Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Ecotoxicol Environ Saf ; 278: 116465, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38749198

ABSTRACT

5-Fluorouracil (5-FU), a chemotherapeutic drug used to treat a variety of cancers, can enter the environment through different routes, causing serious public health and environmental concerns. It has been reported that 5-FU exposure adversely affects male reproductive function, and its effects on this system cannot be avoided. In this study, using western blotting and quantitative polymerase chain reaction studies, we found that 5-FU promoted testicular injury by inducing oxidative stress, which was accompanied by the inhibition of nuclear factor erythroid 2-related factor 2/antioxidant response element signaling. Accumulation of reactive oxygen species (ROS) aggravated 5-FU-mediated mitochondrial dysfunction and apoptosis in murine cell lines and testes, indicating oxidative stress and mitochondrial-dependent apoptotic signaling play crucial roles in the damage of spermatogenic cells caused. N-Acetyl-L-cysteine, an antioxidant that scavenges intracellular ROS, protected spermatogenic cells from 5-FU-induced oxidative damage and mitochondrial dysfunction, revealing the important role of ROS in testicular dysfunction caused by 5-FU. We found that 5-FU exposure induces testicular cell apoptosis through ROS-mediated mitochondria pathway in mice. In summary, our findings revealed the reproductive toxicological effect of 5-FU on mice and its mechanism, provided basic data reference for adverse ecological and human health outcomes associated with 5-FU contamination or poisoning.


Subject(s)
Apoptosis , DNA Damage , Fluorouracil , Mitochondria , Oxidative Stress , Reactive Oxygen Species , Testis , Animals , Male , Fluorouracil/toxicity , Oxidative Stress/drug effects , Mice , Testis/drug effects , Testis/pathology , Mitochondria/drug effects , Reactive Oxygen Species/metabolism , Apoptosis/drug effects , Reproduction/drug effects , Cell Line
3.
Cell Death Discov ; 9(1): 358, 2023 Sep 28.
Article in English | MEDLINE | ID: mdl-37770442

ABSTRACT

Ferroptosis is a type of programmed cell death mediated by iron-dependent lipid peroxidation that leads to excessive lipid peroxidation in different cells. Ferroptosis is distinct from other forms of cell death and is associated with various diseases. Iron is essential for spermatogenesis and male reproductive function. Therefore, it is not surprising that new evidence supports the role of ferroptosis in testicular injury. Although the molecular mechanism by which ferroptosis induces disease is unknown, several genes and pathways associated with ferroptosis have been linked to testicular dysfunction. In this review, we discuss iron metabolism, ferroptosis, and related regulatory pathways. In addition, we analyze the endogenous and exogenous factors of ferroptosis in terms of iron metabolism and testicular dysfunction, as well as summarize the relationship between ferroptosis and male reproductive dysfunction. Finally, we discuss potential strategies to target ferroptosis for treating male reproductive diseases and provide new directions for preventing male reproductive diseases.

4.
J Transl Med ; 20(1): 262, 2022 06 07.
Article in English | MEDLINE | ID: mdl-35672760

ABSTRACT

BACKGROUND: Previous studies have indicated that chronic emotional stressors likely participate in the occurrence of cancers. However, direct evidence connecting stress and colorectal cancer development remains almost completely unexplored. METHODS: Chronic stress mouse model was used to investigate the influence of stress on tumorigenesis. Several major agonists and antagonists of adrenergic receptors were applied to investigate the effects of ß-adrenergic signaling on the development of CRC. Chromatin immunoprecipitation assays (CHIP) were used to investigate the binding of p53 and CEBPB to TRIM2 promoter. Mammosphere cultures, Cell Counting Kit-8 (CCK-8) assay, colony-formation assay, scratch wound healing assays, qPCR, immunofluorescence, coimmunoprecipitation and western blotting were used to explore the effect of stress-induced epinephrine on the CEBPB/TRIM2/P53 axis and the progress of CRC cells. RESULTS: In this study, we found that stress-induced epinephrine (EPI) promotes the proliferation, metastasis and CSC generation of CRC primarily through the ß2-adrenergic receptor. Furthermore, our studies also confirmed that chronic stress decreased the stability of p53 protein by promoting p53 ubiquitination. Results of transcriptome sequencing indicated that TRIM2 was overexpressed in cells treated with EPI. Further studies indicated that TRIM2 could regulate the stability of p53 protein by promoting p53 ubiquitination. Finally, we further proved that CEBPB was regulated by EPI and acts as the upstream transcription factor of TRIM2. CONCLUSIONS: Our studies proved that stress-induced EPI promotes the development and stemness of CRC through the CEBPB/TRIM2/P53 axis.


Subject(s)
Colorectal Neoplasms , Tumor Suppressor Protein p53 , Animals , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Colorectal Neoplasms/pathology , Epinephrine/pharmacology , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Mice , Tripartite Motif Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/metabolism
5.
Semin Liver Dis ; 42(2): 151-158, 2022 05.
Article in English | MEDLINE | ID: mdl-35738256

ABSTRACT

With the spread of coronavirus disease 2019 (COVID-19) worldwide, extrapulmonary lesions, including liver dysfunction, have attracted growing attention. The mechanisms underlying liver dysfunction in COVID-19 remain unclear. The reported prevalence of liver dysfunction varies widely across studies. In addition, its impact on clinical outcomes and its recovery after discharge are still controversial. In this review, pathological and laboratory findings were analyzed to reveal the potential mechanisms of COVID-19-induced liver injury from onset to recovery. Four patterns of liver damage were summarized according to the pathological findings, including hypoxemia and shock changes, vascular thrombosis and vascular damage, bile duct damage, and other histological changes. With a strict definition, the prevalence of liver dysfunction was not as high as reported. Meanwhile, liver dysfunction improved during the process of recovery. Nevertheless, the definite liver dysfunction was significantly associated with severe clinical course, which should not be ignored.


Subject(s)
COVID-19 , Liver Diseases , Thrombosis , COVID-19/complications , Humans
6.
Front Immunol ; 13: 740960, 2022.
Article in English | MEDLINE | ID: mdl-35350786

ABSTRACT

Background: N6-methyladenosine (m6A) and 5-methylcytosine (m5C) can modify long non-coding RNAs (lncRNAs), thereby affecting tumorigenesis and tumor progression. However, there is a lack of knowledge regarding the potential roles and cross-talk of m6A- and m5C-related lncRNAs in the tumor microenvironment (TME) and their effect on prognosis. Methods: We systematically evaluated the expression patterns of m6A- and m5C-related lncRNAs in 1358 colorectal cancer (CRC) samples from four datasets. Consensus clustering was conducted to identify molecular subtypes of CRC, and the clinical significance, TME, tumor-infiltrating immune cells (TIICs), and immune checkpoints in the different molecular subtypes were analyzed. Finally, we established a m6A- and m5C-related lncRNA signature and a prognostic nomogram. Results: We identified 141 m6A- and m5C-related lncRNAs by co-expression analysis, among which 23 lncRNAs were significantly associated with the overall survival (OS) of CRC patients. Two distinct molecular subtypes (cluster A and cluster B) were identified, and these two distinct molecular subtypes could predict clinicopathological features, prognosis, TME stromal activity, TIICs, immune checkpoints. Next, a m6A- and m5C-related lncRNA signature for predicting OS was constructed, and its predictive capability in CRC patients was validated. We then constructed a highly accurate nomogram for improving the clinical applicability of the signature. Analyses of clinicopathological features, prognosis, TIICs, cancer stem cell (CSC), and drug response revealed significant differences between two risk groups. In addition, we found that patients with a low-risk score exhibited enhanced response to anti-PD-1/L1 immunotherapy. Functional enrichment analysis showed that these lncRNAs related to the high-risk group were involved in the development and progression of CRC. Conclusions: We conducted a comprehensive analysis of m6A- and m5C-related lncRNAs in CRC and revealed their potential functions in predicting tumor-immune-stromal microenvironment, clinicopathological features, and prognosis, and determined their role in immunotherapy. These findings may improve our understanding of the cross-talk between m6A- and m5C-related lncRNAs in CRC and pave a new road for prognosis assessment and more effective immunotherapy strategies.


Subject(s)
Colorectal Neoplasms , RNA, Long Noncoding , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Colorectal Neoplasms/pathology , Humans , Prognosis , RNA, Long Noncoding/analysis , RNA, Long Noncoding/genetics , Tumor Microenvironment/genetics
7.
Genes (Basel) ; 12(9)2021 08 31.
Article in English | MEDLINE | ID: mdl-34573357

ABSTRACT

BACKGROUND: The N6-methyladenosine (m6A) RNA modification can modify long non-coding RNAs (lncRNAs), thereby affecting the tumorigenesis and progression of tumors. However, the underlying role of m6A-modified lncRNAs in colorectal cancer (CRC) remains largely unknown. Therefore, our aim was to assess the prognostic value of m6A-modified lncRNAs in CRC patients. METHODS: The gene expression and clinicopathological data of CRC were extracted from The Cancer Genome Atlas (TCGA) database. Pearson correlation analysis was used to investigate the m6A-modified lncRNAs. Consensus clustering was conducted to identify molecular subtypes of CRC, and the clinical significance of molecular subtypes was identified. The least absolute shrinkage and selection operator analysis (LASSO) was applied to establish a risk signature. Finally, a prognostic nomogram with risk score and clinicopathological variables was established. RESULTS: In total, 29 m6A-modified lncRNAs were identified as prognostic lncRNAs. Two molecular clusters were identified and significant differences were found with respect to clinicopathological features and prognosis. Cluster1 is associated with poor overall survival (OS), down-regulation of Programmed cell death ligand-1 (PD-L1) expression, lower immune score, and less immune cell infiltration. Then, an m6A-modified lncRNA signature for predicting OS was constructed in the TCGA training cohort. The signature demonstrated favorable prediction performance in both training and validation sets. Compared with low-risk patients, patients with high risk showed worse clinical outcomes, lower immune scores, and downregulated PD-L1 expression. Further analysis indicated that the signature was an independent prognostic indicator, and then a prognostic nomogram based on risk score, tumor location, and tumor stage was established. CONCLUSIONS: Our study identified a seven m6A-modified lncRNA signature and established a prognostic nomogram that reliably predicts OS in CRC. These findings may improve the understanding of m6A modifications in CRC and provide insights into the prognosis and treatment strategy of CRC.


Subject(s)
Biomarkers, Tumor/metabolism , Colorectal Neoplasms/mortality , Gene Expression Regulation, Neoplastic , RNA, Long Noncoding/metabolism , Adenosine/analogs & derivatives , Adenosine/metabolism , Biomarkers, Tumor/analysis , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Datasets as Topic , Epigenesis, Genetic , Female , Humans , Kaplan-Meier Estimate , Male , Methylation , Middle Aged , Nomograms , Risk Assessment/methods
8.
BMC Infect Dis ; 21(1): 821, 2021 Aug 16.
Article in English | MEDLINE | ID: mdl-34399679

ABSTRACT

BACKGROUND: Elderly patients with COVID-19 were shown to have a high case-fatality rate. We aimed to explore the risk factors associated with death in patients over 70 years old (yr). METHODS: In this retrospective study, we enrolled consecutively hospitalized patients over 70 yr with COVID-19 between January 20 and February 15, 2020 in Renmin Hospital of Wuhan University. Epidemiological, demographic, and clinical data were collected. Clinical subtypes, including mild, moderate, severe, and critical types, were used to evaluate the severity of disease. Patients were classified into two groups: survivor and non-survivor groups. Clinical data were compared between the two groups. Univariable and multivariable Cox regression methods were used to explore the risk factors. RESULTS: A total of 147 patients were enrolled. The case-fatality rate was 28.6%. Multivariable Cox proportional hazard regression showed that clinical subtypes, including the severe type (HR = 2.983, 95% CI: 1.231-7.226, P = 0.016) and the critical type (HR = 3.267, 95%CI: 1.009-10.576, P = 0.048), were associated with increasing risk of death when compared with the general type. Blood urea nitrogen greater than 9.5 mmol/L (HR = 2.805, 95% CI: 1.141-6.892, P = 0.025) on admission was an independent risk factor for death among laboratory findings. CONCLUSION: The patients over 70 yr with COVID-19 had a high case-fatality rate. The risk factors, including clinical subtypes and blood urea nitrogen greater than 9.5 mmol/L, could help physicians to identify elderly patients with poor clinical outcomes at an early stage.


Subject(s)
COVID-19/mortality , Aged , Aged, 80 and over , COVID-19/ethnology , China/epidemiology , Female , Hospital Mortality , Humans , Male , Prognosis , Retrospective Studies , Risk Factors , SARS-CoV-2
9.
Theranostics ; 11(2): 700-714, 2021.
Article in English | MEDLINE | ID: mdl-33391500

ABSTRACT

Rationale: PLAGL2 (pleomorphic adenoma gene like-2), a zinc finger PLAG transcription factor, is aberrantly expressed in several malignant tumors. However, the biological roles of PLAGL2 and its underlying mechanism in gastric cancer (GC) remain unclear. Methods: A series of experiments in vitro and in vivo were conducted to reveal the role of PLAGL2 in GC progression. Results: The data revealed that PLAGL2 promotes GC cell proliferation, migration, invasion, and EMT in vitro and in vivo. Mechanistically, we demonstrated the critical role of PLAGL2 in the stabilization of snail family transcriptional repressor 1 (Snail1) and promoting Snail1-mediated proliferation and migration of GC cells. PLAGL2 activated the transcription of deubiquitinase USP37, which then interacted with and deubiquitinated Snail1 protein directly. In addition, GSK-3ß-dependent phosphorylation of Snail1 protein is essential for USP37-mediated Snail1 deubiquitination regulation. Conclusions: In general, PLAGL2 promotes the proliferation and migration of GC cells through USP37-mediated deubiquitination of Snail1 protein. This work provided potential therapeutic targets for GC treatment.


Subject(s)
Biomarkers, Tumor/metabolism , DNA-Binding Proteins/metabolism , Endopeptidases/metabolism , Gene Expression Regulation, Neoplastic , RNA-Binding Proteins/metabolism , Snail Family Transcription Factors/metabolism , Stomach Neoplasms/pathology , Transcription Factors/metabolism , Ubiquitination , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cell Cycle , Cell Movement , Cell Proliferation , DNA-Binding Proteins/genetics , Endopeptidases/genetics , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Prognosis , RNA-Binding Proteins/genetics , Snail Family Transcription Factors/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Survival Rate , Transcription Factors/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
J Exp Clin Cancer Res ; 39(1): 232, 2020 Nov 05.
Article in English | MEDLINE | ID: mdl-33153498

ABSTRACT

BACKGROUND: Transmembrane 4 L six family member 1 (TM4SF1) is upregulated in several epithelial cancers and is closely associated with poor prognosis. However, the role of TM4SF1 and its potential mechanism in colorectal cancer (CRC) remain elusive. METHODS: We investigated the expression of TM4SF1 in the Oncomine, the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases and confirmed the results by immunohistochemistry (IHC), qPCR and Western blotting (WB) of CRC tissues. The effect of TM4SF1 on the epithelial-to-mesenchymal transition (EMT) and cancer stemness of CRC cells was investigated by Transwell, wound healing and sphere formation assays. A series of in vitro and in vivo experiments were conducted to reveal the mechanisms by which TM4SF1 modulates EMT and cancer stemness in CRC. RESULTS: TM4SF1 expression was markedly higher in CRC tissues than in non-tumour tissues and was positively correlated with poor prognosis. Downregulation of TM4SF1 inhibited the migration, invasion and tumour sphere formation of SW480 and LoVo cells. Conversely, TM4SF1 overexpression significantly enhanced the migration, invasion and tumoursphere formation potential of CRC cells, Additionally, TM4SF1 silencing inhibited the EMT mediated by transforming growth factor-ß1 (TGF-ß1). Mechanistically, gene set enrichment analysis (GSEA) predicted that the Wnt signalling pathway was one of the most impaired pathways in TM4SF1-deficient CRC cells compared to controls. The results were further validated by WB, which revealed that TM4SF1 modulated SOX2 expression in a Wnt/ß-catenin activation-dependent manner. Furthermore, we found that knockdown of TM4SF1 suppressed the expression of c-Myc, leading to decreased c-Myc binding to the SOX2 gene promoter. Finally, depletion of TM4SF1 inhibited metastasis and tumour growth in a xenograft mouse model. CONCLUSION: Our study substantiates a novel mechanism by which TM4SF1 maintains cancer cell stemness and EMT via the Wnt/ß-catenin/c-Myc/SOX2 axis during the recurrence and metastasis of CRC.


Subject(s)
Antigens, Surface/metabolism , Colorectal Neoplasms/metabolism , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/physiology , SOXB1 Transcription Factors/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/physiology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition , HCT116 Cells , Heterografts , Humans , Male , Mice , Mice, Nude , Middle Aged , Transfection
11.
Cell Tissue Bank ; 21(4): 667-673, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32870439

ABSTRACT

Rapid freezing and vitrification using sucrose are two simple and cost-effective sperm cryopreservation methods. However, it is still unclear which method is better and what the optimal concentration of sucrose is. This study aimed to determine the optimal sucrose concentration for human sperm cryopreservation and compare the cryoprotective effects of rapid freezing versus vitrification using different closed straw systems in terms of sperm motility and DNA integrity. Our data showed that: (1) The optimal sucrose concentration for vitrification was 0.25 mol/l among the tested 0, 0.125, 0.25 and 0.5 mol/l concentrations; (2) Sperm total motility and progressive motility were cryopreserved significantly better by rapid freezing than vitrification in standard 0.5 ml cryostraws (P < 0.05); and (3) Sperm total motility and progressive motility were cryopreserved significantly better by vitrification in the straw-in-straw system than rapid freezing in the standard 0.5 cryostraw (P < 0.05), but no difference was found in sperm nuclear DNA fragmentation level between the two cryopreservation methods (P > 0.05). It was concluded that sucrose at 0.25 mol/l concentration is suitable for human sperm rapid freezing and vitrification, and sperm cryopreservation can be achieved by rapid freezing using closed standard 0.5 ml straws or by vitrification using the novel straw-in-straw system made of standard 0.25 and 0.5 ml straws.


Subject(s)
Cryopreservation , Freezing , Spermatozoa/physiology , Sucrose/pharmacology , Vitrification , Adult , Chromatin/metabolism , DNA Damage , Humans , Male , Spermatozoa/drug effects , Young Adult
12.
Aging (Albany NY) ; 12(10): 9633-9657, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32413870

ABSTRACT

Evidence has shown that microRNAs (miRNAs) participate in the progression of CRC. Previous studies have indicated that miR-214-3p is abnormally expressed in various malignant tumors. However, the biological function it plays in CRC and the potential mechanism are unclear. Here, we demonstrated that miR-214-3p was obviously downregulated in CRC. Moreover, we found a strong correlation between the miR-214-3p level and tumor size and lymphatic metastasis. Furthermore, when miR-214-3p was decreased by an Lv-miR-214-3p inhibitor, the proliferation and migration of SW480 and HCT116 cells were significantly increased. As expected, the ability of proliferation and migration was significantly suppressed when miR-214-3p was overexpressed in DLD1 cells. According to the dual-luciferase reporter results, PLAGL2 was found to be a direct downstream molecule of miR-214-3p. Chromatin immunoprecipitation (CHIP) confirmed that MYH9, a well-known cytoskeleton molecule in CRC, was a direct targeting gene of PLAGL2. Silencing PLAGL2 or MYH9 could reverse the effect of a miR-214-3p inhibitor on CRC cells. In summary, our studies proved that low expression of miR-214-3p and overexpression of downstream PLAGL2 in CRC indicated a poor prognosis. MiR-214-3p suppressed the malignant behaviors of colorectal cancer by regulating the PLAGL2/MYH9 axis. MiR-214-3p might be a novel therapeutic target or prognostic marker for CRC.


Subject(s)
Cell Proliferation/genetics , Colorectal Neoplasms/genetics , DNA-Binding Proteins/metabolism , MicroRNAs/physiology , Myosin Heavy Chains/metabolism , RNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Aged , Cell Line, Tumor , Female , HCT116 Cells , Humans , Male , Middle Aged , Signal Transduction/genetics
13.
Br J Cancer ; 122(4): 578-589, 2020 02.
Article in English | MEDLINE | ID: mdl-31827238

ABSTRACT

BACKGROUND: We previously demonstrated that the pleomorphic adenoma gene like-2 (PLAGL2) is involved in the pathogenesis of Hirschsprung disease. Enhanced PLAGL2 expression was observed in several malignant tumours. However, the exact function of PLAGL2 and its underlying mechanism in colorectal cancer (CRC) remain largely unknown. METHODS: Immunohistochemical analysis of PLAGL2 was performed. A series of in vitro and in vivo experiments were conducted to reveal the role of PLAGL2 in the progression of CRC. RESULTS: Enhanced PLAGL2 expression was significantly associated with EMT-related proteins in CRC. The data revealed that PLAGL2 promotes CRC cell proliferation, migration, invasion and EMT both in vitro and in vivo. Mechanistically, PLAGL2 promoted the expression of ZEB1. PLAGL2 enhanced the expression and nuclear translocation of ß-catenin by decreasing its phosphorylation. The depletion of ß-catenin neutralised the regulation of ZEB1 that was caused by enhanced PLAGL2 expression. The small-molecule inhibitor PNU-74654, also impaired the enhancement of ZEB1 that resulted from the modified PLAGL2 expression. The depletion of ZEB1 could block the biological function of PLAGL2 in CRC cells. CONCLUSIONS: Collectively, our findings suggest that PLAGL2 mediates EMT to promote colorectal cancer metastasis via ß-catenin-dependent regulation of ZEB1.


Subject(s)
Colorectal Neoplasms/pathology , DNA-Binding Proteins/metabolism , Epithelial-Mesenchymal Transition/physiology , Gene Expression Regulation, Neoplastic/physiology , Neoplasm Invasiveness/genetics , RNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Animals , Cell Proliferation/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , DNA-Binding Proteins/genetics , Heterografts , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , RNA-Binding Proteins/genetics , Transcription Factors/genetics , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism , beta Catenin/genetics , beta Catenin/metabolism
14.
Pediatr Res ; 86(4): 460-470, 2019 10.
Article in English | MEDLINE | ID: mdl-30822775

ABSTRACT

BACKGROUND: The miR-214 has been reported to be associated with various diseases, but its involvement in the pathophysiology of Hirschsprung disease (HSCR) is almost completely unexplored. METHODS: In our study, we conducted a series of experiments to unravel the biological role of miR-214 in the pathophysiology of HSCR. qRT-PCR and western blotting were utilized to investigate the relative expression levels of miR-214, mRNAs, and proteins of related genes in colon tissues from 20 controls without HSCR and 24 patients with HSCR. The potential biological role of miR-214 in two cell lines (SKN-SH and SH-SY5Y) was assessed using the CCK8 assay, EdU staining, transwell assay, and flow cytometry. The dual-luciferase reporter assay was used to confirm PLAGL2 as a common target gene of miR-214. RESULTS: All results suggested that miR-214 is upregulated in HSCR tissue samples compared with controls. Additionally, we found that miR-214 could inhibit cell proliferation and migration by directly downregulating the expression of PLAGL2, and the extent of the miR-214-mediated inhibitory effects could be rescued by a PLAGL2 overexpression plasmid. CONCLUSION: Our results revealed that miR-214 is indeed involved in the pathophysiology of HSCR and suppresses cell proliferation and migration by directly downregulating PLAGL2 in cell models.


Subject(s)
DNA-Binding Proteins/genetics , Hirschsprung Disease/metabolism , MicroRNAs/genetics , RNA-Binding Proteins/genetics , Transcription Factors/genetics , Case-Control Studies , Cell Line , Cell Movement , Cell Proliferation , Child , Child, Preschool , Colon/metabolism , Down-Regulation , Female , Gene Expression Profiling , Humans , Male , Up-Regulation
15.
J Mol Med (Berl) ; 97(1): 89-102, 2019 01.
Article in English | MEDLINE | ID: mdl-30460377

ABSTRACT

Cancer stem cells (CSCs) and epithelial-mesenchymal transition (EMT) play an important role in the metastasis and chemoresistance in the context of colorectal cancer (CRC). Downregulation of death associated protein kinase 1 (DAPK1) may promote metastasis and chemoresistance of cancer cells through various mechanisms. However, the association between DAPK1 and CSCs or EMT has not been explored. In this study, we demonstrated that DAPK1 was associated with elevated stemness of CSCs in patients with CRC. Silencing of DAPK1 in CRC cell lines promoted the metastasis and chemoresistance due to increased stemness of CSCs and enhanced mesenchymal phenotype, an effect that was mediated via activation of the transcription factor, zinc finger E-box binding homeobox 1 (ZEB1). Blockade of this signaling pathway attenuated the stemness of CSCs and rescued the EMT process. DAPK1-ZEB1 may lie at the interface of TGF-ß and WNT pathways and participate in both CSCs and EMT process. Targeted therapies aimed at DAPK1-ZEB1 pathway may inhibit the chemoresistance and metastasis of CRC. KEY MESSAGES: Downregulation of DAPK1 promotes chemoresistance and metastasis of CRC. Inhibition of DAPK1 promotes the stemness of cancer stem cells and EMT process. DAPK1-ZEB1 may lie at the interface of TGF-ß and WNT pathways. DAPK1-ZEB1 participates in both CSCs and EMT process.


Subject(s)
Colorectal Neoplasms/pathology , Death-Associated Protein Kinases/metabolism , Epithelial-Mesenchymal Transition , Neoplastic Stem Cells/pathology , Zinc Finger E-box-Binding Homeobox 1/metabolism , Animals , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Death-Associated Protein Kinases/analysis , Death-Associated Protein Kinases/genetics , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice, Nude , Middle Aged , Neoplastic Stem Cells/metabolism
16.
Cancer Sci ; 110(1): 86-98, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30320939

ABSTRACT

Nuclear factor I/B (NFIB) is a widely studied transcription factor that participates in tumor progression; nevertheless, studies on NFIB in colorectal cancer (CRC) are limited. In our study, Western blot and RT-PCR analyses showed that NFIB was overexpressed in CRC tissues and cell lines, which was consistent with our bioinformatic analysis results. Furthermore, NFIB expression was closely related to the TNM stage of CRC. NFIB promoted cell proliferation and migration and inhibited cell apoptosis in vitro. Meanwhile, we discovered that NFIB accelerated xenograft tumor growth in vivo. In addition, NFIB weakened the sensitivity of CRC cells to 5-fluorouracil (5-FU). NFIB induced epithelial-mesenchymal transition (EMT) by upregulating snail expression, which was accompanied by decreased E-cadherin and Zo-1 expression and increasedd Vimentin expression. Because the Akt pathway plays an important role in CRC progression, we examined whether there was a correlation between NFIB and the Akt pathway in cell proliferation and migration. Our results showed that NFIB promoted cell proliferation and increased 5-FU resistance by activating the Akt pathway. In summary, our findings suggested that NFIB induced EMT of CRC cells via upregulating snail expression and promoted cell proliferation and 5-FU resistance by activating the Akt pathway.


Subject(s)
Cell Proliferation/genetics , Colorectal Neoplasms/genetics , Drug Resistance, Neoplasm/drug effects , Epithelial-Mesenchymal Transition/genetics , Fluorouracil/pharmacology , NFI Transcription Factors/genetics , Animals , Antimetabolites, Antineoplastic/pharmacology , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/therapy , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Male , Mice, Inbred BALB C , Mice, Nude , Middle Aged , NFI Transcription Factors/metabolism , RNA Interference , RNAi Therapeutics/methods , Xenograft Model Antitumor Assays/methods
17.
Oncol Lett ; 15(2): 1573-1579, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29434852

ABSTRACT

Colorectal cancer (CRC) has been one of the most common types of cancer for decades worldwide. The pathogenesis of CRC is associated with the processes of activating oncogenes and inactivating anti-oncogenes. Platelet-derived growth factor-D (PDGF-D) was confirmed to regulate migration, invasion, proliferation, apoptosis and metastasis in various cancer cells. Overexpression of PDGF-D exists in a number of human malignancies, including pancreatic, prostate and breast cancer. However, the expression and function of PDGF-D and its associated molecular mechanism in CRC remain unclear. Thus, the expression of PDGF-D was detected in CRC tissues and human colon cancer lines. Subsequently, the effects of PDGF-D on the invasion, migration and proliferation of cancer cells were investigated. The corresponding molecular mechanism had also been explored. The present study revealed that PDGF-D was upregulated not only in CRC tissues but also in CRC cell lines, and simultaneously, facilitated the processes of migration, invasion and proliferation. Silencing PDGF-D in the SW480 cell line inhibited migration, invasion and proliferation distinctly, with reduced expression of Notch1 and matrix metalloproteinase-9. Furthermore, upregulating PDGF-D in HCT116 cells led to the opposite results. These findings indicate that PDGF-D may be developed into a potential therapeutic target for CRC treatment.

18.
PLoS One ; 12(9): e0184959, 2017.
Article in English | MEDLINE | ID: mdl-28934284

ABSTRACT

OBJECTIVE: One of the critical mechanisms of gastrointestinal cancer pathogenesis is the silencing of death associated protein kinase 1 (DAPK1), which could be caused by aberrant methylation of the promoter. However, the relationship between DAPK1 methylation and the risk of gastrointestinal cancer is still controversial. Hence, we conducted this study to determine the potential correlation. METHODS: Eligible publications were searched in the Pubmed, Embase, and Cochrane Library through November 2016 according to the inclusion criteria and exclusion criteria. Revman 5.3 and Stata 12.0 software were used to analyze the relevant data regarding the association between the frequency of DAPK1 methylation and gastrointestinal cancer. RESULTS: A total of 22 studies with 2406 patients were included in this meta analysis. Methylation of DAPK1 was positively related with the risk of gastrointestinal cancer (odds ratio [OR] = 5.35, 95% confidence interval [CI]: 2.76-10.38, P<0.00001, random effects model). The source of heterogeneity was analyzed by sensitivity analysis and subgroup analysis. After omitting one heterogeneous study, the I2 decreased and the OR increased in pooled analysis. Also, the heterogeneity decreased most significantly in the subgroup of studies that had a sample size of less than 60 cases. Then, the correlations between DAPK1 methylation and clinicopathological features of gastrointestinal cancer were assessed. DAPK1 methylation was positively correlated with the lymph node (N) stage (positive vs. negative, OR = 1.45, 95%CI: 1.01-2.06, P = 0.04, fixed effects model) and poor differentiation (OR = 1.55, 95%CI: 1.02-2.35, P = 0.04, fixed effects model) in gastric cancer, and the association was significant among Asian patients. However, among cases of gastrointestinal cancer, the association between DAPK1 methylation and tumor (T) stage, N stage, distant metastasis (M) stage, and cancer differentiation were not statistically significant. CONCLUSIONS: DAPK1 methylation is a potential biomarker for the early diagnosis of gastrointestinal cancer. Further analysis of the clinicopathological features indicated that aberrant methylation of DAPK1 is positively associated with the tumorigenesis of gastrointestinal cancer, and metastasis of gastric cancer.


Subject(s)
DNA Methylation , Death-Associated Protein Kinases/genetics , Gastrointestinal Neoplasms/genetics , Genetic Predisposition to Disease , Gene Expression Regulation, Neoplastic , Humans , Risk Factors
19.
Medicine (Baltimore) ; 96(50): e9189, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29390331

ABSTRACT

RATIONALE: This article is aimed to retrospect the clinicopathological data of 2 cases of gastric MANENCs. MANEC is a rare biphasic tumor type that is coexistence of dual neuroendocrine and adenocarcinoma differentiation with each composing exceeding 30% volume. Gastric MANEC have just been reported anecdotally in the literature due to their rarity and heterogeneity. According to our study, these neoplasms have 3 different metastasis patterns: only adenocarcinomatous or neuroendocrine carcinoma and both of the 2 components. We first focus on the correlation of metastasis characteristics with prognosis in gastric MANEC, which may be potential implications for the choice of chemotherapy. PATIENT CONCERNS: The 2 cases of patient shared several symptoms: epigastric discomfort, weight loss, hematemesis, or melena. DIAGNOSIS: The 2 patients were diagnosis as MANEC based on the identification of histopathological analysis. In case 1, the poor differentiated adenocarcinoma accounted for 30%, the neuroendocrine part account for 70% and both of the 2 components metastasized to the lymph nodes, whereas in case 2, poorly differentiated adenocarcinoma accounted for 70%, the neuroendocrine part for 30% and only the glandular component invaded regional lymph nodes. INTERVENTIONS: The first patient underwent laparoscopic radical gastrectomy and underwent adjuvant chemotherapy, combination of cisplatin, and etoposide successfully. The second patient received radical gastronomy, and did not receive any chemotherapy due to general weakness. OUTCOMES: The first patient is alive with no evidence of recurrence, and the second patient died 6 months after the operation. LESSONS: The assessment of metastatic sites should be a routine pathological practice, which is crucial for clinical decision-making and the selection of management.


Subject(s)
Adenocarcinoma/pathology , Carcinoma, Neuroendocrine/pathology , Neoplasm Metastasis/pathology , Stomach Neoplasms/pathology , Adenocarcinoma/surgery , Carcinoma, Neuroendocrine/surgery , Fatal Outcome , Humans , Male , Middle Aged , Prognosis , Stomach Neoplasms/surgery
20.
Oncotarget ; 8(6): 9961-9973, 2017 Feb 07.
Article in English | MEDLINE | ID: mdl-28035069

ABSTRACT

Platelet-derived growth factor-D (PDGF-D) plays a crucial role in the progression of several cancers. However, its role in colorectal cancer (CRC) remains unclear. Our study showed that PDGF-D was highly expressed in CRC tissues and was positively associated with the clinicopathological features. Down-regulation of PDGF-D inhibited the tumor growth, migration and angiogenesis of SW480 cells in vitro and in vivo. Whereas up-regulation of PDGF-D promoted the malignant behaviors of HCT116 cells. Moreover, PDGF-D up-regulated the expression of Notch1 and Twist1 in CRC cells. In addition, PDGF-D expression promoted Epithelial to mesenchymal transition (EMT), which was accompanied with decreased E-cadherin and increased Vimentin expression. Consistently, PDGF-D, Notch1, and Twist1 are obviously up-regulated in transforming growth factor-beta 1 (TGF-ß1) treated HCT116 cells. Since Notch1 and Twist1 play an important role in EMT and tumor progression, we examined whether there is a correlation between Notch1 and Twist1 in EMT status. Our results showed that up-regulation of Notch1 was able to rescue the effects of PDGF-D down-regulation on Twist1 expression in SW480 cells, whereas down-regulation of Notch1 reduced Twist1 expression in HCT116 cells. Furthermore, we found that Twist1 promoted EMT and aggressiveness of CRC cells. These results suggest that PDGF-D promotes tumor growth and aggressiveness of CRC, moreover, down-regulation of PDGF-D inactivates Notch1/Twist1 axis, which could reverse EMT and prevent CRC progression.


Subject(s)
Cell Movement , Cell Proliferation , Colorectal Neoplasms/metabolism , Epithelial-Mesenchymal Transition , Lymphokines/metabolism , Neovascularization, Pathologic , Nuclear Proteins/metabolism , Platelet-Derived Growth Factor/metabolism , Receptor, Notch1/metabolism , Twist-Related Protein 1/metabolism , Animals , Antigens, CD , Cadherins/genetics , Cadherins/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition/drug effects , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , HT29 Cells , Humans , Lymphokines/genetics , Male , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Nuclear Proteins/genetics , Platelet-Derived Growth Factor/genetics , RNA Interference , Receptor, Notch1/genetics , Signal Transduction , Time Factors , Transfection , Transforming Growth Factor beta1/pharmacology , Twist-Related Protein 1/genetics , Vimentin/genetics , Vimentin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...