Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Hum Genet ; 143(3): 343-355, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38480539

ABSTRACT

Colorectal cancer (CRC) is the third most prevalent diagnosed cancer in men and second most prevalent cancer in women. H3K27ac alterations are more commonly than gene mutations in colorectal cancer. Most colorectal cancer genes have significant H3K27ac changes, which leads to an over-expression disorder in gene transcription. Over-expression of STEAP3 is involved in a variety of tumors, participating in the regulation of cancer cell proliferation and migration. The purpose of this work is to investigate the role of STEAP3 in the regulation of histone modification (H3K27ac) expression in colon cancer. Bioinformatic ChIP-seq, ChIP-qPCR and ATAC-seq were used to analyze the histone modification properties and gene accessibility of STEAP3. Western blot and qRT-PCR were used to evaluate relative protein and gene expression, respectively. CRISPR/Cas9 technology was used to knockout STEAP3 on colon cancer cells to analyze the effect of ATF3 on STEAP3. STEAP3 was over-expressed in colon cancer and associated with higher metastases and more invasive and worse stage of colon cancer. ChIP-seq and ChIP-qPCR analyses revealed significant enrichment of H3K27ac in the STEAP3 gene. In addition, knocking down STEAP3 significantly inhibits colon cancer cell proliferation and migration and down-regulates H3K27ac expression. ChIP-seq found that ATF3 is enriched in the STEAP3 gene and CRISPR/Cas9 technology used for the deletion of the ATF3 binding site suppresses the expression of STEAP3. Over-expression of STEAP3 promotes colon cancer cell proliferation and migration. Mechanical studies have indicated that H3K27ac and ATF3 are significantly enriched in the STEAP3 gene and regulate the over-expression of STEAP3.


Subject(s)
Cell Movement , Cell Proliferation , Colonic Neoplasms , Gene Expression Regulation, Neoplastic , Histones , Humans , Cell Proliferation/genetics , Cell Movement/genetics , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Colonic Neoplasms/metabolism , Histones/metabolism , Histones/genetics , Acetylation , Female , Cell Line, Tumor , Male , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism
2.
Macromol Rapid Commun ; 44(13): e2300104, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37082932

ABSTRACT

Recent advancements in aggregation-induced emission (AIE) macromolecular materials have brought their attention as potential antibacterial solutions, these materials offer new approaches to cure multidrug-resistant infections and biofilms in bacterial infections as well as real-time monitoring and specific targeting of bacteria. This review provides an overview of the three main categories of AIE macromolecular materials with antibacterial properties; namely AIE-active polymers, AIEgen@polymer complexes, and clusterization-triggered emission (CTE) based polymers. The mechanisms and applications of these materials in antibacterial treatment, wound care, and protective equipment are also discussed. The potential for future developments and application directions of AIE-based antimicrobial materials are finally highlighted.


Subject(s)
Anti-Bacterial Agents , Polymers , Macromolecular Substances/pharmacology , Polymers/pharmacology , Anti-Bacterial Agents/pharmacology , Bacteria
3.
J Cell Mol Med ; 26(24): 5984-5997, 2022 12.
Article in English | MEDLINE | ID: mdl-36420658

ABSTRACT

According to our prior findings, ARID1A expression is decreased in colon cancer, which has a poor prognosis. In this study, we investigated the ARID1A-VIM/CDH1 signalling axis's role in colon cancer proliferation and migration. The differentially expressed genes in cells that might be controlled by ARID1A were discovered by a database screening for ARID1A knockout. qPCR was used to analyse ARID1A and EMT markers expression levels in colon cancer. We utilized siRNA RID1A to explore the influence of ARID1A silencing on EMT in CRC cells. The function of ARID1A in the colon was investigated utilizing the wound healing, transwell and CCK-8 WST- assays. The molecular mechanism by which ARID1A regulates VIM and CDH1 was elucidated using chip-qPCR. Numerous genes involved in EMT were dysregulated in the absence of ARID1A. VIM expression increased in cells lacking ARID1A expression and vice versa. Many COAD samples with high ARID1A mRNA expression had low VIM mRNA expression, despite the relevance. CDH1 gene was positively correlated with ARID1A. Moreover, siRNA-ARID1A-transfected cells accelerated cell migration and invasion and increased cell proliferation rate in vitro. Chip-qPCR analysis showed that ARID1A binds to the promoters of both genes and changes their expression in colon cancer. ARID1A inactivation is associated with VIM activation and CDH1 suppression, which might serve as crucial molecules influencing COAD prognosis, accelerate tumour progression, and shorten patients' survival time, and promote metastases of COAD. Thus, depletion of ARID1A can be therapeutically exploited by targeting downstream effects to improve cancer treatment-related outcomes.


Subject(s)
Colonic Neoplasms , Epithelial-Mesenchymal Transition , Humans , Down-Regulation/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Cell Line, Tumor , Cell Proliferation/genetics , Cell Movement/genetics , Colonic Neoplasms/genetics , RNA, Small Interfering/genetics , RNA, Messenger , DNA-Binding Proteins/genetics , Transcription Factors/genetics , Antigens, CD/genetics , Cadherins/genetics
4.
Cancer Cell Int ; 20: 62, 2020.
Article in English | MEDLINE | ID: mdl-32123520

ABSTRACT

BACKGROUND AND AIMS: microRNAs (miRNAs) have been reported to regulate proliferation and migration by down-regulating the expression of target genes. The aims of this study were to investigate whether miR-4316 inhibited proliferation and migration by downregulating vascular endothelial growth factor A (VEGF-A) and its clinical significance in gastric cancer (GC). METHODS: The clinical tissues of the GC patients for miR-4316 and VEGF-A were detected by qRT-PCR. The protein levels of VEGF-A and c-Met were determined by western blotting. Cell Proliferation, migration, and colony forming assays were conducted to show whether miR-4316 affects proliferation by CCK-8, migration by transwell, wound healing and colony formation assays. The bioinformatic methods and luciferase reporter assay were applied to detect the relationship between miRNA and VEGF-A on its targeting 3-untranslated regions (3-UTRs). CCK-8, colony formation, wound healing, and transwell assay were performed to explore the function of miR-4316. RESULTS: The results of qRT-PCR indicated that miR-4316 expression level was significantly downregulated in human GC tissues and GC cell lines compared with their control. miR-4316 inhibited proliferation, migration and colony formation in GC cell lines by reducing VEGF-A. And western blot results indicated that miR-4316 significantly inhibited GC through repressing VEGF-A and c-Met. The investigation of Luciferase assay indicated that VEGF-A is a direct target gene of miR-4316. CONCLUSIONS: miR-4316 suppressed proliferation and migration of GC through the VEGF-A gene. MiR-4316 acts as a tumor suppressor by targeting VEGF-A and this indicated that MiR-4316 might be a potential therapeutic target for GC.

5.
Mol Cancer ; 16(1): 78, 2017 04 13.
Article in English | MEDLINE | ID: mdl-28403883

ABSTRACT

BACKGROUND: Profiling evidences of selectin demonstrate that they play an crucial role in cancer progression and metastasis. However, DC-SIGNR as a family member of selectin participates in gastric cancer liver metastasis remains unknown. METHODS: The serum level of DC-SIGNR was evaluated in gastric cancer patients by ELISA. Manipulation DC-SIGNR expression in BGC823 and SGC7901 cell lines was mediated by lentivirus. Investigation the biological effects of DC-SIGNR were verified by MTT, wounding and transwell in vitro and experiments on animals to confirm gastric cancer liver metastasis by IVIS. Insights of the mechanism were employed microarray and bioinformatic analysis. Further to confirm the results were conducted by qRT-PCR, western blot and by flow cytometry. RESULTS: DC-SIGNR serum level was significantly increased in gastric cancer patients compared with healthy group. Additionally, DC-SIGNR level was associated with an advanced pathological stage in gastric cancer patients. DC-SIGNR knockdown inhibited the proliferation, migration and invasion of gastric cancer cells in vitro and suppressed the liver metastasis in vivo. While, DC-SIGNR overexpression promoted cell proliferation, migration and invasion. In mechanism, HNRNPKP2 as a lncRNA was upregulated after DC-SIGNR knockdown. Importantly, STAT5A promoted HNRNPKP2 expression after knockdown DC-SIGNR. Furthermore after HNRNPKP2 depletion, the downstream target gene CXCR4 was downregulated. CONCLUSIONS: DC-SIGNR promoted gastric cancer liver metastasis mediated with HNRNPKP2 which expression was regulated by STAT5A. And HNRNPKP2 decreased the expression of downstream target gene CXCR4. These findings indicated potential therapeutic candidates for gastric cancer liver metastasis.


Subject(s)
Cell Adhesion Molecules/genetics , Gene Expression Regulation, Neoplastic , Lectins, C-Type/genetics , Liver Neoplasms/secondary , Receptors, CXCR4/genetics , Receptors, Cell Surface/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Aged , Aged, 80 and over , Animals , Biomarkers, Tumor , Cell Adhesion Molecules/blood , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cluster Analysis , Disease Models, Animal , Female , Gene Expression Profiling , Gene Knockdown Techniques , Heterografts , Humans , Lectins, C-Type/blood , Lectins, C-Type/metabolism , Male , Mice , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , RNA, Long Noncoding/genetics , Receptors, CXCR4/metabolism , Receptors, Cell Surface/blood , Receptors, Cell Surface/metabolism , STAT5 Transcription Factor/metabolism , Stomach Neoplasms/metabolism
6.
Mol Cell Biochem ; 407(1-2): 151-60, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26150177

ABSTRACT

Dendritic cell-specific intercellular adhesion molecule-grabbing nonintegrin-related protein (DC-SIGNR) is a type II transmembrane protein which has been reported to bind a variety of pathogens as well as participate in immunoregulation. But the association between the level of DC-SIGNR and lung cancer is unknown. To investigate the clinical diagnostic significance of DC-SIGNR in lung cancer, we investigated serum DC-SIGNR levels in 173 lung cancer patients and 134 healthy individuals using enzyme-linked immunosorbent assay (ELISA). Results showed that serum DC-SIGNR levels in lung cancer patients were lower than that in healthy controls (P = 0.0003). A cut-off value of 3.8998 ng/L for DC-SIGNR predicted the presence of lung cancer with 78.03% sensitivity and 49.25% specificity (area under the curve = 0.6212, P = 0.0003). Strikingly, serum DC-SIGNR levels were significantly higher in lung cancer patients with brain metastasis compared to those without metastasis (P = 0.0283). Moreover, the serum concentrations of DC-SIGNR in lung cancer patients also correlated significantly with serum natural killer cells percentage (P = 0.0017). In addition, immunohistochemistry assay demonstrated that the expression of DC-SIGNR in lung tissues of 31 lung cancer patients and 13 tuberculosis patients was significantly lower than that in 18 normal lung tissues (P = 0.0418, 0.0289), and there is no significant difference between tuberculosis tissues and lung cancer tissues (P = 0.2696). These results suggest that DC-SIGNR maybe a promising biological molecule that has the potential for clinical research of lung cancer, whereas its underlying roles are needed to be investigated in further studies.


Subject(s)
Brain Neoplasms/secondary , Cell Adhesion Molecules/blood , Down-Regulation , Killer Cells, Natural/metabolism , Lectins, C-Type/blood , Lung Neoplasms/blood , Receptors, Cell Surface/blood , Adult , Aged , Aged, 80 and over , Brain Neoplasms/blood , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/pathology , Male , Middle Aged
7.
Gut ; 62(8): 1169-78, 2013 Aug.
Article in English | MEDLINE | ID: mdl-22637699

ABSTRACT

OBJECTIVE: Adhesion molecules play an important role in tumour metastasis. The liver is a frequent target for the metastasis of several tumour types. However, virtually no liver-specific adhesion molecules have been described in terms of organ-specific metastasis. This study aimed to determine the role of liver sinusoidal endothelial cell lectin (LSECtin) in colon carcinoma metastasis to the liver. DESIGN: The role of LSECtin in colon carcinoma metastasis to the liver was determined by LSECtin knockout nude mice and anti-LSECtin antibody. LSECtin promoting the migration of LS174T and LoVo cells was determined by transwell experiment. The serum levels of soluble LSECtin in patients were elevated by ELISA. RESULTS: LSECtin was found to adhere to LS174T and LoVo colon cancer cells in vitro and in vivo. Deficiency or blocking of LSECtin significantly decreased hepatic metastases of LS174T and LoVo cells. Primary colon cancer cells from patients also exhibited remarkably low rates of hepatic metastasis in LSECtin knockout mice. LSECtin promoted the migration of LS174T and LoVo cells and increased the expression of c-Met in these cells. Serum soluble LSECtin was detected at significantly higher levels in colon cancer patients with or without hepatic metastases compared with healthy controls and was also increased in colon cancer patients with metastases compared with those without metastases. CONCLUSION: The results indicate that LSECtin plays an important role in colorectal carcinoma liver metastasis and may be a promising new target for intervention in metastasis formation.


Subject(s)
Colonic Neoplasms/metabolism , Lectins, C-Type/physiology , Liver Neoplasms/secondary , Receptors, Virus/physiology , Adult , Aged , Animals , Cell Adhesion/physiology , Cell Movement/physiology , Colonic Neoplasms/blood , Colonic Neoplasms/pathology , Female , Humans , Lectins, C-Type/blood , Lectins, C-Type/deficiency , Liver/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Middle Aged , Neoplasm Proteins/blood , Neoplasm Proteins/metabolism , Neoplasm Proteins/physiology , Neoplasm Transplantation , Proto-Oncogene Proteins c-met/metabolism , Receptors, Virus/deficiency , Recombinant Proteins/metabolism , Transplantation, Heterologous
8.
Adv Mater ; 36(14): e2311384, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38178607

ABSTRACT

Achieving efficient near-infrared room-temperature phosphorescence of purely organic phosphors remains scarce and challenging due to strong nonradiative decay. Additionally, the investigation of triplet excimer phosphorescence is rarely reported, despite the fact that excimer, a special emitter commonly formed in crystals with strong π-π interactions, can efficiently change the fluorescent properties of compounds. Herein, a series of dithienopyrrole derivatives with low triplet energy levels and stable triplet states, exhibiting persistent near-infrared room-temperature phosphorescence, is developed. Via the modification of halogen atoms, the crystals display tunable emissions of monomers from 645 to 702 nm, with a maximum lifetime of 3.68 ms under ambient conditions. Notably, excimer phosphorescence can be switched on at low temperatures, enabled by noncovalent interactions rigidifying the matrix and stabilizing triplet excimer. Unprecedentedly, the dynamic transition process is captured between the monomer and excimer phosphorescence with temperature variations, revealing that the unstable triplet excimers in crystals with a tendency to dissociate can result in the effective quench of room-temperature phosphorescence. Excited state transitions across varying environments are elucidated, interpreting the structural dynamics of the triplet excimer and demonstrating strategies for devising novel near-infrared phosphors.

9.
Biochem Cell Biol ; 91(4): 214-20, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23859015

ABSTRACT

Dendritic cell-specific intercellular adhesion molecule-grabbing nonintegrin-related protein (DC-SIGNR), a type II integral membrane protein and a member of the C-type lectins, has been reported to bind various strains of HIV-1, HIV-2, and simian immunodeficiency virus. Serum DC-SIGNR is not currently available for the detection of non-Hodgkin lymphoma (NHL). Using an enzyme-linked immunosorbent assay (ELISA), we assessed the serum levels of DC-SIGNR in 70 cancer patients and 100 healthy controls. Additionally, using immunohistochemistry, we determined the expression of DC-SIGNR in the lymph nodes. Using the ELISA, low serum levels of DC-SIGNR were detected in the patients (median, 4.513 ng·L(-1); range, 1.066-9.232 ng·L(-1); p = 0.0003). Serum concentrations of DC-SIGNR correlated significantly with age (p = 0.0077) and lactic acid dehydrogenase (p = 0.0046) and ß2-microglobulin (p = 0.0491) levels. However, we found no statistically significant correlation between serum DC-SIGNR levels and clinical data such as sex, Ann Arbor stage, B symptoms, and histologic subtypes. Moreover, NHL patients with a lower level of serum DC-SIGNR expression in lymphatic endothelial cells also showed negative immunostaining levels. These results suggest that DC-SIGNR is a biological molecule that may be potentially useful in NHL clinical settings.


Subject(s)
Carboxylic Ester Hydrolases/metabolism , Cell Adhesion Molecules/metabolism , Dendritic Cells/cytology , Integrins/metabolism , Lectins, C-Type/metabolism , Lymphoma, Non-Hodgkin/metabolism , Receptors, Cell Surface/metabolism , beta 2-Microglobulin/metabolism , Adult , Aged , Aged, 80 and over , Cell Adhesion , Dendritic Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Lymph Nodes/metabolism , Male , Middle Aged , Young Adult
10.
J Cancer Res Clin Oncol ; 149(17): 15697-15712, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37670166

ABSTRACT

BACKGROUND: As a phosphorylated protein, NOLC1 is mainly located in the nucleus and is highly expressed in a variety of tumors, participating in the regulation of cell proliferation and aging. This study further investigated the role of NOLC1 in colorectal cancer tumors, aiming to provide sufficient scientific evidence for the clinical treatment of colorectal cancer. METHODS: We used TCGA, GEO, TNMplot, GEPIA, and other databases to explore the expression level of NOLC1 in colorectal cancer patients, as well as the correlation between the clinical characteristics of colorectal cancer patients and their expression, and conducted the prognostic analysis. Immunohistofluorescence (IHF) staining verified the analytical results. Subsequently, KEGG and GO enrichment analysis was used to identify the potential molecular mechanism of NOLC1 promoting the occurrence and development of colorectal cancer. The influence of NOLC1 expression on the immune microenvironment of colorectal cancer patients was further investigated using the TIMER database. GDSC database analysis was used to screen out possible anti-colorectal cancer drugs against NOLC1. Finally, we demonstrated the effect of NOLC1 on the activity and migration of colorectal cancer cells by Edu Cell proliferation assay and Wound Healing assay in vitro. RESULTS: Our results suggest that NOLC1 is overexpressed in colorectal cancer, and that overexpression of NOLC1 is associated with relevant clinical features. NOLC1, as an independent risk factor affecting the prognosis of colorectal cancer patients, can lead to a poor prognosis of colorectal cancer. In addition, NOLC1 may be associated with MCM10, HELLS, NOC3L, and other genes through participating in Wnt signaling pathways and jointly regulate the occurrence and development of colorectal cancer under the influence of the tumor microenvironment and many other influencing factors. Related to NOLC1: Selumetinib, Imatinib, and targeted drugs such as Lapatinib have potential value in the clinical application of colorectal cancer. NOLC1 enhances the proliferation and migration of colorectal cancer cells. CONCLUSIONS: High expression of NOLC1 as an independent prognostic factor for survival in patients with colorectal cancer. NOLC1 enhances the proliferation and migration of colorectal cancer cells. Further studies and clinical trials are needed to confirm the role of NOLC1 in the development and progression of colorectal cancer.


Subject(s)
Aging , Colorectal Neoplasms , Humans , Prognosis , Cell Proliferation , Colorectal Neoplasms/genetics , Databases, Factual , Tumor Microenvironment , Nuclear Proteins , Phosphoproteins
11.
Adv Sci (Weinh) ; 10(33): e2205229, 2023 11.
Article in English | MEDLINE | ID: mdl-37870214

ABSTRACT

Liver metastasis is a common cause of death in progressive colorectal cancer patients, but the molecular mechanisms remain unclear. Here, it is reported that a conserved and oxidative pentose phosphate pathway-associated circular RNA, circNOLC1, plays a crucial role in colorectal cancer liver metastasis. It is found that circNOLC1 silencing reduces the oxidative pentose phosphate pathway-related intermediate metabolites and elevates NADP+ /NADPH ratio and intracellular ROS levels, thereby attenuating colorectal cancer cell proliferation, migration, and liver metastasis. circNOLC1 interacting with AZGP1 to activate mTOR/SREBP1 signaling, or sponging miR-212-5p to upregulate c-Met expression, both of which can further induce G6PD to activate oxidative pentose phosphate pathway in colorectal cancer liver metastasis. Moreover, circNOLC1 is regulated by the transcription factor YY1 and specifically stabilized HuR induces its parental gene mRNA expression. The associations between circNOLC1 and these signaling molecules are validated in primary CRC and corresponding liver metastasis tissues. These findings reveal that circNOLC1 interacting with AZGP1 and circNOLC1/miR-212-5p/c-Met axis plays a key role in oxidative pentose phosphate pathway-mediated colorectal cancer liver metastasis, which may provide a novel target for precision medicine of colorectal cancer.


Subject(s)
Colorectal Neoplasms , Liver Neoplasms , MicroRNAs , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Colorectal Neoplasms/pathology , Pentose Phosphate Pathway , Liver Neoplasms/metabolism , Oxidative Stress , Adipokines/metabolism
12.
Hematol Oncol ; 30(2): 70-5, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21812012

ABSTRACT

The P2X7 receptor (P2X7R) is a nucleotide receptor expressed predominantly on hemopoietic, bone, and epithelial cells. The P2X7R can be activated by extracellular ATP and induces the influx of calcium, releases cytokines, and participates in cell proliferation and apoptosis. CD44 is an adhesion molecule. The effects of CD44 include cell-cell and cell-matrix adhesion interactions, lymphocyte activation, and cell migration. Many studies have shown that P2X7R and CD44 play important roles in hematological malignancies, but no study exists regarding the relationship between P2X7R and CD44. In the present study, we characterized P388D1 cells for the surface expression of CD44 and analyzed ATP-induced shedding. The data showed that P388D1 cells express CD44. Incubation of P388D1 cells with ATP induced a rapid loss of CD44 from the P388D1 cell surface. In addition, using a receptor inhibitor and P2X7R short hairpin RNA, we showed that the loss of CD44 is mediated via the P2X7R. Finally, we demonstrated that activation of P2X7R by ATP induces CD44 shedding.


Subject(s)
Adenosine Triphosphate/physiology , Hyaluronan Receptors/physiology , Leukemia P388/metabolism , Receptors, Purinergic P2X7/physiology , Animals , Cell Line, Tumor , Extracellular Space/metabolism , Hyaluronan Receptors/analysis , Mice
13.
Hematol Oncol ; 30(2): 62-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21812013

ABSTRACT

Apoptosis-associated speck-like protein (ASC) is a bipartite adaptor molecule that participates in inflammation and apoptosis. ASC silencing has been observed in a significant proportion of human cancers. Here, we examined the role of ASC overexpression in the metastasis of the P388D1 murine lymphoma cell line to the liver, lung, spleen and kidney. First, we determined that the P388D1 cells express ASC. Then, ASC overexpression in P388D1 was achieved by transfecting pEGFP-ASC-C2 into the P388D1 cells. Furthermore, after the ASC-overexpressing P388D1 cells were injected into DBA/2 mice through the vena caudalis, their metastasis to the lung and the liver was significantly reduced in the pEGFP-ASC-C2-transfected group. These data indicate that ASC overexpression affects the in vivo metastatic properties of P388D1 cells.


Subject(s)
Cytoskeletal Proteins/physiology , Leukemia P388/pathology , Animals , Apoptosis Regulatory Proteins , CARD Signaling Adaptor Proteins , Cell Line, Tumor , Cytoskeletal Proteins/analysis , Leukemia P388/metabolism , Mice , Mice, Inbred DBA , Neoplasm Metastasis , Transfection
14.
ACS Bio Med Chem Au ; 2(3): 236-257, 2022 Jun 15.
Article in English | MEDLINE | ID: mdl-37101570

ABSTRACT

Cell death is closely related to various diseases, and monitoring and controlling cell death is a promising strategy to develop efficient therapy. Aggregation-induced emission luminogens (AIEgens) are ideal candidates for developing novel theranostic agents because of their intriguing properties in the aggregate state. The rational application of AIE materials in cell death-related research is still in its infancy but has shown great clinical potential. This review discussed the research frontier and our understanding of AIE materials in various subroutines of cell death, including apoptosis, necrosis, immunogenic cell death, pyroptosis, autophagy, lysosome-dependent cell death, and ferroptosis. We hope that the new insights can be offered to this growing field and attract more researchers to provide valuable contributions.

15.
Cell Death Dis ; 13(7): 593, 2022 07 11.
Article in English | MEDLINE | ID: mdl-35821222

ABSTRACT

Liver and lymph node sinusoidal endothelial cell C-type lectin (LSECtin) plays an important regulatory role in a variety of diseases, including tumors. However, the underlying mechanism of LSECtin in gastric cancer (GC) remains largely unknown. In our research, LSECtin promoted the adhesion and invasion of GC cells, and was involved in lymphatic metastasis of GC cells. Mechanistically, LSECtin promoted the adhesion, proliferation and migration of GC cells by downregulating STAT1 expression. The circular RNA circFBXL4, which is regulated by LSECtin, sponges the microRNA miR-146a-5p to regulate STAT1 expression. The promotion of GC cell proliferation, migration and invasion mediated by LSECtin was largely inhibited by circFBXL4 overexpression or miR-146a-5p silencing. Moreover, in its role as a transcription factor, STAT1 modulated the expression of FN1 and CHD4. In conclusion, LSECtin might be involved in the lymphatic metastasis of GC by upregulating the expression of FN1 and CHD4 via the circFBXL4/miR-146a-5p/STAT1 axis, possibly indicating a newly discovered pathogenic mechanism.


Subject(s)
Lectins, C-Type , MicroRNAs , Stomach Neoplasms , Cell Adhesion/physiology , Cell Movement/physiology , Cell Proliferation/physiology , Humans , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Lymphatic Metastasis , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasm Invasiveness , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology
16.
Front Oncol ; 11: 679334, 2021.
Article in English | MEDLINE | ID: mdl-34414106

ABSTRACT

AT-rich interaction domain 1A (ARID1A) is a tumor suppressor gene that mutates in several cancer types, including breast cancer, ovarian cancer, and colorectal cancer (CRC). In colon adenocarcinoma (COAD), the low expression of ARID1A was reported but the molecular reason is unclear. We noticed that ARID1A low expression was associated with increased levels of miR-185 in the COAD. Therefore, this study aims to explore ncRNA-dependent mechanism that regulates ARID1A expression in COAD regarding miR-185. The expression of ARID1A was tested in COAD cell line under the effect of miR-185 mimics compared with inhibitor. The molecular features associated with loss of ARID1A and its association with tumor prognosis were analyzed using multi-platform data from The Cancer Genome Atlas (TCGA), and gene set enrichment analysis (GSEA) to identify potential signaling pathways associated with ARID1A alterations in colon cancer. Kaplan-Meier survival curve showed that a low level of ARID1A was closely related to low survival rate in patients with COAD. Results showed that inhibiting miR-185 expression in the COAD cell line significantly restored the expression of ARID1A. Further, the increased expression of ARID1A significantly improved the prolonged overall survival of COAD. We noticed that there is a possible relationship between ARID1A high expression and tumor microenvironment infiltrating immune cells. Furthermore, the increase of ARID1A in tumor cells enhanced the response of inflammatory chemokines. In conclusion, this study demonstrates that ARID1A is a direct target of miR-185 in COAD that regulates the immune modulations in the microenvironment of COAD.

17.
Cell Biol Int ; 34(12): 1205-11, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20722629

ABSTRACT

The P2X7R (P2X7 receptor) is an ATP-gated cation channel expressed in normal cells that participates in both cell proliferation and apoptosis. Here, we have confirmed P2X7R expression on murine P388D1 lymphoid neoplasm cells. In addition, ATP-stimulated P2X7R expression was found to trigger increased intracellular calcium flux. Furthermore, silencing with short hairpin RNA and blocking with P2X7R antibody significantly reduced the metastasis of P388D1 cells to lymph nodes. These results indicate that inhibition of the expression and function of P2X7R attenuates the metastatic ability of murine lymphoid neoplasm cell line P388D1, which represents a new potential target for anti-metastatic therapy.


Subject(s)
Lymphoma/pathology , Lymphoma/prevention & control , Purinergic P2X Receptor Antagonists/therapeutic use , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/therapeutic use , Animals , Cell Line, Tumor , Lymph Nodes/drug effects , Lymph Nodes/pathology , Lymphatic Metastasis , Lymphocytes/drug effects , Lymphocytes/pathology , Lymphoma/genetics , Mice , Mice, Inbred DBA , Molecular Targeted Therapy , Neoplasm Transplantation , Purinergic P2X Receptor Antagonists/pharmacology , RNA, Small Interfering/pharmacology , RNA, Small Interfering/therapeutic use , Receptors, Purinergic P2X7/genetics , Receptors, Purinergic P2X7/metabolism , Receptors, Purinergic P2X7/physiology
18.
Oncol Lett ; 19(1): 83-92, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31897118

ABSTRACT

The development of multidrug resistance (MDR) has seriously impeded the efficacy of drug treatment of chronic myeloid leukemia (CML). Recent studies have indicated that S100 calcium-binding protein A8 (S100A8) is associated with the occurrence and development of MDR. Traditional Chinese medicine may provide drugs with the potential to be used as multidrug resistance reversal agents with low toxicity and multi-target characteristics. The present study selected K562/DOX cells, a CML drug-resistant cell line, as a research model, and aimed to examine whether curcumin was able to reverse the resistance to doxorubicin (DOX), and elucidate the underlying molecular mechanisms. An MTT cytotoxicity assay indicated that curcumin at 0.5-2 µM reversed DOX resistance with a reversal index of 1.3-9.3. Western blot analysis revealed that curcumin treatment caused a downregulation of the expression of P-glycoprotein (P-gp) and S100A8 in a dose- and time-dependent manner. To study the internal association between S100A8 and P-gp, and the S100A8 role in drug resistance reversal, an RNA knockdown assay was conducted; however, S100A8 did not regulate the expression of P-gp or vice versa. After inhibiting the expression of S100A8 with specific small interfering RNA (si-S100A8), the sensitivity of K562/DOX cells to DOX was enhanced. In addition, si-S100A8 did not increase the intracellular accumulation of DOX, but increased the intracellular free calcium ion content, and the expression and activity of apoptosis-associated proteins, thereby inducing apoptosis. In conclusion, the present study suggested that inhibition of S100A8 expression increased DOX-induced apoptosis, and curcumin acted independently on S100A8 and P-gp to exert its drug resistance reversal effects.

19.
Mol Ther Nucleic Acids ; 21: 480-491, 2020 Sep 04.
Article in English | MEDLINE | ID: mdl-32679543

ABSTRACT

Previous studies have reported that long noncoding RNAs (lncRNAs) have acted as new players during tumorigenesis. Metallothionein also plays an important role in tumor progression. It is mainly considered to be involved in the process of cell proliferation, oxidative stress, and multidrug resistance. However, the potential involvement of metallothionein-related lncRNAs in colon cancer remains poorly understood. In our study, we found that MT1M affected the expression of lncRNA STEAP3-AS1. STEAP3-AS1 is located in physical contiguity with STEAP3 and notably increased in colon cancer tissues and cell lines. STEAP3-AS1 expression was negatively associated with the expression of STEAP3. High levels of STEPA3-AS1 were associated with poor overall survival in colon cancer patients. In in vitro assays, STEAP3-AS1 knockdown could inhibit colon cancer cell proliferation and migration and arrest colon cancer cells at the G0-G1 phase. In tumorigenicity assays, STEAP3-AS1 knockdown could strongly inhibit tumor growth. Mechanistic investigations demonstrated that STEAP3-AS1 downregulation could increase the expression of cyclin-dependent kinase inhibitor 1C (CDKN1C) by STEAP3 upregulation. Overall, we identify the underlying role of MT1M-related lncRNA STEAP3-AS1 in colon cancer progression, which provides a novel strategy for colon cancer therapy.

20.
Cell Death Differ ; 27(1): 379-395, 2020 01.
Article in English | MEDLINE | ID: mdl-31217502

ABSTRACT

DC-SIGN is previously focused on its physiologic and pathophysiologic roles in immune cells. Little is known about whether DC-SIGN is expressed in malignant epithelial cells and how DC-SIGN participates in tumor progression. Here we showed that DC-SIGN expression was increased in metastatic colorectal cancer (CRC) cell lines and patient tissues. The overall survival in CRC patients with positive DC-SIGN was remarkably reduced. Gain of DC-SIGN function facilitated the CRC metastases both in vitro and in vivo, and this effect was reversed by miR-185. DC-SIGN and Lyn interacted physically, and Lyn maintained the stability of DC-SIGN in cells. DC-SIGN activation recruited Lyn and p85 to form the DC-SIGN-Lyn-p85 complex, which promoted CRC metastasis by increasing PI3K/Akt/ß-catenin signaling in tyrosine kinase Lyn-dependent manner. Furthermore, activation of DC-SIGN promoted the transcription of MMP-9 and VEGF by increasing PI3K/Akt/ß-catenin signaling, and induced TCF1/LEF1-mediated suppression of miR-185. Our findings reveal the presence of the DC-SIGN-TCF1/LEF1-miR-185 loop in cancer cells with metastatic traits, implying that it may represent a new pathogenic mechanism of CRC metastasis. This character of the loop promises to provide new targets for blocking CRC invasive and metastatic activity.


Subject(s)
Cell Adhesion Molecules/metabolism , Colorectal Neoplasms/metabolism , Lectins, C-Type/metabolism , MicroRNAs/metabolism , Receptors, Cell Surface/metabolism , Animals , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/physiology , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Feedback, Physiological , Gene Expression Regulation, Neoplastic , Hepatocyte Nuclear Factor 1-alpha/metabolism , Humans , Lectins, C-Type/genetics , Lectins, C-Type/physiology , Lymphoid Enhancer-Binding Factor 1/metabolism , Matrix Metalloproteinase 9/metabolism , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Neoplasm Invasiveness , Neoplasm Metastasis , Phosphatidylinositol 3-Kinases/metabolism , Prognosis , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/physiology , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , beta Catenin/metabolism , src-Family Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL