Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Cell Rep ; 43(3): 113938, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38460130

ABSTRACT

Recent studies suggest that long non-coding RNAs (lncRNAs) contribute to medulloblastoma (MB) formation and progression. We have identified an lncRNA, lnc-HLX-2-7, as a potential therapeutic target in group 3 (G3) MBs. lnc-HLX-2-7 RNA specifically accumulates in the promoter region of HLX, a sense-overlapping gene of lnc-HLX-2-7, which activates HLX expression by recruiting multiple factors, including enhancer elements. RNA sequencing and chromatin immunoprecipitation reveal that HLX binds to and activates the promoters of several oncogenes, including TBX2, LIN9, HOXM1, and MYC. Intravenous treatment with cerium-oxide-nanoparticle-coated antisense oligonucleotides targeting lnc-HLX-2-7 (CNP-lnc-HLX-2-7) inhibits tumor growth by 40%-50% in an intracranial MB xenograft mouse model. Combining CNP-lnc-HLX-2-7 with standard-of-care cisplatin further inhibits tumor growth and significantly prolongs mouse survival compared with CNP-lnc-HLX-2-7 monotherapy. Thus, the lnc-HLX-2-7-HLX-MYC axis is important for regulating G3 MB progression, providing a strong rationale for using lnc-HLX-2-7 as a therapeutic target for G3 MBs.


Subject(s)
Cerebellar Neoplasms , Medulloblastoma , RNA, Long Noncoding , Humans , Mice , Animals , Feedback , Medulloblastoma/genetics , Medulloblastoma/pathology , Oncogenes , Cerebellar Neoplasms/drug therapy , Cerebellar Neoplasms/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Cell Line, Tumor , Transcription Factors/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism
2.
Acta Neuropathol Commun ; 11(1): 203, 2023 Dec 19.
Article in English | MEDLINE | ID: mdl-38115140

ABSTRACT

The prognosis of childhood medulloblastoma (MB) is often poor, and it usually requires aggressive therapy that adversely affects quality of life. microRNA-211 (miR-211) was previously identified as an important regulator of cells that descend from neural cells. Since medulloblastomas primarily affect cells with similar ontogeny, we investigated the role and mechanism of miR-211 in MB. Here we showed that miR-211 expression was highly downregulated in cell lines, PDXs, and clinical samples of different MB subgroups (SHH, Group 3, and Group 4) compared to normal cerebellum. miR-211 gene was ectopically expressed in transgenic cells from MB subgroups, and they were subjected to molecular and phenotypic investigations. Monoclonal cells stably expressing miR-211 were injected into the mouse cerebellum. miR-211 forced expression acts as a tumor suppressor in MB both in vitro and in vivo, attenuating growth, promoting apoptosis, and inhibiting invasion. In support of emerging regulatory roles of metabolism in various forms of cancer, we identified the acyl-CoA synthetase long-chain family member (ACSL4) as a direct miR-211 target. Furthermore, lipid nanoparticle-coated, dendrimer-coated, and cerium oxide-coated miR-211 nanoparticles were applied to deliver synthetic miR-211 into MB cell lines and cellular responses were assayed. Synthesizing nanoparticle-miR-211 conjugates can suppress MB cell viability and invasion in vitro. Our findings reveal miR-211 as a tumor suppressor and a potential therapeutic agent in MB. This proof-of-concept paves the way for further pre-clinical and clinical development.


Subject(s)
Cerebellar Neoplasms , Medulloblastoma , MicroRNAs , Animals , Humans , Mice , Cell Line, Tumor , Cell Proliferation , Cerebellar Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Homeostasis , Ligases/genetics , Ligases/metabolism , Medulloblastoma/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Quality of Life
3.
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
4.
Acta Neuropathol Commun ; 11(1): 38, 2023 03 10.
Article in English | MEDLINE | ID: mdl-36899402

ABSTRACT

Medulloblastoma (MB) develops through various genetic, epigenetic, and non-coding (nc) RNA-related mechanisms, but the roles played by ncRNAs, particularly circular RNAs (circRNAs), remain poorly defined. CircRNAs are increasingly recognized as stable non-coding RNA therapeutic targets in many cancers, but little is known about their function in MBs. To determine medulloblastoma subgroup-specific circRNAs, publicly available RNA sequencing (RNA-seq) data from 175 MB patients were interrogated to identify circRNAs that differentiate between MB subgroups. circ_63706 was identified as sonic hedgehog (SHH) group-specific, with its expression confirmed by RNA-FISH analysis in clinical tissue samples. The oncogenic function of circ_63706 was characterized in vitro and in vivo. Further, circ_63706-depleted cells were subjected to RNA-seq and lipid profiling to identify its molecular function. Finally, we mapped the circ_63706 secondary structure using an advanced random forest classification model and modeled a 3D structure to identify its interacting miRNA partner molecules. Circ_63706 regulates independently of the host coding gene pericentrin (PCNT), and its expression is specific to the SHH subgroup. circ_63706-deleted cells implanted into mice produced smaller tumors, and mice lived longer than parental cell implants. At the molecular level, circ_63706-deleted cells elevated total ceramide and oxidized lipids and reduced total triglyceride. Our study implicates a novel oncogenic circular RNA in the SHH medulloblastoma subgroup and establishes its molecular function and potential as a future therapeutic target.


Subject(s)
Cerebellar Neoplasms , Medulloblastoma , MicroRNAs , Child , Humans , Animals , Mice , RNA, Circular/genetics , Medulloblastoma/genetics , Hedgehog Proteins/metabolism , MicroRNAs/genetics , Cerebellar Neoplasms/genetics
5.
Neurooncol Adv ; 4(1): vdac120, 2022.
Article in English | MEDLINE | ID: mdl-36267874

ABSTRACT

Background: Although some of the regulatory genes, signaling pathways, and gene regulatory networks altered in medulloblastomas (MB) are known, the roles of non-coding RNAs, particularly long non-coding RNAs (lncRNAs), are poorly described. Here we report that the lncRNA SPRIGHTLY (SPRY4-IT1) gene is upregulated in group 4 medulloblastoma (G4 MB). Methods: SPRIGHTLY expression was assessed in MB subgroup patient-derived xenografts, cell lines, and patient samples. The effect of SPRIGHTLY hemizygous deletion on proliferation, invasion, apoptosis, and colony formation were assessed in vitro and on tumor growth in vivo. dChIRP pull-down assays were used to assess SPRIGHTLY-binding partners, confirmed by immunoprecipitation. SMYD3 ΔE5 transcripts were examined in cell lines and publicly available RNA-seq data. Pathway analysis was performed by phospho-kinase profiling and RNA-seq. Results: CRISPR/Cas9 deletion of SPRIGHTLY reduced cell viability and invasion and increased apoptosis in G4 MB cell lines in vitro. SPRIGHTLY hemizygous-deleted G4 MB cells injected into mouse cerebellums produced smaller tumors than those derived from parental cells expressing both copies of SPRIGHTLY. SPRIGHTLY lncRNA bound to the intronic region of the SMYD3 pre-mRNA transcript. SPRIGHTLY also interacted with PTPB1 protein to regulate SMYD3 exon skipping to produce an aberrant protein. SPRIGHTLY-driven SMYD3 regulation enhanced the expression of EGFR pathway genes in G4 MB cell lines and activated cell coagulation/hemostasis-related gene expression, suggesting a novel oncogenic role in G4 MB. Conclusions: These results demonstrate the importance of SPRIGHTLY lncRNA as a promoter of G4 MB and the role of the SPRIGHTLY-SMYD3-PTPB1 axis as an important oncogenic regulator in MB.

6.
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
8.
Acta Neuropathol Commun ; 10(1): 25, 2022 02 24.
Article in English | MEDLINE | ID: mdl-35209946

ABSTRACT

Medulloblastoma (MB) is the most common malignant brain tumor in children. There remains an unmet need for diagnostics to sensitively detect the disease, particularly recurrences. Cerebrospinal fluid (CSF) provides a window into the central nervous system, and liquid biopsy of CSF could provide a relatively non-invasive means for disease diagnosis. There has yet to be an integrated analysis of the transcriptomic, metabolomic, and lipidomic changes occurring in the CSF of children with MB. CSF samples from patients with (n = 40) or without (n = 11; no cancer) MB were subjected to RNA-sequencing and high-resolution mass spectrometry to identify RNA, metabolite, and lipid profiles. Differentially expressed transcripts, metabolites, and lipids were identified and their biological significance assessed by pathway analysis. The DIABLO multivariate analysis package (R package mixOmics) was used to integrate the molecular changes characterizing the CSF of MB patients. Differentially expressed transcripts, metabolites, and lipids in CSF were discriminatory for the presence of MB but not the exact molecular subtype. One hundred and ten genes and ten circular RNAs were differentially expressed in MB CSF compared with normal, representing TGF-ß signaling, TNF-α signaling via NF-kB, and adipogenesis pathways. Tricarboxylic acid cycle and other metabolites (malate, fumarate, succinate, α-ketoglutarate, hydroxypyruvate, N-acetyl-aspartate) and total triacylglycerols were significantly upregulated in MB CSF compared with normal CSF. Although separating MBs into subgroups using transcriptomic, metabolomic, and lipid signatures in CSF was challenging, we were able to identify a group of omics signatures that could separate cancer from normal CSF. Metabolic and lipidomic profiles both contained indicators of tumor hypoxia. Our approach provides several candidate signatures that deserve further validation, including the novel circular RNA circ_463, and insights into the impact of MB on the CSF microenvironment.


Subject(s)
Cerebellar Neoplasms , Medulloblastoma , Cerebellar Neoplasms/metabolism , Child , Humans , Hypoxia , Lipids , Medulloblastoma/metabolism , Metabolomics/methods , RNA , Tumor Microenvironment
10.
BMC Surg ; 21(1): 213, 2021 Apr 26.
Article in English | MEDLINE | ID: mdl-33902547

ABSTRACT

BACKGROUND: We retrospectively reviewed and consecutively collected the clinical data of distal gastric cancer patients who received surgical treatment, and we discuss the safety and feasibility of double layered end-to-end anastomosis with continuous manual suture to complete digestive tract reconstruction in totally laparoscopic distal gastrectomy. METHODS: We reviewed the clinical data of 41 patients with distal gastric cancer from the gastroenterology department of the Second Affiliated Hospital of Dalian Medical University, from September 2018 to August 2019, who underwent totally laparoscopic distal gastrectomy. During the operation, the method of double layered end-to-end anastomosis with continuous manual suture was used for Billroth type I anastomosis to complete digestive tract reconstruction. All patients have been given a follow-up visit and gastroscopy three months after the operation. The peri-operative clinical information and postoperative follow-up information were collected for analysis, and the clinical application value was evaluated. RESULTS: General information: male(n = 27), female(n = 14), age = 65.02(SD 9.94) years, and BMI = 23.52(SD 2.56) kg/m2, Tumor location: antrum(32,78.0%), angle (6,14.6%), and body (3,7.3%). Clinical stage: I (27, 65.9%), II (7, 17.1%), and III (7, 17.1%). Operative information: operation time = 154.51(SD 33.37) min, anastomosis time = 26.88(SD 5.11) min; intraoperative bleeding = 66.34(SD 48.81) ml; first postoperative ambulation Median = 1(IQR 0) d, first postoperative flatus Median = 3(IQR 2) d, first postoperative diet Median = 3(IQR 1) d, postoperative hospital stay Median = 7(IQR 2) d, and total hospitalization cost = 10,935.00(SD 2205.72)USD. Differentiation degree: high and high-moderate (3,7.32%), moderate and poor-moderate (24, 58.54%), poor differentiation (14, 34.15%), dissected lymph nodes Median = 31(IQR 17), and positive lymph nodes Median = 0(IQR 1). Pathological stage: IA (20, 48.78%), IB (3, 7.32%), IIA (4, 9.76%), IIB (5, 12.20%), IIIA (1, 2.44%), IIIB (3, 7.32%), and IIIC (5, 12.20%). Complications (n = 4): lung infection (1, 2.44%), anastomotic leakage (1, 2.44%), and gastroparesis (2, 4.88%). CONCLUSION: It is safe and feasible in clinical treatment to apply the method of double layered end-to-end anastomosis with continuous manual suture to complete digestive tract reconstruction in totally laparoscopic distal gastrectomy.


Subject(s)
Laparoscopy , Stomach Neoplasms , Aged , Anastomosis, Surgical , Female , Gastrectomy , Gastrointestinal Tract , Humans , Male , Retrospective Studies , Stomach Neoplasms/surgery , Sutures
11.
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.

12.
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
13.
Front Oncol ; 9: 1258, 2019.
Article in English | MEDLINE | ID: mdl-31799200

ABSTRACT

Cancer cells adopt glycolysis to facilitate the generation of biosynthetic substrates demanded by cell proliferation and growth, and to adapt to stress conditions such as excessive reactive oxygen species (ROS) accumulation. TIGAR (TP53-induced glycolysis and apoptosis regulator) is a fructose-2,6-bisphosphatase that is regulated by p53. TIGAR functions to inhibit glycolysis and promote antioxidative activities, which assists the generation of NADPH to maintain the levels of GSH and thus reduces intracellular ROS. However, the functions of TIGAR in gastric cancer (GC) remain unclear. TIGAR expression levels were detected by immunoblotting and immunohistochemistry in gastric cancer samples, along with four established cell lines of GC. The functions of TIGAR were determined by utilizing shRNA-mediated knockdown experiments. The NADPH/NADP+ ratio, ROS, mitochondrial ATP production, and phosphorus oxygen ratios were determined in TIGAR-depleted cells. Xenograft experiment was conducted with BALB/c nude mice. TIGAR was up-regulated compared with corresponding non-cancerous tissues in primary GCs. TIGAR knockdown significantly reduced cell proliferation and increased apoptosis. TIGAR protected cancer cells from oxidative stress-caused damages, but also glycolysis defects. TIGAR also increased the production of NADPH in gastric cancer cells. TIGAR knockdown led to increased ROS production, elevated mitochondrial ATP production, and phosphorus oxygen ratios. The prognosis of high TIGAR expression patients was significantly poorer than those with low TIGAR expression. Taken together, TIGAR exhibits oncogenic features in GC, which can be evaluated as a target for intervention in the treatment of GC.

14.
Oncol Lett ; 14(3): 2813-2821, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28927040

ABSTRACT

Dendritic cell-specific intercellular adhesion molecule-3-grabbing nonintegrin-related protein (DC-SIGNR) is a type II transmembrane protein that has been reported to bind to various pathogens and participate in immunoregulation and tumorigenesis. However, further research is required to investigate whether the level of DC-SIGNR and cervical cancer are associated. The present study aimed to explore the clinical diagnostic significance of DC-SIGNR in cervical cancer. Immunohistochemical staining of DC-SIGNR was performed in samples from 25 patients with early stage cervical cancer, 14 patients with cervical intraepithelial neoplasia (CIN) and cervical polyp samples from 15 individuals. DC-SIGNR expression in cervical cancer tissue was significantly higher compared with that in CIN and cervical polyp tissue (P=0.0184 and P=0.0236, respectively). However, there was no significant difference in DC-SIGNR expression between CIN and cervical polyp tissue (P=0.8103). Additionally, the serum DC-SIGNR levels in 84 cervical cancer patients and 69 healthy female individuals were measured using an ELISA. Serum (s)DC-SIGNR levels were significantly higher in cervical cancer patients compared with healthy female individuals (P<0.0001). A sDC-SIGNR level of 93.7 ng/ml was revealed by receiver operating characteristic curve analysis to predict the presence of cervical cancer with 69.57% sensitivity and 66.67% specificity (area under the curve, 0.6989; P<0.0001). Levels of sDC-SIGNR in cervical cancer patients were also correlated with serum levels of squamous cell carcinoma antigen (r=0.2583; P=0.0348). The results of the present study demonstrate that DC-SIGNR is overexpressed in cervical cancer tissue, and suggest that DC-SIGNR could serve as a biomarker for the early diagnosis of cervical cancer. Nevertheless, further studies are required to demonstrate what role DC-SIGNR serves in cervical cancer.

15.
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
16.
J Hematol Oncol ; 10(1): 28, 2017 01 21.
Article in English | MEDLINE | ID: mdl-28109307

ABSTRACT

BACKGROUND: Tumor metastasis is an essential cause of the poor prognosis of colon cancer. DC-SIGNR is a C-type lectin that is frequently found on human liver sinusoidal endothelial cells. LSECtin, which is a homologue of DC-SIGNR, has been demonstrated to participate in colon cancer liver metastasis. Due to the similarities in the expression pattern and structure of the two proteins, we speculated that DC-SIGNR could also be involved in this process. METHODS: Colon cancer cells were treated with the DC-SIGNR protein or control IgG, after which cell migration, invasion, and morphology were assayed. Xenograft mouse models were used to determine the role of DC-SIGNR in colon cancer liver metastasis in vivo. In addition, a human gene expression array was used to detect differential gene expression in colon cancer cells stimulated with the DC-SIGNR protein. The serum level of DC-SIGNR was examined in colon cancer patients by ELISA, and the significance of DC-SIGNR was determined. RESULTS: In our research, we investigated whether DC-SIGNR promotes colon cancer cell adhesion, migration, and invasion. Knocking down mouse DC-SIGNR decreased the liver metastatic potency of colon cancer cells and increased survival time. Expressing human DC-SIGNR enhanced colon cancer liver metastasis. Furthermore, DC-SIGNR conferred metastatic capability on cancer cells by upregulating various metallothionein isoforms. To validate the above results, we also found that the serum DC-SIGNR level was statistically higher in colon cancer patients with liver metastasis compared with those without metastasis. CONCLUSIONS: These results imply that DC-SIGNR may promote colon carcinoma hepatic metastasis and could serve as a promising therapeutic target for anticancer treatment.


Subject(s)
Cell Adhesion Molecules/pharmacology , Colonic Neoplasms/pathology , Lectins, C-Type/physiology , Liver Neoplasms/secondary , Animals , Cell Adhesion , Cell Adhesion Molecules/therapeutic use , Cell Movement , Disease Models, Animal , Heterografts , Humans , Lectins, C-Type/therapeutic use , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Receptors, Cell Surface/therapeutic use , Tumor Cells, Cultured
17.
World J Gastroenterol ; 19(5): 692-705, 2013 Feb 07.
Article in English | MEDLINE | ID: mdl-23430052

ABSTRACT

AIM: To investigate different methods of creating incomplete intestinal obstruction in a rat model and to compare their electrophysiologic, morphologic and histologic characteristics. METHODS: Rat ileum was partially obstructed by the respective application of: braided silk (penetrated the mesentery and surrounded intestine); half ligation (penetrated directly and ligated 1/2 cross-section of the intestine); wide pipe (6 mm in width, surrounded the intestine); narrow pipe (2 mm in width, surrounded the intestine). A control was also included (no obstruction). Various behavioral and electrophysiologic variables, as well as morphologic and immunohistochemical observations were recorded by blinded investigators at different time points (12, 24, 48, 72 h), including daily general condition, ileal wet weight and circumference, macromorphous and micromorphous intestine, bowel movement capability in vivo and in vitro, slow wave and neural electrical activity, and the number of c-Kit positive interstitial cells of Cajal (ICC). RESULTS: Despite being of a similar general condition, these methods resulted in different levels of obstruction in each group compared with the control at different time points (12, 24, 48, 72 h). However, these fields of the wide pipe rat showed significantly differences when compared with the other three obstructed groups at 12 to 72 h, including macroscopic and histological presentation, intestinal transit ratio and contractility, circumference and wet weight, amplitude and frequency of nerve electrical discharge and slow wave, and ICC numbers (all P < 0.01). CONCLUSION: The wide pipe rat method is significantly more reliable and stable than the other methods of obstruction, demonstrating that use of the wide pipe method can be a useful model of incomplete intestinal obstruction.


Subject(s)
Ileal Diseases/etiology , Ileum/surgery , Intestinal Obstruction/etiology , Animals , Biomarkers/metabolism , Disease Models, Animal , Female , Gastrointestinal Transit , Ileal Diseases/metabolism , Ileal Diseases/pathology , Ileal Diseases/physiopathology , Ileum/innervation , Ileum/pathology , Ileum/physiopathology , Interstitial Cells of Cajal/metabolism , Intestinal Obstruction/metabolism , Intestinal Obstruction/pathology , Intestinal Obstruction/physiopathology , Ligation , Male , Myoelectric Complex, Migrating , Proto-Oncogene Proteins c-kit/metabolism , Rats , Rats, Sprague-Dawley , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...