1
|
Zhang G, Xu Z, Yu M, Gao H. Bcl-2 interacting protein 3 (BNIP3) promotes tumor growth in breast cancer under hypoxic conditions through an autophagy-dependent pathway. Bioengineered 2022; 13:6280-6292. [PMID: 35200106 PMCID: PMC8973668 DOI: 10.1080/21655979.2022.2036399] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Hypoxia-induced autophagy has been implicated in many cancers. Bcl-2 interacting protein 3 (BNIP3) has been associated with hypoxia, whose aberrant expression is involved in the carcinogenesis of breast cancer (BC). Here, we aim to investigate the role of hypoxia-induced autophagy and the mechanistic actions of the bioinformatically identified BNIP3 in BC. The expression pattern of BNIP3 in BC tissues and cell lines was examined using RT-qPCR and Western blot analyses. The binding affinity among BNIP3, BECN1 and BCL-2 was characterized by co-immunoprecipitation. BNIP3 expression was manipulated to assess its effects on BC cell malignant phenotypes, evaluated by cell counting kit-8, Transwell and wound healing assays, and on BC autophagy under hypoxic conditions. A BC tumor xenografts mouse model was further established to substantiate in vitro findings. Up-regulated expression of BNIP3 was found in BC tissues and cell lines, and BNIP3 expression was positively correlated with hypoxia exposure duration. BNIP3 knockdown restricted BC cell proliferation, invasion, and migration under hypoxic conditions. BNIP3 activated BC cell autophagy by inhibiting the binding between BCL-2 and BECN1 under hypoxic conditions. BNIP3-induced autophagy activation enhanced malignant phenotypes of BC cells, thus accelerating the tumorigenesis of BC cells in vivo. These data collectively supported the tumor-promoting role of BNIP3 in autophagy activation of BC under hypoxic conditions, highlighting a potential therapeutic target against BC.
Collapse
Affiliation(s)
- Guipu Zhang
- Department of Breast Surgery, Changzhou Cancer Hospital, Changzhou, China
| | - Zhiyi Xu
- Department of Pathology, Changzhou Cancer Hospital, Changzhou, China
| | - Minjing Yu
- Department of Breast Surgery, Changzhou Cancer Hospital, Changzhou, China
| | - Haiyan Gao
- Department of Breast Surgery, Changzhou Cancer Hospital, Changzhou, China
| |
Collapse
|
2
|
Huang GD, Cui P, Ma GX, Chen FF, Chen ZB, Li XJ, Liao ZJ, Li WP, Li ZY, Chen L. Phragmunis a suppresses glioblastoma through the regulation of MCL1-FBXW7 by blocking ELK1-SRF complex-dependent transcription. Neurochem Int 2021; 147:105051. [PMID: 33979572 DOI: 10.1016/j.neuint.2021.105051] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 04/20/2021] [Accepted: 04/21/2021] [Indexed: 10/21/2022]
Abstract
Glioblastoma (GBM) is a highly aggressive brain tumor. During screening work, we found a new compound named phragmunis A (PGA), which is derived from the fruitbody of Trogia venenata, exhibits a potential cytotoxic effect on patient-derived recurrent GBM cells and temozolomide (TMZ)-resistant cell lines. The present study was designed to investigate the potential molecular mechanism of the anti-glioma effects of PGA in vitro and in vivo. Studies investigating the mechanism revealed that PGA diminished the binding efficiency of ETS family of transcription factor (ELK1) and Serum response factor (SRF), and suppressed ELK1-SRF complex-dependent transcription, which decreased the transcriptional levels of downstream genes Early growth response protein 1 (EGR1)-Polycomb ring finger (BMI1), thus inducing the imbalanced regulation between Myeloid cell leukaemia-1 (MCL1) and F-Box and WD repeat domain containing 7 (FBXW7). Finally, orthotopic xenograft models were established to confirm the anti-glioma effect of PGA on tumour growth. We showed, for the first time, that the cytotoxic effects of PGA occurred by inducing MCL1 inhibition and FBXW7 activation by blocking ELK1-SRF complex-dependent transcription. The blockage of ELK1-mediated transcription resulted in the suppression of EGR1-BMI1, which led to the upregulation of FBXW7 expression and downregulation of MCL1. These findings suggested that PGA could be a therapeutic drug candidate for the treatment of recurrent GBM by targeting the ELK1-SRF complex.
Collapse
Affiliation(s)
- Guo-Dong Huang
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, 3002# Sungang Road, Futian District, Shenzhen, 518035, China
| | - Ping Cui
- Department of Pharmacy, Shenzhen Children's Hospital, Shenzhen, 518038, China
| | - Guo-Xu Ma
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100193, China
| | - Fan-Fan Chen
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, 3002# Sungang Road, Futian District, Shenzhen, 518035, China
| | - Ze-Bin Chen
- Department of Pharmacy, Shenzhen Children's Hospital, Shenzhen, 518038, China
| | - Xue-Juan Li
- Department of Pharmacy, Shenzhen Children's Hospital, Shenzhen, 518038, China
| | - Zi-Jun Liao
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, 3002# Sungang Road, Futian District, Shenzhen, 518035, China
| | - Wei-Ping Li
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, 3002# Sungang Road, Futian District, Shenzhen, 518035, China
| | - Zong-Yang Li
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, 3002# Sungang Road, Futian District, Shenzhen, 518035, China.
| | - Lei Chen
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, 3002# Sungang Road, Futian District, Shenzhen, 518035, China.
| |
Collapse
|
3
|
Scholz N, Kurian KM, Siebzehnrubl FA, Licchesi JDF. Targeting the Ubiquitin System in Glioblastoma. Front Oncol 2020; 10:574011. [PMID: 33324551 PMCID: PMC7724090 DOI: 10.3389/fonc.2020.574011] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 10/07/2020] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma is the most common primary brain tumor in adults with poor overall outcome and 5-year survival of less than 5%. Treatment has not changed much in the last decade or so, with surgical resection and radio/chemotherapy being the main options. Glioblastoma is highly heterogeneous and frequently becomes treatment-resistant due to the ability of glioblastoma cells to adopt stem cell states facilitating tumor recurrence. Therefore, there is an urgent need for novel therapeutic strategies. The ubiquitin system, in particular E3 ubiquitin ligases and deubiquitinating enzymes, have emerged as a promising source of novel drug targets. In addition to conventional small molecule drug discovery approaches aimed at modulating enzyme activity, several new and exciting strategies are also being explored. Among these, PROteolysis TArgeting Chimeras (PROTACs) aim to harness the endogenous protein turnover machinery to direct therapeutically relevant targets, including previously considered "undruggable" ones, for proteasomal degradation. PROTAC and other strategies targeting the ubiquitin proteasome system offer new therapeutic avenues which will expand the drug development toolboxes for glioblastoma. This review will provide a comprehensive overview of E3 ubiquitin ligases and deubiquitinating enzymes in the context of glioblastoma and their involvement in core signaling pathways including EGFR, TGF-β, p53 and stemness-related pathways. Finally, we offer new insights into how these ubiquitin-dependent mechanisms could be exploited therapeutically for glioblastoma.
Collapse
Affiliation(s)
- Nico Scholz
- Department of Biology & Biochemistry, University of Bath, Bath, United Kingdom
| | - Kathreena M. Kurian
- Brain Tumour Research Group, Institute of Clinical Neurosciences, University of Bristol, Bristol, United Kingdom
| | - Florian A. Siebzehnrubl
- Cardiff University School of Biosciences, European Cancer Stem Cell Research Institute, Cardiff, United Kingdom
| | | |
Collapse
|
4
|
Wang W, Liu X, Zheng X, Jin HJ, Li X. Biomineralization: An Opportunity and Challenge of Nanoparticle Drug Delivery Systems for Cancer Therapy. Adv Healthc Mater 2020; 9:e2001117. [PMID: 33043640 DOI: 10.1002/adhm.202001117] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/29/2020] [Indexed: 12/12/2022]
Abstract
Biomineralization is a common process in organisms to produce hard biomaterials by combining inorganic ions with biomacromolecules. Multifunctional nanoplatforms are developed based on the mechanism of biomineralization in many biomedical applications. In the past few years, biomineralization-based nanoparticle drug delivery systems for the cancer treatment have gained a lot of research attention due to the advantages including simple preparation, good biocompatibility, degradability, easy modification, versatility, and targeting. In this review, the research trends of biomineralization-based nanoparticle drug delivery systems and their applications in cancer therapy are summarized. This work aims to promote future researches on cancer therapy based on biomineralization. Rational design of nanoparticle drug delivery systems can overcome the bottleneck in the clinical transformation of nanomaterials. At the same time, biomineralization has also provided new research ideas for cancer treatment, i.e., targeted therapy, which has significantly better performance.
Collapse
Affiliation(s)
- Weicai Wang
- Collaborative Innovation Center of Tumor Marker Detection Technology Equipment and Diagnosis‐Therapy Integration in Universities of Shandong Shandong Province Key Laboratory of Detection Technology for Tumor Makers School of Chemistry and Chemical Engineering Linyi University Linyi Shandong 276005 China
| | - Xiaofan Liu
- Collaborative Innovation Center of Tumor Marker Detection Technology Equipment and Diagnosis‐Therapy Integration in Universities of Shandong Shandong Province Key Laboratory of Detection Technology for Tumor Makers School of Chemistry and Chemical Engineering Linyi University Linyi Shandong 276005 China
| | - Xiangjiang Zheng
- Collaborative Innovation Center of Tumor Marker Detection Technology Equipment and Diagnosis‐Therapy Integration in Universities of Shandong Shandong Province Key Laboratory of Detection Technology for Tumor Makers School of Chemistry and Chemical Engineering Linyi University Linyi Shandong 276005 China
| | - Hyung Jong Jin
- Department of Bioscience and Biotechnology The University of Suwon Hwaseong Gyeonggi‐Do 18323 Republic of Korea
| | - Xuemei Li
- Collaborative Innovation Center of Tumor Marker Detection Technology Equipment and Diagnosis‐Therapy Integration in Universities of Shandong Shandong Province Key Laboratory of Detection Technology for Tumor Makers School of Chemistry and Chemical Engineering Linyi University Linyi Shandong 276005 China
| |
Collapse
|
5
|
Xia Q, Ali S, Liu L, Li Y, Liu X, Zhang L, Dong L. Role of Ubiquitination in PTEN Cellular Homeostasis and Its Implications in GB Drug Resistance. Front Oncol 2020; 10:1569. [PMID: 32984016 PMCID: PMC7492558 DOI: 10.3389/fonc.2020.01569] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 07/21/2020] [Indexed: 12/15/2022] Open
Abstract
Glioblastoma (GB) is the most common and aggressive brain malignancy, characterized by heterogeneity and drug resistance. PTEN, a crucial tumor suppressor, exhibits phosphatase-dependent (PI3K-AKT-mTOR pathway)/independent (nucleus stability) activities to maintain the homeostatic regulation of numerous physiological processes. Premature and absolute loss of PTEN activity usually tends to cellular senescence. However, monoallelic loss of PTEN is frequently observed at tumor inception, and absolute loss of PTEN activity also occurs at the late stage of gliomagenesis. Consequently, aberrant PTEN homeostasis, mainly regulated at the post-translational level, renders cells susceptible to tumorigenesis and drug resistance. Ubiquitination-mediated degradation or deregulated intracellular localization of PTEN hijacks cell growth rheostat control for neoplastic remodeling. Functional inactivation of PTEN mediated by the overexpression of ubiquitin ligases (E3s) renders GB cells adaptive to PTEN loss, which confers resistance to EGFR tyrosine kinase inhibitors and immunotherapies. In this review, we discuss how glioma cells develop oncogenic addiction to the E3s-PTEN axis, promoting their growth and proliferation. Antitumor strategies involving PTEN-targeting E3 ligase inhibitors can restore the tumor-suppressive environment. E3 inhibitors collectively reactivate PTEN and may represent next-generation treatment against deadly malignancies such as GB.
Collapse
Affiliation(s)
- Qin Xia
- School of Life Sciences, Beijing Institute of Technology, Beijing, China
| | - Sakhawat Ali
- School of Life Sciences, Beijing Institute of Technology, Beijing, China
| | - Liqun Liu
- School of Life Sciences, Beijing Institute of Technology, Beijing, China
| | - Yang Li
- School of Life Sciences, Beijing Institute of Technology, Beijing, China
| | - Xuefeng Liu
- School of Electronic and Optical Engineering, Nanjing University of Science and Technology, Nanjing, China
| | - Lingqiang Zhang
- State Key Laboratory of Proteomics, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China
| | - Lei Dong
- School of Life Sciences, Beijing Institute of Technology, Beijing, China
| |
Collapse
|
6
|
Ma J, Weng L, Jia Y, Liu B, Wu S, Xue L, Yin X, Mao A, Wang Z, Shang M. PTBP3 promotes malignancy and hypoxia-induced chemoresistance in pancreatic cancer cells by ATG12 up-regulation. J Cell Mol Med 2020; 24:2917-2930. [PMID: 31989778 PMCID: PMC7077536 DOI: 10.1111/jcmm.14896] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 10/17/2019] [Accepted: 11/23/2019] [Indexed: 01/05/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) tumours exhibit a high level of heterogeneity which is associated with hypoxia and strong resistance to chemotherapy. The RNA splicing protein polypyrimidine tract-binding protein 3 (PTBP3) regulates hypoxic gene expression by selectively binding to hypoxia-regulated transcripts. We have investigated the role of PTBP3 in tumour development and chemotherapeutic resistance in human PDAC tissues and pancreatic cancer cells. In addition, we determined the sensitivity of cancer cells to gemcitabine with differential levels of PTBP3 and whether autophagy and hypoxia affect gemcitabine resistance in vitro. PTBP3 expression was higher in human pancreatic cancer than in paired adjacent tissues. PTBP3 overexpression promoted PDAC proliferation in vitro and tumour growth in vivo, whereas PTBP3 depletion had opposing effects. Hypoxia significantly increased the expression of PTBP3 in pancreatic cancer cells in vitro. Under hypoxic conditions, cells were more resistance to gemcitabine. Knockdown of PTBP3 results in decreased resistance to gemcitabine, which was attributed to attenuated autophagy. We propose that PTBP3 binds to multiple sites in the 3'-UTR of ATG12 resulting in overexpression. PTBP3 increases cancer cell proliferation and autophagic flux in response to hypoxic stress, which contributes to gemcitabine resistance.
Collapse
MESH Headings
- 3' Untranslated Regions/genetics
- Adenocarcinoma/drug therapy
- Adenocarcinoma/genetics
- Adenocarcinoma/pathology
- Animals
- Autophagy/drug effects
- Autophagy/genetics
- Autophagy-Related Protein 12/genetics
- Autophagy-Related Protein 12/metabolism
- Base Sequence
- Carcinoma, Pancreatic Ductal/drug therapy
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/pathology
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Deoxycytidine/analogs & derivatives
- Deoxycytidine/pharmacology
- Deoxycytidine/therapeutic use
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Mice, Inbred BALB C
- Mice, Nude
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/pathology
- Polypyrimidine Tract-Binding Protein/metabolism
- Stress, Physiological/drug effects
- Tumor Hypoxia/drug effects
- Tumor Hypoxia/genetics
- Up-Regulation/drug effects
- Up-Regulation/genetics
- Gemcitabine
Collapse
Affiliation(s)
- Jun Ma
- Department of Interventional RadiologyTongren HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Li Weng
- Department of Interventional RadiologyTongren HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yiping Jia
- Department of Interventional RadiologyTongren HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Bingyan Liu
- Department of Interventional RadiologyTongren HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Shaoqiu Wu
- Department of Interventional RadiologyTongren HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Lei Xue
- Shanghai Key Laboratory of Signaling and Diseases ResearchSchool of Life Science and TechnologyTongji UniversityShanghaiChina
| | - Xiang Yin
- Department of Interventional RadiologyTongren HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Aiwu Mao
- Department of Interventional RadiologyTongren HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Zhongmin Wang
- Department of interventional radiologyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Mingyi Shang
- Department of Interventional RadiologyTongren HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
7
|
Mennerich D, Kubaichuk K, Kietzmann T. DUBs, Hypoxia, and Cancer. Trends Cancer 2019; 5:632-653. [PMID: 31706510 DOI: 10.1016/j.trecan.2019.08.005] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 08/24/2019] [Accepted: 08/27/2019] [Indexed: 02/08/2023]
Abstract
Alterations in protein ubiquitylation and hypoxia are commonly associated with cancer. Ubiquitylation is carried out by three sequentially acting ubiquitylating enzymes and can be opposed by deubiquitinases (DUBs), which have emerged as promising drug targets. Apart from protein localization and activity, ubiquitylation regulates degradation of proteins, among them hypoxia-inducible factors (HIFs). Thereby, various E3 ubiquitin ligases and DUBs regulate HIF abundance. Conversely, several E3s and DUBs are regulated by hypoxia. While hypoxia is a powerful HIF regulator, less is known about hypoxia-regulated DUBs and their impact on HIFs. Here, we review current knowledge about the relationship of E3s, DUBs, and hypoxia signaling. We also discuss the reciprocal regulation of DUBs by hypoxia and use of DUB-specific drugs in cancer.
Collapse
Affiliation(s)
- Daniela Mennerich
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, 90570, Finland
| | - Kateryna Kubaichuk
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, 90570, Finland
| | - Thomas Kietzmann
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, 90570, Finland; Biocenter Oulu, University of Oulu, Oulu, 90570, Finland.
| |
Collapse
|
8
|
Liu L, Shi X, Zhao H, Yang M, Wang C, Liao M, Zhao J. Nicotine induces cell survival and chemoresistance by stimulating Mcl-1 phosphorylation and its interaction with Bak in lung cancer. J Cell Physiol 2019; 234:15934-15940. [PMID: 30741422 DOI: 10.1002/jcp.28251] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 01/10/2019] [Accepted: 01/16/2019] [Indexed: 01/24/2023]
Abstract
Nicotine is a major carcinogen in cigarettes, which can enhance cell proliferation and metastasis and increase the chemoresistance of cancer cells. Our previous data found that nicotine promotes cell survival in lung cancer by affecting the expression of antiapoptotic protein Mcl-1, suggesting that the Mcl-1 may be a therapeutic target for patients with lung cancer. In this study, we found that the effects of drug resistance on nicotine-induced lung cancer cell lines were shown to influence the phosphorylation of Mcl-1. Moreover, nicotine induces Mcl-1 phosphorylation exclusively at the T163 site, which results in enhancement of the antiapoptotic activity of Mcl-1 and increased cell survival. Meanwhile, nicotine can reduce the sensitivity of H1299 cells to CDDP via enhancement of the binding of Mcl-1 to Bak, which inhibits the proapoptotic effect of Bak and ultimately leads to increased survival and drug resistance of lung cancer cells. Thus, nicotine-induced cell survival and chemoresistance may occur in a mechanism by stimulating Mcl-1 phosphorylation and its interaction with Bak, which may contribute to improving the efficacy of chemotherapy in the treatment of human lung cancer.
Collapse
Affiliation(s)
- Ling Liu
- Key Laboratory of Nanobiological Technology of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaqing Shi
- Key Laboratory of Nanobiological Technology of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Huandong Zhao
- Key Laboratory of Nanobiological Technology of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Manyi Yang
- Key Laboratory of Nanobiological Technology of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chengzhi Wang
- Key Laboratory of Nanobiological Technology of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Department of Nephrology Blood Purification Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Mingmei Liao
- Key Laboratory of Nanobiological Technology of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jinfeng Zhao
- Key Laboratory of Nanobiological Technology of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
9
|
Li X, Zhang N, Zhang Y, Jia P, Guo Y, Tian Y, You S, Wu S, Sun Y. E3 ligase Fbw7 participates in oxidative stress‑induced myocardial cell injury via interacting with Mcl‑1. Mol Med Rep 2019; 20:1561-1568. [PMID: 31257502 PMCID: PMC6625382 DOI: 10.3892/mmr.2019.10394] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Accepted: 05/15/2019] [Indexed: 12/14/2022] Open
Abstract
Oxidative stress participates in several heart diseases and is an important mechanism contributing to the pathological alterations of myocardial cell injury. In recent years, ubiquitylation has been demonstrated to be an important biochemical reaction associated with apoptosis. To investigate the effects and interactions of the E3 ligase F-box and WD repeat domain containing 7 (Fbw7) and MCL1 apoptosis regulator, BCL2 family member (Mcl-1) in myocardial cells during oxidative stress, Cell Counting Kit-8, flow cytometry, western blot, reactive oxygen species and co-immunoprecipitation assays were conducted. The current study revealed that Fbw7 may facilitate apoptosis via the Mcl-1-Bax pathway in oxidative stress-induced myocardial H9c2 cell injury. Mcl-1 inhibits the functions of Bcl-2 family members, including the mitochondrial apoptosis factor Bax, to maintain cell viability; however, the present study suggested that Fbw7 may degrade Mcl-1 and impaired this process. Therefore, it may be hypothesized that Fbw-7 promotes myocardial cell injury via interacting with Mcl-1.
Collapse
Affiliation(s)
- Xia Li
- Department of Cardiovascular Medicine, First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Naijin Zhang
- Department of Cardiovascular Medicine, First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Ying Zhang
- Department of Translational Medicine, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Pengyu Jia
- Department of Cardiovascular Medicine, First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yuxuan Guo
- Department of Cardiovascular Medicine, First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yichen Tian
- Department of Cardiovascular Medicine, First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Shilong You
- Department of Cardiovascular Medicine, First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Shaojun Wu
- Department of Cardiovascular Medicine, First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yingxian Sun
- Department of Cardiovascular Medicine, First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
10
|
Ren Y, Bi C, Zhao X, Lwin T, Wang C, Yuan J, Silva AS, Shah BD, Fang B, Li T, Koomen JM, Jiang H, Chavez JC, Pham LV, Sudalagunta PR, Wan L, Wang X, Dalton WS, Moscinski LC, Shain KH, Vose J, Cleveland JL, Sotomayor EM, Fu K, Tao J. PLK1 stabilizes a MYC-dependent kinase network in aggressive B cell lymphomas. J Clin Invest 2018; 128:5517-5530. [PMID: 30260324 PMCID: PMC6264635 DOI: 10.1172/jci122533] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 09/20/2018] [Indexed: 12/13/2022] Open
Abstract
Concordant activation of MYC and BCL-2 oncoproteins in double-hit lymphoma (DHL) results in aggressive disease that is refractory to treatment. By integrating activity-based proteomic profiling and drug screens, polo-like kinase-1 (PLK1) was identified as an essential regulator of the MYC-dependent kinome in DHL. Notably, PLK1 was expressed at high levels in DHL, correlated with MYC expression, and connoted poor outcome. Further, PLK1 signaling augmented MYC protein stability, and in turn, MYC directly induced PLK1 transcription, establishing a feed-forward MYC-PLK1 circuit in DHL. Finally, inhibition of PLK1 triggered degradation of MYC and of the antiapoptotic protein MCL-1, and PLK1 inhibitors showed synergy with BCL-2 antagonists in blocking DHL cell growth, survival, and tumorigenicity, supporting clinical targeting of PLK1 in DHL.
Collapse
Affiliation(s)
- Yuan Ren
- Department of Laboratory Medicine and Hematopathology, Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Chengfeng Bi
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Xiaohong Zhao
- Department of Laboratory Medicine and Hematopathology, Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Tint Lwin
- Department of Laboratory Medicine and Hematopathology, Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Cheng Wang
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Ji Yuan
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | | | | | - Bin Fang
- Proteomics Core Facility, Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Tao Li
- Department of Laboratory Medicine and Hematopathology, Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - John M. Koomen
- Proteomics Core Facility, Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Huijuan Jiang
- Department of Laboratory Medicine and Hematopathology, Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
- Tianjin Medical School, Tianjin, China
| | | | - Lan V. Pham
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Lixin Wan
- Department of Molecular Oncology and
| | - Xuefeng Wang
- Department of Biostatics and Bioinformatics, Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | | | - Lynn C. Moscinski
- Department of Laboratory Medicine and Hematopathology, Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | | | - Julie Vose
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - John L. Cleveland
- Department of Tumor Biology, Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Eduardo M. Sotomayor
- Department of Hematology & Oncology, George Washington University, Washington, DC, USA
| | - Kai Fu
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Jianguo Tao
- Department of Laboratory Medicine and Hematopathology, Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
- Department of Malignant Hematology, and
| |
Collapse
|
11
|
Chong SJF, Lai JXH, Eu JQ, Bellot GL, Pervaiz S. Reactive Oxygen Species and Oncoprotein Signaling-A Dangerous Liaison. Antioxid Redox Signal 2018; 29:1553-1588. [PMID: 29186971 DOI: 10.1089/ars.2017.7441] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
SIGNIFICANCE There is evidence to implicate reactive oxygen species (ROS) in tumorigenesis and its progression. This has been associated with the interplay between ROS and oncoproteins, resulting in enhanced cellular proliferation and survival. Recent Advances: To date, studies have investigated specific contributions of the crosstalk between ROS and signaling networks in cancer initiation and progression. These investigations have challenged the established dogma of ROS as agents of cell death by demonstrating a secondary function that fuels cell proliferation and survival. Studies have thus identified (onco)proteins (Bcl-2, STAT3/5, RAS, Rac1, and Myc) in manipulating ROS level as well as exploiting an altered redox environment to create a milieu conducive for cancer formation and progression. CRITICAL ISSUES Despite these advances, drug resistance and its association with an altered redox metabolism continue to pose a challenge at the mechanistic and clinical levels. Therefore, identifying specific signatures, altered protein expressions, and modifications as well as protein-protein interplay/function could not only enhance our understanding of the redox networks during cancer initiation and progression but will also provide novel targets for designing specific therapeutic strategies. FUTURE DIRECTIONS Not only a heightened realization is required to unravel various gene/protein networks associated with cancer formation and progression, particularly from the redox standpoint, but there is also a need for developing more sensitive tools for assessing cancer redox metabolism in clinical settings. This review attempts to summarize our current knowledge of the crosstalk between oncoproteins and ROS in promoting cancer cell survival and proliferation and treatment strategies employed against these oncoproteins. Antioxid. Redox Signal.
Collapse
Affiliation(s)
- Stephen Jun Fei Chong
- 1 Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore
| | - Jolin Xiao Hui Lai
- 1 Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore
| | - Jie Qing Eu
- 2 Cancer Science Institute , Singapore, Singapore
| | - Gregory Lucien Bellot
- 1 Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore .,3 Department of Hand and Reconstructive Microsurgery, National University Health System , Singapore, Singapore
| | - Shazib Pervaiz
- 1 Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore .,4 NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore , Singapore, Singapore .,5 National University Cancer Institute, National University Health System , Singapore, Singapore .,6 School of Biomedical Sciences, Curtin University , Perth, Australia
| |
Collapse
|
12
|
Wu DM, Hong XW, Wen X, Han XR, Wang S, Wang YJ, Shen M, Fan SH, Zhuang J, Zhang ZF, Shan Q, Li MQ, Hu B, Sun CH, Lu J, Zheng YL. MCL1 gene silencing promotes senescence and apoptosis of glioma cells via inhibition of the PI3K/Akt signaling pathway. IUBMB Life 2018; 71:81-92. [PMID: 30296359 DOI: 10.1002/iub.1944] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 07/27/2018] [Accepted: 08/17/2018] [Indexed: 12/13/2022]
Abstract
Glioma is known to be the most prevalent primary brain tumor. In recent years, there has been evidence indicating myeloid cell leukemia-1 (MCL1) plays a role in brain glioblastoma. Therefore, the present study was conducted with aims of exploring the ability of MCL1 silencing to influence glioma cell senescence and apoptosis through the mediation of the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) signaling pathway. Glioma and tumor-adjacent tissues were collected in order to detect the presence of higher levels of MCL1 protein expression. Next, the mRNA and protein expression of MCL1, PI3K, Akt, B cell lymphoma 2 (Bcl2), Bcl2-associated X (Bax), B lymphoma Mo-MLV insertion region 1 homolog (Bmi-1), and phosphatase and tensin homolog (PTEN) were determined. Cell counting kit-8 assay was applied to detect cell proliferation, β-galactosidase staining for cell senescence, and flow cytometry for cell cycle entry and apoptosis. Initially, the results revealed higher positive expression rate of MCL1 protein, increased mRNA and protein expression of MCL1, PI3K, Akt, Bmi-1, and Bcl-2 and decreased that of Bax and PTEN in human glioma tissues. The silencing of MCL1 resulted in a decrease in mRNA and protein expression of PI3K, Akt, Bmi-1, and Bcl-2 and an increase in Bax and PTEN expressions in glioma cells. Moreover, silencing of MCL1 also inhibited cell proliferation and cell cycle entry in glioma cells, and promoted glioma cell senescence and apoptosis. In conclusion, the aforementioned results collectively suggested that the silencing of MCL1 promotes senescence and apoptosis in glioma cells through inhibiting the PI3K/Akt signaling pathway. Thus, decreasing the expression of MCL1 might have therapeutic functions in glioma. © 2018 IUBMB Life, 71(1):81-92, 2019.
Collapse
Affiliation(s)
- Dong-Mei Wu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, 221116, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, 221116, Jiangsu Province, People's Republic of China
| | - Xiao-Wu Hong
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Xin Wen
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, 221116, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, 221116, Jiangsu Province, People's Republic of China
| | - Xin-Rui Han
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, 221116, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, 221116, Jiangsu Province, People's Republic of China
| | - Shan Wang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, 221116, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, 221116, Jiangsu Province, People's Republic of China
| | - Yong-Jian Wang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, 221116, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, 221116, Jiangsu Province, People's Republic of China
| | - Min Shen
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, 221116, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, 221116, Jiangsu Province, People's Republic of China
| | - Shao-Hua Fan
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, 221116, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, 221116, Jiangsu Province, People's Republic of China
| | - Juan Zhuang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, 221116, China.,School of Environment Science and Spatial Informatics, China University of Mining and Technology, Xuzhou, 221008, China.,Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, School of Life Sciences, Huaiyin Normal University, Huaian, 223300, China
| | - Zi-Feng Zhang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, 221116, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, 221116, Jiangsu Province, People's Republic of China
| | - Qun Shan
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, 221116, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, 221116, Jiangsu Province, People's Republic of China
| | - Meng-Qiu Li
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, 221116, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, 221116, Jiangsu Province, People's Republic of China
| | - Bin Hu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, 221116, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, 221116, Jiangsu Province, People's Republic of China
| | - Chun-Hui Sun
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, 221116, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, 221116, Jiangsu Province, People's Republic of China
| | - Jun Lu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, 221116, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, 221116, Jiangsu Province, People's Republic of China
| | - Yuan-Lin Zheng
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, 221116, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, 221116, Jiangsu Province, People's Republic of China
| |
Collapse
|
13
|
Mata-Garrido J, Tapia O, Casafont I, Berciano MT, Cuadrado A, Lafarga M. Persistent accumulation of unrepaired DNA damage in rat cortical neurons: nuclear organization and ChIP-seq analysis of damaged DNA. Acta Neuropathol Commun 2018; 6:68. [PMID: 30049290 PMCID: PMC6062993 DOI: 10.1186/s40478-018-0573-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 07/19/2018] [Indexed: 01/09/2023] Open
Abstract
Neurons are highly vulnerable to DNA damage induced by genotoxic agents such as topoisomerase activity, oxidative stress, ionizing radiation (IR) and chemotherapeutic drugs. To avert the detrimental effects of DNA lesions in genome stability, transcription and apoptosis, neurons activate robust DNA repair mechanisms. However, defective DNA repair with accumulation of unrepaired DNA are at the basis of brain ageing and several neurodegenerative diseases. Understanding the mechanisms by which neurons tolerate DNA damage accumulation as well as defining the genomic regions that are more vulnerable to DNA damage or refractory to DNA repair and therefore constitute potential targets in neurodegenerative diseases are essential issues in the field. In this work we investigated the nuclear topography and organization together with the genome-wide distribution of unrepaired DNA in rat cortical neurons 15 days upon IR. About 5% of non-irradiated and 55% of irradiated cells accumulate unrepaired DNA within persistent DNA damage foci (PDDF) of chromatin. These PDDF are featured by persistent activation of DNA damage/repair signaling, lack of transcription and localization in repressive nuclear microenvironments. Interestingly, the chromatin insulator CTCF is concentrated at the PDDF boundaries, likely contributing to isolate unrepaired DNA from intact transcriptionally active chromatin. By confining damaged DNA, PDDF would help preserving genomic integrity and preventing the production of aberrant proteins encoded by damaged genes. ChIP-seq analysis of genome-wide γH2AX distribution revealed a number of genomic regions enriched in γH2AX signal in IR-treated cortical neurons. Some of these regions are in close proximity to genes encoding essential proteins for neuronal functions and human neurodegenerative disorders such as epm2a (Lafora disease), serpini1 (familial encephalopathy with neuroserpin inclusion bodies) and il1rpl1 (mental retardation, X-linked 21). Persistent γH2AX signal close to those regions suggests that nearby genes could be either more vulnerable to DNA damage or more refractory to DNA repair.
Collapse
|
14
|
Yuan C, Pu L, He Z, Wang J. BNIP3/Bcl-2-mediated apoptosis induced by cyclic tensile stretch in human cartilage endplate-derived stem cells. Exp Ther Med 2017; 15:235-241. [PMID: 29375685 PMCID: PMC5763692 DOI: 10.3892/etm.2017.5372] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 08/24/2017] [Indexed: 01/07/2023] Open
Abstract
The present study aimed to investigate the molecular mechanisms of cyclic stretch-induced apoptosis in human intervertebral disc cartilage endplate-derived stem cells (CESCs). CESCs were stretched by the Flexercell-4000™ Tension Plus system, the effect on cell viability was measured by a Cell Counting Kit-8 assay, while cell apoptosis was detected by flow cytometry. Western blot analysis was used to evaluate the expression of B-cell lymphoma 2 (Bcl-2)/adenovirus E1B 19 kDa interacting protein 3 (BNIP3), Bcl-2, Bcl-2 homologous antagonist killer (Bak), Bcl-2-associated X protein (Bax), Bcl extra large (Bcl-xl) and the activity of caspase-3, while Z-VAD-FMK was used to inhibit caspase-3. Compared with the control group, the cell viability decreased in a time-dependent manner after stretching. Furthermore, cell apoptosis and the activity of caspase-3 were increased in a time-dependent manner. The ratio of pro-death factor BNIP3 to anti-apoptotic protein Bcl-2 was significantly increased. When cells were stretched for 36 h, the apoptosis-associated proteins Bak and Bax were increased, while Bcl-xl was decreased. The viability and apoptotic ratio of stretched cells were significantly restored after caspase-3 was repressed. In conclusion, cyclic tensile stretch induced apoptosis of CESCs, which was probably due to upregulation of the expression of BNIP3.
Collapse
Affiliation(s)
- Chao Yuan
- Department of Orthopaedics, Xinqiao Hospital, The Third Military Medical University, Chongqing 400037, P.R. China
| | - Luqiao Pu
- Department of Orthopaedics, Xinqiao Hospital, The Third Military Medical University, Chongqing 400037, P.R. China
| | - Zhiliang He
- Department of Orthopaedics, Xinqiao Hospital, The Third Military Medical University, Chongqing 400037, P.R. China
| | - Jian Wang
- Department of Orthopaedics, Xinqiao Hospital, The Third Military Medical University, Chongqing 400037, P.R. China
| |
Collapse
|
15
|
Du Y, Li J, Xu T, Zhou DD, Zhang L, Wang X. MicroRNA-145 induces apoptosis of glioma cells by targeting BNIP3 and Notch signaling. Oncotarget 2017; 8:61510-61527. [PMID: 28977881 PMCID: PMC5617441 DOI: 10.18632/oncotarget.18604] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 05/22/2017] [Indexed: 01/21/2023] Open
Abstract
MicroRNAs (miRNAs) are involved in the pathogenesis of various human cancers. Here we show that miR-145 expression is decreased in human glioma samples, rat glioma tissues, and glioma cell lines, while expression of BNIP3 is increased. Over-expression of miR-145 or suppression of BNIP3 induced glioma cell apoptosis. BNIP3 is localized in the nucleus in glioma cells, and miR-145 inhibits BNIP3 expression by binding to the 3’ untranslated region of its mRNA. Interestingly, miR-145 and BNIP3 regulate glioma cell apoptosis by modulating Notch signaling. These results indicate that miR-145 increases glioma cell apoptosis by inhibiting BNIP3 and Notch signaling, and suggest that miR-145 may serve as a novel therapeutic target for malignant glioma.
Collapse
Affiliation(s)
- Yan Du
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei 230032, China.,Institute for Liver Disease of Anhui Medical University, Anhui Medical University, Hefei 230032, China
| | - Juan Li
- Anhui Provincial Hospital, Hefei 230032, China
| | - Tao Xu
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei 230032, China.,Institute for Liver Disease of Anhui Medical University, Anhui Medical University, Hefei 230032, China
| | - Dan-Dan Zhou
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei 230032, China.,Institute for Liver Disease of Anhui Medical University, Anhui Medical University, Hefei 230032, China
| | - Lei Zhang
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei 230032, China.,Institute for Liver Disease of Anhui Medical University, Anhui Medical University, Hefei 230032, China
| | - Xiao Wang
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| |
Collapse
|