1
|
Wang J, Ruan GX, Li Y, Xiao X, Zhu Z, Chen W, Huang H, Zhang R, Wang R, Chen M, Guo L, Li Y, Xu S, Ou X. Minor Splicing Factor RNPC3 Is Essential for the Germinal Center B Cell Response. Eur J Immunol 2025; 55:e202451508. [PMID: 40170400 DOI: 10.1002/eji.202451508] [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: 09/09/2024] [Revised: 03/07/2025] [Accepted: 03/18/2025] [Indexed: 04/03/2025]
Abstract
Germinal center (GC) response ensures the generation of diverse and high-affinity antibodies during the T cell-dependent (TD) immune response. This process is controlled by coordinated transcriptional and posttranscriptional gene regulatory mechanisms. Minor intron splicing is known to be involved in posttranscriptional regulation of gene expression. RNA-binding region (RNP1, RRM) containing 3 (RNPC3) is a minor spliceosome component involved in stabilizing the U11/U12 di-snRNP complex, which is essential for minor intron splicing. However, it remains unclear if RNPC3 and RNPC3-related gene regulatory mechanisms are important for the TD immune response. In this study, we conditionally ablated RNPC3 in activated B cells and showed that the mutant mice had defective antibody generation due to impaired GC B cell response. We demonstrate that RNPC3 deficiency inhibits the proliferation and promotes the apoptosis of activated B cells. Mechanistically, we show that RNPC3 regulates the development of GC B cells in a minor spliceosome-dependent manner by controlling the removal of minor introns from minor intron-containing genes associated with cell proliferation and apoptosis. Our study thus uncovers a previously unappreciated role for RNPC3 in regulating GC B cell response.
Collapse
Affiliation(s)
- Jing Wang
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Gui-Xin Ruan
- Department of Basic Medicine, School of Medicine, Taizhou University, Taizhou, China
| | - Yuxing Li
- School of Biological and Pharmaceutical Engineering, Lanzhou Jiaotong University, Lanzhou, China
| | - Xiong Xiao
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Zhijian Zhu
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Wenjing Chen
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Hengjun Huang
- Jiangxi Province Key Laboratory of Traditional Chinese Medicine Pharmacology, Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, Nanchang, China
- Jiangxi Health Industry Institute of Traditional Chinese Medicine, Nanchang, China
| | - Rui Zhang
- School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Ruisi Wang
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Meiyuan Chen
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Ling Guo
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Yan Li
- Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Shengli Xu
- Singapore Immunology Network (SIgN), Agency for Science, Technology, and Research (A*STAR), Singapore, Republic of Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Xijun Ou
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
2
|
Pan Y, Zeng W, Liang T, Nie X, Liu K, Chen H, Luo N, Zhu X, Tian K, Chen Y. RNF38 promotes gilteritinib resistance in acute myeloid leukemia via inducing autophagy by regulating ubiquitination of LMX1A. Cell Biol Toxicol 2024; 40:105. [PMID: 39604755 PMCID: PMC11602842 DOI: 10.1007/s10565-024-09936-8] [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: 07/01/2024] [Accepted: 10/16/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND Gilteritinib is a commonly used targeted drug for acute myeloid leukemia (AML), but the emergence of gilteritinib resistance greatly reduces the therapeutic effect. RING finger protein 38 (RNF38), a protein with RING Finger domain and E3 ubiquitin ligase activity, has been implicated in tumorigenesis and drug resistance. However, the role and mechanism of RNF38 in the gilteritinib resistance of AML remains unclear. METHODS Normal AML cells were treated with gilteritinib to construct gilteritinib-resistant cells (MV4-11/Gilteritinib and MOLM-13/Gilteritinib). CCK8 assay, TUNEL staining and EdU assay were used to assess gilteritinib resistance, cell apoptosis and proliferation. The protein levels of autophagy-related markers, RNF38 and LIM homeobox transcription factor 1 alpha (LMX1A) were determined by western blot. Also, RNF38 and LMX1A mRNA levels were tested using qRT-PCR. Autophagic flux was assessed using mRFP-GFP-LC3 labeling, and autophagosome numbers was counted under transmission electron microscopy. Co-IP assay was employed to analyze interaction between RNF38 and LMX1A. The effects of LMX1A and RNF38 on AML tumorigenesis were analyzed by in vivo experiments. RESULTS In gilteritinib-resistant AML cells, autophagy-related markers, mRFP-GFP-LC3 signals and autophagosome numbers were significantly enhanced. Autophagy inhibitor 3-MA could suppress gilteritinib resistance in AML cells. RNF38 knockdown inhibited gilteritinib resistance and autophagy in AML cells. Mechanistically, RNF38 reduced LMX1A expression by inducing its ubiquitination. RNF38 overexpression reversed the inhibitory effect of LMX1A on gilteritinib resistance and autophagy in AML cells, as well as AML tumor growth in vivo, while these effects could be abolished by proteasome inhibitor MG132. CONCLUSIONS RNF38 induced autophagy to promote gilteritinib resistance in AML by increasing the ubiquitination of LMX1A.
Collapse
MESH Headings
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Autophagy/drug effects
- Humans
- Drug Resistance, Neoplasm/drug effects
- Animals
- Cell Line, Tumor
- Ubiquitination/drug effects
- Transcription Factors/metabolism
- Transcription Factors/genetics
- Mice
- Pyrazines/pharmacology
- Aniline Compounds/pharmacology
- Apoptosis/drug effects
- Cell Proliferation/drug effects
- Ubiquitin-Protein Ligases/metabolism
- Ubiquitin-Protein Ligases/genetics
- Xenograft Model Antitumor Assays
- Mice, Nude
Collapse
Grants
- 2022B967, 2021B363 Science and Technology Program of Jiangxi Provincial Administration of Traditional Chinese Medicine
- 2022B967, 2021B363 Science and Technology Program of Jiangxi Provincial Administration of Traditional Chinese Medicine
- 2022B967, 2021B363 Science and Technology Program of Jiangxi Provincial Administration of Traditional Chinese Medicine
- 2022B967, 2021B363 Science and Technology Program of Jiangxi Provincial Administration of Traditional Chinese Medicine
- 2022B967, 2021B363 Science and Technology Program of Jiangxi Provincial Administration of Traditional Chinese Medicine
- 2022B967, 2021B363 Science and Technology Program of Jiangxi Provincial Administration of Traditional Chinese Medicine
- 2022B967, 2021B363 Science and Technology Program of Jiangxi Provincial Administration of Traditional Chinese Medicine
- 2022B967, 2021B363 Science and Technology Program of Jiangxi Provincial Administration of Traditional Chinese Medicine
- 2022B967, 2021B363 Science and Technology Program of Jiangxi Provincial Administration of Traditional Chinese Medicine
- 2022B967, 2021B363 Science and Technology Program of Jiangxi Provincial Administration of Traditional Chinese Medicine
- 202212543, 202410096 Science and Technology Plan Project of Jiangxi Provincial Health Care Commission
- 202212543, 202410096 Science and Technology Plan Project of Jiangxi Provincial Health Care Commission
- 202212543, 202410096 Science and Technology Plan Project of Jiangxi Provincial Health Care Commission
- 202212543, 202410096 Science and Technology Plan Project of Jiangxi Provincial Health Care Commission
- 202212543, 202410096 Science and Technology Plan Project of Jiangxi Provincial Health Care Commission
- 202212543, 202410096 Science and Technology Plan Project of Jiangxi Provincial Health Care Commission
- 202212543, 202410096 Science and Technology Plan Project of Jiangxi Provincial Health Care Commission
- 202212543, 202410096 Science and Technology Plan Project of Jiangxi Provincial Health Care Commission
- 202212543, 202410096 Science and Technology Plan Project of Jiangxi Provincial Health Care Commission
- 202212543, 202410096 Science and Technology Plan Project of Jiangxi Provincial Health Care Commission
- GZWJW202402160 Science and Technology Plan Project of Ganzhou Health Care Commission
- GZWJW202402160 Science and Technology Plan Project of Ganzhou Health Care Commission
- GZWJW202402160 Science and Technology Plan Project of Ganzhou Health Care Commission
- GZWJW202402160 Science and Technology Plan Project of Ganzhou Health Care Commission
- GZWJW202402160 Science and Technology Plan Project of Ganzhou Health Care Commission
- GZWJW202402160 Science and Technology Plan Project of Ganzhou Health Care Commission
- GZWJW202402160 Science and Technology Plan Project of Ganzhou Health Care Commission
- GZWJW202402160 Science and Technology Plan Project of Ganzhou Health Care Commission
- GZWJW202402160 Science and Technology Plan Project of Ganzhou Health Care Commission
- GZWJW202402160 Science and Technology Plan Project of Ganzhou Health Care Commission
Collapse
Affiliation(s)
- Yiyun Pan
- Department of Oncology, Ganzhou Cancer Hospital, Gannan Medical University, Ganzhou City, 341000, Jiangxi Province, P.R. China
- Suzhou Medical College of Soochow University, Suzhou City, 215123, Jiangsu Province, P.R. China
- The Endemic Disease (Thalassemia) Clinical Research Center of Jiangxi Province, Ganzhou City, 341000, Jiangxi Province, P.R. China
| | - Wen Zeng
- Department of Surgical Oncology, Ganzhou Cancer Hospital, Gannan Medical University, Ganzhou City, 341000, Jiangxi Province, P.R. China
| | - Ting Liang
- Department of Pathology, Ganzhou Cancer Hospital, Gannan Medical University, Ganzhou City, 341000, Jiangxi Province, P.R. China
| | - Xiaoming Nie
- Department of Surgical Oncology, Ganzhou Cancer Hospital, Gannan Medical University, Ganzhou City, 341000, Jiangxi Province, P.R. China
| | - Kang Liu
- Department of Thoracic Surgery, Fuyang Sixth People's Hospital, Fuyang City, 236000, Anhui Province, P.R. China
| | - Hailong Chen
- Department of Oncology, Ganzhou Cancer Hospital, Gannan Medical University, Ganzhou City, 341000, Jiangxi Province, P.R. China
| | - Nengping Luo
- Department of Pharmacy, Ganzhou Cancer Hospital, Gannan Medical University, Ganzhou City, 341000, Jiangxi Province, P.R. China
| | - Xiaodan Zhu
- Department of Hematology, The First Affiliated Hospital of Gannan Medical University, Ganzhou City, 341000, Jiangxi Province, P.R. China
| | - Keqiang Tian
- Department of Surgical Oncology, Ganzhou Cancer Hospital, Gannan Medical University, Ganzhou City, 341000, Jiangxi Province, P.R. China.
| | - Yijian Chen
- Suzhou Medical College of Soochow University, Suzhou City, 215123, Jiangsu Province, P.R. China.
- Department of Hematology, The First Affiliated Hospital of Gannan Medical University, Ganzhou City, 341000, Jiangxi Province, P.R. China.
- The Endemic Disease (Thalassemia) Clinical Research Center of Jiangxi Province, Ganzhou City, 341000, Jiangxi Province, P.R. China.
| |
Collapse
|
3
|
Wei Z, Su L, Gao S. The roles of ubiquitination in AML. Ann Hematol 2024; 103:3413-3428. [PMID: 37603061 DOI: 10.1007/s00277-023-05415-y] [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: 04/25/2023] [Accepted: 08/10/2023] [Indexed: 08/22/2023]
Abstract
Acute myeloid leukemia (AML) is a heterogeneously malignant disorder resulting in poor prognosis. Ubiquitination, a major post-translational modification (PTM), plays an essential role in regulating various cellular processes and determining cell fate. Despite these initial insights, the precise role of ubiquitination in AML pathogenesis and treatment remains largely unknown. In order to address this knowledge gap, we explore the relationship between ubiquitination and AML from the perspectives of signal transduction, cell differentiation, and cell cycle control; and try to find out how this relationship can be utilized to inform new therapeutic strategies for AML patients.
Collapse
Affiliation(s)
- Zhifeng Wei
- Department of Hematology, The First Hospital of Jilin University, Changchun, China
| | - Long Su
- Department of Hematology, The First Hospital of Jilin University, Changchun, China
| | - Sujun Gao
- Department of Hematology, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
4
|
Zangooie A, Tavoosi S, Arabhosseini M, Halimi A, Zangooie H, Baghsheikhi AH, Rahgozar S, Ahmadvand M, Jarrahi AM, Salehi Z. Ubiquitin-specific proteases (USPs) in leukemia: a systematic review. BMC Cancer 2024; 24:894. [PMID: 39048945 PMCID: PMC11270844 DOI: 10.1186/s12885-024-12614-x] [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: 04/20/2024] [Accepted: 07/09/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND Leukemia, a type of blood cell cancer, is categorized by the type of white blood cells affected (lymphocytes or myeloid cells) and disease progression (acute or chronic). In 2020, it ranked 15th among the most diagnosed cancers and 11th in cancer-related deaths globally, with 474,519 new cases and 311,594 deaths (GLOBOCAN2020). Research into leukemia's development mechanisms may lead to new treatments. Ubiquitin-specific proteases (USPs), a family of deubiquitinating enzymes, play critical roles in various biological processes, with both tumor-suppressive and oncogenic functions, though a comprehensive understanding is still needed. AIM This systematic review aimed to provide a comprehensive review of how Ubiquitin-specific proteases are involved in pathogenesis of different types of leukemia. METHODS We systematically searched the MEDLINE (via PubMed), Scopus, and Web of Science databases according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines (PRISMA) to identify relevant studies focusing on the role of USPs in leukemia. Data from selected articles were extracted, synthesized, and organized to present a coherent overview of the subject matter. RESULTS The review highlights the crucial roles of USPs in chromosomal aberrations, cell proliferation, differentiation, apoptosis, cell cycle regulation, DNA repair, and drug resistance. USP activity significantly impacts leukemia progression, inhibition, and chemotherapy sensitivity, suggesting personalized diagnostic and therapeutic approaches. Ubiquitin-specific proteases also regulate gene expression, protein stability, complex formation, histone deubiquitination, and protein repositioning in specific leukemia cell types. CONCLUSION The diagnostic, prognostic, and therapeutic implications associated with ubiquitin-specific proteases (USPs) hold significant promise and the potential to transform leukemia management, ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Alireza Zangooie
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Shima Tavoosi
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Mahan Arabhosseini
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Aram Halimi
- Research Center for Social Determinants of Health, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Epidemiology, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Helia Zangooie
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Soheila Rahgozar
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Mohammad Ahmadvand
- Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology, and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Zahra Salehi
- Hematology, Oncology and Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Nunes FM, Apolónio JD, Mota-Pinto A, Leão R. Epigenetic alterations in urothelial bladder cancer associated with disease outcomes. Int J Urol 2024; 31:220-229. [PMID: 37961796 DOI: 10.1111/iju.15335] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 10/19/2023] [Indexed: 11/15/2023]
Abstract
OBJECTIVES Bladder cancer (BLCA) is a molecular heterogeneous disease with known genetic distinctive signatures. However, DNA methylation is highly prevalent across a wide range of tumors, suggesting its potential in oncogenesis. Here, we aimed to interrogate the role of nine epigenetic alterations as diagnostic and prognostic markers in BLCA. METHODS DNA methylation, gene expression, and clinicopathological information were retrieved from The Cancer Genome Atlas data portal. Methylation values and gene expression were assessed to determine their association with normal and malignant tissue. Additionally, we studied the association between methylation values and clinicopathological variables. For the prognostic model, Kaplan-Meier Survival curves were generated. Lastly, univariate and multivariate analysis were performed to evaluate the simultaneous impact of methylation and clinicopathological variables on the risk of tumor progression and survival. RESULTS Nine CpG sites' methylation β -values involved in our study demonstrated different methylation signatures between normal and malignant urothelium. Hypermethylated CpGs were overrepresented in tumor tissue (p < 0.0001). Opposingly, 4 CpG sites showed lower methylation values in tumor samples (p < 0.0001). Cg12743248high and cg17192862low are risk factors for progression-free survival, whereas cg12374721high (HR:3.003 (1.283-7.030)) also demonstrated to be the most valuable independent risk factor for disease progression and a risk factor for overall survival. CONCLUSIONS We have identified that methylated cg12374721 shows promise as a diagnostic and independent prognostic marker in BLCA progression.
Collapse
Affiliation(s)
| | - Joana Dias Apolónio
- Algarve Biomedical Center Research Institute (ABC-RI), University of Algarve, Faro, Portugal
- Algarve Biomedical Center, Faro, Portugal
| | | | - Ricardo Leão
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
6
|
Ng A, Lovat F, Shih AJ, Ma Y, Pekarsky Y, DiCaro F, Crichton L, Sharma E, Yan XJ, Sun D, Song T, Zou YR, Will B, Croce CM, Chiorazzi N. Complete miRNA-15/16 loss in mice promotes hematopoietic progenitor expansion and a myeloid-biased hyperproliferative state. Proc Natl Acad Sci U S A 2023; 120:e2308658120. [PMID: 37844234 PMCID: PMC10614620 DOI: 10.1073/pnas.2308658120] [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: 05/29/2023] [Accepted: 09/13/2023] [Indexed: 10/18/2023] Open
Abstract
Dysregulated apoptosis and proliferation are fundamental properties of cancer, and microRNAs (miRNA) are critical regulators of these processes. Loss of miR-15a/16-1 at chromosome 13q14 is the most common genomic aberration in chronic lymphocytic leukemia (CLL). Correspondingly, the deletion of either murine miR-15a/16-1 or miR-15b/16-2 locus in mice is linked to B cell lymphoproliferative malignancies. However, unexpectedly, when both miR-15/16 clusters are eliminated, most double knockout (DKO) mice develop acute myeloid leukemia (AML). Moreover, in patients with CLL, significantly reduced expression of miR-15a, miR-15b, and miR-16 associates with progression of myelodysplastic syndrome to AML, as well as blast crisis in chronic myeloid leukemia. Thus, the miR-15/16 clusters have a biological relevance for myeloid neoplasms. Here, we demonstrate that the myeloproliferative phenotype in DKO mice correlates with an increase of hematopoietic stem and progenitor cells (HSPC) early in life. Using single-cell transcriptomic analyses, we presented the molecular underpinning of increased myeloid output in the HSPC of DKO mice with gene signatures suggestive of dysregulated hematopoiesis, metabolic activities, and cell cycle stages. Functionally, we found that multipotent progenitors (MPP) of DKO mice have increased self-renewing capacities and give rise to significantly more progeny in the granulocytic compartment. Moreover, a unique transcriptomic signature of DKO MPP correlates with poor outcome in patients with AML. Together, these data point to a unique regulatory role for miR-15/16 during the early stages of hematopoiesis and to a potentially useful biomarker for the pathogenesis of myeloid neoplasms.
Collapse
Affiliation(s)
- Anita Ng
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research Northwell Health, Manhasset, NY11030
| | - Francesca Lovat
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH43210
| | - Andrew J. Shih
- Boas Center for Human Genetics and Genomics, The Feinstein Institutes for Medical Research Northwell Health, Manhasset, NY11030
| | - Yuhong Ma
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY10461
| | - Yuri Pekarsky
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH43210
| | - Frank DiCaro
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research Northwell Health, Manhasset, NY11030
| | - Lita Crichton
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research Northwell Health, Manhasset, NY11030
| | - Esha Sharma
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research Northwell Health, Manhasset, NY11030
| | - Xiao Jie Yan
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research Northwell Health, Manhasset, NY11030
| | - Daqian Sun
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY10461
| | - Tengfei Song
- The Center for Autoimmune, Musculoskeletal, and Hematopoietic Diseases, The Feinstein Institutes for Medical Research Northwell Health, Manhasset, NY11030
| | - Yong-Rui Zou
- The Center for Autoimmune, Musculoskeletal, and Hematopoietic Diseases, The Feinstein Institutes for Medical Research Northwell Health, Manhasset, NY11030
- Departments of Medicine and Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY11549
| | - Britta Will
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY10461
| | - Carlo M. Croce
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH43210
| | - Nicholas Chiorazzi
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research Northwell Health, Manhasset, NY11030
- Departments of Medicine and Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY11549
| |
Collapse
|
7
|
Deng T, Zhong P, Lou R, Yang X. RNF220 promotes gastric cancer growth and stemness via modulating the USP22/wnt/β-catenin pathway. Tissue Cell 2023; 83:102123. [PMID: 37295272 DOI: 10.1016/j.tice.2023.102123] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 05/15/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023]
Abstract
Gastric cancer (GC) is a prevalent malignancy that seriously threatens the health and life of patients. Although Ring finger 220 (RNF220) has been demonstrated to participate in the development of various cancers, its role and mechanism in GC remain undiscovered. The expression of RNF220 was determined by The Cancer Genome Atlas (TCGA) database and Western blot. Additionally, the overall survival (OS) and post-progression survival (PPS) were analyzed based on the levels of RNF220 in the TCGA database. The role and mechanism of RNF220 in growth and stemness were investigated using cell counting kit-8, colony formation, sphere-formation, co-immunoprecipitation, and Western blot experiments. Furthermore, the role of RNF220 was investigated in a xenografted mouse model. The expression of RNF220 was found to be upregulated in GC, which predicted unfavorable OS and PPS in patients with GC. Knockdown of RNF220 reduced cell viability, colony numbers, numbers of spheres formation, and the relative protein levels of Nanog, Sox2, and Oct4 in both AGS and MKN-45 cells. Moreover, overexpression of RNF220 increased cell viability and the numbers of spheres formation in MKN-45 cells. Mechanistically, RNF220 bound to USP22, and interference of RNF220 downregulated the Wnt/β-catenin axis via USP22, which was confirmed by the overexpression of USP22 in both cell lines. Furthermore, silencing of RNF220 significantly decreased tumor volume and weight, the level of Ki-67, and the relative protein levels of USP22, β-catenin, c-myc, Nanog, Sox2, and Oct4. Taken together, downregulation of RNF220 suppressed GC cell growth and stemness by downregulating the USP22/Wnt/β-catenin axis.
Collapse
Affiliation(s)
- Taozhi Deng
- Department of Gastroenterology, Hainan Cancer Hospital, Haikou, Hainan 570000, China
| | - Ping Zhong
- Department of Gastroenterology, Hainan Cancer Hospital, Haikou, Hainan 570000, China
| | - Runlong Lou
- Department of Gastroenterology, Hainan Cancer Hospital, Haikou, Hainan 570000, China
| | - Xiaojun Yang
- Department of Gastroenterology, Suzhou BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Suzhou, Jiangsu 215010, China.
| |
Collapse
|
8
|
The Systematic Analyses of RING Finger Gene Signature for Predicting the Prognosis of Patients with Hepatocellular Carcinoma. JOURNAL OF ONCOLOGY 2022; 2022:2466006. [PMID: 36199791 PMCID: PMC9529411 DOI: 10.1155/2022/2466006] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 09/08/2022] [Indexed: 12/24/2022]
Abstract
RING finger (RNF) proteins are frequently dysregulated in human malignancies and are tightly associated with tumorigenesis. However, the expression profiles of RNF genes in hepatocellular carcinoma (HCC) and their relations with prognosis remain undetermined. Here, we aimed at constructing a prognostic model according to RNF genes for forecasting the outcomes of HCC patients using the data from The Cancer Genome Atlas (TCGA) program. We collected HCC datasets to validate the values of our model in predicting prognosis of HCC patients from International Cancer Genome Consortium (ICGC) platform. Then, functional experiments were carried out to explore the roles of the representative RNF in HCC progression. A total of 107 differentially expressed RNFs were obtained between TCGA-HCC tumor and normal tissues. After comprehensive evaluation, a prognostic signature composed of 11 RNFs (RNF220, RNF25, TRIM25, BMI1, RNF216P1, RNF115, RNF2, TRAIP, RNF157, RNF145, and RNF19B) was constructed based on TCGA cohort. Then, the Kaplan-Meier (KM) curves and the receiver operating characteristic curve (ROC) were employed to evaluate predictive power of the prognostic model in testing cohort (TCGA) and validation cohort (ICGC). The KM and ROC curves illustrated the good predictive power in testing and validation cohort. The areas under the ROC curve are 0.77 and 0.76 in these two cohorts, respectively. Among the prognostic signature genes, BMI1 was selected as a representative for functional study. We found that BMI1 protein level was significantly upregulated in HCC tissues. Moreover, the inhibitor of BMI1, PTC-209, displayed an excellent anti-HCC effect in vitro. Enrichment analysis of BMI1 downstream targets showed that BMI1 might be involved in tumor immunotherapy. Together, our overall analyses revealed that the 11-RNFs prognostic signature might provide us latent chances for evaluating HCC prognosis and developing novel HCC therapy.
Collapse
|
9
|
Yan J, Tan M, Yu L, Jin X, Li Y. Ring finger 220 promotes the stemness and progression of colon cancer cells via Ubiquitin specific peptidase 22-BMI1 axis. Bioengineered 2021; 12:12060-12069. [PMID: 34753387 PMCID: PMC8809949 DOI: 10.1080/21655979.2021.2003664] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/02/2021] [Accepted: 11/03/2021] [Indexed: 12/03/2022] Open
Abstract
Colorectal cancer (CRC) is ranked as the third most common malignancy worldwide. Therefore, it is urgent to screen novel and effective molecular drug targets for colorectal cancer therapeutics. In this study, the specific role and related mechanism underlying Ring finger (RNF) 220 in colon cancer were investigated. Firstly, RT-PCR assay was used to compare differences between expression levels of RNF220 in colorectal tumor and normal tissues. Western blot and RT-PCR assays were applied to examine the protein levels of RNF220 in normal colonic mucosa and colorectal cancer cells. We found that RNF220 was upregulated in colorectal cancer in patients and cell models. RNF220 promoted the proliferation and migration, invasion of colorectal cancer cells through BrdU incorporation, clone formation, transwell and wound healing assays. Spheroid formation and western blot assays illustrated that RNF220 promoted the stemness of colorectal cancer cells. Moreover, we found that RNF220 regulated BMI1 expression through USP22 by western blot. Finally, we discovered that RNF220 facilitated tumor growth in vivo through establishment of subcutaneous xenograft tumor mice model. In conclusion, RNF220 promoted the stemness and progression of colon cancer cells via the USP22-BMI1 axis.
Collapse
Affiliation(s)
- Jianwen Yan
- Department of Surgery 1, Guilin Tcm Hospital of China, Guilin City, Guangxi Zhuang Autonomous Region, China
| | - Min Tan
- Department of Surgery 1, Guilin Tcm Hospital of China, Guilin City, Guangxi Zhuang Autonomous Region, China
| | - Lin Yu
- Department of Surgery 1, Guilin Tcm Hospital of China, Guilin City, Guangxi Zhuang Autonomous Region, China
| | - Xichao Jin
- Department of Surgery 1, Guilin Tcm Hospital of China, Guilin City, Guangxi Zhuang Autonomous Region, China
| | - Yangcheng Li
- Department of General Surgery, Nantong Tumor Hospital, Nantong City, Jiangsu Province, China
| |
Collapse
|
10
|
Ma P, Mao B. The many faces of the E3 ubiquitin ligase, RNF220, in neural development and beyond. Dev Growth Differ 2021; 64:98-105. [PMID: 34716995 DOI: 10.1111/dgd.12756] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/30/2021] [Accepted: 10/02/2021] [Indexed: 11/28/2022]
Abstract
Ubiquitin modification plays important roles in many cellular processes that are fundamental for vertebrate embryo development, such as cell division, differentiation, and migration. Aberrant function or deregulation of ubiquitination enzymes can cause developmental disorders, cancer progression, and neurodegenerative diseases in humans. RING finger protein 220 (RNF220) is an evolutionarily conserved RING-type ubiquitin E3 ligase. Recent studies have revealed the roles and mechanisms of RNF220 and its partner protein, zinc finger C4H2-type containing protein (ZC4H2), in embryonic development and human diseases. Using mouse and zebrafish models, it has been shown that RNF220 regulates sonic hedgehog (Shh) signaling via Gli and embryonic ectoderm development (EED), a polycomb repressive complex 2 (PRC2) component, during ventral neural patterning and cerebellum development. In addition, RNF220 also regulates the development and functions of central noradrenergic and motor neurons in mice. By stabilizing β-catenin and signal transducer and activator of transcription 1 (STAT1), RNF220 is also involved in Wnt and interferon (IFN)-STAT1 signaling and thus the regulation of tumorigenesis and immune response, respectively. In humans, both RNF220 and ZC4H2 mutations have been reported to be associated with diseases accompanied by complicated neural defects. In this review, we summarize the current knowledge of RNF220 with special emphasis on its roles and mechanisms of action in signal transduction, vertebrate neural development, and related human disorders.
Collapse
Affiliation(s)
- Pengcheng Ma
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Bingyu Mao
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
| |
Collapse
|
11
|
Liu X, Liu X, Cai M, Luo A, He Y, Liu S, Zhang X, Yang X, Xu L, Jiang H. CircRNF220, not its linear cognate gene RNF220, regulates cell growth and is associated with relapse in pediatric acute myeloid leukemia. Mol Cancer 2021; 20:139. [PMID: 34702297 PMCID: PMC8549339 DOI: 10.1186/s12943-021-01395-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 07/17/2021] [Indexed: 02/08/2023] Open
Abstract
Background Circular RNAs (circRNAs) constitute a family of transcripts with unique structures and have been confirmed to be critical in tumorigenesis and to be potential biomarkers or therapeutic targets. However, only a few circRNAs have been functionally characterized in pediatric acute myeloid leukemia (AML). Methods Here, we investigated the expression pattern of circRNAs in pediatric AML using a circRNA microarray. The characteristics, potential diagnostic value, and prognostic significance of circRNF220 were evaluated. A series of functional experiments were performed to investigate the role of circRNF220 in primary pediatric AML cells. Then we investigated the aberrant transcriptional networks regulated by circRNF220 in primary AML cells by RNA-seq. Furthermore, biotin RNA pulldown assays were implemented to verify the relationship between circRNF220 and miR-30a. Results We identified a circRNA, circRNF220, which was specifically abundant in and accumulated in the peripheral blood and bone marrow of pediatric patients with AML. It could distinguish AML from ALL and other hematological malignancies with high sensitivity and specificity. Significantly, circRNF220 expression independently predicted prognosis, while high expression of circRNF220 was an unfavorable prognostic marker for relapse. Furthermore, we characterized the function of circRNF220 and found that circRNF220 knockdown specifically inhibited proliferation and promoted apoptosis in AML cell lines and primary cells. Mechanistically, circRNF220 may act as an endogenous sponge of miR-30a to sequester miR-30a and inhibit its activity, which increases the expression of its targets MYSM1 and IER2 and implicated in AML relapse. Conclusions Collectively, these findings demonstrated that circRNF220 could be highly efficient and specific for the accurate diagnosis of pediatric AML, with implications for relapse prediction. Supplementary Information The online version contains supplementary material available at 10.1186/s12943-021-01395-7.
Collapse
Affiliation(s)
- Xiaodan Liu
- Division of Birth Cohort Study, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.,Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Zhujiang Newtown, Tianhe District, Guangzhou, 510623, Guangdong, China
| | - Xiaoping Liu
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Zhujiang Newtown, Tianhe District, Guangzhou, 510623, Guangdong, China.,Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Mansi Cai
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Zhujiang Newtown, Tianhe District, Guangzhou, 510623, Guangdong, China.,Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Ailing Luo
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Zhujiang Newtown, Tianhe District, Guangzhou, 510623, Guangdong, China.,Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yingyi He
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Zhujiang Newtown, Tianhe District, Guangzhou, 510623, Guangdong, China
| | - Sha Liu
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Zhujiang Newtown, Tianhe District, Guangzhou, 510623, Guangdong, China
| | - Xiaohong Zhang
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Zhujiang Newtown, Tianhe District, Guangzhou, 510623, Guangdong, China
| | - Xu Yang
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Zhujiang Newtown, Tianhe District, Guangzhou, 510623, Guangdong, China.,Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Ling Xu
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Zhujiang Newtown, Tianhe District, Guangzhou, 510623, Guangdong, China.
| | - Hua Jiang
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Zhujiang Newtown, Tianhe District, Guangzhou, 510623, Guangdong, China.
| |
Collapse
|