Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Transl Oncol ; 46: 102017, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38852277

ABSTRACT

BACKGROUND: Neoadjuvant chemotherapy (NACT) is routinely used to treat patients with advanced gastric cancer (AGC). However, the identification of reliable markers to determine which AGC patients would benefit from NACT remains challenging. METHODS: A systematic screening of plasma proteins between NACT-sensitive and NACT-resistant AGC patients was performed by a mass spectrometer (n = 6). The effect of the most differential plasma protein was validated in two independent cohorts with AGC patients undergoing NACT (ELISA cohort: n = 155; Validated cohort: n = 203). The expression of this candidate was examined in a cohort of AGC tissues using immunohistochemistry (n = 34). The mechanism of this candidate on 5-Fluorouracil (5-FU) resistance was explored by cell-biology experiments in vitro and vivo. RESULTS: A series of differential plasma proteins between NACT-sensitive and NACT-resistant AGC patients was identified. Among them, plasma HIST1H2BK was validated as a significant biomarker for predicting NACT response and prognosis. Moreover, HIST1H2BK was over-expression in NACT-resistant tissues compared to NACT-sensitive tissues in AGC. Mechanistically, HIST1H2BK inhibited 5-FU-induced apoptosis by upregulating A2M transcription and then activating LRP/PI3K/Akt pathway, thereby promoting 5-FU resistance in GC cells. Intriguingly, HIST1H2BK-overexpressing 5-FU-resistant GC cells propagated resistance to 5-FU-sensitive GC cells through the secretion of HIST1H2BK. CONCLUSION: This study highlights significant differences in plasma protein profiles between NACT-resistant and NACT-sensitive AGC patients. Plasma HIST1H2BK emerged as an effective biomarker for achieving more accurate NACT in AGC. The mechanism of intracellular and secreted HIST1H2BK on 5-FU resistance provided a novel insight into chemoresistance in AGC.

2.
J Transl Med ; 21(1): 632, 2023 09 17.
Article in English | MEDLINE | ID: mdl-37718450

ABSTRACT

BACKGROUND: Ladinin-1 (LAD1), an anchoring filament protein, has been associated with several cancer types, including cancers of the colon, lungs, and breast. However, it is still unclear how and why LAD1 causes gastric cancer (GC). METHODS: Multiple in vitro and in vivo, functional gains and loss experiments were carried out in the current study to confirm the function of LAD1. Mass spectrometry was used to find the proteins that interact with LAD1. Immunoprecipitation analyses revealed the mechanism of LAD1 involved in promoting aggressiveness. RESULTS: The results revealed that the LAD1 was overexpressed in GC tissues, and participants with increased LAD1 expression exhibited poorer disease-free survival (DFS) and overall survival (OS). Functionally, LAD1 promotes cellular invasion, migration, proliferation, and chemoresistance in vivo and in vitro in the subcutaneous patient-and cell-derived xenograft (PDX and CDX) tumor models. Mechanistically, LAD1 competitively bound to Vimentin, preventing it from interacting with the E3 ubiquitin ligase macrophage erythroblast attacher (MAEA), which led to a reduction in K48-linked ubiquitination of Vimentin and an increase in Vimentin protein levels in GC cells. CONCLUSIONS: In conclusion, the current investigation indicated that LAD1 has been predicted as a possible prognostic biomarker and therapeutic target for GC due to its ability to suppress Vimentin-MAEA interaction.


Subject(s)
Stomach Neoplasms , Humans , Animals , Ubiquitin , Vimentin , Ubiquitination , Breast , Disease Models, Animal
3.
J Transl Med ; 21(1): 670, 2023 09 27.
Article in English | MEDLINE | ID: mdl-37759298

ABSTRACT

BACKGROUND: Gastric cancer (GC) is a fatal cancer with unclear pathogenesis. In this study, we explored the function and potential mechanisms of intercellular adhesion molecule 2 (ICAM2) in the development and advancement of GC. METHODS: Quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting were performed to quantify ICAM2 expression in harvested GC tissues and cultured cell lines. Immunohistochemical analyses were conducted on a GC tissue microarray to quantify ICAM2 expression and explore its implication on the prognosis of GC patients. In vitro experiments were carried out to reveal the biological functions of ICAM2 in GC cell lines. Further, in vivo experiments were conducted using xenograft models to assess the impact of ICAM2 on GC development and metastasis. Western blot, immunofluorescence, immunoprecipitation, luciferase assay, chromatin immunoprecipitation, and ubiquitination analysis were employed to investigate the underlying mechanisms. RESULTS: ICAM2 expression was downregulated in GC, positively correlating with advanced T stage, distant metastasis, advanced clinical stage, vessel invasion, and shorter patient survival time. ICAM2 overexpression suppressed the proliferation, migration, invasion, metastasis of GC cells as well as their ability to form tumors, whereas ICAM2 knockdown yielded opposite results. Erythroblast transformation-specific-related gene (ERG) as a transcription factor promoted the transcription of ICAM2 by binding to the crucial response element localized within its promoter region. Further analysis revealed that ICAM2 reduced radixin (RDX) protein stability and expression. In these cells, ICAM2 bound to the RDX protein to promote the ubiquitination and degradation of RDX via NEDD4 Like E3 Ubiquitin Protein Ligase (NEDD4L), and this post-translational modification resulted in the inhibition of GC. CONCLUSIONS: In summary, this study demonstrates that ICAM2, which is induced by ERG, suppresses GC progression by enhancing the ubiquitination and degradation of RDX in a NEDD4L-dependent manner. Therefore, these results suggest that ICAM2 is a potential prognostic marker and a therapeutic target for GC.


Subject(s)
Stomach Neoplasms , Humans , Stomach Neoplasms/genetics , Prospective Studies , Ubiquitination , Cell Adhesion Molecules , Transcriptional Regulator ERG
4.
Cell Signal ; 111: 110866, 2023 11.
Article in English | MEDLINE | ID: mdl-37619822

ABSTRACT

BACKGROUND: While ADAMTS12 (A disintegrin and metalloproteinase with thrombospondin motifs 12) has been established as an important regulator of gastrointestinal tumor development and angiogenic activity, the precise mechanistic functions of ADAMTS12 have yet to be fully clarified in gastric cancer (GC). Accordingly, this study was developed to explore the molecular functions of ADAMTS12 in GC and to examine its utility as a biomarker associated with chemoresistance and prognostic outcomes in this cancer type. METHODS: The ability of ADAMTS12 to modulate the proliferative, migratory, invasive, chemoresistant, and tube formation activity of tumor cells was assessed in vivo and in vitro through gain- and loss-of-function approaches. Correlations between ADAMTS12, CD31, and VEGF expression levels in GC patient tumor tissue samples from individuals that did and did not undergo neoadjuvant chemotherapy (NAC) treatment were analyzed via immunohistochemical staining. RESULTS: These analyses revealed the ability of ADAMTS12 to promote in vivo and in vitro cellular proliferative and angiogenic activity, promoting the activation of ERK and the consequent upregulation of VEGF, thereby inducing angiogenesis and decreasing GC cell oxaliplatin sensitivity. A positive correlation between ADAMTS12 levels and both the expression of VEGF as well as the density of microvessels was observed in GC patient tumor tissues. Moreover, those GC patients exhibiting higher intratumoral ADAMTS12 expression exhibited worse responses to NAC treatment and worse overall survival outcomes. CONCLUSIONS: These findings suggest that ADAMTS12 can modulate signaling via the MAPK/VEGF axis in GC cells to enhance tumor cell resistance to oxaliplatin treatment under hypoxic and normoxic conditions. Elevated ADAMTS12 levels can additionally predict vascular abnormalities, worse survival outcomes, and chemoresistance in patients with GC.


Subject(s)
Stomach Neoplasms , Humans , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , Oxaliplatin/pharmacology , Oxaliplatin/therapeutic use , Vascular Endothelial Growth Factor A/metabolism , Up-Regulation , Drug Resistance, Neoplasm , Cell Line, Tumor , ADAMTS Proteins/metabolism
5.
Int J Mol Sci ; 23(19)2022 Sep 20.
Article in English | MEDLINE | ID: mdl-36232317

ABSTRACT

A disintegrin and metalloproteinase with thrombospondin motifs 16 (ADAMTS16) has been reported to be involved in the pathogenesis of solid cancers. However, its role in gastric cancer (GC) is unclear. In this study, the role of ADAMTS16 in gastric cancer was investigated. The effects of ADAMTS16 on cell migration, invasion, and proliferation were investigated by functional experiments in vivo and in vitro. Downstream signal pathways of ADAMTS16 were confirmed by using bioinformatics analysis, co-immunoprecipitation, and immunofluorescence. Meanwhile, bioinformatics analysis, qRT-PCR, western blot, and dual-luciferase reporter gene analysis assays were used to identify ADAMTS16 targets. The expression of ADAMTS16 in GC was analyzed in public datasets. The expression of ADAMTS16 and its correlations with the clinical characteristics of GC were investigated by immunohistochemistry. Ectopic ADAMTS16 expression significantly promoted tumor cell migration, invasion, and growth. Bioinformatics analysis and western blot showed that ADAMTS16 upregulated the IFI27 protein through the NF-κb pathway, which was confirmed by immunofluorescence and western blot. Dual-luciferase reporter gene analysis identified a binding site between P65 and IFI27 that may be directly involved in the transcriptional regulation of IFI27. IFI27 knockdown reversed the promoting effect of ADAMTS16 on cell invasion, migration, and proliferation indicating that ADAMTS16 acts on GC cells by targeting the NF-κb/IFI27 axis. ADAMTS16 was associated with poor prognosis in clinical characteristics. ADAMTS16 promotes cell migration, invasion, and proliferation by targeting IFI27 through the NF-κB pathway and is a potential progressive and survival biomarker of GC.


Subject(s)
MicroRNAs , Stomach Neoplasms , ADAMTS Proteins/genetics , ADAMTS Proteins/metabolism , Carcinogenesis/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Disintegrins , Gene Expression Regulation, Neoplastic , Humans , Membrane Proteins/metabolism , MicroRNAs/genetics , NF-kappa B/genetics , NF-kappa B/metabolism , Signal Transduction , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Thrombospondins/metabolism
6.
Gastric Cancer ; 25(6): 1002-1016, 2022 11.
Article in English | MEDLINE | ID: mdl-35925524

ABSTRACT

BACKGROUND: A disintegrin and metalloproteinase with thrombospondin motifs 10 (ADAMTS10) plays a role in extracellular matrix and correlates with Weill-Marchesani syndrome. However, its role in gastric cancer remains unknown. Thus, we started this research to unveil the role of ADAMTS10 in gastric cancer (GC). METHODS: The expression of ADAMTS10 in GC was analyzed by immunohistochemical staining and quantitative RT-PCR (qRT-PCR). The effects of ADAMTS10 inhibiting GC cell progression were conducted by functional experiments in vitro and in vivo. Flow cytometry was used to discover changing of cell cycle, apoptosis and ROS by ADAMTS10 in GC cell. Western blot was applied to identify targets of ADAMTS10. Western blot, qRT-PCR and flow cytometry were applied to discover the effect of ADAMT10 on THP1. RESULTS: ADAMTS10 expression was downregulated in GC tissue and patients with low ADAMTS10 levels had poorer overall survival. ADAMTS10 overexpression altered cell cycle, promoted apoptosis, and inhibited proliferation, migration, and invasion in vitro and in vivo. ADAMTS10 regulated TXNIP and ROS through the JAK/STAT/c-MYC pathway. Decreasing TXNIP and ROS reversed the inhibitory effect of ADAMTS10 on cell migration and invasion in vitro. ADAMTS10 secreted by GC cells was absorbed by THP1 and regulated TXNIP and ROS in THP1. ADAMTS10 secreted by GC cells inhibited macrophage M2 polarization. CONCLUSIONS: These results suggest that ADAMTS10 targets TXNIP and ROS via the JAK/STAT/c-MYC pathway and that may play important roles in GC progression and macrophage polarization which indicates that ADAMTS10 can be a potential survival marker for gastric cancer.


Subject(s)
Stomach Neoplasms , Humans , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disintegrins/metabolism , Gene Expression Regulation, Neoplastic , Macrophages/pathology , Reactive Oxygen Species/metabolism , Stomach Neoplasms/pathology , Thrombospondins/metabolism , Tumor Microenvironment , Proto-Oncogene Proteins c-myc
7.
Cancer Cell Int ; 20: 427, 2020.
Article in English | MEDLINE | ID: mdl-32884449

ABSTRACT

BACKGROUND: Circular RNAs (circRNAs) are a novel class of noncoding RNAs. Increasing evidence indicates that circRNAs play an important role in the occurrence and development of tumors. However, the role of circRNA hsa_circ_0044556 in the progression of colorectal cancer (CRC) remains unclear. METHODS: First, we searched for differentially expressed circRNAs using a circRNA microarray in paired CRC and adjacent normal tissues. The circRNA hsa_circ_0044556 was screened out from the existing CRC circRNA microarray in the Gene Expression Omnibus database and our microarray. The clinical significance of hsa_circ_0044556 expression level in CRC patients was then investigated. Finally, the functions of the targets of this circRNA were determined in CRC cell lines. RESULTS: Hsa_circ_0044556 was highly expressed in CRC patients and was positively correlated with tumor stage and lymph node metastasis. In CRC cell lines, the proliferation, migration, and invasion of cancer cells were inhibited by knocking down hsa_circ_0044556 expression. CONCLUSION: Hsa_circ_0044556 promoted the progression of CRC. It is possible that hsa_circ_0044556 will become a novel biomarker or therapeutic target for CRC.

8.
J BUON ; 24(4): 1470-1475, 2019.
Article in English | MEDLINE | ID: mdl-31646793

ABSTRACT

PURPOSE: Colon cancer (CC) is one of the deadly malignancies and the second most frequent cancer in the world. The development of drug resistance and dearth of the viable drug options form a serious obstacle in the treatment of CC. Herein, the anticancer potential of Zingiberene was examined against the CC cells. METHODS: The proliferation rate of the CC cells was assessed by cell counting assay. Autophagy was detected by transmission electron microscopy (TEM). The transfected cells were then treated with varied concentrations of Zingiberene (0, 10, 20 and 40 µM) for 24 h and monitored by fluorescent microscopy. Cell cycle analysis was performed by flow cytometry. Protein expression was determined by immunoblotting. RESULTS: Zingiberene could considerably inhibit the proliferation of CC cells. The anticancer activity of Zingiberene against the HT-29 CC cells was found to be due to induction of autophagy. The Zingiberene triggered autophagy was also linked with increase in the expression of LC3-II and decrease in p62 expression. However, no apparent effects were observed on the LC3-I expression. Moreover, it was found that zingiberine also caused activation of autophagy-related caspases in the HT-29 cells. Further, it was found that Zingiberene could inhibit the mTOR/PI3K/AKT signalling pathway in the CC cells. Zingiberene also suppressed the weight and volume of the xenografted tumors concentration-dependently. CONCLUSIONS: These results indicate that Zingiberene may inhibit the growth of CC in vitro and in vivo and may be used for the development of systemic therapy against CC.


Subject(s)
Autophagy/drug effects , Cell Proliferation/drug effects , Colonic Neoplasms/drug therapy , Monocyclic Sesquiterpenes/pharmacology , Animals , Apoptosis/drug effects , Caspase 2/genetics , Cell Line, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Cysteine Endopeptidases/genetics , Heterografts , Humans , Mice , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/genetics
9.
Mol Biol Rep ; 41(7): 4601-11, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24740148

ABSTRACT

Mannose receptor (MR) plays a significant role in innate immune responses to pathogens in vertebrates. Here we characterized the first teleost MR from Megalobrama amblycephala, named maMR and its expression patterns were investigated. The full-length maMR consists of 5,295 bp encoding a putative protein of 1,433 amino acids. The predicted amino acid sequences showed that maMR contained a signal peptide, a cysteine-rich domain, a single fibronectin type II domain, eight tandemly arranged C-type lectin-like domains, a transmembrane domain and a C-terminal cytoplasmic domain. Phylogenetic analysis revealed the highest similarity of maMR with Danio rerio MR predicted by computational analysis. The maMR-mRNAs were ubiquitously transcribed in different tissues, However the highest transcripts were observed in head kidney. Transcripts of maMR significantly increased at the late stages of embryo and continued to be at the high levels after hatching. The maMR transcripts were significantly increased in M. amblycephala after stimulation with killed Aeromonas hydrophila.


Subject(s)
Cloning, Molecular/methods , Cyprinidae/genetics , Gene Expression , Lectins, C-Type/genetics , Mannose-Binding Lectins/genetics , Receptors, Cell Surface/genetics , Aeromonas hydrophila/physiology , Amino Acid Sequence , Animals , Base Sequence , Chickens , Cyprinidae/immunology , Cyprinidae/microbiology , Fish Diseases/immunology , Fish Diseases/microbiology , Humans , Lectins, C-Type/chemistry , Lectins, C-Type/immunology , Mannose Receptor , Mannose-Binding Lectins/chemistry , Mannose-Binding Lectins/immunology , Mice , Molecular Sequence Data , Protein Structure, Tertiary , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL
...