Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Adv Sci (Weinh) ; 11(24): e2309298, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38639382

ABSTRACT

M2-polarized tumor-associated macrophages (M2 TAMs) promote cancer progression. Exosomes mediate cellular communication in the tumor microenvironment (TME). However, the roles of exosomes from M2 TAMs in gastric cancer progression are unclear. Herein, it is reported that M2 TAMs-derived exosomes induced aerobic glycolysis in gastric cancer cells and enhanced their proliferation, metastasis, and chemoresistance in a glycolysis-dependent manner. It is identified that MALAT1 (metastasis-associated lung adenocarcinoma transcript 1) is enriched in M2 TAM exosomes and confirmed that MALAT1 transfer from M2 TAMs to gastric cancer cells via exosomes mediates this effect. Mechanistically, MALAT1 interacted with the δ-catenin protein and suppressed its ubiquitination and degradation by ß-TRCP. In addition, MALAT1 upregulated HIF-1α expression by acting as a sponge for miR-217-5p. The activation of ß-catenin and HIF-1α signaling pathways by M2 TAM exosomes collectively led to enhanced aerobic glycolysis in gastric cancer cells. Finally, a dual-targeted inhibition of MALAT1 in both gastric cancer cells and macrophages by exosome-mediated delivery of siRNA remarkably suppressed gastric cancer growth and improved chemosensitivity in mouse tumor models. Taken together, these results suggest that M2 TAMs-derived exosomes promote gastric cancer progression via MALAT1-mediated regulation of glycolysis. The findings offer a potential target for gastric cancer therapy.


Subject(s)
Disease Progression , Exosomes , Glycolysis , RNA, Long Noncoding , Stomach Neoplasms , Tumor-Associated Macrophages , Stomach Neoplasms/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/immunology , Exosomes/metabolism , Exosomes/genetics , Humans , Mice , Animals , Tumor Microenvironment/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Cell Line, Tumor , Disease Models, Animal , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Cell Proliferation/genetics
2.
Gene ; 910: 148331, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38438055

ABSTRACT

Compelling evidence has identified circRNAs as crucial regulators in initiation and progression of various cancers, including gastric cancer (GC). However, the function and regulatory mechanisms of circRNAs in GC remain largely unknown. In this study, attention is paid to a novel circular RNA circ1811, which exerts significant downregulated expression in GC tissues compared with adjacent non-cancerous tissues. The expression of circ1811 in GC tumor tissues is negatively correlated with the extent of lymphatic metastasis in GC patients. Overexpression of circ1811 inhibited GC cell proliferation, migration and invasion while promoting apoptosis, whereas knockdown of circ1811 led to the opposite effects. AGO2 RIP and dual luciferase reporter assays indicated that circ1811 directly sponges miR-632 to upregulate the expression of DAPK1. Collectively, circ1811 acts as a tumor-suppressor for GC progression by regulating the miR-632/DAPK1 axis. Our findings suggest the potential of circ1811 as ideal biomarker and therapeutic target for GC.


Subject(s)
MicroRNAs , Stomach Neoplasms , Humans , Stomach Neoplasms/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Circular/genetics , Lymphatic Metastasis , Cell Proliferation/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Cell Movement/genetics , Death-Associated Protein Kinases/genetics , Death-Associated Protein Kinases/metabolism
3.
Clin Chim Acta ; 554: 117773, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38199579

ABSTRACT

BACKGROUND: Gastric cancer (GC) is the third leading cause of cancer-related death worldwide. Sensitive and accurate biomarkers can greatly aid in early diagnosis and favorable prognosis. Neutrophils are the most abundant immune cells in human circulation and play a critical role in tumor progression. Neutrophil-derived exosomes (Neu-Exo) contain abundant bioactive molecules and are critically involved in disease progression. METHODS: We proposed a Dynabeads-based (CD66b antibody-coupled) separation and detection system for Neu-Exo analysis. Dual antibody-assisted fluorescent Dynabeads was established to detect Neu-Exo abundance. MiRNA signature of Neu-Exo was identified by RNA sequencing. QRT-PCR and droplet digital PCR (ddPCR) were used for candidate miRNA detection and the potential of Neu-Exo miRNAs in the diagnosis of gastric cancer was evaluated. RESULTS: Dual antibody-assisted fluorescent Dynabeads obtained a detection limit of 7.8 × 105 particles/mL of Neu-Exo and a recovery rate of 81 % under optimized conditions. ROC curve indicated that the abundance of CD66b+ Neu-Exo could well distinguish GC patients from healthy controls (HC) (AUC > 0.8). Additionally, miR-223-3p was found among the top differentially expressed miRNAs in Neu-Exo and presented superior diagnostic value in gastric cancer. Droplet digital PCR (ddPCR) significantly improved the diagnostic efficiency to differentiate GC patients from HC and benign gastric diseases (BGD) patients (AUC > 0.9). CONCLUSION: The Dynabeads-based separation and detection system, assisted with ddPCR analysis, provides a promising platform to enrich Neu-Exo and analyze miRNA profile for gastric cancer liquid biopsy.


Subject(s)
Exosomes , MicroRNAs , Stomach Neoplasms , Humans , MicroRNAs/genetics , Stomach Neoplasms/diagnosis , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Neutrophils/pathology , Biomarkers, Tumor/genetics , Polymerase Chain Reaction , Exosomes/genetics , Exosomes/pathology
4.
Cell Death Dis ; 14(10): 693, 2023 10 21.
Article in English | MEDLINE | ID: mdl-37865637

ABSTRACT

The interaction between tumor cells and stromal cells within the tumor microenvironment plays a critical role in cancer progression. Mesenchymal stem cells (MSCs) are important tumor stromal cells that exhibit pro-oncogenic activities when reprogrammed by the tumor. However, the precise mechanisms underlying MSC reprogramming in gastric cancer remain not well understood. QRT-PCR, western blot, and immunohistochemistry were used to examine gene and protein expression levels. In vitro and in vivo experiments were conducted to assess the biological functions of gastric cancer cells. RNA-sequencing, RNA immunoprecipitation (RIP), and meRIP assays were performed to investigate underlying molecular mechanisms. We found a significant increase in the expression and N6-methyladenosine (m6A) modification levels of colony-stimulating factor 2 (CSF2) in gastric cancer MSCs. CSF2 gene overexpression induced the reprogramming of normal MSCs into cancer-promoting MSCs, thereby enhancing the proliferation, migration, and drug resistance of gastric cancer cells through the secretion of various pro-inflammatory factors. Additionally, we demonstrated that the m6A reader IGF2BP2 bound to and stabilized CSF2 mRNA in gastric cancer MSCs. Notably, overexpression of IGF2BP2 mimicked the effect of CSF2 on MSCs, promoting gastric cancer progression. Finally, we unveiled that CSF2 induced the ubiquitination of Notch1 to reprogram MSCs. Our study highlights a critical role of IGF2BP2-mediated m6A modification of CSF2 in reprogramming MSCs, which presents a promising therapeutic target for gastric cancer.


Subject(s)
Stomach Neoplasms , Humans , Stomach Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Tumor Microenvironment , RNA-Binding Proteins/genetics
5.
Cancer Cell Int ; 23(1): 151, 2023 Aug 01.
Article in English | MEDLINE | ID: mdl-37525152

ABSTRACT

BACKGROUND: Exosome, a component of liquid biopsy, loaded protein, DNA, RNA and lipid gradually emerges as biomarker in tumors. However, exosomal circRNAs as biomarker and function mechanism in gastric cancer (GC) are not well understood. METHODS: Differentially expressed circRNAs in GC and healthy people were screened by database. The identification of hsa_circ_000200 was verified by RNase R and sequencing, and the expression of hsa_circ_000200 was evaluated using qRT-PCR. The biological function of hsa_circ_000200 in GC was verified in vitro. Western blot, RIP, RNA fluorescence in situ hybridization, and double luciferase assay were utilized to explore the potential mechanism of hsa_circ_000200. RESULTS: Hsa_circ_000200 up-regulated in GC tissue, serum and serum exosomes. Hsa_circ_000200 in serum exosomes showed better diagnostic ability than that of tissues and serum. Combined with clinicopathological parameters, its level was related to invasion depth, TNM staging, and distal metastasis. Functionally, knockdown of hsa_circ_000200 inhibited GC cells proliferation, migration and invasion in vitro, while its overexpression played the opposite role. Importantly, exosomes with up-regulated hsa_circ_000200 promoted the proliferation and migration of co-cultured GC cells. Mechanistically, hsa_circ_000200 acted as a "ceRNA" for miR-4659a/b-3p to increase HBEGF and TGF-ß/Smad expression, then promoted the development of GC. CONCLUSIONS: Our findings suggest that hsa_circ_000200 promotes the progression of GC through hsa_circ_000200/miR-4659a/b-3p/HBEGF axis and affecting the expression of TGF-ß/Smad. Serum exosomal hsa_circ_000200 may serve as a potential biomarker for GC.

6.
Curr Stem Cell Res Ther ; 18(6): 853-863, 2023.
Article in English | MEDLINE | ID: mdl-36424798

ABSTRACT

INTRODUCTION: Resident mesenchymal stem cells (MSCs) in the tumor microenvironment play an important role in tumor progression. Up to now, the mechanism of resident MSCs promoting gastric cancer cell migration remains unclear. METHODS: We tested the migration ability of gastric cancer cells by transwell assays in this study. The inflammatory factors secreted by MSCs were detected by Luminex and ELISA. The activation of NF-κB signaling was detected by western blot. The exosomes derived from MSCs were isolated and identified by transmission electron microscope, nano-sight and western blot. The expression of miR-374a-5p was confirmed by qRT-PCR and its downstream target HAPLN1 by luciferase reporter assay. The expression of adhesion molecules of gastric cancer cells was detected by flow cytometry. RESULTS: MiR-374a-5p could regulate the expression of inflammatory factors by activating NF-κB signaling. The increase of MCP-1 and the decrease of IFN-γ promoted the migration of gastric cancer cells. The miR-374a-5p in MSCs could be encapsulated and delivered to gastric cancer cells by exosomes derived from MSCs. Exogenous miR-374a-5p up-regulated the expression of adhesion molecules in gastric cancer cells by targeting HAPLN1. And miR-374a-5p-enriched exosomes also promoted the migration of gastric cancer cells. CONCLUSION: MiR-374a-5p promoted gastric cancer metastasis, and resident MSCs in the gastric cancer microenvironment played a major role in the regulation of gastric cancer metastasis. The study will provide new ideas and potential targets for the prevention and treatment of gastric cancer metastasis.


Subject(s)
Exosomes , Mesenchymal Stem Cells , MicroRNAs , Stomach Neoplasms , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , NF-kappa B/genetics , Cell Line, Tumor , Exosomes/genetics , Exosomes/metabolism , Mesenchymal Stem Cells/metabolism , Tumor Microenvironment/genetics
7.
Front Oncol ; 12: 1008843, 2022.
Article in English | MEDLINE | ID: mdl-36185262

ABSTRACT

As an abundant component of tumor microenvironment, cancer-associated fibroblasts (CAFs) are heterogeneous cell populations that play important roles in tumor development, progression and therapeutic resistance. Multiple sources of cells can be recruited and educated to become CAFs, such as fibroblasts, mesenchymal stem cells and adipocytes, which may explain the phenotypic and functional heterogeneity of CAFs. It is widely believed that CAFs regulate tumor progression by remodeling extracellular matrix, promoting angiogenesis, and releasing soluble cytokines, making them a promising cancer therapy target. In this review, we discussed about the origin, subpopulation, and functional heterogeneity of CAFs, with particular attention to recent research advances and clinical therapeutic potential of CAFs in cancer.

8.
Front Cell Dev Biol ; 9: 765635, 2021.
Article in English | MEDLINE | ID: mdl-34805173

ABSTRACT

RNA N6-methyladenosine (m6A) modification has important regulatory roles in determining cell fate. The reversible methylation process of adding and removing m6A marks is dynamically regulated by a fine-tuned coordination of many enzymes and binding proteins. Stem cells have self-renewal and pluripotent potential and show broad prospects in regenerative medicine and other fields. Stem cells have also been identified in cancer, which is linked to cancer metastasis, therapy resistance, and recurrence. Herein, we aimed to review the molecular mechanism that controls the reversible balance of m6A level in stem cells and the effect of m6A modification on the balance between pluripotency and differentiation. Additionally, we also elaborated the association between aberrant m6A modification and the maintenance of cancer stem cells in many cancers. Moreover, we discussed about the clinical implications of m6A modification in cancer stem cells for cancer diagnosis and therapy.

9.
Front Cell Dev Biol ; 8: 788, 2020.
Article in English | MEDLINE | ID: mdl-32903528

ABSTRACT

In response to tumor signals, mesenchymal stem cells (MSCs) are recruited to tumor sites and activated to promote tumor progression. Emerging evidences suggest that in addition to tumor cells, non-tumor cells in tumor microenvironment could also interact with MSCs to regulate their phenotype and function. However, the mechanism for MSCs regulation in gastric cancer has not been fully understood. In this study, we reported that tumor-educated neutrophils (TENs) induced the transformation of MSCs into cancer-associated fibroblasts (CAFs) which in turn remarkably facilitated gastric cancer growth and metastasis. Mechanistic study showed that TENs exerted their effects by secreting inflammatory factors including IL-17, IL-23 and TNF-α, which triggered the activation of AKT and p38 pathways in MSCs. Pre-treatment with neutralizing antibodies to these inflammatory factors or pathway inhibitors reversed TENs-induced transformation of MSCs to CAFs. Taken together, these data suggest that TENs promote gastric cancer progression through the regulation of MSCs/CAFs transformation.

10.
Oncogenesis ; 9(7): 63, 2020 Jul 06.
Article in English | MEDLINE | ID: mdl-32632106

ABSTRACT

Deregulated expression of chemokines in tumor microenvironment contributes to tumor metastasis by targeting distinct cells. Epithelial-derived neutrophil-activating peptide-78 (ENA78/CXCL5) is upregulated in many cancers and involved in tumor progression. The role and underlying mechanism of CXCL5 in gastric cancer (GC) metastasis remain unclear. In this study, we reported that the expression of CXCL5 was elevated in tumor tissues and positively associated with lymphatic metastasis and tumor differentiation. Stimulation by recombinant human CXCL5 (rhCXCL5) induced epithelial-mesenchymal transition (EMT) in GC cells through the activation of ERK pathway, which enhanced their migration and invasion abilities. The culture supernatant from tumor tissues also enhanced the migration and invasion abilities of GC cells, however, this effect was reversed by pre-treatment with CXCL5 neutralizing antibody. Further studies showed that rhCXCL5 could induce the expression of IL-6 and IL-23 in neutrophils through the activation of ERK and p38 signaling pathways, which in turn facilitated GC cell migration and invasion. The culture supernatant from tumor tissues showed similar effects on neutrophils in a CXCL5-dependent manner. Blockade of IL-6 and IL-23 with neutralizing antibodies reversed the induction of EMT and the increased migration and invasion abilities in GC cells by CXCL5-activated neutrophils. Moreover, CXCL5 activated neutrophils could promote gastric cancer metastasis in vivo. Taken together, our results indicate that CXCL5 acts on gastric cancer cells to induce EMT and mediates pro-tumor activation of neutrophils, which synergistically promotes the metastatic ability of GC cells.

11.
Front Oncol ; 10: 629, 2020.
Article in English | MEDLINE | ID: mdl-32477934

ABSTRACT

Neutrophils are prominent components of solid tumors and exhibit distinct phenotypes in different tumor milieu. We have previously shown that tumor extracellular vesicles (EVs) could induce pro-tumor activation of neutrophils; however, the role of tumor EV-elicited neutrophils in tumor immunity remains unclear. Herein, we reported that gastric cancer cell-derived EVs (GC-EVs) induced the expression of programmed death-ligand 1 (PD-L1) on neutrophils. GC-EVs transported high-mobility group box-1 (HMGB1) to activate signal transducer and activator of transcription 3 (STAT3) and upregulate PD-L1 gene expression in neutrophils. Blocking STAT3 pathway and silencing HMGB1 reversed GC-EV-induced PD-L1 expression on neutrophils. GC-EV-elicited neutrophils suppressed T cell proliferation, activation, and function in vitro, which could be antagonized by a specific PD-L1 antibody. Furthermore, GC tissue-derived EVs also showed similar effects. Taken together, our results indicate that EVs from the GC microenvironment induce PD-L1 expression on neutrophils to suppress T-cell immunity, which provides a new insight into the pro-tumor roles of neutrophils in GC and sheds light on the multifaceted roles of EVs in orchestrating an immunosuppressive microenvironment.

12.
Cancer Cell Int ; 20: 188, 2020.
Article in English | MEDLINE | ID: mdl-32489324

ABSTRACT

BACKGROUND: The stem cell factor SALL4 is reactivated in human cancers. SALL4 plays diverse roles in tumor growth, metastasis, and drug resistance, but its role in tumor metabolism has not been well characterized. METHODS: The glycolytic levels of gastric cancer cells were detected by glucose uptake, lactate production, lactate dehydrogenase activity, ATP level, and hexokinase activity. QRT-PCR and western blot were used to detect the changes in the expression of glycolytic genes and proteins. The downstream target genes of SALL4 were identified by microarray. The regulation of hexokinase II (HK-2) by SALL4 was analyzed by luciferase reporter assay and chromatin immunoprecipitation assay. Transwell migration assay, matrigel invasion assay, cell counting assay and colony formation assay were used to study the roles of HK-2 regulation by SALL4 in gastric cancer cells in vitro. The effects of SALL4 on glycolysis and gastric cancer progression in vivo were determined by subcutaneous xenograft and peritoneal metastasis tumor models in nude mice. RESULTS: SALL4 knockdown inhibited glucose uptake, lactate production, lactate dehydrogenase activity, ATP level and hexokinase activity in gastric cancer cells, and decreased the expression of glycolytic genes and proteins. Microarray analysis showed that SALL4 knockdown affected glycolysis-related pathway. The regulation of HK-2 gene expression by SALL4 was confirmed by luciferase reporter assay and chromatin immunoprecipitation assay. HK-2 knockdown abrogated the promotion of glycolysis by SALL4 in gastric cancer cells, indicating that HK-2 acts as a downstream effector of SALL4. Moreover, HK-2 knockdown reversed the promoting role of SALL4 in gastric cancer cell proliferation, migration and invasion, suggesting that SALL4 drives gastric cancer progression by upregulating HK-2. CONCLUSIONS: SALL4 promotes gastric cancer progression through HK-2-mediated glycolysis, which reveals a new mechanism for the oncogenic roles of SALL4 in cancer.

13.
Hum Cell ; 33(3): 652-662, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32350750

ABSTRACT

The tumor microenvironment (TM) is an essential factor of tumor progression. Mesenchymal stem cells (MSCs) are important components of the TM and play critical roles in cancer metastasis. Resveratrol (RES) is a potential antitumor drug that has attracted extensive attention. However, it remains unclear whether RES can exert its antitumor activity by targeting MSCs located in the TM. In this study, we demonstrated that the conditioned medium of gastric-cancer-derived MSCs (GC-MSCs) promoted gastric cancer (GC) metastasis and facilitated the progression of epithelialmesenchymal transition (EMT) of GC cells. However, after pretreatment with RES, the prometastatic effect of GC-MSCs on GC cells was reversed. Furthermore, RES reduced GC-MSC (IL-6, IL-8, MCP-1, VEGF) gene expression and protein secretion, and counteracted the activation of the GC-MSC-induced Wnt/ß-catenin signaling of GC cells, with less ß-catenin nuclear transport and declined expression of ß-catenin, CD44, and CyclinD3 in GC cells. Re-expression of ß-catenin impaired the inhibitory effect of RES on GC cells. In conclusion, RES restricted the mobility increase of GC cells and reversed the progress of EMT induced by GC-MSCs by inactivating the Wnt/ß-catenin signaling. GC-MSCs are promising target for RES in the inhibition of GC metastasis.


Subject(s)
Mesenchymal Stem Cells/physiology , Neoplasm Metastasis/drug therapy , Resveratrol/therapeutic use , Stomach Neoplasms/drug therapy , Antineoplastic Agents, Phytogenic , Cell Line, Tumor , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Gene Expression/drug effects , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , Mesenchymal Stem Cells/pathology , Molecular Targeted Therapy , Phytotherapy , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Tumor Microenvironment , Wnt Proteins/metabolism , beta Catenin/genetics , beta Catenin/metabolism
14.
Cell Death Dis ; 10(10): 752, 2019 10 03.
Article in English | MEDLINE | ID: mdl-31582742

ABSTRACT

Long non-coding RNAs (lncRNAs) have been suggested as important regulators of cancer development and progression in hepatocellular carcinoma (HCC). Nevertheless, the clinical value and biological roles of LINC00978 in HCC remain unclear. In this study, we detected the expression of LINC00978 in tumor tissues and serum of HCC patients, examined the roles of LINC00978 in HCC progression and elucidated the underlying molecular mechanisms. We found that LINC00978 expression was upregulated in tumor tissues and serum of HCC patients. Higher serum levels of LINC00978 could distinguish HCC patients from hepatitis and liver cirrhosis patients and healthy controls. LINC00978 knockdown inhibited HCC cell proliferation, migration and invasion while promoted cell cycle arrest and apoptosis. Overexpression of LINC00978 led to the opposite effects. LINC00978 knockdown also inhibited HCC growth and metastasis in mouse tumor models. Mechanistically, LINC00978 bound to EZH2 and mediated its accumulation at the promoter region of p21 and E-cadherin genes, leading to the trimethylation of H27K3 and the inhibition of p21 and E-cadherin expression. Moreover, the simultaneous depletion of p21 and E-cadherin expression reversed the inhibitory effects of LINC00978 knockdown on HCC cell proliferation, migration, and invasion. Taken together, these findings suggest that LINC00978 promotes HCC progression by inhibiting p21 and E-cadherin expression via EZH2-mediated epigenetic silencing. LINC00978 may represent a novel biomarker for HCC diagnosis, prognosis, and therapy.


Subject(s)
Carcinoma, Hepatocellular/genetics , Disease Progression , Liver Neoplasms/genetics , RNA, Long Noncoding/metabolism , Animals , Antigens, CD , Apoptosis/genetics , Cadherins , Carcinogenesis/genetics , Carcinogenesis/pathology , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cyclin-Dependent Kinase Inhibitor p21 , Down-Regulation/genetics , Enhancer of Zeste Homolog 2 Protein , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Liver Neoplasms/blood , Liver Neoplasms/pathology , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Protein Binding/genetics , RNA, Long Noncoding/genetics , Transcription, Genetic , Up-Regulation/genetics
15.
Mol Ther Nucleic Acids ; 18: 320-331, 2019 Dec 06.
Article in English | MEDLINE | ID: mdl-31614322

ABSTRACT

Chemoresistance is one of the causes associated with poor prognosis in gastric cancer. MicroRNAs (miRNAs) are important regulators of chemoresistance. Exosome-mediated delivery of anti-cancer molecules and drugs have emerged as a new approach for cancer therapy. We first examined the expression of miR-374a-5p in gastric cancer serum by qRT-PCR and explored the clinicopathological parameters. We then performed in vitro cell and molecular studies, including CCK-8 assay, flow cytometry, qRT-PCR, and western blot, to determine the roles of miR-374a-5p in gastric cancer chemoresistance and identified its downstream target by luciferase reporter assay. We also used in vivo animal studies to evaluate the therapeutic efficacy of miR-374a-5p inhibitor and exosome-mediated delivery of miR-374a-5p inhibitor in gastric cancer. miR-374a-5p expression level was elevated in gastric cancer serum, and its upregulation predicted poor prognosis. miR-374a-5p overexpression promoted while miR-374a-5p knockdown inhibited gastric cancer chemoresistance in vitro and in vivo. miR-374a-5p bound to Neurod1 to antagonize its effect on chemoresistance. Exosome-mediated delivery of miR-374a-5p inhibitor could increase Neurod1 expression, promote cell apoptosis, and suppress chemoresistance. miR-374a-5p had a promoting role in gastric cancer chemoresistance, which would provide a novel biomarker for gastric cancer diagnosis and prognosis and offer a potential target for gastric cancer drug resistance therapy.

16.
Aging (Albany NY) ; 11(10): 3376-3391, 2019 05 30.
Article in English | MEDLINE | ID: mdl-31147526

ABSTRACT

MircoRNA-335 (miR-335) has been reported as a significant cancer-associated microRNA, which was often epigenetically silenced and acted as a tumor suppressor gene in diverse human solid tumors. Conversely, recent studies show that miR-335 overexpression was identified in both adult and pediatric acute myeloid leukemia (AML), suggesting that it might play an oncogenic role of miR-335 in AML. However, the role of miR-335 during leukemogenesis remains to be elucidated. MiR-335/ID4 expression was detected by real-time quantitative PCR and/or western blot. Survival analysis was performed to explore the association between miR-335/ID4 expression and the prognosis, and further validated by public databases. Gain-of-function experiments determined by cell proliferation, apoptosis, and differentiation were conducted to investigate the biological functions of miR-335/ID4. Herein, we found that miR-335 expression, independent of its methylation, was significantly increased and negatively correlated with reduced ID4 expression in AML. Moreover, aberrant miR-335/ID4 expression independently affected chemotherapy response and leukemia-free/overall survival in patients with AML. Gain-of-function experiments in vitro showed the oncogenic role of miR-335 by affecting cell apoptosis and proliferation in AML, and could be rescued by ID4 restoration. Mechanistically, we identified and verified that miR-335/ID4 contributed to leukemogenesis through activating PI3K/Akt signaling pathway. Collectively, aberrant miR-335/ID4 expression was an independent prognostic biomarker in AML. MiR-335/ID4 dysregulation facilitated leukemogenesis through the activation of PI3K/Akt signaling pathway.


Subject(s)
Inhibitor of Differentiation Proteins/metabolism , Leukemia, Myeloid, Acute/etiology , Leukemia, Myeloid, Acute/metabolism , MicroRNAs/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Case-Control Studies , China/epidemiology , Female , HL-60 Cells , Humans , Induction Chemotherapy , K562 Cells , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Young Adult
17.
J Transl Med ; 17(1): 102, 2019 03 29.
Article in English | MEDLINE | ID: mdl-30922328

ABSTRACT

BACKGROUND: Leukemia stem cell (LSC)-enriched genes have been shown to be highly prognostic in acute myeloid leukemia (AML). However, the prognostic value of tumor suppressor genes (TSGs) that are repressed early in LSC remains largely unknown. METHODS: We compared the public available expression/methylation profiling data of LSCs with that of hematopoietic stem cells (HSCs), in order to identify potential tumor suppressor genes in LSC. The prognostic relevance of PCDH17 was analyzed on a cohort of 173 AML patients from The Cancer Genome Atlas (TCGA), and further validated in three independent cohorts (n = 339). RESULTS: We identified protocadherin17 (PCDH17) and demonstrated that it was significantly down-regulated and hypermethylated in LSCs compared with HSCs. Our analyses of primary AML patient samples also confirmed these deregulations. Clinically, low PCDH17 expression was associated with female sex (P = 0.01), higher WBC (P < 0.0001), higher percentages of blasts in bone marrow (BM) and peripheral blood (PB) (P = 0.04 and < 0.001, respectively), presence of FLT3-internal tandem duplications (P = 0.002), mutated NPM1 (P = 0.02), and wild-type TP53 (P = 0.005). Moreover, low PCDH17 expression predicted worse overall survival (OS) in four independent cohorts as well as in the molecularly defined subgroups of AML patients. In multivariable analyses, low PCDH17 expression retained independent prognostic value for OS. Biologically, PCDH17 expression-associated gene signatures were characterized by deregulations of EMT- and Wnt pathway-related genes. CONCLUSIONS: PCDH17 gene was silenced by DNA methylation in AML. Low PCDH17 expression is associated with distinct clinical and biological features and improves risk stratification in patients with AML.


Subject(s)
Cadherins/genetics , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/genetics , Neoplastic Stem Cells/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Cadherins/metabolism , Case-Control Studies , Cohort Studies , DNA Methylation , Down-Regulation/genetics , Epigenesis, Genetic , Female , Gene Expression Regulation, Leukemic , HL-60 Cells , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Neoplastic Stem Cells/pathology , Nucleophosmin , Prognosis , Survival Analysis , Transcriptome , Young Adult
18.
J Cell Mol Med ; 23(5): 3317-3324, 2019 05.
Article in English | MEDLINE | ID: mdl-30793488

ABSTRACT

The clinical activity of decitabine (5-aza-2-deoxycytidine, DAC), a hypomethylating agent, has been demonstrated in acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) patients. However, secondary resistance to this agent often occurs during treatment and leads to treatment failure. It is important to clarify the mechanisms underlying the resistance for improving the efficacy. In this study, by gradually increasing concentration after a continuous induction of DAC, we established the DAC-resistant K562 cell line (K562/DAC) from its parental cell line K562. The proliferation and survival rate of K562/DAC was significantly increased, whereas the apoptosis rate was remarkably decreased than that of K562 after DAC treatment. In K562/DAC, a total of 108 genes were upregulated and 118 genes were downregulated by RNA-Seq. In addition, we also observed aberrant expression of DDX43/H19/miR-186 axis (increased DDX43/H19 and decreased miR-186) in K562/DAC cells. Ectopic expression of DDX43 in parental K562 cells rendered cells resistant to the DAC. Taken together, we successfully established DAC-resistant K562 cell line which can serve as a good model for investigating DAC resistance mechanisms, and DDX43/H19/miR-186 may be involved in DAC resistance in K562.


Subject(s)
Decitabine/pharmacology , Drug Resistance, Neoplasm/drug effects , DEAD-box RNA Helicases/metabolism , Gene Expression Regulation, Leukemic/drug effects , Humans , K562 Cells , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasm Proteins/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
19.
Am J Transl Res ; 10(10): 3224-3232, 2018.
Article in English | MEDLINE | ID: mdl-30416663

ABSTRACT

MicroRNAs (miRNAs) play important roles in cell transformation and carcinogenesis. We have previously established a tumor cell line K3 transformed from rat bone marrow-derived mesenchymal stem cells (rBM-MSCs). However, the underlying mechanism involved in MSC transformation remains unclear. Herein, we identified the key miRNAs that regulate the transformation of rBM-MSCs, and clarified their biological roles. Microarray and qRT-PCR results showed an increased expression of miR-374 but decreased expressions of miR-199a, miR-145, miR-34a, and miR-214 in K3 cells compared to rBM-MSCs. MiR-374 overexpression in rBM-MSCs increased the colony number and the proportion of the cells in S-phase. In addition, miR-374 overexpression reduced E-cadherin expression and increased N-cadherin expression in rBM-MSCs, promoting the migration ability of these cells. On the contrary, miR-374 knockdown in K3 cells led to impaired proliferation and migration capacities. Furthermore, wnt5a was identified as a target gene of miR-374. MiR-374 overexpression upregulated ß-catenin expression in rBM-MSCs while miR-374 knockdown downregulated that in K3 cells. In conclusion, miR-374 promotes the proliferation and migration of transformed MSCs by regulating Wnt5a/ß-catenin signaling pathway, which provides evidence for the contribution of miRNA to MSC transformation and suggests a new role of miR-374 in cancer development and progression.

20.
Oncol Rep ; 38(5): 2959-2966, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28901479

ABSTRACT

Emerging evidence has revealed that neutrophils have phenotypic and functional plasticity. Neutrophils could be polarized towards a pro-tumor phenotype by tumor-derived factors. In the present study, we investigated the role of the interaction with neutrophils on the functions of gastric cancer cells in vitro. Human promyelocytic leukemia HL-60 cells were induced to differentiate into neutrophil-like cells (HL-60N) using dimethyl sulfoxide (DMSO). Human gastric cancer cells were co-cultured with HL-60N cells or treated with the conditioned medium (CM) of cancer-activated HL-60N cells. The migration and invasion of gastric cancer cells were significantly enhanced while their proliferation was minimally altered. The expression of pro-inflammatory factors including IL-6, IL-8, IL-1ß, and TNFα was significantly increased in cancer-activated HL-60N cells, which induced the activation of the ERK pathway and epithelial-mesenchymal transition (EMT) in gastric cancer cells. Blocking the ERK pathway activation reversed the promoting effects of cancer-activated HL-60N cells on gastric cancer cell migration and invasion. In addition, mouse gastric cancer cell derived CM could also increase the expression of pro-inflammatory factors in mouse bone marrow neutrophils, which in turn enhanced the migration and invasion of mouse gastric cancer cells. Collectively, our findings revealed that the interaction with neutrophils promoted gastric cancer cell migration and invasion through the activation of the ERK pathway and the induction of EMT, indicating that neutrophils may play an important role in gastric cancer metastasis. Therefore, targeting neutrophil-cancer cell interaction may provide a new strategy for the treatment of gastric cancer.


Subject(s)
Epithelial-Mesenchymal Transition , Interleukins/metabolism , Neutrophils/cytology , Stomach Neoplasms/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Differentiation , Cell Line, Tumor , Cell Movement , Cell Survival , Coculture Techniques , Culture Media, Conditioned/pharmacology , Dimethyl Sulfoxide/pharmacology , HL-60 Cells , Humans , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , Interleukins/genetics , MAP Kinase Signaling System , Mice , Neutrophils/immunology , Neutrophils/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/immunology , Tumor Necrosis Factor-alpha/genetics
SELECTION OF CITATIONS
SEARCH DETAIL