Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Sheng Li Xue Bao ; 74(5): 783-791, 2022 Oct 25.
Article in Chinese | MEDLINE | ID: mdl-36319101

ABSTRACT

As COVID-19 pandemic approaching its third year, more data have shown that obesity and hypertriglyceridemia are the high-risk factors for the major complications such as acute respiratory distress syndrome, thromboembolism, coagulopathy and cytokine storm, which are responsible for the majority of disease severity and mortality. In this review article, we have analyzed the public available clinical reports and laboratory research results of the COVID-19 studies by researchers and clinicians of many nations around the world. Many of these reports covered COVID-19 patients of different ethnic groups. We suggested that obesity and high triglycerides are high risks for severe COVID-19 and death. We also summarized the possible underlying molecular mechanism likely connecting the severe COVID-19 with obesity and hypertriglyceridemia. From public health perspective, we highlight the importance of the healthy diet and lifestyle in fighting against SARS-CoV-2 virus in long period of time.


Subject(s)
COVID-19 , Hypertriglyceridemia , Humans , Pandemics , SARS-CoV-2 , Obesity , Severity of Illness Index
2.
Front Immunol ; 13: 1013049, 2022.
Article in English | MEDLINE | ID: mdl-36389799

ABSTRACT

Background: Thyroid cancer (THCA) is the most prevalent malignant disease of the endocrine system, in which 5-year survival can attain about 95%, but patients with metastasis have a poor prognosis. Very little is known about the role of CAPN8 in the metastasis of THCA. In particular, the effect of CAPN8 on the tumor immune microenvironment (TIME) and immunotherapy response is unclear. Material and methods: Multiome datasets and multiple cohorts were acquired for analysis. Firstly, the expression and the prognostic value of CAPN8 were explored in public datasets and in vitro tumor tissues. Then, hierarchical clustering analysis was performed to identify the immune subtypes of THCA according to the expression of CAPN8 and the activities of related pathways. Subsequent analyses explored the different patterns of TIME, genetic alteration, DNA replication stress, drug sensitivity, and immunotherapy response among the three immune phenotypes. Finally, five individual cohorts of thyroid cancer were utilized to test the robustness and extrapolation of the three immune clusters. Results: CAPN8 was found to be a significant risk factor for THCA with a markedly elevated level of mRNA and protein in tumor tissues. This potential oncogene could induce the activation of epithelial-mesenchymal transition and E2F-targeted pathways. Three subtypes were identified for THCA, including immune exhausted, inflamed, and immune desert phenotypes. The exhausted type was characterized by a markedly increased expression of inhibitory receptors and infiltration of immune cells but was much more likely to respond to immunotherapy. The immune desert type was resistant to common chemotherapeutics with extensive genomic mutation and copy number variance. Conclusion: The present study firstly explored the role of CAPN8 in the metastasis of THCA from the aspects of TIME. Three immune subtypes were identified with quite different patterns of prognosis, immunotherapy response, and drug sensitivity, providing novel insights for the treatment of THCA and helping understand the cross-talk between CAPN8 and tumor immune microenvironment.


Subject(s)
Thyroid Neoplasms , Humans , Thyroid Neoplasms/genetics , Tumor Microenvironment , Prognosis , Immunotherapy , DNA Copy Number Variations
3.
Am J Transl Res ; 14(8): 5441-5454, 2022.
Article in English | MEDLINE | ID: mdl-36105009

ABSTRACT

OBJECTIVE: Breast cancer, as a malignancy with the highest incidence and mortality in women, seriously threatens women's life and health. Pieces of evidence have suggested that long non-coding RNAs (lncRNAs) possess important roles in regulating the occurrence and development of breast cancer. METHODS: RT-qPCR was used to explore the expression levels of MIR4435-2HG, miR-22-3p and TMEM9B in breast cancer tissues and cell lines. Cell viability, proliferation, migration and invasion were assessed by CCK-8 assay, Colony formation assay, Wound healing assay and Transwell assay, respectively. The effect of MIR4435-2HG on EMT progress was explored by Immunofluorescence assay and Western blot. RNA pull-down analysis and Dual-luciferase reporter assay were performed to validate the interaction between MIR4435-2HG and miR-22-3p, as well as miR-22-3p and TMEM9B. RESULTS: MIR4435-2HG was notably up-regulated in breast cancer tissues and cell lines. Additionally, down-regulation of MIR4435-2HG restrained the viability, proliferation, migration, invasion and EMT of breast cancer cells. MiR-22-3p expression was down-regulated in breast cancer tissues and cell lines, and negatively associated with MIR4435-2HG expression. Over-expression of miR-22-3p obviously inhibited the viability, proliferation, migration, invasion and EMT of breast cancer cell lines. Furthermore, TMEM9B was up-regulated in breast cancer tissues and cell lines and negatively associated with miR-22-3p expression. TMEM9B inhibition partially restored the effects of MIR4435-2HG/miR-22-3p on the viability, proliferation, migration, invasion and EMT of breast cancer cell lines. CONCLUSION: MIR4435-2HG plays a potential tumor-promoting role in the occurrence and development of breast cancer, possibly by regulating the miR-22-3p/TMEM9B axis.

4.
Ann Transl Med ; 10(15): 833, 2022 Aug.
Article in English | MEDLINE | ID: mdl-36035010

ABSTRACT

Background: Ferroptosis is a kind of programmed cell death that is characterized by iron dependence. It differs from apoptosis, necrosis, autophagy, pyroptosis, and other types of cell death. Some studies have found that most of the genes involved in the regulation of ferroptosis or act as markers of ferroptosis are related to the poor prognosis of cancer patients. Methods: This study evaluated the expression, mutation, and copy number variation (CNV) of 60 previously reported ferroptosis genes in breast cancer samples from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. Unsupervised clustering of breast cancer samples with ferroptosis genes was performed, followed by enrichment analysis with Gene Set Variation Analysis (GSVA), mutation display, and correlation analysis of clinical characteristics. Based on the analysis of differences among groups, the ferroptosis-related genes were identified, and the consistent clustering of breast cancer samples was performed. The characteristic genes were screened by stochastic forest algorithm and COX analysis, and a ferroptosis score (ferr.score) model was constructed to evaluate the prognosis of breast cancer patients. Results: Copy number amplification and deletion of ferroptosis genes are common in breast cancer. Breast cancer patients grouped by ferroptosis gene clusters showed significant differences in survival, immune cell infiltration, and enriched Kyoto Encyclopedia of Genes and Genomes (KEGG) signaling pathways. The ferroptosis-related differential genes were identified by comparison among clustering groups of ferroptosis gene. Characteristic genes were screened from these ferroptosis-related differential genes to construct the ferr.score model. The scoring model could accurately distinguish and predict the survival prognosis and immunotherapy efficacy in breast cancer patients. Conclusions: Ferroptosis plays an important role in the occurrence and development of tumors. According to the ferr.score model, the breast cancer samples can be divided into two groups with significantly different prognoses. These results provide novel insights and ideas for immunotherapy in breast cancer patients.

5.
Transl Lung Cancer Res ; 11(4): 632-646, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35529787

ABSTRACT

Background: Ferroptosis is a novel iron-dependent cell death, and an increasing number of studies have shown that long non-coding RNA (lncRNAs) are involved in the ferroptosis process. However, studies on ferroptosis-related lncRNAs in lung squamous cell carcinoma (LUSC) are limited. In addition, the prognostic role of ferroptosis-related lncRNAs and their relationship with the immune microenvironment and methylation of LUSC is unclear. This study aimed to investigate the potential prognostic value of ferroptosis-related lncRNAs and their involved biological functions in LUSC. Methods: The Cancer Genome Atlas (TCGA) database and the FerrDb website were used to obtain ferroptosis-related genes for LUSC. The "limma" R package and Pearson analysis were used to find ferroptosis-related lncRNAs. The biological functions of the characterized lncRNAs were analyzed by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). We evaluated the prognostic power of this model using Kaplan-Meier analysis, receiver operating characteristic (ROC), and decision curve analysis (DCA). Univariate and multifactor Cox (proportional-hazards) risk model and a nomogram were produced using risk models and clinicopathological parameters for further verification. In addition, the relationship between characterized lncRNAs and tumor immune infiltration and methylation was also discussed. Results: We identified 29 characterized lncRNAs to produce prognostic risk models. Kaplan-Meier analysis revealed the high-risk group was associated with poor prognosis in LUSC (P<0.001), and ROC (AUC =0.658) and DCA suggested that risk models could predict prognosis. Univariate and multifactorial Cox as well as nomogram further validated the prognostic model (P<0.001). Gene set enrichment analysis (GSEA) showed that the high-risk group was associated with pro-tumor pathways and high-frequency mutations in TP53 were present in both groups. Single sample gene set enrichment analysis (ssGSEA) showed significant differences in immune cell infiltration subtypes and corresponding functions between the two groups. Some immune checkpoint and methylation-related genes were significantly different between the two groups (P<0.05). Conclusions: We investigated the potential mechanisms of LUSC development from the perspective of ferroptosis-related lncRNAs, providing new insights into LUSC research, and identified 29 lncRNAs as biomarkers to predict the prognosis of LUSC patients.

6.
Ann Transl Med ; 8(8): 559, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32775360

ABSTRACT

[This corrects the article DOI: 10.21037/atm.2019.11.105.].

7.
Ann Transl Med ; 8(7): 519, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32395563

ABSTRACT

[This corrects the article DOI: 10.21037/atm.2019.11.106.].

8.
Ann Transl Med ; 7(22): 681, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31930082

ABSTRACT

BACKGROUND: CDK14 has significant involvement in tumorigenesis of cancers including hepatocellular carcinoma, gastric carcinoma and breast cancer. In esophageal cancer, CDK14 is useful as a prognostic marker and as a predictor of response to chemotherapy. However, the exact mechanism of CDK14 n chemotherapy for esophageal squamous cell carcinoma (ESCC) has not been explored. METHODS: Western blots and immunohistochemistry (IHC) analysis were performed to analyse the expression of CDK14 in ESCC. Co-immunoprecipitation and immunofluorescence assays were used to explore the mechanism of CDK14 involvement in ESCC. Colony formation assays and proliferation assays were used to investigate the function of CDK14 in ESCC. At last, we constructed two truncated mutants of CDK14 by the PCR technology to research the functional structural domain. RESULTS: Western blots and IHC analysis showed that CDK14 expression was higher n tumor tissues and cell lines than that in normal tissues. IHC staining revealed that CDK14 positively correlated with clinical pathological variables of tumor size (P=0.001), tumor grade (P=0.004), Ki-67 (P=0.012) and survival (P=0.000). Immunoprecipitation and immunofluorescence assays revealed that CDK-activating kinase (CAK), namely CDK7/CCNH complex physically interacted and was collocated with CDK14 in the cell nucleus. This direct interaction increased CDK14 phosphorylation and inhibited Rb function through phosphorylation. In vitro starvation and refeeding assays demonstrated that CDK14 expression was related to proliferation of ESCC cells. Overexpression of CDK14 in Eca109 cells increased colony formation and reduced sensitivity to cisplatin. Overexpressing CDK7 with CDK14 strengthened these effects, demonstrating that CDK7 was a major component in CDK14 activation. CONCLUSIONS: Expression of CDK14 worsened the effects of cisplatin chemotherapy by promoting ESCC proliferation.

9.
Ann Transl Med ; 7(22): 682, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31930083

ABSTRACT

BACKGROUND: Aberrant DNA methylation plays a crucial part in cancer progression through the silencing of gene expression. The purpose of this article was to investigate the DNA methylation-driven genes in breast invasive carcinoma (BRCA) by using integrated bioinformatics analysis and in vitro experiments. METHODS: The methylation and expression profile data of BRCA patients were downloaded from the TCGA database. Besides, the MethylMix algorithm was performed to distinguish differentially methylation-driven genes. Moreover, methylation-specific PCR was used to test the methylation-driven genes. RESULTS: A total of 218 differentially expressed methylation-driven genes were obtained. Then, four of these genes were applied to establish a prognostic risk model. Moreover, we found that hypermethylation was in the CpG islands of the promoter of COX7A1 gene in BRCA tissues. Furthermore, we found that COX7A1 was significantly down-regulated BRCA tissues and the COX7A1 expression level was markedly increased in BRCA cells after 5-Aza-dC treatment. CONCLUSIONS: Our study reveals that aberrant promoter hypermethylation is critical for COX7A1 gene silencing in BRCA and that COX7A1 emerge as a new biomarker and therapeutic target for BRCA.

10.
Ann Transl Med ; 7(22): 683, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31930084

ABSTRACT

BACKGROUND: This study analyzed the clinical data and general information of breast cancer patients who were admitted by the Affiliated Hospital of Nantong University and underwent lumpectomy, followed by sentinel lymph node biopsy (SLNB) to investigate the effect of tumor location on the sentinel lymph node (SLN) detection rate, obtain a clear understanding of the SLNB procedure and further promote the use of this procedure in the local area. METHODS: This study involved a total of 118 patients who were diagnosed with breast cancer and admitted by the Affiliated Hospital of Nantong University for lumpectomy and SLNB between July 2015 and June 2019. An analysis was conducted to explore the role of tumor location in the detection of SLNs. RESULTS: Tumor location was associated with the success rate of post-lumpectomy SLNB. In the case of tumor location in the upper outer quadrant (UOQ) of the breast near the axilla, the SLN detection rate was relatively low. In contrast, when a tumor occurred in any of the other quadrants or the UOQ next to the areola, the tumor location had no significant impact on the SLN detection rate. SLNB indicated that 102 out of the 118 patients had SLNs, with the detection rate of 86.4%. Particularly, for patients whose tumors were located in the UOQ near their axillae, the SLN detection rate was 30% (3/10). As to tumor location in other quadrants or the UOQ next to the areola, the SLN detection rate was up to 90.8% (99/109). CONCLUSIONS: The performance of post-lumpectomy SLNB is associated with tumor location. SLNB is recommended when the tumor site lies in the upper inner/lower outer/lower inner quadrants (UIQ/LOQ/LIQ) of the breast or the UOQ next to the areola. If the SLNB result turns out to be negative, there is no need to perform axillary lymph node dissection (ALND). For tumor location in the UOQ of the breast, especially when it is near the axilla, SLNB is not a favorable option after lumpectomy. It is recommended that the patient receive a core needle biopsy (CNB) before SLNB.

11.
J Hematol Oncol ; 11(1): 17, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29422082

ABSTRACT

BACKGROUND: Chromobox protein homolog 7 (CBX7), a member of the polycomb group (PcG) family of proteins, is involved in the regulation of cell proliferation and cancer progression. PcG family members, such as BMI, Mel-18, and EZH2, are integral constituents of the polycomb repressive complexes (PRCs) and have been known to regulate cancer stem cell (CSC) phenotype. However, the role of other PRCs' constituents such as CBX7 in the regulation of CSC phenotype remains largely elusive. This study was to investigate the role of CBX7 in regulating stem cell-like properties of gastric cancer and the underlying mechanisms. METHODS: Firstly, the role of CBX7 in regulating stem cell-like properties of gastric cancer was investigated using sphere formation, Western blot, and xenograft tumor assays. Next, RNA interference and ectopic CBX7 expression were employed to determine the impact of CBX7 on the expression of CSC marker proteins and CSC characteristics. The expression of CBX7, its downstream targets, and stem cell markers were analyzed in gastric stem cell spheres, common cancer cells, and gastric cancer tissues. Finally, the pathways by which CBX7 regulates stem cell-like properties of gastric cancer were explored. RESULTS: We found that CBX7, a constituent of the polycomb repressive complex 1 (PRC1), plays an important role in maintaining stem cell-like characteristics of gastric cancer cells via the activation of AKT pathway and the downregulation of p16. Spearman rank correlation analysis showed positive correlations among the expression of CBX7 and phospho-AKT (pAKT), stem cell markers OCT-4, and CD133 in gastric cancer tissues. In addition, CBX7 was found to upregulate microRNA-21 (miR-21) via the activation of AKT-NF-κB pathway, and miR-21 contributes to CBX7-mediated CSC characteristics. CONCLUSIONS: CBX7 positively regulates stem cell-like characteristics of gastric cancer cells by inhibiting p16 and activating AKT-NF-κB-miR-21 pathway.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/genetics , Gene Expression Regulation, Neoplastic , Neoplastic Stem Cells/pathology , Polycomb Repressive Complex 1/metabolism , Signal Transduction , Stomach Neoplasms/pathology , Cell Line, Tumor , Humans , MicroRNAs/genetics , NF-kappa B/metabolism , Neoplastic Stem Cells/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism
12.
Dig Dis Sci ; 62(2): 387-395, 2017 02.
Article in English | MEDLINE | ID: mdl-27995408

ABSTRACT

BACKGROUND: BCCIP was originally identified as a BRCA2 interacting protein in humans and Ustilago maydis. It had low expression in some human cancer tissues. However, recent research indicated that many caretaker genes are also necessary for cell viability and their expression could contribute to tumor progression. AIM: To characterize whether BCCIP is a caretaker gene in esophageal squamous cell carcinoma (ESCC). METHODS: Western blotting and immunohistochemistry were used to measure the expression of BCCIP ß. In vitro studies were used to verify the effects of BCCIP ß in Eca109 cells. RESULTS: Expression of BCCIP ß was notably higher in tumor tissues of ESCC and Eca 109 cells. Meanwhile, the immunohistochemistry stain revealed that BCCIP ß was positively correlated with clinical pathologic variables such as tumor size and tumor grade, as well as Ki-67, and prompted poor prognosis. In vitro studies such as starvation and refeeding assay along with BCCIP ß-shRNA transfection assay demonstrated that BCCIP ß expression promoted proliferation of ESCC cells. In addition, BCCIP ß downregulation by silencing RNA significantly decreased the rate of colony formation, alleviated cellular apoptosis and increased the chemosensitivity of cisplatin. CONCLUSIONS: This research first put forward that BCCIP ß is an oncogene in human ESCC and contributes to the poor outcome of the deadly disease.


Subject(s)
Apoptosis/genetics , Calcium-Binding Proteins/genetics , Carcinoma, Squamous Cell/genetics , Cell Cycle Proteins/genetics , Cell Proliferation/genetics , Drug Resistance, Neoplasm/genetics , Esophageal Neoplasms/genetics , Nuclear Proteins/genetics , Antineoplastic Agents , Blotting, Western , Calcium-Binding Proteins/metabolism , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cisplatin , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma , Female , Flow Cytometry , Fluorescent Antibody Technique , Gene Knockdown Techniques , Humans , Immunohistochemistry , In Vitro Techniques , Male , Middle Aged , Neoplasm Grading , Nuclear Proteins/metabolism , Prognosis , RNA, Small Interfering , Tumor Burden , Tumor Stem Cell Assay
13.
Oncotarget ; 8(5): 8010-8021, 2017 Jan 31.
Article in English | MEDLINE | ID: mdl-28030829

ABSTRACT

Chromobox protein homolog 7 (CBX7), one of the polycomb group (PcG) proteins, is a transcriptional repressor involved in the regulation of cell proliferation and senescence. In the present study, we showed that CBX7 negatively regulates the proliferation, viability, chemoresistance, and migration of pancreatic cancer cells. Overexpression of CBX7 significantly inhibited the proliferation of pancreatic cancer cells in vitro and in vivo. Depletion of CBX7 facilitated their growth. CBX7 also impaired the viability and chemoresistance of pancreatic cancer cells. Transwell assays showed that CBX7 reduces the migratory capacity of pancreatic cancer cells. Of note, CBX7 reduced PTEN/Akt signaling in pancreatic cancer cells by increasing PTEN transcription, suggesting involvement of PTEN/Akt pathway in the tumor suppressive activity of CBX7. In addition, immunohistochemical analysis the CBX7 and PTEN expression in 74 surgically resected pancreatic ductal adenocarcinoma (PDAC) specimens revealed that CBX7 expression is significantly downregulated in pancreatic ductal adenocarcinoma, compared to normal pancreatic tissues. Reduced expression of CBX7 and PTEN was associated with increased malignancy grade in pancreatic adenocarcinoma, whereas maintenance of CBX7 and PTEN expression showed a trend toward a longer survival. These findings suggest CBX7 is an important tumor suppressor that negatively modulates PTEN/Akt signaling during pancreatic tumorigenesis.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Pancreatic Ductal/enzymology , Cell Movement , Cell Proliferation , PTEN Phosphohydrolase/metabolism , Pancreatic Neoplasms/enzymology , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Antimetabolites, Antineoplastic/pharmacology , Biomarkers, Tumor/genetics , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Female , Fluorouracil/pharmacology , Gene Expression Regulation, Neoplastic , Humans , Male , Mice, Nude , Middle Aged , Neoplasm Grading , Neoplasm Invasiveness , PTEN Phosphohydrolase/genetics , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Polycomb Repressive Complex 1/genetics , Signal Transduction , Time Factors , Transfection , Tumor Suppressor Proteins/genetics
14.
J Hematol Oncol ; 9(1): 90, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27644439

ABSTRACT

BACKGROUND: B cell-specific Moloney murine leukemia virus integration site 1 (Bmi-1) plays an important role in regulating stemness in some kinds of cancer. However, the mechanisms remain unclear. This study was to investigate whether and how Bmi-1 regulates stemness of gastric cancer. METHODS: We firstly explored the role of Bmi-1 in regulating stem cell-like features in gastric cancer. Secondly, we screened out its downstream miRNAs and clarified whether these miRNAs are involved in the regulation of stemness. Finally, we investigated the mechanisms how Bmi-1 regulates miRNAs. RESULTS: Bmi-1 positively regulates stem cell-like properties of gastric cancer and upregulates miR-21 and miR-34a. There was a positive correlation between Bmi-1 and miR-21 expression in gastric cancer tissues. MiR-21 mediated the function of Bmi-1 in regulating stem cell-like properties, while miR-34a negatively regulates stem cell-like characteristics via downregulating Bmi-1. Bmi-1 binds to PTEN promoter and directly inhibits PTEN and thereafter activates AKT. Bmi-1 also regulates p53 and PTEN via miR-21. Bmi-1 activated NF-kB via AKT and enhanced the binding of NF-kB to the promoter of miR-21 and miR-34a and increased their expression. CONCLUSIONS: Bmi-1 positively regulates stem cell-like properties via upregulating miR-21, and miR-34a negatively regulates stem cell-like characteristics by negative feedback regulation of Bmi-1 in gastric cancer. Bmi-1 upregulates miR-21 and miR-34a by activating AKT-NF-kB pathway.


Subject(s)
Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , Polycomb Repressive Complex 1/physiology , Stem Cells/pathology , Stomach Neoplasms/pathology , Humans , MicroRNAs/genetics , MicroRNAs/physiology , NF-kappa B/metabolism , PTEN Phosphohydrolase/antagonists & inhibitors , Promoter Regions, Genetic , Proto-Oncogene Proteins c-akt/metabolism , Specimen Handling , Stomach Neoplasms/genetics , Tumor Suppressor Protein p53/metabolism
15.
Neurosci Lett ; 631: 1-6, 2016 Sep 19.
Article in English | MEDLINE | ID: mdl-27507697

ABSTRACT

ANXA10 (annexin A10) is a member of the annexin family, and its biological effects are mediated primarily through the calcium-dependent phospholipid-binding and calcium ion binding. We examined the gene expressions of the L5 spinal cord after spinal nerve ligation (SNL)-induced neuropathic pain in mice by gene chip. The results showed that Anxa10 mRNA was the most upregulated gene in annexin family with 73.7-fold increase. Although previous studies have reported that several annexins are involved in nociceptive pain, the role of Anxa10 in pain remains undefined. We aimed to evaluate the role of ANXA10 in mediating injury-induced heat hyperalgesia and mechanical allodynia. We found that SNL induced persistent upregulation of Anxa10 mRNA and protein in the spinal cord of mice. Moreover, ANXA10 was colocalized with the neuronal marker MAP2 and astrocytic marker glial fibrillary acidic protein (GFAP), but not with microglial marker CD11b. Finally, pretreatment with Anxa10 siRNA partially prevented SNL-induced mechanical allodynia and heat hyperalgesia. Posttreatment with Anxa10 siRNA attenuated SNL-induced neuropathic pain. These findings suggest that ANXA10 might be a novel target in the treatment of neuropathic pain.


Subject(s)
Annexins/physiology , Neuralgia/physiopathology , Spinal Cord/metabolism , Animals , Annexins/genetics , Annexins/metabolism , Astrocytes/metabolism , HEK293 Cells , Humans , Hyperalgesia/genetics , Hyperalgesia/metabolism , Ligation , Male , Mice , Mice, Inbred ICR , Neuralgia/genetics , Neuralgia/metabolism , Neurons/metabolism , RNA, Messenger/metabolism , Spinal Nerves/injuries , Up-Regulation
16.
Tumour Biol ; 37(10): 13799-13809, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27481515

ABSTRACT

Protein kinase C iota (PKCι) has been shown to play an important role in tumorigenesis of many cancers. It was reported that frequent amplification and overexpression of PKCi were correlated with resistance to anoikis in primary esophageal squamous cell carcinomas (ESCC). In this study, we clarified a novel role of PKCι on the cell cycle progression and proliferation in ESCC. Western blot and immunohistochemistry (IHC) analysis showed that the expression of PKCι was higher in ESCC tumor tissues and cell lines. Meanwhile, IHC stain revealed that PKCι was positively correlated with clinical pathologic variables such as tumor size, tumor grade, and tumor invasion, as well as ki67. Immunoprecipitation and immunofluorescence assay revealed that PKCι/CDK7 has the physical interaction and were co-located in the cell nucleus. And this direct interaction could increase the phosphorylation level of CDK7. In vitro studies such as starvation and refeeding assay along with PKCι-shRNA transfection assay demonstrated that PKCι expression promoted proliferation of ESCC cells. And knocking PKCi down by silencing RNA (siRNA) significantly caused cell cycle arrest at G0/G1 phase, decreased rate of colony formation, and alleviated cellular apoptosis. This research provide new insights into PKCi signaling to more deeply understand its cancer-promoting function in ESCC.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/secondary , Cell Proliferation , Cyclin-Dependent Kinases/metabolism , Esophageal Neoplasms/pathology , G1 Phase , Isoenzymes/metabolism , Protein Kinase C/metabolism , S Phase , Apoptosis , Biomarkers, Tumor/genetics , Blotting, Western , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cyclin-Dependent Kinases/genetics , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Female , Flow Cytometry , Follow-Up Studies , Humans , Immunoenzyme Techniques , Isoenzymes/genetics , Male , Middle Aged , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Staging , Prognosis , Protein Kinase C/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Survival Rate , Tumor Cells, Cultured
17.
Tumour Biol ; 36(2): 747-56, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25286760

ABSTRACT

NF45 (also known as ILF2), as one subunit of NF-AT (nuclear factor of activated T cells), repairs DNA breaks, inhibits viral replication, and also functions as a negative regulator in the microRNA processing pathway in combination with NF90. Recently, it was found that implicated in the mitotic control of HeLa cells and deletion of endogenous NF45 decreases growth of HeLa cells. While the role of NF45 in cancer biology remains under debate. In this study, we analyzed the expression and clinical significance of NF45 in esophageal squamous cell carcinoma ESCC. The expression of NF45 was evaluated by Western blot in 8 paired fresh ESCC tissues and immunohistochemistry on 105 paraffin-embedded slices. NF45 was highly expressed in ESCC and significantly associated with ESCC cells tumor stage and Ki-67. Besides, high NF45 expression was an independent prognostic factor for ESCC patients' poor survival. To determine whether NF45 could regulate the proliferation of ESCC cells, we increased endogenous NF45 and analyzed the proliferation of TE1 ESCC cells using Western blot, CCK8, flow cytometry assays and colony formation analyses, which together indicated that overexpression of NF45 favors cell cycle progress of TE1 ESCC cells. While knockdown of NF45 resulted in cell cycle arrest at G0/G1-phase and thus abolished the cell growth. These findings suggested that NF45 might play an important role in promoting the tumorigenesis of ESCC, and thus be a promising therapeutic target to prevent ESCC progression.


Subject(s)
Apoptosis/genetics , Biomarkers, Tumor/biosynthesis , Carcinoma, Squamous Cell/genetics , Esophageal Neoplasms/genetics , Nuclear Factor 45 Protein/biosynthesis , Biomarkers, Tumor/genetics , Carcinogenesis , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Cell Proliferation/genetics , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HeLa Cells , Humans , Nuclear Factor 45 Protein/genetics , Prognosis
18.
J Pathol ; 235(4): 656-67, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25382750

ABSTRACT

The tumour suppressor p53 plays an important role in tumourigenesis. Besides inducing apoptosis, it regulates cellular senescence, which constitutes an important barrier to tumourigenesis. The mechanism of regulation of cellular senescence by p53 and its downstream pathway are poorly understood. Here, we report that the ubiquitin domain-containing 1 (UBTD1) gene, a new downstream target of p53, induces cellular senescence and acts as a novel tumour suppressor by a mechanism that depends on p53. Expression of UBTD1 increased upon cellular senescence induced by serial passageing of cultures, as well as by exposure to DNA-damageing drugs that induce premature senescence. Over-expression of UBTD1 induces senescence in human fibroblasts and cancer cells and attenuation of the transformed phenotype in cancer cells. UBTD1 is down-regulated in gastric and colorectal cancer tissues, and its lower expression correlates with a more aggressive phenotype and worse prognosis. Multivariate analysis revealed that UBTD1 expression was an independent prognostic factor for gastric cancer patients. Furthermore, UBTD1 increased the stability of p53 protein, by promoting the degradation of Mdm2 protein. Importantly, UBTD1 and p53 function mutually depend on each other in regulating cellular senescence and proliferation. Thus, our data suggest that, upon DNA damage, p53 induction by UBTD1 creates a positive feedback mechanism to further increase p53 expression. Our results establish UBTD1 as a regulator of cellular senescence that mediates p53 function, and provide insights into the mechanism of Mdm2 inhibition that impacts p53 dynamics during cellular senescence and tumourigenesis.


Subject(s)
Cellular Senescence , Colorectal Neoplasms/enzymology , Fibroblasts/enzymology , Proto-Oncogene Proteins c-mdm2/metabolism , Stomach Neoplasms/enzymology , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitins/metabolism , Antineoplastic Agents/pharmacology , Binding Sites , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cellular Senescence/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , DNA Damage , Dose-Response Relationship, Drug , Feedback, Physiological , Fibroblasts/drug effects , Fibroblasts/pathology , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Multivariate Analysis , Phenotype , Prognosis , Promoter Regions, Genetic , Protein Stability , Proteolysis , Proto-Oncogene Proteins c-mdm2/genetics , RNA Interference , Risk Factors , Signal Transduction , Stomach Neoplasms/genetics , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Time Factors , Transfection , Tumor Suppressor Protein p53/genetics , Ubiquitination , Ubiquitins/genetics
19.
J Surg Res ; 181(2): 342-54, 2013 May.
Article in English | MEDLINE | ID: mdl-22857917

ABSTRACT

BACKGROUND: Caspase 3 associated with apoptosis and inflammation plays a key role in ischemia-reperfusion injury. The efficacy of naked caspase 3 small interfering RNA (siRNA) has been proved in an isolated porcine kidney perfusion model but not in autotransplantation. MATERIALS AND METHODS: The left kidney was retrieved from mini pigs and infused with the University of Wisconsin solution with or without 0.3mg of caspase 3 siRNA into the renal artery with the renal artery and vein clamped for 24-h cold storage (CS). After right nephrectomy, the left kidney was autotransplanted into the right for 48 h without systemic treatment of siRNA. RESULTS: Fluorescent dye-labeled caspase 3 siRNA was visualized in the post-CS kidneys but was weakened after transplantation. The expression of caspase 3 messenger RNA and precursor was downregulated by siRNA in the post-CS kidneys. In the siRNA-preserved posttransplant kidneys, however, the caspase 3 messenger RNA and active subunit were upregulated with further decreased precursor but increased active caspase 3+ cells, apoptotic cells, and myeloperoxidase+ cells. Moreover, the renal tissue damage was aggravated by siRNA, whereas the renal function was not significantly changed. CONCLUSIONS: Naked caspase 3 siRNA administered into the kidney was effective in cold preservation but not enough to protect posttransplant kidneys, which might be because of systemic complementary responses overcoming local effects.


Subject(s)
Caspase 3/genetics , Cold Ischemia/methods , Down-Regulation/drug effects , Kidney Transplantation/methods , Kidney/drug effects , RNA, Small Interfering/pharmacology , Reperfusion Injury/prevention & control , Animals , Apoptosis/drug effects , Biomarkers/metabolism , Blotting, Western , Caspase 3/metabolism , In Situ Nick-End Labeling , Infusions, Intra-Arterial , Kidney/metabolism , Male , RNA, Small Interfering/administration & dosage , Swine , Transplantation, Autologous/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...