1
|
Rithvik A, Wadhavane S, Rasool M. Decoding poly (RC)-binding protein 1 (PCBP1), the underrated guard at the foothill of ferroptosis. Pathol Res Pract 2025; 266:155771. [PMID: 39700662 DOI: 10.1016/j.prp.2024.155771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 12/21/2024]
Abstract
PCBP1 is a multifunctional adaptor protein, whose function as an iron chaperone and epigenetic regulator of several chemical messengers involved in ferroptosis has garnered much attention. Herein, this review, several attempts have been made to simplify our understanding of the complex roles of PCBP1. The review begins by elucidating the relevance of PCBP1 in key events governing ferroptosis. We expeditiously shed light on some of the important mechanisms that have critical implications for the ferroptosis landscape. For instance, senescence, EMT, hypoxia, and regulation of the cell cycle and immune checkpoints, among others, have been demonstrated to influence ferroptosis sensitivity to varying degrees. Thus, this review entails a conscious attempt to carefully examine the relevance of PCBP1 in such potential mechanisms. Furthermore, we investigated the therapeutic relevance of PCBP1 in tumor biology and autoimmunity, while underscoring the contrasting perspective of ferroptosis targeting across the disease spectrum. Finally, we debate the different strategies that can be exploited to target PCBP1 in promoting or inhibiting ferroptosis.
Collapse
Affiliation(s)
- Arulkumaran Rithvik
- Immunopathology Lab, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India
| | - Sakshi Wadhavane
- Immunopathology Lab, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India
| | - Mahaboobkhan Rasool
- Immunopathology Lab, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India.
| |
Collapse
|
2
|
Wang F, Fan Y, Li Y, Zhou Y, Wang X, Zhu M, Chen X, Xue Y, Shen C. Identification of differentially expressed genes of blood leukocytes for Schizophrenia. Front Genet 2024; 15:1398240. [PMID: 38988837 PMCID: PMC11233772 DOI: 10.3389/fgene.2024.1398240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 06/03/2024] [Indexed: 07/12/2024] Open
Abstract
Background Schizophrenia (SCZ) is a severe neurodevelopmental disorder with brain dysfunction. This study aimed to use bioinformatic analysis to identify candidate blood biomarkers for SCZ. Methods The study collected peripheral blood leukocyte samples of 9 SCZ patients and 20 healthy controls for RNA sequencing analysis. Bioinformatic analyses included differentially expressed genes (DEGs) analysis, pathway enrichment analysis, and weighted gene co-expression network analysis (WGCNA). Results This study identified 1,205 statistically significant DEGs, of which 623 genes were upregulated and 582 genes were downregulated. Functional enrichment analysis showed that DEGs were mainly enriched in cell chemotaxis, cell surface, and serine peptidase activity, as well as involved in Natural killer cell-mediated cytotoxicity. WGCNA identified 16 gene co-expression modules, and five modules were significantly correlated with SCZ (p < 0.05). There were 106 upregulated genes and 90 downregulated genes in the five modules. The top ten genes sorted by the Degree algorithm were RPS28, BRD4, FUS, PABPC1, PCBP1, PCBP2, RPL27A, RPS21, RAG1, and RPL27. RAG1 and the other nine genes belonged to the turquoise and pink module respectively. Pathway enrichment analysis indicated that these 10 genes were mainly involved in processes such as Ribosome, cytoplasmic translation, RNA binding, and protein binding. Conclusion This study finds that the gene functions in key modules and related enrichment pathways may help to elucidate the molecular pathogenesis of SCZ, and the potential of key genes to become blood biomarkers for SCZ warrants further validation.
Collapse
Affiliation(s)
- Feifan Wang
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yao Fan
- Department of Clinical Epidemiology, Jiangsu Province Geriatric Institute, Geriatric Hospital of Nanjing Medical University, Nanjing, China
| | - Yinghui Li
- Department of Medical Psychology, Huai'an Third Hospital, Huai'an, China
| | - Yuan Zhou
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xin Wang
- Department of Medical Laboratory, Huai'an Third Hospital, Huai'an, China
| | - Mengya Zhu
- Department of Medical Laboratory, Huai'an Third Hospital, Huai'an, China
| | - Xuefei Chen
- Department of Medical Laboratory, Huai'an Third Hospital, Huai'an, China
| | - Yong Xue
- Department of Medical Laboratory, Huai'an Third Hospital, Huai'an, China
| | - Chong Shen
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
3
|
Su R, Kang X, Niu Y, Zhao T, Wang H. PCBP1 interacts with the HTLV-1 Tax oncoprotein to potentiate NF-κB activation. Front Immunol 2024; 15:1375168. [PMID: 38690287 PMCID: PMC11058652 DOI: 10.3389/fimmu.2024.1375168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 03/25/2024] [Indexed: 05/02/2024] Open
Abstract
Human T-cell leukemia virus type 1 (HTLV-1) is the etiological agent of adult T-cell leukemia/lymphoma. The HTLV-1 Tax constitutively activates nuclear factor-κB (NF-κB) to promote the survival and transformation of HTLV-1-infected T cells. Despite extensive study of Tax, how Tax interacts with host factors to regulate NF-κB activation and HTLV-1-driven cell proliferation is not entirely clear. Here, we showed that overexpression of Poly (rC)-binding protein 1 (PCBP1) promoted Tax-mediated IκB kinase (IKK)-NF-κB signaling activation, whereas knockdown of PCBP1 attenuated Tax-dependent IKK-NF-κB activation. However, Tax activation of HTLV-1 long terminal repeat was unaffected by PCBP1. Furthermore, depletion of PCBP1 led to apoptosis and reduced proliferation of HTLV-1-transformed cells. Mechanistically, PCBP1 interacted and co-localized with Tax in the cytoplasm, and PCBP1 KH3 domain was indispensable for the interaction between PCBP1 and Tax. Moreover, PCBP1 facilitated the assembly of Tax/IKK complex. Collectively, our results demonstrated that PCBP1 may exert an essential effect in Tax/IKK complex combination and subsequent NF-κB activation, which provides a novel insight into the pathogenetic mechanisms of HTLV-1.
Collapse
Affiliation(s)
- Rui Su
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
- Henan Key Laboratory of Immunology and Targeted Drug, Xinxiang Medical University, Xinxiang, China
| | - Xue Kang
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
- Henan Key Laboratory of Immunology and Targeted Drug, Xinxiang Medical University, Xinxiang, China
| | - Yifan Niu
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
- Henan Key Laboratory of Immunology and Targeted Drug, Xinxiang Medical University, Xinxiang, China
| | - Tiesuo Zhao
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, China
| | - Hui Wang
- Henan Key Laboratory of Immunology and Targeted Drug, Xinxiang Medical University, Xinxiang, China
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
4
|
Song Y, Wang L, Xu M, Lu X, Wang Y, Zhang L. Molecular and functional characterization of porcine poly C binding protein 1 (PCBP1). BMC Vet Res 2024; 20:25. [PMID: 38218813 PMCID: PMC10787444 DOI: 10.1186/s12917-023-03861-4] [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: 02/13/2023] [Accepted: 12/20/2023] [Indexed: 01/15/2024] Open
Abstract
BACKGROUND Poly C Binding Protein 1 (PCBP1) belongs to the heterogeneous nuclear ribonucleoprotein family. It is a multifunctional protein that participates in several functional circuits and plays a variety of roles in cellular processes. Although PCBP1 has been identified in several mammals, its function in porcine was unclear. RESULTS In this study, we cloned the gene of porcine PCBP1 and analyzed its evolutionary relationships among different species. We found porcine PCBP1 protein sequence was similar to that of other animals. The subcellular localization of PCBP1 in porcine kidney cells 15 (PK-15) cells was analyzed by immunofluorescence assay (IFA) and revealed that PCBP1 was mainly localized to the nucleus. Reverse transcription-quantitative PCR (RT-qPCR) was used to compare PCBP1 mRNA levels in different tissues of 30-day-old pigs. Results indicated that PCBP1 was expressed in various tissues and was most abundant in the liver. Finally, the effects of PCBP1 on cell cycle and apoptosis were investigated following its overexpression or knockdown in PK-15 cells. The findings demonstrated that PCBP1 knockdown arrested cell cycle in G0/G1 phase, and enhanced cell apoptosis. CONCLUSIONS Porcine PCBP1 is a highly conserved protein, plays an important role in determining cell fate, and its functions need further study.
Collapse
Affiliation(s)
- Yue Song
- Molecule Biology Laboratory of Zhengzhou Normal University, Zhengzhou Henan, 450044, China
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, 450046, China
| | - Linqing Wang
- Molecule Biology Laboratory of Zhengzhou Normal University, Zhengzhou Henan, 450044, China.
| | - Menglong Xu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, 450046, China
| | - Xiuxiang Lu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, 450046, China
| | - Yumin Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, 450046, China
| | - Limeng Zhang
- Molecule Biology Laboratory of Zhengzhou Normal University, Zhengzhou Henan, 450044, China
| |
Collapse
|
5
|
Kumar V, Bauer C, Stewart JH. Cancer cell-specific cGAS/STING Signaling pathway in the era of advancing cancer cell biology. Eur J Cell Biol 2023; 102:151338. [PMID: 37423035 DOI: 10.1016/j.ejcb.2023.151338] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 06/27/2023] [Accepted: 07/04/2023] [Indexed: 07/11/2023] Open
Abstract
Pattern-recognition receptors (PRRs) are critical to recognizing endogenous and exogenous threats to mount a protective proinflammatory innate immune response. PRRs may be located on the outer cell membrane, cytosol, and nucleus. The cGAS/STING signaling pathway is a cytosolic PRR system. Notably, cGAS is also present in the nucleus. The cGAS-mediated recognition of cytosolic dsDNA and its cleavage into cGAMP activates STING. Furthermore, STING activation through its downstream signaling triggers different interferon-stimulating genes (ISGs), initiating the release of type 1 interferons (IFNs) and NF-κB-mediated release of proinflammatory cytokines and molecules. Activating cGAS/STING generates type 1 IFN, which may prevent cellular transformation and cancer development, growth, and metastasis. The current article delineates the impact of the cancer cell-specific cGAS/STING signaling pathway alteration in tumors and its impact on tumor growth and metastasis. This article further discusses different approaches to specifically target cGAS/STING signaling in cancer cells to inhibit tumor growth and metastasis in conjunction with existing anticancer therapies.
Collapse
Affiliation(s)
- Vijay Kumar
- Department of Interdisciplinary Oncology, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University Health Science Center (LSUHSC), 1700 Tulane Avenue, New Orleans, LA 70012, USA.
| | - Caitlin Bauer
- Department of Interdisciplinary Oncology, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University Health Science Center (LSUHSC), 1700 Tulane Avenue, New Orleans, LA 70012, USA
| | - John H Stewart
- Department of Interdisciplinary Oncology, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University Health Science Center (LSUHSC), 1700 Tulane Avenue, New Orleans, LA 70012, USA; Louisiana Children's Medical Center Cancer Center, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University Health Science Center (LSUHSC), 1700 Tulane Avenue, New Orleans, LA 70012, USA.
| |
Collapse
|
6
|
Meng Y, Huang K, Shi M, Huo Y, Han L, Liu B, Li Y. Research Advances in the Role of the Tropomyosin Family in Cancer. Int J Mol Sci 2023; 24:13295. [PMID: 37686101 PMCID: PMC10488083 DOI: 10.3390/ijms241713295] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 08/22/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
Cancer is one of the most difficult diseases for human beings to overcome. Its development is closely related to a variety of factors, and its specific mechanisms have been a hot research topic in the field of scientific research. The tropomyosin family (Tpm) is a group of proteins closely related to the cytoskeleton and actin, and recent studies have shown that they play an important role in various cancers, participating in a variety of biological activities, including cell proliferation, invasion, and migration, and have been used as biomarkers for various cancers. The purpose of this review is to explore the research progress of the Tpm family in tumorigenesis development, focusing on the molecular pathways associated with them and their relevant activities involved in tumors. PubMed and Web of Science databases were searched for relevant studies on the role of Tpms in tumorigenesis and development and the activities of Tpms involved in tumors. Data from the literature suggest that the Tpm family is involved in tumor cell proliferation and growth, tumor cell invasion and migration, tumor angiogenesis, tumor cell apoptosis, and immune infiltration of the tumor microenvironment, among other correlations. It can be used as a potential biomarker for early diagnosis, follow-up, and therapeutic response of some tumors. The Tpm family is involved in cancer in a close relationship with miRNAs and LncRNAs. Tpms are involved in tumor tissue invasion and migration as a key link. On this basis, TPM is frequently used as a biomarker for various cancers. However, the specific molecular mechanism of its involvement in cancer progression has not been explained clearly, which remains an important direction for future research.
Collapse
Affiliation(s)
- Yucheng Meng
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School of Stomatology, Lanzhou University, Lanzhou 730030, China; (Y.M.); (K.H.); (M.S.); (Y.H.); (L.H.)
| | - Ke Huang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School of Stomatology, Lanzhou University, Lanzhou 730030, China; (Y.M.); (K.H.); (M.S.); (Y.H.); (L.H.)
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730030, China
| | - Mingxuan Shi
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School of Stomatology, Lanzhou University, Lanzhou 730030, China; (Y.M.); (K.H.); (M.S.); (Y.H.); (L.H.)
| | - Yifei Huo
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School of Stomatology, Lanzhou University, Lanzhou 730030, China; (Y.M.); (K.H.); (M.S.); (Y.H.); (L.H.)
| | - Liang Han
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School of Stomatology, Lanzhou University, Lanzhou 730030, China; (Y.M.); (K.H.); (M.S.); (Y.H.); (L.H.)
| | - Bin Liu
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School of Stomatology, Lanzhou University, Lanzhou 730030, China; (Y.M.); (K.H.); (M.S.); (Y.H.); (L.H.)
| | - Yi Li
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School of Stomatology, Lanzhou University, Lanzhou 730030, China; (Y.M.); (K.H.); (M.S.); (Y.H.); (L.H.)
| |
Collapse
|
7
|
Chu WH, Yang N, Zhang JH, Li Y, Song JL, Deng ZP, Meng N, Zhang J, Zhu KK, Jiang CS. Discovery of tetrahydroisoquinolineindole derivatives as first dual PRMT5 inhibitors/hnRNP E1 upregulators: Design, synthesis and biological evaluation. Eur J Med Chem 2023; 258:115625. [PMID: 37429083 DOI: 10.1016/j.ejmech.2023.115625] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 06/30/2023] [Accepted: 07/04/2023] [Indexed: 07/12/2023]
Abstract
Protein arginine methyltransferase 5 (PRMT5) is an epigenetics related enzyme that has been validated as an important therapeutic target for treating various types of cancer. Upregulation of tumor suppressor hnRNP E1 has also been considered as an effective antitumor therapy. In this study, a series of tetrahydroisoquinolineindole hybrids were designed and prepared, and compounds 3m and 3s4 were found to be selective inhibitors of PRMT5 and upregulators of hnRNP E1. Molecular docking studies indicated that compounds 3m occupied the substrate site of PRMT5 and formed essential interactions with amino acid residues. Furthermore, compounds 3m and 3s4 exerted antiproliferative effects against A549 cells by inducing apoptosis and inhibiting cell migration. Importantly, silencing of hnRNP E1 eliminated the antitumor effect of 3m and 3s4 on the apoptosis and migration in A549 cells, suggesting a regulatory relationship between PRMT5 and hnRNP E1. Additionally, compound 3m exhibited high metabolic stability on human liver microsomes (T1/2 = 132.4 min). In SD rats, the bioavailability of 3m was 31.4%, and its PK profiles showed satisfactory AUC and Cmax values compared to the positive control. These results suggest that compound 3m is the first class of dual PRMT5 inhibitor and hnRNP E1 upregulator that deserves further investigation as a potential anticancer agent.
Collapse
Affiliation(s)
- Wen-Hui Chu
- School of Biological Science and Technology, University of Jinan, Jinan, 250022, China
| | - Na Yang
- School of Biological Science and Technology, University of Jinan, Jinan, 250022, China; Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Jin-He Zhang
- School of Biological Science and Technology, University of Jinan, Jinan, 250022, China
| | - Yue Li
- School of Biological Science and Technology, University of Jinan, Jinan, 250022, China
| | - Jia-Li Song
- School of Biological Science and Technology, University of Jinan, Jinan, 250022, China
| | - Zhi-Peng Deng
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250355, China
| | - Ning Meng
- School of Biological Science and Technology, University of Jinan, Jinan, 250022, China.
| | - Juan Zhang
- School of Biological Science and Technology, University of Jinan, Jinan, 250022, China.
| | - Kong-Kai Zhu
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| | - Cheng-Shi Jiang
- School of Biological Science and Technology, University of Jinan, Jinan, 250022, China.
| |
Collapse
|
8
|
La T, Chen S, Zhao XH, Zhou S, Xu R, Teng L, Zhang YY, Ye K, Xu L, Guo T, Jamaluddin MF, Feng YC, Tang HJ, Wang Y, Xu Q, Gu Y, Cao H, Liu T, Thorne RF, Shao F, Zhang XD, Jin L. LncRNA LIMp27 Regulates the DNA Damage Response through p27 in p53-Defective Cancer Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204599. [PMID: 36638271 PMCID: PMC9982580 DOI: 10.1002/advs.202204599] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 12/06/2022] [Indexed: 06/17/2023]
Abstract
P53 inactivation occurs in about 50% of human cancers, where p53-driven p21 activity is devoid and p27 becomes essential for the establishment of the G1/S checkpoint upon DNA damage. Here, this work shows that the E2F1-responsive lncRNA LIMp27 selectively represses p27 expression and contributes to proliferation, tumorigenicity, and treatment resistance in p53-defective colon adenocarcinoma (COAD) cells. LIMp27 competes with p27 mRNA for binding to cytoplasmically localized hnRNA0, which otherwise stabilizes p27 mRNA leading to cell cycle arrest at the G0/G1 phase. In response to DNA damage, LIMp27 is upregulated in both wild-type and p53-mutant COAD cells, whereas cytoplasmic hnRNPA0 is only increased in p53-mutant COAD cells due to translocation from the nucleus. Moreover, high LIMp27 expression is associated with poor survival of p53-mutant but not wild-type p53 COAD patients. These results uncover an lncRNA mechanism that promotes p53-defective cancer pathogenesis and suggest that LIMp27 may constitute a target for the treatment of such cancers.
Collapse
Affiliation(s)
- Ting La
- Translational Research InstituteHenan Provincial and Zhengzhou City Key laboratory of Non‐coding RNA and Cancer MetabolismHenan International Join Laboratory of Non‐coding RNA and Metabolism in CancerHenan Provincial People's HospitalAcademy of Medical SciencesZhengzhou UniversityZhengzhouHenan450053China
- Noncoding Cancer Biomarkers and Therapeutics GroupCancer Detection & Therapy Research ProgramHunter Medical Research InstituteCallaghanNew South Wales2305Australia
- School of Biomedical Sciences and PharmacyThe University of NewcastleCallaghanNew South Wales2308Australia
- National‐Local Joint Engineering Research Center of Biodiagnosis & BiotherapyThe Second Affiliated HospitalXi'an Jiaotong UniversityXi'anShaanxi710004China
| | - Song Chen
- Translational Research InstituteHenan Provincial and Zhengzhou City Key laboratory of Non‐coding RNA and Cancer MetabolismHenan International Join Laboratory of Non‐coding RNA and Metabolism in CancerHenan Provincial People's HospitalAcademy of Medical SciencesZhengzhou UniversityZhengzhouHenan450053China
- Institute of Medicinal BiotechnologyJiangsu College of NursingHuai'anJiangsu223300China
| | - Xiao Hong Zhao
- Noncoding Cancer Biomarkers and Therapeutics GroupCancer Detection & Therapy Research ProgramHunter Medical Research InstituteCallaghanNew South Wales2305Australia
- School of Biomedical Sciences and PharmacyThe University of NewcastleCallaghanNew South Wales2308Australia
| | - Shuai Zhou
- Translational Research InstituteHenan Provincial and Zhengzhou City Key laboratory of Non‐coding RNA and Cancer MetabolismHenan International Join Laboratory of Non‐coding RNA and Metabolism in CancerHenan Provincial People's HospitalAcademy of Medical SciencesZhengzhou UniversityZhengzhouHenan450053China
| | - Ran Xu
- Noncoding Cancer Biomarkers and Therapeutics GroupCancer Detection & Therapy Research ProgramHunter Medical Research InstituteCallaghanNew South Wales2305Australia
- School of Biomedical Sciences and PharmacyThe University of NewcastleCallaghanNew South Wales2308Australia
| | - Liu Teng
- Translational Research InstituteHenan Provincial and Zhengzhou City Key laboratory of Non‐coding RNA and Cancer MetabolismHenan International Join Laboratory of Non‐coding RNA and Metabolism in CancerHenan Provincial People's HospitalAcademy of Medical SciencesZhengzhou UniversityZhengzhouHenan450053China
| | - Yuan Yuan Zhang
- Noncoding Cancer Biomarkers and Therapeutics GroupCancer Detection & Therapy Research ProgramHunter Medical Research InstituteCallaghanNew South Wales2305Australia
- School of Biomedical Sciences and PharmacyThe University of NewcastleCallaghanNew South Wales2308Australia
| | - Kaihong Ye
- Translational Research InstituteHenan Provincial and Zhengzhou City Key laboratory of Non‐coding RNA and Cancer MetabolismHenan International Join Laboratory of Non‐coding RNA and Metabolism in CancerHenan Provincial People's HospitalAcademy of Medical SciencesZhengzhou UniversityZhengzhouHenan450053China
| | - Liang Xu
- Noncoding Cancer Biomarkers and Therapeutics GroupCancer Detection & Therapy Research ProgramHunter Medical Research InstituteCallaghanNew South Wales2305Australia
- School of Biomedical Sciences and PharmacyThe University of NewcastleCallaghanNew South Wales2308Australia
| | - Tao Guo
- Institute of Future AgricultureNorthwest A&F UniversityYanglingShaanxi712100China
| | - Muhammad Fairuz Jamaluddin
- School of Biomedical Sciences and PharmacyThe University of NewcastleCallaghanNew South Wales2308Australia
| | - Yu Chen Feng
- Noncoding Cancer Biomarkers and Therapeutics GroupCancer Detection & Therapy Research ProgramHunter Medical Research InstituteCallaghanNew South Wales2305Australia
- School of Medicine and Public HealthThe University of NewcastleCallaghanNew South Wales2308Australia
| | - Hai Jie Tang
- Noncoding Cancer Biomarkers and Therapeutics GroupCancer Detection & Therapy Research ProgramHunter Medical Research InstituteCallaghanNew South Wales2305Australia
- School of Biomedical Sciences and PharmacyThe University of NewcastleCallaghanNew South Wales2308Australia
| | - Yanliang Wang
- Department of NephrologyHenan Provincial Key Laboratory of Kidney Disease and ImmunologyHenan Provincial Clinical Research Center for Kidney DiseaseHenan Provincial People's HospitalZhengzhouHenan450053China
| | - Qin Xu
- Department of NephrologyHenan Provincial Key Laboratory of Kidney Disease and ImmunologyHenan Provincial Clinical Research Center for Kidney DiseaseHenan Provincial People's HospitalZhengzhouHenan450053China
| | - Yue Gu
- Department of NephrologyHenan Provincial Key Laboratory of Kidney Disease and ImmunologyHenan Provincial Clinical Research Center for Kidney DiseaseHenan Provincial People's HospitalZhengzhouHenan450053China
| | - Huixia Cao
- Department of NephrologyHenan Provincial Key Laboratory of Kidney Disease and ImmunologyHenan Provincial Clinical Research Center for Kidney DiseaseHenan Provincial People's HospitalZhengzhouHenan450053China
| | - Tao Liu
- Children's Cancer Institute Australia for Medical ResearchUniversity of New South WalesSydneyNew South Wales2750Australia
| | - Rick F. Thorne
- Translational Research InstituteHenan Provincial and Zhengzhou City Key laboratory of Non‐coding RNA and Cancer MetabolismHenan International Join Laboratory of Non‐coding RNA and Metabolism in CancerHenan Provincial People's HospitalAcademy of Medical SciencesZhengzhou UniversityZhengzhouHenan450053China
- School of Biomedical Sciences and PharmacyThe University of NewcastleCallaghanNew South Wales2308Australia
| | - Feng‐Min Shao
- Department of NephrologyHenan Provincial Key Laboratory of Kidney Disease and ImmunologyHenan Provincial Clinical Research Center for Kidney DiseaseHenan Provincial People's HospitalZhengzhouHenan450053China
| | - Xu Dong Zhang
- Translational Research InstituteHenan Provincial and Zhengzhou City Key laboratory of Non‐coding RNA and Cancer MetabolismHenan International Join Laboratory of Non‐coding RNA and Metabolism in CancerHenan Provincial People's HospitalAcademy of Medical SciencesZhengzhou UniversityZhengzhouHenan450053China
- Noncoding Cancer Biomarkers and Therapeutics GroupCancer Detection & Therapy Research ProgramHunter Medical Research InstituteCallaghanNew South Wales2305Australia
- School of Biomedical Sciences and PharmacyThe University of NewcastleCallaghanNew South Wales2308Australia
| | - Lei Jin
- Translational Research InstituteHenan Provincial and Zhengzhou City Key laboratory of Non‐coding RNA and Cancer MetabolismHenan International Join Laboratory of Non‐coding RNA and Metabolism in CancerHenan Provincial People's HospitalAcademy of Medical SciencesZhengzhou UniversityZhengzhouHenan450053China
- Noncoding Cancer Biomarkers and Therapeutics GroupCancer Detection & Therapy Research ProgramHunter Medical Research InstituteCallaghanNew South Wales2305Australia
- School of Medicine and Public HealthThe University of NewcastleCallaghanNew South Wales2308Australia
| |
Collapse
|
9
|
Zhao H, Liu C, Zhao C, Che C, Liu W, Mei Y. Alternatively-spliced lncRNA-PNUTS promotes HCC cell EMT via regulating ZEB1 expression. TUMORI JOURNAL 2023; 109:28-37. [PMID: 35139713 DOI: 10.1177/03008916211072585] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Long non-coding RNAs have been implicated in various cancers as they regulate critical cellular processes such as proliferation, migration, invasion, and apoptosis in tumorous tissues. lncRNA-PNUTS is newly reported as an alternatively-spliced lncRNA from PNUTS pre-mRNA that promotes oncogenesis in breast cancer. However, whether LncRNA-PNUTS plays a role in other forms of cancers, such as liver cancer, remains unknown. METHOD In the current study, we investigated the potential role of lncRNA-PNUTS in hepatocellular carcinoma (HCC). The levels of lncRNA-PNUTS in tumorous tissues obtained from HCC patients were measured. The potential impacts of lncPNUTS on metastasis and invasion were investigated through gain- or loss- of function experiments in cell models of liver cancers, as well as other cellular assays such as trans-well assays and wound-healing assays. RESULTS Here, we report that lncPNUTS was upregulated in human HCC tissues. Loss- and gain-of-function experiments indicated lncPNUTS promoted metastasis and invasion. In addition, ZEB1, which is involved in the activation of epithelial-mesenchymal-transition (EMT), was identified as a downstream target of lncPNUTS. CONCLUSION Our findings indicated lncPNUTS promotes HCC cancer cell metastasis and invasion via targeting ZEB1 to activate the EMT pathway, suggesting that lncPNUTS is a potential prognostic marker and therapeutic target for HCC patients.
Collapse
Affiliation(s)
- Haiyan Zhao
- Medical Faculty of Kunming University of Science and Technology, Kunming, Yunnan, China.,Department of Ophthalmology, The Affiliated Hospital of Kunming University of Science and Technology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Chang Liu
- The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Chongyu Zhao
- The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Chi Che
- The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Wuguang Liu
- The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yan Mei
- Medical Faculty of Kunming University of Science and Technology, Kunming, Yunnan, China.,Department of Ophthalmology, The Affiliated Hospital of Kunming University of Science and Technology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| |
Collapse
|
10
|
Singh DD, Lee HJ, Yadav DK. Clinical updates on tyrosine kinase inhibitors in HER2-positive breast cancer. Front Pharmacol 2022; 13:1089066. [PMID: 36578543 PMCID: PMC9792097 DOI: 10.3389/fphar.2022.1089066] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 11/30/2022] [Indexed: 12/14/2022] Open
Abstract
Breast cancer (BC) is caused by epigenetic modifications and genetic heterogeneity and exhibits various histological feature. HER2+ (Human epidermal growth factor receptor 2) is a more aggressive type of breast cancer, diagnosis and prognosis are difficult for HER2+ BC. Anti-HER2+ inhibitors have been effectively used for patient treatment. High mortality rate is reported in HER2+ BC, due to availability of limited therapeutic options. Despite advances in systemic medications to treat metastatic breast cancer (MBC), HER2-positive MBC is still challenging for patients and treating clinicians. The clinical characteristics of the disease have changed after treatment with HER2-targeted therapy. Various types of Tyrosine kinase inhibitors (TKIs) have been developed to treat patients with HER2+ BC including afatinib, lapatinib, neratinib, tucatinib, and pyrotinib, have been developed as HER2-targeted therapies. The antibody-drug conjugates adotrastuzumab, emtansine, famtrastuzumab, and deruxtecan, as well as the anti-HER2 monoclonal antibody pertuzumab are used in both early-stage and metastatic situations, either alone or in conjunction with chemotherapy and other HER2-targeting therapies. The emergence of drug resistance in anti-HER2 therapies has been observed. To overcome drug resistance and limited efficacy in current treatment options, nano formulations can be used in patients with HER2+ BC treatment. Anti-HER2 ligands can be used in various nano formulations to target HER2 receptors. Here we will discuss, targeted TKIs in patients with HER2+ BC, clinical studies of HER2+ targeted TKIs, mechanisms of resistance to HER2-directed therapies with new implications of TKIs in HER2+ MBC (metastatic breast cancer) and anti-HER2 ligand in various nano formulations to target HER2 receptors.
Collapse
Affiliation(s)
- Desh Deepak Singh
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, India
| | - Hae-Jeung Lee
- Department of Food and Nutrition, College of Bionano Technology, Gachon University, Seongnam-si, Gyeonggi-do, South Korea,*Correspondence: Hae-Jeung Lee, ; Dharmendra Kumar Yadav,
| | - Dharmendra Kumar Yadav
- Department of Pharmacy, Gachon Institute of Pharmaceutical Science, College of Pharmacy, Gachon University, Incheon, South Korea,*Correspondence: Hae-Jeung Lee, ; Dharmendra Kumar Yadav,
| |
Collapse
|
11
|
Wu X, Yang H, Yu X, Qin JJ. Drug-resistant HER2-positive breast cancer: Molecular mechanisms and overcoming strategies. Front Pharmacol 2022; 13:1012552. [PMID: 36210846 PMCID: PMC9540370 DOI: 10.3389/fphar.2022.1012552] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 09/05/2022] [Indexed: 11/24/2022] Open
Abstract
Breast cancer is one of the most common malignancies and the leading cause of cancer-related death in women. HER2 overexpression is a factor for poor prognosis in breast cancer, and anti-HER2 therapy improves survival in these patients. A dual-targeted combination of pertuzumab and trastuzumab, alongside cytotoxic chemotherapy, constitutes the primary treatment option for individuals with early-stage, HER2-positive breast cancer. Antibody-drug conjugate (ADC) and tyrosine kinase inhibitors (TKI) also increase the prognosis for patients with metastatic breast cancer. However, resistance to targeted therapy eventually occurs. Therefore, it is critical to investigate how HER2-positive breast cancer is resistant to targeted therapy and to develop novel drugs or strategies to overcome the resistance simultaneously. This review aims to provide a comprehensive discussion of the HER2-targeted agents currently in clinical practice, the molecular mechanisms of resistance to these drugs, and the potential strategies for overcoming resistance.
Collapse
Affiliation(s)
| | | | - Xingfei Yu
- *Correspondence: Xingfei Yu, ; Jiang-Jiang Qin,
| | | |
Collapse
|
12
|
Lu X, Zhong J, Liu L, Zhang W, Zhao S, Chen L, Wei Y, Zhang H, Wu J, Chen W, Ge F. The function and regulatory mechanism of RNA-binding proteins in breast cancer and their future clinical treatment prospects. Front Oncol 2022; 12:929037. [PMID: 36052258 PMCID: PMC9424610 DOI: 10.3389/fonc.2022.929037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 07/25/2022] [Indexed: 11/19/2022] Open
Abstract
Breast cancer is the most common female malignancy, but the mechanisms regulating gene expression leading to its development are complex. In recent years, as epigenetic research has intensified, RNA-binding proteins (RBPs) have been identified as a class of posttranscriptional regulators that can participate in regulating gene expression through the regulation of RNA stabilization and degradation, intracellular localization, alternative splicing and alternative polyadenylation, and translational control. RBPs play an important role in the development of normal mammary glands and breast cancer. Functional inactivation or abnormal expression of RBPs may be closely associated with breast cancer development. In this review, we focus on the function and regulatory mechanisms of RBPs in breast cancer, as well as the advantages and challenges of RBPs as potential diagnostic and therapeutic targets in breast cancer, and discuss the potential of RBPs in clinical treatment.
Collapse
Affiliation(s)
- Xingjia Lu
- Department of Breast Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, China
- Kunming Medical University, No. 1 School of Clinical Medicine, Kunming, China
| | - Jian Zhong
- Department of Reproductive Medicine, Affiliated Jinling Hospital, Nanjing Medical University, Nanjing, China
- Department of Gynecology, Women’s Hospital of Nanjing Medical University, Nanjing, China
| | - Linlin Liu
- School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Wenzhu Zhang
- Department of Breast Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, China
- Kunming Medical University, No. 1 School of Clinical Medicine, Kunming, China
| | - Shengdi Zhao
- Department of Breast Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, China
- Kunming Medical University, No. 1 School of Clinical Medicine, Kunming, China
| | - Liang Chen
- Department of Breast Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yuxian Wei
- Department of Endocrine Breast Surgery, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hong Zhang
- Department of Breast Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, China
- Kunming Medical University, No. 1 School of Clinical Medicine, Kunming, China
| | - Jingxuan Wu
- Department of Breast Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, China
- Kunming Medical University, No. 1 School of Clinical Medicine, Kunming, China
| | - Wenlin Chen
- Third Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
- *Correspondence: Wenlin Chen, ; Fei Ge,
| | - Fei Ge
- Department of Breast Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, China
- *Correspondence: Wenlin Chen, ; Fei Ge,
| |
Collapse
|
13
|
Zheng Y, Zhou Z, Wei R, Xiao C, Zhang H, Fan T, Zheng B, Li C, He J. The RNA-binding protein PCBP1 represses lung adenocarcinoma progression by stabilizing DKK1 mRNA and subsequently downregulating β-catenin. J Transl Med 2022; 20:343. [PMID: 35907982 PMCID: PMC9338556 DOI: 10.1186/s12967-022-03552-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 07/24/2022] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND PolyC-RNA-binding protein 1 (PCBP1) functions as a tumour suppressor and RNA regulator that is downregulated in human cancers. Here, we aimed to reveal the biological function of PCBP1 in lung adenocarcinoma (LUAD). METHODS First, PCBP1 was identified as an important biomarker that maintains LUAD through The Cancer Genome Atlas (TCGA) project screening and confirmed by immunohistochemistry and qPCR. Via colony formation, CCK8, IncuCyte cell proliferation, wound healing and Transwell assays, we confirmed that PCBP1 was closely related to the proliferation and migration of LUAD cells. The downstream gene DKK1 was discovered by RNA sequencing of PCBP1 knockdown cells. The underlying mechanisms were further investigated using western blot, qPCR, RIP, RNA pulldown and mRNA stability assays. RESULTS We demonstrate that PCBP1 is downregulated in LUAD tumour tissues. The reduction in PCBP1 promotes the proliferation, migration and invasion of LUAD in vitro and in vivo. Mechanistically, the RNA-binding protein PCBP1 represses LUAD by stabilizing DKK1 mRNA. Subsequently, decreased expression of the DKK1 protein relieves the inhibitory effect on the Wnt/β-catenin signalling pathway. Taken together, these results show that PCBP1 acts as a tumour suppressor gene, inhibiting the tumorigenesis of LUAD. CONCLUSIONS We found that PCBP1 inhibits LUAD development by upregulating DKK1 to inactivate the Wnt/β-catenin pathway. Our findings highlight the potential of PCBP1 as a promising therapeutic target.
Collapse
Affiliation(s)
- Yujia Zheng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zheng Zhou
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ran Wei
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chu Xiao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hao Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tao Fan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bo Zheng
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
14
|
Song L, Mao R, Ding L, Tian Z, Zhang M, Wang J, Wang M, Lyu Y, Liu C, Feng M, Jia H, Wang J. hnRNP E1 Regulates HPV16 Oncogene Expression and Inhibits Cervical Cancerization. Front Oncol 2022; 12:905900. [PMID: 35800060 PMCID: PMC9253288 DOI: 10.3389/fonc.2022.905900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/24/2022] [Indexed: 11/15/2022] Open
Abstract
hnRNP E1 (heterogeneous nuclear ribonucleoprotein E1) is an important RNA-binding protein (RBPs) that plays a vital role in tumor development. Human papillomavirus 16 (HPV16) contains numerous sites that can bind to RNA/DNA and may be modified by multiple RBPs, which contribute to HPV gene expression and HPV-associated cancer development. However, the effects of hnRNP E1 on HPV16 oncogenes in the development of cervical lesions remain unclear. A total of 816 participants with different grades of cervical lesions were enrolled in a community-based cohort established in Shanxi Province, China. The Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) databases were used to analyze the association between hnRNP E1 mRNA expression and cervical lesions. Cells with up_ and down_regulated hnRNP E1 were established. hnRNP E1 functions were evaluated using cell counting kit-8, flow cytometry analyses, and chromatin immunoprecipitation sequencing. Our results showed that hnRNP E1 expression was linearly dependent on the severity of the cervical lesions. Low expression of HPV16 E2, high expression of E6, and a low ratio of E2 to E6 could increase the risk of cervical lesions. hnRNP E1 expression was correlated with HPV16 oncogene expression. hnRNP E1-relevant genes were involved in the dopaminergic synapses, Wnt signaling pathway, gnRH secretion, and mTOR signaling pathway. hnRNP E1 significantly inhibited cell proliferation, induced apoptosis, arrested the cell cycle at the G0/G1 stage, and decreased HPV16 E6 expression. Our results indicate that hnRNP E1 could downregulate HPV16 E6 oncogene expression and inhibit cervical cancerization, which sheds new light on preventing the carcinogenicity of HPV across a range of diseases by regulating RNA-binding proteins.
Collapse
Affiliation(s)
- Li Song
- Department of Epidemiology, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Rui Mao
- Questrom School of Business, Boston University, Boston, MA, United States
| | - Ling Ding
- Department of Epidemiology, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Zhiqiang Tian
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mingxuan Zhang
- Department of Epidemiology, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Jiahao Wang
- Department of Epidemiology, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Ming Wang
- Department of Epidemiology, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Yuanjing Lyu
- Department of Epidemiology, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Chunliang Liu
- Department of Epidemiology, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Meijuan Feng
- Department of Epidemiology, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Haixia Jia
- Department of Epidemiology, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Jintao Wang
- Department of Epidemiology, School of Public Health, Shanxi Medical University, Taiyuan, China
- *Correspondence: Jintao Wang,
| |
Collapse
|
15
|
Kaida D, Shida K. Spliceostatin A stabilizes CDKN1B mRNA through the 3′ UTR. Biochem Biophys Res Commun 2022; 608:39-44. [DOI: 10.1016/j.bbrc.2022.03.085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 03/16/2022] [Indexed: 11/02/2022]
|
16
|
Peng K, Chen X, Lin A, Tong Z, Lin W. PolyC-RNA-binding protein 1 (PCBP1) enhances tropomyosin 3 (TPM3) mRNA stability to promote the progression of esophageal squamous cell carcinoma. Bioengineered 2022; 13:8581-8592. [PMID: 35287546 PMCID: PMC9161940 DOI: 10.1080/21655979.2022.2053801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
The molecular etiology of esophageal squamous cell carcinoma (ESCC) has not been fully elucidated. Understanding the molecular mechanisms and finding new therapeutic targets for ESCC are of crucial importance. PolyC-RNA-binding protein 1 (PCBP1) is an RNA-binding protein. Here, we found overexpressed PCBP1 in esophageal cancer tissues by quantitative polymerase chain reaction (qPCR) and western blotting analysis. PCBP1 knockdown significantly attenuated migratory and invasion abilities of ESCC cells. Mechanistically, PCBP1 bound directly to tropomyosin 3 (TPM3) mRNA, which was verified by RNA–protein immunoprecipitation (RIP) assay. PCBP1 knockdown markedly reduced messenger RNA (mRNA) levels of TPM3. After inhibiting intracellular mRNA synthesis with actinomycin D (ActD), it was found that PCBP1 knockdown contributed to a significant decrease in TPM3 mRNA degradation. Furthermore, PCBP1 promoted migration and invasion of EC cells by directly binding to the 3’UTR of TPM3 mRNA, increasing TPM3 mRNA stability. Taken together, PCBP1 acting as a pro-oncogenic factor enhances TPM3 mRNA stability by directly binding to the 3’UTR of TPM3 mRNA in esophageal squamous cell carcinoma. Our findings provide a new perspective for understanding the molecular mechanism of esophageal carcinogenesis, and PCBP1 is a promising therapeutic target.
Collapse
Affiliation(s)
- Kaiming Peng
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou350001, Fujian Province, China
| | - Xiaoqiang Chen
- Department of Otolaryngology, Fujian Medical University Union Hospital, Fuzhou350001, Fujian Province, China
| | - Anqin Lin
- Department of surgery, Fujian Medical University Union Hospital, Fuzhou350001, Fujian Province, China
| | - Zhangwei Tong
- Fujian Medical University Union Hospital, Fujian Medical University, Fuzhou350001, Fujian Province, China
| | - Wenwei Lin
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou350001, Fujian Province, China
| |
Collapse
|
17
|
Lin L, Li H, Shi D, Liu Z, Wei Y, Wang W, Wu D, Li B, Guo Q. Depletion of C12orf48 inhibits gastric cancer growth and metastasis via up-regulating Poly r(C)-Binding Protein (PCBP) 1. BMC Cancer 2022; 22:123. [PMID: 35100974 PMCID: PMC8802463 DOI: 10.1186/s12885-022-09220-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 01/19/2022] [Indexed: 01/20/2023] Open
Abstract
Background Gastric cancer remains a major cause of cancer-related death worldwide. C12orf48, also named PARP1 binding protein, is over-expressed in several cancers. However, the expression profile and potential roles of C12orf48 in gastric cancer are largely unknown. Methods We used bioinformatics approaches and tissue microarray immunohistochemistry to analyze the expression profile of C12orf48 in gastric cancer tissues. Plasmid-mediated over-expression or knockdown were performed. CCK-8 assays and flow cytometry were employed to evaluate cellular proliferation and apoptosis respectively. Transwell assays were used to assess migrative and invasive abilities. The roles of C12orf48 were also evaluated in a xenograft tumor model. Results We found that C12orf48 was over-expressed in gastric cancer tissue, which associated with advanced stage and poor prognosis. In vitro and in vivo experiments showed depletion of C12orf48 attenuated cancer growth, while facilitated apoptosis. Further, the expression of Poly r(C)-Binding Protein (PCBP) 1 was found negatively regulated by C12orf48. Intended up-regulation of PCBP1 prevented C12orf48-mediated proliferation and rescued cells from apoptosis. Besides, C12orf48 promoted cellular migration and invasion, with E-cadherin down-regulated while vimentin and N-cadherin up-regulated, which was reversed by up-regulated PCBP1. Conclusions Our findings indicate that depletion of C12orf48 inhibited gastric cancer growth and metastasis via up-regulating PCBP1. Targeting C12orf48-PCBP1 axis may be a potential therapeutic strategy. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09220-0.
Collapse
Affiliation(s)
- Lele Lin
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Zhejiang University School of Medicine, 88# Jiefang RoadZhejiang Province, Hangzhou City, 310000, P. R. China
| | - Hongbo Li
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Zhejiang University School of Medicine, 88# Jiefang RoadZhejiang Province, Hangzhou City, 310000, P. R. China
| | - Dike Shi
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Zhejiang University School of Medicine, 88# Jiefang RoadZhejiang Province, Hangzhou City, 310000, P. R. China
| | - Zhiqiang Liu
- Department of General Surgery, Anyang Tumor Hospital, 1# North Huanbin Road, Henan Province, 455000, Anyang City, PR China
| | - Yunhai Wei
- Department of Gastrointestinal Surgery, Huzhou Central Hospital, 198# Hongqi RoadZhejiang Province, Huzhou City, 31300, P. R. China
| | - Wei Wang
- Department of Gastrointestinal Surgery, Shaoxing People's Hospital, 568# North Zhongxing RoadZhejiang Province, Shaoxing City, 312000, P. R. China
| | - Dan Wu
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Zhejiang University School of Medicine, 88# Jiefang RoadZhejiang Province, Hangzhou City, 310000, P. R. China
| | - Baozhong Li
- Department of General Surgery, Anyang Tumor Hospital, 1# North Huanbin Road, Henan Province, 455000, Anyang City, PR China.
| | - Qingqu Guo
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Zhejiang University School of Medicine, 88# Jiefang RoadZhejiang Province, Hangzhou City, 310000, P. R. China.
| |
Collapse
|
18
|
Lv C, Sun J, Ye Y, Lin Z, Li H, Liu Y, Mo K, Xu W, Hu W, Draz E, Wang S. LncRNA EIF1AX-AS1 promotes endometrial cancer cell apoptosis by affecting EIF1AX mRNA stabilization. Cancer Sci 2022; 113:1277-1291. [PMID: 35080085 PMCID: PMC8990785 DOI: 10.1111/cas.15275] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 01/11/2022] [Accepted: 01/20/2022] [Indexed: 12/24/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) have been found to play an important role in the occurrence and development of endometrial carcinoma (EC). Here, using RNA sequencing analysis, we systemically screened and identified the lncRNA EIF1AX-AS1, which is aberrantly down-regulated in clinical EC tissues and closely correlated with tumor type. EIF1AX-AS1 markedly inhibited EC cell proliferation and promoted apoptosis in vitro and in vivo. Mechanistically, EIF1AX-AS1 interacts with EIF1AX mRNA and poly C binding protein 1 (PCBP1), which promote eukaryotic translation initiation factor 1A, X-linked (EIF1AX) mRNA degradation. Intriguingly, interaction with IRES-related proteins Y-box binding protein 1 (YBX-1), EIF1AX promotes c-Myc translation through the internal ribosome enter site pathway. c-Myc promotes EIF1AX transcription and thus forms a feed-forward loop to regulate EC cell proliferation. Taken together, these data reveal new insights into the biology driving EC proliferation and highlights the potential of lncRNAs as biomarkers for prognosis and future therapeutic targets for cancer.
Collapse
Affiliation(s)
- Chengyu Lv
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fujian Medical University, Fuzhou, 350122, P. R. China.,Department of Obstetrics and Gynecology, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, 350001, P. R. China
| | - Jiandong Sun
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fujian Medical University, Fuzhou, 350122, P. R. China
| | - Yuhong Ye
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, P. R. China.,Department of Pathology, The First Affiliated Hospital of Fujian Medical University, 350005, Fuzhou, P. R.China
| | - Zihang Lin
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fujian Medical University, Fuzhou, 350122, P. R. China
| | - Hua Li
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fujian Medical University, Fuzhou, 350122, P. R. China.,Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, P. R. China
| | - Yue Liu
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fujian Medical University, Fuzhou, 350122, P. R. China.,Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, P. R. China
| | - Kaien Mo
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fujian Medical University, Fuzhou, 350122, P. R. China
| | - Weiwei Xu
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fujian Medical University, Fuzhou, 350122, P. R. China
| | - Weitao Hu
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fujian Medical University, Fuzhou, 350122, P. R. China
| | - Eman Draz
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, P. R. China.,Human Anatomy and Embryology department, Suez Canal University, 12411, Egypt
| | - Shie Wang
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fujian Medical University, Fuzhou, 350122, P. R. China.,Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, P. R. China
| |
Collapse
|
19
|
Wu J, Wu Y, Guo Q, Wang S, Wu X. RNA-binding proteins in ovarian cancer: a novel avenue of their roles in diagnosis and treatment. J Transl Med 2022; 20:37. [PMID: 35062979 PMCID: PMC8783520 DOI: 10.1186/s12967-022-03245-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 01/11/2022] [Indexed: 12/18/2022] Open
Abstract
Ovarian cancer (OC), an important cause of cancer-related death in women worldwide, is one of the most malignant cancers and is characterized by a poor prognosis. RNA-binding proteins (RBPs), a class of endogenous proteins that can bind to mRNAs and modify (or even determine) the amount of protein they can generate, have attracted great attention in the context of various diseases, especially cancers. Compelling studies have suggested that RBPs are aberrantly expressed in different cancer tissues and cell types, including OC tissues and cells. More specifically, RBPs can regulate proliferation, apoptosis, invasion, metastasis, tumorigenesis and chemosensitivity and serve as potential therapeutic targets in OC. Herein, we summarize what is currently known about the biogenesis, molecular functions and potential roles of human RBPs in OC and their prospects for application in the clinical treatment of OC.
Collapse
Affiliation(s)
- Jiangchun Wu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, People's Republic of China
| | - Yong Wu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, People's Republic of China
| | - Qinhao Guo
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, People's Republic of China
| | - Simin Wang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, People's Republic of China
| | - Xiaohua Wu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
20
|
Zhao H, Wei Z, Shen G, Chen Y, Hao X, Li S, Wang R. Poly(rC)-binding proteins as pleiotropic regulators in hematopoiesis and hematological malignancy. Front Oncol 2022; 12:1045797. [PMID: 36452487 PMCID: PMC9701828 DOI: 10.3389/fonc.2022.1045797] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 10/26/2022] [Indexed: 11/16/2022] Open
Abstract
Poly(rC)-binding proteins (PCBPs), a defined subfamily of RNA binding proteins, are characterized by their high affinity and sequence-specific interaction with poly-cytosine (poly-C). The PCBP family comprises five members, including hnRNP K and PCBP1-4. These proteins share a relatively similar structure motif, with triple hnRNP K homology (KH) domains responsible for recognizing and combining C-rich regions of mRNA and single- and double-stranded DNA. Numerous studies have indicated that PCBPs play a prominent role in hematopoietic cell growth, differentiation, and tumorigenesis at multiple levels of regulation. Herein, we summarized the currently available literature regarding the structural and functional divergence of various PCBP family members. Furthermore, we focused on their roles in normal hematopoiesis, particularly in erythropoiesis. More importantly, we also discussed and highlighted their involvement in carcinogenesis, including leukemia and lymphoma, aiming to clarify the pleiotropic roles and molecular mechanisms in the hematopoietic compartment.
Collapse
Affiliation(s)
- Huijuan Zhao
- Henan International Joint Laboratory of Thrombosis and Hemostasis, Henan University of Science and Technology, Luoyang, China.,Basic Medical College, Henan University of Science and Technology, Luoyang, China
| | - Ziqing Wei
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Guomin Shen
- Henan International Joint Laboratory of Thrombosis and Hemostasis, Henan University of Science and Technology, Luoyang, China.,Basic Medical College, Henan University of Science and Technology, Luoyang, China
| | - Yixiang Chen
- Henan International Joint Laboratory of Thrombosis and Hemostasis, Henan University of Science and Technology, Luoyang, China.,Basic Medical College, Henan University of Science and Technology, Luoyang, China
| | - Xueqin Hao
- Basic Medical College, Henan University of Science and Technology, Luoyang, China
| | - Sanqiang Li
- Basic Medical College, Henan University of Science and Technology, Luoyang, China
| | - Rong Wang
- Department of Clinical Laboratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
21
|
PCBP1 regulates the transcription and alternative splicing of metastasis‑related genes and pathways in hepatocellular carcinoma. Sci Rep 2021; 11:23356. [PMID: 34857818 PMCID: PMC8640068 DOI: 10.1038/s41598-021-02642-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 11/03/2021] [Indexed: 02/08/2023] Open
Abstract
PCBP1 is a multifunctional RNA-binding protein (RBP) expressed in most human cells and is involved in posttranscriptional gene regulation. PCBP1 regulates the alternative splicing, translation and RNA stability of many cancer-related genes and has been identified as a potential tumour suppressor gene. PCBP1 inhibits the invasion of hepatocellular carcinoma (HCC) cells, but there are few studies on the specific regulatory target and mechanism of RBPs in HCC, and it is unclear whether PCBP1 plays a role in tumour metastasis as a splicing factor. We analysed the regulation of gene expression by PCBP1 at the transcriptional level. We obtained and analysed PCBP1-knockdown RNA-seq data and eCLIP-seq data of PCBP1 in HepG2 cells and found that PCBP1 widely regulates the alternative splicing and expression of genes enriched in cancer-related pathways, including extracellular matrix, cell adhesion, small molecule metabolic process and apoptosis. We validated five regulated alternative splicing events affected by PCBP1 using RT-qPCR and found that there was a significant difference in the expression of APOC1 and SPHK1 between tumour and normal tissues. In this study, we provided convincing evidence that human PCBP1 profoundly regulates the splicing of genes associated with tumour metastasis. These findings provide new insight into potential markers or therapeutic targets for HCC treatment.
Collapse
|
22
|
Tian J, Ma C, Yang L, Sun Y, Zhang Y. Prognostic Value and Immunological Characteristics of a Novel RNA Binding Protein Signature in Cutaneous Melanoma. Front Genet 2021; 12:723796. [PMID: 34531901 PMCID: PMC8438157 DOI: 10.3389/fgene.2021.723796] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 08/09/2021] [Indexed: 12/23/2022] Open
Abstract
Background The existing studies indicate that RNA binding proteins (RBPs) are closely correlated with the genesis and development of cancers. However, the role of RBPs in cutaneous melanoma remains largely unknown. Therefore, the present study aims to establish a reliable prognostic signature based on RBPs to distinguish cutaneous melanoma patients with different prognoses and investigate the immune infiltration of patients. Methods After screening RBPs from the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases, Cox and least absolute shrinkage and selection operator (LASSO) regression analysis were then used to establish a prediction model. The relationship between the signature and the abundance of immune cell types, the tumor microenvironment (TME), immune-related pathways, and immune checkpoints were also analyzed. Results In total, 7 RBPs were selected to establish the prognostic signature. Patients categorized as a high-risk group demonstrated worse overall survival (OS) rates compared to those of patients categorized as a low-risk group. The signature was validated in an independent external cohort and indicated a promising prognostic ability. Further analysis indicated that the signature wasan independent prognostic indicator in cutaneous melanoma. A nomogram combining risk score and clinicopathological features was then established to evaluate the 3- and 5-year OS in cutaneous melanoma patients. Analyses of immune infiltrating, the TME, immune checkpoint, and drug susceptibility revealed significant differences between the two groups. GSEA analysis revealed that basal cell carcinoma, notch signaling pathway, melanogenesis pathways were enriched in the high-risk group, resulting in poor OS. Conclusion We established and validated a robust 7-RBP signature that could be a potential biomarker to predict the prognosis and immunotherapy response of cutaneous melanoma patients, which provides new insights into cutaneous melanoma immunotherapeutic strategies.
Collapse
Affiliation(s)
- Jun Tian
- Department of Dermatology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Chongzhi Ma
- Department of Dermatology, The 63600 Hospital of PLA, Lanzhou, China
| | - Li Yang
- Department of Dermatology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Yang Sun
- Department of Oncology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Yuan Zhang
- Department of Oncology, Shaanxi Provincial People's Hospital, Xi'an, China
| |
Collapse
|
23
|
Fabbri L, Chakraborty A, Robert C, Vagner S. The plasticity of mRNA translation during cancer progression and therapy resistance. Nat Rev Cancer 2021; 21:558-577. [PMID: 34341537 DOI: 10.1038/s41568-021-00380-y] [Citation(s) in RCA: 137] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/14/2021] [Indexed: 02/07/2023]
Abstract
Translational control of mRNAs during gene expression allows cells to promptly and dynamically adapt to a variety of stimuli, including in neoplasia in response to aberrant oncogenic signalling (for example, PI3K-AKT-mTOR, RAS-MAPK and MYC) and microenvironmental stress such as low oxygen and nutrient supply. Such translational rewiring allows rapid, specific changes in the cell proteome that shape specific cancer phenotypes to promote cancer onset, progression and resistance to anticancer therapies. In this Review, we illustrate the plasticity of mRNA translation. We first highlight the diverse mechanisms by which it is regulated, including by translation factors (for example, eukaryotic initiation factor 4F (eIF4F) and eIF2), RNA-binding proteins, tRNAs and ribosomal RNAs that are modulated in response to aberrant intracellular pathways or microenvironmental stress. We then describe how translational control can influence tumour behaviour by impacting on the phenotypic plasticity of cancer cells as well as on components of the tumour microenvironment. Finally, we highlight the role of mRNA translation in the cellular response to anticancer therapies and its promise as a key therapeutic target.
Collapse
Affiliation(s)
- Lucilla Fabbri
- Institut Curie, PSL Research University, CNRS UMR3348, INSERM U1278, Orsay, France
- Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, INSERM U1278, Orsay, France
- Equipe Labellisée Ligue Nationale Contre le Cancer, Orsay, France
| | - Alina Chakraborty
- Institut Curie, PSL Research University, CNRS UMR3348, INSERM U1278, Orsay, France
- Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, INSERM U1278, Orsay, France
- Equipe Labellisée Ligue Nationale Contre le Cancer, Orsay, France
| | - Caroline Robert
- INSERM U981, Gustave Roussy Cancer Campus, Villejuif, France
- Université Paris-Sud, Université Paris-Saclay, Kremlin-Bicêtre, France
- Dermato-Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Stéphan Vagner
- Institut Curie, PSL Research University, CNRS UMR3348, INSERM U1278, Orsay, France.
- Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, INSERM U1278, Orsay, France.
- Equipe Labellisée Ligue Nationale Contre le Cancer, Orsay, France.
- Dermato-Oncology, Gustave Roussy Cancer Campus, Villejuif, France.
| |
Collapse
|
24
|
Huynh TK, Huang CH, Chen JY, Yao JH, Yang YS, Wei YL, Chen HF, Chen CH, Tu CY, Hsu YM, Liu LC, Huang WC. MiR-221 confers lapatinib resistance by negatively regulating p27 kip1 in HER2-positive breast cancer. Cancer Sci 2021; 112:4234-4245. [PMID: 34382727 PMCID: PMC8486195 DOI: 10.1111/cas.15107] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 08/02/2021] [Accepted: 08/09/2021] [Indexed: 12/02/2022] Open
Abstract
Development of acquired resistance to lapatinib, a dual epidermal growth factor receptor (EGFR)/human epidermal growth factor receptor 2 (HER2) tyrosine kinase inhibitor, severely limits the duration of clinical response in advanced HER2‐driven breast cancer patients. Although the compensatory activation of the PI3K/Akt survival signal has been proposed to cause acquired lapatinib resistance, comprehensive molecular mechanisms remain required to develop more efficient strategies to circumvent this therapeutic difficulty. In this study, we found that suppression of HER2 by lapatinib still led to Akt inactivation and elevation of FOX3a protein levels, but failed to induce the expression of their downstream pro‐apoptotic effector p27kip1, a cyclin‐dependent kinase inhibitor. Elevation of miR‐221 was found to contribute to the development of acquired lapatinib resistance by targeting p27kip1 expression. Furthermore, upregulation of miR‐221 was mediated by the lapatinib‐induced Src family tyrosine kinase and subsequent NF‐κB activation. The reversal of miR‐221 upregulation and p27kip1 downregulation by a Src inhibitor, dasatinib, can overcome lapatinib resistance. Our study not only identified miRNA‐221 as a pivotal factor conferring the acquired resistance of HER2‐positive breast cancer cells to lapatinib through negatively regulating p27kip1 expression, but also suggested Src inhibition as a potential strategy to overcome lapatinib resistance.
Collapse
Affiliation(s)
- Thanh Kieu Huynh
- Graduate Institute of Biomedical Sciences, Drug Development Center, China Medical University, Taichung, 404, Taiwan.,Center for Molecular Medicine, China Medical University Hospital, Taichung, 404, Taiwan
| | - Chih-Hao Huang
- Graduate Institute of Biomedical Sciences, Drug Development Center, China Medical University, Taichung, 404, Taiwan.,Division of Breast Surgery, China Medical University Hospital, Taichung, 40402, Taiwan
| | - Jhen-Yu Chen
- Graduate Institute of Biomedical Sciences, Drug Development Center, China Medical University, Taichung, 404, Taiwan
| | - Jin-Han Yao
- School of Medicine, China Medical University, Taichung, 404, Taiwan
| | - Yi-Shiang Yang
- Center for Molecular Medicine, China Medical University Hospital, Taichung, 404, Taiwan
| | - Ya-Ling Wei
- Center for Molecular Medicine, China Medical University Hospital, Taichung, 404, Taiwan
| | - Hsiao-Fan Chen
- Center for Molecular Medicine, China Medical University Hospital, Taichung, 404, Taiwan
| | - Chia-Hung Chen
- School of Medicine, China Medical University, Taichung, 404, Taiwan.,Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, China Medical University Hospital, Taichung, 40402, Taiwan
| | - Chih-Yen Tu
- School of Medicine, China Medical University, Taichung, 404, Taiwan.,Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, China Medical University Hospital, Taichung, 40402, Taiwan
| | - Yuan-Man Hsu
- Department of Biological Science and Technology, China Medical University, Taichung, 404, Taiwan.,Department of Animal Science and Technology, Agriculture College, Tunghai University, Taichung, 40704, Taiwan
| | - Liang-Chih Liu
- Division of Breast Surgery, China Medical University Hospital, Taichung, 40402, Taiwan.,School of Medicine, China Medical University, Taichung, 404, Taiwan
| | - Wei-Chien Huang
- Graduate Institute of Biomedical Sciences, Drug Development Center, China Medical University, Taichung, 404, Taiwan.,Center for Molecular Medicine, China Medical University Hospital, Taichung, 404, Taiwan.,The Ph.D. program for Cancer Biology and Drug Discovery, China Medical University and Academia Sinica, Taichung, 404, Taiwan.,Department of Biotechnology, Asia University, Taichung, 413, Taiwan
| |
Collapse
|
25
|
Priyanka P, Sharma M, Das S, Saxena S. The lncRNA HMS recruits RNA-binding protein HuR to stabilize the 3'-UTR of HOXC10 mRNA. J Biol Chem 2021; 297:100997. [PMID: 34302808 PMCID: PMC8363838 DOI: 10.1016/j.jbc.2021.100997] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 07/09/2021] [Accepted: 07/19/2021] [Indexed: 12/27/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) have been reported to drive key cancer pathways but the functions of majority of lncRNAs are unknown making a case for comprehensive functional evaluation of lncRNAs. With an aim to identify lncRNAs dysregulated in human cancers, we analyzed the cancer patient database of lung adenocarcinoma (LUAD), which revealed an upregulated lncRNA, LINC02381 (renamed HOXC10mRNA stabilizing factor or HMS in this study), whose depletion results in proliferation defects and inhibition of colony formation of human cancer cells. In order to identify the binding targets of HMS, we screened for cis-genes and discovered that HOXC10, an oncogene, is downregulated in the absence of HMS. Depletion of HMS does not affect the HOXC10 promoter activity but inhibits the HOXC10 3′-UTR-linked luciferase reporter activity. Since lncRNAs have been known to associate with RNA-binding proteins (RBPs) to stabilize mRNA transcripts, we screened for different RBPs and discovered that HuR, an ELAV family protein, stabilizes HOXC10 mRNA. Using RNA pull-down and deletion mapping experiments, we show that HuR physically interacts with the cytosine-rich stretch of HMS and HOXC10 3′-UTR to stabilize HOXC10 mRNA. HOXC10 is overexpressed in many human cancers, and our discovery highlights that lncRNA HMS sustains the HOXC10 mRNA levels to maintain the invasive phenotypes of cancer cells.
Collapse
Affiliation(s)
- Priyanka Priyanka
- DNA Replication and Cell Cycle Laboratory, National Institute of Immunology, New Delhi, India
| | | | - Sanjeev Das
- DNA Replication and Cell Cycle Laboratory, National Institute of Immunology, New Delhi, India
| | - Sandeep Saxena
- DNA Replication and Cell Cycle Laboratory, National Institute of Immunology, New Delhi, India; Department of Biotechnology, JNU, New Delhi, India.
| |
Collapse
|
26
|
Yuan C, Chen M, Cai X. Advances in poly(rC)-binding protein 2: Structure, molecular function, and roles in cancer. Biomed Pharmacother 2021; 139:111719. [PMID: 34233389 DOI: 10.1016/j.biopha.2021.111719] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 04/29/2021] [Accepted: 05/07/2021] [Indexed: 02/08/2023] Open
Abstract
Poly(rC)-binding protein 2 (PCBP2) is an RNA-binding protein that is characterized by its ability to interact with poly(C) with high affinity in a sequence-specific manner. PCBP2 contains three K homology domains, which are consensus RNA-binding domains that play a role in recognizing and combining with RNA and DNA. The specific structure and localization of PCBP2 lay the foundation for its multiple roles in transcriptional, posttranscriptional, and translational processes, even in iron metabolism. Numerous studies have indicated that PCBP2 expression is increased in many cancer types. PCBP2 is considered as an oncogene that promotes tumorigenesis, development of cancer cells, and metastasis. Here, we summarized the current evidence regarding PCBP2 in the proliferation, migration, invasion of cancer cells, and drug resistance, aiming to clarify the molecular mechanisms of PCBP2 in cancer. Results from this review suggest that an in-depth study of PCBP2 in cancer may provide novel biomarkers for prognostic or therapeutic purposes.
Collapse
Affiliation(s)
- Chendong Yuan
- Department of Vascular Surgery, Zhuji Affiliated Hospital of Shaoxing University, Zhuji, Zhejiang 311800, China.
| | - Mingxiang Chen
- Department of Cardiovascular surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, Yubei 401120, China.
| | - Xiaolu Cai
- Department of Oncological Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China.
| |
Collapse
|
27
|
Wu Y, Zhao H, Zhang EE, Liu N. Identification of PCBP1 as a Novel Modulator of Mammalian Circadian Clock. Front Genet 2021; 12:656571. [PMID: 33841513 PMCID: PMC8034388 DOI: 10.3389/fgene.2021.656571] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 03/05/2021] [Indexed: 11/13/2022] Open
Abstract
The circadian clock governs our daily cycle of behavior and physiology. Previous studies have identified a handful of core clock components and hundreds of circadian modifiers. Here, we report the discovery that poly(C)-binding protein 1 (PCBP1), displaying a circadian expression pattern, was a novel circadian clock regulator. We found that knocking down PCBP1 resulted in period shortening in human U2OS cells, and that manipulations of PCBP1 expression altered the activity of CLOCK/BMAL1 in an E-box-based reporter assay. Further mechanistic study demonstrated that this clock function of PCBP1 appears to work by enhancing the association of Cryptochrome 1 (CRY1) with the CLOCK/BMAL1 complex, thereby negatively regulating the latter’s activation. Co-immunoprecipitation of PCBP1 and core clock molecules confirmed the interactions between PCBP1 and CRY1, and a time-course qPCR assay revealed the rhythmic expression of PCBP1 in mouse hearts in vivo. Given that the RNA interference of mushroom-body expressed (mub), the poly(rC) binding protein (PCBP) homolog of Drosophila, in the clock neurons also led to a circadian phenotype in the locomotor assay, our study deemed PCBP1 a novel clock modifier whose circadian regulatory mechanism is conserved during evolution.
Collapse
Affiliation(s)
- Yaling Wu
- Hubei Engineering Research Center of Special Wild Vegetables Breeding and Comprehensive Utilization Technology, Hubei Normal University, Huangshi, China.,Hubei Key Laboratory of Edible Wild Plants Conservation and Utilization, Hubei Normal University, Huangshi, China.,National Demonstration Center for Experimental Biology Education, Hubei Normal University, Huangshi, China.,College of Life Sciences, Hubei Normal University, Huangshi, China.,National Institute of Biological Sciences, Beijing, China
| | - Haijiao Zhao
- National Institute of Biological Sciences, Beijing, China
| | | | - Na Liu
- Hubei Engineering Research Center of Special Wild Vegetables Breeding and Comprehensive Utilization Technology, Hubei Normal University, Huangshi, China.,Hubei Key Laboratory of Edible Wild Plants Conservation and Utilization, Hubei Normal University, Huangshi, China.,National Demonstration Center for Experimental Biology Education, Hubei Normal University, Huangshi, China.,College of Life Sciences, Hubei Normal University, Huangshi, China.,National Institute of Biological Sciences, Beijing, China
| |
Collapse
|
28
|
Explaining decisions of graph convolutional neural networks: patient-specific molecular subnetworks responsible for metastasis prediction in breast cancer. Genome Med 2021; 13:42. [PMID: 33706810 PMCID: PMC7953710 DOI: 10.1186/s13073-021-00845-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 02/05/2021] [Indexed: 12/19/2022] Open
Abstract
Background Contemporary deep learning approaches show cutting-edge performance in a variety of complex prediction tasks. Nonetheless, the application of deep learning in healthcare remains limited since deep learning methods are often considered as non-interpretable black-box models. However, the machine learning community made recent elaborations on interpretability methods explaining data point-specific decisions of deep learning techniques. We believe that such explanations can assist the need in personalized precision medicine decisions via explaining patient-specific predictions. Methods Layer-wise Relevance Propagation (LRP) is a technique to explain decisions of deep learning methods. It is widely used to interpret Convolutional Neural Networks (CNNs) applied on image data. Recently, CNNs started to extend towards non-Euclidean domains like graphs. Molecular networks are commonly represented as graphs detailing interactions between molecules. Gene expression data can be assigned to the vertices of these graphs. In other words, gene expression data can be structured by utilizing molecular network information as prior knowledge. Graph-CNNs can be applied to structured gene expression data, for example, to predict metastatic events in breast cancer. Therefore, there is a need for explanations showing which part of a molecular network is relevant for predicting an event, e.g., distant metastasis in cancer, for each individual patient. Results We extended the procedure of LRP to make it available for Graph-CNN and tested its applicability on a large breast cancer dataset. We present Graph Layer-wise Relevance Propagation (GLRP) as a new method to explain the decisions made by Graph-CNNs. We demonstrate a sanity check of the developed GLRP on a hand-written digits dataset and then apply the method on gene expression data. We show that GLRP provides patient-specific molecular subnetworks that largely agree with clinical knowledge and identify common as well as novel, and potentially druggable, drivers of tumor progression. Conclusions The developed method could be potentially highly useful on interpreting classification results in the context of different omics data and prior knowledge molecular networks on the individual patient level, as for example in precision medicine approaches or a molecular tumor board. Supplementary Information The online version contains supplementary material available at (10.1186/s13073-021-00845-7).
Collapse
|
29
|
Lee HY, Son SW, Moeng S, Choi SY, Park JK. The Role of Noncoding RNAs in the Regulation of Anoikis and Anchorage-Independent Growth in Cancer. Int J Mol Sci 2021; 22:ijms22020627. [PMID: 33435156 PMCID: PMC7827914 DOI: 10.3390/ijms22020627] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 01/02/2021] [Accepted: 01/06/2021] [Indexed: 12/11/2022] Open
Abstract
Cancer is a global health concern, and the prognosis of patients with cancer is associated with metastasis. Multistep processes are involved in cancer metastasis. Accumulating evidence has shown that cancer cells acquire the capacity of anoikis resistance and anchorage-independent cell growth, which are critical prerequisite features of metastatic cancer cells. Multiple cellular factors and events, such as apoptosis, survival factors, cell cycle, EMT, stemness, autophagy, and integrins influence the anoikis resistance and anchorage-independent cell growth in cancer. Noncoding RNAs (ncRNAs), such as microRNAs (miRNAs) and long noncoding RNAs (lncRNAs), are dysregulated in cancer. They regulate cellular signaling pathways and events, eventually contributing to cancer aggressiveness. This review presents the role of miRNAs and lncRNAs in modulating anoikis resistance and anchorage-independent cell growth. We also discuss the feasibility of ncRNA-based therapy and the natural features of ncRNAs that need to be contemplated for more beneficial therapeutic strategies against cancer.
Collapse
|
30
|
Zhang X, Di C, Chen Y, Wang J, Su R, Huang G, Xu C, Chen X, Long F, Yang H, Zhang H. Multilevel regulation and molecular mechanism of poly (rC)-binding protein 1 in cancer. FASEB J 2020; 34:15647-15658. [PMID: 33058239 DOI: 10.1096/fj.202000911r] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 09/02/2020] [Accepted: 09/15/2020] [Indexed: 01/07/2023]
Abstract
Poly (rC)-binding protein 1 (PCBP1), an RNA- or DNA-binding protein with a relative molecular weight of 38 kDa, which is characterized by downregulation in many cancer types. Numerous cases have indicated that PCBP1 could be considered as a tumor suppressor to inhibit tumorigenesis, development, and metastasis. In the current review, we described the multilevel regulatory roles of PCBP1, including gene transcription, alternative splicing, and translation of many cancer-related genes. Additionally, we also provided a brief overview about the inhibitory effect of PCBP1 on most common tumors. More importantly, we summarized the current research status about PCBP1 in hypoxic microenvironment, autophagy, apoptosis, and chemotherapy of cancer cells, aiming to clarify the molecular mechanisms of PCBP1 in cancer. Taken together, in-depth study of PCBP1 in cancer may provide new ideas for cancer therapy.
Collapse
Affiliation(s)
- Xuetian Zhang
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China.,Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.,School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Cuixia Di
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China.,Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.,School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Yuhong Chen
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China.,Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.,School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Jing Wang
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Ruowei Su
- The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Guomin Huang
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China.,Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.,School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Caipeng Xu
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China.,Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.,School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Xiaohua Chen
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China.,Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.,School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Feng Long
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Hongying Yang
- School of Radiation Medicine and Protection, Medical College of Soochow, Soochow, China
| | - Hong Zhang
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China.,Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.,School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
31
|
The HMGB1-2 Ovarian Cancer Interactome. The Role of HMGB Proteins and Their Interacting Partners MIEN1 and NOP53 in Ovary Cancer and Drug-Response. Cancers (Basel) 2020; 12:cancers12092435. [PMID: 32867128 PMCID: PMC7564582 DOI: 10.3390/cancers12092435] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/22/2020] [Accepted: 08/24/2020] [Indexed: 12/14/2022] Open
Abstract
High mobility group box B (HMGB) proteins are overexpressed in different types of cancers such as epithelial ovarian cancers (EOC). We have determined the first interactome of HMGB1 and HMGB2 in epithelial ovarian cancer (the EOC-HMGB interactome). Libraries from the SKOV-3 cell line and a primary transitional cell carcinoma (TCC) ovarian tumor were tested by the Yeast Two Hybrid (Y2H) approach. The interactome reveals proteins that are related to cancer hallmarks and their expression is altered in EOC. Moreover, some of these proteins have been associated to survival and prognosis of patients. The interaction of MIEN1 and NOP53 with HMGB2 has been validated by co-immunoprecipitation in SKOV-3 and PEO1 cell lines. SKOV-3 cells were treated with different anti-tumoral drugs to evaluate changes in HMGB1, HMGB2, MIEN1 and NOP53 gene expression. Results show that combined treatment of paclitaxel and carboplatin induces a stronger down-regulation of these genes in comparison to individual treatments. Individual treatment with paclitaxel or olaparib up-regulates NOP53, which is expressed at lower levels in EOC than in non-cancerous cells. On the other hand, bevacizumab diminishes the expression of HMGB2 and NOP53. This study also shows that silencing of these genes affects cell-viability after drug exposure. HMGB1 silencing causes loss of response to paclitaxel, whereas silencing of HMGB2 slightly increases sensitivity to olaparib. Silencing of either HMGB1 or HMGB2 increases sensitivity to carboplatin. Lastly, a moderate loss of response to bevacizumab is observed when NOP53 is silenced.
Collapse
|
32
|
Ishii T, Igawa T, Hayakawa H, Fujita T, Sekiguchi M, Nakabeppu Y. PCBP1 and PCBP2 both bind heavily oxidized RNA but cause opposing outcomes, suppressing or increasing apoptosis under oxidative conditions. J Biol Chem 2020; 295:12247-12261. [PMID: 32647012 PMCID: PMC7443489 DOI: 10.1074/jbc.ra119.011870] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 07/06/2020] [Indexed: 12/18/2022] Open
Abstract
PCBP1, a member of the poly(C)-binding protein (PCBP) family, has the capability of binding heavily oxidized RNA and therefore participates in the cellular response to oxidative conditions, helping to induce apoptosis. There are four other members of this family, PCBP2, PCBP3, PCBP4, and hnRNPK, but it is not known whether they play similar roles. To learn more, we first tested their affinity for an RNA strand carrying two 8-oxoguanine (8-oxoG) residues at sites located in close proximity to each other, representative of a heavily oxidized strand or RNA with one 8-oxoG or none. Among them, only PCBP2 exhibited highly selective binding to RNA carrying two 8-oxoG residues similar to that observed with PCBP1. In contrast, PCBP3, PCBP4, and hnRNPK bound RNA with or without 8-oxoG modifications and exhibited slightly increased binding to the former. Mutations in conserved RNA-binding domains of PCBP2 disrupted the specific interaction with heavily oxidized RNA. We next tested PCBP2 activity in cells. Compared with WT HeLa S3 cells, PCBP2-KO cells established by gene editing exhibited increased apoptosis with increased caspase-3 activity and PARP1 cleavage under oxidative conditions, which were suppressed by the expression of WT PCBP2 but not one of the mutants lacking binding activity. In contrast, PCBP1-KO cells exhibited reduced apoptosis with much less caspase-3 activity and PARP cleavage than WT cells. Our results indicate that PCBP2 as well as PCBP1 bind heavily oxidized RNA; however, the former may counteract PCBP1 to suppress apoptosis under oxidative conditions.
Collapse
Affiliation(s)
- Takashi Ishii
- Department of Biochemistry, Fukuoka Dental College, Fukuoka, Japan; Oral Medicine Research Center, Fukuoka Dental College, Fukuoka, Japan
| | - Tatsuhiro Igawa
- Frontier Research Center, Fukuoka Dental College, Fukuoka, Japan
| | - Hiroshi Hayakawa
- Department of Biochemistry, Fukuoka Dental College, Fukuoka, Japan
| | - Tsugumi Fujita
- Department of Biochemistry, Fukuoka Dental College, Fukuoka, Japan
| | - Mutsuo Sekiguchi
- Frontier Research Center, Fukuoka Dental College, Fukuoka, Japan
| | - Yusaku Nakabeppu
- Division of Neurofunctional Genomics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
33
|
Zhang W, Zhang S, Guan W, Huang Z, Kong J, Huang C, Wang H, Yang S. Poly C Binding Protein 1 Regulates p62/SQSTM1 mRNA Stability and Autophagic Degradation to Repress Tumor Progression. Front Genet 2020; 11:930. [PMID: 32922440 PMCID: PMC7457068 DOI: 10.3389/fgene.2020.00930] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 07/27/2020] [Indexed: 12/11/2022] Open
Abstract
Accumulating evidence show that Poly C Binding Protein 1 (PCBP1) is deleted in distinct types of tumors as a novel tumor suppressor, but its tumor suppression mechanism remains elusive. Here, we firstly describe that downregulation of PCBP1 is significantly associated with clinical ovarian tumor progression. Mechanistically, PCBP1 overexpression affects various autophagy-related genes expression at various expression levels to attenuate the intrinsic cell autophagy, including the autophagy-initiating ULK, ATG12, ATG7 as well as the bona fide marker of autophagosome, LC3B. Accordingly, knockdown of the endogenous PCBP1 in turn enhances autophagy and less cell death. Meanwhile, PCBP1 upregulates p62/SQSTM1 via inhibition p62/SQSTM1 autophagolysome and proteasome degradation as well as its mRNA stability, consequently accompanying with the caspase 3 or 8 activation for tumor cell apoptosis. Importantly, clinical ovary cancer sample analysis consistently validates the relevance of PCBP1 expression to both p62/SQSTM1 and caspase-8 to overall survival, and indicates PCBP1 may be a master player to repress tumor initiation. Taken together, our results uncover the tumorigenic mechanism of PCBP1 depletion and suggest that inhibition of tumor cell autophagy with autophagic inhibitors could be an effective therapeutical strategy for PCBP1-deficient tumor.
Collapse
Affiliation(s)
- Wenliang Zhang
- Translational Medicine Centre, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Shaoyang Zhang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Wen Guan
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhicong Huang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jianqiu Kong
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Chunlong Huang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Haihe Wang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Center for Stem Cell Biology and Tissue Engineering, Sun Yat-sen University, Guangzhou, China
| | - Shulan Yang
- Translational Medicine Centre, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Engineering and Technology Research Center for Disease-Model Animals, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
34
|
Yanatori I, Richardson DR, Toyokuni S, Kishi F. The new role of poly (rC)-binding proteins as iron transport chaperones: Proteins that could couple with inter-organelle interactions to safely traffic iron. Biochim Biophys Acta Gen Subj 2020; 1864:129685. [PMID: 32679248 DOI: 10.1016/j.bbagen.2020.129685] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/07/2020] [Accepted: 07/11/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Intracellular iron transport is mediated by iron chaperone proteins known as the poly(rC)-binding proteins (PCBPs), which were originally identified as RNA/DNA-binding molecules. SCOPE OF REVIEW PCBPs assume a role as not only as cytosolic iron carriers, but also as regulators of iron transport and recycling. PCBP1 is involved in the iron storage pathway that involves ferritin, while PCBP2 is involved in processes that include: iron transfer from the iron importer, divalent metal ion transporter 1; iron export mediated by ferroportin-1; and heme degradation via heme oxygenase 1. MAJOR CONCLUSIONS Both PCBP1 and PCBP2 possess iron-binding activity and form hetero/homo dimer complexes. These iron chaperones have a subset of non-redundant functions and regulate iron metabolism independently. GENERAL SIGNIFICANCE This intracellular iron chaperone system mediated by PCBPs provide a transport "gateway" of ferrous iron that may potentially link with dynamic, inter-organelle interactions to safely traffic intracellular iron.
Collapse
Affiliation(s)
- Izumi Yanatori
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Des R Richardson
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan; Centre for Cancer Cell Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane 4111, Queensland, Australia
| | - Shinya Toyokuni
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | | |
Collapse
|
35
|
Smith SM. Molecular biology meets the endocrine pathologist: an appraisal of p27 in thyroid malignancy. DIAGNOSTIC HISTOPATHOLOGY 2020; 26:216-223. [DOI: 10.1016/j.mpdhp.2020.02.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
36
|
Functional expression of ZNF467 and PCBP2 supports adipogenic lineage commitment in adipose-derived mesenchymal stem cells. Gene 2020; 737:144437. [PMID: 32032745 DOI: 10.1016/j.gene.2020.144437] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 02/03/2020] [Indexed: 01/13/2023]
Abstract
Bone marrow-derived mesenchymal stromal/stem cells (BMSCs) have the potential to be employed in many different skeletal therapies. A major limitation to utilizing BMSCs as a therapeutic strategy in human disease and tissue regeneration is the low cell numbers obtained from initial isolation necessitating multiple cell passages that can lead to decreased cell quality. Adipose-derived mesenchymal stromal/stem cells (AMSCs) have been proposed as an alternative cell source for regenerative therapies; however the differentiation capacity of these cells differs from BMSCs. To understand the differences between BMSCs and AMSCs, we compared the global gene expression profiles of BMSCs and AMSCs and identified two genes, PCBP2 and ZNF467 that were differentially expressed between AMSCs and BMSCs. We demonstrate that PCBP2 and ZNF467 impact adipogenic but not osteogenic differentiation, further supporting evidence that AMSCs and BMSCs appear to be adapted to their microenvironment.
Collapse
|
37
|
Inhibition of Caspase-2 Translation by the mRNA Binding Protein HuR: A Novel Path of Therapy Resistance in Colon Carcinoma Cells? Cells 2019; 8:cells8080797. [PMID: 31366165 PMCID: PMC6721497 DOI: 10.3390/cells8080797] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 07/26/2019] [Accepted: 07/29/2019] [Indexed: 12/28/2022] Open
Abstract
An increased expression and cytoplasmic abundance of the ubiquitous RNA binding protein human antigen R (HuR) is critically implicated in the dysregulated control of post- transcriptional gene expression during colorectal cancer development and is frequently associated with a high grade of malignancy and therapy resistance. Regardless of the fact that HuR elicits a broad cell survival program by increasing the stability of mRNAs coding for prominent anti-apoptotic factors, recent data suggest that HuR is critically involved in the regulation of translation, particularly, in the internal ribosome entry site (IRES) controlled translation of cell death regulatory proteins. Accordingly, data from human colon carcinoma cells revealed that HuR maintains constitutively reduced protein and activity levels of caspase-2 through negative interference with IRES-mediated translation. This review covers recent advances in the understanding of mechanisms underlying HuR's modulatory activity on IRES-triggered translation. With respect to the unique regulatory features of caspase-2 and its multiple roles (e.g., in DNA-damage-induced apoptosis, cell cycle regulation and maintenance of genomic stability), the pathophysiological consequences of negative caspase-2 regulation by HuR and its impact on therapy resistance of colorectal cancers will be discussed in detail. The negative HuR-caspase-2 axis may offer a novel target for tumor sensitizing therapies.
Collapse
|
38
|
Nazarov IB, Bakhmet EI, Tomilin AN. KH-Domain Poly(C)-Binding Proteins as Versatile Regulators of Multiple Biological Processes. BIOCHEMISTRY (MOSCOW) 2019; 84:205-219. [PMID: 31221059 DOI: 10.1134/s0006297919030039] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Five known members of the family of KH-domain poly(C)-binding proteins (Pcbp1-4, hnRNP-K) have an unusually broad spectrum of cellular functions that include regulation of gene transcription, regulation of pre-mRNA processing, splicing, mRNA stability, translational silencing and enhancement, the control of iron turnover, and many others. Mechanistically, these proteins act via nucleic acid binding and protein-protein interactions. Through performing these multiple tasks, the KH-domain poly(C)-binding family members are involved in a wide variety of biological processes such as embryonic development, cell differentiation, and cancer. Deregulation of KH-domain protein expression is frequently associated with severe developmental defects and neoplasia. This review summarizes progress in studies of the KH-domain proteins made over past two decades. The review also reports our recent finding implying an involvement of the KH-factor Pcbp1 into control of transition from naïve to primed pluripotency cell state.
Collapse
Affiliation(s)
- I B Nazarov
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, 194064, Russia.
| | - E I Bakhmet
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, 194064, Russia
| | - A N Tomilin
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, 194064, Russia
| |
Collapse
|
39
|
Mohibi S, Chen X, Zhang J. Cancer the'RBP'eutics-RNA-binding proteins as therapeutic targets for cancer. Pharmacol Ther 2019; 203:107390. [PMID: 31302171 DOI: 10.1016/j.pharmthera.2019.07.001] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 07/02/2019] [Indexed: 12/11/2022]
Abstract
RNA-binding proteins (RBPs) play a critical role in the regulation of various RNA processes, including splicing, cleavage and polyadenylation, transport, translation and degradation of coding RNAs, non-coding RNAs and microRNAs. Recent studies indicate that RBPs not only play an instrumental role in normal cellular processes but have also emerged as major players in the development and spread of cancer. Herein, we review the current knowledge about RNA binding proteins and their role in tumorigenesis as well as the potential to target RBPs for cancer therapeutics.
Collapse
Affiliation(s)
- Shakur Mohibi
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California at Davis, United States
| | - Xinbin Chen
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California at Davis, United States
| | - Jin Zhang
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California at Davis, United States.
| |
Collapse
|
40
|
Meng N, Gong Y, Mu X, Wang YH, Su L, Jiang CS, Zhang H. Novel Role of Heterogeneous Nuclear Ribonucleoprotein E1 in Regulation of Apoptosis and Autophagy by a Triazole Derivative in Vascular Endothelial Cells. Int J Biol Sci 2019; 15:1299-1309. [PMID: 31223288 PMCID: PMC6567801 DOI: 10.7150/ijbs.32677] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Accepted: 04/04/2019] [Indexed: 11/08/2022] Open
Abstract
Vascular endothelial cell (VEC) apoptosis and autophagy play an important role in the maintenance of vascular homeostasis. However, the association of molecular mechanisms between vascular endothelial cell apoptosis and autophagy has not been clarified. Here, we identified a novel triazole derivative, JL014, which could inhibit human umbilical vein vascular endothelial cell (HUVEC) apoptosis induced by deprivation of serum and fibroblast growth factor 2 and maintain HUVEC survival by promoting autophagy. Importantly, JL014 increased the mRNA and protein level of heterogeneous nuclear ribonucleoprotein E1 (hnRNP E1) in HUVECs. In addition, knockdown of hnRNP E1 by RNA interference inhibited the effects of JL014 on VEC apoptosis and autophagy. Furthermore, we investigated the effect of JL014 on the expression of HMBOX1, a key VEC apoptosis inhibitor and autophagy inducer by inhibiting mTOR signaling and the level of cleaved caspase-3. Our results demonstrated that JL014 enhanced mRNA transcription and increased protein synthesis of HMBOX1. JL014 also inhibited mTOR signaling and the cleaved caspase-3 level. Mechanistic studies revealed that hnRNP E1 could bind to the promoter and 5'UTR of HMBOX1 and active HMBOX1 expression. Therefore, our results firmly establish hnRNP E1 as a new regulator of VEC apoptosis and autophagy through mediating HMBOX1 expression, and opened the door to a novel therapeutic drug for related vascular diseases.
Collapse
Affiliation(s)
- Ning Meng
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Yan Gong
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Xin Mu
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Yan Hong Wang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Le Su
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan, 250353, China
| | - Cheng Shi Jiang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Hua Zhang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| |
Collapse
|
41
|
Wang X, Guo J, Che X, Jia R. PCBP1 inhibits the expression of oncogenic STAT3 isoform by targeting alternative splicing of STAT3 exon 23. Int J Biol Sci 2019; 15:1177-1186. [PMID: 31223278 PMCID: PMC6567812 DOI: 10.7150/ijbs.33103] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Accepted: 03/28/2019] [Indexed: 01/27/2023] Open
Abstract
STAT3 plays very important roles in the initiation and development of tumors. Despite of extensive studies in repressing its activation and function via multiple ways, so far, there are few effective therapeutic methods to inhibit STAT3 in the clinic. STAT3 has two isoforms generated by alternative splicing of exon 23. STAT3α is the longer isoform and encodes the full-length oncogenic STAT3α protein. STAT3β is shorter and encodes the truncated and tumor-suppressive STAT3β protein. It remains unknown how the alternative splicing of STAT3 exon 23 is regulated. Here, we discovered that there is an exonic splicing suppressor (ESS) in exon 23. Importantly, splicing factor PCBP1 binds to this ESS. Overexpression of PCBP1 significantly reduced the proportion of STAT3α /STAT3β isoforms and the expression of STAT3α protein. Moreover, increased PCBP1 inhibited the growth of oral squamous cell carcinoma and breast cancer cells, and the expression of STAT3 target genes. Our results demonstrated that PCBP1 is the key splicing factor that promotes the switch from oncogenic isoform STAT3α to tumor-suppressive isoform STAT3β. Our results pave the way for finding new anti-STAT3 methods for cancer treatment.
Collapse
Affiliation(s)
- Xiaole Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, PR China
| | - Jihua Guo
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, PR China.,Department of Endodontics, School & Hospital of Stomatology, Wuhan University, Wuhan, PR China
| | - Xiaoxuan Che
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, PR China
| | - Rong Jia
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, PR China
| |
Collapse
|
42
|
Grelet S, Howe PH. hnRNP E1 at the crossroads of translational regulation of epithelial-mesenchymal transition. ACTA ACUST UNITED AC 2019; 5. [PMID: 31681852 PMCID: PMC6824538 DOI: 10.20517/2394-4722.2018.85] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The epithelial-mesenchymal transition (EMT), in which cells undergo a switch from a polarized, epithelial phenotype to a highly motile fibroblastic or mesenchymal phenotype is fundamental during embryonic development and can be reactivated in a variety of diseases including cancer. Spatio-temporally-regulated mechanisms are constantly orchestrated to allow cells to adapt to their constantly changing environments when disseminating to distant organs. Although numerous transcriptional regulatory factors are currently well-characterized, the post-transcriptional control of EMT requires continued investigation. The hnRNP E1 protein displays a major role in the control of tumor cell plasticity by regulating the translatome through multiple non-redundant mechanisms, and this role is exemplified when E1 is absent. hnRNP E1 binding to RNA molecules leads to direct or indirect translational regulation of specific sets of proteins: (1) hnRNP E1 binding to specific targets has a direct role in translation by preventing elongation of translation; (2) hnRNP E1-dependent alternative splicing can prevent the generation of a competing long non-coding RNA that acts as a decoy for microRNAs (miRNAs) involved in translational inhibition of EMT master regulators; (3) hnRNP E1 binding to the 3’ untranslated region of transcripts can also positively regulate the stability of certain mRNAs to improve their translation. Globally, hnRNP E1 appears to control proteome reprogramming during cell plasticity, either by direct or indirect regulation of protein translation.
Collapse
Affiliation(s)
- Simon Grelet
- Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina 29425, USA.,Department of Biochemistry, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | - Philip H Howe
- Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina 29425, USA.,Department of Biochemistry, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| |
Collapse
|
43
|
Li X, Xue Y, Liu X, Zheng J, Shen S, Yang C, Chen J, Li Z, Liu L, Ma J, Ma T, Liu Y. ZRANB2/SNHG20/FOXK1 Axis regulates Vasculogenic mimicry formation in glioma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:68. [PMID: 30744670 PMCID: PMC6371528 DOI: 10.1186/s13046-019-1073-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 01/30/2019] [Indexed: 12/11/2022]
Abstract
Background Glioma is the most common intracranial neoplasm with vasculogenic mimicry formation as one form of blood supply. Many RNA-binding proteins and long non-coding RNAs are involved in tumorigenesis of glioma. Methods The expression of ZRANB2, SNHG20 and FOXK1 in glioma were detected by real-time PCR or western blot. The function of ZRANB2/SNHG20/FOXK1 axis in glioma associated with vasculogenic mimicry formation was analyzed. Results ZRANB2 is up-regulated in glioma tissues and glioma cells. ZRANB2 knockdown inhibits the proliferation, migration, invasion and vasculogenic mimicry formation of glioma cells. ZRANB2 binds to SNHG20 and increases its stability. Knockdown of SNHG20 reduces the degradation of FOXK1 mRNA by SMD pathway. FOXK1 inhibits transcription by binding to the promoters of MMP1, MMP9 and VE-Cadherin and inhibits vasculogenic mimicry formation of glioma cells. Conclusions ZRANB2/SNHG20/FOXK1 axis plays an important role in regulating vasculogenic mimicry formation of glioma, which might provide new targets of glioma therapy. Electronic supplementary material The online version of this article (10.1186/s13046-019-1073-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaozhi Li
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, 110004, China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, China
| | - Yixue Xue
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, 110122, China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, China
| | - Xiaobai Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, 110004, China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, China
| | - Jian Zheng
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, 110004, China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, China
| | - Shuyuan Shen
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, 110122, China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, China
| | - Chunqing Yang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, 110004, China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, China
| | - Jiajia Chen
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, 110122, China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, China
| | - Zhen Li
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, 110004, China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, China
| | - Libo Liu
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, 110122, China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, China
| | - Jun Ma
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, 110122, China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, China
| | - Teng Ma
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, 110122, China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, China
| | - Yunhui Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China. .,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, 110004, China. .,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, China.
| |
Collapse
|