Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Funct Integr Genomics ; 22(4): 481-489, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35322335

ABSTRACT

Sepsis is a life-threatening medical condition caused by a dysregulated host response to infection. Recent studies have found that the expression of miRNAs is associated with the pathogenesis of sepsis and septic shock. Our study aimed to reveal which miRNAs may be involved in the dysregulated immune response in sepsis and how these miRNAs interact with transcription factors (TFs) using a computational approach with in vitro validation studies. To determine the network of TFs, miRNAs, and target genes involved in sepsis, GEO datasets GSE94717 and GSE131761 were used to identify differentially expressed miRNAs and DEGs. TargetScan and miRWalk databases were used to predict biological targets that overlap with the identified DEGs of differentially expressed miRNAs. The TransmiR database was used to predict the differential miRNA TFs that overlap with the identified DEGs. The TF-miRNA-mRNA network was constructed and visualized. Finally, qRT-PCR was used to verify the expression of TFs and miRNA in HUVECs. Between the healthy and sepsis groups, there were 146 upregulated and 98 downregulated DEGs in the GSE131761 dataset, and there were 1 upregulated and 183 downregulated DEMs in the GSE94717 dataset. A regulatory network of the TF-miRna target genes was established. According to the experimental results, RUNX3 was found to be downregulated while MAPK14 was upregulated, which corroborates the result of the computational expression analysis. In a HUVECs model, miR-19b-1-5p and miR-5009-5p were found to be significantly downregulated. Other TFs and miRNAs did not correlate with our bioinformatics expression analysis. We constructed a TF-miRNA-target gene regulatory network and identified potential treatment targets RUNX3, MAPK14, miR-19b-1-5p, and miR-5009-5p. This information provides an initial basis for understanding the complex sepsis regulatory mechanisms.


Subject(s)
MicroRNAs , Mitogen-Activated Protein Kinase 14 , Sepsis , Transcription Factors , Computational Biology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , MicroRNAs/genetics , Mitogen-Activated Protein Kinase 14/genetics , Mitogen-Activated Protein Kinase 14/metabolism , RNA, Messenger/genetics , Sepsis/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
2.
Int J Biol Sci ; 17(3): 781-795, 2021.
Article in English | MEDLINE | ID: mdl-33767588

ABSTRACT

Multi-drug resistance is a major challenge to hepatocellular carcinoma (HCC) treatment, and the over-expression or deletion of microRNA (miRNA) expression is closely related to the drug-resistant properties of various cell lines. However, the underlying molecular mechanisms remain unclear. CCK-8, EdU, flow cytometry, and transmission electron microscopy were performed to determine cell viability, proliferation, apoptosis, autophagic flow, and nanoparticle characterization, respectively. In this study, the results showed that the expression of miR-26b was downregulated following doxorubicin treatment in human HCC tissues. An miR-26b mimic enhanced HCC cell doxorubicin sensitivity, except in the absence of p53 in Hep3B cells. Delivery of the proteasome inhibitor, MG132, reversed the inhibitory effect of miR-26b on the level of p53 following doxorubicin treatment. Tenovin-1 (an MDM2 inhibitor) protected p53 from ubiquitination-mediated degradation only in HepG2 cells with wild type p53. Tenovin-1 pretreatment enhanced HCC cell resistance to doxorubicin when transfected with an miR-26b mimic. Moreover, the miR-26b mimic inhibited doxorubicin-induced autophagy and the autophagy inducer, rapamycin, eliminated the differences in the drug sensitivity effect of miR-26b. In vivo, treatment with sp94dr/miR-26b mimic nanoparticles plus doxorubicin inhibited tumor growth. Our current data indicate that miR-26b enhances HCC cell sensitivity to doxorubicin through diminishing USP9X-mediated p53 de-ubiquitination caused by DNA damaging drugs and autophagy regulation. This miRNA-mediated pathway that modulates HCC will help develop novel therapeutic strategies.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Doxorubicin/therapeutic use , Liver Neoplasms/drug therapy , MicroRNAs/metabolism , Animals , Autophagy , Carcinoma, Hepatocellular/metabolism , Drug Resistance, Neoplasm , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Mice, Inbred BALB C , Tumor Suppressor Protein p53/metabolism , Ubiquitin Thiolesterase/metabolism , Xenograft Model Antitumor Assays
3.
Medicine (Baltimore) ; 99(27): e20759, 2020 Jul 02.
Article in English | MEDLINE | ID: mdl-32629654

ABSTRACT

Sepsis is one of the leading causes of mortality in intensive care units (ICU). The growing incidence rate of sepsis and its high mortality rate result are very important sociosanitary problems. Sepsis is a result of infection which can cause systemic inflammatory and organ failure. But the pathogenesis and the molecular mechanisms of sepsis is still not well understood. The aim of the present study was to identify the candidate key genes in the progression of sepsis.Microarray datasets GSE28750, GSE64457, and GSE95233 were downloaded from Gene Expression Omnibus (GEO) database. The differentially expressed genes (DEGs) were identified, and function enrichment analyses were performed. The protein-protein interaction network (PPI) was constructed and the module analysis was performed using STRING and Cytoscape. Furthermore, to verify the results of the bioinformatics analyses, the expression levels of selected DEGs were quantified by Reverse Transcription-Polymerase Chain Reaction (RT-PCR) in libobolysaccharide (LPS)-induced Human Umbilical Vein Endothelial Cells (HUVECs) to support the result of bioinformatics analysis.Thirteen hub genes were identified and biological process analysis revealed that these genes were mainly enriched in apoptotic process, inflammatory response, innate immune response. Hub genes with high degrees, including MAPK14, SLC2A3, STOM, and MMP8, were demonstrated to have an association with sepsis. Furthermore, RT-PCR results showed that SLC2A3 and MAPK14 were significantly upregulated in the HUVECs induced by LPS compared with controls.In conclusion, DEGs and hub genes identified in the present study help us understand the molecular mechanisms of sepsis, and provide candidate targets for diagnosis and treatment of sepsis.


Subject(s)
Genetic Predisposition to Disease/genetics , Sepsis/genetics , Computational Biology , Humans , Oligonucleotide Array Sequence Analysis , Protein Interaction Domains and Motifs/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sepsis/etiology , Transcriptome
4.
Cell Death Dis ; 10(11): 843, 2019 11 06.
Article in English | MEDLINE | ID: mdl-31695022

ABSTRACT

Doxorubicin is conventionally used in chemotherapy against hepatocellular carcinoma (HCC), but acquired resistance developed during long-term therapy limits its benefits. Autophagy, a conserved catabolic process for cellular self-protection and adaptation to the changing environment, is regarded as a potential clinical target to overcome doxorubicin resistance. In this study, the potential role of miR-223 in modulating doxorubicin-induced autophagy and sensitivity were evaluated in four transfected human HCC cell lines, and the in vivo relevance was assessed using a mouse xenograft model of HCC. We found that the well-defined miR-223 is expressed at low levels in doxorubicin treated HCC cells and that miR-223 overexpression inhibits the doxorubicin-induced autophagy that contributes to chemoresistance. Blockade of autophagic flux by chloroquine resulted in the failure of miR-223 inhibitor to suppress doxorubicin sensitivity of HCC cells. We further identified FOXO3a as a direct downstream target of miR-223 and primary mediator of the regulatory effect of miR-223 on doxorubicin-induced autophagy and chemoresistance in HCC cells. Finally, we confirmed the enhancement of doxorubicin sensitivity by agomiR-223 in xenograft models of HCC. These findings establish a novel miRNA-based approach for autophagy interference to reverse doxorubicin resistance in future chemotherapy regimens against human HCC.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Forkhead Box Protein O3/genetics , Liver Neoplasms/drug therapy , MicroRNAs/genetics , Animals , Apoptosis/drug effects , Autophagy/drug effects , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Proliferation/drug effects , Doxorubicin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Xenograft Model Antitumor Assays
5.
Am J Transl Res ; 11(1): 473-485, 2019.
Article in English | MEDLINE | ID: mdl-30788003

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is well-known to be the most deadly malignancy with the worst survival rate of all cancers. Gemcitabine-based chemotherapy is the most common treatment option for pancreatic ductal adenocarcinoma. However, it offers little therapeutic value in many cases due to the rapid development of chemoresistance. MicroRNAs (miRNAs) have been found to play pivotal roles in the chemotherapeutic resistance of PDAC. In the present study, we examined the molecular basis for the effective combination of OSI-027 and gemcitabine (GEM). Firstly, we identified a specific miRNA expression profile in PDAC cells after treatment with either of these drugs. We found that miR-663a was significantly upregulated after treatment with GEM and downregulated after OSI-027 treatment. With combination of the two drugs, miR-663a level was lower than the GEM group, but higher than the OSI-027 group. Real-time quantitative PCR confirmed these observations. To further establish the role of miR-663a in OSI-027 and GEM resistance in pancreatic cancer, we transfected PDAC cells with miR-663a mimic or miR-663a inhibitor. Cell viability and proliferation assays showed that miR-663a mimic enhanced drug sensitivity, while inhibitor promoted drug resistance. Moreover, we found that the combined effect of OSI-027 and GEM disappeared after inhibiting miR-663a. Our study clearly demonstrates that GEM upregulates miR-663a, thereby promoting the sensitivity of pancreatic cancer cells to OSI-027. Our study suggests that miR-663a expression may be a useful indicator of the potential for chemoresistance and provides a potential new therapeutic target to avert chemoresistance in PDAC.

6.
Oncogene ; 37(36): 4964-4978, 2018 09.
Article in English | MEDLINE | ID: mdl-29786077

ABSTRACT

Tumor self-seeding occurs when circulating malignant cells reinfiltrate the original tumor. The process may breed more aggressive tumor cells, which may contribute to cancer progression. In this study, we observed tumor self-seeding in mouse xenograft models of hepatocellular carcinoma (HCC) for the first time. We confirmed that circulating tumor cell uptake of tumor-derived exosomes, which are increasingly recognized as key instigators of cancer progression by facilitating cell-cell communication, promoted tumor self-seeding by enhancing the invasive and migration capability of recipient HCC cells. Horizontal transfer of exosomal microRNA-25-5p to anoikis-resistant HCC cells significantly enhanced their migratory and invasive abilities, whereas inhibiting microRNA-25-5p alleviated these effects. Our experiments delineate an exosome-based novel pathway employed by functional microRNA from the original tumor cells that can influence the biological fate of circulating tumor cells.


Subject(s)
Carcinoma, Hepatocellular/genetics , Cell Movement/genetics , Exosomes/genetics , Liver Neoplasms/genetics , MicroRNAs/genetics , Animals , Carcinoma, Hepatocellular/pathology , Cell Communication/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/genetics , Heterografts , Human Umbilical Vein Endothelial Cells , Humans , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Signal Transduction/genetics
7.
Sci Rep ; 6: 23269, 2016 Mar 24.
Article in English | MEDLINE | ID: mdl-27011063

ABSTRACT

Hepatocellular carcinoma (HCC) is notoriously refractory to chemotherapy because of its tendency to develop multi-drug resistance (MDR), whose various underlying mechanisms make it difficult to target. The calcium signalling pathway is associated with many cellular biological activities, and is also a critical player in cancer. However, its role in modulating tumour MDR remains unclear. In this study, stimulation by doxorubicin, hypoxia and ionizing radiation was used to induce MDR in HCC cells. A sustained aggregation of intracellular calcium was observed upon these stimuli, while inhibition of calcium signalling enhanced the cells' sensitivity to various drugs by attenuating epithelial-mesenchymal transition (EMT), Hif1-α signalling and DNA damage repair. The effect of calcium signalling is mediated via transient receptor potential canonical 6 (TRPC6), a subtype of calcium-permeable channel. An in vivo xenograft model of HCC further confirmed that inhibiting TRPC6 enhanced the efficacy of doxorubicin. In addition, we deduced that STAT3 activation is a downstream signalling pathway in MDR. Collectively, this study demonstrated that the various mechanisms regulating MDR in HCC cells are calcium dependent through the TRPC6/calcium/STAT3 pathway. We propose that targeting TRPC6 in HCC may be a novel antineoplastic strategy, especially combined with chemotherapy.


Subject(s)
Calcium/metabolism , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , TRPC Cation Channels/metabolism , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Gene Silencing , Humans , Liver Neoplasms/metabolism , TRPC Cation Channels/genetics , TRPC6 Cation Channel
SELECTION OF CITATIONS
SEARCH DETAIL
...