1
|
Li Y, Fang J, Jiao Y, Lv Q, Xiao X, Zheng S, Chen X, Song J, Zhang X, Zhang L, Ma Z, Li C, Hao L. MiR-21-5p enhances differentiation and mitigates oleic acid-induced lipid droplet accumulation in C2C12 myoblasts by targeting FBXO11. Anim Biosci 2025; 38:1279-1290. [PMID: 40045615 PMCID: PMC12061579 DOI: 10.5713/ab.24.0665] [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: 09/24/2024] [Revised: 11/24/2024] [Accepted: 12/30/2024] [Indexed: 05/09/2025] Open
Abstract
OBJECTIVE This study aimed to investigate the role of miRNA 21-5p in regulating the differentiation of C2C12 myoblasts and intramuscular lipid droplets accumulation in myotubes. METHODS The role of miR-21-5p in the proliferation and differentiation of myofibroblasts and intracellular lipid accumulation was analyzed using bioinformatics, CCK-8 assay, quantitative real-time polymerase chain reaction, immunoblotting, immunofluorescence staining, and Oil Red O staining. RESULTS The analysis of porcine BodyMap transcriptome data revealed differential expression of miRNA 21-5p in skeletal muscle and adipose tissue. Bioinformatics analysis combined with a dual-luciferase reporter assay demonstrated that FBXO11 serves as a direct target of miR-21-5p. Transfection experiments involving a miR-21-5p mimic, miR-21-5p inhibitor, and si-FBXO11 in C2C12 cells showed that overexpression of miR-21-5p or silencing of FBXO11 significantly enhanced the proliferation of C2C12 cells, upregulated myogenesis-related factors, and promoted myotube formation. Furthermore, oleic acid-induced lipid accumulation in myotubes was suppressed, accompanied by reduced expression of adipogenesis-related genes. Conversely, inhibition of miR-21-5p expression produced opposite effects. CONCLUSION These findings indicate that miR-21-5p promotes proliferation and differentiation while inhibiting intramyocellular lipid deposition by targeting the 3'-untranslated region of FBXO11 in myogenic cell. The results suggest that miR-21-5p could serve as a potential miRNA biomarker for regulating intramuscular adipogenesis.
Collapse
Affiliation(s)
- Yi Li
- College of Animal Science, Jilin University, Changchun,
China
| | - Jiayuan Fang
- College of Animal Science, Jilin University, Changchun,
China
| | - Yingying Jiao
- College of Animal Science, Jilin University, Changchun,
China
| | - Qinchuan Lv
- College of Animal Science, Jilin University, Changchun,
China
| | - Xingyu Xiao
- College of Animal Science, Jilin University, Changchun,
China
| | - Shuo Zheng
- College of Animal Science, Jilin University, Changchun,
China
| | - Xi Chen
- College of Animal Science, Jilin University, Changchun,
China
| | - Jie Song
- College of Animal Science, Jilin University, Changchun,
China
| | - Xunming Zhang
- College of Animal Science, Jilin University, Changchun,
China
| | - Libo Zhang
- College of Animal Science, Jilin University, Changchun,
China
| | - Ze Ma
- College of Animal Science, Jilin University, Changchun,
China
| | - Changhong Li
- College of Life Science, Baicheng Normal University, Baicheng,
China
| | - Linlin Hao
- College of Animal Science, Jilin University, Changchun,
China
| |
Collapse
|
2
|
Lin Z, Liang F, Hong G, Jiang X, Zhang Q, Wang M. TACC3 enhances glycolysis in bladder cancer cells through inducing acetylation of c-Myc. Cell Death Dis 2025; 16:311. [PMID: 40246827 PMCID: PMC12006502 DOI: 10.1038/s41419-025-07645-6] [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: 09/06/2024] [Revised: 04/05/2025] [Accepted: 04/07/2025] [Indexed: 04/19/2025]
Abstract
The proliferation of bladder cancer (BC) cells is driven by metabolic reprogramming, marked by a glycolytic dependency to sustain uncontrolled growth. While Transforming Acidic Coiled-Coil Containing Protein 3 (TACC3) is known to promote BC progression and correlate with poor prognosis, the mechanisms underlying its upregulation and role in aerobic glycolysis remain unclear. Here, we identify E2F3 as a direct transcriptional activator of TACC3, with its amplification in BC driving elevated TACC3 expression. TACC3 overexpression enhances glycolysis, increasing glucose consumption, lactate production, and expression of glycolytic enzymes (e.g., GLUT1, HK2, PFKFB3), while its knockdown suppresses these effects. Pharmacological inhibition of glycolysis abrogates TACC3-driven tumor growth in vitro and in vivo. Mechanistically, TACC3 interacts with c-Myc, promoting its acetylation at lysine 323 (K323) by recruiting the acetyltransferase PCAF and antagonizing the deacetylase SIRT1. This acetylation stabilizes c-Myc, amplifying its transcriptional activation of glycolytic targets. Our findings establish TACC3 as a critical regulator of c-Myc-driven metabolic reprogramming in BC, highlighting its potential as a therapeutic target to disrupt glycolysis and oncogenic c-Myc signaling.
Collapse
Affiliation(s)
- Zhirui Lin
- Institute of Medical Research, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong Province, People's Republic of China.
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong Province, People's Republic of China.
| | - Falian Liang
- Radiation Oncology Department, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510245, Guangdong Province, People's Republic of China
| | - Gengde Hong
- Radiation Oncology Department, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510245, Guangdong Province, People's Republic of China
| | - Xizhen Jiang
- Radiation Oncology Department, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510245, Guangdong Province, People's Republic of China
| | - Qingling Zhang
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong Province, People's Republic of China.
| | - Mengyao Wang
- Radiation Oncology Department, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510245, Guangdong Province, People's Republic of China.
| |
Collapse
|
3
|
Chen CM, Yu R. A multi-step completion process model of cell plasticity. Brief Bioinform 2025; 26:bbaf165. [PMID: 40223810 PMCID: PMC11995008 DOI: 10.1093/bib/bbaf165] [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: 11/27/2024] [Revised: 02/11/2025] [Accepted: 03/25/2025] [Indexed: 04/15/2025] Open
Abstract
Plasticity is the potential for cells or cell populations to change their phenotypes and behaviors in response to internal or external cues. Plasticity is fundamental to many complex biological processes, yet to date there remains a lack of mathematical models that can elucidate and predict molecular behaviors in a plasticity program. Here, we report a new mathematical framework that models cell plasticity as a multi-step completion process, where the system moves from the initial state along a path guided by multiple intermediate attractors until the final state (i.e. a new homeostasis) is reached. Using omics time-series data as model input, we show that our method fits data well; identifies attractor states by their timing and molecular markers which are well-aligned with domain knowledge; and can make quantitative and time-resolved predictions such as the molecular outcomes of blocking a plasticity program from reaching completion, to an R2 of 0.53-0.63. We demonstrate that application of our model to primary patient-derived data can provide quantitative insights and predictions that may be useful in guiding further research and potential biomedical interventions.
Collapse
Affiliation(s)
- Chen M Chen
- Department of Molecular Developmental Biology, Radboud Institute for Molecular Life Sciences, Faculty of Science, Radboud University, Geert Grooteplein-Zuid 26-28, Nijmegen, 6525GA, The Netherlands
| | - Rosemary Yu
- Department of Molecular Developmental Biology, Radboud Institute for Molecular Life Sciences, Faculty of Science, Radboud University, Geert Grooteplein-Zuid 26-28, Nijmegen, 6525GA, The Netherlands
| |
Collapse
|
4
|
Hu F, Zhao L, Wang J, Li X, Xue Z, Ma Y, Zheng M, Chen C, Tong M, Guo X, Li H, Jin H, Xie Q, Zhang X, Huang C, Huang H. TRIM40 interacts with ROCK1 directly and inhibits colorectal cancer cell proliferation through the c-Myc/p21 axis. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119855. [PMID: 39357549 DOI: 10.1016/j.bbamcr.2024.119855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 09/10/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Colorectal cancer (CRC) is the most common malignancy of the digestive tract, and to date, morbidity and mortality rates remain high. While existing therapeutic methods have achieved certain effective outcomes, there are still many problems in treating this disease. Therefore, it is still urgent to constantly find new therapeutic targets in CRC that could lead to new therapeutics. METHODS Immunohistochemistry, Real-time PCR and Western Blot were employed to measure mRNA and protein levels of the target protein, respectively. The proliferation ability of CRC cells was evaluated using ATP assay, Soft agar assay, and nude mouse subcutaneous tumorigenesis assay. Protein Degradation Assay was conducted to determine protein degradation rate, while Ubiquitination assay was used to assess the ubiquitination modification level of target proteins. Immunoprecipitation assay was used to study protein interactions, and pull-down assay was employed to investigate direct interactions between proteins. RESULTS TRIM40 was significantly down-regulated in CRC tissues, with its expression levels positively correlating with disease prognosis. Using both in vitro and in vivo approaches, it was demonstrated that TRIM40 could significantly inhibit the proliferation of CRC cells. Molecular mechanism studies showed that TRIM40 directly binds to and ubiquitinates ROCK1 protein, accelerating its degradation and subsequently reducing the stability of c-Myc protein. This cascade of events results in the release of transcriptional inhibition of p21 by c-Myc, leading to increased p21 expression and G0/G1 phase arrest in CRC cells. CONCLUSION This research suggests that TRIM40 could be a valuable therapeutic target for the treatment of CRC.
Collapse
Affiliation(s)
- Fangyu Hu
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Lingling Zhao
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Junyu Wang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiaoying Li
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Zixuan Xue
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yimeng Ma
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Minghui Zheng
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Chenglin Chen
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Meiting Tong
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiaohuan Guo
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Hongyan Li
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Honglei Jin
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Qipeng Xie
- Department of Laboratory Medicine, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiaodong Zhang
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Chuanshu Huang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325035, China.
| | - Haishan Huang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| |
Collapse
|
5
|
Kin K, Bhogale S, Zhu L, Thomas D, Bertol J, Zheng WJ, Sinha S, Fakhouri WD. Sequence-to-expression approach to identify etiological non-coding DNA variations in P53 and cMYC-driven diseases. Hum Mol Genet 2024; 33:1697-1710. [PMID: 39017605 PMCID: PMC11413647 DOI: 10.1093/hmg/ddae109] [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: 03/17/2024] [Revised: 06/08/2024] [Accepted: 07/11/2024] [Indexed: 07/18/2024] Open
Abstract
Disease risk prediction based on genomic sequence and transcriptional profile can improve disease screening and prevention. Despite identifying many disease-associated DNA variants, distinguishing deleterious non-coding DNA variations remains poor for most common diseases. In this study, we designed in vitro experiments to uncover the significance of occupancy and competitive binding between P53 and cMYC on common target genes. Analyzing publicly available ChIP-seq data for P53 and cMYC in embryonic stem cells showed that ~344-366 regions are co-occupied, and on average, two cis-overlapping motifs (CisOMs) per region were identified, suggesting that co-occupancy is evolutionarily conserved. Using U2OS and Raji cells untreated and treated with doxorubicin to increase P53 protein level while potentially reducing cMYC level, ChIP-seq analysis illustrated that around 16 to 922 genomic regions were co-occupied by P53 and cMYC, and substitutions of cMYC signals by P53 were detected post doxorubicin treatment. Around 187 expressed genes near co-occupied regions were altered at mRNA level according to RNA-seq data analysis. We utilized a computational motif-matching approach to illustrate that changes in predicted P53 binding affinity in CisOMs of co-occupied elements significantly correlate with alterations in reporter gene expression. We performed a similar analysis using SNPs mapped in CisOMs for P53 and cMYC from ChIP-seq data, and expression of target genes from GTEx portal. We found significant correlation between change in cMYC-motif binding affinity in CisOMs and altered expression. Our study brings us closer to developing a generally applicable approach to filter etiological non-coding variations associated with common diseases.
Collapse
Affiliation(s)
- Katherine Kin
- Department of Diagnostic and Biomedical Sciences, Center for Craniofacial Research, School of Dentistry, University of Texas Health Science Center at Houston, 7500 Cambridge St, Houston, TX 77054, United States
| | - Shounak Bhogale
- Center for Biophysics and Quantitative Biology, University of Illinois at Urbana–Champaign, 600 S Mathews Ave, Urbana, IL 61801, United States
| | - Lisha Zhu
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, 7000 Fannin St #600, Houston, TX 77030, United States
| | - Derrick Thomas
- Department of Diagnostic and Biomedical Sciences, Center for Craniofacial Research, School of Dentistry, University of Texas Health Science Center at Houston, 7500 Cambridge St, Houston, TX 77054, United States
| | - Jessica Bertol
- Department of Diagnostic and Biomedical Sciences, Center for Craniofacial Research, School of Dentistry, University of Texas Health Science Center at Houston, 7500 Cambridge St, Houston, TX 77054, United States
| | - W Jim Zheng
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, 7000 Fannin St #600, Houston, TX 77030, United States
| | - Saurabh Sinha
- Center for Biophysics and Quantitative Biology, University of Illinois at Urbana–Champaign, 600 S Mathews Ave, Urbana, IL 61801, United States
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory University, Georgia Institute of Technology, North Avenue Atlanta, GA 30332, United States
| | - Walid D Fakhouri
- Department of Diagnostic and Biomedical Sciences, Center for Craniofacial Research, School of Dentistry, University of Texas Health Science Center at Houston, 7500 Cambridge St, Houston, TX 77054, United States
- Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin St, Houston, TX 77030, United States
| |
Collapse
|
6
|
Zhao Z, Liu M, Lin Z, Zhu M, Lv L, Zhu X, Fan R, Al-Danakh A, He H, Tan G. The mechanism of USP43 in the development of tumor: a literature review. Aging (Albany NY) 2024; 16:6613-6626. [PMID: 38613804 PMCID: PMC11042928 DOI: 10.18632/aging.205731] [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: 10/19/2023] [Accepted: 03/13/2024] [Indexed: 04/15/2024]
Abstract
Ubiquitination of the proteins is crucial for governing protein degradation and regulating fundamental cellular processes. Deubiquitinases (DUBs) have emerged as significant regulators of multiple pathways associated with cancer and other diseases, owing to their capacity to remove ubiquitin from target substrates and modulate signaling. Consequently, they represent potential therapeutic targets for cancer and other life-threatening conditions. USP43 belongs to the DUBs family involved in cancer development and progression. This review aims to provide a comprehensive overview of the existing scientific evidence implicating USP43 in cancer development. Additionally, it will investigate potential small-molecule inhibitors that target DUBs that may have the capability to function as anti-cancer medicines.
Collapse
Affiliation(s)
- Ziqi Zhao
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian 116011, China
| | - Meichen Liu
- Department of Neurology, The First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian 116011, China
| | - Zhikun Lin
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian 116011, China
- Liaoning Key Laboratory of Molecular Targeted Drugs in Hepatobiliary and Pancreatic Cancer, Dalian 116000, China
| | - Mengru Zhu
- Department of Plastic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian 116011, China
| | - Linlin Lv
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian 116011, China
| | - Xinqing Zhu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian 116011, China
| | - Rui Fan
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, National, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Abdullah Al-Danakh
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian 116011, China
| | - Hui He
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian 116011, China
| | - Guang Tan
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian 116011, China
- Liaoning Key Laboratory of Molecular Targeted Drugs in Hepatobiliary and Pancreatic Cancer, Dalian 116000, China
| |
Collapse
|
7
|
Sheppard AJ, Delgado K, Barfield AM, Xu Q, Massey PA, Dong Y, Barton RS. Rapamycin Inhibits Senescence and Improves Immunomodulatory Function of Mesenchymal Stem Cells Through IL-8 and TGF-β Signaling. Stem Cell Rev Rep 2024; 20:816-826. [PMID: 38340274 PMCID: PMC10984889 DOI: 10.1007/s12015-024-10682-x] [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] [Accepted: 01/16/2024] [Indexed: 02/12/2024]
Abstract
Mesenchymal stromal cells (MSCs) grown in high-density monolayers (sheets) are promising vehicles for numerous bioengineering applications. When MSC sheets are maintained in prolonged cultures, they undergo rapid senescence, limiting their downstream efficacy. Although rapamycin is a potential agent that can inhibit senescence in cell cultures, no study has investigated rapamycin's effect on MSCs grown in high-density culture and its effect on downstream target gene expression. In this study, placental-derived MSCs (PMSCs) were seeded at high density to generate PMSC sheets in 24 hours and were then treated with rapamycin or vehicle for up to 7 days. Autophagy activity, cell senescence and apoptosis, cell size and granularity, and senescence-associated cytokines (IL-6 and IL-8) were analyzed. Differential response in gene expression were assessed via microarray analysis. Rapamycin significantly increased PMSC sheet autophagy activity, inhibited cellular senescence, decreased cell size and granularity at all timepoints. Rapamycin also significantly decreased the number of cells in late apoptosis at day 7 of sheet culture, as well as caspase 3/7 activity at all timepoints. Notably, while rapamycin decreased IL-6 secretion, increased IL-8 levels were observed at all timepoints. Microarray analysis further confirmed the upregulation of IL-8 transcription, as well as provided a list of 396 genes with 2-fold differential expression, where transforming growth factor-β (TGF-β) signaling were identified as important upregulated pathways. Rapamycin both decreased senescence and has an immunomodulatory action of PMSCs grown in sheet culture, which will likely improve the chemotaxis of pro-healing cells to sites of tissue repair in future bioengineering applications.
Collapse
Affiliation(s)
- Aaron J Sheppard
- School of Medicine, LSU Health Shreveport, Shreveport, LA, USA
- Department of Orthopedic Surgery, LSU Health Shreveport, Shreveport, LA, USA
| | - Kristin Delgado
- School of Medicine, LSU Health Shreveport, Shreveport, LA, USA
| | | | - Qinqin Xu
- Department of Orthopedic Surgery, LSU Health Shreveport, Shreveport, LA, USA
| | - Patrick A Massey
- Department of Orthopedic Surgery, LSU Health Shreveport, Shreveport, LA, USA
| | - Yufeng Dong
- Department of Orthopedic Surgery, LSU Health Shreveport, Shreveport, LA, USA.
| | - Richard S Barton
- Department of Orthopedic Surgery, LSU Health Shreveport, Shreveport, LA, USA
| |
Collapse
|
8
|
Song XQ, Chen BB, Jin YM, Wang CY. DNMT1-mediated epigenetic suppression of FBXO32 expression promoting cyclin dependent kinase 9 (CDK9) survival and esophageal cancer cell growth. Cell Cycle 2024; 23:262-278. [PMID: 38597826 PMCID: PMC11057636 DOI: 10.1080/15384101.2024.2309022] [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: 06/16/2023] [Revised: 12/11/2023] [Accepted: 12/25/2023] [Indexed: 04/11/2024] Open
Abstract
Esophageal cancer (EC) is a common and serious form of cancer, and while DNA methyltransferase-1 (DNMT1) promotes DNA methylation and carcinogenesis, the role of F-box protein 32 (FBXO32) in EC and its regulation by DNMT1-mediated methylation is still unclear. FBXO32 expression was examined in EC cells with high DNMT1 expression using GSE163735 dataset. RT-qPCR assessed FBXO32 expression in normal and EC cells, and impact of higher FBXO32 expression on cell proliferation, migration, and invasion was evaluated, along with EMT-related proteins. The xenograft model established by injecting EC cells transfected with FBXO32 was used to evaluate tumor growth, apoptosis, and tumor cells proliferation and metastasis. Chromatin immunoprecipitation (ChIP) assay was employed to study the interaction between DNMT1 and FBXO32. HitPredict, co-immunoprecipitation (Co-IP), and Glutathione-S-transferase (GST) pulldown assay analyzed the interaction between FBXO32 and cyclin dependent kinase 9 (CDK9). Finally, the ubiquitination assay identified CDK9 ubiquitination, and its half-life was measured using cycloheximide and confirmed through western blotting. DNMT1 negatively correlated with FBXO32 expression in esophageal cells. High FBXO32 expression was associated with better overall survival in patients. Knockdown of DNMT1 in EC cells increased FBXO32 expression and suppressed malignant phenotypes. FBXO32 repressed EC tumor growth and metastasis in mice. Enrichment of DNMT1 in FBXO32 promoter region led to increased DNA methylation and reduced transcription. Mechanistically, FBXO32 degraded CDK9 through promoting its ubiquitination.
Collapse
Affiliation(s)
- Xian-Qiang Song
- Department of Radiotherapy, Qinhuai Medical District, General Hospital of Eastern Theater Command, Nanjing, PR China
| | - Bin-Bin Chen
- Departments of Laboratory Medicine, Qinhuai Medical District, General Hospital of Eastern Theater Command, Nanjing, PR China
| | - Yong-Mei Jin
- Department of Cardiothoracic Surgery, Qinhuai Medical District, General Hospital of Eastern Theater Command, Nanjing, PR China
| | - Chang-Yong Wang
- Department of Cardiothoracic Surgery, Qinhuai Medical District, General Hospital of Eastern Theater Command, Nanjing, PR China
| |
Collapse
|
9
|
Li M, Yu J, Ju L, Wang Y, Jin W, Zhang R, Xiang W, Ji M, Du W, Wang G, Qian K, Zhang Y, Xiao Y, Wang X. USP43 stabilizes c-Myc to promote glycolysis and metastasis in bladder cancer. Cell Death Dis 2024; 15:44. [PMID: 38218970 PMCID: PMC10787741 DOI: 10.1038/s41419-024-06446-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 01/15/2024]
Abstract
A hallmark of tumor cells, including bladder cancer (BLCA) cells, is metabolic reprogramming toward aerobic glycolysis (Warburg effect). The classical oncogene MYC, which is crucial in regulating glycolysis, is amplified and activated in BLCA. However, direct targeting of the c-Myc oncoprotein, which regulates glycolytic metabolism, presents great challenges and necessitates the discovery of a more clarified regulatory mechanism to develop selective targeted therapy. In this study, a siRNA library targeting deubiquitinases identified a candidate enzyme named USP43, which may regulate glycolytic metabolism and c-Myc transcriptional activity. Further investigation using functional assays and molecular studies revealed a USP43/c-Myc positive feedback loop that contributes to the progression of BLCA. Moreover, USP43 stabilizes c-Myc by deubiquitinating c-Myc at K148 and K289 primarily through deubiquitinase activity. Additionally, upregulation of USP43 protein in BLCA increased the chance of interaction with c-Myc and interfered with FBXW7 access and degradation of c-Myc. These findings suggest that USP43 is a potential therapeutic target for indirectly targeting glycolytic metabolism and the c-Myc oncoprotein consequently enhancing the efficacy of bladder cancer treatment.
Collapse
Affiliation(s)
- Mingxing Li
- Department of Urology, Laboratory of Precision Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jingtian Yu
- Department of Urology, Laboratory of Precision Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lingao Ju
- Department of Biological Repositories, Human Genetic Resources Preservation Center of Hubei Province, Hubei Key Laboratory of Urological Diseases, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yejinpeng Wang
- Department of Urology, Laboratory of Precision Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wan Jin
- Department of Biological Repositories, Human Genetic Resources Preservation Center of Hubei Province, Hubei Key Laboratory of Urological Diseases, Zhongnan Hospital of Wuhan University, Wuhan, China
- Euler Technology, ZGC Life Sciences Park, Beijing, China
| | - Renjie Zhang
- Department of Urology, Laboratory of Precision Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wan Xiang
- Department of Biological Repositories, Human Genetic Resources Preservation Center of Hubei Province, Hubei Key Laboratory of Urological Diseases, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Meng Ji
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wenzhi Du
- Department of Urology, Laboratory of Precision Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
- Department of Urology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Gang Wang
- Department of Urology, Laboratory of Precision Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
- Department of Biological Repositories, Human Genetic Resources Preservation Center of Hubei Province, Hubei Key Laboratory of Urological Diseases, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Kaiyu Qian
- Department of Urology, Laboratory of Precision Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
- Department of Biological Repositories, Human Genetic Resources Preservation Center of Hubei Province, Hubei Key Laboratory of Urological Diseases, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yi Zhang
- Euler Technology, ZGC Life Sciences Park, Beijing, China.
- Center for Quantitative Biology, School of Life Sciences, Peking University, Beijing, China.
| | - Yu Xiao
- Department of Biological Repositories, Human Genetic Resources Preservation Center of Hubei Province, Hubei Key Laboratory of Urological Diseases, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Xinghuan Wang
- Department of Urology, Laboratory of Precision Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China.
- Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China.
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China.
| |
Collapse
|
10
|
Zeng Y, Jiang H, Zhang X, Xu J, Wu X, Xu Q, Cai W, Ying H, Zhou R, Ding Y, Ying K, Song X, Chen Z, Zeng L, Zhao L, Yu F. Canagliflozin reduces chemoresistance in hepatocellular carcinoma through PKM2-c-Myc complex-mediated glutamine starvation. Free Radic Biol Med 2023; 208:571-586. [PMID: 37696420 DOI: 10.1016/j.freeradbiomed.2023.09.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 09/13/2023]
Abstract
Cisplatin (CPT) is one of the standard treatments for hepatocellular carcinoma (HCC). However, its use is limits as a monotherapy due to drug resistance, and the underlying mechanism remains unclear. To solve this problem, we tried using canagliflozin (CANA), a clinical drug for diabetes, to reduce chemoresistance to CPT, and the result showed that CANA could vigorously inhibit cell proliferation and migration independent of the original target SGLT2. Mechanistically, CANA reduced aerobic glycolysis in HCC by targeting PKM2. The downregulated PKM2 directly bound to the transcription factor c-Myc in the cytoplasm to form a complex, which upregulated the level of phosphorylated c-Myc Thr58 and promoted the ubiquitination and degradation of c-Myc. Decreased c-Myc reduced the expression of GLS1, a key enzyme in glutamine metabolism, leading to impaired glutamine utilization. Finally, intracellular glutamine starvation induced ferroptosis and sensitized HCC to CPT. In conclusion, our study showed that CANA re-sensitized HCC to CPT by inducing ferroptosis through dual effects on glycolysis and glutamine metabolism. This is a novel mechanism to increase chemosensitivity, which may provide compatible chemotherapy drugs for HCC.
Collapse
Affiliation(s)
- Yuan Zeng
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Haoran Jiang
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiangting Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jun Xu
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiao Wu
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qian Xu
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weimin Cai
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Huiya Ying
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ruoru Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yingrong Ding
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Kanglei Ying
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xian Song
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhuoyan Chen
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Liuwei Zeng
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Luying Zhao
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Fujun Yu
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
11
|
Ferreira RP, Duarte JA. Protein Turnover in Skeletal Muscle: Looking at Molecular Regulation towards an Active Lifestyle. Int J Sports Med 2023; 44:763-777. [PMID: 36854391 DOI: 10.1055/a-2044-8277] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
Skeletal muscle is a highly plastic tissue, able to change its mass and functional properties in response to several stimuli. Skeletal muscle mass is influenced by the balance between protein synthesis and breakdown, which is regulated by several signaling pathways. The relative contribution of Akt/mTOR signaling, ubiquitin-proteasome pathway, autophagy among other signaling pathways to protein turnover and, therefore, to skeletal muscle mass, differs depending on the wasting or loading condition and muscle type. By modulating mitochondria biogenesis, PGC-1α has a major role in the cell's bioenergetic status and, thus, on protein turnover. In fact, rates of protein turnover regulate differently the levels of distinct protein classes in response to atrophic or hypertrophic stimuli. Mitochondrial protein turnover rates may be enhanced in wasting conditions, whereas the increased turnover of myofibrillar proteins triggers muscle mass gain. The present review aims to update the knowledge on the molecular pathways implicated in the regulation of protein turnover in skeletal muscle, focusing on how distinct muscle proteins may be modulated by lifestyle interventions with emphasis on exercise training. The comprehensive analysis of the anabolic effects of exercise programs will pave the way to the tailored management of muscle wasting conditions.
Collapse
Affiliation(s)
- Rita Pinho Ferreira
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Jose Alberto Duarte
- TOXRUN - Toxicology Research Unit, University Institute of Health Sciences, CESPU, CRL, Gandra, Portugal
- CIAFEL, Faculty of Sports, University of Porto and Laboratory for Integrative and Translational Research in Population Health (ITR), Porto, Portugal
| |
Collapse
|
12
|
Kin K, Bhogale S, Zhu L, Thomas D, Bertol J, Zheng WJ, Sinha S, Fakhouri WD. Sequence-to-expression approach to identify etiological non-coding DNA variations in P53 and cMYC-driven diseases. RESEARCH SQUARE 2023:rs.3.rs-3037310. [PMID: 37503250 PMCID: PMC10371153 DOI: 10.21203/rs.3.rs-3037310/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Background and methods Disease risk prediction based on DNA sequence and transcriptional profile can improve disease screening, prevention, and potential therapeutic approaches by revealing contributing genetic factors and altered regulatory networks. Despite identifying many disease-associated DNA variants through genome-wide association studies, distinguishing deleterious non-coding DNA variations remains poor for most common diseases. We previously reported that non-coding variations disrupting cis-overlapping motifs (CisOMs) of opposing transcription factors significantly affect enhancer activity. We designed in vitro experiments to uncover the significance of the co-occupancy and competitive binding and inhibition between P53 and cMYC on common target gene expression. Results Analyzing publicly available ChIP-seq data for P53 and cMYC in human embryonic stem cells and mouse embryonic cells showed that ~ 344-366 genomic regions are co-occupied by P53 and cMYC. We identified, on average, two CisOMs per region, suggesting that co-occupancy is evolutionarily conserved in vertebrates. Our data showed that treating U2OS cells with doxorubicin increased P53 protein level while reducing cMYC level. In contrast, no change in protein levels was observed in Raji cells. ChIP-seq analysis illustrated that 16-922 genomic regions were co-occupied by P53 and cMYC before and after treatment, and substitutions of cMYC signals by P53 were detected after doxorubicin treatment in U2OS. Around 187 expressed genes near co-occupied regions were altered at mRNA level according to RNA-seq data. We utilized a computational motif-matching approach to determine that changes in predicted P53 binding affinity by DNA variations in CisOMs of co-occupied elements significantly correlate with alterations in reporter gene expression. We performed a similar analysis using SNPs mapped in CisOMs for P53 and cMYC from ChIP-seq data in U2OS and Raji, and expression of target genes from the GTEx portal. Conclusions We found a significant correlation between change in motif-predicted cMYC binding affinity by SNPs in CisOMs and altered gene expression. Our study brings us closer to developing a generally applicable approach to filter etiological non-coding variations associated with P53 and cMYC-dependent diseases.
Collapse
Affiliation(s)
- Katherine Kin
- Department of Diagnostic and Biomedical Sciences, Center for Craniofacial Research, School of Dentistry, University of Texas Health Science Center at Houston
| | | | - Lisha Zhu
- School of Biomedical Informatics, University of Texas Health Science Center at Houston
| | - Derrick Thomas
- Department of Diagnostic and Biomedical Sciences, Center for Craniofacial Research, School of Dentistry, University of Texas Health Science Center at Houston
| | - Jessica Bertol
- Department of Diagnostic and Biomedical Sciences, Center for Craniofacial Research, School of Dentistry, University of Texas Health Science Center at Houston
| | - W Jim Zheng
- School of Biomedical Informatics, University of Texas Health Science Center at Houston
| | - Saurabh Sinha
- The Wallace H. Coulter Department of Biomedical Engineering
| | - Walid D Fakhouri
- Department of Diagnostic and Biomedical Sciences, Center for Craniofacial Research, School of Dentistry, University of Texas Health Science Center at Houston
| |
Collapse
|
13
|
Han D, Wang L, Jiang S, Yang Q. The ubiquitin-proteasome system in breast cancer. Trends Mol Med 2023:S1471-4914(23)00096-5. [PMID: 37328395 DOI: 10.1016/j.molmed.2023.05.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/12/2023] [Accepted: 05/17/2023] [Indexed: 06/18/2023]
Abstract
Ubiquitin-proteasome system (UPS) is a selective proteolytic system that is associated with the expression or function of target proteins and participates in various physiological and pathological processes of breast cancer. Inhibitors targeting the 26S proteasome in combination with other drugs have shown promising therapeutic effects in the clinical treatment of breast cancer. Moreover, several inhibitors/stimulators targeting other UPS components are also effective in preclinical studies, but have not yet been applied in the clinical treatment of breast cancer. Therefore, it is vital to comprehensively understand the functions of ubiquitination in breast cancer and to identify potential tumor promoters or tumor suppressors among UPS family members, with the aim of developing more effective and specific inhibitors/stimulators targeting specific components of this system.
Collapse
Affiliation(s)
- Dianwen Han
- Department of Breast Surgery, General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Lijuan Wang
- Pathology Tissue Bank, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Shan Jiang
- Department of Breast Surgery, General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Qifeng Yang
- Department of Breast Surgery, General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Pathology Tissue Bank, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Research Institute of Breast Cancer, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
14
|
Xu K, Liu Q, Huang W, Chu Y, Fan W, Liu J, He Y, Huang F. Promotive Effect of FBXO32 on the Odontoblastic Differentiation of Human Dental Pulp Stem Cells. Int J Mol Sci 2023; 24:ijms24097708. [PMID: 37175415 PMCID: PMC10178205 DOI: 10.3390/ijms24097708] [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: 03/17/2023] [Revised: 04/11/2023] [Accepted: 04/15/2023] [Indexed: 05/15/2023] Open
Abstract
Odontoblastic differentiation of human dental pulp stem cells (hDPSCs) is crucial for the intricate formation and repair processes in dental pulp. Until now, the literature is not able to demonstrate the role of ubiquitination in the odontoblastic differentiation of hDPSCs. This study investigated the role of F-box-only protein 32 (FBXO32), an E3 ligase, in the odontoblastic differentiation of hDPSCs. The mRNA expression profile was obtained from ribonucleic acid sequencing (RNA-Seq) data and analyzed. Immunofluorescence and immunohistochemical staining identify the FBXO32 expression in human dental pulp and hDPSCs. Small-hairpin RNA lentivirus was used for FBXO32 knockdown and overexpression. Odontoblastic differentiation of hDPSCs was determined via alkaline phosphatase activity, Alizarin Red S staining, and mRNA and protein expression levels were detected using real-time quantitative polymerase chain reaction and Western blotting. Furthermore, subcutaneous transplantation in nude mice was performed to evaluate the role of FBXO32 in mineralization in vivo using histological analysis. FBXO32 expression was upregulated in the odontoblast differentiated hDPSCs as evidenced by RNA-Seq data analysis. FBXO32 was detected in hDPSCs and the odontoblast layer of the dental pulp. Increased FBXO32 expression in hDPSCs during odontoblastic differentiation was confirmed. Through lentivirus infection method, FBXO32 downregulation in hDPSCs attenuated odontoblastic differentiation in vitro and in vivo, whereas FBXO32 upregulation promoted the hDPSCs odontoblastic differentiation, without affecting proliferation and migration. This study demonstrated, for the first time, the promotive role of FBXO32 in regulating the odontoblastic differentiation of hDPSCs, thereby providing novel insights into the regulatory mechanisms during odontoblastic differentiation in hDPSCs.
Collapse
Affiliation(s)
- Ke Xu
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| | - Qin Liu
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| | - Wushuang Huang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| | - Yanhao Chu
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| | - Wenguo Fan
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| | - Jiawei Liu
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| | - Yifan He
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| | - Fang Huang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| |
Collapse
|
15
|
Chen Z, Ren D, Lv J, Xu Y, Xie M, He X, Shi W, Qian Q, Jing A, Ma X, Qin J, Ding Y, Geng T, Ma J, Liu W, Liu S, Ji J. The atypical ubiquitin ligase RNF31 stabilizes c-Myc via epigenetic inactivation of FBXO32 nd promotes cancer development. Cell Signal 2023; 107:110677. [PMID: 37028779 DOI: 10.1016/j.cellsig.2023.110677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 03/10/2023] [Accepted: 04/03/2023] [Indexed: 04/07/2023]
Abstract
RNF31, an atypical E3 ubiquitin ligase of the RING-between-RING protein family, is one of the important components of the linear ubiquitin chain complex LUBAC. It plays a carcinogenic role in a variety of cancers by promoting cell proliferation, invasion and inhibiting apoptosis. However, the specific molecular mechanism by which RNF31 exerts its cancer-promoting effects is still unclear. By analyzing the expression profile of RNF31-depleted cancer cells, we found that loss of RNF31 significantly resulted in the inactivation of the c-Myc pathway. We further showed that RNF31 played an important role in the maintenance of c-Myc protein levels in cancer cells by extending the half-life of c-Myc protein and reducing its ubiquitination. c-Myc protein levels are tightly regulated by the ubiquitin proteasome, in which the E3 ligase FBXO32 is required to mediate its ubiquitin-dependent degradation. We found that RNF31 inhibited the transcription of FBXO32 through EZH2-mediated trimethylation of histone H3K27 in the FBXO32 promoter region, leading to the stabilization and activation of c-Myc protein. Under this circumstance, the expression of FBXO32 was significantly increased in RNF31-deficient cells, promoting the degradation of c-Myc protein, inhibiting cell proliferation and invasion, increasing cell apoptosis, and ultimately blocking the progression of tumors. Consistent with these results, the reduced malignancy phenotype caused by RNF31 deficiency could be partially reversed by overexpression of c-Myc or further knockdown of FBXO32. Together, our results reveal a key association between RNF31 and epigenetic inactivation of FBXO32 in cancer cells, and suggest that RNF31 may be a promising target for cancer therapy.
Collapse
Affiliation(s)
- Zefeng Chen
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Dexu Ren
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Jinyu Lv
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Yuxin Xu
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Mengru Xie
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Xingbei He
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Wen Shi
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Qilan Qian
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Aixin Jing
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Xinhui Ma
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Jingting Qin
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Yuanyuan Ding
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Ting Geng
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Jinming Ma
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Wei Liu
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Shunfang Liu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road 1095, Wuhan 430030, P.R. China.
| | - Jing Ji
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China.
| |
Collapse
|
16
|
Xiong Y, Wang L, Xu S, Fu B, Che Y, Zaky MY, Tian R, Yao R, Guo D, Sha Z, Lin F, Lin X, Wu H. Small molecule Z363 co-regulates TAF10 and MYC via the E3 ligase TRIP12 to suppress tumour growth. Clin Transl Med 2023; 13:e1153. [PMID: 36639831 PMCID: PMC9839843 DOI: 10.1002/ctm2.1153] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/17/2022] [Accepted: 08/12/2022] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND The MYC oncoprotein, also known as the master regulator of genes, is a transcription factor that regulates numerous physiological processes, including cell cycle control, apoptosis, protein synthesis and cell adhesion, among others. MYC is overexpressed in approximately 70% of human cancers. Given its pervasive role in cancer biology, MYC down-regulation has become an attractive cancer treatment strategy. METHODS The CRISPR/Cas9 method was used to produce KO cell models. Western blot was used to analyzed the expressions of MYC and TATA-binding proteinassociated factors 10 (TAF10) in cancer cells (MCF7, A549, HepG2 cells) Cell culture studies were performed to determine the mechanisms by which small molecules (Z363119456, Z363) affects MYC and TAF10 expressions and functions. Mouse studies were carried out to investigate the impact of Z363 regulation on tumor growth. RESULTS Z363 activate Thyroid hormone Receptor-interacting Protein 12 (TRIP12), which phosphorylates MYC at Thr58, resulting in MYC ubiquitination and degradation and thereby regulating MYC target genes. Importantly, TRIP12 also induces TAF10 degradation, which reduces MYC protein levels. TRIP12, an E3 ligase, controls MYC levels both directly and indirectly by inhibiting MYC or TAF10 activity. CONCLUSIONS In summary,these results demonstrate the anti-cancer properties of Z363, a small molecule that is co-regulated by TAF10 and MYC.
Collapse
Affiliation(s)
- Yan Xiong
- Department of Physiology, School of Life SciencesChongqing UniversityChongqingChina
| | - Lulu Wang
- Department of Physiology, School of Life SciencesChongqing UniversityChongqingChina
| | - Shiyao Xu
- Department of Physiology, School of Life SciencesChongqing UniversityChongqingChina
| | - Beibei Fu
- Department of Physiology, School of Life SciencesChongqing UniversityChongqingChina
| | - Yuchen Che
- Department of Physiology, School of Life SciencesChongqing UniversityChongqingChina
| | - Mohamed Y. Zaky
- Molecular Physiology DivisionZoology DepartmentFaculty of ScienceBeni‐Suef UniversityBeni‐SuefEgypt,Department of OncologyFaculty of MedicineLinköping UniversitySweden,Department of Biomedical and Clinical SciencesFaculty of MedicineLinköping UniversitySweden
| | - Rong Tian
- Department of Biomedical and Clinical SciencesFaculty of MedicineLinköping UniversitySweden
| | - Rui Yao
- Department of PathologyChongqing Hygeia HospitalChongqingChina
| | - Dong Guo
- Department of Physiology, School of Life SciencesChongqing UniversityChongqingChina
| | - Zhou Sha
- Department of Physiology, School of Life SciencesChongqing UniversityChongqingChina
| | - Feng Lin
- Department of Physiology, School of Life SciencesChongqing UniversityChongqingChina
| | - Xiaoyuan Lin
- Department of Physiology, School of Life SciencesChongqing UniversityChongqingChina
| | - Haibo Wu
- Department of Physiology, School of Life SciencesChongqing UniversityChongqingChina
| |
Collapse
|
17
|
Targeting CSC-related transcription factors by E3 ubiquitin ligases for cancer therapy. Semin Cancer Biol 2022; 87:84-97. [PMID: 36371028 DOI: 10.1016/j.semcancer.2022.11.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 10/07/2022] [Accepted: 11/07/2022] [Indexed: 11/10/2022]
Abstract
Evidence has revealed that transcription factors play essential roles in regulation of multiple cellular processes, including cell proliferation, metastasis, EMT, cancer stem cells and chemoresistance. Dysregulated expression levels of transcription factors contribute to tumorigenesis and malignant progression. The expression of transcription factors is tightly governed by several signaling pathways, noncoding RNAs and E3 ubiquitin ligases. Cancer stem cells (CSCs) have been validated in regulation of tumor metastasis, reoccurrence and chemoresistance in human cancer. Transcription factors have been verified to participate in regulation of CSC formation, including Oct4, SOX2, KLF4, c-Myc, Nanog, GATA, SALL4, Bmi-1, OLIG2, POU3F2 and FOX proteins. In this review article, we will describe the critical role of CSC-related transcription factors. We will further discuss which E3 ligases regulate the degradation of these CSC-related transcription factors and their underlying mechanisms. We also mentioned the functions and mechanisms of EMT-associated transcription factors such as ZEB1, ZEB2, Snail, Slug, Twist1 and Twist2. Furthermore, we highlight the therapeutic potential via targeting E3 ubiquitin ligases for modulation of these transcription factors.
Collapse
|
18
|
Androgen-Responsive Oncogenic lncRNA RP11-1023L17.1 Enhances c-Myc Protein Stability in Prostate Cancer. Int J Mol Sci 2022; 23:ijms232012219. [PMID: 36293081 PMCID: PMC9603324 DOI: 10.3390/ijms232012219] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/10/2022] [Accepted: 10/11/2022] [Indexed: 11/17/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) have been found as novel participants in the pathophysiology of prostate cancer (PCa), which is predominantly regulated by androgen and its receptor. The biological function of androgen-responsive lncRNAs remains poorly understood. Here, we identified that lncRNA RP11-1023L17.1, which is highly expressed in PCa. RP11-1023L17.1 expression, can be directly repressed by the androgen receptor in PCa cells. RP11-1023L17.1 depletion inhibited the proliferation, migration, and cell cycle progression, and promoted the apoptosis of PCa cells, indicating that RP11-1023L17.1 acts as an oncogene in PCa cells. Microarray results revealed that RP11-1023L17.1 depletion downregulated the c-Myc transcription signature in PCa cells. RP11-1023L17.1 depletion-induced cellular phenotypes can be overcome by ectopically overexpressed c-Myc. Mechanistically, RP11-1023L17.1 represses FBXO32 mRNA expression, thereby enhancing c-Myc protein stability by blocking FBXO32-mediated c-Myc degradation. Our findings reveal the previously unrecognized roles of RP11-1023L17.1 in c-Myc-dependent PCa tumorigenesis.
Collapse
|
19
|
RAI14 Promotes Melanoma Progression by Regulating the FBXO32/c-MYC Pathway. Int J Mol Sci 2022; 23:ijms231912036. [PMID: 36233342 PMCID: PMC9569902 DOI: 10.3390/ijms231912036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 11/17/2022] Open
Abstract
Melanoma originates from the malignant transformation of melanocytes. Compared with other skin cancers, melanoma has a higher fatality rate. The 5-year survival rate of patients with early-stage primary melanoma through surgical resection can reach more than 90%. However, the 5-year survival rate of patients with metastatic melanoma is only 25%. Therefore, accurate assessment of melanoma progression is critical. Previous studies have found that Retinoic Acid Induced 14(RAI14) is critical in tumorigenesis. However, the biological function of RAI14 for the development of melanoma is unclear. In this study, RAI14 is highly expressed in melanoma and correlated with prognosis. The expression of RAI14 can affect the proliferation, migration and invasion of melanoma cells. F-Box Protein 32(FBXO32) is an E3 ubiquitin ligase of c-MYC. We found that RAI14 affects the transcriptional expression of FBXO32 and regulates the stability of c-MYC. These results suggest that RAI14 play an important role in the growth of melanoma and is expected to be a therapeutic target for melanoma.
Collapse
|
20
|
FBXO32 targets PHPT1 for ubiquitination to regulate the growth of EGFR mutant lung cancer. Cell Oncol (Dordr) 2022; 45:293-307. [PMID: 35411430 DOI: 10.1007/s13402-022-00669-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Phosphohistidine phosphatase 1 (PHPT1) is an oncogene that has been reported to participate in multiple tumorigenic processes. As yet, however, the role of PHPT1 in lung cancer development remains uncharacterized. METHODS RNA sequencing assay and 18 pairs of tumor and normal tissues from patients were analyzed to reveal the upregulation of PHPT1 in lung cancer, followed by confirming the biological function in vitro and in vivo. Next, Gene Set Enrichment Analysis, lung cancer samples, apoptosis assay, mass spectrometry experiments and western blotting were used to investigate the molecular mechanism underlying PHPT1 driven progression in epidermal growth factor receptor (EGFR)-mutant lung cancer. Finally, we performed cellular and animal experiments to explore the tumor suppressive function of F-box protein 32 (FBXO32). RESULTS We found that PHPT1 is overexpressed in lung cancer patients and correlates with a poor overall survival. In addition, we found that the expression of PHPT1 is elevated in EGFR-mutant lung cancer cells and primary patient samples. Inhibition of PHPT1 expression in EGFR mutant lung cancer cells significantly decreased their proliferation and clonogenicity, and suppressed their in vitro tumor growth. Mechanistic studies revealed that activation of the ERK/MAPK pathway is driven by PHPT1. PHPT1 is required for maintaining drug resistance to erlotinib in EGFR mutant lung cancer cells. We found that FBXO32 acts as an E3 ubiquitin ligase for PHPT1, and that knockdown of FBXO32 leads to PHPT1 accumulation, activation of the ERK/MAPK pathway and promotion of the proliferation, clonogenicity and growth of lung cancer cells. CONCLUSIONS Our findings indicate that PHPT1 may serve as a biomarker and therapeutic target for acquired erlotinib resistance in lung cancer patients carrying EGFR mutations.
Collapse
|
21
|
Gong RH, Chen M, Huang C, Wong HLX, Kwan HY, Bian Z. Combination of artesunate and WNT974 induces KRAS protein degradation by upregulating E3 ligase ANACP2 and β-TrCP in the ubiquitin–proteasome pathway. Cell Commun Signal 2022; 20:34. [PMID: 35305671 PMCID: PMC8934478 DOI: 10.1186/s12964-022-00834-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 01/29/2022] [Indexed: 12/01/2022] Open
Abstract
Background KRAS mutation is one of the dominant gene mutations in colorectal cancer (CRC). Up to present, targeting KRAS for CRC treatment remains a clinical challenge. WNT974 (LGK974) is a porcupine inhibitor that interferes Wnt signaling pathway. Artesunate (ART) is a water-soluble semi-synthetic derivative of artemisinin. Methods The synergistic effect of ART and WNT974 combination in reducing CRC cell viability was determined by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. RT-PCR was utilized for the mRNA levels of KRAS, CUL7, ANAPC2, UBE2M, RNF123, SYVN1, or β-TrCP. Western blot assay was utilized for the protein levels of NRAS, HRAS, KRAS, ANAPC2, β-TrCP, GSK-3β, p-Akt (Ser473), t-Akt, p-PI3K (Tyr458), t-PI3K, p-mTOR (Ser2448), t-mTOR. Xenograft mouse model assay was performed for the anti-CRC effect of combination of ART and WNT974 in vivo. IHC assay was utilized for the levels of KRAS, β-TrCP, GSK-3β or ANAPC2 in tumor tissues. Results Our study shows that the combination of WNT974 and ART exhibits synergistic effect in reducing CRC growth. The combination treatment significantly reduces KRAS protein level and activity in CRC cells. Interestingly, the combination treatment increases E3 ligases ANAPC2 expression. Our data show that overexpression of ANAPC2 significantly reduces KRAS protein levels, which is reversed by MG132. Knockdown of ANAPC2 in CRC abolishes the combination treatment-reduce KRAS expression. Besides, the treatment also increases the expressions of GSK-3β and E3 ligase β-TrCP that is known to degrade GSK-3β-phosphorylated KRAS protein. Knockdown of β-TrCP- and inhibition of GSK-3β abolish the combination treatment-induce KRAS ubiquitination and reduction in expression. Last but not least, combination treatment suppresses PI3K/Akt/m-TOR signaling pathway. Conclusions Our data clearly show that the combination treatment significantly enhances KRAS protein degradation via the ubiquitination ubiquitin–proteasome pathway, which is also demonstrated in xenograft mouse model. The study provides strong scientific evidence for the development of the combination of WNT974 and ART as KRAS-targeting therapeutics for CRC treatment. Video Abstract
Supplementary Information The online version contains supplementary material available at 10.1186/s12964-022-00834-2.
Collapse
|
22
|
Nagasaka M, Inoue Y, Yoshida M, Miyajima C, Morishita D, Tokugawa M, Nakamoto H, Sugano M, Ohoka N, Hayashi H. The deubiquitinating enzyme USP17 regulates c‐Myc levels and controls cell proliferation and glycolysis. FEBS Lett 2022; 596:465-478. [DOI: 10.1002/1873-3468.14296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/09/2022] [Accepted: 01/12/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Mai Nagasaka
- Department of Cell Signaling Graduate School of Pharmaceutical Sciences Nagoya City University 467‐8603 Nagoya Japan
| | - Yasumichi Inoue
- Department of Cell Signaling Graduate School of Pharmaceutical Sciences Nagoya City University 467‐8603 Nagoya Japan
- Department of Innovative Therapeutics Sciences Cooperative Major in Nanopharmaceutical Sciences Graduate School of Pharmaceutical Sciences Nagoya City University 467‐8603 Nagoya Japan
| | - Manaka Yoshida
- Department of Cell Signaling Graduate School of Pharmaceutical Sciences Nagoya City University 467‐8603 Nagoya Japan
| | - Chiharu Miyajima
- Department of Cell Signaling Graduate School of Pharmaceutical Sciences Nagoya City University 467‐8603 Nagoya Japan
- Department of Innovative Therapeutics Sciences Cooperative Major in Nanopharmaceutical Sciences Graduate School of Pharmaceutical Sciences Nagoya City University 467‐8603 Nagoya Japan
| | - Daisuke Morishita
- Department of Cell Signaling Graduate School of Pharmaceutical Sciences Nagoya City University 467‐8603 Nagoya Japan
- Chordia Therapeutics Inc 251‐0012 Kanagawa Japan
| | - Muneshige Tokugawa
- Department of Cell Signaling Graduate School of Pharmaceutical Sciences Nagoya City University 467‐8603 Nagoya Japan
| | - Haruna Nakamoto
- Department of Cell Signaling Graduate School of Pharmaceutical Sciences Nagoya City University 467‐8603 Nagoya Japan
| | - Mayumi Sugano
- Department of Cell Signaling Graduate School of Pharmaceutical Sciences Nagoya City University 467‐8603 Nagoya Japan
| | - Nobumichi Ohoka
- Division of Molecular Target and Gene Therapy Products National Institute of Health Sciences 210‐9501 Kanagawa Japan
| | - Hidetoshi Hayashi
- Department of Cell Signaling Graduate School of Pharmaceutical Sciences Nagoya City University 467‐8603 Nagoya Japan
- Department of Innovative Therapeutics Sciences Cooperative Major in Nanopharmaceutical Sciences Graduate School of Pharmaceutical Sciences Nagoya City University 467‐8603 Nagoya Japan
| |
Collapse
|
23
|
Islam S, Dutta P, Sahay O, Gopalakrishnan K, Roy Muhury S, Parameshwar P, Shetty P, Santra MK. Feedback-regulated transcriptional repression of FBXO31 by c-Myc triggers ovarian cancer tumorigenesis. Int J Cancer 2021; 150:1512-1524. [PMID: 34706096 DOI: 10.1002/ijc.33854] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 07/30/2021] [Accepted: 09/02/2021] [Indexed: 11/08/2022]
Abstract
FBXO31, a member of F-box protein family, has been shown to play an important role in preventing tumorigenesis by preserving genomic stability during cell proliferations as well as upon genotoxic stresses. Inactivation of FBXO31 due to loss of heterozygosity is associated with various cancers, including ovarian cancer, one of the deadliest forms of gynecological cancers. However, the role and regulation of FBXO31 in ovarian cancer remained elusive. Here, using biochemical and molecular biology techniques, we show that c-Myc suppresses the mRNA levels of FBXO31 in ovarian cancer cell lines and mouse model. Chromatin immunoprecipitation experiment showed that c-Myc is recruited to the promoter region of FBXO31 and prevents FBXO31 mRNA synthesis. In contrast, FBXO31 maintains the c-Myc expression at an optimum through proteasome pathway. FBXO31 interacts with and facilitates the polyubiquitination of c-Myc through the SCF complex and thereby inhibits ovarian cancer growth both in vitro and in vivo. Moreover, FBXO31-mediated proteasomal degradation of c-Myc is unique. Unlike other negative regulators, FBXO31 recognizes c-Myc in phosphorylation independent manner to direct its degradation. Further, expression levels analysis revealed that c-Myc and FBXO31 share a converse correlation of expression in ovarian cancer cell lines and patient samples. We observed an increase in the expression levels of c-Myc with a concomitant decrease in the levels of FBXO31 in higher grades of ovarian cancer patient samples. In conclusion, our study demonstrated that oncogene c-Myc impairs the tumor-suppressive functions of FBXO31 to promote ovarian cancer progression, and therefore c-Myc-FBXO31 axis can be explored to develop better cancer therapy. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Sehbanul Islam
- National Centre for Cell Science, NCCS Complex, Ganeshkhind Road, Pune, Maharashtra, India.,Department of Biotechnology, Savitribai Phule Pune University, Pune, Maharashtra, India
| | - Parul Dutta
- National Centre for Cell Science, NCCS Complex, Ganeshkhind Road, Pune, Maharashtra, India.,Department of Biotechnology, Savitribai Phule Pune University, Pune, Maharashtra, India
| | - Osheen Sahay
- National Centre for Cell Science, NCCS Complex, Ganeshkhind Road, Pune, Maharashtra, India.,Department of Biotechnology, Savitribai Phule Pune University, Pune, Maharashtra, India
| | - K Gopalakrishnan
- National Centre for Cell Science, NCCS Complex, Ganeshkhind Road, Pune, Maharashtra, India
| | - Sushrita Roy Muhury
- National Centre for Cell Science, NCCS Complex, Ganeshkhind Road, Pune, Maharashtra, India
| | - Parinitha Parameshwar
- Department of Pathology, SDM College of Medical Sciences & Hospital, Sattur, Dharwad, India
| | - Praveenkumar Shetty
- K. S. Hegde Medical Academy, NITTE (Deemed to be University), University Enclave, Medical Sciences Complex, Dheralakatte, Mangalore, India
| | - Manas Kumar Santra
- National Centre for Cell Science, NCCS Complex, Ganeshkhind Road, Pune, Maharashtra, India
| |
Collapse
|
24
|
Sun Z, Liu X, Chen M, Zhang H, Zeng X. Overexpression of RNF126 is associated with poor prognosis and contributes to the progression of lung adenocarcinoma. Biomark Med 2021; 15:1345-1355. [PMID: 34533052 DOI: 10.2217/bmm-2020-0798] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Aim: To document the expression and function of RNF126 in lung adenocarcinoma (LAD). Materials & methods: A total of 102 LAD patients were enrolled in this retrospective study. The mRNA and protein levels of RNF126 were tested via RT-qPCR and immunohistochemical staining, respectively. Univariate and multivariate analyses were conducted to exploring the clinical significance of RNF126 in the prognosis. Knockdown and overexpression strategies were used to validate the tumor-related roles of RNF126 both in vitro and in vivo. Results: RNF126 was highly expressed and was an independent predictor of a poor prognosis for LAD patients. We also revealed that RNF126 knockdown suppressed proliferation of LAD cells and xenografts. Conclusion: RNF126 is a potential survival predictor and therapeutic target for LAD.
Collapse
Affiliation(s)
- Zhaona Sun
- Department of Cardiology, Yidu Central Hospital of Weifang, Weifang, 262500, China
| | - Xin Liu
- Department of Emergency Medicine, Yidu Central Hospital of Weifang, Weifang, 262500, China
| | - Meiyuan Chen
- Department of Hepatobiliary Surgery, Yidu Central Hospital of Weifang, Weifang, 262500, China
| | - Hong Zhang
- Second Ward of Tuberculosis Medicine, Linyi People's Hospital, Linyi, 276000, China
| | - Xiancong Zeng
- Department of Respiratory Medicine, Shiyan People's Hospital, Shiyan, 442000, China
| |
Collapse
|
25
|
Zhu L, Chen Z, Zang H, Fan S, Gu J, Zhang G, Sun KDY, Wang Q, He Y, Owonikoko TK, Ramalingam SS, Sun SY. Targeting c-Myc to Overcome Acquired Resistance of EGFR Mutant NSCLC Cells to the Third-Generation EGFR Tyrosine Kinase Inhibitor, Osimertinib. Cancer Res 2021; 81:4822-4834. [PMID: 34289988 PMCID: PMC8448971 DOI: 10.1158/0008-5472.can-21-0556] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 06/15/2021] [Accepted: 07/20/2021] [Indexed: 12/24/2022]
Abstract
Osimertinib (AZD9291 or TAGRISSO) is a promising and approved third-generation EGFR tyrosine kinase inhibitor (TKI) for treating patients with advanced non-small cell lung cancer (NSCLC) harboring EGFR-activating mutations or the resistant T790M mutation. However, the inevitable emergence of acquired resistance limits its long-term efficacy. A fuller understanding of the mechanism of action of osimertinib and its linkage to acquired resistance will enable the development of more efficacious therapeutic strategies. Consequently, we have identified a novel connection between osimertinib or other EGFR-TKIs and c-Myc. Osimertinib rapidly and sustainably decreased c-Myc levels primarily via enhancing protein degradation in EGFR-mutant (EGFRm) NSCLC cell lines and xenograft tumors. c-Myc levels were substantially elevated in different EGFRm NSCLC cell lines with acquired resistance to osimertinib in comparison with their corresponding parental cell lines and could not be reduced any further by osimertinib. Consistently, c-Myc levels were elevated in the majority of EGFRm NSCLC tissues relapsed from EGFR-TKI treatment compared with their corresponding untreated baseline c-Myc levels. Suppression of c-Myc through knockdown or pharmacologic targeting with BET inhibitors restored the response of resistant cell lines to osimertinib. These findings indicate that c-Myc modulation mediates the therapeutic efficacy of osimertinib and the development of osimertinib acquired resistance. Furthermore, they establish c-Myc as a potential therapeutic target and warrant clinical testing of BET inhibition as a potential strategy to overcome acquired resistance to osimertinib or other EGFR inhibitors. SIGNIFICANCE: This study demonstrates a critical role of c-Myc modulation in mediating therapeutic efficacy of osimertinib including osimertinib acquired resistance and suggests targeting c-Myc as a potential strategy to overcome osimertinib acquired resistance.
Collapse
Affiliation(s)
- Lei Zhu
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia
| | - Zhen Chen
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia
| | - Hongjing Zang
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Songqing Fan
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Jiajia Gu
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia
| | - Guojing Zhang
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia
| | - Kevin D-Y Sun
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia
| | - Qiming Wang
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Yong He
- Department of Respiratory Disease, Daping Hospital, Chongqing, China
| | - Taofeek K Owonikoko
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia
| | - Suresh S Ramalingam
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia
| | - Shi-Yong Sun
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia.
| |
Collapse
|
26
|
Wang H, Liu H, Zhao L, Luo S, Akinyemiju T, Hwang S, Yue Y, Wei Q. Association of genetic variants of FBXO32 and FOXO6 in the FOXO pathway with breast cancer risk. Mol Carcinog 2021; 60:661-670. [PMID: 34197655 DOI: 10.1002/mc.23331] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 05/26/2021] [Accepted: 06/06/2021] [Indexed: 01/10/2023]
Abstract
Forkhead box class O (FOXO) transcription factors play a pivotal role in regulating a variety of biological processes, including organismal development, cell signaling, cell metabolism, and tumorigenesis. Therefore, we hypothesize that genetic variants in FOXO pathway genes are associated with breast cancer (BC) risk. To test this hypothesis, we conducted a large meta-analysis using 14 published genome-wide association study (GWAS) data sets in the Discovery, Biology, and Risk of Inherited Variants in Breast Cancer (DRIVE) study. We assessed associations between 5214 (365 genotyped in DRIVE and 4849 imputed) common single-nucleotide polymorphisms (SNPs) in 55 FOXO pathway genes and BC risk. After multiple comparison corrections by the Bayesian false-discovery probability method, we found five SNPs to be significantly associated with BC risk. In stepwise multivariate logistic regression analysis with adjustment for age, principal components, and previously published SNPs in the same data set, three independent SNPs (i.e., FBXO32 rs10093411 A>G, FOXO6 rs61229336 C>T, and FBXO32 rs62521280 C>T) remained to be significantly associated with BC risk (p = 0.0008, 0.0011, and 0.0017, respectively). Additional expression quantitative trait loci analysis revealed that the FBXO32 rs62521280 T allele was associated with decreased messenger RNA (mRNA) expression levels in breast tissue, while the FOXO6 rs61229336 T allele was found to be associated with decreased mRNA expression levels in the whole blood cells. Once replicated by other investigators, these genetic variants may serve as new biomarkers for BC risk.
Collapse
Affiliation(s)
- Haijiao Wang
- Department of Gynecology Oncology, The First Hospital of Jilin University, Changchun, Jilin, China.,Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina, USA.,Department of Population Health Sciences, Duke University School of Medicine, Durham, North Carolina, USA
| | - Hongliang Liu
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina, USA.,Department of Population Health Sciences, Duke University School of Medicine, Durham, North Carolina, USA
| | - Lingling Zhao
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina, USA.,Department of Population Health Sciences, Duke University School of Medicine, Durham, North Carolina, USA.,Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Sheng Luo
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Tomi Akinyemiju
- Department of Population Health Sciences, Duke University School of Medicine, Durham, North Carolina, USA
| | - Shelley Hwang
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Ying Yue
- Department of Gynecology Oncology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Qingyi Wei
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina, USA.,Department of Population Health Sciences, Duke University School of Medicine, Durham, North Carolina, USA.,Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA.,Duke Global Health Institute, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
27
|
Sun XX, Li Y, Sears RC, Dai MS. Targeting the MYC Ubiquitination-Proteasome Degradation Pathway for Cancer Therapy. Front Oncol 2021; 11:679445. [PMID: 34178666 PMCID: PMC8226175 DOI: 10.3389/fonc.2021.679445] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/24/2021] [Indexed: 12/23/2022] Open
Abstract
Deregulated MYC overexpression and activation contributes to tumor growth and progression. Given the short half-life and unstable nature of the MYC protein, it is not surprising that the oncoprotein is highly regulated via diverse posttranslational mechanisms. Among them, ubiquitination dynamically controls the levels and activity of MYC during normal cell growth and homeostasis, whereas the disturbance of the ubiquitination/deubiquitination balance enables unwanted MYC stabilization and activation. In addition, MYC is also regulated by SUMOylation which crosstalks with the ubiquitination pathway and controls MYC protein stability and activity. In this mini-review, we will summarize current updates regarding MYC ubiquitination and provide perspectives about these MYC regulators as potential therapeutic targets in cancer.
Collapse
Affiliation(s)
- Xiao-Xin Sun
- Department of Molecular & Medical Genetics, School of Medicine and the OHSU Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| | - Yanping Li
- Department of Molecular & Medical Genetics, School of Medicine and the OHSU Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| | - Rosalie C Sears
- Department of Molecular & Medical Genetics, School of Medicine and the OHSU Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| | - Mu-Shui Dai
- Department of Molecular & Medical Genetics, School of Medicine and the OHSU Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
28
|
The Role of GSK-3β in the Regulation of Protein Turnover, Myosin Phenotype, and Oxidative Capacity in Skeletal Muscle under Disuse Conditions. Int J Mol Sci 2021; 22:ijms22105081. [PMID: 34064895 PMCID: PMC8151958 DOI: 10.3390/ijms22105081] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/03/2021] [Accepted: 05/10/2021] [Indexed: 12/12/2022] Open
Abstract
Skeletal muscles, being one of the most abundant tissues in the body, are involved in many vital processes, such as locomotion, posture maintenance, respiration, glucose homeostasis, etc. Hence, the maintenance of skeletal muscle mass is crucial for overall health, prevention of various diseases, and contributes to an individual’s quality of life. Prolonged muscle inactivity/disuse (due to limb immobilization, mechanical ventilation, bedrest, spaceflight) represents one of the typical causes, leading to the loss of muscle mass and function. This disuse-induced muscle loss primarily results from repressed protein synthesis and increased proteolysis. Further, prolonged disuse results in slow-to-fast fiber-type transition, mitochondrial dysfunction and reduced oxidative capacity. Glycogen synthase kinase 3β (GSK-3β) is a key enzyme standing at the crossroads of various signaling pathways regulating a wide range of cellular processes. This review discusses various important roles of GSK-3β in the regulation of protein turnover, myosin phenotype, and oxidative capacity in skeletal muscles under disuse/unloading conditions and subsequent recovery. According to its vital functions, GSK-3β may represent a perspective therapeutic target in the treatment of muscle wasting induced by chronic disuse, aging, and a number of diseases.
Collapse
|
29
|
Bedada FB, Ntekim OE, Nwulia EO, Fungwe TV, Nadarajah SR, Obisesan TO. Exercise Training-Increased FBXO32 and FOXO1 in a Gender-Dependent Manner in Mild Cognitively Impaired African Americans: GEMS-1 Study. Front Aging Neurosci 2021; 13:641758. [PMID: 33935685 PMCID: PMC8079639 DOI: 10.3389/fnagi.2021.641758] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/18/2021] [Indexed: 01/29/2023] Open
Abstract
The ubiquitin proteasome system (UPS) and FOXOs transcription factors play a pivotal role in cellular clearance and minimizing the accumulation of Aβ in neurodegeneration (ND). In African Americans (AAs) with mild cognitive impairment (MCI), the role of components of UPS and FOXOs; and whether they are amenable to exercise effects is unknown. We hypothesized that exercise can enhance cellular clearance systems during aging and ND by increasing expressions of FBXO32 and FOXO1. To test this hypothesis, we used TaqMan gene expression analysis in peripheral blood (PB) to investigate the component of UPS and FOXOs; and provide mechanistic insight at baseline, during exercise, and in both genders. At baseline, levels of FBXO32 were higher in women than in men. In our attempt to discern gender-specific exercise-related changes, we observed that levels of FBXO32 increased in men but not in women. Similarly, levels of FOXO1 increased in men only. These data suggest that a graded dose of FBXO32 and FOXO1 may be beneficial when PB cells carrying FBXO32 and FOXO1 summon into the brain in response to Alzheimer's disease (AD) perturbation (docking station PB cells). Our observation is consistent with emerging studies that exercise allows the trafficking of blood factors. Given the significance of FBXO32 and FOXO1 to ND and associated muscle integrity, our findings may explain, at least in part, the benefits of exercise on memory, associated gait, and balance perturbation acknowledged to herald the emergence of MCI.
Collapse
Affiliation(s)
- Fikru B. Bedada
- Department of Clinical Laboratory Sciences, College of Nursing and Allied Health Sciences, Howard University, Washington, DC, United States
| | - Oyonumo E. Ntekim
- Department of Nutritional Sciences, College of Nursing and Allied Health Sciences, Howard University, Washington, DC, United States
| | - Evaristus O. Nwulia
- Department of Psychiatry, Howard University Hospital, Washington, DC, United States
| | - Thomas V. Fungwe
- Department of Nutritional Sciences, College of Nursing and Allied Health Sciences, Howard University, Washington, DC, United States
| | - Sheeba Raaj Nadarajah
- Department of Nursing, College of Nursing and Allied Health Sciences, Howard University, Washington, DC, United States
| | - Thomas O. Obisesan
- Division of Geriatrics, Department of Medicine, Howard University Hospital, Washington, DC, United States
| |
Collapse
|
30
|
Schukur L, Zimmermann T, Niewoehner O, Kerr G, Gleim S, Bauer-Probst B, Knapp B, Galli GG, Liang X, Mendiola A, Reece-Hoyes J, Rapti M, Barbosa I, Reschke M, Radimerski T, Thoma CR. Identification of the HECT E3 ligase UBR5 as a regulator of MYC degradation using a CRISPR/Cas9 screen. Sci Rep 2020; 10:20044. [PMID: 33208877 PMCID: PMC7676242 DOI: 10.1038/s41598-020-76960-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 11/02/2020] [Indexed: 02/06/2023] Open
Abstract
MYC oncoprotein is a multifunctional transcription factor that regulates the expression of a large number of genes involved in cellular growth, proliferation and metabolism. Altered MYC protein level lead to cellular transformation and tumorigenesis. MYC is deregulated in > 50% of human cancers, rendering it an attractive drug target. However, direct inhibition of this class of proteins using conventional small molecules is challenging due to their intrinsically disordered state. To discover novel posttranslational regulators of MYC protein stability and turnover, we established a genetic screen in mammalian cells by combining a fluorescent protein-based MYC abundance sensor, CRISPR/Cas9-based gene knockouts and next-generation sequencing. Our screen identifies UBR5, an E3 ligase of the HECT-type family, as a novel regulator of MYC degradation. Even in the presence of the well-described and functional MYC ligase, FBXW7, UBR5 depletion leads to accumulation of MYC in cells. We demonstrate interaction of UBR5 with MYC and reduced K48-linked ubiquitination of MYC upon loss of UBR5 in cells. Interestingly, in cancer cell lines with amplified MYC expression, depletion of UBR5 resulted in reduced cell survival, as a consequence of MYC stabilization. Finally, we show that MYC and UBR5 are co-amplified in more than 40% of cancer cells and that MYC copy number amplification correlates with enhanced transcriptional output of UBR5. This suggests that UBR5 acts as a buffer in MYC amplified settings and protects these cells from apoptosis.
Collapse
Affiliation(s)
- Lina Schukur
- Novartis Institutes for Biomedical Research (NIBR) Oncology, Novartis, Basel, Switzerland. .,Pharmaceutical Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland.
| | - Tamara Zimmermann
- Novartis Institutes for Biomedical Research (NIBR) Oncology, Novartis, Basel, Switzerland
| | - Ole Niewoehner
- NIBR Chemical Biology and Therapeutics, Novartis, Cambridge, USA
| | - Grainne Kerr
- Novartis Institutes for Biomedical Research (NIBR) Oncology, Novartis, Basel, Switzerland
| | - Scott Gleim
- NIBR Chemical Biology and Therapeutics, Novartis, Cambridge, USA
| | - Beatrice Bauer-Probst
- Novartis Institutes for Biomedical Research (NIBR) Oncology, Novartis, Basel, Switzerland
| | - Britta Knapp
- NIBR Chemical Biology and Therapeutics, Novartis, Cambridge, USA
| | - Giorgio G Galli
- Novartis Institutes for Biomedical Research (NIBR) Oncology, Novartis, Basel, Switzerland
| | - Xiaoyou Liang
- NIBR Chemical Biology and Therapeutics, Novartis, Cambridge, USA
| | - Angelica Mendiola
- Genomics Institute of the Novartis Research Foundation, LaJolla, USA
| | - John Reece-Hoyes
- NIBR Chemical Biology and Therapeutics, Novartis, Cambridge, USA
| | - Melivoia Rapti
- Novartis Institutes for Biomedical Research (NIBR) Oncology, Novartis, Basel, Switzerland
| | - Ines Barbosa
- Novartis Institutes for Biomedical Research (NIBR) Oncology, Novartis, Basel, Switzerland
| | - Markus Reschke
- Novartis Institutes for Biomedical Research (NIBR) Oncology, Novartis, Basel, Switzerland
| | - Thomas Radimerski
- Novartis Institutes for Biomedical Research (NIBR) Oncology, Novartis, Basel, Switzerland.,Basilea Pharmaceutica, Basel, Switzerland
| | - Claudio R Thoma
- NIBR Chemical Biology and Therapeutics, Novartis, Cambridge, USA.
| |
Collapse
|
31
|
Mirzoev TM. Skeletal Muscle Recovery from Disuse Atrophy: Protein Turnover Signaling and Strategies for Accelerating Muscle Regrowth. Int J Mol Sci 2020; 21:ijms21217940. [PMID: 33114683 PMCID: PMC7663166 DOI: 10.3390/ijms21217940] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/08/2020] [Accepted: 10/23/2020] [Indexed: 12/15/2022] Open
Abstract
Skeletal muscle fibers have a unique capacity to adjust their metabolism and phenotype in response to alternations in mechanical loading. Indeed, chronic mechanical loading leads to an increase in skeletal muscle mass, while prolonged mechanical unloading results in a significant decrease in muscle mass (muscle atrophy). The maintenance of skeletal muscle mass is dependent on the balance between rates of muscle protein synthesis and breakdown. While molecular mechanisms regulating protein synthesis during mechanical unloading have been relatively well studied, signaling events implicated in protein turnover during skeletal muscle recovery from unloading are poorly defined. A better understanding of the molecular events that underpin muscle mass recovery following disuse-induced atrophy is of significant importance for both clinical and space medicine. This review focuses on the molecular mechanisms that may be involved in the activation of protein synthesis and subsequent restoration of muscle mass after a period of mechanical unloading. In addition, the efficiency of strategies proposed to improve muscle protein gain during recovery is also discussed.
Collapse
Affiliation(s)
- Timur M Mirzoev
- Myology Laboratory, Institute of Biomedical Problems RAS, Moscow 123007, Russia
| |
Collapse
|
32
|
Li Z, Fan S, Wang J, Chen X, Liao Q, Liu X, Ouyang G, Cao H, Xiao W. Zebrafish F-box Protein fbxo3 Negatively Regulates Antiviral Response through Promoting K27-Linked Polyubiquitination of the Transcription Factors irf3 and irf7. THE JOURNAL OF IMMUNOLOGY 2020; 205:1897-1908. [DOI: 10.4049/jimmunol.2000305] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 07/29/2020] [Indexed: 12/23/2022]
|
33
|
Cai EY, Kufeld MN, Schuster S, Arora S, Larkin M, Germanos AA, Hsieh AC, Beronja S. Selective Translation of Cell Fate Regulators Mediates Tolerance to Broad Oncogenic Stress. Cell Stem Cell 2020; 27:270-283.e7. [PMID: 32516567 PMCID: PMC7993921 DOI: 10.1016/j.stem.2020.05.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 03/13/2020] [Accepted: 05/13/2020] [Indexed: 12/23/2022]
Abstract
Human skin tolerates a surprisingly high burden of oncogenic lesions. Although adult epidermis can suppress the expansion of individual mutant clones, the mechanisms behind tolerance to oncogene activation across broader regions of tissue are unclear. Here, we uncover a dynamic translational mechanism that coordinates oncogenic HRAS-induced hyperproliferation with loss of progenitor self-renewal to restrain aberrant growth and tumorigenesis. We identify translation initiator eIF2B5 as a central co-regulator of HRAS proliferation and cell fate choice. By coupling in vivo ribosome profiling with genetic screening, we provide direct evidence that oncogene-induced loss of progenitor self-renewal is driven by eIF2B5-mediated translation of ubiquitination genes. Ubiquitin ligase FBXO32 specifically inhibits epidermal renewal without affecting overall proliferation, thus restraining HRAS-driven tumorigenesis while maintaining normal tissue growth. Thus, oncogene-driven translation is not necessarily inherently tumor promoting but instead can manage widespread oncogenic stress by steering progenitor fate to prolong normal tissue growth.
Collapse
Affiliation(s)
- Elise Y Cai
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Medical Scientist Training Program, University of Washington, Seattle, WA 98195, USA; Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA 98195, USA
| | - Megan N Kufeld
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Samantha Schuster
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA 98195, USA
| | - Sonali Arora
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Madeline Larkin
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Alexandre A Germanos
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA 98195, USA
| | - Andrew C Hsieh
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Slobodan Beronja
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| |
Collapse
|
34
|
Yumimoto K, Yamauchi Y, Nakayama KI. F-Box Proteins and Cancer. Cancers (Basel) 2020; 12:cancers12051249. [PMID: 32429232 PMCID: PMC7281081 DOI: 10.3390/cancers12051249] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/09/2020] [Accepted: 05/12/2020] [Indexed: 12/20/2022] Open
Abstract
Controlled protein degradation is essential for the operation of a variety of cellular processes including cell division, growth, and differentiation. Identification of the relations between ubiquitin ligases and their substrates is key to understanding the molecular basis of cancer development and to the discovery of novel targets for cancer therapeutics. F-box proteins function as the substrate recognition subunits of S-phase kinase-associated protein 1 (SKP1)−Cullin1 (CUL1)−F-box protein (SCF) ubiquitin ligase complexes. Here, we summarize the roles of specific F-box proteins that have been shown to function as tumor promoters or suppressors. We also highlight proto-oncoproteins that are targeted for ubiquitylation by multiple F-box proteins, and discuss how these F-box proteins are deployed to regulate their cognate substrates in various situations.
Collapse
|
35
|
The Impact of Transcription Factor Prospero Homeobox 1 on the Regulation of Thyroid Cancer Malignancy. Int J Mol Sci 2020; 21:ijms21093220. [PMID: 32370142 PMCID: PMC7247360 DOI: 10.3390/ijms21093220] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 04/30/2020] [Accepted: 04/30/2020] [Indexed: 12/11/2022] Open
Abstract
Transcription factor Prospero homeobox 1 (PROX1) is continuously expressed in the lymphatic endothelial cells, playing an essential role in their differentiation. Many reports have shown that PROX1 is implicated in cancer development and acts as an oncoprotein or suppressor in a tissue-dependent manner. Additionally, the PROX1 expression in many types of tumors has prognostic significance and is associated with patient outcomes. In our previous experimental studies, we showed that PROX1 is present in the thyroid cancer (THC) cells of different origins and has a high impact on follicular thyroid cancer (FTC) phenotypes, regulating migration, invasion, focal adhesion, cytoskeleton reorganization, and angiogenesis. Herein, we discuss the PROX1 transcript and protein structures, the expression pattern of PROX1 in THC specimens, and its epigenetic regulation. Next, we emphasize the biological processes and genes regulated by PROX1 in CGTH-W-1 cells, derived from squamous cell carcinoma of the thyroid gland. Finally, we discuss the interaction of PROX1 with other lymphatic factors. In our review, we aimed to highlight the importance of vascular molecules in cancer development and provide an update on the functionality of PROX1 in THC biology regulation.
Collapse
|
36
|
Deng L, Meng T, Chen L, Wei W, Wang P. The role of ubiquitination in tumorigenesis and targeted drug discovery. Signal Transduct Target Ther 2020; 5:11. [PMID: 32296023 PMCID: PMC7048745 DOI: 10.1038/s41392-020-0107-0] [Citation(s) in RCA: 475] [Impact Index Per Article: 95.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/12/2019] [Accepted: 12/17/2019] [Indexed: 02/08/2023] Open
Abstract
Ubiquitination, an important type of protein posttranslational modification (PTM), plays a crucial role in controlling substrate degradation and subsequently mediates the "quantity" and "quality" of various proteins, serving to ensure cell homeostasis and guarantee life activities. The regulation of ubiquitination is multifaceted and works not only at the transcriptional and posttranslational levels (phosphorylation, acetylation, methylation, etc.) but also at the protein level (activators or repressors). When regulatory mechanisms are aberrant, the altered biological processes may subsequently induce serious human diseases, especially various types of cancer. In tumorigenesis, the altered biological processes involve tumor metabolism, the immunological tumor microenvironment (TME), cancer stem cell (CSC) stemness and so on. With regard to tumor metabolism, the ubiquitination of some key proteins such as RagA, mTOR, PTEN, AKT, c-Myc and P53 significantly regulates the activity of the mTORC1, AMPK and PTEN-AKT signaling pathways. In addition, ubiquitination in the TLR, RLR and STING-dependent signaling pathways also modulates the TME. Moreover, the ubiquitination of core stem cell regulator triplets (Nanog, Oct4 and Sox2) and members of the Wnt and Hippo-YAP signaling pathways participates in the maintenance of CSC stemness. Based on the altered components, including the proteasome, E3 ligases, E1, E2 and deubiquitinases (DUBs), many molecular targeted drugs have been developed to combat cancer. Among them, small molecule inhibitors targeting the proteasome, such as bortezomib, carfilzomib, oprozomib and ixazomib, have achieved tangible success. In addition, MLN7243 and MLN4924 (targeting the E1 enzyme), Leucettamol A and CC0651 (targeting the E2 enzyme), nutlin and MI-219 (targeting the E3 enzyme), and compounds G5 and F6 (targeting DUB activity) have also shown potential in preclinical cancer treatment. In this review, we summarize the latest progress in understanding the substrates for ubiquitination and their special functions in tumor metabolism regulation, TME modulation and CSC stemness maintenance. Moreover, potential therapeutic targets for cancer are reviewed, as are the therapeutic effects of targeted drugs.
Collapse
Affiliation(s)
- Lu Deng
- College of Animal Science and Technology, Northwest A&F University, Yangling Shaanxi, 712100, China.
| | - Tong Meng
- Division of Spine, Department of Orthopedics, Tongji Hospital Affiliated to Tongji University School of Medicine, 389 Xincun Road, Shanghai, China
| | - Lei Chen
- Division of Laboratory Safety and Services, Northwest A&F University, Yangling Shaanxi, 712100, China
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Ping Wang
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
37
|
Yan H, Jiang J, Du A, Gao J, Zhang D, Song L. Genistein Enhances Radiosensitivity of Human Hepatocellular Carcinoma Cells by Inducing G2/M Arrest and Apoptosis. Radiat Res 2020; 193:286-300. [PMID: 32017668 DOI: 10.1667/rr15380.1] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
New radiosensitizers are urgently needed for radiation therapy patients with localized hepatocellular carcinoma (HCC) that is refractory to radical surgery. We previously found that genistein, a major soy isoflavone, exerts radioprotective effects on L-02 normal liver cells at low concentrations. Here, we report that 5 µM genistein shows less harm to L-02 cells than HCC cells and that it significantly enhances the radiosensitivity of HCC cells by enhancing DNA damage, chromosomal aberrations and cell cycle arrest at G2/M phase and by exacerbating apoptosis. Mechanistically, genistein aggravates X-ray-induced decreases in the levels of phospho-Bad (Ser136) but enhances the levels of phospho-Chk2 (Thr68), phospho-ATM (Ser1981) and γ-H2AX. Micro-array analysis indicated that downregulation of POU6F and CCNE2 expression and upregulation of FBXO32 and cyclin B1 expression might play vital roles in genistein-induced radiosensitivity. These findings suggest genistein as an interesting candidate for adjuvant radiotherapy for HCC and indicate that genistein causes less harm to normal cells than HCC cells by inducing G2/M arrest and apoptosis.
Collapse
Affiliation(s)
- Hongli Yan
- Department of Laboratory Medicine, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Jing Jiang
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiaotong University, Shanghai 200240, China
| | - Aiying Du
- Department of Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Science, Shanghai, China
| | - Jinli Gao
- Department of Pathology, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Daisong Zhang
- Department of Surgery, Penglai People's Hospital, Penglai 265600, Shandong Province, China
| | - Lihua Song
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiaotong University, Shanghai 200240, China
| |
Collapse
|
38
|
Chen Y, Sun XX, Sears RC, Dai MS. Writing and erasing MYC ubiquitination and SUMOylation. Genes Dis 2019; 6:359-371. [PMID: 31832515 PMCID: PMC6889025 DOI: 10.1016/j.gendis.2019.05.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 05/23/2019] [Accepted: 05/29/2019] [Indexed: 12/22/2022] Open
Abstract
The transcription factor c-MYC (MYC thereafter) controls diverse transcription programs and plays a key role in the development of many human cancers. Cells develop multiple mechanisms to ensure that MYC levels and activity are precisely controlled in normal physiological context. As a short half-lived protein, MYC protein levels are tightly regulated by the ubiquitin proteasome system. Over a dozen of ubiquitin ligases have been found to ubiquitinate MYC whereas a number of deubiquitinating enzymes counteract this process. Recent studies show that SUMOylation and deSUMOylation can also regulate MYC protein stability and activity. Interestingly, evidence suggests an intriguing crosstalk between MYC ubiquitination and SUMOylation. Deregulation of the MYC ubiquitination-SUMOylation regulatory network may contribute to tumorigenesis. This review is intended to provide the current understanding of the complex regulation of the MYC biology by dynamic ubiquitination and SUMOylation and their crosstalk.
Collapse
Affiliation(s)
- Yingxiao Chen
- Departments of Molecular & Medical Genetics, School of Medicine, OHSU Knight Cancer Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Xiao-Xin Sun
- Departments of Molecular & Medical Genetics, School of Medicine, OHSU Knight Cancer Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Rosalie C Sears
- Departments of Molecular & Medical Genetics, School of Medicine, OHSU Knight Cancer Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Mu-Shui Dai
- Departments of Molecular & Medical Genetics, School of Medicine, OHSU Knight Cancer Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| |
Collapse
|
39
|
Wang C, Li X, Zhang J, Ge Z, Chen H, Hu J. EZH2 contributes to 5-FU resistance in gastric cancer by epigenetically suppressing FBXO32 expression. Onco Targets Ther 2018; 11:7853-7864. [PMID: 30464532 PMCID: PMC6225849 DOI: 10.2147/ott.s180131] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Background Increasing evidence suggests the involvement of enhancer of zeste homologue 2 (EZH2) in chemoresistance of cancer treatment. Nevertheless, its function and molecular mechanisms in gastric cancer (GC) chemoresistance are still not well elucidated. Materials and methods In the present study, we investigated the functional role of EZH2 in 5-fluorouracil (5-FU) resistance of GC cells and discovered the underlying molecular mechanism. Results Results revealed that EZH2 was upregulated in 5-FU-resistant GC tissues and cell lines. High ZEH2 expression was correlated with poor prognosis of GC patients. EZH2 knockdown enhanced 5-FU sensitivity of AGS/5-FU and SGC-7901/5-FU cells. Moreover, EZH2 could epigenetically suppress FBXO32 expression. FBXO32 overexpression could mimic the functional role of downregulated EZH2 in 5-FU resistance. FBXO32 knockdown counteracted the inductive effect of EZH2 inhibition on 5-FU sensitivity of AGS/5-FU and SGC-7901/5-FU cells. Furthermore, EZH2 knockdown facilitated 5-FU sensitivity of 5-FU-resistant GC cells in vivo. Conclusion In summary, EZH2 depletion overcame 5-FU resistance in GC by epigenetically silencing FBXO32, providing a novel therapeutic target for GC chemoresistance.
Collapse
Affiliation(s)
- Chenyu Wang
- Department of Anorectal, Huaihe Hospital of Henan University, Kaifeng, 475000, People's Republic of China,
| | - Xingwang Li
- Department of Anorectal, Huaihe Hospital of Henan University, Kaifeng, 475000, People's Republic of China,
| | - Junjie Zhang
- Department of Anorectal, Huaihe Hospital of Henan University, Kaifeng, 475000, People's Republic of China,
| | - Zheng Ge
- Department of Anorectal, Huaihe Hospital of Henan University, Kaifeng, 475000, People's Republic of China,
| | - Hejin Chen
- Department of Anorectal, Huaihe Hospital of Henan University, Kaifeng, 475000, People's Republic of China,
| | - Junhong Hu
- Department of Anorectal, Huaihe Hospital of Henan University, Kaifeng, 475000, People's Republic of China,
| |
Collapse
|
40
|
Yang C, Fan P, Zhu S, Yang H, Jin X, Wu H. 3F-Box protein 32 degrades ataxia telangiectasia and Rad3-related and regulates DNA damage response induced by gemcitabine in pancreatic cancer. Oncol Lett 2018; 15:8878-8884. [PMID: 29805624 PMCID: PMC5950567 DOI: 10.3892/ol.2018.8367] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 02/19/2018] [Indexed: 12/26/2022] Open
Abstract
Ataxia telangiectasia and Rad3-related (ATR) activates checkpoint kinase 1 (CHK1) following replication fork stalling, leading to cell cycle arrest. ATR-CHK1 pathway components are considered to be promising therapeutic targets to enhance the effectiveness of replication inhibitors. The present study revealed that F-Box protein 32 (FBXO32) regulated ATR expression in pancreatic cancer PANC-1 and MIA PaCa-2 cells. Additionally, FBXO32 interacts with ATR in PANC-1 cells and ATR is a degradation substrate of E3 ubiquitin ligase FBXO32. Furthermore, FBXO32 regulated the DNA damage response induced by gemcitabine in PANC-1 cells. Taken together, the results of the present study suggested that FBXO32, as an E3 ubiquitin ligase of ATR, regulates the DNA damage response induced by gemcitabine in pancreatic cancer.
Collapse
Affiliation(s)
- Chong Yang
- Organ Transplantation Center, Hospital of The University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Ping Fan
- Department of Digestive Surgical Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Shikai Zhu
- Organ Transplantation Center, Hospital of The University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Hongji Yang
- Organ Transplantation Center, Hospital of The University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Xin Jin
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Heshui Wu
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
41
|
McGirr JA, Martin CH. Parallel evolution of gene expression between trophic specialists despite divergent genotypes and morphologies. Evol Lett 2018; 2:62-75. [PMID: 30283665 PMCID: PMC6089502 DOI: 10.1002/evl3.41] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 12/22/2017] [Accepted: 01/03/2018] [Indexed: 12/20/2022] Open
Abstract
Parallel evolution of gene expression commonly underlies convergent niche specialization, but parallel changes in expression could also underlie divergent specialization. We investigated divergence in gene expression and whole-genome genetic variation across three sympatric Cyprinodon pupfishes endemic to San Salvador Island, Bahamas. This recent radiation consists of a generalist and two derived specialists adapted to novel niches: a scale-eating and a snail-eating pupfish. We sampled total mRNA from all three species at two early developmental stages and compared gene expression with whole-genome genetic differentiation among all three species in 42 resequenced genomes. Eighty percent of genes that were differentially expressed between snail-eaters and generalists were up or down regulated in the same direction between scale-eaters and generalists; however, there were no fixed variants shared between species underlying these parallel changes in expression. Genes showing parallel evolution of expression were enriched for effects on metabolic processes, whereas genes showing divergent expression were enriched for effects on cranial skeleton development and pigment biosynthesis, reflecting the most divergent phenotypes observed between specialist species. Our findings reveal that even divergent niche specialists may exhibit convergent adaptation to higher trophic levels through shared genetic pathways. This counterintuitive result suggests that parallel evolution in gene expression can accompany divergent ecological speciation during adaptive radiation.
Collapse
Affiliation(s)
- Joseph A. McGirr
- Department of BiologyUniversity of North Carolina at Chapel HillChapel HillNorth Carolina27514
| | - Christopher H. Martin
- Department of BiologyUniversity of North Carolina at Chapel HillChapel HillNorth Carolina27514
| |
Collapse
|
42
|
van Doeselaar S, Burgering BMT. FOXOs Maintaining the Equilibrium for Better or for Worse. Curr Top Dev Biol 2018; 127:49-103. [PMID: 29433740 DOI: 10.1016/bs.ctdb.2017.10.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
A paradigm shift is emerging within the FOXO field and accumulating evidence indicates that we need to reappreciate the role of FOXOs, at least in cancer development. Here, we discuss the possibility that FOXOs are both tumor suppressors as well as promoters of tumor progression. This is mostly dependent on the biological context. Critical to this dichotomous role is the notion that FOXOs are central in preserving cellular homeostasis in redox control, genomic stability, and protein turnover. From this perspective, a paradoxical role in both suppressing and enhancing tumor progression can be reconciled. As many small molecules targeting the PI3K pathway are developed by big pharmaceutical companies and/or are in clinical trial, we will discuss what the consequences may be for the context-dependent role of FOXOs in tumor development in treatment options based on active PI3K signaling in tumors.
Collapse
Affiliation(s)
- Sabina van Doeselaar
- Molecular Cancer Research, Center Molecular Medicine, Oncode Institute, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Boudewijn M T Burgering
- Molecular Cancer Research, Center Molecular Medicine, Oncode Institute, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
43
|
FBXO32 promotes microenvironment underlying epithelial-mesenchymal transition via CtBP1 during tumour metastasis and brain development. Nat Commun 2017; 8:1523. [PMID: 29142217 PMCID: PMC5688138 DOI: 10.1038/s41467-017-01366-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 09/12/2017] [Indexed: 12/12/2022] Open
Abstract
The set of events that convert adherent epithelial cells into migratory cells are collectively known as epithelial–mesenchymal transition (EMT). EMT is involved during development, for example, in triggering neural crest migration, and in pathogenesis such as metastasis. Here we discover FBXO32, an E3 ubiquitin ligase, to be critical for hallmark gene expression and phenotypic changes underlying EMT. Interestingly, FBXO32 directly ubiquitinates CtBP1, which is required for its stability and nuclear retention. This is essential for epigenetic remodeling and transcriptional induction of CtBP1 target genes, which create a suitable microenvironment for EMT progression. FBXO32 is also amplified in metastatic cancers and its depletion in a NSG mouse xenograft model inhibits tumor growth and metastasis. In addition, FBXO32 is essential for neuronal EMT during brain development. Together, these findings establish that FBXO32 acts as an upstream regulator of EMT by governing the gene expression program underlying this process during development and disease. Epithelial-to-mesenchymal transition (EMT) regulates both processes of organism development and changes in cell state causing disease. Here, the authors show that an E3 ubiquitin ligase, FBXO32, regulates EMT via CtBP1 and the transcriptional program, and also mediates cancer metastatic burden and neurogenesis.
Collapse
|
44
|
SPSB3 targets SNAIL for degradation in GSK-3β phosphorylation-dependent manner and regulates metastasis. Oncogene 2017; 37:768-776. [PMID: 29059170 DOI: 10.1038/onc.2017.370] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 08/11/2017] [Accepted: 09/04/2017] [Indexed: 12/11/2022]
Abstract
Epithelial-mesenchymal transition (EMT) is a process during which normal epithelial cells acquire mesenchymal characteristics. EMT has a critical role in various human diseases especially in cancer. EMT facilitates tumor initiation and progression by mediating cancer cell stemness and motility. Zinc finger transcription factor SNAIL is one of the most important initiators of EMT. Therefore, it is of great significance to understand the regulating mechanism of SNAIL. In this study, we carried out a luciferase-based genome-wide screening using small interfering RNA library against ~200 of E3 ligases and ubiquitin-related genes and identified SOCS box protein SPSB3 as a novel E3 ligase component that targets SNAIL into polyubiquitination and degradation in response to GSK-3β phosphorylation of SNAIL. Functionally, we observed that SPSB3 overexpression greatly inhibits tumor metastasis by regulating SNAIL degradation both in vitro and in vivo. The expression of SPSB3 and SNAIL are negatively correlated in human esophageal squamous cell carcinoma tissues, and low SPSB3 expression indicates lymph node metastasis. Moreover, high SPSB3 expression indicates good survivals in various kinds of cancer. Collectively, these findings suggest that SPSB3-mediated SNAIL degradation has a vital role in regulating EMT and cancer progression.
Collapse
|
45
|
Geng C, Kaochar S, Li M, Rajapakshe K, Fiskus W, Dong J, Foley C, Dong B, Zhang L, Kwon OJ, Shah SS, Bolaki M, Xin L, Ittmann M, O’Malley BW, Coarfa C, Mitsiades N. SPOP regulates prostate epithelial cell proliferation and promotes ubiquitination and turnover of c-MYC oncoprotein. Oncogene 2017; 36:4767-4777. [PMID: 28414305 PMCID: PMC5887163 DOI: 10.1038/onc.2017.80] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 01/16/2017] [Accepted: 02/26/2017] [Indexed: 12/13/2022]
Abstract
The E3 ubiquitin ligase adaptor speckle-type POZ protein (SPOP) is frequently dysregulated in prostate adenocarcinoma (PC), via either somatic mutations or mRNA downregulation, suggesting an important tumour suppressor function. To examine its physiologic role in the prostate epithelium in vivo, we generated mice with prostate-specific biallelic ablation of Spop. These mice exhibited increased prostate mass, prostate epithelial cell proliferation, and expression of c-MYC protein compared to littermate controls, and eventually developed prostatic intraepithelial neoplasia (PIN). We found that SPOPWT can physically interact with c-MYC protein and, upon exogenous expression in vitro, can promote c-MYC ubiquitination and degradation. This effect was attenuated in PC cells by introducing PC-associated SPOP mutants or upon knockdown of SPOP via short-hairpin-RNA, suggesting that SPOP inactivation directly increases c-MYC protein levels. Gene Set Enrichment Analysis revealed enrichment of Myc-induced genes in transcriptomic signatures associated with SPOPMT. Likewise, we observed strong inverse correlation between c-MYC activity and SPOP mRNA levels in two independent PC patient cohorts. The core SPOPMT;MYCHigh transcriptomic response, defined by the overlap between the SPOPMT and c-MYC transcriptomic programmes, was also associated with inferior clinical outcome in human PCs. Finally, the organoid-forming capacity of Spop-null murine prostate cells was more sensitive to c-MYC inhibition than that of Spop-WT cells, suggesting that c-MYC upregulation functionally contributes to the proliferative phenotype of Spop knock-out prostates. Taken together, our data highlight SPOP as an important regulator of luminal epithelial cell proliferation and c-MYC expression in prostate physiology, identify c-MYC as a novel bona fide SPOP substrate, and help explain the frequent inactivation of SPOP in human PC. We propose SPOPMT-induced stabilization of c-MYC protein as a novel mechanism that can increase total c-MYC levels in PC cells, in addition to amplification of c-MYC locus.
Collapse
Affiliation(s)
- Chuandong Geng
- Dept. of Medicine, Houston, TX 77030
- Dept. of Molecular and Cellular Biology, Houston, TX 77030
| | - Salma Kaochar
- Dept. of Medicine, Houston, TX 77030
- Dept. of Molecular and Cellular Biology, Houston, TX 77030
| | - Min Li
- Dept. of Medicine, Houston, TX 77030
- Dept. of Molecular and Cellular Biology, Houston, TX 77030
| | | | - Warren Fiskus
- Dept. of Medicine, Houston, TX 77030
- Dept. of Molecular and Cellular Biology, Houston, TX 77030
| | - Jianrong Dong
- Dept. of Molecular and Cellular Biology, Houston, TX 77030
| | - Christopher Foley
- Dept. of Medicine, Houston, TX 77030
- Dept. of Molecular and Cellular Biology, Houston, TX 77030
| | - Boming Dong
- Dept. of Medicine, Houston, TX 77030
- Dept. of Molecular and Cellular Biology, Houston, TX 77030
| | - Li Zhang
- Dept. of Molecular and Cellular Biology, Houston, TX 77030
| | - Oh-Joon Kwon
- Dept. of Molecular and Cellular Biology, Houston, TX 77030
| | - Shrijal S. Shah
- Dept. of Medicine, Houston, TX 77030
- Dept. of Molecular and Cellular Biology, Houston, TX 77030
| | - Menaka Bolaki
- Dept. of Medicine, Houston, TX 77030
- Dept. of Molecular and Cellular Biology, Houston, TX 77030
| | - Li Xin
- Dept. of Molecular and Cellular Biology, Houston, TX 77030
- Dan L. Duncan Cancer Center, Houston, TX 77030
| | - Michael Ittmann
- Dan L. Duncan Cancer Center, Houston, TX 77030
- Dept. of Pathology and Immunology and Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX 77030
| | | | - Cristian Coarfa
- Dept. of Molecular and Cellular Biology, Houston, TX 77030
- Dan L. Duncan Cancer Center, Houston, TX 77030
| | - Nicholas Mitsiades
- Dept. of Medicine, Houston, TX 77030
- Dept. of Molecular and Cellular Biology, Houston, TX 77030
- Dan L. Duncan Cancer Center, Houston, TX 77030
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
46
|
Kim D, Hong A, Park HI, Shin WH, Yoo L, Jeon SJ, Chung KC. Deubiquitinating enzyme USP22 positively regulates c-Myc stability and tumorigenic activity in mammalian and breast cancer cells. J Cell Physiol 2017; 232:3664-3676. [PMID: 28160502 DOI: 10.1002/jcp.25841] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 02/03/2017] [Accepted: 02/03/2017] [Indexed: 12/22/2022]
Abstract
The proto-oncogene c-Myc has a pivotal function in growth control, differentiation, and apoptosis and is frequently affected in human cancer, including breast cancer. Ubiquitin-specific protease 22 (USP22), a member of the USP family of deubiquitinating enzymes (DUBs), mediates deubiquitination of target proteins, including histone H2B and H2A, telomeric repeat binding factor 1, and cyclin B1. USP22 is also a component of the mammalian SAGA transcriptional co-activating complex. In this study, we explored the functional role of USP22 in modulating c-Myc stability and its physiological relevance in breast cancer progression. We found that USP22 promotes deubiquitination of c-Myc in several breast cancer cell lines, resulting in increased levels of c-Myc. Consistent with this, USP22 knockdown reduces c-Myc levels. Furthermore, overexpression of USP22 stimulates breast cancer cell growth and colony formation, and increases c-Myc tumorigenic activity. In conclusion, the present study reveals that USP22 in breast cancer cell lines increases c-Myc stability through c-Myc deubiquitination, which is closely correlated with breast cancer progression.
Collapse
Affiliation(s)
- Dongyeon Kim
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Ahyoung Hong
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Hye In Park
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Woo Hyun Shin
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Lang Yoo
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Seo Jeong Jeon
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Kwang Chul Chung
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| |
Collapse
|
47
|
FBXO32 suppresses breast cancer tumorigenesis through targeting KLF4 to proteasomal degradation. Oncogene 2017; 36:3312-3321. [PMID: 28068319 DOI: 10.1038/onc.2016.479] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 11/10/2016] [Accepted: 11/15/2016] [Indexed: 12/21/2022]
Abstract
Krüppel-like factor 4 (KLF4, GKLF) is a zinc-finger transcription factor involved in a large variety of cellular processes, including apoptosis, cell cycle progression, as well as stem cell renewal. KLF4 is critical for cell fate decision and has an ambivalent role in tumorigenesis. Emerging data keep reminding us that KLF4 dysregulation either facilitates or impedes tumor progression, making it important to clarify the regulating network of KLF4. Like most transcription factors, KLF4 has a rather short half-life within the cell and its turnover must be carefully orchestrated by ubiquitination and ubiquitin-proteasome system. To better understand the mechanism of KLF4 ubiquitination, we performed a genome-wide screen of E3 ligase small interfering RNA library based on western blot and identified SCF-FBXO32 to be a new E3 ligase, which is responsible for KLF4 ubiquitination and degradation. The F-box domain is critical for FBXO32-dependent KLF4 ubiquitination and degradation. Furthermore, we demonstrated that FBXO32 physically interacts with the N-terminus (1-60 aa) of KLF4 via its C-terminus (228-355 aa) and directly targets KLF4 for ubiquitination and degradation. We also found out that p38 mitogen-activated protein kinase pathway may be implicated in FBXO32-mediated ubiquitination of KLF4, as p38 kinase inhibitor coincidently abrogates endogenous KLF4 ubiquitination and degradation, as well as FBXO32-dependent exogenous KLF4 ubiquitination and degradation. Finally, FBXO32 inhibits colony formation in vitro and primary tumor initiation and growth in vivo through targeting KLF4 into degradation. Our findings thus further elucidate the tumor-suppressive function of FBXO32 in breast cancer. These results expand our understanding of the posttranslational modification of KLF4 and of its role in breast cancer development and provide a potential target for diagnosis and therapeutic treatment of breast cancer.
Collapse
|
48
|
Jin J, Zhao L, Li Z. The E3 ubiquitin ligase RNF135 regulates the tumorigenesis activity of tongue cancer SCC25 cells. Cancer Med 2016; 5:3140-3146. [PMID: 27709798 PMCID: PMC5119969 DOI: 10.1002/cam4.832] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Revised: 06/11/2016] [Accepted: 06/27/2016] [Indexed: 12/13/2022] Open
Abstract
Several E3 ubiquitin ligases have been confirmed that they are related to the tumorigenesis. This study aims to find the tongue cancer‐related E3 ubiquitin ligase. The E3 ubiquitin ligase library was screened. The effect of candidate molecule on tongue cancer was validated through cell viability, cell proliferation, colony formation, invasive assay in vitro, and the xenograft model in vivo. The E3 ubiquitin ligase RNF135 significantly promoted the expression of PTEN and TP53 in SCC25 cells. The overexpression of RNF135 inhibited the viability, proliferation, and invasion of SCC25 cells. Knockdown of RNF135 had the opposite effects. Furthermore, RNF135 regulates the tumorigenesis activity of SCC25 cells in vivo. Our results demonstrated that RNF135 had the potential to affect the development of the tongue cancer in vitro. The further in vivo study is helpful to fully understand the role of it.
Collapse
Affiliation(s)
- Jian Jin
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.,Department of Oral and Maxillofacial Trauma and Plastic Aesthetic Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Liya Zhao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.,Department of Oral and Maxillofacial Trauma and Plastic Aesthetic Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Zubing Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.,Department of Oral and Maxillofacial Trauma and Plastic Aesthetic Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| |
Collapse
|
49
|
Gao R, Wang L, Cai H, Zhu J, Yu L. E3 Ubiquitin Ligase RLIM Negatively Regulates c-Myc Transcriptional Activity and Restrains Cell Proliferation. PLoS One 2016; 11:e0164086. [PMID: 27684546 PMCID: PMC5042457 DOI: 10.1371/journal.pone.0164086] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 09/19/2016] [Indexed: 11/19/2022] Open
Abstract
RNF12/RLIM is a RING domain-containing E3 ubiquitin ligase whose function has only begun to be elucidated recently. Although RLIM was reported to play important roles in some biological processes such as imprinted X-chromosome inactivation and regulation of TGF-β pathway etc., other functions of RLIM are largely unknown. Here, we identified RLIM as a novel E3 ubiquitin ligase for c-Myc, one of the most frequently deregulated oncoproteins in human cancers. RLIM associates with c-Myc in vivo and in vitro independently of the E3 ligase activity of RLIM. Moreover, RLIM promotes the polyubiquitination of c-Myc protein independently of Ser62 and Thr58 phosphorylation of c-Myc. However, RLIM-mediated ubiquitination does not affect c-Myc stability. Instead, RLIM inhibits the transcriptional activity of c-Myc through which RLIM restrains cell proliferation. Our results suggest that RLIM may function as a tumor suppressor by controlling the activity of c-Myc oncoprotein.
Collapse
Affiliation(s)
- Rui Gao
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, P.R. China
| | - Lan Wang
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, P.R. China
- Shanghai Center for Systems Biomedicine, Key Laboratory of Systems Biomedicine Ministry of Education, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Hao Cai
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, P.R. China
| | - Jingjing Zhu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Long Yu
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, P.R. China
| |
Collapse
|
50
|
Abstract
Ubiquitination plays a key and complex role in the regulation of c-Myc stability, transactivation, and oncogenic activity. c-Myc is ubiquitinated by a number of ubiquitin ligases (E3s), such as SCF(Fbw7) and SCF(Skp2). Depending on the E3s, ubiquitination can either positively or negatively regulate c-Myc levels and activity. Meanwhile, c-Myc ubiquitination can be reversed by deubiquitination. An early study showed that USP28 deubiquitinates c-Myc via interacting with Fbw7α whereas a recent study reveals that USP37 deubiquitinates c-Myc independently of Fbw7 and c-Myc phosphorylation. Consequently, both USP28 and USP37 stabilize c-Myc and enhance its activity. We recently found the nucleolar USP36 as a novel c-Myc deubiquitinase that controls the end-point of c-Myc degradation pathway in the nucleolus. Here we briefly review the current understanding of ubiquitination and deubiquitination regulation of c-Myc and further discuss the USP36-c-Myc regulatory pathway.
Collapse
Affiliation(s)
- Xiao-Xin Sun
- a Departments of Molecular & Medical Genetics ; School of Medicine and the OHSU Knight Cancer Institute; Oregon Health & Science University ; Portland , OR USA
| | - Rosalie C Sears
- a Departments of Molecular & Medical Genetics ; School of Medicine and the OHSU Knight Cancer Institute; Oregon Health & Science University ; Portland , OR USA
| | - Mu-Shui Dai
- a Departments of Molecular & Medical Genetics ; School of Medicine and the OHSU Knight Cancer Institute; Oregon Health & Science University ; Portland , OR USA
| |
Collapse
|