1
|
Chen Z, Ju H, Zhao T, Yu S, Li P, Jia J, Li N, Jing X, Tan B, Li Y. hsa_circ_0092306 Targeting miR-197-3p Promotes Gastric Cancer Development by Regulating PRKCB in MKN-45 Cells. MOLECULAR THERAPY-NUCLEIC ACIDS 2019; 18:617-626. [PMID: 31689616 PMCID: PMC6838893 DOI: 10.1016/j.omtn.2019.08.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 07/11/2019] [Accepted: 08/07/2019] [Indexed: 12/24/2022]
Abstract
Gastric cancer (GC) is one of the most common cancers worldwide and is thus a global cancer burden. Here, we focused on a novel circular RNA hsa_circ_0092306 and explored the potential molecular mechanism to provide a new target for and novel insights into GC treatment. The GEO microarray was mined and analyzed with R software. Sanger sequencing and RNase R assay were applied to verify the identification of hsa_circ_0092306. Quantitative real-time PCR and western blot were performed to measure the mRNA and protein levels. Pull-down and luciferase reporter assays were conducted to confirm the target relationships. Annexin V-PI apoptosis flow cytometry, 3-(4,5Dimethylthiazol- yl)-2,5Dimethylthiazol-2-yl)-2,5diphenyltetrazolium bromide (MTT), wound healing, and Transwell assays were applied to detect cell apoptosis, viability, migration, and invasion in MKN-45 cells, respectively. A xenograft in vivo experiment was conducted to confirm the cell experiment results. hsa_circ_0092306 was upregulated in GC tissues and GC cells, and promoted GC development in MKN-45 cells. hsa_circ_0092306 inhibited tumor suppressor miR-197-3p expression but promoted tumor promotor protein kinase C beta (PRKCB) expression in MKN-45 cells. hsa_circ_0092306 and PRKCB had a common target (miR-197-3p) and were negatively related to miR-197-3p expression. hsa_circ_0092306 promoted the development of GC by regulating the pathway of miR-197-3p/PRKCB in MKN-45 cells.
Collapse
Affiliation(s)
- Zihao Chen
- Graduate School of Hebei Medical University, Shijiazhuang 050017, Hebei, China; The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei, China
| | - Hongping Ju
- School of Medicine, Kunming University, Kunming 650214, Yunnan, China; The Respiratory System Disease Prevention and Control of Public Service Platform of Science and Technology in Yunnan Province, Kunming 650214, Yunnan, China.
| | - Ting Zhao
- Graduate School of Hebei Medical University, Shijiazhuang 050017, Hebei, China
| | - Shan Yu
- School of Medicine, Kunming University, Kunming 650214, Yunnan, China
| | - Ping Li
- School of Medicine, Kunming University, Kunming 650214, Yunnan, China
| | - Jing Jia
- School of Medicine, Kunming University, Kunming 650214, Yunnan, China
| | - Nan Li
- School of Medicine, Kunming University, Kunming 650214, Yunnan, China
| | - Xiaojie Jing
- Department of Medicine, The People's Hospital of Economic and Technological Development Zone, Kunming 650217, Yunnan, China
| | - Bibo Tan
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei, China
| | - Yong Li
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei, China.
| |
Collapse
|
2
|
Kong J, Wang L, Ren L, Yan Y, Cheng Y, Huang Z, Shen F. Triptolide induces mitochondria-mediated apoptosis of Burkitt's lymphoma cell via deacetylation of GSK-3β by increased SIRT3 expression. Toxicol Appl Pharmacol 2018; 342:1-13. [DOI: 10.1016/j.taap.2018.01.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 01/15/2018] [Accepted: 01/18/2018] [Indexed: 12/16/2022]
|
3
|
Huang W, Mehta D, Sif S, Kent LN, Jacob ST, Ghoshal K, Mehta KD. Dietary fat/cholesterol-sensitive PKCβ-RB signaling: Potential role in NASH/HCC axis. Oncotarget 2017; 8:73757-73765. [PMID: 29088742 PMCID: PMC5650297 DOI: 10.18632/oncotarget.17890] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 03/30/2017] [Indexed: 12/14/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a frequent form of cancer with a poor prognosis, and environmental factors significantly contribute to the risk. Despite knowledge that a Western-style diet is a risk factor in the development of nonalcoholic steatohepatitis (NASH) and subsequent progression to HCC, diet-induced signaling changes are not well understood. Understanding molecular mechanisms altered by diet is crucial for developing preventive and therapeutic strategies. We have previously shown that diets enriched with high-fat and high-cholesterol, shown to produce NASH and HCC, induce hepatic protein kinase C beta (PKCβ) expression in mice, and a systemic loss of PKCβ promotes hepatic cholesterol accumulation in response to this diet. Here, we sought to determine how PKCβ and diet functionally interact during the pathogenesis of NASH and how it may promote hepatic carcinogenesis. We found that diet-induced hepatic PKCβ expression is accompanied by an increase in phosphorylation of Ser780 of retinoblastoma (RB) protein. Intriguingly, PKCβ-/- livers exhibited reduced RB protein levels despite increased transcription of the RB gene. It is also accompanied by reduced RBL-1 with no significant effect on RBL-2 protein levels. We also found reduced expression of the PKCβ in HCC compared to non-tumorous liver in human patients. These results raise an interesting possibility that diet-induced PKCβ activation represents an important mediator in the functional wiring of cholesterol metabolism and tumorigenesis through modulating stability of cell cycle-associated proteins. The potential role of PKCβ in the suppression of tumorigenesis is discussed.
Collapse
Affiliation(s)
- Wei Huang
- Department of Biological Chemistry and Pharmacology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Devina Mehta
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Said Sif
- Department of Biological Chemistry and Pharmacology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Lindsey N Kent
- Department of Cancer Genetics, OSU Comprehensive Cancer Center, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Samson T Jacob
- Department of Cancer Genetics, OSU Comprehensive Cancer Center, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Kalpana Ghoshal
- Department of Cancer Genetics, OSU Comprehensive Cancer Center, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Kamal D Mehta
- Department of Biological Chemistry and Pharmacology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, Ohio, USA
| |
Collapse
|
4
|
Xue K, Gu JJ, Zhang Q, Mavis C, Hernandez-Ilizaliturri FJ, Czuczman MS, Guo Y. Vorinostat, a histone deacetylase (HDAC) inhibitor, promotes cell cycle arrest and re-sensitizes rituximab- and chemo-resistant lymphoma cells to chemotherapy agents. J Cancer Res Clin Oncol 2016; 142:379-87. [PMID: 26314218 DOI: 10.1007/s00432-015-2026-y] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 08/04/2015] [Indexed: 12/29/2022]
Abstract
PURPOSE Preclinical models of chemotherapy resistance and clinical observations derived from the prospective multicenter phase III collaborative trial in relapsed aggressive lymphoma (CORAL) study demonstrated that primary refractory/relapsed B cell diffuse large B cell lymphoma has a poor clinical outcome with current available second-line treatments. Preclinically, we found that rituximab resistance is associated with a deregulation on the mitochondrial potential rendering lymphoma cells resistant to chemotherapy-induced apoptotic stimuli. There is a dire need to develop agents capable to execute alternative pathways of cell death in an attempt to overcome chemotherapy resistance. Posttranscriptional histone modification plays an important role in regulating gene transcription and is altered by histone acetyltransferases (HATs) and histone deacetylases (HDACs). HDACs regulate several key cellular functions, including cell proliferation, cell cycle, apoptosis, angiogenesis, migration, antigen presentation, and/or immune regulation. Given their influence in multiple regulatory pathways, HDAC inhibition is an attractive strategy to evaluate its anti-proliferation activity in cancer cells. To this end, we studied the anti-proliferation activity and mechanisms of action of suberoylanilide hydroxamic acid (SAHA, vorinostat) in rituximab-chemotherapy-resistant preclinical models. METHODS A panel of rituximab-chemotherapy-sensitive (RSCL) and rituximab-chemotherapy-resistant cell lines (RRCL) and primary tumor cells isolated from relapsed/refractory B cell lymphoma patients were exposed to escalating doses of vorinostat. Changes in mitochondrial potential, ATP synthesis, and cell cycle distribution were determined by Alamar blue reduction, Titer-Glo luminescent assays, and flow cytometric, respectively. Protein lysates were isolated from vorinostat-exposed cells, and changes in members of Bcl-2 family, cell cycle regulatory proteins, and the acetylation status of histone H3 were evaluated by Western blotting. Finally, cell lines were pre-exposed to vorinostat for 48 h and subsequently exposed to several chemotherapy agents (cisplatin, etoposide, or gemcitabine); changes in cell viability were determined by CellTiter-Glo(®) luminescence assay (Promega, Fitchburg, WI), and synergistic activity was evaluated using the CalcuSyn software. RESULTS Vorinostat induced dose-dependent cell death in RRCL and in primary tumor cells. In addition, in vitro exposure of RRCL to vorinostat resulted in an increase in p21 and acetylation of histone H3 leading to G1 cell cycle arrest. Vorinostat exposure resulted in apoptosis in RSCL cell lines but not in RRCL. This finding suggests that in RRCL, vorinostat induces cell death by alternative pathways (i.e., irreversible cell cycle arrest). Of interest, vorinostat was found to reverse acquired chemotherapy resistance in RRCL. CONCLUSIONS Our data suggest that vorinostat is active in RRCL with a known defective apoptotic machinery, it can active alternative cell death pathways. Given the multiple pathways affected by HDAC inhibition, vorinostat can potentially be used to overcome acquired resistant to chemotherapy in aggressive B cell lymphoma.
Collapse
Affiliation(s)
- Kai Xue
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Juan J Gu
- Departments of Medicine and Immunology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Qunling Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Cory Mavis
- Departments of Medicine and Immunology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | | | - Myron S Czuczman
- Departments of Medicine and Immunology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Ye Guo
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
| |
Collapse
|
5
|
Saba NS, Angelova M, Lobelle-Rich PA, Levy LS. Disruption of pre-B-cell receptor signaling jams the WNT/β-catenin pathway and induces cell death in B-cell acute lymphoblastic leukemia cell lines. Leuk Res 2015; 39:S0145-2126(15)30355-6. [PMID: 26298175 DOI: 10.1016/j.leukres.2015.08.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 08/04/2015] [Accepted: 08/06/2015] [Indexed: 11/28/2022]
Abstract
Targeting components of the B-cell receptor (BCR) pathway have dramatically improved clinical outcomes in a variety of B-cell malignancies. Despite the well-documented pathogenic role of BCR precursor (pre-BCR) pathway in B-cell acute lymphoblastic leukemia (B-ALL), there is limited available data of therapies that aim to disrupt this pathway. To investigate the role of protein kinase Cβ (PKCβ), a crucial mediator of BCR and pre-BCR signaling, in B-ALL survival, we studied the activity of the PKCβ selective inhibitor enzastaurin (ENZ) in seven B-ALL cell lines. Treatment with ENZ resulted in a dose- and time-dependent growth inhibition in all cell lines with a relatively higher efficacy in pro-B ALL with translocation t(4;11)(q21;q23). The mechanism of growth inhibition was by apoptotic induction and cell cycle arrest. A rapid reduction in phosphorylation of AKT and its downstream target glycogen synthase kinase 3β (GSK3β) were observed at 30min after treatment and remaining for 48h. The reduction in GSK3β phosphorylation was associated with a paradoxical accumulation of β-catenin, which was due to a transient loss of β-catenin phosphorylation at ser33-37. In addition, accumulation of β-catenin was associated with downregulation of c-Myc, upregulatiuon of c-Jun, and a subsequent protective effect on the tumor suppressor p73. Data in this paper were presented in part at 2012 American Society of Hematology Annual Meeting, abstract 1350.
Collapse
Affiliation(s)
- Nakhle S Saba
- Section of Hematology and Medical Oncology, Department of Medicine, Tulane University, New Orleans, LA 70112, USA.
| | - Magdalena Angelova
- Department of Microbiology and Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Patricia A Lobelle-Rich
- Section of Hematology and Medical Oncology, Department of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Laura S Levy
- Department of Microbiology and Immunology, Tulane University, New Orleans, LA 70112, USA
| |
Collapse
|
6
|
Ma D, Fang Q, Wang P, Gao R, Wu W, Lu T, Cao L, Hu X, Wang J. Induction of heme oxygenase-1 by Na+-H+ exchanger 1 protein plays a crucial role in imatinib-resistant chronic myeloid leukemia cells. J Biol Chem 2015; 290:12558-71. [PMID: 25802333 DOI: 10.1074/jbc.m114.626960] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Indexed: 12/18/2022] Open
Abstract
Resistance toward imatinib (IM) and other BCR/ABL tyrosine kinase inhibitors remains troublesome in the treatment of advanced stage chronic myeloid leukemia (CML). The aim of this study was to estimate the reversal effects of down-regulation of Na(+)/H(+) exchanger 1 (NHE1) on the chemoresistance of BCR-ABL-positive leukemia patients' cells and cell lines. After treatment with the specific NHE1 inhibitor cariporide to decrease intracellular pH (pHi), the heme oxygenase-1 (HO-1) levels of the K562R cell line and cells from IM-insensitive CML patients decreased. HO-1, as a Bcr/Abl-dependent survival molecule in CML cells, is important for the resistance to tyrosine kinase inhibitors in patients with newly diagnosed CML or IM-resistant CML. Silencing PKC-β and Nrf-2 or treatment with inhibitors of p38 pathways obviously blocked NHE1-induced HO-1 expression. Furthermore, treatment with HO-1 or p38 inhibitor plus IM increased the apoptosis of the K562R cell line and IM-insensitive CML patients' cells. Inhibiting HO-1 enhanced the activation of caspase-3 and poly(ADP-ribose) polymerase-1. Hence, the results support the anti-apoptotic role of HO-1 induced by NHE1 in the K562R cell line and IM-insensitive CML patients and provide a mechanism by which inducing HO-1 expression via the PKC-β/p38-MAPK pathway may promote tumor resistance to oxidative stress.
Collapse
Affiliation(s)
- Dan Ma
- From the Department of Hematology, Affiliated Hospital of Guiyang Medical University, Guiyang 550004, China, Department of Pharmacy, Affiliated BaiYun Hospital of Guiyang Medical University, Guiyang 550014, China
| | - Qin Fang
- Department of Pharmacy, Affiliated BaiYun Hospital of Guiyang Medical University, Guiyang 550014, China, Department of Pharmacy, Affiliated Hospital of Guiyang Medical University, Guiyang 550004, China, and
| | - Ping Wang
- From the Department of Hematology, Affiliated Hospital of Guiyang Medical University, Guiyang 550004, China, Guizhou Province Hematopoietic Stem Cell Transplantation Center and Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guiyang 550004, China
| | - Rui Gao
- From the Department of Hematology, Affiliated Hospital of Guiyang Medical University, Guiyang 550004, China, Guizhou Province Hematopoietic Stem Cell Transplantation Center and Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guiyang 550004, China
| | - Weibing Wu
- From the Department of Hematology, Affiliated Hospital of Guiyang Medical University, Guiyang 550004, China, Guizhou Province Hematopoietic Stem Cell Transplantation Center and Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guiyang 550004, China
| | - Tangsheng Lu
- School of Pharmacy, Guiyang Medical University, Guiyang 550004, China
| | - Lu Cao
- School of Pharmacy, Guiyang Medical University, Guiyang 550004, China
| | - Xiuying Hu
- From the Department of Hematology, Affiliated Hospital of Guiyang Medical University, Guiyang 550004, China, Guizhou Province Hematopoietic Stem Cell Transplantation Center and Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guiyang 550004, China
| | - Jishi Wang
- From the Department of Hematology, Affiliated Hospital of Guiyang Medical University, Guiyang 550004, China, Guizhou Province Hematopoietic Stem Cell Transplantation Center and Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guiyang 550004, China,
| |
Collapse
|
7
|
Current world literature. Curr Opin Oncol 2012; 24:587-95. [PMID: 22886074 DOI: 10.1097/cco.0b013e32835793f1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|