Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
Add more filters

Publication year range
1.
Biochem Pharmacol ; 197: 114940, 2022 03.
Article in English | MEDLINE | ID: mdl-35120895

ABSTRACT

Programmed death ligand-1 (PD-L1) and indoleamine 2, 3-dioxygenase 1 (IDO1) are immune checkpoints induced by interferon-γ (IFN-γ) in the tumor microenvironment, leading to immune escape of tumors. Myricetin (MY) is a flavonoid distributed in many edible and medicinal plants. In this study, MY was identified to inhibit IFN-γ-induced PD-L1 expression in human lung cancer cells. It also reduced the expression of IDO1 and the production of kynurenine which is the product catalyzed by IDO1, while didn't show obvious effect on the expression of major histocompatibility complex-I (MHC-I), a crucial molecule for antigen presentation. In addition, the function of T cells was evaluated using a co-culture system consist of lung cancer cells and the Jurkat-PD-1 T cell line overexpressing PD-1. MY restored the survival, proliferation, CD69 expression and interleukin-2 (IL-2) secretion of Jurkat-PD-1 T cells suppressed by IFN-γ-treated lung cancer cells. Mechanistically, IFN-γ up-regulated PD-L1 and IDO1 at the transcriptional level through the JAK-STAT-IRF1 axis, which was targeted and inhibited by MY. Together, our research revealed a new mechanism of MY mediated anti-tumor activity and highlighted the potential implications of MY in tumor immunotherapy.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , Flavonoids/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Interferon-gamma/pharmacology , Lung Neoplasms/metabolism , A549 Cells , B7-H1 Antigen/biosynthesis , B7-H1 Antigen/genetics , Coculture Techniques , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic/physiology , HCT116 Cells , HEK293 Cells , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Jurkat Cells , Tumor Microenvironment/drug effects , Tumor Microenvironment/physiology
2.
Int J Mol Sci ; 23(4)2022 Feb 18.
Article in English | MEDLINE | ID: mdl-35216384

ABSTRACT

Zinc has been suggested to play a role in carcinogenesis and tumor progression. Serum zinc levels of lung cancer patients are for example lower than in healthy individuals. The activation and expression of the epidermal growth factor receptor (EGFR), which plays a role in tumor biology, are presumably influenced by zinc. EGFR activation influences cell adhesion and immune escape. This study provides insights into the impacts of zinc on the EGFR activation and expression of downstream proteins such as E-cadherin and PD-L1 in the alveolar carcinoma cell line A549. To model chronic changes in zinc homeostasis, A549 cells were cultured in media with different zinc contents. EGFR surface expression of unstimulated and stimulated A549 cells was determined by flow cytometry. EGFR phosphorylation as well as the protein expression of E-cadherin and PD-L1 were analyzed by Western blot. In our hands, chronic zinc deficiency led to increased EGFR surface expression, decreased E-cadherin protein expression and increased PD-L1 protein expression. Zinc supplementation decreased EGFR surface expression and PD-L1 protein expression. In summary, zinc-deficient A549 cells may display a more malignant phenotype. Thus, future clinical research should further focus on the possible benefits of restoring disturbed zinc homeostasis, especially in lung cancer patients.


Subject(s)
Zinc/metabolism , A549 Cells , B7-H1 Antigen/metabolism , Cadherins/metabolism , Cell Line, Tumor , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic/physiology , Humans , Lung Neoplasms/metabolism , Signal Transduction/physiology
3.
Chem Biol Interact ; 351: 109705, 2022 Jan 05.
Article in English | MEDLINE | ID: mdl-34656559

ABSTRACT

Non-small cell lung cancer (NSCLC) is one of the prevalent and deadly cancers worldwide. Cisplatin (CDDP) has been used as a standard adjuvant therapy for advanced NSCLC patients, while chemoresistance is one of the most challenging problems to limit its clinical application. Our data showed that the expression of visfatin was significantly increased in CDDP resistant NSCLC cells as compared with that in their parental cells, while knockdown of visfatin or its neutralization antibody can restore the CDDP sensitivity of resistant NSCLC cells. The upregulation of visfatin in CDDP resistant NSCLC cells was due to the increased mRNA stability and promoter activity. Further, we found that signal transducer and activator of transcription 3 (STAT3), which was increased in chemoresistant cells, can increase the transcription of visfatin. While tristetraprolin (TTP), which can decease mRNA stability of visfatin, was decreased in chemoresistant cells. Inhibition of STAT3 or over expression of TTP can restore CDDP sensitivity of resistant NSCLC cells. Collectively, our data showed that STAT3 and TTP-regulated expression of visfatin was involved in CDDP resistance of NSCLC cells. It indicated that targeted inhibition of visfatin should be a potential approach to overcome CDDP resistance of NSCLC treatment.


Subject(s)
Carcinoma, Non-Small-Cell Lung/physiopathology , Cytokines/metabolism , Drug Resistance, Neoplasm/physiology , Lung Neoplasms/physiopathology , Nicotinamide Phosphoribosyltransferase/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/physiology , Humans , RNA Stability/physiology , STAT3 Transcription Factor/metabolism , Tristetraprolin/metabolism , Up-Regulation/physiology
4.
Int J Biol Sci ; 17(12): 3133-3144, 2021.
Article in English | MEDLINE | ID: mdl-34421355

ABSTRACT

Sorafenib is the standard first-line drug for the treatment of advanced hepatocellular carcinoma (HCC), however, its therapeutic efficacy is not satisfactory due to primary or secondary resistance of HCC cells. In the present study, we identified Metaxin 1 (MTX1) as a new regulator of sorafenib resistance in HCC through genome-scale CRISPR activation (CRISPRa) screening. We found that MTX1 was frequently upregulated in HCC tissues and overexpression of MTX1 promoted HCC cell proliferation in vitro and in vivo. As well, MTX1 overexpression increased cell growth rate and decreased cell apoptosis upon sorafenib treatment. Consistently, the resistance induced by MTX1 was also observed in subcutaneous xenograft tumor model. Clinically, high expression of MTX1 was closely related with poor outcomes in HCC patients who received sorafenib treatment. Mechanistically, overexpression of MTX1 could promote HCC cell autophagy via interacting with and inhibiting CDGSH iron sulfur domain 1 (CISD1), an autophagy negative regulator. Taken together, our findings suggest that MTX1 is upregulated in HCC and contributes to sorafenib resistance via a possible mechanism involving CISD1 mediated autophagy.


Subject(s)
Antineoplastic Agents/therapeutic use , Autophagy/drug effects , Carcinoma, Hepatocellular/genetics , Drug Resistance, Neoplasm/drug effects , Liver Neoplasms/genetics , Mitochondrial Membrane Transport Proteins/genetics , Sorafenib/therapeutic use , Animals , CRISPR-Cas Systems , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Cell Proliferation , Cells, Cultured , Flow Cytometry , Gain of Function Mutation , Gene Expression Regulation, Neoplastic/physiology , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Mice , Mice, Nude , Microscopy, Electron , Real-Time Polymerase Chain Reaction , Xenograft Model Antitumor Assays
5.
Chem Biol Interact ; 347: 109602, 2021 Sep 25.
Article in English | MEDLINE | ID: mdl-34331906

ABSTRACT

Breast cancer is the most common cancer among females and the leading cause of cancer-related deaths. Approximately 70 % of breast cancers are estrogen receptor (ER) positive. An ER antagonist such as tamoxifen is used as adjuvant therapy in ER-positive patients. The major problem with endocrine therapy is the emergence of acquired resistance in approximately 40 % of patients receiving tamoxifen. Metabolic alteration is one of the hallmarks of cancer cells. Rapidly proliferating cancer cells require increased nutritional support to fuel various functions such as proliferation, cell migration, and metastasis. Recent studies have established that the metabolic state of cancer cells influences their susceptibility to chemotherapeutic drugs and that cancer cells reprogram their metabolism to develop into resistant phenotypes. In this review, we discuss the major findings on metabolic pathway alterations in tamoxifen-resistant (TAMR) breast cancer and the molecular mechanisms known to regulate the expression and function of metabolic enzymes and the respective metabolite levels upon tamoxifen treatment. It is anticipated that this in-depth analysis of specific metabolic pathways in TAMR cancer might be exploited therapeutically.


Subject(s)
Breast Neoplasms/metabolism , Drug Resistance, Neoplasm/physiology , Tamoxifen/therapeutic use , Amino Acids/metabolism , Animals , Breast Neoplasms/drug therapy , Cell Line, Tumor , Energy Metabolism/physiology , Gene Expression Regulation, Neoplastic/physiology , Humans , Lipid Metabolism/physiology , Nucleic Acids/metabolism
6.
Pathol Res Pract ; 221: 153337, 2021 May.
Article in English | MEDLINE | ID: mdl-33798911

ABSTRACT

BACKGROUND: Papillary thyroid microcarcinoma (PTM) belongs to papillary carcinomas whose length is about 1.0 cm. According to previous studies, FOXE1 is a transcription factor involved in the progression of papillary thyroid carcinoma (PTC). However, its detailed upstream mechanism remains unknown in PTM. OBJECTIVE: Our study aimed at detecting and verifying the up-regulation of FOXE1 in PTM cell lines. METHODS: FXOE1 expression was detected in PTM and normal cells through RT-qPCR. Loss-of-function experiments were conducted to identify the effect of silenced FOXE1 on cell proliferation, apoptosis, migration and invasion. Mechanism experiments were carried out to explore the upstream molecular mechanism of FOXE1. RESULTS: Knockdown of FOXE1 could lead to the inhibition on cell proliferation, migration and invasion while positively regulating cell apoptosis. Importantly, Yin-Yang-1 (YY1) could boost the transcription of FOXE1, thereby upregulating FOXE1. Also, the binding potential of miR-129-5p to FOXE1 was identified in PTM cells and MiR-129-5p could target FOXE1. In addition, the cellular processes in PTM were hindered with the increase of miR-129-5p expression level. CONCLUSION: Our research suggested that the up-regulation of FOXE1 is regulated by YY1 and miR-129-5p, which may contribute to PTM progression.


Subject(s)
Carcinoma, Papillary/pathology , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic/physiology , MicroRNAs/metabolism , Thyroid Neoplasms/pathology , YY1 Transcription Factor/metabolism , Cell Line, Tumor , Disease Progression , Humans , Up-Regulation
7.
Neurochem Res ; 45(12): 2894-2902, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32990800

ABSTRACT

Glioblastoma is one of the most frequent and aggressive primary tumor of glial brain tumors. Long non-coding RNA Prostate cancer-associated ncRNA transcript 6 (PCAT6) has been identified to influence the progression of many cancers, but its expression and functions in glioblastoma remain unclear. In this study, we intended to investigate the expression, functions and the corresponding mechanisms of PCAT6 in glioblastoma. We observed that PCAT6 expression was upregulated in glioblastoma tissues and cell lines and its high expression was due to the transcriptional activation by Yin Yang 1. miR-513 was a target of PCAT6 and Insulin like growth factor 2 mRNA binding protein 1 (IGF2BP1) was a target of miR-513. Hence, PCAT6 upregulated IGF2BP1 expression via miR-513 in a competing endogenous RNAs manner. PCAT6 and IGF2BP1 functioned as oncogenes while miR-513 acted as a tumor suppressor gene in glioblastoma. PCAT6 and miR-513 modulated the proliferation and survival of glioblastoma cells via AKT signaling by mediating IGF2BP1. IGF2BP1 raised the expression of PCAT6 by increasing its stability. In conclusion, our results indicate that PCAT6/miR-513/IGF2BP1 positive feedback loop plays a crucial role in facilitating glioblastoma progression.


Subject(s)
Glioblastoma/metabolism , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , RNA-Binding Proteins/metabolism , YY1 Transcription Factor/metabolism , Apoptosis/physiology , Cell Line, Tumor , Cell Proliferation/physiology , Disease Progression , Gene Expression Regulation, Neoplastic/physiology , Glioblastoma/genetics , Humans , RNA Stability/physiology , Up-Regulation
8.
Acta Histochem ; 122(6): 151576, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32778238

ABSTRACT

BACKGROUND: In recent years, microRNAs (miRNAs) are reported to act as molecular biomarkers for cancer diagnosis, treatment, and prognosis (including liver cancer) and to be involved in the development and progression of cancer and other physiological and pathological changes. However, the role of miR-34a-5p in liver cancer is still largely unknown. METHODS: In our study, the expression of miR-34a-5p in liver cancer tissues and HCC cell lines was detected by qRT-PCR. The CCK-8, scratch wound-healing motility and Transwell assays were used to evaluate the effect on cell proliferation, migration and invasion. The expression of YY1, E-cadherin, N-cadherin and vimentin was analysed by western blotting. The dual luciferase assay was performed to confirm whether YY1 is a target of miR-34a-5p. The combination of YY1 and MYCT1 was detected by chromatin immunoprecipitation (ChIP) assay. RESULTS: The results showed that miR-34a-5p was downregulated in liver cancer tissues and HCC cell lines. Overexpression of miR-34a-5p inhibited the proliferation, migration and invasion of liver cancer cells. YY1 was a direct target of miR-34a-5p, and the expression of YY1 could reverse the influence of miR-34a-5p on the proliferation, migration and invasion of liver cancer cells. YY1 inhibited MYCT1 expression by directly binding to its promoter region, and knockdown of MYCT1 reversed the influence of miR-34a-5p on the proliferation, migration and invasion of liver cancer cells. CONCLUSION: Our results suggest that miR-34a-5p could inhibit the invasion and metastasis of hepatoma cells by targeting YY1-mediated MYCT1 transcriptional repression.


Subject(s)
Liver Neoplasms/metabolism , Liver Neoplasms/pathology , YY1 Transcription Factor/metabolism , Blotting, Western , Cell Line, Tumor , Chromatin Immunoprecipitation , Disease Progression , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , Humans , Liver Neoplasms/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasm Metastasis/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Prognosis , Reverse Transcriptase Polymerase Chain Reaction , Sincalide/metabolism , Wound Healing/genetics , Wound Healing/physiology , YY1 Transcription Factor/genetics
9.
J Hematol Oncol ; 12(1): 103, 2019 10 17.
Article in English | MEDLINE | ID: mdl-31623653

ABSTRACT

BACKGROUND: Circular RNAs (circRNAs) represent a type of endogenous noncoding RNAs that are generated by back-splicing events and favor repetitive sequences. Recent studies have reported that cancer-associated chromosomal translocations could juxtapose distant complementary repetitive intronic sequences, resulting in the aberrant formation of circRNAs. However, among the reported fusion genes, only a small number of circRNAs were found to originate from fusion regions during gene translocation. We question if circRNAs could also originate from fusion partners during gene translocation. METHODS: Firstly, we designed divergent primers for qRT-PCR to identify a circRNA circAF4 in AF4 gene and investigated the expression pattern in different types of leukemia samples. Secondly, we designed two small interfering RNAs specially targeting the back-spliced junction point of circAF4 for functional studies. CCK8 cell proliferation and cell cycle assay were performed, and a NOD-SCID mouse model was used to investigate the contribution of circAF4 in leukemogenesis. Finally, luciferase reporter assay, AGO2 RNA immunoprecipitation (RIP), and RNA Fluorescent in Situ Hybridization (FISH) were performed to confirm the relationship of miR-128-3p, circAF4, and MLL-AF4 expression. RESULTS: We discovered a circRNA, named circAF4, originating from the AF4 gene, a partner of the MLL fusion gene in MLL-AF4 leukemia. We showed that circAF4 plays an oncogenic role in MLL-AF4 leukemia and promotes leukemogenesis in vitro and in vivo. More importantly, knockdown of circAF4 increases the leukemic cell apoptosis rate in MLL-AF4 leukemia cells, while no effect was observed in leukemia cells that do not carry the MLL-AF4 translocation. Mechanically, circAF4 can act as a miR-128-3p sponge, thereby releasing its inhibition on MLL-AF4 expression. We finally analyzed most of the MLL fusion genes loci and found that a number of circRNAs could originate from these partners, suggesting the potential roles of fusion gene partner-originating circRNAs (named as FP-circRNAs) in leukemia with chromosomal translocations. CONCLUSION: Our findings demonstrate that the abnormal elevated expression of circAF4 regulates the cell growth via the circAF4/miR-128-3p/MLL-AF4 axis, which could contribute to leukemogenesis, suggesting that circAF4 may be a novel therapeutic target of MLL-AF4 leukemia.


Subject(s)
Gene Expression Regulation, Neoplastic/physiology , Leukemia/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Oncogene Proteins, Fusion/metabolism , RNA, Circular/metabolism , Animals , Apoptosis , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Line , Cell Proliferation , Genetic Predisposition to Disease , Humans , Male , Mice , Mice, SCID , MicroRNAs/genetics , MicroRNAs/metabolism , Myeloid-Lymphoid Leukemia Protein/genetics , Neoplasms, Experimental , Oncogene Proteins, Fusion/genetics
10.
Integr Cancer Ther ; 18: 1534735419864434, 2019.
Article in English | MEDLINE | ID: mdl-31370719

ABSTRACT

Objective: To identify prognostic biomarkers and drugs that target them in colon adenocarcinoma (COAD) based on the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus databases. Methods: The TCGA dataset was used to identify the top 50 upregulated differentially expressed genes (DEGs), and Gene Expression Omnibus profiles were used for validation. Survival analyses were conducted with the TCGA dataset using the RTCGAToolbox package in the R software environment. Drugs targeting the candidate prognostic biomarkers were searched in the DrugBank and herbal databases. Results: Among the top 50 upregulated DEGs in patients with COAD in the TCGA dataset, the Wnt signaling pathway and cytokine-cytokine receptor interactions and pathways in cancer Kyoto Encyclopedia of Genes and Genomes pathway analysis were enriched in DEGs. Tissue development and regulation of cell proliferation were the main Gene Ontology biological processes associated with upregulated DEGs. MYC and KLK6 were overexpressed in tumors validated in the TCGA, GSE41328, and GSE113513 databases (all P < .001) and were significantly associated with overall survival in patients with COAD (P = .021 and P = .047). Nadroparin and benzamidine were identified as inhibitors of MYC and KLK6 in DrugBank, and 8 herbs targeting MYC, including Da Huang (Radix Rhei Et Rhizome), Hu Zhang (Polygoni Cuspidati Rhizoma Et Radix), Huang Lian (Coptidis Rhizoma), Ban Xia (Arum Ternatum Thunb), Tu Fu Ling (Smilacis Glabrae Rhixoma), Lei Gong Teng (Tripterygii Radix), Er Cha (Catechu), and Guang Zao (Choerospondiatis Fructus), were identified. Conclusion: MYC and KLK6 may serve as candidate prognostic predictors and therapeutic targets in patients with COAD.


Subject(s)
Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Biomarkers, Tumor/metabolism , Colon/metabolism , Colon/pathology , Colonic Neoplasms/pathology , Colonic Neoplasms/metabolism , Computational Biology/methods , Gene Expression Regulation, Neoplastic/physiology , Humans , Kallikreins/metabolism , Prognosis , Survival Analysis , Transcriptome/physiology , Up-Regulation/physiology , Wnt Signaling Pathway/physiology
11.
Hepatol Int ; 13(4): 440-453, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31250351

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is the second leading cause of cancer death worldwide. Chemotherapy is an alternative treatment for advanced HCCs, but chemo-resistance prevents cancer therapies from achieving stable and complete responses. Understanding the underlying mechanisms in chemo-resistance is critical to improve the efficacy of HCC. METHODS: The expression levels of Id-1 and CCN2 were detected in large cohorts of HCCs, and functional analyses of Id-1 and CCN2 were performed both in vitro and in vivo. cDNA microarrays were performed to evaluate the alterations of expression profiling of HCC cells with overexpression of CCN2. Finally, the role of downstream signaling of MAPK/Id-1 signaling pathway in oxaliplatin resistance were also explored. RESULTS: The increased expression of Id-1 and CCN2 were closely related to oxaliplatin resistance in HCC. Upregulation of CCN2 and Id-1 was independently associated with shorter survival and increased recurrence in HCC patients, and significantly enhanced oxaliplatin resistance and promoted lung metastasis in vivo, whereas knock-down of their expression significantly reversed the chemo-resistance and inhibited HCC cell stemness. cDNA microarrays and PCR revealed that Id-1 and MAPK pathway were the downstream signaling of CCN2. CCN2 significantly enhanced oxaliplatin resistance by activating the MAPK/Id-1 signaling pathway, and Id-1 could upregulate CCN2 in a positive feedback manner. CONCLUSIONS: CCN2/MAPK/Id-1 loop feedback amplification is involved in oxaliplatin resistance, and the combination of oxaliplatin with inhibitor of CCN2 or MAPK signaling could provide a promising approach to ameliorating oxaliplatin resistance in HCC.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Connective Tissue Growth Factor/metabolism , Liver Neoplasms/drug therapy , Oxaliplatin/therapeutic use , Adult , Aged , Animals , Biomarkers, Tumor/metabolism , Butadienes/pharmacology , Cell Line, Tumor , Down-Regulation/drug effects , Drug Resistance, Neoplasm , Feedback/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Heterografts , Humans , MAP Kinase Signaling System/drug effects , Male , Mice, Nude , Middle Aged , Mitogen-Activated Protein Kinase 1/metabolism , Neoplasm Recurrence, Local , Neoplasm Transplantation , Neoplastic Stem Cells/drug effects , Nitriles/pharmacology , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/metabolism , Sorafenib/pharmacology , Up-Regulation/drug effects
12.
J Cell Biochem ; 120(10): 18415-18424, 2019 10.
Article in English | MEDLINE | ID: mdl-31190375

ABSTRACT

Lacking of diagnostic and prognostic biomarkers is a significant reason for the poor prognosis of patients with triple-negative breast cancer (TNBC). MicroRNAs (miRNAs) have been discovered to engage in the tumorigenesis and development of TNBC. miR-5590-3p has been found to be involved in the development of gastric cancer, but its role and underlying mechanism in TNBC remain obscure. In this study, it was discovered that miR-5590-3p was downregulated in TNBC tissues and cells. Function assays confirmed that miR-5590-3p overexpression inhibited cell proliferation, migration, and epithelial-mesenchymal transition (EMT) process as well as promoted cell apoptosis in TNBC. Moreover, YY1 could bind with the promoter of miR-5590-3p and overexpression of YY1 inhibited the transcription of miR-5590-3p. It was found that YY1 acted as a downstream target gene to bind with miR-5590-3p and was negatively regulated by miR-5590-3p. Finally, it was discovered that overexpression of YY1 could partially rescue the miR-5590-3p overexpression-mediated inhibitive effect on TNBC progression. Taken together these results, it can be concluded that miR-5590-3p-YY1 feedback loop promoted the proliferation and migration of TNBC.


Subject(s)
Cell Movement/physiology , Cell Proliferation/physiology , MicroRNAs/metabolism , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , YY1 Transcription Factor/metabolism , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Chromatin Immunoprecipitation , Female , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Triple Negative Breast Neoplasms/pathology , YY1 Transcription Factor/genetics
13.
Nat Commun ; 10(1): 663, 2019 02 08.
Article in English | MEDLINE | ID: mdl-30737378

ABSTRACT

The biological role of miR-500a-5p has not yet been reported in the context of colorectal cancer (CRC). Here, we show that miR-500a-5p expression is decreased in CRC tissues compared with adjacent normal tissues. Low miR-500a-5p expression is associated with malignant progression. Moreover, transfection of CRC cells with miR-500a-5p induces G0/G1 cell cycle arrest and inhibits their growth and migration. Mechanistically, miR-500a-5p directly targets HDAC2 and inhibits HDAC2-mediated proliferation in CRC in nude mice. Furthermore, YY1 binds to the promoter of miR-500a-5p and negatively regulates its transcription. Restoration of miR-500a-5p expression is up-regulated via the p300/YY1/HDAC2 complex. Besides, therapeutic delivery of miR-500a-5p significantly suppresses tumour development in a xenograft tumour model and a HDAC2 inhibitor FK228-treated CRC model. Our studies demonstrate that miR-500a-5p functions as a tumour suppressor in CRC by targeting the p300/YY1/HDAC2 axis, which contributes to the development of and provides new potential candidates for CRC therapy.


Subject(s)
Colorectal Neoplasms/metabolism , E1A-Associated p300 Protein/metabolism , Histone Deacetylase 2/metabolism , MicroRNAs/metabolism , YY1 Transcription Factor/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Cell Line , Cell Line, Tumor , Cell Proliferation/genetics , Cell Proliferation/physiology , Colorectal Neoplasms/genetics , E1A-Associated p300 Protein/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , HCT116 Cells , Histone Deacetylase 2/genetics , Humans , In Situ Nick-End Labeling , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Signal Transduction/genetics , Signal Transduction/physiology , YY1 Transcription Factor/genetics
14.
Int J Cancer ; 142(7): 1392-1404, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29168185

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is one of the malignant lethal tumors. It has been reported that the transcriptional regulator Yin Yang-1 (YY1) suppressed the invasion and metastasis of PDAC. However, the function of YY1 on proliferation and migration of pancreatic cancer remains to be clarified. In this study, we found that YY1 overexpression or knockdown can inhibit or promote the proliferation and migration of pancreatic cancer cells. Digital gene expression sequencing indicates that cyclin-dependent kinase inhibitor 3 (CDKN3) may be the candidate target gene of YY1. Then we found that YY1 can downregulate the expression of CDKN3 by directly binding to the promoter region of CDKN3. Silencing CDKN3 expression could inhibit the ability of cell proliferation and migration and overexpression of CDKN3 could restore the effects induced by YY1 overexpression in pancreatic cancer cells. The expression levels of YY1 and CDKN3 were negatively correlated in pancreatic cancer tissues and PDAC patients with higher levels of CDKN3 have poor prognosis. Vitro and vivo study show that CDKN3 can form a complex with MdM2-P53, thus leading to inhibiting the expression of P21, which is the target gene of P53, and finally facilitates the cell cycle to promote the proliferation of pancreatic cancer cells. Hence, YY1 can directly regulate the expression of CDKN3 and participate in the cycle of pancreatic cancer cells, which can inhibit the progression of pancreatic cancer. These results reveal that YY1-CDKN3-MDM2/P53-P21 axis is involved in pancreatic tumorigenesis, which may develop new methods for human pancreatic cancer therapy.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Gene Expression Regulation, Neoplastic/physiology , Pancreatic Neoplasms/pathology , Signal Transduction/physiology , YY1 Transcription Factor/metabolism , Animals , Cell Movement , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor Proteins/metabolism , Dual-Specificity Phosphatases/metabolism , Heterografts , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Proto-Oncogene Proteins c-mdm2/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Tumor Suppressor Protein p53/metabolism
15.
PLoS One ; 12(8): e0182149, 2017.
Article in English | MEDLINE | ID: mdl-28787001

ABSTRACT

Cancer stem cell survival is the leading factor for tumor recurrence after tumor-suppressive treatments. Therefore, specific and efficient inhibitors of cancer stemness must be discovered for reducing tumor recurrence. YM155 has been indicated to significantly reduce stemness-derived tumorsphere formation. However, the pharmaceutical mechanism of YM155 against cancer stemness is unclear. This study investigated the potential mechanism of YM155 against cancer stemness in lung cancer. Tumorspheres derived from epidermal growth factor receptor (EGFR)-mutant HCC827 and EGFR wild-type A549 cells expressing higher cancer stemness markers (CD133, Oct4, and Nanog) were used as cancer stemness models. We observed that EGFR autophosphorylation (Y1068) was higher in HCC827- and A549-derived tumorspheres than in parental cells; this autophosphorylation induced tumorsphere formation by activating G9a-mediated stemness. Notably, YM155 inhibited tumorsphere formation by blocking the autophosphorylation of EGFR and the EGFR-G9a-mediated stemness pathway. The chemical and genetic inhibition of EGFR and G9a revealed the significant role of the EGFR-G9a pathway in maintaining the cancer stemness property. In conclusion, this study not only revealed that EGFR could trigger tumorsphere formation by elevating G9a-mediated stemness but also demonstrated that YM155 could inhibit this formation by simultaneously blocking EGFR autophosphorylation and G9a activity, thus acting as a potent agent against lung cancer stemness.


Subject(s)
Antineoplastic Agents/pharmacology , ErbB Receptors/metabolism , Histocompatibility Antigens/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Imidazoles/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Naphthoquinones/pharmacology , Afatinib , Cell Line, Tumor , Drug Evaluation, Preclinical , ErbB Receptors/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Methylation/drug effects , Octamer Transcription Factor-3/metabolism , Phosphorylation/drug effects , Phosphorylation/physiology , Quinazolines/pharmacology , RNA, Messenger/metabolism
16.
Vet Pathol ; 54(2): 212-217, 2017 03.
Article in English | MEDLINE | ID: mdl-27698080

ABSTRACT

The expression of tyrosine kinase receptors is attracting major interest in human and veterinary oncological pathology because of their role as targets for adjuvant therapies. Little is known about tyrosine kinase receptor (TKR) expression in canine liposarcoma (LP), a soft tissue sarcoma. The aim of this study was to evaluate the immunohistochemical expression of the TKRs fibroblast growth factor receptor 1 (FGFR1) and platelet-derived growth factor receptor-ß (PDGFRß); their ligands, fibroblast growth factor 2 (FGF2) and platelet-derived growth factor B (PDGFB); and c-kit in canine LP. Immunohistochemical labeling was categorized as high or low expression and compared with the mitotic count and MIB-1-based proliferation index. Fifty canine LPs were examined, classified, and graded. Fourteen cases were classified as well differentiated, 7 as myxoid, 25 as pleomorphic, and 4 as dedifferentiated. Seventeen cases were grade 1, 26 were grade 2, and 7 were grade 3. A high expression of FGF2, FGFR1, PDGFB, and PDGFRß was identified in 62% (31/50), 68% (34/50), 81.6% (40/49), and 70.8% (34/48) of the cases, respectively. c-kit was expressed in 12.5% (6/48) of the cases. Mitotic count negatively correlated with FGF2 ( R = -0.41; P < .01), being lower in cases with high FGF2 expression, and positively correlated with PDGFRß ( R = 0.33; P < .01), being higher in cases with high PDGFRß expression. No other statistically significant correlations were identified. These results suggest that the PDGFRß-mediated pathway may have a role in the progression of canine LP and may thus represent a promising target for adjuvant cancer therapies.


Subject(s)
Dog Diseases/metabolism , Fibroblast Growth Factor 2/metabolism , Liposarcoma/veterinary , Proto-Oncogene Proteins c-sis/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Animals , Dog Diseases/pathology , Dogs , Fibroblast Growth Factor 2/genetics , Gene Expression Regulation, Enzymologic/physiology , Gene Expression Regulation, Neoplastic/physiology , Liposarcoma/metabolism , Proto-Oncogene Proteins c-sis/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Platelet-Derived Growth Factor beta/genetics
17.
Neuroendocrinology ; 105(1): 54-66, 2017.
Article in English | MEDLINE | ID: mdl-27442441

ABSTRACT

Cancer immunotherapy is becoming a cornerstone in the clinical care of cancer patients due to the breakthrough trials with immune checkpoint blockade antibodies and chimeric antigen receptor T cells. The next breakthrough in cancer immunotherapy is likely to be oncolytic viruses engineered to selectively kill tumor cells and deceive the immune system to believe that the tumor is a foreign entity that needs to be eradicated. We have developed AdVince, an oncolytic adenovirus for treatment of liver metastases from neuroendocrine tumor (NET). AdVince includes the gene promoter from human chromogranin A for selective replication in neuroendocrine cells, miR122 target sequences for reduced liver toxicity, and a cell-penetrating peptide in the capsid for increased infectivity of tumor cells and optimized spread within tumors. This paper describes the preclinical evaluation of AdVince on freshly isolated human gastrointestinal NET cells resected from liver metastases and freshly isolated human hepatocytes as well as in fresh human blood. AdVince selectively replicates in and kills NET cells. Approximately 73-fold higher concentration of AdVince is needed to induce a similar level of cytotoxicity in NET cells as in hepatocytes. AdVince did not activate complement or induce considerable amount of proinflammatory cytokines or chemokines in human blood. The data presented herein indicate that AdVince can be safely evaluated in a phase I/IIa clinical trial for patients with liver-dominant NET.


Subject(s)
Carcinoma, Neuroendocrine/pathology , Gene Expression Regulation, Neoplastic/physiology , Liver Neoplasms/secondary , Liver Neoplasms/therapy , Neuroendocrine Tumors/pathology , Oncolytic Virotherapy/methods , Carcinoma, Neuroendocrine/blood , Cell Line, Tumor , Cytokines/genetics , Cytokines/metabolism , Drug Evaluation, Preclinical , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hepatocytes/drug effects , Hepatocytes/virology , Humans , Liver Neoplasms/blood , Neuroendocrine Tumors/blood , Oncolytic Viruses/physiology , Receptors, Complement/metabolism , Time Factors
18.
Cancer Res ; 77(1): 100-111, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27793842

ABSTRACT

KRAS activation occurring in more than 90% of pancreatic ductal adenocarcinomas (PDAC) drives progression and metastasis, but the underlying mechanisms involved in these processes are still poorly understood. Here, we show how KRAS acts through inflammatory NF-κB signaling to activate the transcription factor YY1, which represses expression of the tumor suppressor gene miR-489. In PDAC cells, repression of miR-489 by KRAS signaling inhibited migration and metastasis by targeting the extracellular matrix factors ADAM9 and MMP7. miR-489 downregulation elevated levels of ADAM9 and MMP7, thereby enhancing the migration and metastasis of PDAC cells. Together, our results establish a pivotal mechanism of PDAC metastasis and suggest miR-489 as a candidate therapeutic target for their attack. Cancer Res; 77(1); 100-11. ©2016 AACR.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Gene Expression Regulation, Neoplastic/physiology , MicroRNAs/metabolism , Pancreatic Neoplasms/pathology , Signal Transduction/physiology , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Heterografts , Humans , Mice , Mice, Inbred NOD , Mice, SCID , MicroRNAs/genetics , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Polymerase Chain Reaction , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Transcriptome , YY1 Transcription Factor/genetics , YY1 Transcription Factor/metabolism
19.
PLoS One ; 11(11): e0165830, 2016.
Article in English | MEDLINE | ID: mdl-27824899

ABSTRACT

The effects of sarcosine on the processes driving prostate cancer (PCa) development remain still unclear. Herein, we show that a supplementation of metastatic PCa cells (androgen independent PC-3 and androgen dependent LNCaP) with sarcosine stimulates cells proliferation in vitro. Similar stimulatory effects were observed also in PCa murine xenografts, in which sarcosine treatment induced a tumor growth and significantly reduced weight of treated mice (p < 0.05). Determination of sarcosine metabolism-related amino acids and enzymes within tumor mass revealed significantly increased glycine, serine and sarcosine concentrations after treatment accompanied with the increased amount of sarcosine dehydrogenase. In both tumor types, dimethylglycine and glycine-N-methyltransferase were affected slightly, only. To identify the effects of sarcosine treatment on the expression of genes involved in any aspect of cancer development, we further investigated expression profiles of excised tumors using cDNA electrochemical microarray followed by validation using the semi-quantitative PCR. We found 25 differentially expressed genes in PC-3, 32 in LNCaP tumors and 18 overlapping genes. Bioinformatical processing revealed strong sarcosine-related induction of genes involved particularly in a cell cycle progression. Our exploratory study demonstrates that sarcosine stimulates PCa metastatic cells irrespectively of androgen dependence. Overall, the obtained data provides valuable information towards understanding the role of sarcosine in PCa progression and adds another piece of puzzle into a picture of sarcosine oncometabolic potential.


Subject(s)
Cell Cycle/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Genes, Neoplasm/physiology , Prostatic Neoplasms/metabolism , Sarcosine/pharmacology , Animals , Cell Cycle/physiology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/physiology , Glycine N-Methyltransferase/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Polymerase Chain Reaction , Prostatic Neoplasms/physiopathology , Sarcosine/metabolism , Sarcosine Dehydrogenase/metabolism , Transcriptome , Up-Regulation
20.
Oncotarget ; 6(41): 43182-201, 2015 Dec 22.
Article in English | MEDLINE | ID: mdl-26673823

ABSTRACT

Despite intensive study, many mysteries remain about the MYCN oncogene's functions. Here we focus on MYCN's role in neuroblastoma, the most common extracranial childhood cancer. MYCN gene amplification occurs in 20% of cases, but other recurrent somatic mutations are rare. This scarcity of tractable targets has hampered efforts to develop new therapeutic options. We employed a multi-level omics approach to examine MYCN functioning and identify novel therapeutic targets for this largely un-druggable oncogene. We used systems medicine based computational network reconstruction and analysis to integrate a range of omic techniques: sequencing-based transcriptomics, genome-wide chromatin immunoprecipitation, siRNA screening and interaction proteomics, revealing that MYCN controls highly connected networks, with MYCN primarily supressing the activity of network components. MYCN's oncogenic functions are likely independent of its classical heterodimerisation partner, MAX. In particular, MYCN controls its own protein interaction network by transcriptionally regulating its binding partners.Our network-based approach identified vulnerable therapeutically targetable nodes that function as critical regulators or effectors of MYCN in neuroblastoma. These were validated by siRNA knockdown screens, functional studies and patient data. We identified ß-estradiol and MAPK/ERK as having functional cross-talk with MYCN and being novel targetable vulnerabilities of MYCN-amplified neuroblastoma. These results reveal surprising differences between the functioning of endogenous, overexpressed and amplified MYCN, and rationalise how different MYCN dosages can orchestrate cell fate decisions and cancerous outcomes. Importantly, this work describes a systems-level approach to systematically uncovering network based vulnerabilities and therapeutic targets for multifactorial diseases by integrating disparate omic data types.


Subject(s)
Genes, myc/physiology , Neuroblastoma/genetics , Nuclear Proteins/physiology , Oncogene Proteins/physiology , Protein Interaction Maps/physiology , Blotting, Western , Chromatin Immunoprecipitation , Computational Biology/methods , Gene Expression Regulation, Neoplastic/physiology , High-Throughput Nucleotide Sequencing/methods , Humans , N-Myc Proto-Oncogene Protein , Neuroblastoma/metabolism , Neuroblastoma/pathology , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Proteomics/methods , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL