Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Plant Cell Rep ; 43(4): 102, 2024 Mar 18.
Article in English | MEDLINE | ID: mdl-38499710

ABSTRACT

KEY MESSAGE: The transcriptomic, phenotypic and metabolomic analysis of transgenic plants overexpressing GhMPK31 in upland cotton revealed the regulation of H2O2 burst and the synthesis of defensive metabolites by GhMPK31. Mitogen-activated protein kinases (MAPKs) are a crucial class of protein kinases, which play an essential role in various biological processes in plants. Upland cotton (G. hirsutum) is the most widely cultivated cotton species with high economic value. To gain a better understanding of the role of the MAPK gene family, we conducted a comprehensive analysis of the MAPK gene family in cotton. In this study, a total of 55 GhMPK genes were identified from the whole genome of G. hirsutum. Through an investigation of the expression patterns under diverse stress conditions, we discovered that the majority of GhMPK family members demonstrated robust responses to abiotic stress, pathogen stress and pest stress. Furthermore, the overexpression of GhMPK31 in cotton leaves led to a hypersensitive response (HR)-like cell death phenotype and impaired the defense capability of cotton against herbivorous insects. Transcriptome and metabolomics data analysis showed that overexpression of GhMPK31 enhanced the expression of H2O2-related genes and reduced the accumulation of defensive related metabolites. The direct evidence of GhMPK31 interacting with GhRBOHB (H2O2-generating protein) were found by Y2H, BiFC, and LCI. Therefore, we propose that the increase of H2O2 content caused by overexpression of GhMPK31 resulted in HR-like cell death in cotton leaves while reducing the accumulation of defensive metabolites, ultimately leading to a decrease in the defense ability of cotton against herbivorous insects. This study provides valuable insights into the function of MAPK genes in plant resistance to herbivorous insects.


Subject(s)
Gossypium , Hydrogen Peroxide , Gossypium/metabolism , Hydrogen Peroxide/metabolism , Gene Expression Profiling , Transcriptome , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Gene Expression Regulation, Plant , Plant Proteins/genetics , Plant Proteins/metabolism , Phylogeny
2.
Sensors (Basel) ; 24(6)2024 Mar 16.
Article in English | MEDLINE | ID: mdl-38544170

ABSTRACT

In recent years, the rapid prevalence of high-definition video in Internet of Things (IoT) systems has been directly facilitated by advances in imaging sensor technology. To adapt to limited uplink bandwidth, most media platforms opt to compress videos to bitrate streams for transmission. However, this compression often leads to significant texture loss and artifacts, which severely degrade the Quality of Experience (QoE). We propose a latent feature diffusion model (LFDM) for compressed video quality enhancement, which comprises a compact edge latent feature prior network (ELPN) and a conditional noise prediction network (CNPN). Specifically, we first pre-train ELPNet to construct a latent feature space that captures rich detail information for representing sharpness latent variables. Second, we incorporate these latent variables into the prediction network to iteratively guide the generation direction, thus resolving the problem that the direct application of diffusion models to temporal prediction disrupts inter-frame dependencies, thereby completing the modeling of temporal correlations. Lastly, we innovatively develop a Grouped Domain Fusion module that effectively addresses the challenges of diffusion distortion caused by naive cross-domain information fusion. Comparative experiments on the MFQEv2 benchmark validate our algorithm's superior performance in terms of both objective and subjective metrics. By integrating with codecs and image sensors, our method can provide higher video quality.

3.
Gut ; 72(4): 624-637, 2023 04.
Article in English | MEDLINE | ID: mdl-36002248

ABSTRACT

OBJECTIVE: Many cancers engage embryonic genes for rapid growth and evading the immune system. SOX9 has been upregulated in many tumours, yet the role of SOX9 in mediating immunosuppressive tumour microenvironment is unclear. Here, we aim to dissect the role of SOX9-mediated cancer stemness attributes and immunosuppressive microenvironment in advanced gastric adenocarcinoma (GAC) for novel therapeutic discoveries. METHODS: Bulk RNAseq/scRNA-seq, patient-derived cells/models and extensive functional studies were used to identify the expression and functions of SOX9 and its target genes in vitro and in vivo. Immune responses were studied in PBMCs or CD45+ immune cells cocultured with tumour cells with SOX9high or knockout and the KP-Luc2 syngeneic models were used for efficacy of combinations. RESULTS: SOX9 is one of the most upregulated SOX genes in GAC and highly expressed in primary and metastatic tissues and associated with poor prognosis. Depletion of SOX9 in patient-derived GAC cells significantly decreased cancer stemness attributes, tumour formation and metastases and consistently increased CD8+ T cell responses when cocultured with PBMCs/CD45+ cells from GAC patients. RNA sequencing identified the leukaemia inhibitory factor (LIF) as the top secreted molecule regulated by SOX9 in tumour cells and was enriched in malignant ascites and mediated SOX9-induced M2 macrophage repolarisation and inhibited T cell function. CONCLUSION: Epithelial SOX9 is critical in suppressing CD8+ T cell responses and modified macrophage function in GAC through the paracrine LIF factor. Cotargeting LIF/LIFR and CSF1R has great potential in targeting SOX9-mediated cancer stemness, T cell immunosuppression and metastases suggesting the novel combination therapy against advanced GAC.


Subject(s)
Adenocarcinoma , Stomach Neoplasms , Humans , Tumor Microenvironment , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Immunosuppressive Agents , Immunosuppression Therapy , SOX9 Transcription Factor/genetics
4.
Gut ; 71(3): 467-478, 2022 03.
Article in English | MEDLINE | ID: mdl-33785559

ABSTRACT

BACKGROUND: Gastric adenocarcinoma (GAC) is a lethal disease with limited therapeutic options. Genetic alterations in chromatin remodelling gene AT-rich interactive domain 1A (ARID1A) and mTOR pathway activation occur frequently in GAC. Targeting the mechanistic target of rapamycin (mTOR) pathway in unselected patients has failed to show survival benefit. A deeper understanding of GAC might identify a subset that can benefit from mTOR inhibition. METHODS: Genomic alterations in ARID1A were analysed in GAC. Mouse gastric epithelial cells from CK19-Cre-Arid1Afl/fl and wild-type mice were used to determine the activation of oncogenic genes due to loss of Arid1A. Functional studies were performed to determine the significance of loss of ARID1A and the sensitivity of ARID1A-deficient cancer cells to mTOR inhibition in GAC. RESULTS: More than 30% of GAC cases had alterations (mutations or deletions) of ARID1A and ARID1A expression was negatively associated with phosphorylation of S6 and SOX9 in GAC tissues and patient-derived xenografts (PDXs). Activation of mTOR signalling (increased pS6) and SOX9 nuclear expression were strongly increased in Arid1A-/- mouse gastric tissues which could be curtailed by RAD001, an mTOR inhibitor. Knockdown of ARID1A in GAC cell lines increased pS6 and nuclear SOX9 and increased sensitivity to an mTOR inhibitor which was further amplified by its combination with fluorouracil both in vitro and in vivo in PDXs. CONCLUSIONS: The loss of ARID1A activates pS6 and SOX9 in GAC, which can be effectively targeted by an mTOR inhibitor. Therefore, our studies suggest a new therapeutic strategy of clinically targeting the mTOR pathway in patients with GAC with ARID1A deficiency.


Subject(s)
Adenocarcinoma/etiology , DNA-Binding Proteins/physiology , SOX9 Transcription Factor/physiology , Signal Transduction/physiology , Stomach Neoplasms/etiology , TOR Serine-Threonine Kinases/physiology , Transcription Factors/physiology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Cell Culture Techniques , Cell Line, Tumor , Cell Proliferation , Mice , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology
5.
Small ; 17(37): e2102323, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34288454

ABSTRACT

The relative low hole mobility of p-channel building block device challenges the continued miniaturization of modern electronic chips. Metal-semiconductor junction is always an efficient strategy to control the carrier concentration of channel semiconductor, benefiting the carrier mobility regulation of building block device. In this work, complementary metal oxide semiconductor (CMOS)-compatible metals are selected to deposit on the surface of the important p-channel building block of GaSb nanowire field-effect-transistors (NWFETs), demonstrating the efficient strategy of hole mobility enhancement by metal-semiconductor junction. When deposited with lower work function metal of Al, the peak hole mobility of GaSb NWFET can be enhanced to as high as ≈3372 cm2 V-1 s-1 , showing three times than the un-deposited one. The as-studied metal-semiconductor junction is also efficient for the hole mobility enhancement of other p-channel devices, such as GaAs NWFET, GaAs film FET, and WSe2 FET. With the enhanced mobility, the as-constructed CMOS inverter shows good invert characteristics, showing a relatively high gain of ≈18.1. All results may be regarded as important advances to the next-generation electronics.

6.
Cancer Sci ; 110(7): 2145-2155, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31087525

ABSTRACT

Gastric cancer (GC) is a common cause of cancer-related death worldwide. As a result of the lack of reliable diagnostic or prognostic biomarkers for GC, patient prognosis is still poor. Therefore, there is an urgent need for studies examining the underlying pathogenesis of GC in order to find effective biomarkers. LRRN1 (leucine-rich repeat neuronal protein-1) is a type I transmembrane protein that plays an important role in the process of nerve development and regeneration. However, its role in cancer, especially in GC, remains unclear. In the present study, we found that LRRN1 expression is upregulated in GC tissues and that high LRRN1 expression is associated with poor prognosis. siRNA and shRNA-mediated knockdowns of LRRN1 expression promoted GC cell apoptosis and activation of the Fas/FasL pathway. LRRN1 knockdown also resulted in upregulation of JUN, a subunit of the transcription factor AP-1 (activator protein-1). This suggests that LRRN1 suppresses GC cell apoptosis by downregulating AP-1, resulting in inhibition of the Fas/FasL pathway. These results confirm that LRRN1 plays a significant role in GC pathogenesis. Moreover, LRRN1 may be a potential prognostic biomarker and therapeutic target for GC.


Subject(s)
Membrane Proteins/genetics , Membrane Proteins/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Signal Transduction , Stomach Neoplasms/pathology , Up-Regulation , Animals , Apoptosis , Cell Line, Tumor , Fas Ligand Protein/metabolism , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Male , Mice , Neoplasm Transplantation , Nerve Tissue Proteins , Prognosis , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Survival Analysis , Transcription Factor AP-1/metabolism , fas Receptor/metabolism
7.
Ann Surg Oncol ; 26(11): 3745-3755, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31087180

ABSTRACT

BACKGROUND: A recent study showed that circulating exosomal PD-L1 is an effective predictor for anti-PD-1 therapy in melanomas. Exosomal PD-L1 induced immunosuppression microenvironments in cancer patients. However, its prognostic value and immunosuppressive effect in gastric cancer (GC) were poorly understood. METHODS: We retrospectively evaluated the prognostic value of exosomal PD-L1 and soluble PD-L1 in preoperative plasma of 69 GC patients. The correlation between exosomal PD-L1 and the T cell counts or cytokine in the plasma was evaluated in 31 metastatic GC patients before chemotherapy. RESULTS: Overall survival (OS) was significantly lower in the high exosomal PD-L1 group compared with the low exosomal PD-L1 group (P = 0.004). Exosomal PD-L1 was an independent prognostic factor in GC (n = 69, 95% confidence interval = 1.142-7.669, P = 0.026). However, soluble PD-L1 showed no correlation with OS (P = 0.139). Additionally, exosomal PD-L1 in the plasma samples of 31 metastatic GC patients was negatively associated with CD4+ T cell count (P = 0.001, R = - 0.549), CD8+ T-cell count (P = 0.054, R = - 0.349), and granzyme B (P = 0.002, R = - 0.537), indicating that exosomal PD-L1 was associated with immunosuppressive status of GC patients. GC cells also secreted exosomal PD-L1 and were positively associated with the amount of PD-L1 in corresponding GC cell lines. Besides, exosomal PD-L1 significantly decreased T-cell surface CD69 and PD-1 expressions compared with soluble PD-L1 due to its stable and MHC-I expression. CONCLUSIONS: Overall, exosomal PD-L1 predicts the worse survival and reflects the immune status in GC patients, resulting from a stronger T-cell dysfunction due to its stable and MHC-I expression.


Subject(s)
Adenocarcinoma/secondary , B7-H1 Antigen/blood , B7-H1 Antigen/immunology , Exosomes/metabolism , Immunosuppression Therapy , Stomach Neoplasms/pathology , Adenocarcinoma/blood , Adenocarcinoma/immunology , Adenocarcinoma/surgery , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/blood , Biomarkers, Tumor/immunology , Female , Follow-Up Studies , Humans , Lymphatic Metastasis , Male , Middle Aged , Prognosis , Retrospective Studies , Stomach Neoplasms/blood , Stomach Neoplasms/immunology , Stomach Neoplasms/surgery , Survival Rate
8.
Cell Biol Int ; 43(1): 33-43, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30468278

ABSTRACT

Bufalin, a natural small-molecule compound derived from the traditional Chinese medicine Chan su, has shown promising anti-cancer effects against a broad variety of cancer cells through different mechanisms. It has been reported to induce autophagy in gastric cancer cells. However, the molecular mechanism involved is not fully elucidated. In the present study, we aimed to investigate the molecular mechanism by which bufalin induce autophagy in human gastric cancer cells. We found that bufalin induced apoptosis and autophagy in gastric cancer cells, and autophagy prevented human gastric cancer cells from undergoing apoptosis. Bufalin treatment changed the expression of autophagy-related proteins. Moreover, phosphorylated Akt, mTOR, and p70S6K were all significantly decreased, while phosphorylated ERK1/2 was increased by bufalin. Pretreatment of MGC803 cells with the ERK1/2-specific inhibitor PD98059 led to the down-regulation of LC3 II. Further study showed that Cbl-b positively regulated autophagy by suppressing mTOR and enhancing ERK1/2 activation. Therefore, our data provide evidence that bufalin induces autophagy in MGC803 cells via both Akt/mTOR/p70S6K and ERK signaling pathways, and Cbl-b-mediated suppression of mTOR and activation of ERK1/2 might play an important role.


Subject(s)
Autophagy/drug effects , Bufanolides/pharmacology , MAP Kinase Signaling System/drug effects , Proto-Oncogene Proteins c-cbl/metabolism , Stomach Neoplasms/pathology , TOR Serine-Threonine Kinases/metabolism , Apoptosis/drug effects , Autophagy-Related Proteins/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Humans , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Stomach Neoplasms/ultrastructure
9.
Cell Biol Int ; 43(2): 98-107, 2019 Feb.
Article in English | MEDLINE | ID: mdl-29972257

ABSTRACT

Although epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) could greatly improve the prognosis of NSCLC patients harboring activating EGFR mutations, drug resistance still remains a major obstacle to successful treatment. Our previous study found that the EGFR-TKI icotinib could upregulate the expression of Casitas-B-lineage lymphoma protein-B (Cbl-b), an E3 ubiquitin ligase. In the present study, we aimed to clarify the potential role of Cbl-b in the resistance to icotinib, and the underlying mechanisms using EGFR-mutant cell lines. We found that icotinib inhibited the proliferation of mutant-EGFR NSCLC cells (PC9 and HCC827), and upregulated the expression of Cbl-b at both the protein and mRNA levels. Cbl-b knockdown decreased the sensitivity of PC9 and HCC827 cells to icotinib, and partially restored icotinib-inhibited AKT activation in PC9 cells. On the contrary, Cbl-b overexpression could partly reverse the drug resistance in PC9 icotinib-resistant cells (PC9/IcoR). Moreover, overexpressing p65, the main member of transcription factor NF-κB family, reversed the icotinib-mediated upregulation of Cbl-b. Collectively, these data suggest that icotinib could upregulate Cbl-b mediated by NF-κB inhibition, and Cbl-b contribute to the icotinib sensitivity in EGFR-mutant NSCLC cells. This study highlights that low expression of Cbl-b might be the key obstacles in the efficacy of icotinib therapy.


Subject(s)
Crown Ethers/pharmacology , Drug Resistance, Neoplasm/genetics , NF-kappa B/metabolism , Proto-Oncogene Proteins c-cbl/metabolism , Quinazolines/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mutation , NF-kappa B/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-cbl/antagonists & inhibitors , Proto-Oncogene Proteins c-cbl/genetics , RNA Interference , RNA, Small Interfering/metabolism , Up-Regulation/drug effects
10.
Exp Cell Res ; 373(1-2): 180-187, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30367831

ABSTRACT

Although anti-programmed death ligand-1 (PD-L1) therapy has shown light in treatment of gastric cancer, only a limited number of patients respond to the treatment. In addition to its immunosuppressive effect, PD-L1 is involved in other functions of tumor cells. Previously study showed that PD-L1 promoted EMT in lung cancer cells. However, the other effect and role of PD-L1 in gastric cancer remains unclear. In the present study, we first demonstrated that PD-L1 promoted the proliferation and migration in gastric cancer cell lines. We found that another STAT family member, STAT5a, is involved in regulating the expression of PD-L1 in gastric cancer. Additionally, Cbl-b interacted and ubiquitated STAT5a, down-regulated the expression of STAT5a and PD-L1. Moreover, bioinformatics predictions and experimental data showed that Cbl-b is a target gene of the microRNA miR-940. We further found that miR-940 promoted the proliferation and migration of gastric cancer in vivo and in vitro. Taken together, our findings suggest that miR-940/Cbl-b/STAT5a axis regulated the expression of PD-L1, which promotes the proliferation and migration of gastric cancer cells.


Subject(s)
B7-H1 Antigen/metabolism , MicroRNAs/metabolism , Stomach Neoplasms/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , B7-H1 Antigen/biosynthesis , B7-H1 Antigen/physiology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Jurkat Cells , Mice, Inbred BALB C , Mice, Nude , Proto-Oncogene Proteins c-cbl/genetics , Proto-Oncogene Proteins c-cbl/metabolism , STAT5 Transcription Factor/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Stomach Neoplasms/physiopathology , Tumor Suppressor Proteins/metabolism , Ubiquitination , Up-Regulation
11.
Exp Cell Res ; 368(2): 194-201, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29723509

ABSTRACT

A variety of solid tumors are surrounded by a hypoxic microenvironment, which is known to be associated with high metastatic capability and resistance to various clinical therapies, contributing to a poor survival rate for cancer patients. Although the majority of previous studies on tumor-associated hypoxia have focused on acute hypoxia, chronic hypoxia more closely mimics the actual hypoxic microenvironment of a tumor. In this study, two novel hypoxia-resistant gastric cancer (HRGC) cell lines which could grow normally in 2% oxygen were established. The long non-coding RNA UCA1 was upregulated in HRGC cells, which promoted their migration. Bioinformatics analysis and a luciferase reporter assay showed that miR-7-5p could bind to specific sites of UCA1 to regulate the target EGFR through competitive endogenous RNA function. UCA1 directly interacted with miR-7-5p and decreased the binding of miR-7-5p to the EGFR 3'-untranslated region, which suppressed the degradation of EGFR mRNA by miR-7-5p. Therefore, long-term hypoxia induced UCA1 to promote cell migration by enhancing the expression of EGFR. This study thus reveals a new mechanism by which a hypoxic microenvironment promotes tumor metastasis, and highlights UCA1 as a potential biomarker for predicting the metastasis of gastric cancer to guide clinical treatment.


Subject(s)
Cell Hypoxia/genetics , Cell Movement/genetics , Drug Resistance, Neoplasm/genetics , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Stomach Neoplasms/genetics , Up-Regulation/genetics , 3' Untranslated Regions/genetics , Cell Line, Tumor , Cell Proliferation/genetics , ErbB Receptors/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , RNA, Messenger/genetics , Transcriptional Activation/genetics , Tumor Microenvironment/genetics
12.
Exp Cell Res ; 368(2): 184-193, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29709516

ABSTRACT

Cancer-associated fibroblasts (CAFs) are major components of the tumor stroma and regulators of tumor progression. However, the molecular mechanism by which CAFs promote gastric cancer progression should be further explored. In our study, we found that interleukin-11 (IL-11) secretion was significantly increased when CAFs were co-cultured with gastric cancer cells. Co-culture system-derived IL-11 promoted the migration and invasion of gastric cancer cells, whereas the increase of migration and invasion was attenuated by a neutralizing antibody of IL-11 or inhibition of JAK/STAT3 and MAPK/ERK pathways with specific inhibitors. Taken together, these results revealed that CAFs play a significant role in the gastric cancer progression in the tumor microenvironment through IL-11-STAT3/ERK signaling by upregulating MUC1. Also, IL-11 targeted therapy can achieve an effective treatment against gastric cancer indirectly by exerting their action on stromal fibroblasts.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Interleukin-11/metabolism , Mucin-1/metabolism , Neoplasm Metastasis/pathology , Stomach Neoplasms/metabolism , Up-Regulation/physiology , Animals , Cancer-Associated Fibroblasts/pathology , Cell Line, Tumor , Coculture Techniques/methods , Female , Humans , MAP Kinase Signaling System/physiology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness/pathology , STAT3 Transcription Factor/metabolism , Signal Transduction/physiology , Stomach Neoplasms/pathology , Transcriptional Activation/physiology , Tumor Microenvironment/physiology
13.
Br J Cancer ; 118(11): 1453-1463, 2018 05.
Article in English | MEDLINE | ID: mdl-29755114

ABSTRACT

BACKGROUND: BRAF mutation is associated with poor clinical outcome of patients with malignant tumours, and mediates resistance to chemotherapy and targeted therapy. This study aimed to determine whether V600E mutant and wild type BRAF colorectal cancers exhibit distinct sensitivities to the dual BRAF inhibitor AZ304. METHODS: Kinase activity was assessed by the AlphaScreen assay. Then, MTT assay, EdU assay, colony-formation assay and Western blot were performed to evaluate the anti-tumour effects of AZ304 in vitro. In vivo efficacy was investigated by xenograft analysis and immunohistochemistry. RESULTS: AZ304 exerted potent inhibitory effects on both wild type and V600E mutant forms of the serine/threonine-protein kinase BRAF, with IC50 values of 79 nM and 38 nM, respectively. By suppressing ERK phosphorylation, AZ304 effectively inhibited a panel of human cancer cell lines with different BRAF and RAS genetic statuses. In selected colorectal cancer cell lines, AZ304 significantly inhibited cell growth in vitro and in vivo, regardless of BRAF genetic status. In addition, the EGFR inhibitor Cetuximab enhanced the potency of AZ304 independently of BRAF mutational status. CONCLUSIONS: The BRAF inhibitor AZ304 has broad spectrum antitumour activity, which is significantly enhanced by combination with Cetuximab in colorectal cancers in vitro and in vivo.


Subject(s)
Cetuximab/administration & dosage , Colorectal Neoplasms/drug therapy , Extracellular Signal-Regulated MAP Kinases/metabolism , Protein Kinase Inhibitors/administration & dosage , Proto-Oncogene Proteins B-raf/genetics , Animals , Caco-2 Cells , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cetuximab/pharmacology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Drug Synergism , Female , HT29 Cells , Humans , Mice , Mutation , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Xenograft Model Antitumor Assays , ras Proteins/genetics
14.
BMC Cancer ; 18(1): 1138, 2018 Nov 20.
Article in English | MEDLINE | ID: mdl-30453894

ABSTRACT

BACKGROUND: Multiple primary malignant tumors (MPMT) refers to the presence of two or more primary cancers of different organs in the same patient. MPMT is a sparse disease in the past, but there has been a gradual increase in the morbidity. Since multiple primary malignant tumors treatment methods differ, it is essential for clinicians to be able to distinguish between separate primary lesions and metastasis. CASE PRESENTATION: We present the case of a 57-year-old woman with MPMT presenting with cancer in the left breast and synchronous double primary lung adenocarcinomas. We used IHC and epidermal growth factor receptor(EGFR)mutation to analyze genomic alteration profiles in the patient to validate the difference among the pathological assessments and the clinical differences between double primary lesions of lung and breast. EGFR gene analysis of breast cancer lesion revealed no mutations. The left and right lower lobe lung adenocarcinomas contained EGFR gene mutations: an L858R point mutation in exon 21 in the left lesion and a deletion mutation in exon 19 in the right lesion. The breast cancer and both lung adenocarcinomas were surgically resected. To date, the patient has remained disease-free. CONCLUSIONS: Both pathological and molecular assessment adapted in the current study appeared necessary. Mutational analysis of the EGFR gene provided important information not only in the diagnosis and but also in the treatment of MPMT.


Subject(s)
Adenocarcinoma/genetics , Breast Neoplasms/genetics , Lung Neoplasms/genetics , Mutation , Neoplasms, Multiple Primary/genetics , Adenocarcinoma/pathology , Breast Neoplasms/pathology , DNA Mutational Analysis , ErbB Receptors/genetics , Female , Humans , Lung Neoplasms/pathology , Neoplasms, Multiple Primary/pathology
15.
BMC Cancer ; 18(1): 462, 2018 04 24.
Article in English | MEDLINE | ID: mdl-29690901

ABSTRACT

BACKGROUND: Programmed death-ligand 1 (PD-L1) is an immunosuppressor that plays an important role in cancer treatments. Although majority of the studies demonstrated that PD-L1 expression was regulated by cellular intrinsic and extrinsic controls, and IFN-γ was a key molecule of extrinsic control, other studies imply that other cytokines play important roles in PD-L1 expression. In this study, we investigated the regulation of PD-L1 by chemokine signaling pathway in gastric cancer (GC) cells. METHODS: Bioinformatics was used to explore the PD-L1-related genes in GC and propose a hypothesis. PD-L1 and CXCR3 expression were detected by western blot in SGC7901 and MKN74 cell lines. Meanwhile, PD-L1 and CXCR3 expressions were immunohistochemically assessed for their relevance. Moreover, PD-L1, pSTAT3 and pAkt were detected after treatment with CXCL9/10/11. Furthermore,PD-L1, pSTAT3 and pAkt were evaluated after blocking chemokine signaling in SGC7901 cells. RESULTS: Based on online database analysis, CXCL9/10/11-CXCR3 is proposed to upregulate PD-L1 expression by activating the STAT and PI3K-Akt pathways. This hypothesis was confirmed by in vitro and vivo experiments. CXCR3 and PD-L1 were expressed in GC cell lines and tissues, and the expression of CXCR3 and PD-L1 was positively related. PD-L1 was upregulated after treatment with CXCL9/10/11, accompanied by activation of STAT3 and Akt. After blocking chemokine signaling, upregulation of PD-L1 and activation of STAT3 and Akt were diminished. CONCLUSIONS: CXCL9/10/11-CXCR3 upregulated the expression of PD-L1 by activating the STAT and PI3K-Akt signaling pathways in GC cells. There was a significant positive correlation between the expression of PD-L1 and CXCR3 in gastric cancer patient tissues.


Subject(s)
B7-H1 Antigen/genetics , Chemokines, CXC/metabolism , Gene Expression Regulation, Neoplastic , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , B7-H1 Antigen/metabolism , Biomarkers , Cell Line, Tumor , Chemokine CXCL10/metabolism , Chemokine CXCL11/metabolism , Chemokine CXCL9/metabolism , Computational Biology/methods , Humans , Immunohistochemistry , Molecular Sequence Annotation , Neoplasm Staging , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, CXCR3/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Stomach Neoplasms/pathology
16.
BMC Cancer ; 18(1): 681, 2018 Jun 25.
Article in English | MEDLINE | ID: mdl-29940895

ABSTRACT

BACKGROUND: MicroRNAs can be used in the prognosis of malignancies; however, their regulatory mechanisms are unknown, especially in pancreatic ductal adenocarcinoma (PDAC). METHODS: In 120 PDAC specimens, miRNA levels were assessed by quantitative real time polymerase chain reaction (qRT-PCR). Then, the role of miR-29b-2-5p in cell proliferation was evaluated both in vitro (Trypan blue staining and cell cycle analysis in the two PDAC cell lines SW1990 and Capan-2) and in vivo using a xenograft mouse model. Next, bioinformatics methods, a luciferase reporter assay, Western blot, and immunohistochemistry (IHC) were applied to assess the biological effects of Cbl-b inhibition by miR-29b-2-5p. Moreover, the relationship between Cbl-b and p53 was evaluated by immunoprecipitation (IP), Western blot, and immunofluorescence. RESULTS: From the 120 PDAC patients who underwent surgical resection, ten patients with longest survival and ten with shortest survival were selected. We found that high miR-29b-2-5p expression was associated with good prognosis (p = 0.02). The validation cohort confirmed miR-29b-2-5p as an independent prognostic factor in PDAC (n = 100, 95% CI = 0.305-0.756, p = 0.002). Furthermore, miR-29b-2-5p inhibited cell proliferation, induced cell cycle arrest, and promoted apoptosis both in vivo and in vitro. Interestingly, miR-29b-2-5p directly bound the Cbl-b gene, down-regulating its expression and reducing Cbl-b-mediated degradation of p53. Meanwhile, miR-29b-2-5p expression was negatively correlated with Cbl-b in PDAC tissues (r = - 0.33, p = 0.001). CONCLUSIONS: Taken together, these findings indicated that miR-29b-2-5p improves prognosis in PDAC by targeting Cbl-b to promote p53 expression, and would constitute an important prognostic factor in PDAC.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adenocarcinoma/pathology , Carcinoma, Pancreatic Ductal/pathology , MicroRNAs/physiology , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-cbl/genetics , Animals , Cell Line, Tumor , Cell Proliferation , Female , G1 Phase Cell Cycle Checkpoints , Humans , Mice , Mice, Inbred BALB C , Prognosis , Tumor Suppressor Protein p53/physiology
17.
Cell Biol Int ; 42(5): 506-514, 2018 May.
Article in English | MEDLINE | ID: mdl-29274253

ABSTRACT

Tumor necrosis factor-alpha-related apoptosis-inducing ligand (TRAIL) triggers apoptosis by inducing the death-inducing signaling complex (DISC) formation. Recently, TNFα-induced protein 3 (TNFAIP3, A20) was reported to prevent TRAIL-induced caspase 8 cleavage in the DISC by mediating ubiquitination of RIP1 in glioblastoma. However, whether A20 regulates caspase 8 cleavage in the DISC when TRAIL induces apoptosis in gastric cancer cells is unknown. In the present study, A20 interacted with RIP1 and DR4 in MGC803 and SGC7901 gastric cancer cells. Treatment with TRAIL promoted the A20-mediated polyubiquitination of RIP1, caspase 8 translocation into the DISC, and the interaction between caspase 8 and ubiquitinated RIP1. Inhibition of A20 expression prevented the polyubiquitination of RIP1 and promoted caspase 8 cleavage. Moreover, our data clarified that A20 is a target of miR-200a. Overexpression of miR-200a inhibited A20 expression and polyubiquitination of RIP1 and then promoted cleavage of caspase 8 and TRAIL-triggered apoptosis. Taken together, our results indicate that miR-200a enhanced TRAIL-triggered apoptosis in gastric cancer cells by targeting A20.


Subject(s)
Antineoplastic Agents/pharmacology , MicroRNAs/metabolism , Stomach Neoplasms/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism , Apoptosis , Caspase 8/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Stomach Neoplasms/genetics , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Ubiquitination
18.
Cell Biol Int ; 42(10): 1377-1385, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29957841

ABSTRACT

ß-Elemene, an anti-cancer drug extracted from traditional Chinese medicinal herb, showed anti-tumor effects on gastric cancer cells. Our previous studies reported gastric cancer cells are insensitive to TRAIL. However, whether ß-elemene could enhance anti-cancer effects of TRAIL on gastric cancer cells is unknown. In our present study, ß-elemene prevented gastric cancer cell viability in dose-dependent manner, and when combined with TRAIL, obviously inhibited proliferation and promoted apoptosis in gastric cancer cells. Compared to ß-elemene or TRAIL alone, treatment with ß-elemene and TRAIL obviously promoted DR5 clustering as well as translocation of Caspase-8, DR5 and FADD into lipid rafts. This led to cleavage of Caspase-8 and the formation of death-inducing signaling complex (DISC) in lipid rafts. The cholesterol-sequestering agent nystatin partially reversed DR5 clustering and DISC formation, preventing apoptosis triggered by the combination of ß-elemene and TRAIL. Our results suggest that ß-elemene increases the sensitivity of gastric cancer cells to TRAIL partially by promoting the formation of DISC in lipid rafts.


Subject(s)
Sesquiterpenes/metabolism , Sesquiterpenes/pharmacology , Stomach Neoplasms/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Caspase 8/metabolism , Cell Line, Tumor , Cell Survival/drug effects , China , Death Domain Receptor Signaling Adaptor Proteins/drug effects , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Membrane Microdomains , Signal Transduction/drug effects , Stomach Neoplasms/pathology , TNF-Related Apoptosis-Inducing Ligand/drug effects , TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology
19.
BMC Cancer ; 17(1): 492, 2017 Jul 19.
Article in English | MEDLINE | ID: mdl-28724364

ABSTRACT

BACKGROUND: Although the efficacy of tamoxifen (TAM) for breast cancer has been attributed to inducing cell cycle arrest and apoptosis by inhibiting estrogen receptor (ER) signaling, recent evidence indicates that TAM also possesses ER-independent antitumor activity through an unclear mechanism. The present study investigated the anti-tumor mechanism of TAM on mesenchymal triple-negative breast cancer (TNBC). METHODS: The inhibitory effect of TAM on tumor migration and metastasis was analyzed by transwell chamber in vitro and by murine xenograft model in vivo. The promoter sequence of miR-200c was predicted by an online CpG island predictor. Relative expression of miR-200c was measured by quantitative real-time PCR. RESULTS: After treatment with TAM, mesenchymal TNBC cells (MCF-7/ADR and MDA-MB-231) morphologically changed from mesenchymal to epithelial types. Meanwhile, cell migration ability was also significantly decreased in ER-positive breast cancer cells after exposure to TAM. Consistent with these in-vitro results, TAM significantly suppressed lung metastasis rate of mesenchymal TNBC cells in murine xenograft tumors. miRNA array analysis of two types of breast cancer cells showed that miR-200c expression was inhibited in mesenchymal TNBC cells, but increased after TAM treatment due to demethylation of miR-200c promoters. CONCLUSIONS: Our results indicate that TAM inhibits cell migration and enhances chemosensitivity of mesenchymal TNBC cells by reversing their EMT-like property; and that this EMT-reversal effect results from upregulation of miR-200c through demethylating its promoter. To our knowledge, this is the first explanation of a non-ER-related mechanism for the effect of TAM on mesenchymal TNBC cells.


Subject(s)
Epithelial-Mesenchymal Transition/drug effects , MicroRNAs/drug effects , Tamoxifen/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Animals , Antineoplastic Agents, Hormonal , Cell Line, Tumor , Cell Movement/drug effects , DNA Demethylation , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Mice , MicroRNAs/chemistry , MicroRNAs/genetics , MicroRNAs/metabolism , Promoter Regions, Genetic , Signal Transduction , Tamoxifen/therapeutic use , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
20.
Cell Biol Int ; 41(7): 749-760, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28403518

ABSTRACT

Lipid rafts provide a biological platform for apoptosis induced by tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). We previously reported that insulin-like growth factor 1 receptor (IGF1R) translocation into lipid rafts helped to explain TRAIL resistance. However, it was not clear whether TRAIL resistance was caused by the interaction of IGF1R with caveolin-1 (CAV1) and the non-receptor tyrosine kinase SRC in lipid rafts of gastric cancer cells. Here, we observed high IGF1R expression in TRAIL-resistant gastric cancer cells, and showed that IGF1R combined with both CAV1 and SRC in a native complex. TRAIL was shown to promote the formation of the IGF1R/CAV1/SRC tri-complex and the activation of these three molecules. Knockdown of IGF1R or CAV1 or inhibition of SRC activity reduced the formation of this tri-complex and enhanced TRAIL-induced apoptosis. Furthermore, the overexpression of microRNA-194 reversed TRAIL resistance by reducing IGF1R expression. In summary, TRAIL increased formation of the IGF1R/CAV1/SRC tri-complex and the activation of downstream survival pathways, leading to TRAIL resistance in gastric cancer cells.


Subject(s)
Apoptosis/drug effects , Caveolin 1/metabolism , Receptors, Somatomedin/metabolism , Stomach Neoplasms/drug therapy , TNF-Related Apoptosis-Inducing Ligand/pharmacology , src-Family Kinases/metabolism , Cell Line, Tumor , Humans , Membrane Microdomains/metabolism , MicroRNAs/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Receptor, IGF Type 1/metabolism , Signal Transduction/drug effects , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , TNF-Related Apoptosis-Inducing Ligand/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL