Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 19 de 19
1.
Apoptosis ; 2024 Apr 19.
Article En | MEDLINE | ID: mdl-38641760

To investigate the protective role of immune response gene 1 (IRG1) and exogenous itaconate in autoimmune hepatitis (AIH) and elucidate the underlying mechanisms. Wild-type and IRG1-/- AIH mouse models were established, and samples of liver tissue and ocular blood were collected from each group of mice to assess the effects of IRG1/itaconate on the expression of pro- and anti-inflammatory cytokines. The levels of liver enzymes and related inflammatory factors were determined using enzyme-linked immunosorbent assay and real-time quantitative polymerase chain reaction (PCR). Liver histomorphology was detected through hematoxylin and eosin staining and then scored for liver injury, and the infiltration levels of tissue-resident memory T (TRM) cells and related molecules in the liver tissue were detected through immunofluorescence staining in vitro. RNA sequencing and gene enrichment analysis were conducted to identify the corresponding molecules and pathways, and lentiviral transfection was used to generate TRM cell lines with IRG1, Jak3, Stat3, and p53 knockdown. Real-time quantitative PCR and western blot were performed to detect the expression levels of relevant mRNAs and proteins in the liver tissue and cells. The percentage of apoptotic cells was determined using flow cytometry. IRG1/itaconate effectively reduced the release of pro-inflammatory cytokines and the pathological damage to liver tissue, thereby maintaining normal liver function. At the same time, IRG1/itaconate inhibited the JAK3/STAT3 signaling pathway, regulated the expression of related downstream proteins, and inhibited the proliferation and promoted the apoptosis of CD69+CD103+CD8+ TRM cells. For the first time, P53 was found to act as a downstream molecule of the JAK3/STAT3 pathway and was regulated by IRG1/itaconate to promote the apoptosis of CD8+ TRM cells. IRG1/itaconate can alleviate concanavalin A-induced autoimmune hepatitis in mice by inhibiting the proliferation and promoting the apoptosis of CD69+CD103+CD8+ TRM cells via the JAK3/STAT3/P53 pathway.

2.
Mol Biol Rep ; 51(1): 533, 2024 Apr 20.
Article En | MEDLINE | ID: mdl-38642169

BACKGROUND: Sepsis may be linked to oxidative stress and can be controlled by itaconate, an activator of the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway. Nevertheless, the itaconate impact on sepsis-associated acute kidney injury (SA-AKI) has yet to be definitively established. METHODS: We employed SA-AKI mouse model through a cecal ligation and puncture (CLP) procedure for the in vivo investigation of the potential nephroprotective effect of itaconate in this study. A plasmid was transfected into RAW264.7 cells to examine the Nrf2 pathway function after itaconate administration. Finally, the immune-responsive gene 1-knockout (IRG1-/-) mice were used to study the itaconate impacts on oxidative stress-induced SA-AKI. RESULTS: We have shown that 4-octyl itaconate (OI) significantly reduced CD11b-positive macrophage aggregation and activated the Nrf2 pathway in the bone marrow-derived macrophages (BMDM). The impacts of Nrf2 inhibitor ML385 on the anti-inflammatory and antioxidant properties of itaconate were found to be partial. OI inhibited lipopolysaccharide-induced oxidative stress injury in RAW264.7 macrophages and activated Nrf2 in the nucleus to hinder the expression of nuclear factor kappa B p65, thereby suppressing oxidative stress injury in the macrophages. Additionally, the introduction of the transfected plasmid resulted in a partial inhibition of the anti-inflammatory impact of itaconate. The kidney injury caused by sepsis exhibited greater severity in the IRG1-/- mice than in the wild type mice. Exogenous OI partially attenuated the kidney injury induced by sepsis in the IRG1-/- mice and suppressed the oxidative stress injury in macrophages. CONCLUSIONS: This investigation offers new proof to support the itaconate function in the development and progression of SA-AKI and shows a new possible therapeutic agent for the SA-AKI treatment.


Acute Kidney Injury , Sepsis , Succinates , Mice , Animals , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Macrophage Activation , Oxidative Stress , Acute Kidney Injury/etiology , Anti-Inflammatory Agents/pharmacology , Sepsis/complications
3.
Int J Mol Sci ; 25(5)2024 Feb 29.
Article En | MEDLINE | ID: mdl-38474064

Our previous study has reported that metastasis-associated protein 2 (MTA2) plays essential roles in tumorigenesis and aggressiveness of gastric cancer (GC). However, the underlying molecular mechanisms of MTA2-mediated GC and its upstream regulation mechanism remain elusive. In this study, we identified a novel circular RNA (circRNA) generated from the MTA2 gene (circMTA2) as a crucial regulator in GC progression. CircMTA2 was highly expressed in GC tissues and cell lines, and circMTA2 promoted the proliferation, invasion, and metastasis of GC cells both in vitro and in vivo. Mechanistically, circMTA2 interacted with ubiquitin carboxyl-terminal hydrolase L3 (UCHL3) to restrain MTA2 ubiquitination and stabilize MTA2 protein expression, thereby facilitating tumor progression. Moreover, circMTA2 was mainly encapsulated and transported by exosomes to promote GC cell progression. Taken together, these findings uncover that circMTA2 suppresses MTA2 degradation by interacting with UCHL3, thereby promoting GC progression. In conclusion, we identified a cancer-promoting axis (circMTA2/UCHL3/MTA2) in GC progression, which paves the way for us to design and synthesize targeted inhibitors as well as combination therapies.


Stomach Neoplasms , Humans , Stomach Neoplasms/pathology , Repressor Proteins/genetics , Cell Line, Tumor , Histone Deacetylases/metabolism , Proteolysis , Gene Expression Regulation, Neoplastic , Cell Proliferation/genetics , Ubiquitin Thiolesterase/metabolism
4.
Cancer Sci ; 114(11): 4184-4201, 2023 Nov.
Article En | MEDLINE | ID: mdl-37702298

Although PARP inhibitor (PARPi) has been proven to be a promising anticancer drug in cancer patients harboring BRCA1/2 mutation, it provides limited clinical benefit in colorectal cancer patients with a low prevalence of BRCA1/2 mutations. In our study, we found PARPi talazoparib significantly induced cellular senescence via inhibiting p53 ubiquitination and activating p21. Furthermore, CDK4/6i palbociclib amplified this therapy-induced senescence (TIS) in vitro and in vivo. Mechanistically, talazoparib and palbociclib combination induced senescence-associated secretory phenotype (SASP), and characterization of SASP components revealed type I interferon (IFN)-related mediators, which were amplified by cGAS/STING signaling. More importantly, RNA sequencing data indicated that combination therapy activated T cell signatures and combination treatment transformed the tumor microenvironment (TME) into a more antitumor state with increased CD8 T cells and natural killer (NK) cells and decreased macrophages and granulocytic myeloid-derived suppressor cells (G-MDSCs). Moreover, clearance of the TIS cells by αPD-L1 promoted survival in immunocompetent mouse colorectal cancer models. Collectively, we elucidated the synergistic antitumor and immunomodulatory mechanisms of the talazoparib-palbociclib combination. Further combination with PD-L1 antibody might be a promising "one-two punch" therapeutic strategy for colorectal cancer patients.


Antineoplastic Agents , Colorectal Neoplasms , Animals , Mice , Humans , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , BRCA1 Protein , B7-H1 Antigen , BRCA2 Protein , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/drug therapy , Nucleotidyltransferases , Tumor Microenvironment , Cyclin-Dependent Kinase 4
5.
J Cancer ; 14(2): 262-274, 2023.
Article En | MEDLINE | ID: mdl-36741260

Gastric cancer (GC) is a leading cause of cancer-related death worldwide. Transcription factors (TFs) are essential gene expression regulators, and play critical roles in cancer development. However, the biological actions and prognostic value of TFs in GC remain unclear. In this study, we identified a risk model based on a 14-TF signature to predict recurrence-free survival in patients with GC. We further analyzed the ability of 14-TF to predict recurrence-free survival time in GC and found that a higher expression level of metastasis-associated protein 2 (MTA2) was associated with shorter overall survival and disease-free survival time in GC. Through in vitro and in vivo experiments, we demonstrated that MTA2 significantly promotes GC growth and metastasis. Furthermore, we identified MTA2 binding to the promoter of minichromosome maintenance deficient 5 (MCM5), thereby promoting GC progression. Overall, these findings strongly support the prognostic potential of the 14-TFs signature and suggest that targeting MTA2 may be a promising strategy to treat GC.

6.
Front Oncol ; 12: 844135, 2022.
Article En | MEDLINE | ID: mdl-35480096

MUS81 is a critical endonuclease involved in heterodimer formation with Eme1/Mms4 and an important DNA damage repair regulatory molecule. Our previous study suggested that MUS81 was overexpressed and its high expression was positively correlated with gastric cancer metastasis. However, the therapeutic potential of targeting MUS81 in gastric cancer requires further exploration. Therefore, in this study, the Cancer Genome Atlas (TCGA) data were analyzed and showed that MUS81 is a key regulator of cell cycle distribution and DNA damage repair in gastric cancer. In vitro and in vivo, MUS81 knockdown significantly enhanced the anticancer effect of the PARP inhibitor talazoparib. Mechanistically, MUS81 inhibition impaired the activation of the ATR/CHK1 cell cycle signaling pathway and promoted gastric cancer cells with talazoparib-induced DNA damage to continue mitosis. Moreover, addition of the bromodomain-containing protein 4 inhibitor AZD5153 increased the anticancer effect of talazoparib via MUS81 inhibition in gastric cancer cells, and this combination effect was largely impaired when MUS81 was knocked down. In conclusion, these data suggested that MUS81 regulated ATR/CHK1 activation, a key signaling pathway in the G2M checkpoint, and targeting MUS81 enhanced the antitumor efficacy of talazoparib. Therefore, AZD5153 combined with talazoparib may represent a promising therapeutic strategy for patients with MUS81 proficient gastric cancer.

7.
BMJ Open ; 11(12): e053068, 2021 12 17.
Article En | MEDLINE | ID: mdl-34921080

BACKGROUND: The early spatiotemporal transmission of COVID-19 remains unclear. The community to healthcare agencies and back to community (CHC) model was tested in our study to simulate the early phase of COVID-19 transmission in Wuhan, China. METHODS: We conducted a retrospective study. COVID-19 case series reported to the Municipal Notifiable Disease Report System of Wuhan from December 2019 to March 2020 from 17 communities were collected. Cases from healthcare workers (HW) and from community members (CM) were distinguished by documented occupations. Overall spatial and temporal relationships between HW and CM COVID-19 cases were visualised. The CHC model was then simulated. The turning point separating phase 1 and phase 2 was determined using a quadratic model. For phases 1 and 2, linear regression was used to quantify the relationship between HW and CM COVID-19 cases. RESULTS: The spatial and temporal distributions of COVID-19 cases between HWs and CMs were closely correlated. The turning point was 36.85±18.37 (range 15-70). The linear model fitted well for phase 1 (mean R2=0.98) and phase 2 (mean R2=0.93). In phase 1, the estimated [Formula: see text]s were positive (from 18.03 to 94.99), with smaller [Formula: see text]s (from 2.98 to 15.14); in phase 2, the estimated [Formula: see text]s were negative (from -4.22 to -81.87), with larger [Formula: see text]s (from 5.37 to 78.12). CONCLUSION: Transmission of COVID-19 from the community to healthcare agencies and back to the community was confirmed in Wuhan. Prevention and control measures for COVID-19 in hospitals and among HWs are crucial and warrant further attention.


COVID-19 , China , Delivery of Health Care , Humans , Retrospective Studies , SARS-CoV-2
8.
Opt Express ; 29(23): 37979-37986, 2021 Nov 08.
Article En | MEDLINE | ID: mdl-34808859

Tunable single-longitudinal-mode (SLM) ring Ho:YLF laser with intra-cavity isolator is investigated for 2.05 µm and 2.06 µm band based on single resonator, which is realized dual-band SLM laser conversion by a polarizer. Up to 548 mW SLM power with beam quality factor M2 of 1.1 is achieved at wavelength of 2064.63 nm, and the corresponding slope efficiency is 26.7%. Wavelength tuning ranges from 2063.91 nm to 2065.71 nm and 2050.65nm to 2053.15nm can be demonstrated. The highest SLM power around P12 and R30 CO2 absorption peak of 2064.41 nm and 2050.96 nm are 540 mW and 500 mW, respectively. The power instability within 30 minutes is around 0.14%. As we know, dual-band switched Ho:YLF laser operation at SLM with wavelength tunability is reported for the first time for the potential application of CO2 differential absorption lidar.

9.
EMBO Mol Med ; 13(5): e13524, 2021 05 07.
Article En | MEDLINE | ID: mdl-33821572

Pancreatic beta cells undergo compensatory proliferation in the early phase of type 2 diabetes. While pathways such as FoxM1 are involved in regulating compensatory beta cell proliferation, given the lack of therapeutics effectively targeting beta cell proliferation, other targetable pathways need to be identified. Herein, we show that Pbk, a serine/threonine protein kinase, is essential for high fat diet (HFD)-induced beta cell proliferation in vivo using a Pbk kinase deficiency knock-in mouse model. Mechanistically, JunD recruits menin and HDAC3 complex to the Pbk promoter to reduce histone H3 acetylation, leading to epigenetic repression of Pbk expression. Moreover, menin inhibitor (MI) disrupts the menin-JunD interaction and augments Pbk transcription. Importantly, MI administration increases beta cell proliferation, ameliorating hyperglycemia, and impaired glucose tolerance (IGT) in HFD-induced diabetic mice. Notably, Pbk is required for the MI-induced beta cell proliferation and improvement of IGT. Together, these results demonstrate the repressive role of the menin/JunD/Pbk axis in regulating HFD-induced compensatory beta cell proliferation and pharmacologically regulating this axis may serve as a novel strategy for type 2 diabetes therapy.


Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Insulin-Secreting Cells/cytology , Mitogen-Activated Protein Kinase Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Cell Proliferation , Diet, High-Fat/adverse effects , Histone Deacetylases , Mice , Proto-Oncogene Proteins c-jun
10.
Mol Med Rep ; 17(2): 3344-3349, 2018 Feb.
Article En | MEDLINE | ID: mdl-29257266

Wee1 is an oncogenic nuclear kinase, which can regulate the cell cycle as a crucial G2M checkpoint. Overexpression of Wee1 can be observed in various cancer types, which may lead to a poor prognosis, but the potential therapeutic value of Wee1 in colorectal cancer has not been fully studied. In the present study, the role of Wee1 in colonic cancer was investigated. Wee1 inhibition by small interfering RNA was demonstrated to significantly restrain cancer cell proliferation and sensitize the p53 mutant colonic cancer cell lines HT29 and SW480 to the effect of treatment with ionizing radiation. The anticancer effect of the Wee1 inhibitor MK1775 was investigated in these two colonic cancer cell lines. MK1775 was demonstrated to induce significant DNA damage, suppress cell viability and induce apoptosis. In addition, MK1775 sensitized HT29 and SW480 cells to the effect of irinotecan. Annexin V/propidium iodide staining demonstrated that combination therapy can induce increased apoptosis compared with MK1775 or irinotecan monotherapy. The results of western blot analysis also indicated increased expression of the DNA damage marker histone H2AX, and apoptosis­associated protein cleaved caspase 3, in HT29 and SW480 cells. In conclusion, the present study indicated that Wee1 may be a valuable target for treatment of p53 mutant colonic cancer.


Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/analogs & derivatives , Cell Cycle Proteins/antagonists & inhibitors , Colonic Neoplasms/drug therapy , Enzyme Inhibitors/pharmacology , Nuclear Proteins/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Tumor Suppressor Protein p53/genetics , Apoptosis/drug effects , Camptothecin/pharmacology , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , DNA Damage/drug effects , Humans , Irinotecan , Mutation , Nuclear Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Pyrimidinones
11.
PLoS One ; 12(7): e0180173, 2017.
Article En | MEDLINE | ID: mdl-28742860

MicroRNA-101 has been reported as an important factor in carcinogenesis of several malignant tumors. However, its actual role in prognosis among solid malignancies remains unclear. Accordingly, we performed this meta-analysis aiming to identify prognostic significance of miR-101 in solid tumor. Pooled hazard ratios (HRs) with 95% confidence intervals (CIs) for overall survival (OS) or disease-free survival (DFS)/metastasis-free survival (MFS)/progression-free survival (PFS)/relapse-free survival (RFS)/time-to progression (TTP) were estimated with random effects or fixed effects models on the basis of heterogeneity. Subgroup analysis, sensitive analysis and meta-regression analysis were also conducted to clarify the possible confounding factors and investigate the source of heterogeneity. Publication bias was evaluated by using Begg's and Egger's tests. A total of 21 studies containing 3753 cases were selected into our quantitative analysis via electronic database search. A lower expression of miR-101 was significantly associated with worse OS (HR = 0.66, 95%CI [0.52-0.85], P = 0.001) and PFS (HR = 0.70, 95%CI [0.51-0.95], P = 0.023) in patients with solid tumor. The under-expression of miRNA-101 is a credible indicator of poorer prognosis in several of solid malignancies.


Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Neoplasms/diagnosis , Neoplasms/genetics , Biomarkers, Tumor/genetics , Disease Progression , Disease-Free Survival , Down-Regulation , Humans , Neoplasm Recurrence, Local/diagnosis , Neoplasm Recurrence, Local/genetics , Prognosis , Proportional Hazards Models , Publication Bias , Survival Analysis
12.
Am J Cancer Res ; 7(3): 473-483, 2017.
Article En | MEDLINE | ID: mdl-28401005

Globally, gastric cancer is the second leading cause of cancer deaths because of the lack of effective treatments for patients with advanced tumors when curative surgery is not possible. Thus, there is an urgent need to identify molecular targets in gastric cancer that can be used for developing novel therapies and prolonging patient survival. Checkpoint kinase 1 (Chk1) is a crucial regulator of cell cycle transition in DNA damage response (DDR). In our study, we report that Chk1 plays an important role in promoting gastric cancer cell survival and growth, which serves as an effective therapeutic target in gastric cancer. First, Chk1 ablation by small interfering RNA could significantly inhibit cell proliferation and sensitize the effects of ionizing radiation (IR) treatment in both p53 wild type gastric cancer cell line AGS, and p53 mutant cell line MKN1. Secondly, we tested the anticancer effects of Chk1 chemical inhibitor LY2606368, which is a novel Chk1/2 targeted drug undergoing clinical trials in many malignant diseases. We found that LY2606368 can induce DNA damage, and remarkably suppress cancer proliferation and induce apoptosis in AGS and MKN1 cells. Moreover, we identified that LY2606368 can significantly inhibit homologous recombination (HR) mediated DNA repair and thus showed marked synergistic anticancer effect in combination with poly (ADP-ribose) polymerase 1 (PARP1) inhibitor BMN673 in both in vitro studies and in vivo experiments using a gastric cancer PDx model. The synergy between LY2606368 and PARP1 was likely caused by impaired the G2M checkpoint due to LY2606368 treatment, which forced mitotic entry and cell death in the presence of BMN673. In conclusion, we propose that Chk1 is a valued target for gastric cancer treatment, especially Chk1 inhibitor combined with PARP inhibitor may be a more effective therapeutic strategy in gastric cancer.

13.
Oncotarget ; 8(17): 28125-28134, 2017 Apr 25.
Article En | MEDLINE | ID: mdl-28427226

MiR-152 has been reported may be involved in carcinogenesis in gastric cancer. However, its role has not been comprehensively investigated in gastric cancer. We found miR-152 in human gastric cancer tissues were significantly lower than that in matched adjacent normal tissues. Meanwhile, lower miR-152 was also found in gastric cancer cell lines. The stage, tumor size and lymph node metastasis rate were significant higher in low-miR-152 group in clinical patients. Furthermore, there was a marked correlation between the levels of miR-152 and B7-H1 mRNA in gastric cancer tissues. Mechanistically, miR-152 directly bind to B7-H1 3' untranslated region in gastric cancer cell and inhibited B7-H1 expression. Functional study demonstrated that elevation of miR-152 enhanced T cells proliferation and effector cytokines production via inhibiting B7-H1/PD-1 pathway. In conclusion, our work identified a novel mechanism by which immune response is increased by expression of miR-152 via targeting B7-H1. MiR-152 may be a potential therapeutic approach for gastric cancer.


B7-H1 Antigen/genetics , Gene Expression Regulation, Neoplastic , Immunomodulation/genetics , MicroRNAs/genetics , RNA Interference , Stomach Neoplasms/genetics , Stomach Neoplasms/immunology , Aged , Aged, 80 and over , Biomarkers, Tumor , Case-Control Studies , Cell Line, Tumor , Cell Proliferation , Female , Humans , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Stomach Neoplasms/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Tumor Burden
14.
Cancer Biol Ther ; 18(4): 229-236, 2017 04 03.
Article En | MEDLINE | ID: mdl-28281917

The prevalence of neuroendocrine tumors (NETs) has recently been increasing. Although various drugs such as Octreotide and its analogs show certain efficacy, NETs in many patients progress and metastasize. It is desirable to develop new interventions to improve the therapy. Here we show that human neuroendocrine tumor BON cells are resistant to several drugs commonly used for NET therapy, including Octreotide that activates somatostatin receptor-induced anti-proliferation, and Capecitabine and Temozolimide that damage DNA. In contrast, an inhibitor (IBET) to an epigenetic regulator, Brd4 that binds acetylated histones and upregulates transcription of multiple genes including protooncogene c-Myc, potently inhibited the NET cells. We found that IBET increased the protein levels of cyclin-dependent kinase (CDK) inhibitor p27kip/cip (or p27), but not its mRNA levels. Moreover, the p27 induction at protein level by IBET was at least partly through increasing the protein stability of p27. The increased protein stability of p27 likely resulted from IBET-mediated suppression of Skp2, an E3 ligase that can mediate p27 degradation by increasing its ubiquitinylation. These findings unravel a new mechanism whereby the IBET-induced repression of proliferation of neuroendocrine cells.


Antineoplastic Agents/pharmacology , Benzodiazepines/pharmacology , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Neuroendocrine Tumors/drug therapy , Neuroendocrine Tumors/pathology , Nuclear Proteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Antineoplastic Agents/therapeutic use , Benzodiazepines/therapeutic use , Capecitabine/pharmacology , Capecitabine/therapeutic use , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation/drug effects , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , Drug Resistance, Neoplasm , Epigenesis, Genetic/drug effects , Humans , Octreotide/pharmacology , Octreotide/therapeutic use , Protein Stability/drug effects , RNA, Messenger/metabolism , S-Phase Kinase-Associated Proteins/metabolism , Temozolomide , Transcriptional Activation/drug effects , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/drug effects , Up-Regulation
15.
PLoS One ; 12(1): e0168822, 2017.
Article En | MEDLINE | ID: mdl-28056089

The most common cause of gastric cancer is infection with helicobacter pylori (HP), but the associated molecular mechanism is not well understood. In the present study, we found a marked increase in the expression of B7-H1, a member of the B7 co-stimulatory family of molecules that bind to programmed death-1 (PD-1) and play a critical immunoregulatory role in the cell-mediated immune response, in HP-positive gastric cancer tissue. Infection of cultured gastric cancer cells with HP promoted B7-H1 expression and inhibited miR-152 and miR-200b expression. We further demonstrated that these two miRNAs targeted B7-H1 mRNA and suppressed B7-H1 expression in gastric cancer cells. Finally, B7-H1 expression was found to correlate with miR-152 and miR-200b levels in gastric tumor tissues from human patients. Our findings suggest a novel mechanism by which HP infection promotes gastric cancer and also suggest potential targets, i.e., miR-152 and miR-200b, for the prevention and treatment of gastric cancer.


B7-H1 Antigen/metabolism , Helicobacter pylori/pathogenicity , MicroRNAs/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/microbiology , 3' Untranslated Regions/genetics , B7-H1 Antigen/genetics , Cell Line, Tumor , Flow Cytometry , Fluorescent Antibody Technique , Humans , MicroRNAs/genetics , Real-Time Polymerase Chain Reaction , Stomach Neoplasms/genetics
16.
Oxid Med Cell Longev ; 2016: 9203716, 2016.
Article En | MEDLINE | ID: mdl-26881046

Maresin 1 (MaR 1) was recently reported to have protective properties in several different animal models of acute inflammation by inhibiting inflammatory response. However, its function in acute liver injury is still unknown. To address this question, we induced liver injury in BALB/c mice with intraperitoneal injection of carbon tetrachloride with or without treatment of MaR 1. Our data showed that MaR 1 attenuated hepatic injury, oxidative stress, and lipid peroxidation induced by carbon tetrachloride, as evidenced by increased thiobarbituric acid reactive substances and reactive oxygen species levels were inhibited by treatment of MaR 1. Furthermore, MaR 1 increased activities of antioxidative mediators in carbon tetrachloride-treated mice liver. MaR 1 decreased indices of inflammatory mediators such as tumor necrosis factor-α, interleukin-6, interleukin-1ß, monocyte chemotactic protein 1, myeloperoxidase, cyclooxygenase-2, and inducible nitric oxide synthase. Administration of MaR 1 inhibited activation of nuclear factor kappa B (NF-κb) and mitogen-activated protein kinases (MAPKs) in the liver of CCl4 treated mice. In conclusion, these results suggested the antioxidative, anti-inflammatory properties of MaR 1 in CCl4 induced liver injury. The possible mechanism is partly implicated in its abilities to inhibit ROS generation and activation of NF-κb and MAPK pathway.


Docosahexaenoic Acids/pharmacology , Liver/pathology , Alanine Transaminase/blood , Animals , Antioxidants/metabolism , Aspartate Aminotransferases/blood , Carbon Tetrachloride , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cyclooxygenase 2/metabolism , Cytokines/blood , Docosahexaenoic Acids/administration & dosage , Glutathione/metabolism , Hep G2 Cells , Humans , Inflammation Mediators/metabolism , Liver/drug effects , Male , Mice, Inbred BALB C , Mitogen-Activated Protein Kinases/metabolism , NF-KappaB Inhibitor alpha/metabolism , Nitric Oxide Synthase Type II/metabolism , Oxidative Stress/drug effects , Peroxidase/metabolism , Phosphorylation/drug effects , Reactive Oxygen Species/metabolism , Thiobarbituric Acid Reactive Substances/metabolism , Transcription Factor RelA/metabolism
17.
Mol Med Rep ; 13(3): 2850-6, 2016 Mar.
Article En | MEDLINE | ID: mdl-26847687

Monoacylglycerol lipase (MAGL) is involved in the degradation of triacylglycerol. Previous studies have demonstrated that MAGL regulates tumor growth and metastasis via fatty acid networks, and is associated with colorectal cancer. JZL184 is a MAGL inhibitor, which in the present study was administered to colorectal cancer cell lines, resulting in decreased tumor proliferation, increased apoptosis and increased tumor cell sensitivity to 5-fluorouracil. B­cell lymphoma 2 (Bcl­2) and Bcl­2­associated X protein (Bax) are key proteins in apoptosis. The expression levels of Bcl­2/Bax were determined in colorectal cancer cell lines following JZL184 administration, and it was observed that the mRNA and protein expression levels of Bcl­2 were decreased, whereas the expression levels of Bax were increased. These results indicated that JZL184 may induce tumor cell apoptosis by regulating the expression of Bcl­2 and Bax. Epithelial-mesenchymal transition (EMT) is closely associated with metastasis. Administration of JZL184 in various malignant colorectal cancer cell lines suppressed migration and altered the expression of EMT markers; E­cadherin was increased, whereas the expression levels of vimentin and zinc finger protein SNAI1 were decreased. These results suggested that JZL184 was able to regulate the EMT process, in order to control the migration of colorectal cancer cells, particularly in tumors with a stronger metastatic capability. Therefore, in colorectal cancer, MAGL may be considered a potential therapeutic target and JZL184 may be a possible therapeutic agent.


Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Benzodioxoles/pharmacology , Cell Movement/drug effects , Piperidines/pharmacology , Cell Proliferation , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Drug Screening Assays, Antitumor , HCT116 Cells , Humans , Monoacylglycerol Lipases/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2-Associated X Protein/metabolism
18.
Mediators Inflamm ; 2016: 3798465, 2016.
Article En | MEDLINE | ID: mdl-28042205

Sepsis, frequently caused by infection of bacteria, is considered as an uncontrollable systematic inflammation response syndrome (SIRS). Maresin 1 (Mar1) is a new proresolving mediator with potent anti-inflammatory effect in several animal models. However, its effect in sepsis is still not investigated. To address this question, we developed sepsis model in BALB/c mice by cecal ligation and puncture (CLP) with or without Mar1 treatment. Our data showed that Mar1 markedly improved survival rate and decreased the levels of proinflammatory cytokines in CLP mice such as interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), and interleukin-1ß (IL-1ß). Furthermore, Mar1 reduced serum level of lipopolysaccharide (LPS) and enhanced the bacteria clearance in mice sepsis model. Moreover, Mar1 attenuated lung injury and decreased level of alanine transaminase (ALT), aspartate transaminase (AST), creatinine (Cre), and blood urea nitrogen (BUN) in serum in mice after CLP surgery. Treatment with Mar1 inhibited activation of nuclear factor kappa B (NF-κb) pathway. In conclusion, Mar1 exhibited protective effect in sepsis by reducing LPS, bacteria burden in serum, inhibiting inflammation response, and improving vital organ function. The possible mechanism is partly involved in inhibition of NF-κb activation.


Docosahexaenoic Acids/pharmacology , Inflammation/drug therapy , Sepsis/metabolism , Animals , Blood Urea Nitrogen , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Kidney/metabolism , Lipopolysaccharides/blood , Liver/metabolism , Mice , Mice, Inbred BALB C , NF-kappa B/antagonists & inhibitors , Sepsis/prevention & control , Systemic Inflammatory Response Syndrome/metabolism , Tumor Necrosis Factor-alpha/metabolism , Weight Loss
19.
Oncol Lett ; 9(4): 1833-1838, 2015 Apr.
Article En | MEDLINE | ID: mdl-25789052

Cancer stem-like cells (CSCs) are a rare tumorigenic population with the ability to self-renew in numerous cancer types. Their existence is considered a pivotal contributor to tumor recurrence. B7-H1 is a ligand of inhibitory inducible co-stimulator (ICOS) that is broadly expressed on various human cancers. ICOS acts as a ligand of programmed death-1 (PD-1) on T cells, induces the immune escape of cancer cells and also acts as a receptor mediating anti-apoptotic effects on cancer cells. However, the expression and function of B7-H1 on CSCs is not yet clear. In the present study, gastric cancer samples were collected and the B7-H1 expression in gastric cancer CSCs was detected. Ki67, a proliferation marker, was found to be expressed at a higher rate in B7-H1+ CSCs compared with the B7-H1- counterparts. SGC-7901 cells, a gastric cancer cell line, were cultured in serum-free medium to form sphere cells that possessed stem cell characteristics and could express B7-H1 with the stimulation of interferon-γ. The proliferative ability of sphere cells was enhanced following B7-H1 activation with recombinant PD-1 in vivo and in vitro. This effect could be eliminated by neutralizing B7-H1. Overall, B7-H1 can act as a stimulating receptor for CSCs, and induce CSC proliferation. Blocking B7-H1 on CSCs may possess therapeutic potential for treating gastric cancer.

...