1
|
Yang W, Wang Q, Li Q, Han Y, Zhang Y, Zhu L, Zhu L, Piao J. Knockdown of PAIP1 Inhibits Breast Cancer Cell Proliferation by Regulating Cyclin E2 mRNA Stability. Mol Carcinog 2024; 63:2392-2400. [PMID: 39259041 DOI: 10.1002/mc.23817] [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: 07/05/2024] [Revised: 08/14/2024] [Accepted: 08/20/2024] [Indexed: 09/12/2024]
Abstract
Polyadenylate-binding protein-interacting protein 1 (PAIP1) is a protein that modulates translation initiation in eukaryotic cells. Studies have shown that PAIP1 was overexpressed in various type of cancers, and drives cancer progression by promoting cancer cell proliferation, invasion, and migration. In our previous study, we identified that PAIP1 was overexpressed in breast cancer, and the expression was correlated with poor prognosis. However, the biological function of PAIP1 in breast cancer has not been clearly understood. In this study, we constructed PAIP1 specifically silenced breast cancer cells. Then, cell proliferation, cell cycle distribution, and apoptosis were detected in PAIP1 knockdown cells. RNA-seq analysis was performed to predict the downstream target of PAIP1, and the molecular mechanism was explored. As a results, we found that knockdown of PAIP1 repressed cell proliferation, induced cell cycle arrest, and triggers apoptosis. Xenograft mouse model showed that knockdown of PAIP1 inhibits cell growth in vivo. RNA-seq predicted that CCNE2 mRNA was one of the downstream targets of PAIP1. In addition, we identified that knockdown of PAIP1-inhibited cell proliferation through modulating cyclin E2 expression. Mechanically, knockdown of PAIP1 reduces the expression of cyclin E2 by regulating the mRNA stability of cyclin E2. Moreover, in breast cancer tissues, we found that the expression of PAIP1 was positively correlated with cyclin E2. Taken together, our findings establish the role and mechanism of PAIP1 in breast cancer progression, indicating that PAIP1 would be a new therapeutic target for breast cancer treatment.
Collapse
Affiliation(s)
- Wenqing Yang
- Key Laboratory of Pathobiology, State Ethnic Affairs Commission, Yanbian University, Yanji, China
- Department of Pathology, Medical School of Yanbian University, Yanji, China
| | - Qingkun Wang
- Key Laboratory of Pathobiology, State Ethnic Affairs Commission, Yanbian University, Yanji, China
- Department of Pathology, Medical School of Yanbian University, Yanji, China
| | - Qi Li
- Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, China
| | - Yue Han
- Key Laboratory of Pathobiology, State Ethnic Affairs Commission, Yanbian University, Yanji, China
- Department of Pathology, Medical School of Yanbian University, Yanji, China
| | - Yu Zhang
- Key Laboratory of Pathobiology, State Ethnic Affairs Commission, Yanbian University, Yanji, China
- Department of Pathology, Medical School of Yanbian University, Yanji, China
| | - Lu Zhu
- Key Laboratory of Pathobiology, State Ethnic Affairs Commission, Yanbian University, Yanji, China
- Department of Pathology, Medical School of Yanbian University, Yanji, China
| | - Lianhua Zhu
- Key Laboratory of Pathobiology, State Ethnic Affairs Commission, Yanbian University, Yanji, China
- Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, China
| | - Junjie Piao
- Key Laboratory of Pathobiology, State Ethnic Affairs Commission, Yanbian University, Yanji, China
- Department of Pathology, Medical School of Yanbian University, Yanji, China
| |
Collapse
|
2
|
Zheng J, Zhang X, Xue Y, Shao W, Wei Y, Mi S, Yang X, Hu L, Zhang Y, Liang M. PAIP1 binds to pre-mRNA and regulates alternative splicing of cancer pathway genes including VEGFA. BMC Genomics 2024; 25:926. [PMID: 39363305 PMCID: PMC11451205 DOI: 10.1186/s12864-024-10530-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 06/14/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND Poly (A) binding protein interacting protein 1 (PAIP1) has been shown to causally contribute to the development and progression of cancer. However, the mechanisms of the PAIP1 regulation in tumor cells remain poorly understood. RESULTS Here, we used a recently developed UV cross-linking and RNA immunoprecipitation method (iRIP-seq) to map the direct and indirect interaction sites between PAIP1 and RNA on a transcriptome-wide level in HeLa cells. We found that PAIP1 not only binds to 3'UTRs, but also to pre-mRNAs/mRNAs with a strong bias towards the coding region and intron. PAIP1 binding sites are enriched in splicing enhancer consensus GA-rich motifs. RNA-seq analysis revealed that PAIP1 selectively modulates the alternative splicing of genes in some cancer hallmarks including cell migration, the mTOR signaling pathway and the HIF-1 signaling pathway. PAIP1-regulated alternative splicing events were strongly associated with PAIP1 binding, demonstrating that the binding may promote selection of the nearby splice sites. Deletion of a PAIP1 binding site containing seven repeats of GA motif reduced the PAIP1-mediated suppression of the exon 6 inclusion in a VEGFA mRNA isoform. Proteomic analysis of the PAIP1-interacted proteins revealed the enrichment of the spliceosome components and splicing factors. CONCLUSIONS These findings suggest that PAIP1 is both a polyadenylation and alternative splicing regulator, that may play a large role in RNA processing via its role in alternative splicing regulation.
Collapse
Affiliation(s)
- Jianfeng Zheng
- Department of Laboratory Medicine, Baoan Central Hospital of Shenzhen, Shenzhen, 518102, Guangdong, P.R. China
- Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, 541004, Guangxi, China
| | - Xiaoyu Zhang
- First department of infection, second affiliated hospital of Harbin medical university, 246 Xuefu Road, Harbin, 150000, Heilongjiang, China
| | - Yaqiang Xue
- Center for Genome Analysis, ABLife Inc, Optics Valley International Biomedical Park, Building 18-1, East Lake High-Tech Development Zone, Wuhan, 430075, Hubei, China
- ABLife BioBigData Institute, 388 Gaoxin 2nd Road, Wuhan, 430075, Hubei, China
| | - Wenhua Shao
- Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, 541004, Guangxi, China
| | - Yaxun Wei
- Center for Genome Analysis, ABLife Inc, Optics Valley International Biomedical Park, Building 18-1, East Lake High-Tech Development Zone, Wuhan, 430075, Hubei, China
| | - Sisi Mi
- Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, 541004, Guangxi, China
| | - Xiaojie Yang
- Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, 541004, Guangxi, China
| | - Linan Hu
- Harbin Center for Disease Prevention and Control, Harbin, 150056, Heilongjiang, China
| | - Yi Zhang
- Center for Genome Analysis, ABLife Inc, Optics Valley International Biomedical Park, Building 18-1, East Lake High-Tech Development Zone, Wuhan, 430075, Hubei, China.
- ABLife BioBigData Institute, 388 Gaoxin 2nd Road, Wuhan, 430075, Hubei, China.
| | - Ming Liang
- First department of infection, second affiliated hospital of Harbin medical university, 246 Xuefu Road, Harbin, 150000, Heilongjiang, China.
| |
Collapse
|
3
|
Mallikarjuna T, Thummadi NB, Vindal V, Manimaran P. Prioritizing cervical cancer candidate genes using chaos game and fractal-based time series approach. Theory Biosci 2024; 143:183-193. [PMID: 38807013 DOI: 10.1007/s12064-024-00418-3] [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: 08/17/2023] [Accepted: 05/14/2024] [Indexed: 05/30/2024]
Abstract
Cervical cancer is one of the most severe threats to women worldwide and holds fourth rank in lethality. It is estimated that 604, 127 cervical cancer cases have been reported in 2020 globally. With advancements in high throughput technologies and bioinformatics, several cervical candidate genes have been proposed for better therapeutic strategies. In this paper, we intend to prioritize the candidate genes that are involved in cervical cancer progression through a fractal time series-based cross-correlations approach. we apply the chaos game representation theory combining a two-dimensional multifractal detrended cross-correlations approach among the known and candidate genes involved in cervical cancer progression to prioritize the candidate genes. We obtained 16 candidate genes that showed cross-correlation with known cancer genes. Functional enrichment analysis of the candidate genes shows that they involve GO terms: biological processes, cell-cell junction assembly, cell-cell junction organization, regulation of cell shape, cortical actin cytoskeleton organization, and actomyosin structure organization. KEGG pathway analysis revealed genes' role in Rap1 signaling pathway, ErbB signaling pathway, MAPK signaling pathway, PI3K-Akt signaling pathway, mTOR signaling pathway, Acute myeloid leukemia, chronic myeloid leukemia, Breast cancer, Thyroid cancer, Bladder cancer, and Gastric cancer. Further, we performed survival analysis and prioritized six genes CDH2, PAIP1, BRAF, EPB41L3, OSMR, and RUNX1 as potential candidate genes for cervical cancer that has a crucial role in tumor progression. We found that our study through this integrative approach an efficient tool and paved a new way to prioritize the candidate genes and these genes could be evaluated experimentally for potential validation. We suggest this may be useful in analyzing the nucleotide sequences and protein sequences for clustering, classification, class affiliation, etc.
Collapse
Affiliation(s)
- T Mallikarjuna
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Gachibowli, Hyderabad, 500046, India
| | - N B Thummadi
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Gachibowli, Hyderabad, 500046, India
| | - Vaibhav Vindal
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Gachibowli, Hyderabad, 500046, India
| | - P Manimaran
- School of Physics, University of Hyderabad, Gachibowli, Hyderabad, Telangana, 500046, India.
| |
Collapse
|
4
|
Kosasih HJ, Healey G, Brennan MS, Bjelosevic S, Sadras T, Jalud FB, Ibnat T, Ng AP, Mayoh C, Mao J, Tax G, Ludlow LEA, Johnstone RW, Herold MJ, Khaw SL, de Bock CE, Ekert PG. A novel MYB::PAIP1 oncogenic fusion in pediatric blastic plasmacytoid dendritic cell neoplasm (BPDCN) is dependent on BCL2 expression and is sensitive to venetoclax. Hemasphere 2024; 8:e1. [PMID: 38435422 PMCID: PMC10878182 DOI: 10.1002/hem3.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 12/16/2023] [Indexed: 03/05/2024] Open
Affiliation(s)
- Hansen J. Kosasih
- Murdoch Children's Research InstituteParkvilleVictoriaAustralia
- Children's Cancer Institute, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
| | - Gerry Healey
- The Walter and Eliza Hall Institute of Medical ResearchParkvilleVictoriaAustralia
- Olivia Newton‐John Cancer Research InstituteHeidelbergVictoriaAustralia
| | - Margs S. Brennan
- The Walter and Eliza Hall Institute of Medical ResearchParkvilleVictoriaAustralia
- Department of Medicine Huddinge, Centre for Haematology and Regenerative MedicineKarolinska InstitutetStockholmSweden
| | - Stefan Bjelosevic
- Peter MacCallum Cancer CentreMelbourneVictoriaAustralia
- The Sir Peter MacCallum Department of OncologyUniversity of MelbourneParkvilleVictoriaAustralia
| | - Teresa Sadras
- Peter MacCallum Cancer CentreMelbourneVictoriaAustralia
- The Sir Peter MacCallum Department of OncologyUniversity of MelbourneParkvilleVictoriaAustralia
| | | | - Tasnia Ibnat
- Peter MacCallum Cancer CentreMelbourneVictoriaAustralia
| | - Ashley P. Ng
- The Walter and Eliza Hall Institute of Medical ResearchParkvilleVictoriaAustralia
- The Sir Peter MacCallum Department of OncologyUniversity of MelbourneParkvilleVictoriaAustralia
- Department of BiologyThe University of MelbourneParkvilleVictoriaAustralia
| | - Chelsea Mayoh
- Children's Cancer Institute, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
| | - Jie Mao
- Children's Cancer Institute, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
| | - Gabor Tax
- Children's Cancer Institute, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
- School of Clinical Medicine, UNSW Medicine & HealthUNSW SydneySydneyNew South WalesAustralia
| | - Louise E. A. Ludlow
- Murdoch Children's Research InstituteParkvilleVictoriaAustralia
- Department of PaediatricsUniversity of MelbourneParkvilleVictoriaAustralia
| | - Ricky W. Johnstone
- Peter MacCallum Cancer CentreMelbourneVictoriaAustralia
- The Sir Peter MacCallum Department of OncologyUniversity of MelbourneParkvilleVictoriaAustralia
| | - Marco J. Herold
- The Walter and Eliza Hall Institute of Medical ResearchParkvilleVictoriaAustralia
- Olivia Newton‐John Cancer Research InstituteHeidelbergVictoriaAustralia
- Department of Medical BiologyUniversity of MelbourneParkvilleVictoriaAustralia
- School of Cancer MedicineLa Trobe UniversityHeidelbergVictoriaAustralia
| | - Seong L. Khaw
- Murdoch Children's Research InstituteParkvilleVictoriaAustralia
| | - Charles E. de Bock
- Children's Cancer Institute, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
- School of Women's and Children's HealthUNSW SydneyKensingtonNew South WalesAustralia
| | - Paul G. Ekert
- Children's Cancer Institute, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
- Peter MacCallum Cancer CentreMelbourneVictoriaAustralia
- The Sir Peter MacCallum Department of OncologyUniversity of MelbourneParkvilleVictoriaAustralia
- School of Clinical Medicine, UNSW Medicine & HealthUNSW SydneySydneyNew South WalesAustralia
| |
Collapse
|
5
|
Xue M, Cong F, Zheng W, Xu R, Liu X, Bao H, Sung YY, Xi Y, He F, Ma J, Yang X, Ge W. Loss of Paip1 causes translation reduction and induces apoptotic cell death through ISR activation and Xrp1. Cell Death Discov 2023; 9:288. [PMID: 37543696 PMCID: PMC10404277 DOI: 10.1038/s41420-023-01587-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 08/07/2023] Open
Abstract
Regulation of protein translation initiation is tightly associated with cell growth and survival. Here, we identify Paip1, the Drosophila homolog of the translation initiation factor PAIP1, and analyze its role during development. Through genetic analysis, we find that loss of Paip1 causes reduced protein translation and pupal lethality. Furthermore, tissue specific knockdown of Paip1 results in apoptotic cell death in the wing imaginal disc. Paip1 depletion leads to increased proteotoxic stress and activation of the integrated stress response (ISR) pathway. Mechanistically, we show that loss of Paip1 promotes phosphorylation of eIF2α via the kinase PERK, leading to apoptotic cell death. Moreover, Paip1 depletion upregulates the transcription factor gene Xrp1, which contributes to apoptotic cell death and eIF2α phosphorylation. We further show that loss of Paip1 leads to an increase in Xrp1 translation mediated by its 5'UTR. These findings uncover a novel mechanism that links translation impairment to tissue homeostasis and establish a role of ISR activation and Xrp1 in promoting cell death.
Collapse
Affiliation(s)
- Maoguang Xue
- Division of Human Reproduction and Developmental Genetics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
- Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Fei Cong
- Division of Human Reproduction and Developmental Genetics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
- Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Wanling Zheng
- Division of Human Reproduction and Developmental Genetics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
- Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Ruoqing Xu
- Division of Human Reproduction and Developmental Genetics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
- Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Xiaoyu Liu
- Division of Human Reproduction and Developmental Genetics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
- Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Hongcun Bao
- Division of Human Reproduction and Developmental Genetics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
- Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Ying Ying Sung
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | - Yongmei Xi
- Division of Human Reproduction and Developmental Genetics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
- Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Feng He
- Division of Human Reproduction and Developmental Genetics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
- Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Jun Ma
- Division of Human Reproduction and Developmental Genetics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China.
- Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China.
| | - Xiaohang Yang
- Division of Human Reproduction and Developmental Genetics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China.
- Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China.
| | - Wanzhong Ge
- Division of Human Reproduction and Developmental Genetics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China.
- Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China.
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China.
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
| |
Collapse
|
6
|
Zheng J, Fan W, Zhang X, Quan W, Wu Y, Shu M, Chen M, Liang M. PAIP1 regulates expression of immune and inflammatory response associated genes at transcript level in liver cancer cell. PeerJ 2023; 11:e15070. [PMID: 37101794 PMCID: PMC10124545 DOI: 10.7717/peerj.15070] [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: 10/28/2021] [Accepted: 02/23/2023] [Indexed: 04/28/2023] Open
Abstract
Poly(A) binding protein interacting protein 1 (PAIP1) is a translation regulator and also regulate the decay of mRNA. PAIP1 has also been reported to be a marker of increased invasive potential of liver cancer. However, the roles and underlying molecular mechanism of PAIP1 in liver cancer is still unclear. Here, cell viability and the gene expression profile of liver cancer line HepG2 transfected with PAIP1 siRNA was compared with cells transfected with non-targeting control siRNA. The results showed that PAIP1 knockdown inhibited cell viability, and extensively affects expression of 893 genes at transcriptional level in HepG2 cells. Gene function analysis showed that a large number of PAIP1 up-regulated genes were enriched in term of DNA-dependent transcription and the down-regulated genes were enriched in some pathways including immune response and inflammatory response. qPCR confirmed that PAIP1 knockdown positively regulated the expression of selected immune and inflammatory factor genes in HepG2 cells. Expression analysis of TCGA revealed that PAIP1 had positive correlations with two immune associated genes IL1R2 and PTAFR in liver tumor tissue. Taken together, our results demonstrated that PAIP1 was not only a translation regulator, but also a transcription regulator in liver cancer. Moreover, PAIP1 could function as a regulatory factor of immune and inflammatory genes in liver cancer. Thus, our study provides important cues for further study on the regulatory mechanism of PAIP1 in liver cancer.
Collapse
Affiliation(s)
- Jianfeng Zheng
- Department of Laboratory Medicine, Baoan Central Hospital of Shenzhen, The Fifth Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| | - Weiwei Fan
- Department of Infectious Medicine, Heilongjiang Provincial Hospital, Harbin, Heilongjiang, China
| | - Xiaoyu Zhang
- First Department of Infection, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Weili Quan
- Center for Genome Analysis, ABLife Inc., Wuhan, Hubei, China
- ABLife BioBigData Institute, Wuhan, Hubei, China
| | - Yunfei Wu
- Center for Genome Analysis, ABLife Inc., Wuhan, Hubei, China
| | - Mengni Shu
- First Department of Infection, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Moyang Chen
- First Department of Infection, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Ming Liang
- First Department of Infection, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
7
|
Zhang N, Chen X. PAIP1 is a novel oncogene in human hepatocellular carcinoma. Discov Oncol 2022; 13:132. [PMID: 36436074 PMCID: PMC9702235 DOI: 10.1007/s12672-022-00530-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 04/12/2022] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Poly(A)-binding protein interacting protein 1 (PAIP1) is a translational initiation regulatory factor that has been reported as oncogene in multiple malignant diseases. However, its role in hepatocellular carcinoma (HCC) and the potential mechanisms have not been explored. METHODS PAIP1 expression level in HCC cell lines were detected by real-time quantitative PCR and western blotting. The proliferation and colony formation of HCC cell lines were detected by MTT and colony formation assay. The apoptosis and cell cycle were detected by flow cytometry. The volume and growth rate of the xenograft tumors were observed. The potential mechanism of PAIP1 was analyzed by miRNA Microarray Analysis and TargetScan analysis. RESULTS PAIP1 is significantly upregulated in HCC cell lines. PAIP1 knockdown dramatically inhibits cell proliferation and colony formation, induces apoptosis and alters the cell cycle distribution by increasing the G2/M cell percentage. Moreover, PAIP1 knockdown significantly reduces tumorigenesis in a murine transplantation model. Bioinformatics and immunoblotting analysis reveal that PAIP1 knockdown dysregulates cyclin D pathway-related proteins. CONCLUSION PAIP1 plays an oncogenic role in hepatocellular carcinoma.
Collapse
Affiliation(s)
- Nuobei Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006, China
| | - Xin Chen
- Department of Nuclear Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China.
| |
Collapse
|
8
|
Circ_0005576 Exerts an Oncogenic Role in Cervical Cancer via miR-1305-Dependent Regulation of PAIP1. Reprod Sci 2022; 29:2647-2658. [PMID: 35378711 PMCID: PMC9444835 DOI: 10.1007/s43032-022-00925-y] [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] [Received: 11/29/2021] [Accepted: 03/16/2022] [Indexed: 11/05/2022]
Abstract
Cervical cancer (CC) is a leading cause of high morbidity and mortality in women worldwide. Circular RNAs (circRNAs) are considered to be essential regulators of various cancers, including CC. The purpose of this study was to investigate the role and mechanism of circ_0005576 in CC progression. The levels of circ_0005576, miR-1305, and poly(A)-binding protein-interacting protein 1 (PAIP1) were detected by quantitative real-time PCR (qRT-PCR) or western blot assay. The stability and location of circ_0005576 were determined by ribonuclease R (RNase R) assay and subcellular fractionation distribution assay, respectively. Cell proliferation was evaluated by CCK-8 assay, EDU incorporation assay, and colony formation assay. Cell migration and invasion were assessed by transwell assay. The interactions between miR-1305 and circ_0005576 or PAIP1 were validated by dual-luciferase reporter assay. The protein expression of cyclin D1, vimentin, and matrix metallopeptidase 9 (MMP9) was tested by western blot. Moreover, mice xenograft models were constructed to analyze tumor growth in vivo. Circ_0005576 and PAIP1 were upregulated, while miR-1305 was downregulated in CC tissues and cells. Circ_0005576 was a stable circRNA that was mainly distributed in the cytoplasm of cells. Knockdown of circ_0005576 suppressed the proliferation, migration, and invasion of CC cells, while the silence of miR-1305 facilitated the development of CC cells. Meanwhile, circ_0005576 could sponge miR-1305 to promote PAIP1 expression. Furthermore, PAIP1 overexpression relieved the influence of circ_0005576 silence on the growth of CC cells. Additionally, circ_0005576 silence hindered CC tumor growth in vivo. Circ_0005576 depletion suppressed tumor development in CC by regulating the miR-1305/PAIP1 axis, suggesting that circ_0005576 might be a potential biomarker for CC treatment.
Collapse
|
9
|
Effect of PAIP1 on the metastatic potential and prognostic significance in oral squamous cell carcinoma. Int J Oral Sci 2022; 14:9. [PMID: 35153296 PMCID: PMC8841500 DOI: 10.1038/s41368-022-00162-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 01/13/2022] [Indexed: 11/09/2022] Open
Abstract
AbstractPoly Adenylate Binding Protein Interacting protein 1 (PAIP1) plays a critical role in translation initiation and is associated with the several cancer types. However, its function and clinical significance have not yet been described in oral squamous cell carcinoma (OSCC) and its associated features like lymph node metastasis (LNM). Here, we used the data available from Gene Expression Omnibus (GEO), The Cancer Genome Atlas (TCGA), and Clinical Proteomic Tumor Analysis Consortium (CPTAC) to analyze PAIP1 expression in oral cancer. The publicly available data suggests that PAIP1 mRNA and protein levels were increased in OSCC. The high PAIP1 expression was more evident in samples with advanced stage, LNM, and worse pattern of invasion. Moreover, the in vitro experiments revealed that PAIP1 knockdown attenuated colony forming, the aggressiveness of OSCC cell lines, decreasing MMP9 activity and SRC phosphorylation. Importantly, we found a correlation between PAIP1 and pSRC through the analysis of the IHC scores and CPTAC data in patient samples. Our findings suggest that PAIP1 could be an independent prognostic factor in OSCC with LNM and a suitable therapeutic target to improve OSCC patient outcomes.
Collapse
|
10
|
Zarei Ghobadi M, Emamzadeh R. Integration of gene co-expression analysis and multi-class SVM specifies the functional players involved in determining the fate of HTLV-1 infection toward the development of cancer (ATLL) or neurological disorder (HAM/TSP). PLoS One 2022; 17:e0262739. [PMID: 35041720 PMCID: PMC8765610 DOI: 10.1371/journal.pone.0262739] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 01/04/2022] [Indexed: 11/19/2022] Open
Abstract
Human T-cell Leukemia Virus type-1 (HTLV-1) is an oncovirus that may cause two main life-threatening diseases including a cancer type named Adult T-cell Leukemia/Lymphoma (ATLL) and a neurological and immune disturbance known as HTLV-1 Associated Myelopathy/Tropical Spastic Paraparesis (HAM/TSP). However, a large number of the infected subjects remain as asymptomatic carriers (ACs). There is no comprehensive study that determines which dysregulated genes differentiate the pathogenesis routes toward ATLL or HAM/TSP. Therefore, two main algorithms including weighted gene co-expression analysis (WGCNA) and multi-class support vector machines (SVM) were utilized to find major gene players in each condition. WGCNA was used to find the highly co-regulated genes and multi-class SVM was employed to identify the most important classifier genes. The identified modules from WGCNA were validated in the external datasets. Furthermore, to find specific modules for ATLL and HAM/TSP, the non-preserved modules in another condition were found. In the next step, a model was constructed by multi-class SVM. The results revealed 467, 3249, and 716 classifiers for ACs, ATLL, and HAM/TSP, respectively. Eventually, the common genes between the WGCNA results and classifier genes resulted from multi-class SVM that also determined as differentially expressed genes, were identified. Through these step-wise analyses, PAIP1, BCAS2, COPS2, CTNNB1, FASLG, GTPBP1, HNRNPA1, RBBP6, TOP1, SLC9A1, JMY, PABPC3, and PBX1 were found as the possible critical genes involved in the progression of ATLL. Moreover, FBXO9, ZNF526, ERCC8, WDR5, and XRCC3 were identified as the conceivable major involved genes in the development of HAM/TSP. These genes can be proposed as specific biomarker candidates and therapeutic targets for each disease.
Collapse
Affiliation(s)
- Mohadeseh Zarei Ghobadi
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Rahman Emamzadeh
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
- * E-mail: ,
| |
Collapse
|
11
|
Huang Z, Liu Z, Cheng X, Han Z, Li J, Xia T, Gao Y, Wei L. Prognostic significance of HSF2BP in lung adenocarcinoma. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1559. [PMID: 34790765 PMCID: PMC8576644 DOI: 10.21037/atm-21-4935] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 10/13/2021] [Indexed: 02/01/2023]
Abstract
Background Recent studies have demonstrated that upregulation of heat shock transcription factor 2 binding protein (HSF2BP) may promote genomic instability, thereby leading to the development of tumors and also providing a potential target for biological antitumor therapy. However, the role of HSF2BP has so far remained unclear in lung adenocarcinoma (LUAD). Methods To explore the function of HSF2BP in LUAD, we collected transcriptome data for 551 lung samples from The Cancer Genome Atlas (TCGA) database and methylation data for 461 lung samples from the University of California Santa Cruz (UCSC) genome database, in addition to corresponding clinical information. We used bioinformatic approaches to systematically explore the role of HSF2BP in LUAD, including Gene Set Enrichment Analysis (GSEA), coexpression analysis, the Tumor IMmune Estimation Resource (TIMER) tool, Connectivity Map (CMap) analysis, and a meta-analysis involving three Gene Expression Omnibus (GEO) datasets and one TCGA dataset. Results Our results found that upregulation of HSF2BP in LUAD was an independent risk factor for the prognosis and diagnosis of LUAD. GSEA analysis showed HSF2BP expression was associated with vital signaling pathways, including the cell cycle, P53 signaling pathway, and homologous recombination. Coexpression analysis revealed 10 HSF2BP-associated genes, including oncogenes and tumor suppressor genes. Additionally, we found that HSF2BP expression was negatively correlated with B-cell infiltration and had a potential interaction with CD80 in LUAD, which may play an important role in tumor immune escape. Finally, we identified four small-molecule drugs which show promise for LUAD treatment. Conclusions The present study found that elevated HSF2BP posed a threat to prognosis in LUAD patients. HSF2BP might have been involved in tumorigenesis by influencing genomic stability and contributing to tumor immune evasion in the tumor immune microenvironment of LUAD. These findings suggest that HSF2BP may provide a vulnerable target for improving and enhancing treatment of LUAD.
Collapse
Affiliation(s)
- Zhendong Huang
- Department of Thoracic Surgery, Zhengzhou Key Laboratory for Surgical Treatment for End-Stage Lung Disease, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Zhendong Liu
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital, Henan International Joint Laboratory of Intelligentized Orthopedics Innovation and Transformation, Henan Key Laboratory for Intelligent Precision Orthopedics, Zhengzhou University People's Hospital, People's Hospital of Henan University, Zhengzhou, China
| | - Xingbo Cheng
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital, Henan International Joint Laboratory of Intelligentized Orthopedics Innovation and Transformation, Henan Key Laboratory for Intelligent Precision Orthopedics, Zhengzhou University People's Hospital, People's Hospital of Henan University, Zhengzhou, China
| | - Zhibin Han
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital, Henan International Joint Laboratory of Intelligentized Orthopedics Innovation and Transformation, Henan Key Laboratory for Intelligent Precision Orthopedics, Zhengzhou University People's Hospital, People's Hospital of Henan University, Zhengzhou, China
| | - Jiwei Li
- Department of Thoracic Surgery, Zhengzhou Key Laboratory for Surgical Treatment for End-Stage Lung Disease, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Tian Xia
- Department of Thoracic Surgery, Zhengzhou Key Laboratory for Surgical Treatment for End-Stage Lung Disease, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Yanzheng Gao
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital, Henan International Joint Laboratory of Intelligentized Orthopedics Innovation and Transformation, Henan Key Laboratory for Intelligent Precision Orthopedics, Zhengzhou University People's Hospital, People's Hospital of Henan University, Zhengzhou, China
| | - Li Wei
- Department of Thoracic Surgery, Zhengzhou Key Laboratory for Surgical Treatment for End-Stage Lung Disease, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| |
Collapse
|
12
|
Thummadi NB, T M, Vindal V, P M. Prioritizing the candidate genes related to cervical cancer using the moment of inertia tensor. Proteins 2021; 90:363-371. [PMID: 34468998 DOI: 10.1002/prot.26226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/07/2021] [Accepted: 08/16/2021] [Indexed: 12/24/2022]
Abstract
It is well known that cervical cancer poses the fourth most malignancy threat to women worldwide among all cancer types. There is a tremendous improvement in realizing the underlying molecular associations in cervical cancer. Several studies reported pieces of evidence for the involvement of various genes in the disease progression. However, with the ever-evolving bioinformatics tools, there has been an upsurge in predicting numerous genes responsible for cervical cancer progression and making it highly complex to target the genes for further evaluation. In this article, we prioritized the candidate genes based on the sequence similarity analysis with known cancer genes. For this purpose, we used the concept of the moment of inertia tensor, which reveals the similarities between the protein sequences more efficiently. Tensor for moment of inertia explores the similarity of the protein sequences based on the physicochemical properties of amino acids. From our analysis, we obtained 14 candidate cervical cancer genes, which are highly similar to known cervical cancer genes. Further, we analyzed the GO terms and prioritized these genes based on the number of hits with biological process, molecular functions, and their involvement in KEGG pathways. We also discussed the evidence-based involvement of the prioritized genes in other cancers and listed the available drugs for those genes.
Collapse
Affiliation(s)
- Neelesh Babu Thummadi
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Gachibowli, Hyderabad, India
| | - Mallikarjuna T
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Gachibowli, Hyderabad, India
| | - Vaibhav Vindal
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Gachibowli, Hyderabad, India
| | - Manimaran P
- School of Physics, University of Hyderabad, Gachibowli, Hyderabad, India
| |
Collapse
|
13
|
Kim H, Jung W, Kim A, Kim HK, Kim BH. High Paip1 Expression as a Potential Prognostic Marker in Hepatocellular Carcinoma. In Vivo 2021; 34:2491-2497. [PMID: 32871777 DOI: 10.21873/invivo.12065] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 07/12/2020] [Accepted: 07/13/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND/AIM Translation plays an important role in the carcinogenesis of various human tumors. Paip1 and eIF4A1 are translation-associated proteins that mediate the function of eukaryotic initiation factor 4F complex. This study aimed to analyse the relationship between the expression status of Paip1 and eIF4A1 and clinicopathologic features in hepatocellular carcinoma (HCC). MATERIALS AND METHODS Immunohistochemical analysis was used to evaluate the expression status of Paip1 and eIF4A1. Two pathologists independently interpreted the immunostained slides. The prognostic value of Paip1 and eIF4A1 was evaluated by the Kaplan-Meier plotter. RESULTS Among 173 HCC patients, 28 (16.1%) and 46 (26.6%) belonged in the Paip1 and eIF4A1 high-expression groups. High expression of Paip1 and eIF4A1 was associated with advanced TNM stage and more frequent vascular tumor invasion. Univariate analysis indicated that high Paip1 expression was associated with worse five-year overall survival (OS). Public dataset analysis by Kaplan-Meier plotter revealed that high mRNA expression of Paip1, and not of eIF4A1, was significantly associated with worse five-year OS and disease-free survival. CONCLUSION Paip1 expression has a potential prognostic value in human HCC.
Collapse
Affiliation(s)
- Hayeon Kim
- Department of Pathology, Korea University Guro Hospital, Seoul, Republic of Korea
| | - Wonkyung Jung
- Department of Pathology, Sure Quest Lab, Yongin, Republic of Korea
| | - Aeree Kim
- Department of Pathology, Korea University Guro Hospital, Seoul, Republic of Korea
| | - Han Kyeom Kim
- Department of Pathology, Korea University Guro Hospital, Seoul, Republic of Korea
| | - Baek-Hui Kim
- Department of Pathology, Korea University Guro Hospital, Seoul, Republic of Korea
| |
Collapse
|
14
|
Bi J, Ma H, Liu Y, Huang A, Xiao Y, Shu WJ, Du H, Zhang T. Upregulation of PAIP1 promotes the gallbladder tumorigenesis through regulating PLK1 level. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:991. [PMID: 34277791 PMCID: PMC8267329 DOI: 10.21037/atm-21-2417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/15/2021] [Indexed: 12/17/2022]
Abstract
Background Increasing evidence suggests that elevated expression of polyA-binding protein-interacting protein 1 (PAIP1) is associated with cancer development and progression. However, how PAIP1 promotes gallbladder cancer (GBC) is still unclear. Methods Two GBC tissue-derived cell lines, NOZ and GBC-SD cells, were used in this study. Assays of cell proliferation, colony formation, apoptosis, and xenograft tumor model were performed to examine the tumorigenic effects of PAIP1. Immunohistochemical (IHC) staining was used to examine the expression level of PAIP1 in both patient GBC tissues and mouse tumors. Microarray and bioinformatics analysis were used to explore the targets of PAIP1. Quantitative polymerase chain reaction (qPCR) and western blot analysis were used to validate PAIP1-mediated targets. Results We found that upregulated PAIP1 expression was correlated with GBC. Knockdown of PAIP1 in gallbladder cells alleviated cell proliferation, promoted apoptosis, and inhibited xenograft tumor growth. Gene microarray analysis showed that stable silencing of PAIP1 altered various gene expressions. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis suggested that PAIP1 regulates cell cycle progression. Finally, we found that the PLK1 kinase, a key regulator of cell cycle, was regulated by PAIP1 at the transcriptional and protein levels. PLK1 level was positively correlated with PAIP1 level in both mouse tumors and GBC tissues. PAIP1 interacted with PLK1, and rescue of PAIP1 could recover PLK1 protein level and inhibit apoptosis. Conclusions Our data suggest that PAIP1 contributes to GBC progression likely through regulating PLK1 level. Since upregulated PAIP1 expression is positively associated with GBC, PAIP1 may act as a clinical prognostic biomarker of GBC.
Collapse
Affiliation(s)
- Jianping Bi
- Hubei Key Laboratory of Cell Homeostasis, RNA Institute, College of Life Sciences, Wuhan University, Wuhan, China
| | - Hong Ma
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yafei Liu
- Hubei Key Laboratory of Cell Homeostasis, RNA Institute, College of Life Sciences, Wuhan University, Wuhan, China
| | - Ai Huang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Xiao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wen-Jie Shu
- Hubei Key Laboratory of Cell Homeostasis, RNA Institute, College of Life Sciences, Wuhan University, Wuhan, China
| | - Haining Du
- Hubei Key Laboratory of Cell Homeostasis, RNA Institute, College of Life Sciences, Wuhan University, Wuhan, China
| | - Tao Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
15
|
Wang Q, Li D, Guo A, Li M, Li L, Zhou J, Mishra SK, Li G, Duan Y, Li Q. Whole-genome resequencing of Dulong Chicken reveal signatures of selection. Br Poult Sci 2020; 61:624-631. [PMID: 32627575 DOI: 10.1080/00071668.2020.1792832] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
1. Dulong Chickens (DLCs) live at high altitude (~3000 m) and humidity (~90%), are endemic to the Yunnan province, and have gradually developed unique physiological characteristics, but their genetic basis is still unclear. Using the fixation index (FST ) approach, based on whole-genome resequencing, DLCs were analysed to uncover the genomic architecture of the population and candidate genes involved in selection during domestication. 2. A total of 469 candidate genes were obtained to be putatively under selection in DLCs. Further investigations revealed the genic footprint for local adaptation (high-altitude and high-humidity) as the genic signatures that are involved in economic traits (related to egg production). 3. Candidate genes were identified that may be associated with disease resistance, aggressiveness, small body size and positive selection of vision in DLCs. 4. These data revealed loci of selective signals that operate during selection for production at high altitude and humidity.
Collapse
Affiliation(s)
- Q Wang
- Key Laboratory for Forest Resources Conservation and Utilization in the Southwest Mountains of China (Southwest Forestry University), Ministry of Education , Kunming, China.,Life Science College, Southwest Forestry University , Kunming, China
| | - D Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University , Chengdu, China
| | - A Guo
- Key Laboratory for Forest Resources Conservation and Utilization in the Southwest Mountains of China (Southwest Forestry University), Ministry of Education , Kunming, China.,Life Science College, Southwest Forestry University , Kunming, China
| | - M Li
- School of Mathematics and Computer Science, Yunnan Nationalities University , Kunming, China
| | - L Li
- Life Science College, Southwest Forestry University , Kunming, China
| | - J Zhou
- Life Science College, Southwest Forestry University , Kunming, China
| | - S K Mishra
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University , Chengdu, China
| | - G Li
- Key Laboratory for Forest Resources Conservation and Utilization in the Southwest Mountains of China (Southwest Forestry University), Ministry of Education , Kunming, China.,Life Science College, Southwest Forestry University , Kunming, China
| | - Y Duan
- Technology Center, China Tobacco Yunnan Industrial Co., Ltd ., Kunming, China
| | - Q Li
- Key Laboratory for Forest Resources Conservation and Utilization in the Southwest Mountains of China (Southwest Forestry University), Ministry of Education , Kunming, China.,Life Science College, Southwest Forestry University , Kunming, China.,Kunming Xianghao Technology Co. Ltd ., Kunming, China
| |
Collapse
|
16
|
Xie H, Yang L, Hu Q, Song Y, Wang X, Zhou L, Li L. Effects of inducing apoptosis and inhibiting proliferation of siRNA on polyadenylate-binding protein-interacting protein 1 in tongue cell carcinoma. Head Neck 2020; 42:3623-3637. [PMID: 32827170 DOI: 10.1002/hed.26423] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 05/15/2020] [Accepted: 08/03/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND It has been reported that the polyadenylate-binding protein-interacting protein 1 (PAIP1) pathway is closely connected with the progression of some malignant tumors. Here we examined the potential functional mechanism of PAIP1 in tongue squamous cell carcinoma (TSCC). METHODS PAIP1 was knocked down in TSCC cell lines and proliferation and apoptosis in vitro analyzed. The molecular features of TSCC were determined using quantitative proteome and succinylome analyses. The results were confirmed in the mouse model. RESULTS PAIP1 promoted cell proliferation and inhibited apoptosis. Its knockdown decreased Ki67 and Pcna expressions and increased Bax/Bcl2 index and Caspase-3 expression. Bioinformatics analysis for proteomics revealed that PAIP1 knockdown correlated with the changes in differential protein expression. CONCLUSIONS Upregulation of PAIP1 induces cell proliferation and inhibits apoptosis in TSCC; PAIP1 might be a diagnostic biomarker and a significant drug target.
Collapse
Affiliation(s)
- Huixu Xie
- State Key Laboratory of Oral Diseases, Department of Head and Neck Oncology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lisa Yang
- State Key Laboratory of Oral Diseases, Department of Head and Neck Oncology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Sun Yat-sen University, Guangzhou, China
| | - Qin Hu
- State Key Laboratory of Oral Diseases, Department of Head and Neck Oncology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yingqi Song
- Department of Pharmacology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Xiaoyi Wang
- State Key Laboratory of Oral Diseases, Department of Head and Neck Oncology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Liming Zhou
- Department of Pharmacology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Longjiang Li
- State Key Laboratory of Oral Diseases, Department of Head and Neck Oncology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
17
|
Lian S, Li L, Zhou Y, Liu Z, Wang L. The co-expression networks of differentially expressed RBPs with TFs and LncRNAs related to clinical TNM stages of cancers. PeerJ 2019; 7:e7696. [PMID: 31576243 PMCID: PMC6753928 DOI: 10.7717/peerj.7696] [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: 05/22/2019] [Accepted: 08/19/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND RNA-binding proteins (RBPs) play important roles in cellular homeostasis by regulating the expression of thousands of transcripts, which have been reported to be involved in human tumorigenesis. Despite previous reports of the dysregulation of RBPs in cancers, the degree of dysregulation of RBPs in cancers and the intrinsic relevance between dysregulated RBPs and clinical TNM information remains unknown. Furthermore, the co-expressed networks of dysregulated RBPs with transcriptional factors and lncRNAs also require further investigation. RESULTS Here, we firstly analyzed the deviations of expression levels of 1,542 RBPs from 20 cancer types and found that (1) RBPs are dysregulated in almost all 20 cancer types, especially in BLCA, COAD, READ, STAD, LUAD, LUSC and GBM with proportion of deviation larger than 300% compared with non-RBPs in normal tissues. (2) Up- and down-regulated RBPs also show opposed patterns of differential expression in cancers and normal tissues. In addition, down-regulated RBPs show a greater degree of dysregulated expression than up-regulated RBPs do. Secondly, we analyzed the intrinsic relevance between dysregulated RBPs and clinical TNM information and found that (3) Clinical TNM information for two cancer types-CHOL and KICH-is shown to be closely related to patterns of differentially expressed RBPs (DE RBPs) by co-expression cluster analysis. Thirdly, we identified ten key RBPs (seven down-regulated and three up-regulated) in CHOL and seven key RBPs (five down-regulated and two up-regulated) in KICH by analyzing co-expression correlation networks. Fourthly, we constructed the co-expression networks of key RBPs between 1,570 TFs and 4,147 lncRNAs for CHOL and KICH, respectively. CONCLUSIONS These results may provide an insight into the understanding of the functions of RBPs in human carcinogenesis. Furthermore, key RBPs and the co-expressed networks offer useful information for potential prognostic biomarkers and therapeutic targets for patients with cancers at the N and M stages in two cancer types CHOL and KICH.
Collapse
Affiliation(s)
- Shuaibin Lian
- College of Physics and Electronic Engineering, XinYang Normal University, Xinyang, HeNan, China
| | - Liansheng Li
- College of Life Sciences, XinYang Normal University, Xinyang, HeNan, China
| | - Yongjie Zhou
- College of Physics and Electronic Engineering, XinYang Normal University, Xinyang, HeNan, China
| | - Zixiao Liu
- College of Physics and Electronic Engineering, XinYang Normal University, Xinyang, HeNan, China
| | - Lei Wang
- College of Life Sciences, XinYang Normal University, Xinyang, HeNan, China
| |
Collapse
|
18
|
Wang Q, Han A, Chen L, Sun J, Lin Z, Zhang X, Ren X. Paip1 overexpression is involved in the progression of gastric cancer and predicts shorter survival of diagnosed patients. Onco Targets Ther 2019; 12:6565-6576. [PMID: 31496746 PMCID: PMC6701649 DOI: 10.2147/ott.s202698] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 07/10/2019] [Indexed: 01/06/2023] Open
Abstract
Background Gastric cancer (GC) is a major leading cause of cancer mortality worldwide. Polyadenylate (poly(A))-binding protein (PABP)-interacting protein 1 (Paip1) is a key regulator in the initiation of translation; however, its role in GC remains to be investigated. Purpose The purpose of this study is to determine Paip1 expression levels and investigate its underlying molecular mechanism in GC. Patients and methods In the present study, a total of 90 GC samples and 90 adjacent noncancerous tissues were used to examine the expression of Paip1. In order to gain a deep insight into the molecular mechanism of Paip1 in GC, the Paip1 siRNA sequences were transfected into GC cell lines (MGC-803 and SGC-7901), respectively. Meanwhile, Paip1 plasmid was used to mediate overexpression of Paip1. Cell proliferation were examined via colony formation assay, EdU assay and flow cytometry assay. Cell metastasis were discovered via wound healing assay and Transwell assays. In addition, key EMT makers were detected by Western blotting assay. Results In this study, Paip1 expression was observed to be upregulated in GC and was associated with shorter overall survival. Knockdown of Paip1 inhibited cell proliferation, migration and caused cell cycle arrest in GC cells, whereas its overexpression reversed these effects. Another mechanistic study showed that Paip1 overexpression promoted EMT progression and regulated its targets expression. Conclusion High expression of Paip1 plays a significant role in the progression of GC and may be a potential biomarker of poor prognosis as well as a therapeutic target.
Collapse
Affiliation(s)
- Qianrong Wang
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Yanji 133002, People's Republic of China
| | - Anna Han
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Yanji 133002, People's Republic of China
| | - Liyan Chen
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Yanji 133002, People's Republic of China.,Key Laboratory of the Science and Technology Department of Jilin Province, Yanji 133002, People's Republic of China
| | - Jie Sun
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Yanji 133002, People's Republic of China
| | - Zhenhua Lin
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Yanji 133002, People's Republic of China.,Key Laboratory of the Science and Technology Department of Jilin Province, Yanji 133002, People's Republic of China
| | - Xianglan Zhang
- Department of Pathology, Yanbian University Hospital, Yanji City, Jilin Province, People's Republic of China.,Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, Korea
| | - Xiangshan Ren
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Yanji 133002, People's Republic of China.,Key Laboratory of the Science and Technology Department of Jilin Province, Yanji 133002, People's Republic of China
| |
Collapse
|
19
|
Xu W, Wang Y, Wang Y, Lv S, Xu X, Dong X. Screening of differentially expressed genes and identification of NUF2 as a prognostic marker in breast cancer. Int J Mol Med 2019; 44:390-404. [PMID: 31198978 PMCID: PMC6605639 DOI: 10.3892/ijmm.2019.4239] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 05/29/2019] [Indexed: 12/24/2022] Open
Abstract
The aims of the present study were to screen differentially expressed genes (DEGs) in breast cancer (BC) and investigate NDC80 kinetochore complex component (NUF2) as a prognostic marker of BC in detail. A total of four BC microarray datasets, downloaded from the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) databases, were used to screen DEGs. A total of 190 DEGs with the same expression trends were identified in the 4 datasets, including 65 upregulated and 125 downregulated DEGs. Functional and pathway enrichment analyses were performed using the Database for Annotation, Visualization and Integrated Discovery. The upregulated DEGs were enriched for 10 Gene Ontology (GO) terms and 7 pathways, and the downregulated DEGs were enriched for 10 GO terms and 10 pathways. A protein‑protein interaction network containing 149 nodes and 930 edges was constructed using the Search Tool for the Retrieval of Interacting Genes, and 2 functional modules were identified using the MCODE plugin of Cytoscape. Based on an in‑depth analysis of module 1 and literature mining, NUF2 was selected for further research. Oncomine database analysis and reverse transcription‑quantitative PCR showed that NUF2 is significantly upregulated in BC tissues. In analyses of correlations between NUF2 and clinical pathological characteristics, NUF2 was significantly associated with the malignant features of BC. Using 5 additional datasets from GEO, it was demonstrated that NUF2 has a significant prognostic role in both ER‑positive and ER‑negative BC. A Gene Set Enrichment Analysis indicated that NUF2 may regulate breast carcinogenesis and progression via cell cycle‑related pathways. The results of the present study demonstrated that NUF2 is overexpressed in BC and is significantly associated with its multiple pathological features and prognosis.
Collapse
Affiliation(s)
- Wenjie Xu
- Department of Clinical Laboratory Center, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, P.R. China
| | - Yizhen Wang
- Department of Clinical Laboratory Center, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, P.R. China
| | - Yanan Wang
- Department of Clinical Laboratory Center, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, P.R. China
| | - Shanmei Lv
- Department of Clinical Laboratory Center, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, P.R. China
| | - Xiuping Xu
- Department of Clinical Laboratory Center, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, P.R. China
| | - Xuejun Dong
- Department of Clinical Laboratory Center, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, P.R. China
| |
Collapse
|
20
|
Li N, Piao J, Wang X, Kim KY, Bae JY, Ren X, Lin Z. Paip1 Indicated Poor Prognosis in Cervical Cancer and Promoted Cervical Carcinogenesis. Cancer Res Treat 2019; 51:1653-1665. [PMID: 31010277 PMCID: PMC6790838 DOI: 10.4143/crt.2018.544] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 04/17/2019] [Indexed: 12/30/2022] Open
Abstract
Purpose This study was aimed to investigate the role of poly(A)-binding protein-interacting protein 1 (Paip1) in cervical carcinogenesis. Materials and Methods The expression of Paip1 in normal cervical epithelial tissues and cervical cancer (CC) tissues were detected by immunohistochemistry. In vivo and in vitro assays were performed to validate effect of Paip1 on CC progression. Results Paip1 was found to be up-regulated in CC, which was linked with shorter survival. Knockdown of Paip1 inhibited cell growth, induced apoptosis and cell cycle arrest in CC cells, whereas its overexpression reversed these effects. The in vivo tumor model confirmed the pro-tumor role of Paip1 in CC growth. Conclusion Altogether, the investigation demonstrated the clinical significance of Paip1 expression, which prompted that the up-regulated of Paip1 can presumably be a potential prognostic and progression marker for CC.
Collapse
Affiliation(s)
- Nan Li
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, China.,Key Laboratory of the Science and Technology, Department of Jilin Province, Yanji, China
| | - Junjie Piao
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, China.,Key Laboratory of the Science and Technology, Department of Jilin Province, Yanji, China
| | - Xinyue Wang
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, China.,Key Laboratory of the Science and Technology, Department of Jilin Province, Yanji, China
| | - Ki-Yeol Kim
- Brain Korea 21 Project, Yonsei University College of Dentistry, Seoul, Korea
| | - Jung Yoon Bae
- Department of Oral Pathology, Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, Korea
| | - Xiangshan Ren
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, China.,Key Laboratory of the Science and Technology, Department of Jilin Province, Yanji, China
| | - Zhenhua Lin
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, China.,Key Laboratory of the Science and Technology, Department of Jilin Province, Yanji, China
| |
Collapse
|
21
|
Role of Paip1 on angiogenesis and invasion in pancreatic cancer. Exp Cell Res 2019; 376:198-209. [DOI: 10.1016/j.yexcr.2019.01.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 01/11/2019] [Accepted: 01/28/2019] [Indexed: 12/11/2022]
|
22
|
Wang Y, Piao J, Wang Q, Cui X, Meng Z, Jin T, Lin Z. Paip1 predicts poor prognosis and promotes tumor progression through AKT/GSK-3β pathway in lung adenocarcinoma. Hum Pathol 2018; 86:233-242. [PMID: 30496797 DOI: 10.1016/j.humpath.2018.11.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 11/12/2018] [Accepted: 11/16/2018] [Indexed: 12/19/2022]
Abstract
The expression and biological function of Paip1 remain poorly understood in most human cancers. The objective of this research is to investigate its clinical significance and roles in lung adenocarcinoma (LADC). Immunohistochemistry was used to determine Paip1 expression in 58 cases of LADC patients with strict follow-up and 60 cases of adjacent normal lung tissues. Paip1 protein was upregulated in 77.6% (45/58) LADC tissues compared with adjacent normal lung tissues. The overexpression of Paip1 was significantly correlated with histologic grade, clinical stage, and poor prognosis. Small interfering RNA-mediated transfection was performed in A549 and H1299 cells. Paip1 depletion attenuated the proliferation and migration of A549 and H1299 cells. Paip1 also changed the expression of epithelial-to-mesenchymal transition markers including E-cadherin, Vimentin, Slug, and Snail. Furthermore, Paip1 regulated AKT/GSK-3β oncogenic signaling pathways. In conclusions, Paip1 expression is frequently upregulated in LADC, and its overexpression correlates with poor prognosis in LADC patients. Attenuated Paip1 expression suppresses proliferation and epithelial-to-mesenchymal transition-related migration of A549 and H1299 cells by regulating the AKT/GSK-3β signaling pathway.
Collapse
Affiliation(s)
- Yixuan Wang
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Yanji 133002, China; Key Laboratory of the Science and Technology Department of Jilin Province, Yanji 133002, China
| | - Junjie Piao
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Yanji 133002, China; Key Laboratory of the Science and Technology Department of Jilin Province, Yanji 133002, China
| | - Qianrong Wang
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Yanji 133002, China; Key Laboratory of the Science and Technology Department of Jilin Province, Yanji 133002, China
| | - Xuelian Cui
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Yanji 133002, China; Key Laboratory of the Science and Technology Department of Jilin Province, Yanji 133002, China
| | - Ziqi Meng
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Yanji 133002, China; Key Laboratory of the Science and Technology Department of Jilin Province, Yanji 133002, China
| | - Tiefeng Jin
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Yanji 133002, China; Key Laboratory of the Science and Technology Department of Jilin Province, Yanji 133002, China.
| | - Zhenhua Lin
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Yanji 133002, China; Key Laboratory of the Science and Technology Department of Jilin Province, Yanji 133002, China.
| |
Collapse
|