Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
J Cell Mol Med ; 28(7): e18190, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38494844

ABSTRACT

Systemic lupus erythematosus (SLE), a multifactorial autoimmune disease, can affect the brain and cause neuropsychiatric dysfunction, also named neuropsychiatric lupus (NPSLE). Microglial activation is observed in NPSLE patients. However, the mechanisms regulating microglia-mediated neurotoxicity in NPSLE remain elusive. Here, we showed that M1-like proinflammatory cytokine levels were increased in the cerebrospinal fluid (CSF) of SLE patients, especially those with neuropsychiatric symptoms. We also demonstrated that MRL/lpr lupus mice developed anxiety-like behaviours and cognitive deficits in the early and active phases of lupus, respectively. An increase in microglial number was associated with upregulation of proinflammatory cytokines in the MRL/lpr mouse brain. RNA sequencing revealed that genes associated with phagocytosis and M1 polarization were upregulated in microglia from lupus mice. Functionally, activated microglia induced synaptic stripping in vivo and promoted neuronal death in vitro. Finally, tofacitinib ameliorated neuropsychiatric disorders in MRL/lpr mice, as evidenced by reductions in microglial number and synaptic/neuronal loss and alleviation of behavioural abnormalities. Thus, our results indicated that classically activated (M1) microglia play a crucial role in NPSLE pathogenesis. Minocycline and tofacitinib were found to alleviate NPSLE by inhibiting micrglial activation, providing a promising therapeutic strategy.


Subject(s)
Lupus Erythematosus, Systemic , Lupus Vasculitis, Central Nervous System , Humans , Mice , Animals , Microglia , Depression/drug therapy , Mice, Inbred MRL lpr , Brain , Lupus Erythematosus, Systemic/genetics , Cytokines
2.
Front Immunol ; 14: 1231611, 2023.
Article in English | MEDLINE | ID: mdl-37841281

ABSTRACT

Introduction: Systemic lupus erythematosus (SLE) is an autoimmune disease involving many systems and organs, and individuals with SLE exhibit unique cancer risk characteristics. The significance of the basement membrane (BM) in the occurrence and progression of human autoimmune diseases and tumors has been established through research. However, the roles of BM-related genes and their protein expression mechanisms in the pathogenesis of SLE and pan-cancer development has not been elucidated. Methods: In this study, we applied bioinformatics methods to perform differential expression analysis of BM-related genes in datasets from SLE patients. We utilized LASSO logistic regression, SVM-RFE, and RandomForest to screen for feature genes and construct a diagnosis model for SLE. In order to attain a comprehensive comprehension of the biological functionalities of the feature genes, we conducted GSEA analysis, ROC analysis, and computed levels of immune cell infiltration. Finally, we sourced pan-cancer expression profiles from the TCGA and GTEx databases and performed pan-cancer analysis. Results: We screened six feature genes (AGRN, PHF13, SPOCK2, TGFBI, COL4A3, and COLQ) to construct an SLE diagnostic model. Immune infiltration analysis showed a significant correlation between AGRN and immune cell functions such as parainflammation and type I IFN response. After further gene expression validation, we finally selected AGRN for pan-cancer analysis. The results showed that AGRN's expression level varied according to distinct tumor types and was closely correlated with some tumor patients' prognosis, immune cell infiltration, and other indicators. Discussion: In conclusion, BM-related genes play a pivotal role in the pathogenesis of SLE, and AGRN shows immense promise as a target in SLE and the progression of multiple tumors.


Subject(s)
Autoimmune Diseases , Interferon Type I , Lupus Erythematosus, Systemic , Neoplasms , Humans , Computational Biology , DNA-Binding Proteins , Interferon Type I/physiology , Neoplasms/genetics , Proteoglycans , Risk Factors , Transcription Factors , Agrin/metabolism
3.
Front Endocrinol (Lausanne) ; 13: 939303, 2022.
Article in English | MEDLINE | ID: mdl-36147579

ABSTRACT

Objective: To explore the value of the optimal parameters of shear wave elastography (SWE) to enhance the identification of benign and malignant thyroid nodules by C-TIRADS. Methods: The two-dimensional ultrasonography images and SWE images of 515 patients with a total of 586 thyroid nodules were retrospectively analyzed. The nodules were divided into the D ≤10 mm and D >10 mm groups according to size and were graded by C-TIRADS. With the pathological results as the gold standard, the receiver operating characteristic (ROC) curves were drawn, and the area under the curve (AUC) was calculated to compare the diagnostic performances of C-TIRADS, SWE, and the combination of the two on the benign and malignant thyroid nodules. Results: The ROC showed that the AUC of the maximum elastic modulus (0.875) was higher than that of the mean elastic modulus (0.798) and elasticity ratio (0.772), with an optimal cutoff point of 51 kPa, which was the optimal parameter to distinguish the malignant from the benign nodules (P < 0.001). In the D ≤10 mm group, the AUC of TIRADS combined with SWE (0.955) was elevated by 0.172 compared with the application of C-TIRADS alone (0.783), and the difference was statistically significant (P < 0.05). In the D >10 mm group, the AUC of TIRADS combined with SWE (0.904) was elevated by 0.076 compared with the application of C-TIRADS alone (0.828), and the difference was statistically significant (P < 0.05). Among all nodules, the application of C-TIRADS alone had a sensitivity of 88.14%, a specificity of 74.56%, and an accuracy of 85.50% in diagnosing benign and malignant thyroid nodules, while the sensitivity, specificity, and accuracy were 93.22%, 90.35%, and 92.66%, respectively, in combination with SWE. Conclusion: The diagnostic performance of SWE in combination with TIRADS was better than that of SWE or C-TIRADS alone. Here, SWE enhanced the diagnostic performance of C-TIRADS for the benign and malignant thyroid nodules, most significantly for nodules with D ≤10 mm.


Subject(s)
Elasticity Imaging Techniques , Thyroid Nodule , Elasticity Imaging Techniques/methods , Humans , ROC Curve , Retrospective Studies , Thyroid Nodule/diagnostic imaging , Thyroid Nodule/pathology , Ultrasonography/methods
4.
Int J Gen Med ; 14: 5665-5673, 2021.
Article in English | MEDLINE | ID: mdl-34557021

ABSTRACT

OBJECTIVE: To investigate the diagnostic performance of the ADNEX model in the International Ovarian Tumor Analysis diagnostic models for ovarian tumors and further explore its application value in the staging of ovarian tumors. METHODS: A total of 224 patients who underwent ultrasound for evaluation of adnexal masses and were treated surgically owing to adnexal masses from January 2018 to June 2020 in our hospital were selected for research on the diagnostic accuracy of the ADNEX model. The clinical information and ultrasonographic findings of the patients were collected, and the pathological diagnosis was taken as the gold standard. According to the ADNEX model, the ovarian tumors were divided into five subtypes: benign and borderline, stage I, stage II-IV, and metastatic cancer. The sensitivity, specificity, positive predictive value, negative predictive value, diagnostic odds ratio, and area under the receiver operating characteristics curve (AUC) of the ADNEX model were calculated. RESULTS: Of the 224 patients, 119 (53.1%) developed benign tumors and 105 (46.9%) had malignant tumors. When the cut-off value for malignancy risk was 10%, the ADNEX model including CA 125 achieved a sensitivity of 94.3% (95% CI: 88.0-97.9%), specificity of 74.0% (95% CI: 65.1-81.6%), positive predictive value of 76.2% (95% CI: 70.2-81.3%), negative predictive value of 93.6% (95% CI: 87.0-97.0%), diagnostic odds ratio of 45.25, and an AUC of 0.94 (95% CI: 0.90-0.97) for differentiating between benign and malignant ovarian tumors. The AUC in the model excluding CA 125 was 0.93 (95% CI: 0.89-0.96), but the difference was not statistically significant (P=0.20). The accuracy of the ADNEX model for the diagnosis of ovarian tumors of all subtypes exceeds 80% when CA 125 measurements were included in the application, but the sensitivity for diagnosing borderline, stage I, and metastatic ovarian tumors was only 60.0% (95% CI:36.1-80.9%), 28.6% (95% CI:8.4-58.1%) and 45.5% (95% CI:16.7-76.6%). CONCLUSION: The ADNEX model shows good diagnostic performance in differentiating between benign and malignant ovarian tumors. The model has a certain clinical value in the diagnosis of all subtypes of ovarian tumors, but the sensitivity is unsatisfactory for the diagnosis of borderline, stage I, and metastatic ovarian tumors and needs to be verified.

5.
Mol Ther Nucleic Acids ; 23: 1110-1119, 2021 Mar 05.
Article in English | MEDLINE | ID: mdl-33664992

ABSTRACT

Ovarian cancer (OC) is a type of cancer with high prevalence and shocking mortality in women around the world. Radioresistance is a major reason for OC relapse. Mounting studies have shown the significant function of dysregulated microRNAs (miRNAs) in cancer progression and the cellular response to irradiation. The present study inquired about the function and mechanism of microRNA (miR)-4478 in regulating radiosensitivity of OC cells. Results showed that miR-4478 was downregulated in OC, and a low miR-4478 level indicated a disappointing prognosis for OC patients. Besides, in OC cells exposed to irradiation, the expression of miR-4478 decreased over time. Functionally, the upregulation of miR-4478 retarded OC cell proliferation and sensitized OC cells to irradiation. Mechanistically, miR-4478 targeted and inhibited fused in sarcoma (Fus). Additionally, Fus was upregulated in OC and its expression further elevated in OC cells under irradiation. Furthermore, miR-4478 targeted Fus to inhibit autophagy, therefore sensitizing OC cells to irradiation. Collectively, our study uncovered miR-4478 as a novel radiosensitizer by targeting Fus in OC cells, which may shed a new light on developing targets for treating patients with OC, particularly those with radioresistance.

6.
Sci Technol Adv Mater ; 21(1): 856-866, 2021 Feb 01.
Article in English | MEDLINE | ID: mdl-33551680

ABSTRACT

We report the formulation of nanoassemblies (NAs) comprising C225 conjugates Au-PFH-NAs (C-Au-PFH-NAs) for low-intensity focused ultrasound diagnosis ablation of thyroid cancer. C-Au-PFH-NAs showed excellent stability in water, phosphate-buffered saline (PBS), and 20% rat serum. Transmission electron microscopy (TEM) images also revealed the effective construction of C-Au-PFH-NAs as common spherical assemblies. The incubation of C625 thyroid carcinoma with C-Au-PFH-NAs triggers apoptosis, as confirmed by flow cytometry analysis. The C-Au-PFH-NAs exhibited antitumour efficacy in human thyroid carcinoma xenografts, where histopathological results further confirmed these outcomes. Furthermore, we were able to use low-intensity focused ultrasound diagnosis imaging (LIFUS) to examine the efficiency of C-Au-PFH-NAs in thyroid carcinoma in vivo. These findings clearly show that the use of LIFUS agents with high-performance imaging in different therapeutic settings will have extensive potential for future biomedical applications.

7.
Front Cell Dev Biol ; 8: 559, 2020.
Article in English | MEDLINE | ID: mdl-32766240

ABSTRACT

The present study aimed to determine whether icariin could attenuate type 1 diabetic nephropathy (T1DN) induced by streptozotocin (STZ) after 4 weeks or not. Therefore, its therapeutic effect on diabetic kidney disease was investigated in view of reactive oxygen (ROS) and extracellular matrix (ECM) generation in human glomerular mesangial cells under high glucose. To establish the participation and the key role of GPER and Nrf2 in ECM deposition, a combination of G15 (antagonist of GPER) or siGPER and siNrf2 were performed, respectively. The results showed that T1DN can be significantly inhibited by oral icariin, evidenced by improvement of 24 h urinary volume, 24 h proteinuria, microalbuminuria, and histopathological changes of kidney. Icariin decreased the levels of intracellular superoxide anion, impeded the generation of fibronectin and increased the expression and activity of antioxidant enzymes in the human glomerular mesangial cells treated with high glucose. It acted as a GPER activator, increased dissociation of Nrf2/Keap1 complexes, combination of Keap1/p62 complexes, Nrf2 translocation to nuclear, Nrf2/ARE DNA binding activity, and ARE luciferase reporter gene activity in glomerular mesangial cells. The Nrf2 inhibitor ML385 or siNrf2 obviously abolished extracellular matrix (ECM) generation inhibited by icariin. Furthermore, icariin-induced Nrf2 activation was mainly dependent on p62-mediated Keap1 degradation, which functions as an adaptor protein during autophagy. The GPER antagonist G15 and siGPER obviously abolished the above effects by icariin. Taken together, the present study demonstrated that the therapeutic effects of icariin on type 1 diabetic nephropathy in rats via GPER mediated p62-dependent Keap1 degradation and Nrf2 activation.

8.
Onco Targets Ther ; 13: 7585-7598, 2020.
Article in English | MEDLINE | ID: mdl-32821122

ABSTRACT

BACKGROUND: Although gefitinib brings about tremendous advances in the treatment of non-small cell lung cancer (NSCLC) harboring epidermal growth factor receptor (EGFR) mutations, most of patients become incurable due to drug resistance. JuBei oral liquid (JB) has been widely used to treat pneumonia in clinic. Components of JB were reported to induce apoptosis in NSCLC, which indicated that JB could be a potential antitumor agent for NSCLC patients. In this study, we investigated the effect of JB on gefitinib-sensitive PC-9 and gefitinib-resistant PC-9/GR, H1975 cells as well as its underlying molecular mechanisms. METHODS: PC-9, PC-9/GR and H1975 cells were treated with JB, LY294002, SCH772984, gefitinib alone or in combination. Then, cell viability, colony formation, cell death, expression of mitochondria-dependent pathway proteins, expression of EGFR, PI3K/AKT, MAPK signal pathway proteins, Bcl-2 mitochondrial translocation, ROS generation and cell apoptosis were examined by MTT, colony forming, live/dead cell staining, Western blot, immunofluorescence and flow cytometry assay. RESULTS: Our results showed that JB significantly induced cell growth inhibition and apoptotic cell death in PC-9, PC-9/GR and H1975 cells. JB activated mitochondria-mediated apoptotic pathway through inhibiting Bcl-2 mitochondrial translocation while inducing Bax translocated into mitochondria along with accumulated ROS production, thereby increasing the release of cytochrome c, subsequently cleaving procaspase9 into cleaved-caspase9 and then cleaving procaspase3 into cleaved-caspase3. Furthermore, the employment of protein kinase inhibitors LY294002 and SCH772984 revealed that the induction of mitochondria-mediated apoptosis by JB was reliant on inactivation of PI3K/AKT and MAPK signal pathways. Moreover, JB could synergize with gefitinib to induce apoptosis in PC-9, PC-9/GR and H1975 cells. CONCLUSION: These data indicated that JB could be a potential therapeutic agent for NSCLC patients harboring EGFR mutations as well as those under gefitinib resistance.

9.
Cell Death Dis ; 11(7): 580, 2020 07 24.
Article in English | MEDLINE | ID: mdl-32709863

ABSTRACT

Cancer progression including proliferation, metastasis, and chemoresistance has become a serious hindrance to cancer therapy. This phenomenon mainly derives from the innate insensitive or acquired resistance of cancer cells to apoptosis. Ferroptosis is a newly discovered mechanism of programmed cell death characterized by peroxidation of the lipid membrane induced by reactive oxygen species. Ferroptosis has been confirmed to eliminate cancer cells in an apoptosis-independent manner, however, the specific regulatory mechanism of ferroptosis is still unknown. The use of ferroptosis for overcoming cancer progression is limited. Noncoding RNAs have been found to play an important roles in cancer. They regulate gene expression to affect biological processes of cancer cells such as proliferation, cell cycle, and cell death. Thus far, the functions of ncRNAs in ferroptosis of cancer cells have been examined, and the specific mechanisms by which noncoding RNAs regulate ferroptosis have been partially discovered. However, there is no summary of ferroptosis associated noncoding RNAs and their functions in different cancer types. In this review, we discuss the roles of ferroptosis-associated noncoding RNAs in detail. Moreover, future work regarding the interaction between noncoding RNAs and ferroptosis is proposed, the possible obstacles are predicted and associated solutions are put forward. This review will deepen our understanding of the relationship between noncoding RNAs and ferroptosis, and provide new insights in targeting noncoding RNAs in ferroptosis associated therapeutic strategies.


Subject(s)
Disease Progression , Ferroptosis/genetics , Neoplasms/genetics , Neoplasms/pathology , RNA, Untranslated/genetics , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Untranslated/metabolism , Reactive Oxygen Species/metabolism
10.
J Cancer ; 11(2): 334-344, 2020.
Article in English | MEDLINE | ID: mdl-31897229

ABSTRACT

Background: The clinical application of EGFR tyrosine kinase inhibitors is always accompanied by inevitable drug resistance. However, the mechanism remains elusive. In the present study, we investigate the involvement of MAPK/SREBP1 pathway in NSCLC gefitinib resistance and evaluate the synergistic effects of shenqi fuzheng injection (SFI) and gefitinib on NSCLC cells. Methods: To investigate the MAPK/SREBP1 pathway involved in gefitinib resistance, Western blotting was used to examine p-MEK, p-ERK and SREBP1 expression in PC-9 and PC-9/GR cells, MTT was used on cell proliferation, wound healing assay was used on cell migration. To detect the cooperative effects of SFI and gefitinib, clonogenic assay was used on cell proliferation. Apoptosis assay was analyzed by flow cytometry. Immunofluorescence was used to detect gefitinib binding to EGFR. Western blotting was used to detect whether SFI regulate the resistance to gefitinib via the suppression of MAPK/SREBP1 pathway. Results: Our results showed that MAPK/SREBP1 pathway mediated resistance to gefitinib in NSCLC cells. MAPK pathway was found to directly target SREBP1 and inhibition of SREBP1 increased gefitinib sensitivity. In addition, SFI showed cooperative anti-proliferation and pro-apoptosis impacts on gefitinib resistant cells via down-regulating MAPK/SREBP1 pathway. Moreover, the combination of SFI and gefitinib enhanced gefitinib binding to EGFR resulting in the restoration of sensitivity to gefitinib. Conclusions: Taken together, MAPK/SREBP1 pathway could be regarded as the potential treatment target for overcoming resistance to EGFR-TKIs in NSCLC and adjuvant therapy of SFI could be a potential therapeutic strategy for gefitinib resistant treatment.

11.
J Cell Physiol ; 235(4): 3425-3437, 2020 04.
Article in English | MEDLINE | ID: mdl-31556117

ABSTRACT

Glutathione peroxidase 4 (GPX4) has been confirmed to inhibit ferroptosis in cancer cells, however, whether GPX4 serves as an oncogene is not clear. In this study, the expression of GPX4 and its influence to survival of patients with cancer were analyzed via public databases. Furthermore, the epigenetic regulation of GPX4 and the relation between GPX4 and chemoresistance of different anticancer drugs was also detected. Most importantly, cytological assays were performed to investigate the function of GPX4 in cancer cells. The results showed that GPX4 was higher expressed in cancer tissues than normal and was negatively associated with prognosis of patients. Furthermore, at upstream of GPX4 there was low DNA methylation sites and enhanced level of H3K4me3 and H3K27ac, indicating that high level of GPX4 in cancer may resulted from epigenetic regulation. Moreover, GPX4 was positively related to chemoresistance of anticancer drugs L-685458, lapatinib, palbociclib, and topotecan. In addition, GPX4 may potentially be involved in translation of protein, mitochondrial respiratory chain complex I assembly, electron transport oxidative phosphorylation, nonalcoholic fatty liver disease, and metabolic pathways. Finally, we detected that GPX4 inhibited ferroptosis in cancer cells, the inhibition of GPX4 via RSL3 could enhance the anticancer effect of cisplatin in vitro and in vivo. In conclusion, GPX4 acts as an oncogene and inhibits ferroptosis in cancer cells, the anticancer effect of cisplatin can be enhanced by GPX4 inhibition.


Subject(s)
DNA Methylation/drug effects , Ferroptosis/genetics , Neoplasms/drug therapy , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , A549 Cells , Animals , Carbamates/pharmacology , Cell Movement/drug effects , Cisplatin/pharmacology , Dipeptides/pharmacology , Epigenesis, Genetic/drug effects , Ferroptosis/drug effects , Heterografts , Histones/genetics , Humans , Lapatinib/pharmacology , Mice , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasms/genetics , Neoplasms/pathology , Piperazines/pharmacology , Pyridines/pharmacology , Topotecan/pharmacology
12.
Cancer Med ; 8(14): 6358-6369, 2019 10.
Article in English | MEDLINE | ID: mdl-31466138

ABSTRACT

BACKGROUND: As an oncogene, long noncoding RNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) can promote tumor metastasis. Hyperexpression of MALAT1 has been observed in many malignant tumors, including hepatocellular carcinoma (HCC). However, the role and mechanism of MALAT1 in HCC remain unclear. METHODS: Thirty human HCC and paracancerous tissue specimens were collected, and the human hepatoma cell lines Huh7 and HepG2 were cultured according to standard methods. MALAT1 and Snail family zinc finger (Slug) expression were measured by real-time PCR, immunohistochemistry, and western blotting. Luciferase reporter assay and RNA immunoprecipitation (RIP) assay verified the direct interaction between miR-124-3p and Slug(SNAI2) or MALAT1. Wound healing and transwell assays were performed to examine invasion and migration, and a subcutaneous tumor model was established to measure tumor progression in vivo. RESULTS: MALAT1 expression was upregulated in HCC tissues and positively correlated with Slug expression. MALAT1 and miR-124-3p bind directly and reversibly to each other. MALAT1 silencing inhibited cell migration and invasion. miR-124-3p inhibited HCC metastasis by targeting Slug. CONCLUSIONS: MALAT1 regulates Slug through miR-124-3p, affecting HCC cell metastasis. Thus, the MALAT1/miR-124-3p/Slug axis plays an important role in HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , Liver Neoplasms/pathology , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Snail Family Transcription Factors/genetics , Adult , Aged , Animals , Cell Line, Tumor , Disease Models, Animal , Female , Gene Knockdown Techniques , Humans , Immunohistochemistry , Male , Mice , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Prognosis , RNA Interference , ROC Curve , Snail Family Transcription Factors/metabolism , Xenograft Model Antitumor Assays
13.
Oncol Lett ; 15(6): 9436-9442, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29805667

ABSTRACT

MicroRNAs (miRNAs/miRs), which are endogenous non-coding single-stranded RNAs 19-25 nucleotides in length, regulate gene expression by blocking translation or transcription repression. The present study revealed that miR-3160-5p was widely expressed in prostate cancer cells by reverse transcription-quantitative polymerase chain reaction. There was a negative association between the expression of miR-3160-5p and F-box and WD repeat domain containing 8 (Fbxw8) in prostate cancer DU145 cells. A luciferase activity assay was used to verify that Fbxw8 is the target of miR-3160-5p. In the present study, using MTT assay and cell cycle analysis, it was demonstrated that DU145 cell proliferation was repressed and the cell cycle was arrested in the G2/M cell cycle phase with upregulation of miR-3160-5p. Subsequent studies demonstrated that miR-3160-5p regulated the progression of the cell cycle in DU145 prostate cancer cells when the expression levels of phosphorylated cell division cycle (CDC)2, CDC25C and cyclin B1 were directly inhibited. Taken together, these findings revealed the mechanism underlying the role of miR-3160-5p in regulating the proliferation of DU145 cells and indicated that miR-3160-5p may serve as a promising novel therapeutic tool for prostate cancer.

14.
Oncol Lett ; 15(3): 2978-2984, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29435027

ABSTRACT

A number of studies have demonstrated that dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) may be used to evaluate microvessel density (MVD), and may quantitatively reflect tumor angiogenesis. To investigate the dynamics, including angiogenesis and tumor cellularity, of rabbit VX2 tumors during the 4 weeks following tumor implantation, the present study used DCE-MRI combined with diffusion-weighted imaging (DWI) to scan the tumors at 3 days, and then at 1, 2, 3 and 4-week intervals, following tumor implantation. The dynamics, volume transfer coefficient (Ktrans) and apparent diffusion coefficient (ADC) of the tumor parenchyma were analyzed. Furthermore, the associations between Ktrans and MVD at 4 weeks after tumor implantation were analyzed. Tumor Ktrans was positively correlated with MVD at 4 weeks (r=0.674, P<0.001). Following tumor implantation, the tumor Ktrans level rose for 2 weeks and then began to decline, reaching its lowest point at 4 weeks (P<0.001). ADC values at 1 week were higher than at 3 days, but declined thereafter (P<0.001). Tumor necrosis appeared by 1 week after tumor implantation. The necrosis degree of tumor was gradually increased from the occurrence of necrosis within the 4-week time span of the present study (1 vs. 2 weeks, P=0.008; 2 vs. 3 weeks, P<0.001; 3 vs. 4 weeks, P<0.001). The present study identified that tumor angiogenesis is a dynamic process that serves a function in tumor growth, and that DCE-MRI may reflect tumor parenchymal MVD and be useful in evaluating angiogenesis.

15.
Oncotarget ; 7(40): 65553-65567, 2016 10 04.
Article in English | MEDLINE | ID: mdl-27566588

ABSTRACT

EMT (epithelial-mesenchymal transition) occurs in a wide range of tumor types, and has been shown to be crucial for metastasis. Epigenetic modifications of histones contribute to chromatin structure and result in the alterations in gene expression. Tri-methylation of histone H3 lysine 4 (H3K4me3) is associated with the promoters of actively transcribed genes and can serve as a transcriptional on/off switch. RbBP5 is a component of the COMPASS/ -like complex, which catalyzes H3K4me3 formation. In this study, we found that in the process of TGF-Beta1 induced EMT in the prostate cancer cell line DU145, H3K4me3 enrichment and RbBP5 binding increased in the vicinity of Snail (SNAI1) transcription start site. Knocking-down of RbBP5 notably decreased Snail expression and EMT. Recruitment of RbBP5 and formation of H3K4me3 at Snail TSS during EMT depend on binding of SMAD2/3 and CBP at Snail TSS. This study links the SMAD2/3 signal with Snail transcription via a histone modification - H3K4me3. Furthermore, our research also demonstrates that RbBP5 and even WRAD may be a promising therapeutic candidates in treating prostate cancer metastasis, and that DU145 cells maintain their incomplete mesenchymal state in an auto/ paracrine manner.


Subject(s)
Histones/metabolism , Multiprotein Complexes/metabolism , Nuclear Proteins/metabolism , Prostatic Neoplasms/metabolism , Snail Family Transcription Factors/genetics , Cell Line, Tumor , DNA-Binding Proteins , Epigenesis, Genetic , Epithelial-Mesenchymal Transition/genetics , Humans , Male , Methylation , Nuclear Proteins/genetics , Peptide Fragments/metabolism , Prostatic Neoplasms/genetics , Protein Binding , RNA, Small Interfering/genetics , Sialoglycoproteins/metabolism , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transcription Initiation Site , Transforming Growth Factor beta/metabolism
16.
Oncol Rep ; 35(2): 1153-62, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26718770

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) endows cancer cells with enhanced invasive and metastatic potential during cancer progression. Fractalkine, also known as chemokine (C-X3-C motif) ligand 1 (CX3CL1), the only member recognized so far that belongs to the CX3C chemokine subfamily, was reported to participate in the molecular events that regulate cell adhesion, migration and survival of human prostate cancer cells. However, the relationship between CX3CL1 and EMT remains unknown. We treated DU145 and PC-3 cells with CX3CL1 under hypoxic conditions. The migration and invasion abilities of DU145 and PC-3 cells were detected by Transwell assays. Induction of EMT was verified by morphological changes in the DU145 and PC-3 cells and analysis of protein expression of EMT markers such as E-cadherin and vimentin. To identify the involved signaling pathway in CX3CL1-induced EMT, activation of epidermal growth factor receptor (EGFR) was measured using western blot analysis, and Slug expression was detected with or without an EGFR inhibitor prior to CX3CL1 treatment. Concentrations of soluble and total TGF-α in the CX3CL­treated DU145 cells were detected by ELISA. Additionally, we determined the involvement of the TACE/TGF-α/EGFR pathway in CX3CL1­induced EMT using RNA interference and specific inhibitors. CX3CL1 increased the migration and invasiveness of the DU145 and PC-3 cells, and resulted in characteristic alterations of EMT. Our results demonstrated that TACE/TGF-α/EGFR pathway activation and subsequent upregulation of Slug expression were responsible for CX3CL1­induced EMT, and contributed to the migration and inva-sion of prostate cancer cells. Inhibition of TACE/TGF-α/EGFR signaling reversed EMT and led to reduced migration and invasion abilities of the prostate cancer cells. We provide initial evidence that CX3CL1 exposure resulted in EMT occurrence and enhancement of cell migration and invasion through a mechanism involving activation of TACE/TGF-α/EGFR signaling. These findings revealed that CX3CL1 may serve as a new target for the treatment of prostate cancer.


Subject(s)
ADAM Proteins/physiology , Adenocarcinoma/pathology , Chemokine CX3CL1/physiology , Epithelial-Mesenchymal Transition/physiology , ErbB Receptors/physiology , Neoplasm Proteins/physiology , Prostatic Neoplasms/pathology , Signal Transduction/physiology , Transforming Growth Factor alpha/physiology , ADAM Proteins/genetics , ADAM17 Protein , Adenocarcinoma/metabolism , Cell Hypoxia , Cell Line, Tumor , Cell Movement , Humans , Male , Neoplasm Invasiveness , Neoplasm Metastasis , Prostatic Neoplasms/metabolism , RNA Interference , RNA, Small Interfering/genetics , Snail Family Transcription Factors , Transcription Factors/biosynthesis , Transcription Factors/genetics , Up-Regulation
17.
Mol Med Rep ; 12(6): 7907-14, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26496926

ABSTRACT

Hypoxia is a common phenomenon in prostate cancer, which leads to cell proliferation and tumor growth. Fractalkine (FKN) is a membrane­bound chemokine, which is implicated in the progression of human prostate cancer and skeletal metastasis. However, the association between FKN and hypoxia­induced prostate cancer cell proliferation remains to be elucidated. The present study demonstrated that hypoxia induced the expression and secretion of FKN in the DU145 prostate cancer cell line. Furthermore, inhibiting the activity of FKN with the anti­FKN FKN­specific antibody markedly inhibited hypoxia­induced DU145 cell proliferation. Under normoxic conditions, DU145 cell proliferation markedly increased following exogenous administration of human recombinant FKN protein, and the increase was significantly alleviated by anti­FKN, indicating the importance of FKN in DU145 cell proliferation. In addition, subsequent determination of cell cycle distribution and expression levels of two core cell cycle regulators, cyclin E and cyclin­dependent kinase (CDK)2, suggested that FKN promoted the G1/S phase transition by upregulating the expression levels of cyclin E and CDK2. The results of the present study demonstrated that hypoxia led to the upregulation of the secretion and expression of FKN, which enhanced cell proliferation by promoting cell cycle progression in the prostate cancer cells. These findings provide evidence of a novel function for FKN, and suggest that FKN may serve as a potential target for treating androgen­independent prostate cancer.


Subject(s)
Cell Proliferation/genetics , Chemokine CX3CL1/physiology , Prostatic Neoplasms/pathology , Up-Regulation , Cell Hypoxia , Cell Line, Tumor , Chemokine CX3CL1/genetics , Chemokine CX3CL1/metabolism , Cyclin E/metabolism , Cyclin-Dependent Kinase 2/metabolism , G1 Phase Cell Cycle Checkpoints , Gene Expression Regulation, Neoplastic , Humans , Male , Tumor Microenvironment
18.
PLoS One ; 10(10): e0139865, 2015.
Article in English | MEDLINE | ID: mdl-26447479

ABSTRACT

Sensing and responding to endogenous electrical fields are important abilities for cells engaged in processes such as embryogenesis, regeneration and wound healing. Many types of cultured cells have been induced to migrate directionally within electrical fields in vitro using a process known as galvanotaxis. The underlying mechanism by which cells sense electrical fields is unknown. In this study, we assembled a polydimethylsiloxane (PDMS) galvanotaxis system and found that mouse fibroblasts and human prostate cancer PC3 cells migrated to the cathode. By comparing the effects of a pulsed direct current, a constant direct current and an anion-exchange membrane on the directed migration of mouse fibroblasts, we found that these cells responded to the ionic flow in the electrical fields. Taken together, the observed effects of the calcium content of the medium, the function of the store-operated calcium channels (SOCs) and the intracellular calcium content on galvanotaxis indicated that calcium ionic flow from the anode to the cathode within the culture medium permeated the cells through SOCs at the drift velocity, promoting migration toward the cathode. The RTK-PI3K pathway was involved in this process, but the ROCK and MAPK pathways were not. PC3 cells and mouse fibroblasts utilized the same mechanism of galvanotaxis. Together, these results indicated that the signaling pathway responsible for cathode-directed cellular galvanotaxis involved calcium ionic flow from the anode to the cathode within the culture medium, which permeated the cells through SOCs, causing cytoskeletal reorganization via PI3K signaling.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Electricity , Animals , Calcium Channels/chemistry , Calcium Channels/genetics , Cell Line , Cell Movement , Electrochemical Techniques/instrumentation , Electrodes , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Ions/chemistry , Mice , ORAI1 Protein , Phosphatidylinositol 3-Kinases/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction
19.
Oncol Rep ; 33(5): 2622-30, 2015 May.
Article in English | MEDLINE | ID: mdl-25812694

ABSTRACT

OCT4 is an essential transcription factor for maintaining the self-renewal and the pluripotency of embryonic stem cells (ESCs). The human OCT4 gene can generate three mRNA isoforms (OCT4A, OCT4B and OCT4B1) by alternative splicing. OCT4A protein is a transcription factor for the stemness of ESCs, while the function of OCT4B isoforms remains unclear. Most types of cancer express a relatively low level of OCT4 protein, particularly the OCT4B isoforms. In the present study, we found that OCT4A and OCT4B mRNA were co-expressed in several types of tumor cell lines and tumor samples, and we demonstrated that OCT4B functioned as a non-coding RNA, modulating OCT4A expression in an miRNA-dependent manner [competing endogenous RNA (ceRNA) regulation] at the post-transcription level in the tumor cell lines. This is the first time that ceRNA regulation was observed among spliced isoforms of one gene, and may pave the way for identification of new targets for cancer treatment.


Subject(s)
Octamer Transcription Factor-3/genetics , RNA, Messenger/genetics , RNA, Untranslated/genetics , Alternative Splicing/genetics , Cell Line, Tumor , HCT116 Cells , Humans , Protein Isoforms/genetics , RNA Processing, Post-Transcriptional/genetics , Transcription Factors/genetics
20.
Mol Med Rep ; 10(6): 3169-76, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25310235

ABSTRACT

Prostate cancer is the most commonly diagnosed type of cancer and the second leading cause of cancer­associated mortality in males. The efficacy of prostate cancer chemotherapy is frequently impaired by drug resistance; however, the underlying mechanisms of this resistance remain elusive. Sex determining region Y-box 2 (Sox2) is of vital importance in the regulation of stem cell proliferation and carcinogenesis. In the present study, using MTT, clone formation, cell cycle and apoptosis assays, over-expression of Sox2 was demonstrated to enhance the paclitaxel (Pac) resistance of the PC-3 prostate cancer cell line, promoting cell proliferation and exhibiting an anti­apoptotic effect. Western blot analysis revealed that the phosphoinositide 3-kinase/Akt signaling pathway was activated in cells overexpressing Sox2, and by targeting cyclin E and survivin, Sox2 promoted G1/S phase transition and prevented apoptosis under Pac treatment. The present study provided an understanding of Pac resistance in prostate cancer and may indicate novel therapeutic methods for chemoresistant prostate cancer.


Subject(s)
Drug Resistance, Neoplasm/genetics , Paclitaxel/pharmacology , Phosphatidylinositol 3-Kinases/genetics , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins c-akt/genetics , SOXB1 Transcription Factors/genetics , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cyclin E/genetics , G1 Phase/drug effects , G1 Phase/genetics , Humans , Inhibitor of Apoptosis Proteins/genetics , Male , S Phase/drug effects , S Phase/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Survivin
SELECTION OF CITATIONS
SEARCH DETAIL
...