CXADR in ectopic endometrial tissues obtained from women with endometriosis compared with healthy controls. Further in vitro investigation indicated that CXADR regulated the stability and function of the phosphatases and AKT inhibitors PHLPP2 (pleckstrin homology domain and leucine-rich repeat protein phosphatase 2) and PTEN (phosphatase and tensin homolog). Loss of CXADR led to phosphorylation of AKT and glycogen synthase kinase-3ß (GSK-3ß), which resulted in stabilization of an epithelial-mesenchymal transition (EMT) factor, SNAIL1 (snail family transcriptional repressor 1). Therefore, EMT processs was induced, and the proliferation, migration and invasion of Ishikawa cells were enhanced. Over-expression of CXADR showed opposite effects. These findings suggest a previously undefined role of AKT/GSK-3ß signaling axis in regulating EMT and reveal the involvement of a CXADR-induced EMT, in pathogenic progression of endometriosis.


Endometriosis , Proto-Oncogene Proteins c-akt , Female , Humans , Cell Adhesion Molecules , Cell Line, Tumor , Cell Movement , Endometriosis/genetics , Epithelial-Mesenchymal Transition , Glycogen Synthase Kinase 3 beta , Phosphoprotein Phosphatases/pharmacology , Phosphoric Monoester Hydrolases , Proto-Oncogene Proteins c-akt/metabolism , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism
...