Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
Gene ; 813: 146092, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-34896523

ABSTRACT

BACKGROUND: Diffuse large B-cell lymphoma (DLBCL) is the main pathological type of non-Hodgkin lymphoma (NHL). Chromobox (CBX) family proteins are classical components of polycomb group (PcG) complexes in many cancer types, resulting in accelerated carcinogenesis. Nevertheless, the prognostic, functional and expression significance of these CBX family members in DLBCL remain unclear and elusive. METHODS: CBX transcriptional levels were confirmed using Oncomine, Gene Expression Profiling Interactive Analysis (GEPIA) and Cancer Cell Line Encyclopedia (CCLE) databases. The protein levels of CBX family members were analysed using The Human Protein Atlas (HPA) database. Information on the PPI network, functional enrichment, drug sensitivity, prognostic value, miRNA network, protein structure, genetic alteration and immune cell infiltration were generated using the GeneMANIA, Metascape, GSCALite, GEPIA, PDB, cBioPortal, and TIMER databases, and the correlation of these factors with CBX expression levels in DLBCL was assessed. RESULTS: CBX1/2/3/5/6/8 mRNA levels were significantly enhanced in DLBCL tissues compared to corresponding normal tissues. CBX1/3/4/5/8 protein expression levels were obviously increased, whereas CBX7 was obviously decreased. This difference might be attributed to miRNA regulation based on the miRNA network. Overall survival (OS) analysis showed that CBX levels were not correlated with prognosis in DLBCL patients, indicating that CBXs are not good biomarkers for DLBCL patients. Furthermore, functional enrichment analyses indicated that CBXs were closely related to DNA duplex unwinding, covalent chromatin modification, and histone lysine methylation. The levels of CBXs were also significantly associated with diverse immune cell infiltration in DLBCL. CONCLUSIONS: This study reveals that dysregulated CBXs are involved in DLBCL development and might represent potential therapeutic targets for DLBCL.


Subject(s)
Chromobox Protein Homolog 5/genetics , Lymphoma, Large B-Cell, Diffuse/genetics , Polycomb-Group Proteins/genetics , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Chromobox Protein Homolog 5/biosynthesis , Chromobox Protein Homolog 5/metabolism , Databases, Genetic , Databases, Protein , Gene Expression , Humans , Lymphoma, Large B-Cell, Diffuse/metabolism , Mutation , Polycomb Repressive Complex 1/biosynthesis , Polycomb Repressive Complex 1/genetics , Polycomb-Group Proteins/biosynthesis , Polycomb-Group Proteins/metabolism , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Survival Analysis , Transcriptome
2.
Medicine (Baltimore) ; 100(34): e27056, 2021 Aug 27.
Article in English | MEDLINE | ID: mdl-34449496

ABSTRACT

ABSTRACT: Chromobox homolog 8 (CBX8) plays an important role in the occurrence and development of various tumors, and is closely related to the prognosis of patients with cancer. However, the occurrence, development, and prognostic value of CBX8 in cervical cancer have not been reported yet.In this study, immunohistochemistry was used to detect the expression of CBX8 in cervical cancer tissues and the corresponding normal tissues adjacent to the tumor. Furthermore, the relationship between CBX8 and programmed death-ligand 1 (PD-L1) expression, clinicopathological characteristics, and prognosis of cervical cancer were explored, and the prognostic value of CBX8 in cervical cancer was clarified.In this study, the results of immunohistochemistry using tissue chips obtained from patients with cervical cancer showed that CBX8 was highly expressed in cervical cancer tissues, and its expression was proportional to the international federation of gynecology and obstetrics (FIGO) stage. Disease-free and overall survival of patients with high CBX8 expression in cervical cancer were significantly shorter than those of patients with low CBX8 expression. Thus, CBX8 was found to be an independent prognostic factor for patients with cervical cancer. In addition, CBX8 and PD-L1 co-expression model could better predict the prognosis of patients with cervical cancer, and its area under the receiver operating characteristic curve was similar to that of FIGO stage.CBX8 may be an independent prognostic factor for cervical cancer. Moreover, the CBX8 and PD-L1 co-expression model could predict the postoperative survival of patients with cervical cancer objectively and reliably, which will aid clinicians to shunt patients with cervical cancer based on the risk of death, develop a reasonable treatment plan, and provide personalized prognosis.


Subject(s)
B7-H1 Antigen/biosynthesis , Polycomb Repressive Complex 1/biosynthesis , Uterine Cervical Neoplasms/mortality , Uterine Cervical Neoplasms/pathology , Adult , Female , Humans , Kaplan-Meier Estimate , Middle Aged , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Staging , Prognosis , Survival Analysis
3.
Mol Med Rep ; 23(2)2021 02.
Article in English | MEDLINE | ID: mdl-33313949

ABSTRACT

Gastric cancer (GC) is the most common and fast­growing malignancy of the digestive system, which has a high mortality. Chromobox homolog 2 (CBX2) has been reported to be highly expressed in cancer tissues compared with adjacent normal tissues. It has also been established that CBX2 is upregulated in GC cell lines by searching the Cancer Cell Line Encyclopedia. The aim of the present study was to investigate the biomolecular role and underlying mechanism of CBX2 in the proliferation, invasion and migration of GC cells. Short hairpin RNA­CBX2 and yes­associated protein (YAP) overexpression plasmids were constructed to regulate CBX2 and YAP expression, respectively. Additionally, the expression of certain mRNAs and proteins involved in the YAP/ß­catenin pathway and those associated with cell invasion were assessed by western blotting and reverse transcription­quantitative PCR, respectively. The cellular behaviors of MFC cells were analyzed using Cell Counting Kit­8, colony formation wound­healing and Transwell assays. The results of the present study revealed that increased CBX2 expression was observed in GC cell lines compared with normal gastric cells. In addition, CBX2 knockdown inhibited the nuclear cytoplasm translocation of YAP, inducing its phosphorylation, and suppressing the activation of the ß­catenin signaling pathway. The results also demonstrated that CBX2 depletion inhibited the proliferation, migration and invasion of GC cells by inactivating the YAP/ß­catenin pathway. It was determined that CBX2 promoted the proliferation, invasion and migration of GC cells by activating the YAP/ß­catenin pathway, suggesting that CBX2 is involved in the pathogenesis of GC and may represent a novel target for the clinical treatment of GC.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Polycomb Repressive Complex 1/biosynthesis , Polycomb Repressive Complex 1/genetics , Stomach Neoplasms/genetics , beta Catenin/metabolism , Animals , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Mice , Phosphorylation/genetics , Stomach Neoplasms/metabolism , Up-Regulation , YAP-Signaling Proteins
4.
Int J Mol Med ; 47(1): 137-150, 2021 01.
Article in English | MEDLINE | ID: mdl-33236134

ABSTRACT

Overproduction of pro­inflammatory cytokines in the aged, which is called inflammaging, leads to the deterioration of periodontitis. Toll­like receptor 4 (TLR4) plays a role in the regulation of cellular senescence, and its expression increases with age. However, there has been limited research into the molecular mechanisms underlying the onset of periodontal inflammaging, and the interplay between TLR4 and inflammaging. In the present study, wild­type and TLR4 gene knockout mice were used to investigate the activation of the TLR4 pathway in mouse periodontitis and the expression of the nucleotide­binding and oligomerization domain­like receptor 3 (NLRP3) inflammasome, an upstream immune checkpoint during the development of inflammaging. Activation of TLR4 in a mouse model of periodontitis enhanced the expression of a senescence­associated secretory phenotype (SASP), which boosted the inflammaging process. Conversely, TLR4 activation downregulated the expression of B cell­specific Moloney murine leukemia virus integration site 1 (Bmi­1) and promoted the priming of NLRP3 inflammasome, both of which are regulators of SASP. Treating gingival fibroblasts with Bmi­1 inhibitor PTC209, it was demonstrated that TLR4 activated the NLRP3 pathway and the inflammaging process by suppressing Bmi­1. In addition, there was a significant reduction in the expression of Bmi­1 expression in the gingiva of patients with periodontitis compared with healthy controls. In conclusion, the present study demonstrated that TLR4 acted by inhibiting Bmi­1 to enhance the NLRP3 pathway and SASP factors. This cascade of reactions may contribute to the senescence of the periodontium.


Subject(s)
Gene Expression Regulation , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Periodontitis/metabolism , Polycomb Repressive Complex 1/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Toll-Like Receptor 4/metabolism , Animals , Female , Inflammasomes/genetics , Inflammation/genetics , Inflammation/metabolism , Male , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Periodontitis/genetics , Polycomb Repressive Complex 1/genetics , Proto-Oncogene Proteins/genetics , Toll-Like Receptor 4/genetics
5.
Breast Cancer Res Treat ; 182(3): 581-589, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32524353

ABSTRACT

PURPOSE: BMI1, which is a major component of the polycomb group complex 1, is an essential epigenetic repressor of multiple regulatory genes and has been identified as a cancer stem cell (CSC) marker in several cancers. However, its role in breast cancer (BC) remains to be defined. In this study, we have evaluated the prognostic significance of BMI1 among the different molecular subtypes and assessed its association with other breast CSC markers (BCSC). MATERIAL AND METHOD: BMI1 copy number and mRNA was assessed in large and well-characterised cohorts of early-stage BC patients [METABRIC (n = 1980) and the Bc-GenExMiner (n = 9616) databases]. BMI1 protein expression was assessed using tissue microarray and immunohistochemistry in a cohort of 870 invasive BC patients with long-term outcome data and the expression of a panel of BCSC markers was monitored. RESULT: BMI1 expression, prognostic significance and its association with BCSC markers were differed between molecular classes. In the luminal oestrogen receptor-positive (ER+) BC, BMI1 showed significantly higher expression compared to ER- tumours. BMI1 showed positive correlation with favourable prognostic features and it was negatively associated with the expression of key BCSC markers (ALDH1A1, CD24, CD44, CD133, SOX10 and SOX9). High expression of BMI1 was associated with longer breast cancer-specific survival (BCSS) independent of other prognostic variables. In the basal triple negative BC subtype, BMI1 expression showed positive association with CD133 and SOX10 and it was significantly associated with shorter BCSS. CONCLUSION: High BMI1 expression is associated with clinicopathological variables and outcome in BC. However, this association is dependent on the molecular subtypes. Further functional assessment to detect its underlying mechanistic roles in BC subtypes is warranted.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Neoplastic Stem Cells/pathology , Polycomb Repressive Complex 1/metabolism , Biomarkers, Tumor/metabolism , Breast Neoplasms/surgery , Female , Follow-Up Studies , Humans , Middle Aged , Neoplasm Invasiveness , Neoplastic Stem Cells/metabolism , Polycomb Repressive Complex 1/biosynthesis , Prognosis , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Retrospective Studies , Survival Rate
6.
Biomed Pharmacother ; 121: 109602, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31707349

ABSTRACT

Esophageal squamous cell carcinoma (ESCC) is one of the most common cancers in China, accompanied by an extremely high mortality rate. Chlorogenic acid (CGA) is a small-molecule compound, that has been shown to have a wide range of biological activities, including antitumor. However, the efficacy and molecular mechanism of CGA on ESCC remains unknown. In this study, we confirmed the inhibition of proliferation by CGA in ESCC cells, as well as the reduction of ESCC xenograft volume by CGA in vivo. In addition, CGA also suppressed both the migration and invasion of ESCC cells in vitro. In a carcinogen-induced murine model of ESCC, hyperplasia of the esophagus was slowed by CGA, while mice suffering from ESCC that were treated with CGA had longer survival times than mice in the control group. The measurement of pluripotency factors (BMI1, SOX2, OCT4 and Nanog) that are related to poor prognosis revealed reduced expression of both BMI1 and SOX2, but not of OCT4 or Nanog, in ESCC cells, in both a dose- and time-dependent manner. Together, our initial findings demonstrate that CGA suppresses ESCC progression, downregulates the expression of BMI1 and SOX2, and provide an anti-tumor candidate for ESCC therapy.


Subject(s)
Chlorogenic Acid/therapeutic use , Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/metabolism , Growth Inhibitors/therapeutic use , Polycomb Repressive Complex 1/biosynthesis , Proto-Oncogene Proteins/biosynthesis , SOXB1 Transcription Factors/biosynthesis , Animals , Cell Line , Cell Line, Tumor , Chlorogenic Acid/pharmacology , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Down-Regulation/physiology , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/genetics , Esophageal Squamous Cell Carcinoma/drug therapy , Esophageal Squamous Cell Carcinoma/genetics , Gene Expression Regulation, Neoplastic , Growth Inhibitors/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Polycomb Repressive Complex 1/antagonists & inhibitors , Polycomb Repressive Complex 1/genetics , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , SOXB1 Transcription Factors/antagonists & inhibitors , SOXB1 Transcription Factors/genetics
7.
Biomed Res Int ; 2019: 6436825, 2019.
Article in English | MEDLINE | ID: mdl-31211140

ABSTRACT

BACKGROUND: The chromobox (CBX) proteins CBX2, CBX4, CBX6, CBX7, and CBX8, also known as Polycomb (Pc) proteins, are canonical components of the Polycomb repressive complex 1 (PRC1). Abundant evidence indicates that abnormal expression of Pc proteins is associated with a variety of tumors, but their role in the pathogenesis of hepatocellular carcinoma (HCC) has not been fully elucidated. In the present study, we performed a case-control study to investigate the relationship between single nucleotide polymorphisms (SNPs) of CBX genes and HCC. METHODS: Nine SNPs on CBX genes (rs7217395, rs2036316 of CBX2; rs3764374, rs1285251, rs2289728 of CBX4; rs7292074 of CBX6; and rs710190, rs139394, rs5750753 of CBX7) were screened and genotyped using MassARRAY technology in 334 HCC cases and 321 controls. The association between SNPs and their corresponding gene expressions was analyzed through bioinformatics methods using the Ensembl database and Blood eQTL browser online tools. RESULTS: The results indicated that rs2289728 (G>A) of CBX4 (P = 0.03, OR = 0.56, 95% CI: 0.33-0.94) and rs139394 (C>A) of CBX7 (P = 0.02, OR = 0.55, 95% CI: 0.33-0.90) decreased the risk of HCC. Interaction between rs2036316 and HBsAg increased the risk of HCC (P = 0.02, OR = 6.88, 95% CI: 5.20-9.11), whereas SNP-SNP interaction between rs710190 and rs139394 reduced the risk of HCC (P = 0.03, OR = 0.33, 95% CI: 0.12-0.91). Gene expression analyses showed that the rs2289728 A allele and the rs139394 A allele significantly reduced CBX4 and CBX7 expression, respectively. CONCLUSION: Our findings suggest that CBX4 rs2289728 and CBX7 rs139394 are protective SNPs against HCC. The two SNPs may reduce the risk of HCC while suppressing the expression of CBX4 and CBX7.


Subject(s)
Carcinoma, Hepatocellular/genetics , Gene Expression Regulation, Neoplastic , Ligases/genetics , Liver Neoplasms/genetics , Neoplasm Proteins/genetics , Polycomb Repressive Complex 1/genetics , Polycomb-Group Proteins/genetics , Polymorphism, Single Nucleotide , Adult , Alleles , Carcinoma, Hepatocellular/epidemiology , Carcinoma, Hepatocellular/metabolism , Female , Humans , Ligases/biosynthesis , Liver Neoplasms/epidemiology , Liver Neoplasms/metabolism , Male , Middle Aged , Neoplasm Proteins/biosynthesis , Polycomb Repressive Complex 1/biosynthesis , Polycomb-Group Proteins/biosynthesis , Risk Factors
8.
Dev Biol ; 450(1): 34-46, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30851270

ABSTRACT

Many organisms both undergo dramatic morphological changes during post-embryonic development and also regenerate lost structures, but the roles of epigenetic regulators in such processes are only beginning to be understood. In the present study, the functions of two histone modifiers were examined during metamorphosis and larval limb regeneration in the red flour beetle Tribolium castaneum. Polycomb (Pc), a member of Polycomb repressive complex 1 (PRC1), and Enhancer of zeste (E(z)), a member of Polycomb repressive complex 2 (PRC2), were silenced in larvae using RNA interference. In the absence of Pc, the head appendages of adults transformed into a leg-like morphology, and the legs and wings assumed a metathoracic identity, indicating that Pc acts to specify proper segmental identity. Similarly, silencing of E(z) led to homeotic transformation of legs and wings. Additional defects were also observed in limb patterning as well as eye morphogenesis, indicating that PcG proteins play critical roles in imaginal precursor cells. In addition, larval legs and antennae failed to re-differentiate when either Pc or E(z) was knocked down, indicating that histone modification is necessary for proper blastema growth and differentiation. These findings indicate that PcG proteins play extensive roles in postembryonic plasticity of imaginal precursor cells.


Subject(s)
Hindlimb/physiology , Insect Proteins/biosynthesis , Morphogenesis , Polycomb Repressive Complex 1/biosynthesis , Polycomb Repressive Complex 2/biosynthesis , Regeneration , Tribolium/metabolism , Animals , Insect Proteins/genetics , Larva/genetics , Larva/metabolism , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 2/genetics , Tribolium/genetics
9.
Tumour Biol ; 41(3): 1010428319840473, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30915904

ABSTRACT

Human papillomavirus is detected in over 50% of oropharyngeal squamous cell carcinomas. Human papillomavirus-positive oropharyngeal squamous cell carcinomas differ from human papillomavirus-negative tumors, and both expression patterns are classified as distinct entities. The Bmi-1 oncogene is a well-known member of the mammalian polycomb-group family. HESC5:3 and HES77 are newly developed monoclonal antibodies produced against undifferentiated embryonic stem cells. Our aim was to explore their roles in both human papillomavirus-positive and -negative oropharyngeal squamous cell carcinomas. Our cohort comprised 202 consecutive oropharyngeal squamous cell carcinoma patients diagnosed and treated with curative intent. We used tissue microarray tumor blocks to study the immunohistochemical expression of Bmi-1, HESC5:3, and HES77. We compared the expressions of these stem cell markers with p16 immunoexpression and human papillomavirus status, as well as with other characteristics of the tumor, and with patients' clinical data and follow-up data. Human papillomavirus- and p16-positive tumors expressed less Bmi-1 and more HESC5:3 than the negative tumors. HES77 expression was high in human papillomavirus-positive oropharyngeal squamous cell carcinoma, but it did not correlate with p16 positivity. In our multivariable model, Bmi-1 and HESC5:3 were still associated with human papillomavirus, but the association between human papillomavirus and HES77 remained absent. In conclusion, Bmi-1, HESC5:3, and HES77 may have a different role in human papillomavirus-positive and human papillomavirus-negative tumors. There was no correlation between Bmi-1, HESC5:3, and HES77 expression and survival.


Subject(s)
Carcinoma, Squamous Cell/pathology , Cyclin-Dependent Kinase Inhibitor Proteins/biosynthesis , Oropharyngeal Neoplasms/pathology , Oropharyngeal Neoplasms/virology , Papillomaviridae/isolation & purification , Polycomb Repressive Complex 1/biosynthesis , Antibodies, Monoclonal/immunology , Carcinoma, Squamous Cell/virology , Cyclin-Dependent Kinase Inhibitor Proteins/genetics , Epithelial-Mesenchymal Transition/genetics , Humans , Papillomavirus Infections/virology
10.
Cell Death Dis ; 10(2): 52, 2019 01 18.
Article in English | MEDLINE | ID: mdl-30718464

ABSTRACT

Polycomb group family is a class of proteins that have important roles in both physiological and pathological processes, and its family member Chromobox homolog 8 (CBX8) regulates cell differentiation, aging, and cell cycle progression in numerous carcinomas; however, the effects and underlying mechanisms of CBX8 in hepatocellular carcinoma (HCC) are rarely reported. We found that CBX8 expression in clinical HCC specimens correlates inversely with patient survival. In HCC cells, we found that enforced overexpression of CBX8 induces epithelial-mesenchymal transition, invasive migration, and stem cell-like traits, which are associated with increased tumor growth and metastasis in mice. Conversely, CBX8 silencing inhibits the aggressive phenotype of HCC cells that have high CBX8 expression. Mechanistically, CBX8 modulates H3K27me3 in the gene promoter of bone morphogenetic protein 4 (BMP4), which is associated with active BMP4 transcription and, consequently, the activation of Smads and mitogen-activated protein kinases. BMP4 expression reverses the effects of CBX8 silencing in inhibiting epithelial-mesenchymal transition, stemness, and metastasis. Our results establish CBX8 as a critical driver of HCC stem cell-like and metastatic behaviors and characterize its role in modulating BMP4 expression. These findings have implications for the targeting of CBX8 as an approach to HCC prognosis and treatment.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Polycomb Repressive Complex 1/genetics , Animals , Bone Morphogenetic Protein 4/biosynthesis , Bone Morphogenetic Protein 4/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Hep G2 Cells , Heterografts , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Polycomb Repressive Complex 1/biosynthesis , Prognosis , Transfection
11.
Cell Rep ; 26(7): 1906-1918.e8, 2019 02 12.
Article in English | MEDLINE | ID: mdl-30759399

ABSTRACT

In this study, we demonstrate that, among all five CBX Polycomb proteins, only CBX7 possesses the ability to control self-renewal of human hematopoietic stem and progenitor cells (HSPCs). Xenotransplantation of CBX7-overexpressing HSPCs resulted in increased multi-lineage long-term engraftment and myelopoiesis. Gene expression and chromatin analyses revealed perturbations in genes involved in differentiation, DNA and chromatin maintenance, and cell cycle control. CBX7 is upregulated in acute myeloid leukemia (AML), and its genetic or pharmacological repression in AML cells inhibited proliferation and induced differentiation. Mass spectrometry analysis revealed several non-histone protein interactions between CBX7 and the H3K9 methyltransferases SETDB1, EHMT1, and EHMT2. These CBX7-binding proteins possess a trimethylated lysine peptide motif highly similar to the canonical CBX7 target H3K27me3. Depletion of SETDB1 in AML cells phenocopied repression of CBX7. We identify CBX7 as an important regulator of self-renewal and uncover non-canonical crosstalk between distinct pathways, revealing therapeutic opportunities for leukemia.


Subject(s)
Hematopoietic Stem Cells/metabolism , Polycomb Repressive Complex 1/metabolism , Stem Cells/metabolism , Animals , Female , Fetal Blood/cytology , Fetal Blood/metabolism , HEK293 Cells , HL-60 Cells , Hematopoietic Stem Cells/cytology , Heterografts , Histone-Lysine N-Methyltransferase/metabolism , Humans , K562 Cells , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Polycomb Repressive Complex 1/biosynthesis , Polycomb Repressive Complex 1/genetics , Stem Cells/cytology , Transcription, Genetic
12.
Pathol Res Pract ; 215(2): 243-250, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30420101

ABSTRACT

BACKGROUND: Previous studies indicated that miR-218 was deregulated in gastric cancer patients and correlated with tumor invasion and prognosis. The aim of this study was to clarify the effect of miR-218 on the malignant behavior of gastric cancer and its role in regulating Bmi-1/Akt signaling pathway. MATERIALS AND METHODS: We used miR-218 mimic to transfect gastric cancer cell lines AGS and SGC-7901, and the overexpression efficiency was validated using qRT-PCR assay. MTT assay and Transwell chamber system were performed to detect the effect of miR-218 on cell proliferation, invasion and migration on gastric cancer. Western blot and qRT-PCR assay was used to test the role of miR-218 in regulating Bmi-1/Akt signaling pathway. RESULTS: As shown in our research, ectopic expression of miR-218 in gastric cancer cells inhibits the proliferation, invasion and migration of gastric cancer cells. In addition, miR-218 re-expression inhibits the expression of Bmi-1 and its downstream target p-Akt473, as well as MMPs and EMT process. CONCLUSIONS: miR-218 inhibits the proliferation, invasion and migration of gastric cancer cells through modulating EMT process and the expression of MMPs via Bmi-1/Akt signaling pathway.


Subject(s)
Adenocarcinoma/genetics , Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic/genetics , MicroRNAs/genetics , Stomach Neoplasms/genetics , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Humans , Kaplan-Meier Estimate , Polycomb Repressive Complex 1/biosynthesis , Proto-Oncogene Proteins c-akt/biosynthesis , Signal Transduction/genetics , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology
13.
Cancer Res ; 78(7): 1751-1765, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29374066

ABSTRACT

Colorectal cancer includes an invasive stem-like/mesenchymal subtype, but its genetic drivers, functional, and clinical relevance are uncharacterized. Here we report the definition of an altered miRNA signature defining this subtype that includes a major genomic loss of miR-508. Mechanistic investigations showed that this miRNA affected the expression of cadherin CDH1 and the transcription factors ZEB1, SALL4, and BMI1. Loss of miR-508 in colorectal cancer was associated with upregulation of the novel hypoxia-induced long noncoding RNA AK000053. Ectopic expression of miR-508 in colorectal cancer cells blunted epithelial-to-mesenchymal transition (EMT), stemness, migration, and invasive capacity in vitro and in vivo In clinical colorectal cancer specimens, expression of miR-508 negatively correlated with stemness and EMT-associated gene expression and positively correlated with patient survival. Overall, our results showed that miR-508 is a key functional determinant of the stem-like/mesenchymal colorectal cancer subtype and a candidate therapeutic target for its treatment.Significance: These results define a key functional determinant of a stem-like/mesenchymal subtype of colorectal cancers and a candidate therapeutic target for its treatment. Cancer Res; 78(7); 1751-65. ©2018 AACR.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition/genetics , Mesenchymal Stem Cells/cytology , MicroRNAs/genetics , Animals , Antigens, CD/biosynthesis , Caco-2 Cells , Cadherins/biosynthesis , Cell Line, Tumor , Cell Movement/genetics , HCT116 Cells , HT29 Cells , Humans , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Neoplasm Invasiveness/genetics , Neoplasm Transplantation , Polycomb Repressive Complex 1/biosynthesis , RNA, Long Noncoding/biosynthesis , RNA, Long Noncoding/genetics , Transcription Factors/biosynthesis , Transplantation, Heterologous , Zinc Finger E-box-Binding Homeobox 1/biosynthesis
14.
Stem Cells ; 36(4): 562-573, 2018 04.
Article in English | MEDLINE | ID: mdl-29282831

ABSTRACT

The outermost layer of the eye, the cornea, is renewed continuously throughout life. Stem cells of the corneal epithelium reside in the limbus at the corneal periphery and ensure homeostasis of the central epithelium. However, in young mice, homeostasis relies on cells located in the basal layer of the central corneal epithelium. Here, we first studied corneal growth during the transition from newborn to adult and assessed Keratin 19 (Krt19) expression as a hallmark of corneal maturation. Next, we set out to identify a novel marker of murine corneal epithelial progenitor cells before, during and after maturation, and we found that Bmi1 is expressed in the basal epithelium of the central cornea and limbus. Furthermore, we demonstrated that Bmi1+ cells participated in tissue replenishment in the central cornea. These Bmi1+ cells did not maintain homeostasis of the cornea for more than 3 months, reflecting their status as progenitor rather than stem cells. Finally, after injury, Bmi1+ cells fueled homeostatic maintenance, whereas wound closure occurred via epithelial reorganization. Stem Cells 2018;36:562-573.


Subject(s)
Cornea/metabolism , Corneal Injuries/metabolism , Gene Expression Regulation , Polycomb Repressive Complex 1/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Stem Cells/metabolism , Wound Healing , Animals , Cornea/pathology , Corneal Injuries/genetics , Corneal Injuries/pathology , Mice , Mice, Inbred ICR , Mice, Mutant Strains , Polycomb Repressive Complex 1/genetics , Proto-Oncogene Proteins/genetics , Stem Cells/pathology
15.
J Ethnopharmacol ; 213: 328-339, 2018 Mar 01.
Article in English | MEDLINE | ID: mdl-29051115

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Schisandra chinensis (Turcz.) Baill (SC), Lycium chinense Mill (LC) and Eucommia ulmoides Oliv (EU) are representative tonic herbal medicines that help to strengthen body muscles and bones making them stronger according to the Donguibogam, a tradition medical book of the Joseon Dynasty in Korea. AIM OF THE STUDY: To evaluate effects of an herbal formula consisting of SC, LC and EU on muscle atrophy in C2C12 myotubes and in a rat model of immobilization-induced muscle atrophy. MATERIALS AND METHODS: Muscle atrophy was developed by cast immobilization of unilateral hindlimb on rats for 3 weeks. Treatments were administered orally 14 times over 3 weeks. After treatments, we compared the change of body weight, muscle weight, grip strength, muscle fiber size, muscle fiber type shift by Grip strength meter, H&E stain and ATPase stain. And western blot was used for evaluating molecular mechanism in muscle atrophy on C2C12 cells. RESULTS: When taken individually, SC was the most effective of the three in inhibiting tumor necrosis factor alpha (TNF-α)-induced degeneration of C2C12 myogenesis. The formulation with a mass ratio of 2:1:1 SC: LC: EU (SSLE) was more effective against TNF-α-induced muscle atrophy than was a 1:1:1 SC: LC: EU (SLE) formula or any of the single herbal extracts. In a rat model of disuse muscle atrophy, the SSLE formula significantly inhibited reductions in muscle weight, grip strength and muscle fiber size induced by hindlimb immobilization, in a dose-dependent manner. The formula also inhibited immobilization-induced shifting of the muscle fiber type in soleus muscle. Treatment with SSLE inhibited TNF-α-induced expression of the atrogenes atrogin-1 and muscle RING-finger protein 1 in C2C12 cells. The SSLE formula also increased myoblast differentiation markers (myoD and myogenin) and activation of the Akt and mammalian target of rapamycin (mTOR) signaling pathway. CONCLUSION: These findings suggest that the SSLE formula prevents muscle atrophy through inhibition of the ubiquitin-proteasome system as well as upregulation of myoblast differentiation and muscle protein synthesis in C2C12 cells. Taken together, we conclude that the SSLE formula is invaluable for the development of therapeutic medicines to prevent disuse muscle atrophy and its accompanying muscle weakness.


Subject(s)
Eucommiaceae , Lycium , Muscular Atrophy/drug therapy , Phytotherapy , Schisandra , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Hand Strength , Hindlimb Suspension , Male , Mice , Muscle Fibers, Skeletal/drug effects , Muscle Proteins/biosynthesis , Muscle, Skeletal/drug effects , Plant Extracts/therapeutic use , Polycomb Repressive Complex 1/biosynthesis , Rats , SKP Cullin F-Box Protein Ligases/biosynthesis , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism , Ubiquitin-Protein Ligases/biosynthesis
16.
Cell Tissue Res ; 370(3): 427-440, 2017 12.
Article in English | MEDLINE | ID: mdl-28986650

ABSTRACT

Homeostatic cell turnover has been extensively characterized in mammals. In their adult tissues, lost or aging differentiated cells are replenished by a self-renewing cohort of stem cells. The stem cells have been particularly well studied in the intestine and are clearly identified by the expression of marker genes including Lgr5 and Bmi1. It is, however, unknown if the established principles of tissue renewal learned from mammals would be operating in non-mammalian systems. Here, we study homeostatic cell turnover in the sea cucumber digestive tube, the organ with high tissue plasticity even in adult animals. Both the luminal epithelium and mesothelium express orthologs of mammalian Lgr5 and Bmi1. However, unlike in mammals, there is no segregation of these positively labeled cells to specific regions in the luminal epithelium, where most of the cell proliferation would take place. In the mesothelium, the cells expressing the stem cell markers are tentatively identified as peritoneocytes. There are significant differences among the five anatomical gut regions in cell renewal dynamics and stem factor expression. The cloaca differs from the rest of the digestive tube as the region with the highest expression of the Lgr5 ortholog, lowest level of Bmi1 and the longest retention of BrdU-labeled cells.


Subject(s)
Epithelial Cells/metabolism , Gastrointestinal Tract/metabolism , Polycomb Repressive Complex 1/biosynthesis , Receptors, G-Protein-Coupled/biosynthesis , Sea Cucumbers/metabolism , Stem Cell Factor/biosynthesis , Stem Cells/metabolism , Animals , Cell Proliferation , Epithelium/metabolism , Mitogen-Activated Protein Kinase 7/genetics , Polycomb Repressive Complex 1/genetics , Receptors, G-Protein-Coupled/genetics , Stem Cells/cytology
17.
Tumour Biol ; 39(7): 1010428317718414, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28720065

ABSTRACT

MiR-139-5p has been reported to be overexpressed in many types of cancers, but its role in bladder cancer has not been elucidated yet. Here, we report that miR-139-5p functions as a tumor suppressor in bladder cancer and inhibits the cancer stem cell self-renewal by targeting Bmi1 directly. We found that miR-139-5p expression was significantly downregulated in the bladder cancer specimens compared with that in adjacent normal tissues. In vitro, restoration of miR-139-5p expression significantly inhibited the proliferation of bladder cancer cells. Mechanism analysis revealed that miR-139-5p could decrease Bmi1 protein levels by binding to the 3' untranslated region of Bmi1 messenger RNA. Stem cell-related proteins such as c-MYC, NANOG, OCT4, and KLF4 and signaling pathways such as Wnt signaling were suppressed by restoration of miR-139-5p in bladder cancer cells. In addition, miR-139-5p expression also blocked self-renewal of bladder cancer stem cells by inhibiting Bmi1. In summary, our study supports that miR-139-5p acts as a tumor suppressor in bladder cancer development and suppresses cancer stem cell property of bladder cancer. Our study also suggests that miR-139-5p has the potential to be used as a therapeutic molecule for bladder cancer treatment.


Subject(s)
Cell Proliferation/genetics , MicroRNAs/genetics , Polycomb Repressive Complex 1/biosynthesis , Urinary Bladder Neoplasms/genetics , Apoptosis/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Self Renewal/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Kruppel-Like Factor 4 , Male , Neoplastic Stem Cells/pathology , Polycomb Repressive Complex 1/genetics , Urinary Bladder Neoplasms/pathology , Wnt Signaling Pathway/genetics
18.
Target Oncol ; 12(4): 449-462, 2017 08.
Article in English | MEDLINE | ID: mdl-28589491

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) represents one of the most lethal cancers worldwide due to therapy resistance and disease recurrence. Tumor relapse following treatment could be driven by the persistence of liver cancer stem-like cells (CSCs). The protein BMI1 is a member of the polycomb epigenetic factors governing cellular self-renewal, proliferation, and stemness maintenance. BMI1 expression also correlates with poor patient survival in various cancer types. OBJECTIVE: We aimed to elucidate the extent to which BMI1 can be used as a potential therapeutic target for CSC eradication in HCC. METHODS: We have recently participated in characterizing the first known pharmacological small molecule inhibitor of BMI1. Here, we synthesized a panel of novel BMI1 inhibitors and examined their ability to alter cellular growth and eliminate cancer progenitor/stem-like cells in HCC with different p53 backgrounds. RESULTS: Among various molecules examined, RU-A1 particularly downregulated BMI1 expression, impaired cell viability, reduced cell migration, and sensitized HCC cells to 5-fluorouracil (5-FU) in vitro. Notably, long-term analysis of HCC survival showed that, unlike chemotherapy, RU-A1 effectively reduced CSC content, even as monotherapy. BMI1 inhibition with RU-A1 diminished the number of stem-like cells in vitro more efficiently than the model compound C-209, as demonstrated by clonogenic assays and impairment of CSC marker expression. Furthermore, xenograft assays in zebrafish showed that RU-A1 abrogated tumor growth in vivo. CONCLUSIONS: This study demonstrates the ability to identify agents with the propensity for targeting CSCs in HCC that could be explored as novel treatments in the clinical setting.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Polycomb Repressive Complex 1/antagonists & inhibitors , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/pharmacology , Animals , Antineoplastic Agents/chemistry , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , HEK293 Cells , Hep G2 Cells , Humans , Immunohistochemistry , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Polycomb Repressive Complex 1/biosynthesis , Polycomb Repressive Complex 1/genetics , Small Molecule Libraries/chemistry , Xenograft Model Antitumor Assays , Zebrafish
19.
Oncotarget ; 8(15): 25384-25394, 2017 Apr 11.
Article in English | MEDLINE | ID: mdl-28445986

ABSTRACT

Polycomb group member protein BMI1 is involved in maintaining cell identity, proliferation, differentiation and human oncogenesis. In the present study, we determined BMI1 mRNA expression in whole blood and evaluated the impact of the expression level on the treatment response and survival of 96 advanced NSCLC patients treated with first-line platinum-based chemotherapy. We also determined BMI1 mRNA expression in primary tumors from 22 operable NSCLC patients treated with radical surgery. We found that compared with control subjects, BMI1 mRNA expression in whole blood of advanced NSCLC patients was decreased (P<0.001). Similarly, we observed decreased BMI1 mRNA expression in primary tumors compared to normal lungs from operable NSCLC patients (P=0.001). We found high BMI1 mRNA expression in blood was associated with longer progression-free survival (PFS) (P=0.049) and overall survival (OS) (P=0.012) in advanced NSCLC patients treated with first-line platinum-based chemotherapy. However, no association between the BMI1 mRNA level and response to chemotherapy was found (P=0.21). Multivariate Cox proportional hazards regression analysis showed elevated BMI1 mRNA level in whole blood was an independent prognostic factor for longer PFS (P=0.012) and OS (P<0.001). In conclusion, BMI1 mRNA expression in whole blood might represent a new biomarker for the diagnosis and prognosis of NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , Polycomb Repressive Complex 1/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/blood , Adult , Aged , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/pathology , Female , Humans , Lung Neoplasms/blood , Lung Neoplasms/pathology , Male , Middle Aged , Polycomb Repressive Complex 1/biosynthesis , Polycomb Repressive Complex 1/blood , Prognosis , RNA, Messenger/genetics
20.
Mol Oncol ; 11(2): 194-207, 2017 02.
Article in English | MEDLINE | ID: mdl-28079973

ABSTRACT

Extracellular signal-regulated kinase 3 (ERK3) is an atypical mitogen-activated protein kinase (MAPK), whose biological activity is tightly regulated by its cellular abundance. Recent studies have revealed that ERK3 is upregulated in multiple cancers and promotes cancer cell migration/invasion and drug resistance. Little is known, however, about how ERK3 expression level is upregulated in cancers. Here, we have identified the oncogenic polycomb group protein BMI1 as a positive regulator of ERK3 level in head and neck cancer cells. Mechanistically, BMI1 upregulates ERK3 expression by suppressing the tumor suppressive microRNA (miRNA) let-7i, which directly targets ERK3 mRNA. ERK3 then acts as an important downstream mediator of BMI1 in promoting cancer cell migration. Importantly, ERK3 protein level is positively correlated with BMI1 level in head and neck tumor specimens of human patients. Taken together, our study revealed a molecular pathway consisting of BMI1, miRNA let-7i, and ERK3, which controls the migration of head and neck cancer cells, and suggests that ERK3 kinase is a potential new therapeutic target in head and neck cancers, particularly those with BMI1 overexpression.


Subject(s)
Cell Movement , Head and Neck Neoplasms/metabolism , MAP Kinase Signaling System , MicroRNAs/biosynthesis , Mitogen-Activated Protein Kinase 6/biosynthesis , Neoplasm Proteins/biosynthesis , Polycomb Repressive Complex 1/biosynthesis , RNA, Neoplasm/biosynthesis , HeLa Cells , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Humans , MicroRNAs/genetics , Mitogen-Activated Protein Kinase 6/genetics , Neoplasm Proteins/genetics , Polycomb Repressive Complex 1/genetics , RNA, Neoplasm/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...