Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Nature ; 567(7747): 257-261, 2019 03.
Article in English | MEDLINE | ID: mdl-30814741

ABSTRACT

Hepatocellular carcinoma is the third leading cause of deaths from cancer worldwide. Infection with the hepatitis B virus is one of the leading risk factors for developing hepatocellular carcinoma, particularly in East Asia1. Although surgical treatment may be effective in the early stages, the five-year overall rate of survival after developing this cancer is only 50-70%2. Here, using proteomic and phospho-proteomic profiling, we characterize 110 paired tumour and non-tumour tissues of clinical early-stage hepatocellular carcinoma related to hepatitis B virus infection. Our quantitative proteomic data highlight heterogeneity in early-stage hepatocellular carcinoma: we used this to stratify the cohort into the subtypes S-I, S-II and S-III, each of which has a different clinical outcome. S-III, which is characterized by disrupted cholesterol homeostasis, is associated with the lowest overall rate of survival and the greatest risk of a poor prognosis after first-line surgery. The knockdown of sterol O-acyltransferase 1 (SOAT1)-high expression of which is a signature specific to the S-III subtype-alters the distribution of cellular cholesterol, and effectively suppresses the proliferation and migration of hepatocellular carcinoma. Finally, on the basis of a patient-derived tumour xenograft mouse model of hepatocellular carcinoma, we found that treatment with avasimibe, an inhibitor of SOAT1, markedly reduced the size of tumours that had high levels of SOAT1 expression. The proteomic stratification of early-stage hepatocellular carcinoma presented in this study provides insight into the tumour biology of this cancer, and suggests opportunities for personalized therapies that target it.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Molecular Targeted Therapy/trends , Proteomics , Animals , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Cell Growth Processes , Cell Movement , Hepatitis B virus/pathogenicity , Humans , Liver Neoplasms/pathology , Liver Neoplasms/virology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Staging , Prognosis , Sterol O-Acyltransferase/genetics
2.
J Proteome Res ; 21(8): 1916-1929, 2022 08 05.
Article in English | MEDLINE | ID: mdl-35820117

ABSTRACT

The liver plays a unique role as a metabolic center of the body, and also performs other important functions such as detoxification and immune response. Here, we establish a cell type-resolved healthy human liver proteome including hepatocytes (HCs), hepatic stellate cells (HSCs), Kupffer cells (KCs), and liver sinusoidal endothelial cells (LSECs) by high-resolution mass spectrometry. Overall, we quantify total 8354 proteins for four cell types and over 6000 proteins for each cell type. Analysis of this data set and regulatory pathway reveals the cellular labor division in the human liver follows the pattern that parenchymal cells make the main components of pathways, but nonparenchymal cells trigger these pathways. Human liver cells show some novel molecular features: HCs maintain KCs and LSECs homeostasis by producing cholesterol and ketone bodies; HSCs participate in xenobiotics metabolism as an agent deliverer; KCs and LSECs mediate immune response through MHC class II-TLRs and MHC class I-TGFß cascade, respectively; and KCs play a central role in diurnal rhythms regulation through sensing diurnal IGF and temperature flux. Together, this work expands our understandings of liver physiology and provides a useful resource for future analyses of normal and diseased livers.


Subject(s)
Endothelial Cells , Proteome , Endothelial Cells/metabolism , Hepatic Stellate Cells , Hepatocytes/metabolism , Humans , Kupffer Cells , Proteome/genetics , Proteome/metabolism
3.
Mol Cancer ; 21(1): 111, 2022 05 10.
Article in English | MEDLINE | ID: mdl-35538475

ABSTRACT

BACKGROUND: Sunitinib resistance can be classified into primary and secondary resistance. While accumulating research has indicated several underlying factors contributing to sunitinib resistance, the precise mechanisms in renal cell carcinoma are still unclear. METHODS: RNA sequencing and m6A sequencing were used to screen for functional genes involved in sunitinib resistance. In vitro and in vivo experiments were carried out and patient samples and clinical information were obtained for clinical analysis. RESULTS: We identified a tumor necrosis factor receptor-associated factor, TRAF1, that was significantly increased in sunitinib-resistant cells, resistant cell-derived xenograft (CDX-R) models and clinical patients with sunitinib resistance. Silencing TRAF1 increased sunitinib-induced apoptotic and antiangiogenic effects. Mechanistically, the upregulated level of TRAF1 in sunitinib-resistant cells was derived from increased TRAF1 RNA stability, which was caused by an increased level of N6-methyladenosine (m6A) in a METTL14-dependent manner. Moreover, in vivo adeno-associated virus 9 (AAV9) -mediated transduction of TRAF1 suppressed the sunitinib-induced apoptotic and antiangiogenic effects in the CDX models, whereas knockdown of TRAF1 effectively resensitized the sunitinib-resistant CDXs to sunitinib treatment. CONCLUSIONS: Overexpression of TRAF1 promotes sunitinib resistance by modulating apoptotic and angiogenic pathways in a METTL14-dependent manner. Targeting TRAF1 and its pathways may be a novel pharmaceutical intervention for sunitinib-treated patients.


Subject(s)
Adenosine , Carcinoma, Renal Cell , Kidney Neoplasms , Methyltransferases , Sunitinib , TNF Receptor-Associated Factor 1 , Adenosine/analogs & derivatives , Angiogenesis Inhibitors/pharmacology , Apoptosis/drug effects , Carcinoma, Renal Cell/blood supply , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Humans , Kidney Neoplasms/blood supply , Kidney Neoplasms/drug therapy , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Male , Methyltransferases/metabolism , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Sunitinib/pharmacology , TNF Receptor-Associated Factor 1/genetics , TNF Receptor-Associated Factor 1/metabolism
4.
J Gene Med ; 24(2): e3397, 2022 02.
Article in English | MEDLINE | ID: mdl-34751492

ABSTRACT

BACKGROUND: Aberrant expression of m6A-related proteins contributes to the occurrence and progression of non-small cell lung cancer (NSCLC). Current studies mainly focus on single m6A regulatory genes and their underlying mechanisms, and the expression of multiple m6A regulatory proteins in NSCLC remains unclear. Therefore, it is necessary to systematically examine these proteins, particularly in clinical specimens. METHODS: Bioinformatic analysis was used to determine the expression of m6A regulatory genes and their correlation with common gene mutations, such as TP53, EGFR and KRAS, using The Cancer Genome Atlas (TCGA) and the AE-meta databases. Immunohistochemistry was employed to analyze the protein expression of m6A regulatory proteins in 61 benign lung tissues and 316 NSCLC tissues. Statistical analysis was performed to calculate the correlation between the expression of m6A regulatory proteins and clinicopathological features, survival, and common gene mutations in lung carcinoma patients. RESULTS: Analysis of the mRNA levels of 13 core m6A regulators, using information from TCGA and the AE-meta databases, revealed that YTHDF1 levels were upregulated in NSCLC compared to those in adjacent normal tissues. Immunohistochemical staining showed that the expression of METTL3, ALKBH5, YTHDC2 and YTHDF1 was significantly upregulated in NSCLC tissues. Further analyses demonstrated a positive correlation between differentially expressed m6A regulatory proteins, including METTL3, ALKBH5, YTHDC2 and YTHDF1, and the poor clinicopathological features and survival of NSCLC patients. According to the statistics of NSCLC patients enrolled in the present study, the protein levels of METTL3 in patients with EGFR exon-19 mutation were higher than those in patients with wild-type EGFR. CONCLUSIONS: Our results indicate that m6A regulators, including METTL3, ALKBH5, YTHDC2 and YTHDF1, could serve as predictive markers of NSCLC, which will facilitate the early detection and diagnosis of NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Adenosine/genetics , Adenosine/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , ErbB Receptors/genetics , Genes, Regulator , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Methyltransferases/genetics
5.
Invest New Drugs ; 38(6): 1847-1853, 2020 12.
Article in English | MEDLINE | ID: mdl-32363427

ABSTRACT

Purpose Apatinib, a new tyrosine kinase inhibitor targeting vascular endothelial growth factor receptor-2, has shown promising efficacy against several solid cancers, but evidence of its efficacy against relapsed and refractory nasopharyngeal carcinoma is limited. We investigated the efficacy and safety of apatinib for relapsed and refractory nasopharyngeal carcinoma in an open-label, single-arm, phase II clinical trial. Fifty-one patients with relapsed and refractory nasopharyngeal carcinoma in the First Affiliated Hospital, Zhengzhou University, who met the inclusion criteria were enrolled in the study. All patients received apatinib at an initial dose of 500 mg daily (1 cycle = 28 days). The primary and secondary endpoints were overall response rate, progression-free survival, and overall survival. We evaluated treatment effects and recorded apatinib-related adverse events by performing regular follow-ups and workup. The overall response rate (complete and partial responses) was 31.37% (16/51). The median overall survival and progression-free survival were 16 (95% CI, 9.32-22.68) and 9 months (95% CI, 5.24-12.76), respectively. Most patients tolerated treatment-related adverse events of grades 1 and 2; hypertension (29, 56.86%), proteinuria (25, 49.02%), and hand-foot syndrome (27, 52.94%) were the most common adverse events. There were no treatment-related deaths. Apatinib showed good efficacy and safety in patients with relapsed and refractory NPC.


Subject(s)
Antineoplastic Agents/therapeutic use , Nasopharyngeal Carcinoma/drug therapy , Nasopharyngeal Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Pyridines/therapeutic use , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Adolescent , Adult , Aged , Antineoplastic Agents/adverse effects , Drug Resistance, Neoplasm , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Progression-Free Survival , Protein Kinase Inhibitors/adverse effects , Pyridines/adverse effects , Recurrence , Young Adult
6.
FASEB J ; 33(5): 6365-6377, 2019 05.
Article in English | MEDLINE | ID: mdl-30802150

ABSTRACT

Chemotherapy is one of the vital treatments for gastric cancer (GC) patients, especially those suffering advanced stages. Chemoresistance results in tumor relapse, leading to poor prognosis in GC patients; thus, identifying key regulators in this process might provide novel clues for GC therapy. Herein, we identify hyaluronan-mediated motility receptor (HMMR) as a key regulator of chemoresistance in GC. HMMR was found to be substantially up-regulated in 5-fluorouracil (5-Fu)-resistant GC biopsies and cell lines. High expression of HMMR significantly correlates with tumor relapse and predicts poorer prognosis in GC patients. Moreover, we observed that HMMR induced epithelial-mesenchymal transition and increased the cancer stem cell properties of GC, thus rendering resistance to chemotherapy. Importantly, silencing of HMMR effectively increased the susceptibility to 5-Fu therapy both in vitro and in vivo. Furthermore, we demonstrated that HMMR activates the TGF-ß/Smad2 signaling pathway, which was required for the HMMR-mediated oncogenic effects and exhibited significant clinical relevance with HMMR expression. These findings reveal a critical role for HMMR in the chemoresistance of GC and suggest that HMMR might be a potential prognostic marker or therapeutic target against the disease.-Zhang, H., Ren, L., Ding, Y., Li, F., Chen, X., Ouyang, Y., Zhang, Y., Zhang, D. Hyaluronan-mediated motility receptor confers resistance to chemotherapy via TGFß/Smad2-induced epithelial-mesenchymal transition in gastric cancer.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Epithelial-Mesenchymal Transition , Extracellular Matrix Proteins/metabolism , Fluorouracil/pharmacology , Hyaluronan Receptors/metabolism , Neoplasm Proteins/metabolism , Signal Transduction/drug effects , Smad2 Protein/metabolism , Stomach Neoplasms/drug therapy , Transforming Growth Factor beta/metabolism , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Extracellular Matrix Proteins/genetics , Gene Expression Regulation, Neoplastic , Humans , Hyaluronan Receptors/genetics , Mice, Knockout , Neoplasm Proteins/genetics , Signal Transduction/genetics , Smad2 Protein/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Transforming Growth Factor beta/genetics , Xenograft Model Antitumor Assays
7.
Mol Cell Proteomics ; 17(6): 1067-1083, 2018 06.
Article in English | MEDLINE | ID: mdl-29472430

ABSTRACT

Aberrant kinases contribute to cancer survival and proliferation. Here, we quantitatively characterized phosphoproteomic changes in an HBx-transgenic mouse model of hepatocellular carcinoma (HCC) using high-resolution mass spectrometry, profiled 22,539 phosphorylation sites on 5431 proteins. Using a strategy to interpret kinase- substrate relations in HCC and to uncover predominant kinases in tumors, our results, revealed elevated kinase activities of Src family kinases (SFKs), PKCs, MAPKs, and ROCK2 in HCC, representatives of which were further validated in cell models and clinical HBV-positive HCC samples. Inhibitor combinations targeting Src and PKCs or ROCK2 both synergized significantly to inhibit cell growth. In addition, we demonstrated that phosphorylation at Src Ser17 directly affects its kinase activity. Our phosphoproteome data facilitated the construction of a detailed molecular landscape in HCC and should serve as a resource for the cancer community. Our strategy is generally applicable to targeted therapeutics, also highlights potential mechanisms of kinase regulation.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Phosphoproteins/metabolism , Protein Kinases/metabolism , Animals , Male , Mice, Transgenic , Phosphorylation , Proteome
8.
J Proteome Res ; 17(1): 710-721, 2018 01 05.
Article in English | MEDLINE | ID: mdl-29116813

ABSTRACT

Mass-spectrometry-based phosphoproteomic workflows traditionally require efficient prefractionation and enrichment of phosphopeptides to gain an in-depth, global, and unbiased systematic investigation of phosphoproteome. Here we present TiO2 with tandem fractionation (TAFT) approach, which combines titanium dioxide (TiO2) enrichment and tandem high-pH reverse-phase (HpRP) for phosphoproteome analysis in a high-throughput manner; the entire workflow takes only 3 h to complete without laborious phosphopeptide preparation. We applied this approach to HeLa and HepG2.2.15 cells to characterize the capability of TAFT approach, which enables deep identification and quantification of more than 14 000 unique phosphopeptides in a single sample from 1 mg of protein as starting materials in <4 h of MS measurement. In total, we identified and quantified 21 281 phosphosites in two cell lines with >91% selectivity and high quantitative reproducibility (average Pearson correlation is 0.90 between biological replicates). More generally, the presented approach enables rapid, deep, and reproducible phosphoproteome analysis in a high-throughput manner with low cost, which should facilitate our understanding of signaling networks in a wide range of biological systems or the process of clinical applications.


Subject(s)
Phosphopeptides/analysis , Proteomics/methods , Titanium , HeLa Cells , Hep G2 Cells , Humans , Reproducibility of Results , Signal Transduction , Time Factors
9.
Int J Cancer ; 143(10): 2602-2613, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30151890

ABSTRACT

Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer with high proliferative and metastatic phenotypes. CDCA7, a new member of the cell division cycle associated family of genes, is involved in embryonic development and dysregulated in various types of human cancer. However, the biological role and molecular mechanism of CDCA7 in TNBC have not been defined. Herein, we found that CDCA7 was preferentially and markedly expressed in TNBC cell lines and tissues. High expression of CDCA7 was associated with metastatic relapse status and predicted poorer disease-free survival in patients with TNBC. We observed that CDCA7 silencing in TNBC cell lines effectively impaired cell proliferation, invasion and migration in vitro. Importantly, depletion of CDCA7 strongly reduced the tumorigenicity and distant colonization capacities of TNBC cells in vivo. Furthermore, CDCA7 increased the expression of EZH2, a marker of aggressive breast cancer that is involved in tumor progression, by enhancing the transcriptional activity of its promoter. This increase in EZH2 expression was essential for the CDCA7-mediated effects on TNBC progression. Finally, our immunohistochemical analysis revealed that the CDCA7/EZH2 axis was clinical relevant. These findings suggest CDCA7 plays a crucial role in TNBC progression by transcriptionally upregulating EZH2 and might be a potential prognostic factor and therapeutic target in TNBC.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/biosynthesis , Nuclear Proteins/biosynthesis , Triple Negative Breast Neoplasms/metabolism , Animals , Cell Line, Tumor , Disease Progression , Enhancer of Zeste Homolog 2 Protein/genetics , Female , Heterografts , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Nuclear Proteins/genetics , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Up-Regulation
10.
Nucleic Acids Res ; 43(21): 10474-91, 2015 Dec 02.
Article in English | MEDLINE | ID: mdl-26405199

ABSTRACT

PIWI interacting RNAs (piRNAs) are highly expressed in germline cells and are involved in maintaining genome integrity by silencing transposons. These are also involved in DNA/histone methylation and gene expression regulation in somatic cells of invertebrates. The functions of piRNAs in somatic cells of vertebrates, however, remain elusive. We found that snoRNA-derived and C (C')/D' (D)-box conserved piRNAs are abundant in human CD4 primary T-lymphocytes. piRNA (piR30840) significantly downregulated interleukin-4 (IL-4) via sequence complementarity binding to pre-mRNA intron, which subsequently inhibited the development of Th2 T-lymphocytes. Piwil4 and Ago4 are associated with this piRNA, and this complex further interacts with Trf4-Air2-Mtr4 Polyadenylation (TRAMP) complex, which leads to the decay of targeted pre-mRNA through nuclear exosomes. Taken together, we demonstrate a novel piRNA mechanism in regulating gene expression in highly differentiated somatic cells and a possible novel target for allergy therapeutics.


Subject(s)
Exosome Multienzyme Ribonuclease Complex/metabolism , Gene Expression Regulation , Interleukin-4/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Asthma/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Nucleus/metabolism , Cells, Cultured , Down-Regulation , HEK293 Cells , Humans , Interleukin-4/metabolism , Introns , RNA Precursors/metabolism , RNA Stability , RNA, Small Interfering/chemistry , RNA, Small Nucleolar/chemistry , Th2 Cells/immunology
11.
Proteomics ; 14(17-18): 1977-86, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24995952

ABSTRACT

Activated hepatic stellate cell (HSC) is the main myofibroblast cell in the liver fibrosis (LF). An important characteristic of the recovery of LF is not only the apoptosis of activated HSCs but also reversal of myofibroblast-like phenotype to a quiescent-like phenotype. Understanding the changes of secreted proteins in the reversion of activated HSCs may provide the broader view of cellular regulatory networks and discover candidate markers or targets for therapeutic strategies of LF. In this study, stable isotope labeling with amino acids (SILAC) combined with linear ion trap-Fourier transform ion cyclotron resonance mass spectrometer (LTQ-FT MS) was performed on in vitro activated HSCs and reverted HSCs to obtain a proteomic view of secretory proteins. In total, 330 proteins showed significant differences in reverted HSCs. Among these, 109 upregulated proteins were mainly involved in amino acid metabolism pathway and glucose metabolism pathway using GeneGO/MetaCore software, while 221 downregulated proteins are closely associated with HSCs activation, such as cytoskeleton remodeling, chemokines, and cell adhesion. Additionally, a set of novel proteins associated with HSCs activation and reversion were validated by Western blotting in the cell secretion and in the sera of LF, including vitronectin, laminin beta 1, and ubiquitin conjugation factor E4B. Our study provided the valuable insight into the mechanisms in the reversion of activated HSCs and identified some potential biomarkers of LF in clinical studies. All MS data have been deposited in the ProteomeXchange with identifier PXD000773 (http://proteomecentral.proteomexchange.org/dataset/PXD000773).


Subject(s)
Hepatic Stellate Cells/metabolism , Isotope Labeling/methods , Proteome/analysis , Proteome/chemistry , Proteomics/methods , Blotting, Western , Cell Line , Hepatic Stellate Cells/chemistry , Humans , Mass Spectrometry/methods , Proteins/analysis , Proteins/chemistry , Proteins/classification , Reproducibility of Results
12.
J Proteome Res ; 13(5): 2409-19, 2014 May 02.
Article in English | MEDLINE | ID: mdl-24717071

ABSTRACT

Comprehensively identifying gene expression in both transcriptomic and proteomic levels of one tissue is a prerequisite for a deeper understanding of its biological functions. Alternative splicing and RNA editing, two main forms of transcriptional processing, play important roles in transcriptome and proteome diversity and result in multiple isoforms for one gene, which are hard to identify by mass spectrometry (MS)-based proteomics approach due to the relative lack of isoform information in standard protein databases. In our study, we employed MS and RNA-Seq in parallel into mouse liver tissue and captured a considerable catalogue of both transcripts and proteins that, respectively, covered 60 and 34% of protein-coding genes in Ensembl. We then developed a bioinformatics workflow for building a customized protein database that for the first time included new splicing-derived peptides and RNA-editing-caused peptide variants, allowing us to more completely identify protein isoforms. Using this experimentally determined database, we totally identified 150 peptides not present in standard biological databases at false discovery rate of <1%, corresponding to 72 novel splicing isoforms, 43 new genetic regions, and 15 RNA-editing sites. Of these, 11 randomly selected novel events passed experimental verification by PCR and Sanger sequencing. New discoveries of gene products with high confidence in two omics levels demonstrated the robustness and effectiveness of our approach and its potential application into improve genome annotation. All the MS data have been deposited to the iProx ( http://ww.iprox.org ) with the identifier IPX00003601.


Subject(s)
Gene Expression Profiling/methods , Liver/metabolism , Proteins/genetics , Proteins/metabolism , Proteomics/methods , Alternative Splicing , Amino Acid Sequence , Animals , Base Sequence , Databases, Protein , Male , Mass Spectrometry , Mice, Inbred C57BL , Molecular Sequence Data , Peptides/genetics , Peptides/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Editing , Sequence Analysis, RNA
13.
PLoS Pathog ; 8(11): e1003036, 2012.
Article in English | MEDLINE | ID: mdl-23209409

ABSTRACT

The in vivo kinetics of antigen-presenting cells (APCs) in patients with advanced and convalescent tuberculosis (TB) is not well characterized. In order to target Mycobacterium tuberculosis (MTB) peptides- and HLA-DR-holding monocytes and macrophages, 2 MTB peptide-specific CD4(+) T-cell receptor (TCR) tetramers eu and hu were successfully constructed. Peripheral blood (PBL) samples from inpatients with advanced pulmonary TB (PTB) were analyzed using flow cytometry, and the percentages of tetramer-bound CD14(+) monocytes ranged from 0.26-1.44% and 0.21-0.95%, respectively; significantly higher than those measured in PBL samples obtained from non-TB patients, healthy donors, and umbilical cords. These tetramers were also able to specifically detect macrophages in situ via immunofluorescent staining. The results of the continuous time-point tracking of the tetramer-positive rates in PBL samples from active PTB outpatients undergoing treatment show that the median percentages were at first low before treatment, increased to their highest levels during the first month, and then began to decrease during the second month until finally reaching and maintaining a relatively low level after 3-6 months. These results suggest that there is a relatively low level of MTB-specific monocytes in advanced and untreated patients. Further experiments show that MTB induces apoptosis in CD14(+) cells, and the percentage of apoptotic monocytes dramatically decreases after treatment. Therefore, the relatively low level of MTB-specific monocytes is probably related to the apoptosis or necrosis of APCs due to live bacteria and their growth. The bactericidal effects of anti-TB drugs, as well as other unknown factors, would induce a peak value during the first month of treatment, and a relatively low level would be subsequently reached and maintained until all of the involved factors reached equilibrium. These tetramers have diagnostic potential and can provide valuable insights into the mechanisms of antigen presentation and its relationship with TB infection and latent TB infection.


Subject(s)
Apoptosis/immunology , CD4-Positive T-Lymphocytes/immunology , Monocytes/immunology , Mycobacterium tuberculosis/immunology , Receptors, Antigen, T-Cell/immunology , Tuberculosis, Pulmonary/immunology , Antitubercular Agents/administration & dosage , Apoptosis/drug effects , Female , Humans , Macrophages/immunology , Macrophages/pathology , Male , Monocytes/pathology , Time Factors , Tuberculosis, Pulmonary/drug therapy , Tuberculosis, Pulmonary/pathology
14.
Sci Rep ; 14(1): 8013, 2024 04 05.
Article in English | MEDLINE | ID: mdl-38580754

ABSTRACT

Hepatocellular carcinoma (HCC) seriously threatens human health, mostly developed from liver fibrosis or cirrhosis. Since diethylnitrosamine (DEN) and carbon tetrachloride (CCl4)-induced HCC mouse model almost recapitulates the characteristic of HCC with fibrosis and inflammation, it is taken as an essential tool to investigate the pathogenesis of HCC. However, a comprehensive understanding of the protein expression profile of this model is little. In this study, we performed proteomic analysis of this model to elucidate its proteomic characteristics. Compared with normal liver tissues, 432 differentially expressed proteins (DEPs) were identified in tumor tissues, among which 365 were up-regulated and 67 were down-regulated. Through Gene Ontology (GO) analysis, Ingenuity Pathway Analysis (IPA), protein-protein interaction networks (PPI) analysis and Gene-set enrichment analysis (GSEA) analysis of DEPs, we identified two distinguishing features of DEN and CCl4-induced HCC mouse model in protein expression, the upregulation of actin cytoskeleton and branched-chain amino acids metabolic reprogramming. In addition, matching DEPs from the mouse model to homologous proteins in the human HCC cohort revealed that the DEN and CCl4-induced HCC mouse model was relatively similar to the subtype of HCC with poor prognosis. Finally, combining clinical information from the HCC cohort, we screened seven proteins with prognostic significance, SMAD2, PTPN1, PCNA, MTHFD1L, MBOAT7, FABP5, and AGRN. Overall, we provided proteomic data of the DEN and CCl4-induced HCC mouse model and highlighted the important proteins and pathways in it, contributing to the rational application of this model in HCC research.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms, Experimental , Liver Neoplasms , Mice , Animals , Humans , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/chemically induced , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Proteomics , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/metabolism , Diethylnitrosamine/adverse effects , Liver Cirrhosis/pathology , Disease Models, Animal , Fatty Acid-Binding Proteins
15.
Eur J Med Res ; 29(1): 151, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38429762

ABSTRACT

BACKGROUND: Urosepsis is a life-threatening organ disease in which pathogenic microorganisms in the urine enter the blood through the vessels, causing an imbalance in the immune response to infection. The aim of this study was to elucidate the role of testicular orphan receptor 4 (TR4) in urosepsis. METHODS: The role of TR4 in the progression and prognosis of urosepsis was confirmed by analyzing data from online databases and clinical human samples. To mimic urosepsis, we injected E. coli bacteria into the renal pelvis of mice to create a urosepsis model. Hematoxylin and eosin staining was used to observe histopathological changes in urosepsis. The effects of the upregulation or downregulation of TR4 on macrophage pyroptosis were verified in vitro. Chromatin immunoprecipitation assay was used to verify the effect of TR4 on Gasdermin D (GSDMD) transcription. RESULTS: TR4 was more highly expressed in the nonsurviving group than in the surviving group. Furthermore, overexpressing TR4 promoted inflammatory cytokine expression, and knocking down TR4 attenuated inflammatory cytokine expression. Mechanistically, TR4 promoted pyroptosis by regulating the expression of GSDMD in urosepsis. Furthermore, we also found that TR4 knockdown protected mice from urosepsis induced by the E. coli. CONCLUSIONS: TR4 functions as a key regulator of urosepsis by mediating pyroptosis, which regulates GSDMD expression. Targeting TR4 may be a potential strategy for urosepsis treatment.


Subject(s)
Body Fluids , Sepsis , Animals , Humans , Mice , Cytokines , Eosine Yellowish-(YS) , Escherichia coli , Gasdermins , Phosphate-Binding Proteins/genetics , Sepsis/complications , Sepsis/genetics
16.
Cell Death Dis ; 14(8): 568, 2023 08 26.
Article in English | MEDLINE | ID: mdl-37633920

ABSTRACT

Sustained activation of DNA damage response (DDR) signaling has been demonstrated to play vital role in chemotherapy failure in cancer. However, the mechanism underlying DDR sustaining in cancer cells remains unclear. In the current study, we found that the expression of the DDUP microprotein, encoded by the CTBP1-DT lncRNA, drastically increased in cisplatin-resistant ovarian cancer cells and was inversely correlated to cisplatin-based therapy response. Using a patient-derived human cancer cell model, we observed that DNA damage-induced DDUP foci sustained the RAD18/RAD51C and RAD18/PCNA complexes at the sites of DNA damage, consequently resulting in cisplatin resistance through dual RAD51C-mediated homologous recombination (HR) and proliferating cell nuclear antigen (PCNA)-mediated post-replication repair (PRR) mechanisms. Notably, treatment with an ATR inhibitor disrupted the DDUP/RAD18 interaction and abolished the effect of DDUP on prolonged DNA damage signaling, which resulted in the hypersensitivity of ovarian cancer cells to cisplatin-based therapy in vivo. Altogether, our study provides insights into DDUP-mediated aberrant DDR signaling in cisplatin resistance and describes a potential novel therapeutic approach for the management of platinum-resistant ovarian cancer.


Subject(s)
Ovarian Neoplasms , RNA, Long Noncoding , Female , Humans , Cisplatin/therapeutic use , DNA-Binding Proteins/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Proliferating Cell Nuclear Antigen , RNA, Long Noncoding/genetics , Ubiquitin-Protein Ligases , Micropeptides
17.
Int J Biol Sci ; 19(1): 167-182, 2023.
Article in English | MEDLINE | ID: mdl-36594094

ABSTRACT

Drug resistance presents a major obstacle in the treatment of genitourinary cancers. Exosomes as the medium of intercellular communication serve important biological functions and play essential roles in pathological processes, including drug response. Through the transfer of bioactive cargoes, exosomes can modulate drug resistance via multiple mechanisms. This review attempts to elucidate the mechanisms of exosomal cargoes with reference to tumor drug resistance, their role in genitourinary cancers, and their potential clinical applications as candidate biomarkers in liquid biopsy.


Subject(s)
Exosomes , Neoplasms , Urogenital Neoplasms , Humans , Biomarkers , Drug Resistance, Neoplasm/genetics , Liquid Biopsy , Urogenital Neoplasms/pathology , Neoplasms/drug therapy , Biomarkers, Tumor
18.
Clin Transl Med ; 13(1): e1156, 2023 01.
Article in English | MEDLINE | ID: mdl-36597139

ABSTRACT

BACKGROUND: Circular RNAs (circRNAs) have been reported to play a significant role in tumorigenesis. However, the detailed function of circRNA in prostate cancer (PCa) is still largely unknown. METHODS: We quantified circTFDP2 expression in PCa tissues and adjacent normal tissues using quantitative reverse transcription-polymerase chain reaction (qRT-PCR). Colony formation, Cell Counting Kit-8 (CCK-8), flow cytometry, transwell, and in vivo progression and metastasis assays were applied to reveal the proliferation and metastatic abilities of circTFDP2 in PCa cells. Mass spectrometry, RNA pulldown, RNA-immunoprecipitation (RIP), western blotting and immunofluorescence were used for the mechanistic studies. qRT-PCR and RIP assays were used to explore the regulatory role of eIF4A3 in the biogenesis of circTFDP2. Finally, functional assays showed the effect of circTFDP2-containing exosomes on PCa cell progression. RESULTS: circTFDP2 was upregulated in PCa tissues compared with adjacent normal tissues. Furthermore, high circTFDP2 expression was positively correlated with the Gleason score. Functionally, circTFDP2 promoted PCa cell proliferation and metastasis both in vivo and in vitro. Mechanistically, circTFDP2 interacted with poly(ADP-ribose) polymerase 1 (PARP1) protein in its DNA-binding domain to prevent it from active caspase-3-dependent cleavage, and finally relieved PCa cells from DNA damage. In addition, RNA-binding protein eIF4A3 can interact with the flanking region of circTFDP2 and promote the biogenesis of circTFDP2. Moreover, exosome-derived circTFDP2 promoted PCa cell progression. CONCLUSIONS: In general, our study demonstrated that circTFDP2 promoted PCa cell progression through the PARP1/DNA damage axis, which may be a promising therapeutic target for PCa.


Subject(s)
Exosomes , Prostatic Neoplasms , Male , Humans , Caspase 3 , Exosomes/metabolism , Disease Progression , Cell Line, Tumor , Cell Movement/genetics , Prostatic Neoplasms/metabolism , RNA , RNA, Circular/genetics , Poly (ADP-Ribose) Polymerase-1/genetics
19.
PeerJ ; 10: e13922, 2022.
Article in English | MEDLINE | ID: mdl-35999846

ABSTRACT

Purpose: We aimed to establish a cholesterogenic gene signature to predict the prognosis of young breast cancer (BC) patients and then verified it using cell line experiments. Methods: In the bioinformatic section, transcriptional data and corresponding clinical data of young BC patients (age ≤ 45 years) were downloaded from The Cancer Genome Atlas (TCGA) database for training set. Differentially expressed genes (DEGs) were compared between tumour tissue (n = 183) and normal tissue (n = 30). By using univariate Cox regression and multi COX regression, a five-cholesterogenic-gene signature was established to predict prognosis. Subgroup analysis and external validations of GSE131769 from the Gene Expression Omnibus (GEO) were performed to verify the signature. Subsequently, in experiment part, cell experiments were performed to further verify the biological roles of the five cholesterogenic genes in BC. Results: In the bioinformatic section, a total of 97 upregulated genes and 124 downregulated cholesterogenic genes were screened as DEGs in the TCGA for training the model. A risk scoring signature contained five cholesterogenic genes (risk score = -1.169 × GRAMD1C -0.992 × NFKBIA + 0.432 × INHBA + 0.261 × CD24 -0.839 × ACSS2) was established, which could differentiate the prognosis of young BC patients between high-risk and low-risk group (<0.001). The prediction value of chelesterogenic gene signature in excellent with AUC was 0.810 in TCGA dataset. Then the prediction value of the signature was verified in GSE131769 with P = 0.033. In experiment part, although the downregulation of CD24, GRAMD1C and ACSS2 did not significantly affect cell viability, NFKBIA downregulation promoted the viability, colony forming ability and invasion capability of BC cells, while INHBA downregulation had the opposite effects. Conclusion: The five-cholesterogenic-gene signature had independent prognostic value and robust reliability in predicting the prognosis of young BC patients. The cell experiment results suggested that NFKBIA played a protective role, while INHBA played the pro-cancer role in breast cancer.


Subject(s)
Breast Neoplasms , Humans , Middle Aged , Female , Breast Neoplasms/genetics , Reproducibility of Results , Prognosis , Cell Line , Cell Survival
20.
DNA Cell Biol ; 41(12): 1075-1083, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36454274

ABSTRACT

Cancer stem cells (CSCs) drive tumor relapse, which is a major clinical challenge in colon cancer. Targeting CSCs presents a great opportunity in eradicating cancer cells and thus treatment of patients with cancer. However, the epigenetic control of the CSC signature and key molecules involved in colon cancer remains undefined. In this study, we demonstrated that alpha-1,3-glucosyltransferase (ALG8) is upregulated in colon cancer tissues compared with normal tissues. Overexpression of the ALG8 gene predicted poor overall survival and disease-free survival in colon cancer patients. Silencing of the ALG8 gene repressed the stemness of colon tumor cells. Xenograft mice transplanted with ALG8-deficient tumor cells significantly alleviated tumor burden and prolonged survival in comparison with control mice. Further analysis showed that ALG8 gene promoted cancer stemness through inducing glycosylation of LRP6, which activates the WNT/beta-catenin signaling pathway. Importantly, attenuation of the glycosylation using tunicamycin abrogated the effect of ALG8 gene on cancer stemness. Taken together, our findings demonstrated that ALG8 enhances colon tumorigenesis by activating the WNT/beta-catenin signaling pathway. Therefore, ALG8 gene is a potential therapeutic target in colon cancer.


Subject(s)
Colonic Neoplasms , Wnt Signaling Pathway , Humans , Mice , Animals , Wnt Signaling Pathway/genetics , Glycosylation , Neoplasm Recurrence, Local/genetics , Neoplastic Stem Cells , Colonic Neoplasms/metabolism , beta Catenin/genetics , beta Catenin/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Glucosyltransferases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL