Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Cancer Lett ; 597: 217062, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38878852

ABSTRACT

Immune checkpoint inhibitors (ICIs) have transformed cancer therapy, yet persistent challenges such as low response rate and significant heterogeneity necessitate attention. The pivotal role of the major histocompatibility complex (MHC) in ICI efficacy, its intricate impacts and potentials as a prognostic marker, warrants comprehensive exploration. This study integrates single-cell RNA sequencing (scRNA-seq), bulk RNA-seq, and spatial transcriptomic analyses to unveil pan-cancer immune characteristics governed by the MHC transcriptional feature (MHC.sig). Developed through scRNA-seq analysis of 663,760 cells across diverse cohorts and validated in 30 solid cancer types, the MHC.sig demonstrates a robust correlation between immune-related genes and infiltrating immune cells, highlighting its potential as a universal pan-cancer marker for anti-tumor immunity. Screening the MHC.sig for therapeutic targets using CRISPR data identifies potential genes for immune therapy synergy and validates its predictive efficacy for ICIs responsiveness across diverse datasets and cancer types. Finally, analysis of cellular communication patterns reveals interactions between C1QC+macrophages and malignant cells, providing insights into potential therapeutic agents and their sensitivity characteristics. This comprehensive analysis positions the MHC.sig as a promising marker for predicting immune therapy outcomes and guiding combinatorial therapeutic strategies.

2.
Chemistry ; : e202402085, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38926940

ABSTRACT

We described a copper(I)-catalyzed atom economic and selective hydroamination-cyclization of alkynyl-tethered quinazolinones to prepare a variety of indole-fused pyrazino[1,2-a]quinazolinones in good to excellent yields ranging from 39% to 99% under mild reaction conditions. Control experiments revealed that coordination-directed method of quinazolinone moiety with copper(I) was important for the selective hydroamination-cyclization of alkynes at the N1-atom instead of N3-atom of quinazolinone. The reaction could be easily performed at gram scales and some prepared indole-fused pyrazino[1,2-a]quinazolinones with donating groups on the indole moiety showed a distinct fluorescence emission wavelength with blue shift under the acid conditions.

3.
Cancer Lett ; 578: 216460, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37863352

ABSTRACT

Coiled-Coil Domain-Containing (CCDC) is a large class of structural proteins containing left-handed supercoiled structure. The clinical value and the functional implication of CCDC in colorectal cancer (CRC) remain unknown. Based on the genetic, transcriptional, and clinical data from The Cancer Genome Atlas, five of thirty-six CCDC proteins were differentially expressed in the CRC and associated with the survival of patients with CRC. A CCDC-score model was established to evaluate the prognosis of patients. The potential function of Coiled-Coil Domain-Containing 154 (CCDC154) was investigated using bioinformatical methods, which unveiled that high expression of CCDC154 indicates poor survival for patients with CRC and correlates with low infiltration of CD8+ T cells and high infiltration of neutrophils, indicating that CCDC154 enhances tumor growth and metastasis. CCDC154 interacts with Minichromosome Maintenance Complex Component 2 (MCM2) protein and promotes malignant phenotype via MCM2. We validated the expression level and survival prediction value of CCDC154 in clinical samples, and analyzed its co-expression of MCM2, Ki-67 and p53. This work discloses the role of CCDC in clinical setting and CCDC154 functions in CRC.


Subject(s)
Cell Cycle Proteins , Colorectal Neoplasms , Humans , CD8-Positive T-Lymphocytes/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Minichromosome Maintenance Complex Component 2/genetics , Minichromosome Maintenance Complex Component 2/metabolism , Prognosis
4.
World J Gastroenterol ; 29(4): 706-730, 2023 Jan 28.
Article in English | MEDLINE | ID: mdl-36742169

ABSTRACT

BACKGROUND: The diagnostic and economic value of carcinoembryonic antigen (CEA), carbohydrate antigen 19-9 (CA19-9) and CA72-4 for gastrointestinal malignant tumors lacked evaluation in a larger scale. AIM: To reassess the diagnostic and economic value of the three tumor biomarkers. METHODS: A retrospective analysis of all 32857 subjects who underwent CEA, CA19-9, CA72-4, gastroscopy and colonoscopy from October 2006 to May 2018 was conducted. Then, we assessed the discrimination and clinical usefulness. Total cost, cost per capita and cost-effectiveness ratios were used to evaluate the economic value of two schemes (gastrointestinal endoscopy for all people without blood tests vs both gastroscopy and colonoscopy when blood tests were positive). RESULTS: The analysis of 32857 subjects showed that CEA was a qualified biomarker for colorectal cancer (CRC), while the diagnostic efficiencies of CA72-4 were catastrophic for all gastrointestinal cancers (GICs). Regarding early diagnosis, only CEA could be used for early CRC. The combination of biomarkers didn't greatly increase the area under the curve. The economic indicators of CEA were superior to those of CA19-9, CA72-4 and any combination. At the threshold of 1.8 µg/L to 10.4 µg/L, all four indicators of CEA were lower than those in the scheme that conducted gas-trointestinal endoscopy only. Subgroup analysis implied that the health checkup of CEA for people above 65 years old was economically valuable. CONCLUSION: CEA had qualified diagnostic value for CRC and superior economic value for GICs, especially for elderly health checkup subjects. CA72-4 was not suitable as a diagnostic biomarker.


Subject(s)
Gastrointestinal Neoplasms , Stomach Neoplasms , Humans , Aged , CA-19-9 Antigen , Carcinoembryonic Antigen , Retrospective Studies , Stomach Neoplasms/pathology , Prognosis , Antigens, Tumor-Associated, Carbohydrate , Biomarkers, Tumor , Gastrointestinal Neoplasms/diagnosis , Carbohydrates
5.
Front Immunol ; 13: 979521, 2022.
Article in English | MEDLINE | ID: mdl-36569910

ABSTRACT

Background: Metabolic reprogramming is a feature of cancer. However, colon cancer subtypes based on the glycolysis‒cholesterol synthesis axis have not been identified, and little is known about connections between metabolic features and the tumor microenvironment. Methods: Data for 430 colon cancer cases were extracted from The Cancer Genome Atlas, including transcriptome data, clinical information, and survival outcomes. Glycolysis and cholesterol synthesis-related gene sets were obtained from the Molecular Signatures Database for a gene set variation analysis. The relationship between the genomic landscape and immune landscape were investigated among four metabolic subtypes. Hub genes were determined. The clinical significance of candidate hub gene was evaluated in 264 clinical samples and potential functions were validated in vitro and in vivo. Results: Colon cancer cases were clustered into four metabolic subtypes: quiescent, glycolytic, cholesterogenic, and mixed. The metabolic subtypes differed with respect to the immune score, stromal score, and estimate score using the ESTIMATE algorithm, cancer-immunity cycle, immunomodulator signatures, and signatures of immunotherapy responses. Patients in the cholesterogenic group had better survival outcomes than those for other subtypes, especially glycolytic. The glycolytic subtype was related to unfavorable clinical characteristics, including high mutation rates in TTN, APC, and TP53, high mutation burden, vascular invasion, right colon cancer, and low-frequency microsatellite instability. GGH, CACNG4, MME, SLC30A2, CKMT2, SYN3, and SLC22A31 were identified as differentially expressed both in glycolytic-cholesterogenic subgroups as well as between colon cancers and healthy samples, and were involved in glycolysis‒cholesterol synthesis. GGH was upregulated in colon cancer; its high expression was correlated with CD4+ T cell infiltration and longer overall survival and it was identified as a favorable independent prognostic factor. The overexpression of GGH in colon cancer-derived cell lines (SW48 and SW480) inhibited PKM, GLUT1, and LDHA expression and decreased the extracellular lactate content and intracellular ATP level. The opposite effects were obtained by GGH silencing. The phenotype associated with GGH was also validated in a xenograft nude mouse model. Conclusions: Our results provide insight into the connection between metabolism and the tumor microenvironment in colon cancer and provides preliminary evidence for the role of GGH, providing a basis for subsequent studies.


Subject(s)
Colonic Neoplasms , gamma-Glutamyl Hydrolase , Animals , Mice , Humans , gamma-Glutamyl Hydrolase/genetics , gamma-Glutamyl Hydrolase/metabolism , Tumor Microenvironment/genetics , Colonic Neoplasms/pathology , Glycolysis , Cholesterol , Creatine Kinase, Mitochondrial Form/metabolism
6.
Front Immunol ; 13: 868480, 2022.
Article in English | MEDLINE | ID: mdl-35572523

ABSTRACT

Background: Although checkpoint blockade is a promising approach for the treatment of hepatocellular carcinoma (HCC), subsets of patients expected to show a response have not been established. As T cell-mediated tumor killing (TTK) is the fundamental principle of immune checkpoint inhibitor therapy, we established subtypes based on genes related to the sensitivity to TKK and evaluated their prognostic value for HCC immunotherapies. Methods: Genes regulating the sensitivity of tumor cells to T cell-mediated killing (referred to as GSTTKs) showing differential expression in HCC and correlations with prognosis were identified by high-throughput screening assays. Unsupervised clustering was applied to classify patients with HCC into subtypes based on the GSTTKs. The tumor microenvironment, metabolic properties, and genetic variation were compared among the subgroups. A scoring algorithm based on the prognostic GSTTKs, referred to as the TCscore, was developed, and its clinical and predictive value for the response to immunotherapy were evaluated. Results: In total, 18 out of 641 GSTTKs simultaneously showed differential expression in HCC and were correlated with prognosis. Based on the 18 GSTTKs, patients were clustered into two subgroups, which reflected distinct TTK patterns in HCC. Tumor-infiltrating immune cells, immune-related gene expression, glycolipid metabolism, somatic mutations, and signaling pathways differed between the two subgroups. The TCscore effectively distinguished between populations with different responses to chemotherapeutics or immunotherapy and overall survival. Conclusions: TTK patterns played a nonnegligible role in formation of TME diversity and metabolic complexity. Evaluating the TTK patterns of individual tumor will contribute to enhancing our cognition of TME characterization, reflects differences in the functionality of T cells in HCC and guiding more effective therapy strategies.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/pathology , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/pathology , T-Lymphocytes/pathology , Tumor Microenvironment/genetics
7.
Cell Death Dis ; 13(3): 237, 2022 03 14.
Article in English | MEDLINE | ID: mdl-35288533

ABSTRACT

Solute carrier family 25 (SLC25) encodes transport proteins at the inner mitochondrial membrane and functions as carriers for metabolites. Although SLC25 genetic variants correlate with human metabolic diseases, their roles in colon cancer remain unknown. Cases of colon cancer were retrieved from The Cancer Genome Atlas, and the transcriptionally differentially expressed members (DEMs) of SLC25 were identified. DNA level alterations, clinicopathological characteristics, and clinical survival were also investigated. A risk score model based on the DEMs was constructed to further evaluate their prognostic values in a clinical setting. The results were preliminarily validated using bioinformatic analysis of datasets from the Gene Expression Omnibus, immunohistochemical evaluations in clinical specimens, and functional experiments in colon cancer-derived cell lines. Thirty-seven DEMs were identified among 53 members of SLC25. Eight of 37 DEMs were introduced into a risk score model using integrated LASSO regression and multivariate Cox regression. Validated by GSE395282 and GSE175356, DEMs with high-risk scores were associated with the phenotypes of increasing tumor immune infiltration and decreasing glycolysis and apoptosis contents. SLC25A5 was downregulated in cancer, and its upregulation was related to better overall survival in patients from public datasets and in clinical cases. High SLC25A5 expression was an independent prognostic factor for 79 patients after surgical treatment. A negative correlation between CD8 and SLC25A5 was determined in specimens from 106 patients with advanced colon cancer. SLC25A5 attenuated cell proliferation, upregulated the expression of programmed cell death-related signatures, and exerted its biological function by inhibiting the MAPK signaling pathway. Our study reveals that mitochondrial SLC25 has prognostic value in patients with colon cancer. The bioinformatic analyses by following verification in situ and in vitro provide direction for further functional and mechanistic studies on the identified member of SLC25.


Subject(s)
Colonic Neoplasms , Computational Biology , Carcinogenesis/metabolism , Cell Transformation, Neoplastic/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Humans , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Membranes/metabolism
8.
J Transl Med ; 20(1): 68, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35115027

ABSTRACT

BACKGROUND: Interferon regulatory factor 2 (IRF-2) acts as an anti-oncogene in gastric cancer (GC); however, the underlying mechanism remains unknown. METHODS: This study determined the expression of IRF-2 in GC tissues and adjacent non-tumor tissues using immunohistochemistry (IHC) and explored the predictive value of IRF-2 for the prognoses of GC patients. Cell function and xenograft tumor growth experiments in nude mice were performed to test tumor proliferation ability, both in vitro and in vivo. Chromatin immunoprecipitation sequencing (ChIP-Seq) assay was used to verify the direct target of IRF-2. RESULTS: We found that IRF-2 expression was downregulated in GC tissues and was negatively correlated with the prognoses of GC patients. IRF-2 negatively affected GC cell proliferation both in vitro and in vivo. ChIP-Seq assay showed that IRF-2 could directly activate AMER-1 transcription and regulate the Wnt/ß-catenin signaling pathway, which was validated using IHC, in both tissue microarray and xenografted tumor tissues, western blot analysis, and cell function experiments. CONCLUSIONS: Increased expression of IRF-2 can inhibit tumor growth and affect the prognoses of patients by directly regulating AMER-1 transcription in GC and inhibiting the Wnt/ß-catenin signaling pathway.


Subject(s)
Stomach Neoplasms , Adaptor Proteins, Signal Transducing , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Humans , Interferon Regulatory Factor-2/genetics , Interferon Regulatory Factor-2/metabolism , Mice , Mice, Nude , Stomach Neoplasms/pathology , Tumor Suppressor Proteins , Wnt Signaling Pathway , beta Catenin/metabolism
9.
J Transl Med ; 19(1): 379, 2021 09 06.
Article in English | MEDLINE | ID: mdl-34488791

ABSTRACT

BACKGROUND: Since interferon regulatory factor (IRF) family functions in immune response to viral infection, its role in colorectal cancer (CRC) has not been inspected before. This study tries to investigate members of IRF family using bioinformatics approaches in aspect of differential expressions, biological function, tumor immune infiltration and clinical prognostic value for patients with CRC. METHODS: Transcriptome profiles data, somatic mutations and clinical information of CRC were obtained from COAD/READ dataset of The Cancer Genome Atlas (TCGA) as a training set. Gene expression data (GSE17536 and GSE39582) were downloaded from the Gene Expression Omnibus as a validating set. A random forest algorithm was used to score the risk for every case. Analyzing gene and function enrichment, constructing protein-protein interaction and noncoding RNA network, identifying hub-gene, characterizing tumor immune infiltration, evaluating differences in tumor mutational burden (TMB) and sensitivity to chemotherapeutics or immunotherapy were performed by a series of online tools and R packages. Immunohistochemical (IHC) examinations were carried out validation in tissue samples. RESULTS: Principal-component analysis (PCA) suggested that the transcript expression levels of nine members of IRF family differed between normal colorectum and CRC. The risk score constructed by IRF family not only acted as an independent factor for predicting survival in CRC patients with different biological processes, signaling pathways and TMB, but also indicated different immunotherapy response with diverse immune and stromal cells infiltration. IRF3 and IRF7 were upregulated in CRC and suggested a shorter survival time in patients with CRC. Differentially expressed members of IRF family exhibited varying degrees of immune cell infiltration. IHC analysis showed a positive association between IRF3 and IRF7 expression and tumor-infiltrating immune cells, including CD4+ T cell and CD68+ macrophages. CONCLUSIONS: On account of differential expression, IRF family members can help to predict both response to immunotherapy and clinical prognosis of patients with CRC. Our bioinformatic investigation not only gives a preliminary picture of the genetic features as well as tumor microenvironment, but it may provide a clue for further experimental exploration and verification on IRF family members in CRC.


Subject(s)
Colorectal Neoplasms , Interferon Regulatory Factors , Biomarkers, Tumor , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Humans , Interferon Regulatory Factors/genetics , Prognosis , Tumor Microenvironment
10.
World J Pediatr ; 17(4): 385-393, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34331676

ABSTRACT

BACKGROUND: Antibiotics, a common strategy used for neonatal infection, show consistent effect on the gut microbiota of neonates. Supplementation with probiotics has become increasingly popular in mitigating the loss of the gut microbiota. However, no clear consensus recommending the use of probiotics in the infection of neonates currently exists. This study examined the effects of probiotics on the gut microbiota of infectious neonates when used concurrently with or during the recovery period following antibiotic therapy. METHODS: Fifty-five full-term neonates diagnosed with neonatal infections were divided into the following groups: NI (no intervention, antibiotic therapy only), PCA (probiotics used concurrently with antibiotics), and PAA (probiotics used after antibiotics). The NI group received antibiotic treatment (piperacillin-tazobactam) for 1 week and the PCA group received antibiotic treatment together with probiotics (Bifidobacterium longum, Lactobacillus acidophilus, and Enterococcus faecalis) for 1 week. The PAA group received antibiotic treatment for 1 week followed by probiotics for 1 week. Fecal samples were collected at four time nodes: newborn, 1 week, 2 weeks, and 42 days after birth. The composition of the gut microbiota was determined by the high-throughput sequencing of 16S rRNA amplicons. RESULTS: Antibiotic exposure was found to dramatically alter gut microbiota, with a significant decrease of Bifidobacterium and Lactobacillus. The use of probiotics did not restore the overall diversity of the gut microbiota. However, using probiotics simultaneously with the antibiotics was found to be beneficial for the gut microbiota as compared to delaying the use of probiotics to follow treatment with antibiotics, particularly in promoting the abundance of Bifidobacterium. CONCLUSIONS: These results suggest that the early use of probiotics may have a potential ability to remodel the gut microbiota during recovery from antibiotic treatment. However, further study is required to fully understand the long-term effects including the clinical benefits.


Subject(s)
Gastrointestinal Microbiome , Probiotics , Anti-Bacterial Agents/therapeutic use , Dietary Supplements , Humans , Infant, Newborn , RNA, Ribosomal, 16S
11.
J Integr Neurosci ; 19(3): 421-428, 2020 Sep 30.
Article in English | MEDLINE | ID: mdl-33070520

ABSTRACT

The relationship between chronic bacterial colonization in the brain and Alzheimer's disease is attracting extensive attention. Recent studies indicated that the components of bacterial biofilm drive the amyloid-ß production. Muramyl dipeptide, the minimal bioactive peptidoglycan motif common to all bacteria, contributes to the development of many central inflammatory and neurodegenerative disorders. However, the involvement of Muramyl dipeptide in amyloid-ß production is not completely defined. In our present study, wild type mice received an intracerebroventricular injection of normal saline or Muramyl dipeptide. Data showed that the production of Aß1-42 oligomers was significantly increased after Muramyl dipeptide injection in the wild type mice or incubation of the SH-SY5Y cells with Muramyl dipeptide. Moreover, the action of Muramyl dipeptide was dose- and time-dependent. The above results suggested a possibility that the Muramyl dipeptide-induced Aß1-42 oligomer production might be related to the NOD2/p-p38 MAPK/BACE1 pathway. To confirm this, the SH-SY5Y cells were transfected with siRNA NOD2. Data showed that the transfected SH-SY5Y cells exhibited decreased expression of Aß1-42 oligomer, NOD2, p-p38 MAPK, and BACE1 after treatment with Muramyl dipeptide. Finally, SH-SY5Y cells were pretreated with SB203580, an inhibitor of the p-38-MAPK pathway. The results indicated that these pretreated SH-SY5Y cells exhibited decreased expression of Aß1-42 oligomer, p-p38 MAPK, and BACE1 after treatment with Muramyl dipeptide. In conclusion, these results suggested that Muramyl dipeptide was the trigger factor for Aß1-42 oligomer production, which probably acts via the NOD2/p-p38 MAPK/BACE1 signaling pathway.


Subject(s)
Acetylmuramyl-Alanyl-Isoglutamine/administration & dosage , Amyloid beta-Peptides/metabolism , MAP Kinase Signaling System , Peptide Fragments/metabolism , Amyloid Precursor Protein Secretases/metabolism , Animals , Aspartic Acid Endopeptidases/metabolism , Cell Line, Tumor , Humans , Male , Mice , Nod2 Signaling Adaptor Protein/metabolism
12.
Int Immunopharmacol ; 83: 106396, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32193103

ABSTRACT

OBJECTIVE: To elucidate the effects of ISO-α-acids (IAAs), a PPAR-γ agonist, on ICH rats and its potential mechanism. MATERIAL AND METHODS: The Sprague Dawley rats ICH model was induced by stereotactic injecting of 100 µl autologous artery blood. Ninety male rats were randomly allocated to five groups: autologous blood and IAAs (IAA); received autologous blood, IAAs and PPAR-γ inhibitor (IAA + GW9662); autologous blood and normal Saline (Saline); only autologous blood (Mock); and only needle injection (Sham). Neurological functions were assessed by mNSS. Hematoma volume, brain water content, surface proteins and inflammatory factors were detected. The microglia anti-inflammatory abilities were also evaluated. RESULTS: IAAs were able to significantly decrease ICH rat's mNSS scores, alleviate brain water content, improve hematoma resolution than Saline, Mock (p < 0.05). More "M2" microglial/macrophage can be induced by IAAs. The expression of CD 36 was statistically higher in IAA than other groups (p < 0.05). Injection of IAAs led to a greatly increasing in CD 11b and CD 206 double-positive anti-inflammatory type microglial/macrophage, moreover, a reduction of inflammatory cytokines expression (p < 0.05). Such protective effects can be relieved by GW9662. CONCLUSIONS: This is the first study to elucidate the relationship between IAAs and ICH. IAAs were able to accelerate hematoma absorption, alleviate brain edema, suppress peri-hematoma inflammations and finally improved the outcome of ICH rats. The phenotype was due to the IAAs induction of "M2" microglial/macrophage via activating of PPAR-γ and increasing CD 36 expression.


Subject(s)
Brain Edema/drug therapy , Cerebral Hemorrhage/drug therapy , Hematoma/drug therapy , Indoleacetic Acids/therapeutic use , Microglia/immunology , Plant Extracts/therapeutic use , Animals , Anti-Inflammatory Agents/therapeutic use , CD36 Antigens/genetics , CD36 Antigens/metabolism , Cell Differentiation , Cytokines/metabolism , Disease Models, Animal , Humans , Humulus/immunology , Indoleacetic Acids/pharmacology , PPAR gamma/agonists , Rats , Rats, Sprague-Dawley , Signal Transduction , Th2 Cells/immunology , Up-Regulation
13.
Dig Dis Sci ; 65(1): 168-177, 2020 01.
Article in English | MEDLINE | ID: mdl-31350707

ABSTRACT

PURPOSE: The interferon regulatory factor 2 (IRF-2) acted as a tumor suppressor. We inspected IRF-2 as a predictor of prognosis in gastric cancer (GC) patients and tried to find out the potential molecular mechanism. METHODS: In this study, the association between IRF-2 expression and clinical or prognosis significance was investigated in 86 pairs of tumor and the adjacent normal gastric tissues from GC patients. After establishing the stable cell lines, the Transwell assays were deduced to evaluate the malignancy of tumor. Then, microarray assay was carried out and the GO/KEGG pathway analyses were conducted to identify IRF-2's target gene. The relationship between IRF-2 and matrix metalloproteinases 1 (MMP-1) was also investigated by the immunohistochemistry in 15 pairs of tumor and adjacent normal gastric tissues. RESULTS: We found that IRF-2 expression level in GC was significantly correlated with the prognosis of the patients. Transwell assays suggested an impaired ability of invasion and migration in IRF-2-overexpressed GC cells and a progressive malignant phenotype in IRF-2-knockdown GC cells. Ninety differentially expressed genes were found between IRF-2-overexpressed GC cells and its normal control sets by microarray. We demonstrated that MMP-1 was canonical in the network of differentially expressed genes by GO and KEGG pathway analysis and its expression level was markedly decreased in IRF-2-overexpressed cells of MKN-45 and increased in IRF-2-knockdown cells of SGC-7901. The expression of MMP-1 was inversely correlated with IRF-2 in GAC TMA specimens. CONCLUSION: IRF-2 may inhibit GC progression by down-regulating MMP-1 level.


Subject(s)
Cell Movement , Interferon Regulatory Factor-2/metabolism , Matrix Metalloproteinase 1/metabolism , Stomach Neoplasms/enzymology , Aged , Cell Line, Tumor , Down-Regulation , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Interferon Regulatory Factor-2/genetics , Male , Matrix Metalloproteinase 1/genetics , Middle Aged , Neoplasm Invasiveness , Signal Transduction , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology
14.
Med Sci Monit ; 24: 6255-6263, 2018 Sep 08.
Article in English | MEDLINE | ID: mdl-30194286

ABSTRACT

BACKGROUND The aim of this study was to investigate the effect of the JAK2/STAT3 pathway on the proliferation, cell cycle distribution, apoptosis, and oxidative stress of Raji cells via regulating HSP70 expression. MATERIAL AND METHODS Raji cells were divided into Blank, HSP70 siRNA, NC siRNA, AG490 (a JAK2/STAT3 signaling pathway inhibitor), and HSP70 siRNA + rh JAK2 (recombinant human JAK2) groups. HSP70 expression was detected by quantitative real-time reverse transcription-PCR (qRT-PCR); the expression levels of HSP70 and JAK2/STAT3 pathway-related proteins were evaluated by Western blotting; cell proliferation was assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays; cell cycle distribution was observed by flow cytometry; cell apoptosis was tested by Annexin V-FITC/PI and Hoechst 33342/PI staining; reactive oxygen species (ROS) production was measured by dichloro-dihydro-fluorescein diacetate (DCFH-DA) assays; and MDA content and SOD and GSH-Px activities were determined using detection kits. RESULTS AG490 obviously down-regulated HSP70 expression, inhibited proliferation, induced cell cycle arrest at the G0/G1 phase, and promoted apoptosis in Raji cells; these effects were similar to the effects of HSP70 siRNA. Furthermore, ROS production and MDA content were increased in Raji cells treated with HSP70 siRNA or AG490, while SOD and GSH-Px activities were reduced. Raji cells in the HSP70 siRNA + rh JAK2 group did not significantly differ from those in the Blank group in regards to proliferation, cell cycle, apoptosis, and oxidative stress. CONCLUSIONS Blocking the JAK2/STAT3 signaling pathway may inhibit proliferation, induce cell cycle arrest, and promote oxidative stress and apoptosis in Raji cells via the down-regulation of HSP70.


Subject(s)
Burkitt Lymphoma/metabolism , HSP70 Heat-Shock Proteins/metabolism , Janus Kinase 2/metabolism , Oxidative Stress/physiology , STAT3 Transcription Factor/metabolism , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Burkitt Lymphoma/pathology , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Down-Regulation/drug effects , HSP70 Heat-Shock Proteins/genetics , Humans , Oxidative Stress/drug effects , Oxidative Stress/genetics , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Tyrphostins/pharmacology
15.
J Cancer ; 9(16): 2876-2884, 2018.
Article in English | MEDLINE | ID: mdl-30123356

ABSTRACT

Background and Aim: MicroRNAs, dysregulated in the circulation of esophageal squamous cell carcinoma (ESCC) patient, have been assumed to be with great potential in the diagnosis and prognosis of esophageal cancer. We aimed to review previous articles on ESCC. Methods: A search of electronic databases was performed before Nov 12, 2017. We summarized the identification of microRNA imbalance in the blood of ESCC compared with the healthy controls, with the objective to evaluate the efficiency of microRNAs in diagnosing and forecasting ESCC. Results: A total of 35 studies investigating plasma or serum microRNAs were included in the meta-analysis. Based on the consequences of the quality assessment of each study, the articles involved were appropriate for quantitative synthesis. For diagnostic meta-analysis. The overall pooled sensitivity, specificity, and area under the curve of circulating microRNA is 0.794 (95% CI: 0.765 - 0.820), 0.779 (95%CI: 0.746 - 0.808), 0.86 (95%CI: 0.82 - 0.88). The diagnostic value of each microRNA was calculated respectively. For prognostic meta-analysis, the overall pooled hazard ratios of higher microRNA expression in circulation was 1.34 (95% CI: 1.14-1.58), which could significantly predict poorer survival in ESCC. Conclusions: Circulating microRNAs distinguish patients with ESCC from healthy controls with high sensitivity and specificity, compared to other invasive currently used screening methods. Simultaneously, there was prognostic value for the prognosis of ESCC.

16.
J Gastroenterol Hepatol ; 33(11): 1844-1852, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29744928

ABSTRACT

BACKGROUND AND AIM: Irritable bowel syndrome (IBS) is a highly prevalent chronic functional gastrointestinal disorder. Recent studies have showed increasing important role of gut microbiota in the pathophysiological changes of IBS. Our study aims to elaborate the association between intestinal flora with the genesis and the development of IBS. METHODS: Illumina high-throughput sequencing technology was applied to investigate microbial communities of IBS patients and healthy donors. Stool specimens from the IBS-D patients were equally premixed and implanted into germ free C57B/6 mice to construct IBS animal model, and the normal group was also transplanted with normal premixed feces. The post-transplant defecation and intra-epithelial lymphocyte counts were evaluated. Microbial communities were also checked by the illumina high-throughput sequencing technology. RESULTS: Fifteen genuses significantly different were found expressed in the gut flora of IBS patients, and six genuses showed significantly different abundances between the stool specimens of mice of IBS group and normal group. Among these differences, Parasutterella expression was remarkably different in both screening and validation experiments and also related to chronic intestinal inflammation; therefore, Parasutterella expression is considered in association with the development and progression of IBS. CONCLUSION: Parasutterella may be related with the genesis and development of IBS and also associated with chronic intestinal inflammation in IBS patients.


Subject(s)
Betaproteobacteria/pathogenicity , Gastrointestinal Microbiome , Irritable Bowel Syndrome/etiology , Irritable Bowel Syndrome/microbiology , Adolescent , Adult , Aged , Animals , Betaproteobacteria/genetics , Betaproteobacteria/isolation & purification , DNA, Bacterial/isolation & purification , Disease Models, Animal , Disease Progression , Fecal Microbiota Transplantation , Feces/microbiology , High-Throughput Nucleotide Sequencing , Humans , Male , Mice, Inbred C57BL , Middle Aged , Real-Time Polymerase Chain Reaction , Young Adult
17.
BMC Cancer ; 18(1): 415, 2018 04 13.
Article in English | MEDLINE | ID: mdl-29653559

ABSTRACT

BACKGROUND: Many novel diagnostic biomarkers have been developed for gastric cancer (GC) recently. We chose two methods with high diagnostic value, the detection of serum microRNAs and metabolomics based on gas chromatography/mass spectrometry (GC/MS), and aimed to establish appropriate models. METHODS: We reviewed the diagnostic accuracies of all microRNAs identified by previous diagnostic tests. Then appropriate microRNAs and their combinations were validated the diagnostic value. We included 80 patients with GC and 82 healthy controls (HCs) and detected the expression of the microRNAs. GC/MS analysis was conducted, and we used three multivariate statistical analyses to establish diagnostic models. The concentrations of carcinoembryonic antigen (CEA) and carbohydrate antigen 19-9 (CA19-9) were detected for comparison with the novel models. RESULTS: Sixty-seven published studies and 70 microRNAs were finally included in the systematic review. MiR-18a, miR-19a, miR-21, miR-92a, miR-199a and miR-421 were chosen to further validate their diagnostic efficiencies. Five of those microRNAs in GC patients had significantly different expression. The combination of miR-19a and miR-92a had the highest area under the curve (AUC) at 0.850 with a sensitivity of 91.3% and a specificity of 61.0%. The GC/MS analysis performed an excellent diagnostic value and the AUC reached 1.0. CONCLUSION: There is a good potential for microRNAs and GC/MS analysis as new diagnostic methods in view of their high diagnostic value compared with traditional biomarkers.


Subject(s)
Biomarkers, Tumor , Circulating MicroRNA , Metabolomics , MicroRNAs/genetics , Stomach Neoplasms/diagnosis , Stomach Neoplasms/genetics , Case-Control Studies , Circulating MicroRNA/blood , Female , Gas Chromatography-Mass Spectrometry , Gene Expression , Humans , Male , Metabolomics/methods , MicroRNAs/blood , Neoplasm Grading , Neoplasm Staging , Publication Bias , Sensitivity and Specificity , Stomach Neoplasms/blood
18.
Cancer Sci ; 109(4): 1185-1194, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29363233

ABSTRACT

Recently, many new diagnostic biomarkers have been developed for colorectal cancer. We chose 2 methods with high diagnostic efficiency, the detection of serum microRNA and metabolomics based on gas chromatography/mass spectrometry (GC/MS), and aimed to establish appropriate models. We reviewed the diagnostic value of all microRNA identified by previous diagnostic tests. We selected appropriate microRNA to validate their diagnostic efficiency, and determined the optimal combination. We included 85 patients with colorectal cancer (CRC) and 78 healthy controls (HC) and detected the expression of the microRNA. GC/MS analysis was conducted, and we used 3 multivariate statistical methods to establish diagnostic models. The concentrations of carcinoembryonic antigen (CEA) and carbohydrate antigen 19-9 (CA19-9) were detected for comparison with the novel models. Ultimately, 62 published studies and 63 microRNA were included in this review. MiR-21, miR-29a, miR-92a, miR-125b and miR-223 were selected to further validate their diagnostic value. The serum levels of the 5 microRNA in CRC patients were significantly higher than those in the HC. The combination of miR-21, miR-29a, miR-92a and miR-125b had the highest area under the curve (AUC) at 0.952, with a sensitivity of 84.7% and a specificity of 98.7%. The GC/MS analysis exhibited an excellent diagnostic value and the AUC reached 1.0. With regard to traditional biomarkers, the AUC of CEA and CA19-9 were 0.808 and 0.705, respectively. The application prospects are good for microRNA and metabolomics as new diagnostic methods because of their high diagnostic value compared with traditional biomarkers.


Subject(s)
Colorectal Neoplasms/blood , Colorectal Neoplasms/diagnosis , Metabolome/physiology , MicroRNAs/blood , Adult , Area Under Curve , Biomarkers, Tumor/blood , Biomarkers, Tumor/metabolism , CA-19-9 Antigen/metabolism , Case-Control Studies , Early Detection of Cancer/methods , Female , Humans , Male , Metabolomics/methods , Middle Aged , Sensitivity and Specificity
19.
Int Immunopharmacol ; 49: 1-5, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28531759

ABSTRACT

Interferon regulatory factors (IRFs), which have 10 members, belong to the transcription factor family and were named because of the regulation of interferon expression. They play important roles in the immune regulation, cell differentiation, cell apoptosis, and cell cycle regulation. This article will review the functional characteristics and immune activity of the family members, especially in the role of cell differentiation and autoimmune diseases. Intensive studies will help uncover the pathogenesis of the disease in a more comprehensive view, and provide novel targets for disease treatment. But the most important problems yet to solve is IRFs function in the development processes of tumour, and whether IRFs can be an important regulator in tumour immune treatment.


Subject(s)
Autoimmune Diseases/genetics , Carcinogenesis/genetics , Immunity/genetics , Immunotherapy/methods , Interferon Regulatory Factors/genetics , Neoplasms/therapy , Animals , Autoimmune Diseases/therapy , Humans , Interferon Regulatory Factors/metabolism , Molecular Targeted Therapy , Neoplasms/genetics , Neoplasms/immunology , Transcription, Genetic
20.
Dig Liver Dis ; 49(4): 331-337, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28179092

ABSTRACT

BACKGROUND: Many studies have reported significant changes in intestinal microbiota in irritable bowel syndrome (IBS) patients based on quantitative real-time PCR analysis. AIMS: We aimed to review the alterations in intestinal microbiota. METHODS: An online search up to June 9, 2016, was conducted. This systematic review and meta-analysis included differential expression of intestinal microbiota in patients with IBS versus healthy controls (HCs) and subgroup analysis. We assessed the quality of the included studies using an original assessment tool. RESULTS: A total of 13 articles involving 360 IBS patients and 268 healthy controls were included. The quality assessment scores for these articles ranged from 5 to 8. Significant differences in expression in IBS patients were observed for Lactobacillus (SMD=-0.85, P<0.001, I2=28%), Bifidobacterium (SMD=-1.17, P<0.001, I2=79.3%), and Faecalibacterium prausnitzii (SMD=-1.05, P<0.001, I2=0.0%) but not Bacteroides-Prevotella group, Escherichia coli or other genera or species. Subgroup analysis showed that diarrhea-predominant IBS patients had significantly different expression of Lactobacillus (SMD=-1.81, P<0.001) and Bifidobacterium (SMD=-1.45, P<0.001). CONCLUSION: Down-regulation of bacterial colonization including Lactobacillus, Bifidobacterium and F. prausnitzii was observed in IBS patients, particularly in diarrhea-predominant IBS (IBS-D). Microbiota changes participate in the pathogenesis of IBS and may underlie the efficacy of probiotic supplements.


Subject(s)
Bacteria/classification , Gastrointestinal Microbiome/genetics , Irritable Bowel Syndrome/microbiology , Bacteria/isolation & purification , DNA, Bacterial/analysis , Diarrhea/microbiology , Down-Regulation , Feces/microbiology , Humans , Probiotics/therapeutic use , Real-Time Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...