Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
PeerJ ; 11: e16166, 2023.
Article in English | MEDLINE | ID: mdl-37790630

ABSTRACT

Background: Lung cancer has the highest cancer-related mortality worldwide. Lung adenocarcinoma (LUAD) is the most common histological subtype of non-small cell lung cancer (NSCLC). Chromatin licensing and DNA replication factor 1 (CDT1), a key regulator of cell cycle control and replication in eukaryotic cells, has been implicated in various cancer-related processes. Given its significant role in cancer, the focus on CDT1 in this study is justified as it holds promise as a potential biomarker or therapeutic target for cancer treatment. However, its prognostic value in lung adenocarcinoma (LUAD) remains unclear. Methods: Bioinformatics analysis was conducted using data obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) databases were utilized to predict biological processes and signaling pathways, respectively. The LinkedOmics database was employed to identify differentially expressed genes (DEGs) associated with CDT1. Nomograms and Kaplan-Meier plots were generated to assess the survival rates of patients with lung adenocarcinoma (LUAD). To determine the RNA and protein expression levels of CDT1 in LUAD and adjacent normal tissues, quantitative polymerase chain reaction (qPCR) and immunohistochemistry techniques were employed, respectively. Results: CDT1 was upregulated in the vast majority of cancer tissues, based on pan-cancer analysis in TCGA and GEO datasets, as to lung cancer, the level of CDT1 expression was much higher in LUAD tissue than in healthy lung tissue. Our clinical data supported these findings. In our study, we used a specific cutoff value to dichotomize the patient samples into high and low CDT1 expression groups. The Kaplan-Meier survival curve revealed poor survival rates in CDT1 high expression group than the low expression group. To determine if CDT1 expression was an independent risk factor in LUAD patients, univariate and multivariate Cox regression analyses were performed. The result showed that CDT1 was a potential novel prognosis factor for LUAD patients, whose prognosis was poorer when CDT1 expression was higher. Based on functional enrichment analysis, highly expressed DEGs of CDT1-high patients were predicted to be involved in the cell cycle. According to our analysis of immune infiltration, CDT1 exhibited a strong correlation with specific immune cell subsets and was found to be a significant predictor of poor survival in patients with LUAD. Conclusions: Our research found that CDT1 was upregulated in LUAD and that high CDT1 expression predicted poor prognosis. We comprehensively and systematically analyzed the expression level in the datasets as well as in our own clinical samples, we also evaluated the prognostic and diagnostic value of CDT1, and finally, the potential mechanisms of CDT1 in the progression of LUAD. These results suggested that CDT1 may be a prognostic marker and therapeutic target for LUAD.


Subject(s)
Adenocarcinoma of Lung , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Lung Neoplasms/diagnosis , Prognosis , Adenocarcinoma of Lung/genetics , Cell Cycle Proteins/genetics , Computational Biology
2.
Expert Rev Anticancer Ther ; 22(9): 999-1008, 2022 09.
Article in English | MEDLINE | ID: mdl-35833358

ABSTRACT

OBJECTIVES: Radioiodine-refractory differentiated thyroid cancer (RAI-rDTC) has frequently been associated with poor prognosis. We conducted a meta-analysis of published randomized controlled trials to evaluate multi-kinase inhibitors' efficacy and safety profile treatment. METHODS: A comprehensive search was conducted using PubMed, Embase, Cochrane, and Medline databases. The quality of literature and trial risk of bias was assessed using the Cochrane risk of bias tool, while the results of progression-free survival (PFS), overall survival (OS), and adverse events (AEs) were evaluated using RevMan5.3 software. RESULTS: Treatment with MKIs significantly improved PFS and OS, but AEs were significantly higher than those in the control group (P < 0.01). The studies demonstrated the median PFS (HR 0.30, 95% CI: 0.18-0.50, P < 0.00001) and OS (HR 0.70, 95% CI: 0.57-0.88, P = 0.002) in RAI-rDTC patients treated with MKIs, and the median PFS of papillary thyroid carcinoma (HR0.28, 95% CI: 0.22-0.37, P < 0.00001) along with follicular thyroid carcinoma (HR0.14, 95%CI 0.09-0.24, P < 0.00001) were extended. CONCLUSION: MKIs significantly prolonged PFS and OS in patients with RAI-rDTC (P < 0.01). Our recommendation is to use MKIs carefully in patients after evaluating their health status to maximize treatment benefits and minimize adverse effects.


Subject(s)
Antineoplastic Agents , Thyroid Neoplasms , Antineoplastic Agents/adverse effects , Humans , Iodine Radioisotopes/adverse effects , Progression-Free Survival , Thyroid Neoplasms/drug therapy
3.
Front Oncol ; 12: 896024, 2022.
Article in English | MEDLINE | ID: mdl-35619908

ABSTRACT

With the modern technological developments in the diagnosis and treatment of cancer, the survival rate of cancer patients has increased. On the other hand, the incidence of multiple primary tumors is increasing annually. Lynch syndrome (LS), an autosomal dominant disorder with germline mutations in DNA mismatch repair genes, increases the risk of cancer in patients carrying those mutations. In this report, we present an extremely rare case of an 81-year-old male patient with eight primary malignancies and LS. The patient is still alive having survived for more than 41 years since the initial discovery of the first tumor. The eighth and most recently diagnosed primary cancer was a malignant peripheral nerve sheath tumor. Although there have been numerous reports of malignancies in LS, malignant peripheral nerve sheath tumors have not been reported previously with LS. Here, we report, to the best of our knowledge, the first case of a malignant peripheral nerve sheath tumor with LS.

4.
Int J Colorectal Dis ; 37(1): 251-258, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34716473

ABSTRACT

BACKGROUND AND OBJECTIVE: Immune checkpoint inhibitor (ICI) therapies have shown promising prospects in colorectal cancer (CRC) immunotherapy; many clinical trials have been carried out. In this study, we sought to evaluate the efficacy and safety of ICI therapies in CRC by presenting a meta-analysis of relevant studies. METHODS: Databases including PubMed, Embase, Cochrane Library, and Web of Science were systematically searched for studies concerning the efficacy and safety of ICI in colorectal cancer. The reported odds ratio (OR) or weighted mean difference (WMD) with 95% confidence intervals (CIs) of overall survival (OS), progression-free survival (PFS), objective response rate (ORR), disease control rate (DCR), treatment-related adverse events (TRAEs), and TRAEs ≥ 3 in the included studies were analyzed by fixed effects/random effects models. RESULTS: Three studies involving 667 patients with colorectal cancer were included in our meta-analysis. No significant difference between the immune checkpoint inhibitor therapies and conventional therapies in OS (WMD = 0.73, 95% CI - 3.09, 4.54; p = 0.71), in ORR (OR = 1.54, 95% CI 0.98, 2.40; p = 0.06), and in DCR (OR = 0.97, 95% CI 0.36, 2.61; p = 0.95). The median PFS of the ICI therapy group was shorter than that of the conventional therapy group (WMD = - 0.10, 95% CI - 0.18, - 0.02; p = 0.02). At the same time, we also could not find a significant difference between the immune checkpoint inhibitor therapies and conventional therapies in TRAEs (OR = 1.56, 95% CI 0.11, 22.09; p = 0.74) and in TRAEs ≥ 3 (OR = 0.94, 95% CI 0.16, 5.65; p = 0.95). CONCLUSION: Immune checkpoint inhibitor therapies could not improve all survival endpoints to advanced or metastatic colorectal cancer patients. Whether immune checkpoint inhibitors should be the first choice of therapies for colorectal cancer patients with undetermined microsatellite status or not able to determine microsatellite status needs more related studies to prove.


Subject(s)
Colorectal Neoplasms , Immune Checkpoint Inhibitors , Colorectal Neoplasms/drug therapy , Humans , Immunotherapy/adverse effects , Progression-Free Survival
5.
Mol Med Rep ; 24(5)2021 11.
Article in English | MEDLINE | ID: mdl-34558633

ABSTRACT

Radiation­induced lung tissue injury is an important reason for the limited application of radiotherapy on thoracic malignancies. Previously, we reported that administration of Jiawei­Maxing­Shigan decoction (JMSD) attenuated the radiation­induced epithelial­mesenchymal transition (EMT) in alveolar epithelial cells (AECs) via TGF­ß/Smad signaling. The present study aimed to examine the role of protein phosphatase Mg2+/Mn2+­dependent 1A (PPM1A) in the anti­EMT activity of JMSD on AECs. The components in the aqueous extract of JMSD were identified by high­performance liquid chromatography coupled with electrospray mass spectrometry. Primary rat type II AECs were treated with radiation (60Co γ­ray at 8 Gy) and JMSD­medicated serum. PPM1A was overexpressed and knocked down in the AECs via lentivirus transduction and the effects of JMSD administration on the key proteins related to TGF­ß1/Smad signaling were measured by western blotting. It was found that radiation decreased the PPM1A expression in the AECs and JMSD­medicated serum upregulated the PPM1A expressions in the radiation­induced AECs. PPM1A overexpression increased the E­cadherin level but decreased the phosphorylated (p­)Smad2/3, vimentin and α­smooth muscle actin (α­SMA) levels in the AECs. By contrast, the PPM1A knockdown decreased the E­cadherin level and increased the p­Smad2/3, vimentin and α­SMA levels in the AECs and these effects could be blocked by SB431542 (TGF­ß1/Smad signaling inhibitor). JMSD administration increased the E­cadherin level and decreased the p­Smad2/3, vimentin and α­SMA levels in the AECs; however, these effects could be blocked by siPPM1A­2. In conclusion, PPM1A is a key target of JMSD administration for the attenuation of the radiation­induced EMT in primary type II AECs via the TGF­ß1/Smad pathway.


Subject(s)
Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/metabolism , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/radiation effects , Protein Phosphatase 2C/metabolism , Alveolar Epithelial Cells/radiation effects , Animals , Chromatography, High Pressure Liquid , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , Protein Phosphatase 2C/genetics , Rats , Smad Proteins/genetics , Smad Proteins/metabolism , Spectrometry, Mass, Electrospray Ionization
6.
Article in English | MEDLINE | ID: mdl-32256654

ABSTRACT

Irinotecan (CPT-11) is used for therapy of various cancers. However, it has several serious adverse reactions such as diarrhea which is caused by SN-38, the active form of CPT-11. This study aimed to evaluate the effectiveness of Jiawei xianglian decoction (JWXLD), which has been widely used for the treatment of diarrhea in China. In this study, a mouse model with delayed diarrhea was generated by CPT-11. Real-time PCR and enzyme-linked immunosorbent assay (ELISA) were performed to explore intestinal microflora and inflammatory cytokine. Hematoxylin and eosin (H&E) staining was used to analyze tissue morphology. We found that 0.12, 0.23, and 0.46 g JWXLD significantly reduced the severity of CPT-11-induced diarrhea. The levels of Lactobacillus (Lacto) and Bifidobacterium (Bifid) were significantly downregulated by CPT-11, and these effects can be reversed by JWXLD treatment. Furthermore, JWXLD was observed to decrease the activity of ß-glucuronidase (ß-GD). Histopathological data showed that CPT-11 induced severe intestinal mucosal injury, which was characterized as grade 6, and JWXLD significantly alleviated the injury. In addition, CPT-11 increased the productions of tumor necrosis factor-alpha (TNF-α), tumor necrosis factor-beta (TNF-ß), interleukin-6 (IL-6), and interleukin-1 (IL-1), but decreased interleukin-15 (IL-15), interleukin-7 (IL-7), and uridine diphosphate-glucuronosyltransferase 1A1 (UGT1A1). In conclusion, JWXLD can counteract these effects caused by CPT-11 treatment. JWXLD could alleviate CPT-11-induced diarrhea.

7.
Am J Transl Res ; 11(2): 733-743, 2019.
Article in English | MEDLINE | ID: mdl-30899375

ABSTRACT

Radiation-induced lung toxicity, including radiation pneumonitis and pulmonary fibrosis, often occurs in patients receiving radiation therapy. Epithelial-mesenchymal transition (EMT) of alveolar epithelial cells (AECs) plays critical roles in radiation-induced lung toxicity. In the present study, RNA sequencing was applied to examine the whole transcriptomes of human pulmonary AEC cells (HPAEpiC) with or without radiation treatment. We found that cytokine, chemokine and cell adhesion signaling pathways were enriched in radiation-treated cells. CCL2 (C-C Motif Chemokine Ligand 2), CCL5 and CCR4 (C-C Motif Chemokine Receptor 4) were among the top enriched genes in chemokine signaling pathway. The upregulation of CCL2, CCL5 and CCR4 in response to irradiation was confirmed at both mRNA and protein levels by real-time PCR, western blotting and enzyme-linked immunosorbent assay analyses. Ophiopogonin B, a bioactive ingredient of Radix Ophiopogon japonicas, was found to attenuate radiation-induced EMT in HPAEpiC cells as demonstrated by the alteration in cell morphology, and the expression of E-cadherin and Vimentin. Ophiopogonin B could also reduce radiation-induced expression of CCL2, CCL5, CCR4 and phosphorylated ERK (p-ERK). Moreover, CCR4 knockdown, U0126 (a MEK/ERK inhibitor) or ophiopogonin B also partially blocked the EMT promoting effects of CCL2 and CCL5. Our data suggested CCL2, CCL5 and CCR4 may be potential therapeutic targets for radiation-induced lung toxicity. Ophiopogonin B, which could down-regulate CCL2, CCL5 and CCR4, may be a useful radioprotective agent.

8.
J Cell Biochem ; 120(8): 12500-12507, 2019 08.
Article in English | MEDLINE | ID: mdl-30834595

ABSTRACT

Radiation-induced lung injury (RILI) frequently occurs in patients with thoracic malignancies. In response to radiation, alveolar epithelial cells (AEC) undergo epithelial-mesenchymal transition (EMT) and contribute to the pathogenesis of RILI. Insulin-like growth factor binding protein 7 (IGFBP7) is reported as a downstream mediator of transforming growth factor-ß1 (TGF-ß1) pathway, which plays a crucial role in radiation-induced EMT. In the present study, the levels of IGFBP7 and TGF-ß1 were simultaneously increased in experimental RILI models and radiation-treated AEC (human pulmonary alveolar epithelial cells [HPAEpic]). The expression of IGFBP7 in radiation-treated HPAEpic cells was obviously inhibited by the specific inhibitor of TGF-ß receptor antagonist SB431542 and TGF-ß1 neutralizing antibody, and time-dependently enhanced by TGF-ß1 treatment. Moreover, IGFBP7 knockdown significantly attenuated the effects of radiation on morphology change, cell migration, expression of EMT-related markers (E-cadherin, α-SMA, and Vimentin), and phosphorylation of extracellular-signal-regulated kinase (ERK). The effects of IGFBP7 overexpression on the expression of EMT-related markers were partially reversed by the ERK inhibitor PD98059. In conclusion, IGFBP7, was enhanced by TGF-ß1, may be involved in radiation-induced EMT of AEC via the ERK signaling pathway, thus contributing to the pathogenesis of RILI.


Subject(s)
Alveolar Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Insulin-Like Growth Factor Binding Proteins/metabolism , MAP Kinase Signaling System , Alveolar Epithelial Cells/physiology , Alveolar Epithelial Cells/radiation effects , Animals , Cell Line , Cell Movement , Humans , Insulin-Like Growth Factor Binding Proteins/physiology , Rats , Signal Transduction , Transforming Growth Factor beta1/metabolism
9.
Cell Physiol Biochem ; 50(2): 721-733, 2018.
Article in English | MEDLINE | ID: mdl-30308513

ABSTRACT

BACKGROUND/AIMS: Radiotherapy plays a critical role in lung cancer treatment. Radiation can activate transforming growth factor-ß (TGF-ß) signaling and induce the epithelial-mesenchymal transition (EMT), which may lead to distant metastases. MicroRNAs (miRNAs) have been suggested to affect radiotherapy in lung cancer. METHODS: miRNA Next-Generation Sequencing was performed to investigate the effects of irradiation on the miRNA profile of lung cancer A549 cells. The functions of identified miRNA on the radiation induced EMT and TGF-ß activation in A549 cells were then explored. Protein expression was evaluated by western blotting. Immunofluorescence staining was performed to detect the localization of Snail. Luciferase Assay was used to determine the target gene regulated by the identified miRNA. RESULTS: Radiation time-dependently induced EMT in A549 lung cancer cells as indicated by the changes of morphology, the expression of EMT marker proteins (E-cadherin, α-SMA and Vimentin) and the nuclear localization of Snail. Moreover, miR-3591-5p was identified as the most significant increased miRNA in response to radiation, and further experiments indicated that miR-3591-5p was required for radiation induced EMT and TGF-ß/ Smad2/3 activation. Ubiquitin Specific Peptidase 33 (USP33) was a downstream target of miR-3591-5p as predicted by TargetScan and validated by 3' untranslated regions (UTRs) Luciferase Assay. USP33 could deubiquitinate PPM1A (protein phosphatase, Mg2+/Mn2 + dependent 1A), a phosphatase for Smad2/3. Ectopic expression of USP33 or PPM1A partially abolished the effects of miR-3591-5p on EMT and TGF-ß/ Smad2/3 activation. CONCLUSION: The present study revealed the critical role of miR-3591-5p/USP33/PPM1A in radiation-induced EMT via TGF-ß signaling and may suggest novel radiation sensitise strategies for lung cancer.


Subject(s)
Epithelial-Mesenchymal Transition/radiation effects , MicroRNAs/metabolism , Protein Phosphatase 2C/metabolism , Radiation, Ionizing , Ubiquitin Thiolesterase/metabolism , 3' Untranslated Regions , A549 Cells , Actins/metabolism , Antagomirs/metabolism , Cadherins/metabolism , Down-Regulation/drug effects , Humans , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Protein Phosphatase 2C/genetics , Signal Transduction/radiation effects , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Ubiquitin Thiolesterase/genetics , Ubiquitination , Vimentin/metabolism
10.
Biomed Pharmacother ; 95: 1219-1224, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28931214

ABSTRACT

BACKGROUND: Radiation therapy is commonly used to treat thoracic malignancies. However, it may lead to severe lung pneumonitis and ultimately fibrosis. Irradiation has been reported to increase epithelial-mesenchymal transition (EMT) of type II alveolar epithelial cells (AEC), which play an important role in pulmonary fibrosis. The transforming growth factor-ß (TGF-ß) and ERK/glycogen synthase kinase 3ß (GSK3ß) pathways are critically involved in radiation-induced EMT. In the present study, we investigated whether baicalin was a novel therapeutic candidate for radiation-induced EMT in type II AEC. METHODS: Primary type II AEC were isolated and treated with 60Co γ-rays and a series doses of baicalin (2µM, 10µM and 50µM). The ultrastructure and morphology changes were observed by transmission electron microscopy and optical microscopy, respectively. Protein expression was determined by western blotting analysis. Immunofluorescence staining was performed to detect the nuclear translocation of Snail. RESULTS: After irradiation, type II AEC displayed a mesenchymal-like morphology accompanied by a decrease in E-cadherin expression, an increase in the expression of Vimentin and α-SMA. Nuclear translocation of Snail, the activation of TGF-ß/Smad pathway, and the inactivation of GSK3ß were prominent in radiation-treated cells. Baicalin significantly attenuated the effects of radiation on type II AEC. CONCLUSIONS: Baicalin may a useful radioprotective agent through suppressing the EMT of type II AEC.


Subject(s)
Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Epithelial-Mesenchymal Transition , Flavonoids/pharmacology , Glycogen Synthase Kinase 3 beta/metabolism , MAP Kinase Signaling System/drug effects , Transforming Growth Factor beta/metabolism , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/radiation effects , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Gamma Rays , Protein Transport/drug effects , Rats, Sprague-Dawley , Smad Proteins/metabolism , Snail Family Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...