1
|
Shen Y, Ma C, Li X, Li X, Wu Y, Yang T, Hu Y, Liu C, Shen H, Guo P, Shen Y. Generation of B7-H3 isoform regulated by ANXA2/NSUN2/YBX1 axis in human glioma. J Cell Mol Med 2024; 28:e18575. [PMID: 39048916 PMCID: PMC11269050 DOI: 10.1111/jcmm.18575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 07/01/2024] [Accepted: 07/18/2024] [Indexed: 07/27/2024] Open
Abstract
In recent years, in the development of emerging immunotherapy, B7-H3 is also termed as CD276 and has become a novel chimeric antigen receptor (CAR)-T target against glioma and other tumours, and aroused extensive attention. However, B7-H3 has three isoforms (2, 3 and 4Ig) with the controversial expression and elusive function in tumour especially glioma. The current study mainly focuses on the regulatory factors and related mechanisms of generation of different B7-H3 isoforms. First, we have determined that 2Ig is dominant in glioma with high malignancy, and 4Ig is widely expressed, whereas 3Ig shows negative expression in all glioma. Next, we have further found that RNA binding protein annexin A2 (ANXA2) is essential for B7-H3 isoform maintenance, but fail to determine the choice of 4Ig or 2Ig. RNA methyltransferase NOP2/Sun RNA methyltransferase 2 (NSUN2) and 5-methylcytosine reader Y-box binding protein 1 (YBX1) facilitate the production of 2Ig. Our findings have uncovered a series of factors (ANXA2/NSUN2/YBX1) that can determine the alternative generation of different isoforms of B7-H3 in glioma. Our result aims to help peers gain a clearer understanding of the expression and regulatory mechanisms of B7H3 in tumour patients, and to provide better strategies for designing B7H3 as a target in immunotherapy.
Collapse
Affiliation(s)
- Yifen Shen
- Central Laboratory, Suzhou Bay Clinical CollegeXuzhou Medical University, Suzhou Ninth People's HospitalSuzhouJiangsuChina
| | - Chunfang Ma
- Clinical LaboratorySuzhou Ninth People's HospitalSuzhouJiangsuChina
| | - Xiangxiang Li
- Clinical LaboratorySuzhou Ninth People's HospitalSuzhouJiangsuChina
| | - Xiaosong Li
- Department of Anorectal SurgerySuzhou Ninth People's HospitalSuzhouJiangsuChina
| | - Yuxiang Wu
- Department of PathologySuzhou Ninth People's HospitalSuzhouJiangsuChina
| | - Tao Yang
- Department of Medical Cosmetology, Suzhou Ninth People's HospitalSoochow UniversitySuzhouJiangsuChina
| | - Yanping Hu
- Department of Molecular PathologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouHenanChina
| | - Chao Liu
- Central Laboratory, Suzhou Bay Clinical CollegeXuzhou Medical University, Suzhou Ninth People's HospitalSuzhouJiangsuChina
| | - Hao Shen
- Clinical LaboratorySuzhou Ninth People's HospitalSuzhouJiangsuChina
| | - Pin Guo
- Department of NeurosurgeryThe Affiliated Hospital of Qingdao UniversityQingdaoShandongChina
| | - Yihang Shen
- Central Laboratory, Suzhou Bay Clinical CollegeXuzhou Medical University, Suzhou Ninth People's HospitalSuzhouJiangsuChina
| |
Collapse
|
2
|
Zhu S, Jin Q, Zhang S, Song Z, Zhang S, Zhao Z. Integrating Network Pharmacology and Experimental Verification to Explore the Pharmacological Mechanisms of Radix Paeoniae Rubra Against Glioma. Appl Biochem Biotechnol 2024:10.1007/s12010-024-04887-6. [PMID: 38381309 DOI: 10.1007/s12010-024-04887-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2024] [Indexed: 02/22/2024]
Abstract
Glioma has a high mortality and can hardly be completely cured. Radix Paeoniae Rubra (RPR) is a prevalent component in traditional Chinese medicine used for tumor treatments. We explored the mechanism of RPR in treating glioma using network pharmacology and experiments. A network pharmacology approach was used to screen active ingredients, targets of RPR and glioma. We then constructed a herb-active ingredient-target-pathway network and conducted protein-protein interaction (PPI) network analysis, as well as Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. Molecular docking was also performed. Using CCK-8, colony formation, and xenograft experiments, we evaluated the effect of RPR on glioma. The involved pathway and proteins were identified by Western blot. From public databases, we identified nine active RPR ingredients and 40 overlapping targets among 109 RPR targets and 1360 glioma-associated targets. The PPI analysis revealed ten targets, such as AKT1, TP53, and VEGFA, which were identified as hub genes. The results from GO and KEGG analysis highlighted the involvement of the PI3K/AKT pathway. A herb-active ingredient-target-pathway network was constructed. By docking molecular structures, six suitable conformations have been identified. The RPR extract demonstrated anti-tumor properties by inhibiting glioma cell proliferation in vitro and in vivo, likely achieved by suppressing the phosphorylation of the PI3K/AKT signaling pathway. RPR concurrently downregulated the phosphorylation level of AKT1 and the protein expression level of VEGFA, while upregulating the expression of P53 in the U251 cell line. Utilizing network pharmacology and molecular docking, our study not only predicted the impact of RPR on glioma but also delineated the herb-active ingredient-target-pathway network. Experimentally, we confirmed that RPR may exert its anti-tumor properties by inhibiting the phosphorylation of the PI3K/AKT pathway, including AKT1, and by regulating the expression levels of VEGFA and P53.
Collapse
Affiliation(s)
- Siyu Zhu
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Qianxu Jin
- Department of Neurosurgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shiyang Zhang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zihan Song
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shiqi Zhang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zongmao Zhao
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
- Department of Neurosurgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| |
Collapse
|
3
|
Dinh NTM, Nguyen TM, Park MK, Lee CH. Y-Box Binding Protein 1: Unraveling the Multifaceted Role in Cancer Development and Therapeutic Potential. Int J Mol Sci 2024; 25:717. [PMID: 38255791 PMCID: PMC10815159 DOI: 10.3390/ijms25020717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 12/22/2023] [Accepted: 01/02/2024] [Indexed: 01/24/2024] Open
Abstract
Y-box binding protein 1 (YBX1), a member of the Cold Shock Domain protein family, is overexpressed in various human cancers and is recognized as an oncogenic gene associated with poor prognosis. YBX1's functional diversity arises from its capacity to interact with a broad range of DNA and RNA molecules, implicating its involvement in diverse cellular processes. Independent investigations have unveiled specific facets of YBX1's contribution to cancer development. This comprehensive review elucidates YBX1's multifaceted role in cancer across cancer hallmarks, both in cancer cell itself and the tumor microenvironment. Based on this, we proposed YBX1 as a potential target for cancer treatment. Notably, ongoing clinical trials addressing YBX1 as a target in breast cancer and lung cancer have showcased its promise for cancer therapy. The ramp up in in vitro research on targeting YBX1 compounds also underscores its growing appeal. Moreover, the emerging role of YBX1 as a neural input is also proposed where the high level of YBX1 was strongly associated with nerve cancer and neurodegenerative diseases. This review also summarized the up-to-date advanced research on the involvement of YBX1 in pancreatic cancer.
Collapse
Affiliation(s)
- Ngoc Thi Minh Dinh
- College of Pharmacy, Dongguk University, Goyang 10326, Republic of Korea; (N.T.M.D.); (T.M.N.)
| | - Tuan Minh Nguyen
- College of Pharmacy, Dongguk University, Goyang 10326, Republic of Korea; (N.T.M.D.); (T.M.N.)
| | - Mi Kyung Park
- Department of BioHealthcare, Hwasung Medi-Science University, Hwaseong-si 18274, Republic of Korea
| | - Chang Hoon Lee
- College of Pharmacy, Dongguk University, Goyang 10326, Republic of Korea; (N.T.M.D.); (T.M.N.)
| |
Collapse
|
4
|
Lan P, Li M, Wang Y, Wang J, Li L, Zhang S, Zhang X, Ran C, Zheng J, Gong H. Y-box protein-1 modulates circSPECC1 to promote glioma tumorigenesis via miR-615-5p/HIP1/AKT axis. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1902-1912. [PMID: 37994157 PMCID: PMC10753359 DOI: 10.3724/abbs.2023230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 06/13/2023] [Indexed: 11/24/2023] Open
Abstract
Y-box binding protein-1 (YB-1) is upregulated in glioma and plays an important role in its occurrence and drug resistance. However, the involved regulatory processes and downstream pathways are still unclear. Since various circular RNAs (circRNAs) and microRNAs (miRNAs) also play roles in the pathogenesis of glioma, we hypothesize that YB-1 may exert its function through a circRNA-miRNA-protein interaction network. In this study, we use the RNA binding protein immunoprecipitation assay and quantitative reverse transcription polymerase chain reaction to determine the circRNAs involved in the regulation of YB-1 and further elucidate their biological functions. The level of circSPECC1 (hsa_circ_0000745) modulated by YB-1 is significantly upregulated in the U251 and U87 glioma cell lines. Downregulation of circSPECC1 markedly inhibits the proliferation and invasiveness of U251 and U87 cells by inducing apoptosis. Bioinformatics analysis reveals that miR-615-5p could interact with circSPECC1 and huntingtin-interacting protein-1 (HIP-1). Then we determine the interactions between miR-615-5p, circSPECC1, and HIP1 using dual luciferase reporter system and pull-down assays. Mechanistic analysis indicates that the downregulation of circSPECC1 results in a decreased HIP1 expression. This study demonstrates that circSPECC1 modulated by YB-1 is increased in glioma cell lines. In addition, circSPECC1 promotes glioma growth through the upregulation of HIP1 by sponging miR-615-5p and targeting the HIP1/AKT pathway. This indicates that YB-1 and circSPECC1 may both be promising targets for glioma treatment.
Collapse
Affiliation(s)
- Ping Lan
- Department of NephrologyHospital of Nephrologythe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an710061China
| | - Meihe Li
- Department of Renal TransplantationHospital of Nephrologythe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an710061China
| | - Ying Wang
- Department of Renal TransplantationHospital of Nephrologythe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an710061China
| | - Jingwen Wang
- Department of Renal TransplantationHospital of Nephrologythe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an710061China
| | - Luyao Li
- Department of Pathologythe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an710061China
| | - Sha Zhang
- Department of Pathologythe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an710061China
| | - Xuan Zhang
- Department of Pathologythe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an710061China
| | - Caihong Ran
- Department of PathologyNgari Prefecture People’s HospitalNgari Prefecture 859099China
| | - Jin Zheng
- Department of Renal TransplantationHospital of Nephrologythe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an710061China
| | - Huilin Gong
- Department of Pathologythe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an710061China
| |
Collapse
|
5
|
Long S, Yan Y, Xu H, Wang L, Jiang J, Xu Z, Liu R, Zhou Q, Huang X, Chen J, Li Z, Wei W, Li X. Insights into the regulatory role of RNA methylation modifications in glioma. J Transl Med 2023; 21:810. [PMID: 37964279 PMCID: PMC10644640 DOI: 10.1186/s12967-023-04653-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/24/2023] [Indexed: 11/16/2023] Open
Abstract
Epitranscriptomic abnormalities, which are highly prevalent in primary central nervous system malignancies, have been identified as crucial contributors to the development and progression of gliomas. RNA epitranscriptomic modifications, particularly the reversible modification methylation, have been observed throughout the RNA cycle. Epitranscriptomic modifications, which regulate RNA transcription and translation, have profound biological implications. These modifications are associated with the development of several cancer types. Notably, three main protein types-writers, erasers, and readers, in conjunction with other related proteins, mediate these epitranscriptomic changes. This review primarily focuses on the role of recently identified RNA methylation modifications in gliomas, such as N6-methyladenosine (m6A), 5-methylcytosine (m5C), N7-methylguanosine (m7G), and N1-methyladenosine (m1A). We delved into their corresponding writers, erasers, readers, and related binding proteins to propose new approaches and prognostic indicators for patients with glioma.
Collapse
Affiliation(s)
- Shengrong Long
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Brain Research Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yu Yan
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Brain Research Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Hongyu Xu
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Brain Research Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Lesheng Wang
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Brain Research Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Jiazhi Jiang
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Brain Research Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Ziyue Xu
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Brain Research Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Runming Liu
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Brain Research Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Qiangqiang Zhou
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Brain Research Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Xiaopeng Huang
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Brain Research Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Jincao Chen
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Brain Research Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Zhiqiang Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Brain Research Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Wei Wei
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
- Brain Research Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Xiang Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
- Brain Research Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
6
|
Drug Resistance in Medulloblastoma Is Driven by YB-1, ABCB1 and a Seven-Gene Drug Signature. Cancers (Basel) 2023; 15:cancers15041086. [PMID: 36831428 PMCID: PMC9954169 DOI: 10.3390/cancers15041086] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/01/2023] [Accepted: 02/04/2023] [Indexed: 02/10/2023] Open
Abstract
Therapy resistance represents an unmet challenge in the treatment of medulloblastoma. Accordingly, the identification of targets that mark drug-resistant cell populations, or drive the proliferation of resistant cells, may improve treatment strategies. To address this, we undertook a targeted approach focused on the multi-functional transcription factor YB-1. Genetic knockdown of YB-1 in Group 3 medulloblastoma cell lines diminished cell invasion in 3D in vitro assays and increased sensitivity to standard-of-care chemotherapeutic vincristine and anti-cancer agents panobinostat and JQ1. For vincristine, this occurred in part by YB-1-mediated transcriptional regulation of multi-drug resistance gene ABCB1, as determined by chromatin immunoprecipitation. Whole transcriptome sequencing of YB-1 knockdown cells identified a role for YB-1 in the regulation of tumourigenic processes, including lipid metabolism, cell death and survival and MYC and mTOR pathways. Stable cisplatin- and vincristine-tolerant Group 3 and SHH cell lines were generated to identify additional mechanisms driving resistance to standard-of-care medulloblastoma therapy. Next-generation sequencing revealed a vastly different transcriptomic landscape following chronic drug exposure, including a drug-tolerant seven-gene expression signature, common to all sequenced drug-tolerant cell lines, representing therapeutically targetable genes implicated in the acquisition of drug tolerance. Our findings provide significant insight into mechanisms and genes underlying therapy resistance in medulloblastoma.
Collapse
|
7
|
Su T, Guan Q, Cheng H, Zhu Z, Jiang C, Guo P, Tai Y, Sun H, Wang M, Wei W, Wang Q. Functions of G protein-coupled receptor 56 in health and disease. Acta Physiol (Oxf) 2022; 236:e13866. [PMID: 35959520 DOI: 10.1111/apha.13866] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 08/05/2022] [Accepted: 08/08/2022] [Indexed: 01/29/2023]
Abstract
Human G protein-coupled receptor 56 (GPR56) is encoded by gene ADGRG1 from chromosome 16q21 and is homologously encoded in mice, at chromosome 8. Both 687 and 693 splice forms are present in humans and mice. GPR56 has a 381 amino acid-long N-terminal extracellular segment and a GPCR proteolysis site upstream from the first transmembrane domain. GPR56 is mainly expressed in the heart, brain, thyroid, platelets, and peripheral blood mononuclear cells. Accumulating evidence indicates that GPR56 promotes the formation of myelin sheaths and the development of oligodendrocytes in the cerebral cortex of the central nervous system. Moreover, GPR56 contributes to the development and differentiation of hematopoietic stem cells, induces adipogenesis, and regulates the function of immune cells. The lack of GPR56 leads to nervous system dysfunction, platelet disorders, and infertility. Abnormal expression of GPR56 is related to the malignant transformation and tumor metastasis of several cancers including melanoma, neuroglioma, and gastrointestinal cancer. Metabolic disorders and cardiovascular diseases are also associated with dysregulation of GPR56 expression, and GPR56 is involved in the pharmacological resistance to some antidepressant and cancer drug treatments. In this review, the molecular structure, expression profile, and signal transduction of GPR56 are introduced, and physiological and pathological functions of GRP56 are comprehensively summarized. Attributing to its significant biological functions and its long N-terminal extracellular region that interacts with multiple ligands, GPR56 is becoming an attractive therapeutic target in treating neurological and hematopoietic diseases.
Collapse
Affiliation(s)
- Tiantian Su
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui Province, China
| | - Qiuyun Guan
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui Province, China
| | - Huijuan Cheng
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui Province, China
| | - Zhenduo Zhu
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui Province, China
| | - Chunru Jiang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui Province, China
| | - Paipai Guo
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui Province, China
| | - Yu Tai
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui Province, China
| | - Hanfei Sun
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui Province, China
| | - Manman Wang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui Province, China
| | - Wei Wei
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui Province, China
| | - Qingtong Wang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui Province, China
| |
Collapse
|
8
|
Wang JZ, Zhu H, You P, Liu H, Wang WK, Fan X, Yang Y, Xu K, Zhu Y, Li Q, Wu P, Peng C, Wong CC, Li K, Shi Y, Zhang N, Wang X, Zeng R, Huang Y, Yang L, Wang Z, Hui J. Up-regulated YB-1 protein promotes glioblastoma growth through an YB-1/CCT4/mLST8/mTOR pathway. J Clin Invest 2022; 132:146536. [PMID: 35239512 PMCID: PMC9012288 DOI: 10.1172/jci146536] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/01/2022] [Indexed: 11/17/2022] Open
Abstract
The Y-box binding protein 1 (YB-1) is a multi-functional RNA binding protein involved in virtually each step of RNA metabolism. However, the functions and mechanisms of YB-1 in one of the most aggressive cancers, glioblastoma, are not well understood. In this study, we identified that YB-1 protein was markedly overexpressed in glioblastoma and acted as a critical activator of both mTORC1 and mTORC2 signaling. Mechanistically, YB-1 bound the 5' untranslated region (UTR) of the CCT4 mRNA to promote the translation of CCT4, a component of CCT chaperone complex, that in turn activated the mTOR signal pathway by promoting mLST8 folding. In addition, YB-1 autoregulated its own translation by binding to its 5' UTR, leading to sustained activation of mTOR signaling. In glioblastoma patients, the protein level of YB-1 positively correlated with CCT4 and mLST8 expression as well as activated mTOR signaling. Importantly, the administration of RNA decoys specifically targeting YB-1 in a mouse xenograft model resulted in slower tumor growth and better survival. Taken together, these findings uncover a disrupted proteostasis pathway involving YB-1/CCT4/mLST8/mTOR axis in promoting glioblastoma growth, suggesting that YB-1 is a potential therapeutic target for the treatment of glioblastoma.
Collapse
Affiliation(s)
- Jin-Zhu Wang
- Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Hong Zhu
- Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Pu You
- Institute of Brain-Intelligence Technology, Zhangjiang Laboratory, Shanghai, China
| | - Hui Liu
- Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Wei-Kang Wang
- Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Xiaojuan Fan
- CAS-MPG Partner Institute for Computational Biology, Chinese Academy of Sciences, Shanghai, China
| | - Yun Yang
- CAS-MPG Partner Institute for Computational Biology, Chinese Academy of Sciences, Shanghai, China
| | - Keren Xu
- Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Yingfeng Zhu
- Department of Pathology, Fudan University, Shanghai, China
| | - Qunyi Li
- Department of Pharmacy, Fudan University, Shanghai, China
| | - Ping Wu
- National Facility for Protein Science in Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Chao Peng
- National Facility for Protein Science in Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Catherine Cl Wong
- Center for Precision Medicine Multi-Omics Research, Peking University, Beijing, China
| | - Kaicheng Li
- Institute of Brain-Intelligence Technology, Zhangjiang Laboratory, Shanghai, China
| | - Yufeng Shi
- School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Nu Zhang
- Department of Neurosurgery, The 1st Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiuxing Wang
- School of Basic Medical Science, Nanjing Medical University, Nanjing, China
| | - Rong Zeng
- Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Ying Huang
- Department of General Surgery, Shanghai Jiao Tong University, Shanghai, China
| | - Liusong Yang
- Department of Neurosurgery, Fudan University, Shanghai, China
| | - Zefeng Wang
- CAS-MPG Partner Institute for Computational Biology, Chinese Academy of Sciences, Shanghai, China
| | - Jingyi Hui
- Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
9
|
Niwa Y, Kamimura K, Ogawa K, Oda C, Tanaka Y, Horigome R, Ohtsuka M, Miura H, Fujisawa K, Yamamoto N, Takami T, Okuda S, Ko M, Owaki T, Kimura A, Shibata O, Morita S, Sakai N, Abe H, Yokoo T, Sakamaki A, Kamimura H, Terai S. Cyclin D1 Binding Protein 1 Responds to DNA Damage through the ATM–CHK2 Pathway. J Clin Med 2022; 11:jcm11030851. [PMID: 35160302 PMCID: PMC8836734 DOI: 10.3390/jcm11030851] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 01/30/2022] [Accepted: 02/01/2022] [Indexed: 12/14/2022] Open
Abstract
Cyclin D1 binding protein 1 (CCNDBP1) is considered a tumor suppressor, and when expressed in tumor cells, CCNDBP1 can contribute to the viability of cancer cells by rescuing these cells from chemotherapy-induced DNA damage. Therefore, this study focused on investigating the function of CCNDBP1, which is directly related to the survival of cancer cells by escaping DNA damage and chemoresistance. Hepatocellular carcinoma (HCC) cells and tissues obtained from Ccndbp1 knockout mice were used for the in vitro and in vivo examination of the molecular mechanisms of CCNDBP1 associated with the recovery of cells from DNA damage. Subsequently, gene and protein expression changes associated with the upregulation, downregulation, and irradiation of CCNDBP1 were assessed. The overexpression of CCNDBP1 in HCC cells stimulated cell growth and showed resistance to X-ray-induced DNA damage. Gene expression analysis of CCNDBP1-overexpressed cells and Ccndbp1 knockout mice revealed that Ccndbp1 activated the Atm–Chk2 pathway through the inhibition of Ezh2 expression, accounting for resistance to DNA damage. Our study demonstrated that by inhibiting EZH2, CCNDBP1 contributed to the activation of the ATM–CHK2 pathway to alleviate DNA damage, leading to chemoresistance.
Collapse
Affiliation(s)
- Yusuke Niwa
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Niigata, Japan; (Y.N.); (K.O.); (C.O.); (Y.T.); (R.H.); (M.K.); (T.O.); (A.K.); (O.S.); (S.M.); (N.S.); (H.A.); (T.Y.); (A.S.); (H.K.); (S.T.)
| | - Kenya Kamimura
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Niigata, Japan; (Y.N.); (K.O.); (C.O.); (Y.T.); (R.H.); (M.K.); (T.O.); (A.K.); (O.S.); (S.M.); (N.S.); (H.A.); (T.Y.); (A.S.); (H.K.); (S.T.)
- Department of General Medicine, Niigata University School of Medicine, Niigata 951-8510, Niigata, Japan
- Correspondence: ; Tel.: +81-(25)-227-2207
| | - Kohei Ogawa
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Niigata, Japan; (Y.N.); (K.O.); (C.O.); (Y.T.); (R.H.); (M.K.); (T.O.); (A.K.); (O.S.); (S.M.); (N.S.); (H.A.); (T.Y.); (A.S.); (H.K.); (S.T.)
| | - Chiyumi Oda
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Niigata, Japan; (Y.N.); (K.O.); (C.O.); (Y.T.); (R.H.); (M.K.); (T.O.); (A.K.); (O.S.); (S.M.); (N.S.); (H.A.); (T.Y.); (A.S.); (H.K.); (S.T.)
| | - Yuto Tanaka
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Niigata, Japan; (Y.N.); (K.O.); (C.O.); (Y.T.); (R.H.); (M.K.); (T.O.); (A.K.); (O.S.); (S.M.); (N.S.); (H.A.); (T.Y.); (A.S.); (H.K.); (S.T.)
| | - Ryoko Horigome
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Niigata, Japan; (Y.N.); (K.O.); (C.O.); (Y.T.); (R.H.); (M.K.); (T.O.); (A.K.); (O.S.); (S.M.); (N.S.); (H.A.); (T.Y.); (A.S.); (H.K.); (S.T.)
| | - Masato Ohtsuka
- Department of Molecular Life Science, Division of Basic Medical Science and Molecular Medicine, School of Medicine, Tokai University, Isehara 259-1193, Kanagawa, Japan; (M.O.); (H.M.)
| | - Hiromi Miura
- Department of Molecular Life Science, Division of Basic Medical Science and Molecular Medicine, School of Medicine, Tokai University, Isehara 259-1193, Kanagawa, Japan; (M.O.); (H.M.)
| | - Koichi Fujisawa
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Yamaguchi, Japan; (K.F.); (N.Y.); (T.T.)
| | - Naoki Yamamoto
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Yamaguchi, Japan; (K.F.); (N.Y.); (T.T.)
| | - Taro Takami
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Yamaguchi, Japan; (K.F.); (N.Y.); (T.T.)
| | - Shujiro Okuda
- Division of Bioinformatics, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Niigata, Japan;
| | - Masayoshi Ko
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Niigata, Japan; (Y.N.); (K.O.); (C.O.); (Y.T.); (R.H.); (M.K.); (T.O.); (A.K.); (O.S.); (S.M.); (N.S.); (H.A.); (T.Y.); (A.S.); (H.K.); (S.T.)
| | - Takashi Owaki
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Niigata, Japan; (Y.N.); (K.O.); (C.O.); (Y.T.); (R.H.); (M.K.); (T.O.); (A.K.); (O.S.); (S.M.); (N.S.); (H.A.); (T.Y.); (A.S.); (H.K.); (S.T.)
| | - Atsushi Kimura
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Niigata, Japan; (Y.N.); (K.O.); (C.O.); (Y.T.); (R.H.); (M.K.); (T.O.); (A.K.); (O.S.); (S.M.); (N.S.); (H.A.); (T.Y.); (A.S.); (H.K.); (S.T.)
| | - Osamu Shibata
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Niigata, Japan; (Y.N.); (K.O.); (C.O.); (Y.T.); (R.H.); (M.K.); (T.O.); (A.K.); (O.S.); (S.M.); (N.S.); (H.A.); (T.Y.); (A.S.); (H.K.); (S.T.)
| | - Shinichi Morita
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Niigata, Japan; (Y.N.); (K.O.); (C.O.); (Y.T.); (R.H.); (M.K.); (T.O.); (A.K.); (O.S.); (S.M.); (N.S.); (H.A.); (T.Y.); (A.S.); (H.K.); (S.T.)
| | - Norihiro Sakai
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Niigata, Japan; (Y.N.); (K.O.); (C.O.); (Y.T.); (R.H.); (M.K.); (T.O.); (A.K.); (O.S.); (S.M.); (N.S.); (H.A.); (T.Y.); (A.S.); (H.K.); (S.T.)
| | - Hiroyuki Abe
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Niigata, Japan; (Y.N.); (K.O.); (C.O.); (Y.T.); (R.H.); (M.K.); (T.O.); (A.K.); (O.S.); (S.M.); (N.S.); (H.A.); (T.Y.); (A.S.); (H.K.); (S.T.)
| | - Takeshi Yokoo
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Niigata, Japan; (Y.N.); (K.O.); (C.O.); (Y.T.); (R.H.); (M.K.); (T.O.); (A.K.); (O.S.); (S.M.); (N.S.); (H.A.); (T.Y.); (A.S.); (H.K.); (S.T.)
| | - Akira Sakamaki
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Niigata, Japan; (Y.N.); (K.O.); (C.O.); (Y.T.); (R.H.); (M.K.); (T.O.); (A.K.); (O.S.); (S.M.); (N.S.); (H.A.); (T.Y.); (A.S.); (H.K.); (S.T.)
| | - Hiroteru Kamimura
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Niigata, Japan; (Y.N.); (K.O.); (C.O.); (Y.T.); (R.H.); (M.K.); (T.O.); (A.K.); (O.S.); (S.M.); (N.S.); (H.A.); (T.Y.); (A.S.); (H.K.); (S.T.)
| | - Shuji Terai
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Niigata, Japan; (Y.N.); (K.O.); (C.O.); (Y.T.); (R.H.); (M.K.); (T.O.); (A.K.); (O.S.); (S.M.); (N.S.); (H.A.); (T.Y.); (A.S.); (H.K.); (S.T.)
| |
Collapse
|
10
|
Mai S, Liang L, Mai G, Liu X, Diao D, Cai R, Liu L. Development and Validation of Lactate Metabolism-Related lncRNA Signature as a Prognostic Model for Lung Adenocarcinoma. Front Endocrinol (Lausanne) 2022; 13:829175. [PMID: 35422758 PMCID: PMC9004472 DOI: 10.3389/fendo.2022.829175] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 02/21/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Lung cancer has been a prominent research focus in recent years due to its role in cancer-related fatalities globally, with lung adenocarcinoma (LUAD) being the most prevalent histological form. Nonetheless, no signature of lactate metabolism-related long non-coding RNAs (LMR-lncRNAs) has been developed for patients with LUAD. Accordingly, we aimed to develop a unique LMR-lncRNA signature to determine the prognosis of patients with LUAD. METHOD The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases were utilized to derive the lncRNA expression patterns. Identification of LMR-lncRNAs was accomplished by analyzing the co-expression patterns between lncRNAs and LMR genes. Subsequently, the association between lncRNA levels and survival outcomes was determined to develop an effective signature. In the TCGA cohort, Cox regression was enlisted to build an innovative signature consisting of three LMR-lncRNAs, which was validated in the GEO validation cohort. GSEA and immune infiltration analysis were conducted to investigate the functional annotation of the signature and the function of each type of immune cell. RESULTS Fourteen differentially expressed LMR-lncRNAs were strongly correlated with the prognosis of patients with LUAD and collectively formed a new LMR-lncRNA signature. The patients could be categorized into two cohorts based on their LMR-lncRNA signatures: a low-risk and high-risk group. The overall survival of patients with LUAD in the high-risk group was considerably lower than those in the low-risk group. Using Cox regression, this signature was shown to have substantial potential as an independent prognostic factor, which was further confirmed in the GEO cohort. Moreover, the signature could anticipate survival across different groups based on stage, age, and gender, among other variables. This signature also correlated with immune cell infiltration (including B cells, neutrophils, CD4+ T cells, CD8+ T cells, etc.) as well as the immune checkpoint blockade target CTLA-4. CONCLUSION We developed and verified a new LMR-lncRNA signature useful for anticipating the survival of patients with LUAD. This signature could give potentially critical insight for immunotherapy interventions in patients with LUAD.
Collapse
Affiliation(s)
- Shijie Mai
- Department of Thoracic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Liping Liang
- Department of Gastroenterology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Genghui Mai
- Department of Gastroenterology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiguang Liu
- Department of Thoracic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Dingwei Diao
- Department of Thoracic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ruijun Cai
- Department of Thoracic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Le Liu, ; Ruijun Cai,
| | - Le Liu
- Department of Gastroenterology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
- *Correspondence: Le Liu, ; Ruijun Cai,
| |
Collapse
|
11
|
Lei Y, Yang Q, Nie Y, Wan J, Deng M. Small-molecule inhibitor LF3 restrains the development of pulmonary hypertension through the Wnt/β-catenin pathway. Acta Biochim Biophys Sin (Shanghai) 2021; 53:1277-1289. [PMID: 34410330 DOI: 10.1093/abbs/gmab103] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Indexed: 12/23/2022] Open
Abstract
Pulmonary hypertension (PH) associated with congenital heart disease is a progressive hemodynamic disease that can lead to increased pulmonary vascular resistance, vascular remodeling, and even right heart failure and death. LF3 is a novel inhibitor of the reporter gene activity of β-catenin/TCF4 interaction in the Wnt/β-catenin signal pathway. However, whether this action of LF3 can prevent PH development remains unclear. In this study, we investigated the therapeutic effect of LF3 in rat primary pulmonary artery smooth muscle cells (PASMCs) of the PH model. We found that LF3 inhibited the decrease in pulmonary artery acceleration time and ejection time by ultra-high-resolution ultrasound imaging and blocked the increase of pulmonary artery systolic pressure by using the BL420 biological function experimental system and right ventricular hypertrophy index by the electronic scales. Simultaneously, it prevented the increase of α-smooth muscle actin and fibronectin and the decrease of elastin in pulmonary arteries of rats in the PH group, as revealed by an immunohistochemical analysis. Moreover, cell proliferation and migration assays showed that LF3 significantly reduced the proliferation and migration of PASMCs. Western blotting and quantitative real-time polymerase chain reaction analyses revealed that LF3 suppressed the expression of proliferating cell nuclear antigens and Bcl-2 and increased the expression of Bax but did not alter the expressions of β-catenin and TCF4. Taken together, LF3 can reduce the migration and proliferation of PASMCs and induce their apoptosis to prevent the development of PH. It would be worthwhile to explore the potential use of LF3 in the treatment of PH.
Collapse
Affiliation(s)
- Yong Lei
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
- Department of Cardiothoracic Surgery, Nanchong Center Hospital, The Second Clinical College, North Sichuan Medical College, Nanchong 637000, China
| | - Qi Yang
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Yongmei Nie
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
- Key Laboratory of Cardiovascular and Metabolic of Luzhou City, Luzhou 646000, China
| | - Juyi Wan
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
- Key Laboratory of Cardiovascular and Metabolic of Luzhou City, Luzhou 646000, China
- Sichuan Clinical Research Center for Birth Defects, Luzhou 646000, China
| | - Mingbin Deng
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
- Key Laboratory of Cardiovascular and Metabolic of Luzhou City, Luzhou 646000, China
- Sichuan Clinical Research Center for Birth Defects, Luzhou 646000, China
| |
Collapse
|
12
|
Yang JW, Sun C, Jin QY, Qiao XH, Guo XL. Potential therapeutic strategies for targeting Y-box-binding protein 1 in cancers. Curr Cancer Drug Targets 2021; 21:897-906. [PMID: 34465278 DOI: 10.2174/1568009621666210831125001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/27/2021] [Accepted: 06/07/2021] [Indexed: 11/22/2022]
Abstract
As one of the most conservative proteins in evolution, Y-box-binding protein 1 (YB-1) has long been considered as a potential cancer target. YB-1 is usually poorly expressed in normal cells and exerts cellular physiological functions such as DNA repair, pre-mRNA splicing and mRNA stabilizing. In cancer cells, the expression of YB-1 is up-regulated and undergoes nuclear translocation and contributes to tumorigenesis, angiogenesis, tumor proliferation, invasion, migration and chemotherapy drug resistance. During the past decades, a variety of pharmacological tools such as siRNA, shRNA, microRNA, circular RNA, lncRNA and various compounds have been developed to target YB-1 for cancer therapy. In this review, we describe the physiological characteristics of YB-1 in detail, highlight the role of YB-1 in tumors and summarize the current therapeutic methods for targeting YB-1 in cancer.
Collapse
Affiliation(s)
- Jia-Wei Yang
- Department of Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012. China
| | - Chao Sun
- Department of Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012. China
| | - Qiu-Yang Jin
- Department of Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012. China
| | - Xing-Hui Qiao
- Department of Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012. China
| | - Xiu-Li Guo
- Department of Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012. China
| |
Collapse
|
13
|
Zhang Q, Han Z, Zhu Y, Chen J, Li W. Role of hypoxia inducible factor-1 in cancer stem cells (Review). Mol Med Rep 2020; 23:17. [PMID: 33179080 PMCID: PMC7673349 DOI: 10.3892/mmr.2020.11655] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 09/25/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer stem cells (CSCs) have been found to play a decisive role in cancer recurrence, metastasis, and chemo‑, radio‑ and immuno‑resistance. Understanding the mechanism of CSC self‑renewal and proliferation may help overcome the limitations of clinical treatment. The microenvironment of tumor growth consists of a lack of oxygen, and hypoxia has been confirmed to induce cancer cell invasion, metastasis and epithelial‑mesenchymal transition, and is usually associated with poor prognosis and low survival rates. Hypoxia inducible factor‑1 (HIF‑1) can be stably expressed under hypoxia and act as an important molecule to regulate the development of CSCs, but the specific mechanism remains unclear. The present review attempted to explain the role of HIF‑1 in the generation and maintenance of CSCs from the perspective of epigenetics, metabolic reprogramming, tumor immunity, CSC markers, non‑coding RNA and signaling pathways associated with HIF‑1, in order to provide novel targets with HIF‑1 as the core for clinical treatment, and extend the life of patients.
Collapse
Affiliation(s)
- Qi Zhang
- Stem Cell and Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 130061, P.R. China
| | - Zhenzhen Han
- Stem Cell and Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 130061, P.R. China
| | - Yanbo Zhu
- Stem Cell and Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 130061, P.R. China
| | - Jingcheng Chen
- Stem Cell and Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 130061, P.R. China
| | - Wei Li
- Stem Cell and Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 130061, P.R. China
| |
Collapse
|
14
|
Mehta S, Algie M, Al-Jabry T, McKinney C, Kannan S, Verma CS, Ma W, Zhang J, Bartolec TK, Masamsetti VP, Parker K, Henderson L, Gould ML, Bhatia P, Harfoot R, Chircop M, Kleffmann T, Cohen SB, Woolley AG, Cesare AJ, Braithwaite A. Critical Role for Cold Shock Protein YB-1 in Cytokinesis. Cancers (Basel) 2020; 12:cancers12092473. [PMID: 32882852 PMCID: PMC7565962 DOI: 10.3390/cancers12092473] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/21/2020] [Accepted: 08/27/2020] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Y-box-binding protein-1, YB-1, plays an important role in regulating the cell cycle, although precisely how it does the is unknown. Using live cell imaging, we show that YB-1 is essential for initiating the last step of cell division (cytokinesis), required for creation of two daughter cells. Using confocal microscopy we showed that YB-1 regulates the spatial distribution of key proteins essential for cytokinesis to occur and that this required YB-1 to be phosphorylated on several residues. In-silico modeling demonstrated that modifications at these residues resulted in conformational changes in YB-1 protein allowing it to interact with proteins essential for cytokinesis. As many cancers have high levels YB-1 and these are associated with poor prognosis, our data suggest developing small molecule inhibitors to block YB-1 phosphorylation could be a novel approach to cancer therapy. Abstract High levels of the cold shock protein Y-box-binding protein-1, YB-1, are tightly correlated with increased cell proliferation and progression. However, the precise mechanism by which YB-1 regulates proliferation is unknown. Here, we found that YB-1 depletion in several cancer cell lines and in immortalized fibroblasts resulted in cytokinesis failure and consequent multinucleation. Rescue experiments indicated that YB-1 was required for completion of cytokinesis. Using confocal imaging we found that YB-1 was essential for orchestrating the spatio-temporal distribution of the microtubules, β-actin and the chromosome passenger complex (CPC) to define the cleavage plane. We show that phosphorylation at six serine residues was essential for cytokinesis, of which novel sites were identified using mass spectrometry. Using atomistic modelling we show how phosphorylation at multiple sites alters YB-1 conformation, allowing it to interact with protein partners. Our results establish phosphorylated YB-1 as a critical regulator of cytokinesis, defining precisely how YB-1 regulates cell division.
Collapse
Affiliation(s)
- Sunali Mehta
- Department of Pathology, University of Otago, 9016 Dunedin, New Zealand; (M.A.); (C.M.); (K.P.); (L.H.); (M.L.G.); (P.B.); (R.H.); (A.G.W.); (A.B.)
- Maurice Wilkins Centre for Biodiscovery, University of Otago, 9016 Dunedin, New Zealand
- Correspondence: ; Tel.: +64-3-4797169
| | - Michael Algie
- Department of Pathology, University of Otago, 9016 Dunedin, New Zealand; (M.A.); (C.M.); (K.P.); (L.H.); (M.L.G.); (P.B.); (R.H.); (A.G.W.); (A.B.)
- Centre for Protein Research, Department of Biochemistry, University of Otago, 9054 Dunedin, New Zealand;
| | - Tariq Al-Jabry
- Children’s Medical Research Institute, University of Sydney, Westmead, NSW 2145, Australia; (T.S.-J.); (W.M.); (J.Z.); (T.K.B.); (V.P.M.); (M.C.); (S.B.C.); (A.J.C.)
| | - Cushla McKinney
- Department of Pathology, University of Otago, 9016 Dunedin, New Zealand; (M.A.); (C.M.); (K.P.); (L.H.); (M.L.G.); (P.B.); (R.H.); (A.G.W.); (A.B.)
| | - Srinivasaraghavan Kannan
- Department of Biomolecular Modelling and Design, Bioinformatics Institute (A*STAR), 30 Biopolis Street, 07-01 Matrix, Singapore 138671, Singapore; (S.K.); (C.S.V.)
| | - Chandra S Verma
- Department of Biomolecular Modelling and Design, Bioinformatics Institute (A*STAR), 30 Biopolis Street, 07-01 Matrix, Singapore 138671, Singapore; (S.K.); (C.S.V.)
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
- Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore 117543, Singapore
| | - Weini Ma
- Children’s Medical Research Institute, University of Sydney, Westmead, NSW 2145, Australia; (T.S.-J.); (W.M.); (J.Z.); (T.K.B.); (V.P.M.); (M.C.); (S.B.C.); (A.J.C.)
| | - Jessie Zhang
- Children’s Medical Research Institute, University of Sydney, Westmead, NSW 2145, Australia; (T.S.-J.); (W.M.); (J.Z.); (T.K.B.); (V.P.M.); (M.C.); (S.B.C.); (A.J.C.)
| | - Tara K. Bartolec
- Children’s Medical Research Institute, University of Sydney, Westmead, NSW 2145, Australia; (T.S.-J.); (W.M.); (J.Z.); (T.K.B.); (V.P.M.); (M.C.); (S.B.C.); (A.J.C.)
| | - V. Pragathi Masamsetti
- Children’s Medical Research Institute, University of Sydney, Westmead, NSW 2145, Australia; (T.S.-J.); (W.M.); (J.Z.); (T.K.B.); (V.P.M.); (M.C.); (S.B.C.); (A.J.C.)
| | - Kim Parker
- Department of Pathology, University of Otago, 9016 Dunedin, New Zealand; (M.A.); (C.M.); (K.P.); (L.H.); (M.L.G.); (P.B.); (R.H.); (A.G.W.); (A.B.)
| | - Luke Henderson
- Department of Pathology, University of Otago, 9016 Dunedin, New Zealand; (M.A.); (C.M.); (K.P.); (L.H.); (M.L.G.); (P.B.); (R.H.); (A.G.W.); (A.B.)
- Maurice Wilkins Centre for Biodiscovery, University of Otago, 9016 Dunedin, New Zealand
| | - Maree L Gould
- Department of Pathology, University of Otago, 9016 Dunedin, New Zealand; (M.A.); (C.M.); (K.P.); (L.H.); (M.L.G.); (P.B.); (R.H.); (A.G.W.); (A.B.)
| | - Puja Bhatia
- Department of Pathology, University of Otago, 9016 Dunedin, New Zealand; (M.A.); (C.M.); (K.P.); (L.H.); (M.L.G.); (P.B.); (R.H.); (A.G.W.); (A.B.)
| | - Rhodri Harfoot
- Department of Pathology, University of Otago, 9016 Dunedin, New Zealand; (M.A.); (C.M.); (K.P.); (L.H.); (M.L.G.); (P.B.); (R.H.); (A.G.W.); (A.B.)
| | - Megan Chircop
- Children’s Medical Research Institute, University of Sydney, Westmead, NSW 2145, Australia; (T.S.-J.); (W.M.); (J.Z.); (T.K.B.); (V.P.M.); (M.C.); (S.B.C.); (A.J.C.)
| | - Torsten Kleffmann
- Centre for Protein Research, Department of Biochemistry, University of Otago, 9054 Dunedin, New Zealand;
| | - Scott B Cohen
- Children’s Medical Research Institute, University of Sydney, Westmead, NSW 2145, Australia; (T.S.-J.); (W.M.); (J.Z.); (T.K.B.); (V.P.M.); (M.C.); (S.B.C.); (A.J.C.)
| | - Adele G Woolley
- Department of Pathology, University of Otago, 9016 Dunedin, New Zealand; (M.A.); (C.M.); (K.P.); (L.H.); (M.L.G.); (P.B.); (R.H.); (A.G.W.); (A.B.)
- Maurice Wilkins Centre for Biodiscovery, University of Otago, 9016 Dunedin, New Zealand
| | - Anthony J Cesare
- Children’s Medical Research Institute, University of Sydney, Westmead, NSW 2145, Australia; (T.S.-J.); (W.M.); (J.Z.); (T.K.B.); (V.P.M.); (M.C.); (S.B.C.); (A.J.C.)
| | - Antony Braithwaite
- Department of Pathology, University of Otago, 9016 Dunedin, New Zealand; (M.A.); (C.M.); (K.P.); (L.H.); (M.L.G.); (P.B.); (R.H.); (A.G.W.); (A.B.)
- Maurice Wilkins Centre for Biodiscovery, University of Otago, 9016 Dunedin, New Zealand
- Children’s Medical Research Institute, University of Sydney, Westmead, NSW 2145, Australia; (T.S.-J.); (W.M.); (J.Z.); (T.K.B.); (V.P.M.); (M.C.); (S.B.C.); (A.J.C.)
- Malaghan Institute of Medical Research, 6242 Wellington, New Zealand
| |
Collapse
|