1
|
Boka HJ, Engel RM, Georges C, McMurrick PJ, Abud HE. Does side matter? Deciphering mechanisms that underpin side-dependent pathogenesis and therapy response in colorectal cancer. Mol Cancer 2025; 24:130. [PMID: 40312719 PMCID: PMC12046799 DOI: 10.1186/s12943-025-02327-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 04/08/2025] [Indexed: 05/03/2025] Open
Abstract
Colorectal cancer (CRC) is stratified by heterogeneity between disease sites, with proximal right-sided CRC (RCRC) multifactorial in its distinction from distal left-sided CRC (LCRC). Notably, right-sided tumors are associated with aggressive disease characteristics which culminate in poor clinical outcomes for these patients. While factors such as mutational profile and patterns of metastasis have been suggested to contribute to differences in therapy response, the exact mechanisms through which RCRC resists effective treatment have yet to be elucidated. In response, recent analyzes, including those utilizing whole genome sequencing, transcriptional profiling, and single-cell analyses, have demonstrated that key molecular differences exist between disease sites, with differentially expressed genes spanning a diverse range of cellular functions. Here, we review and contextualize the most recent data on molecular biomarkers found to exhibit discordance between RCRC and LCRC, and highlight candidates for further investigation, including those which present promise for future clinical application. Given the present disparity in survival outcomes for RCRC patients, we expect the prognostic biomarkers presented in our review to be useful in establishing future directions for the side-specific treatment of CRC.
Collapse
Affiliation(s)
- Harrison J Boka
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, 3800, Australia
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Department of Surgery, Cabrini Monash University, Cabrini Hospital, Malvern, VIC, 3144, Australia
| | - Rebekah M Engel
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, 3800, Australia
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Department of Surgery, Cabrini Monash University, Cabrini Hospital, Malvern, VIC, 3144, Australia
| | - Christine Georges
- Department of Surgery, Cabrini Monash University, Cabrini Hospital, Malvern, VIC, 3144, Australia
| | - Paul J McMurrick
- Department of Surgery, Cabrini Monash University, Cabrini Hospital, Malvern, VIC, 3144, Australia
| | - Helen E Abud
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, 3800, Australia.
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia.
- Department of Surgery, Cabrini Monash University, Cabrini Hospital, Malvern, VIC, 3144, Australia.
| |
Collapse
|
2
|
Ye Z, Yuan J, Hong D, Xu P, Liu W. Multimodal diagnostic models and subtype analysis for neoadjuvant therapy in breast cancer. Front Immunol 2025; 16:1559200. [PMID: 40170854 PMCID: PMC11958217 DOI: 10.3389/fimmu.2025.1559200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 02/26/2025] [Indexed: 04/03/2025] Open
Abstract
Background Breast cancer, a heterogeneous malignancy, comprises multiple subtypes and poses a substantial threat to women's health globally. Neoadjuvant therapy (NAT), administered prior to surgery, is integral to breast cancer treatment strategies. It aims to downsize tumors, optimize surgical outcomes, and evaluate tumor responsiveness to treatment. However, accurately predicting NAT efficacy remains challenging due to the disease's complexity and the diverse responses across different molecular subtypes. Methods In this study, we harnessed multimodal data, including proteomic, genomic, MRI imaging, and clinical information, sourced from multiple cohorts such as I-SPY2, TCGA-BRCA, GSE161529, and METABRIC. Post data preprocessing, Lasso regression was utilized for feature extraction and selection. Five machine learning algorithms were employed to construct diagnostic models, with pathological complete response (pCR) as the predictive endpoint. Results Our results revealed that the multi-omics Ridge regression model achieved the optimal performance in predicting pCR, with an AUC of 0.917. Through unsupervised clustering using the R package MOVICS and nine clustering algorithms, we identified four distinct multimodal breast cancer subtypes associated with NAT. These subtypes exhibited significant differences in proteomic profiles, hallmark cancer gene sets, pathway activities, tumor immune microenvironments, transcription factor activities, and clinical characteristics. For instance, CS1 subtype, predominantly ER-positive, had a low pCR rate and poor response to chemotherapy drugs, while CS4 subtype, characterized by high immune infiltration, showed a better response to immunotherapy. At the single-cell level, we detected significant heterogeneity in the tumor microenvironment among the four subtypes. Malignant cells in different subtypes displayed distinct copy number variations, differentiation levels, and evolutionary trajectories. Cell-cell communication analysis further highlighted differential interaction patterns among the subtypes, with implications for tumor progression and treatment response. Conclusion Our multimodal diagnostic model and subtype analysis provide novel insights into predicting NAT efficacy in breast cancer. These findings hold promise for guiding personalized treatment strategies. Future research should focus on experimental validation, in-depth exploration of the underlying mechanisms, and extension of these methods to other cancers and treatment modalities.
Collapse
Affiliation(s)
- Zheng Ye
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
- School of Computer Science of Information Technology, Qiannan Normal University for Nationalities, Duyun, Guizhou, China
| | - Jiaqi Yuan
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
| | - Deqing Hong
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
| | - Peng Xu
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
- School of Computer Science of Information Technology, Qiannan Normal University for Nationalities, Duyun, Guizhou, China
| | - Wenbin Liu
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
| |
Collapse
|
3
|
Li S, Jiang W, Chen F, Qian J, Yang J. The critical role of TRIM protein family in intervertebral disc degeneration: mechanistic insights and therapeutic perspectives. Front Cell Dev Biol 2025; 13:1525073. [PMID: 39981097 PMCID: PMC11839679 DOI: 10.3389/fcell.2025.1525073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 01/20/2025] [Indexed: 02/22/2025] Open
Abstract
Intervertebral disc degeneration (IVDD) is a leading cause of chronic back pain, contributing significantly to reduced quality of life and global public health burdens. The TRIM (Tripartite Motif-containing) protein family, with its diverse regulatory roles, has emerged as a key player in critical cellular processes such as inflammation, cell death, and extracellular matrix (ECM) metabolism. Recent findings underscore the involvement of TRIM proteins in IVDD pathogenesis, where they regulate stress responses, maintain cellular homeostasis, and influence the functional integrity of nucleus pulposus (NP) and annulus fibrosus (AF) cells. This review explores the multifaceted roles of TRIM proteins in IVDD, highlighting their contributions to pathological pathways and their potential as therapeutic targets. Advancing our understanding of TRIM protein-mediated mechanisms may pave the way for innovative and precise therapeutic strategies to combat IVDD.
Collapse
Affiliation(s)
- Shangze Li
- Department of Orthopedics, The Second Affiliated Hospital (Shanghai Changzheng Hospital), Naval Medical University, Shanghai, China
| | - Wenli Jiang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Naval Medical University, Shanghai, China
| | - Fei Chen
- Department of Orthopedics, The Second Affiliated Hospital (Shanghai Changzheng Hospital), Naval Medical University, Shanghai, China
| | - Jiao Qian
- Department of Pharmacy, The First Affiliated Hospital (Shanghai Changhai Hospital), Naval Medical University, Shanghai, China
| | - Jun Yang
- Department of Orthopedics, The Second Affiliated Hospital (Shanghai Changzheng Hospital), Naval Medical University, Shanghai, China
| |
Collapse
|
4
|
Zebene ED, Lombardi R, Pucci B, Medhin HT, Seife E, Di Gennaro E, Budillon A, Woldemichael GB. Proteomic Analysis of Biomarkers Predicting Treatment Response in Patients with Head and Neck Cancers. Int J Mol Sci 2024; 25:12513. [PMID: 39684225 DOI: 10.3390/ijms252312513] [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: 10/25/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Head and neck cancers (HNCs) are the sixth most commonly diagnosed cancer and the eighth leading cause of cancer-related mortality worldwide, with squamous cell carcinoma being the most prevalent type. The global incidence of HNCs is steadily increasing, projected to rise by approximately 30% per year by 2030, a trend observed in both developed and undeveloped countries. This study involved serum proteomic profiling to identify predictive clinical biomarkers in cancer patients undergoing chemoradiotherapy (CRT). Fifteen HNC patients at Tikur Anbessa Specialized Hospital, Radiotherapy (RT) center in Addis Ababa were enrolled. Serum samples were collected before and after RT, and patients were classified as responders (R) or non-responders (NR). Protein concentrations in the serum were determined using the Bradford assay, followed by nano-HPLC-MS/MS for protein profiling. Progenesis QI for proteomics identified 55 differentially expressed proteins (DEPs) between R and NR, with a significance of p < 0.05 and a fold-change (FC) ≥ 1.5. The top five-up-regulated proteins included MAD1L1, PSMC2, TRIM29, C5, and SERPING1, while the top five-down-regulated proteins were RYR1, HEY2, HIF1A, TF, and CNN3. Notably, about 16.4% of the DEPs were involved in cellular responses to DNA damage from cancer treatments, encompassing proteins related to deoxyribonucleic acid (DNA) damage sensing, checkpoint activation, DNA repair, and apoptosis/cell cycle regulation. The analysis of the relative abundance of ten proteins with high confidence scores identified three DEPs: ADIPOQ, HEY2, and FUT10 as potential predictive biomarkers for treatment response. This study highlighted the identification of three potential predictive biomarkers-ADIPOQ, HEY2, and FUT10-through serum proteomic profiling in HNC patients undergoing RT, emphasizing their significance in predicting treatment response.
Collapse
Affiliation(s)
- Emeshaw Damtew Zebene
- Nuclear Medicine Unit, Department of Internal Medicine, College of Health Sciences, Addis Ababa University, Addis Ababa 9086, Ethiopia
- Department of Microbial Cellular and Molecular Biology, College of Natural and Computational Sciences, Addis Ababa University, Addis Ababa 9086, Ethiopia
| | - Rita Lombardi
- Experimental Animal Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy
| | - Biagio Pucci
- Experimental Pharmacology Unit-Laboratory of Naples and Mercogliano (AV), Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy
| | - Hagos Tesfay Medhin
- Nuclear Medicine Unit, Department of Internal Medicine, College of Health Sciences, Addis Ababa University, Addis Ababa 9086, Ethiopia
| | - Edom Seife
- Radiotherapy Center, College of Health Sciences, Addis Ababa University, Addis Ababa 9086, Ethiopia
| | - Elena Di Gennaro
- Experimental Pharmacology Unit-Laboratory of Naples and Mercogliano (AV), Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy
| | - Alfredo Budillon
- Scientific Directorate, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy
| | - Gurja Belay Woldemichael
- Department of Microbial Cellular and Molecular Biology, College of Natural and Computational Sciences, Addis Ababa University, Addis Ababa 9086, Ethiopia
| |
Collapse
|
5
|
Kim Y, Min S, Kim S, Lee S, Park YJ, Heo Y, Park S, Park T, Lee JH, Kang H, Ji JH, Cho H. PARP1-TRIM44-MRN loop dictates the response to PARP inhibitors. Nucleic Acids Res 2024; 52:11720-11737. [PMID: 39217466 PMCID: PMC11514498 DOI: 10.1093/nar/gkae756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 07/12/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
PARP inhibitors (PARPi) show selective efficacy in tumors with homologous recombination repair (HRR)-defects but the activation mechanism of HRR pathway in PARPi-treated cells remains enigmatic. To unveil it, we searched for the mediator bridging PARP1 to ATM pathways by screening 211 human ubiquitin-related proteins. We discovered TRIM44 as a crucial mediator that recruits the MRN complex to damaged chromatin, independent of PARP1 activity. TRIM44 binds PARP1 and regulates the ubiquitination-PARylation balance of PARP1, which facilitates timely recruitment of the MRN complex for DSB repair. Upon exposure to PARPi, TRIM44 shifts its binding from PARP1 to the MRN complex via its ZnF UBP domain. Knockdown of TRIM44 in cells significantly enhances the sensitivity to olaparib and overcomes the resistance to olaparib induced by 53BP1 deficiency. These observations emphasize the central role of TRIM44 in tethering PARP1 to the ATM-mediated repair pathway. Suppression of TRIM44 may enhance PARPi effectiveness and broaden their use even to HR-proficient tumors.
Collapse
Affiliation(s)
- Yonghyeon Kim
- Department of Biochemistry, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Sunwoo Min
- Department of Biochemistry, Ajou University School of Medicine, Suwon 16499, Republic of Korea
- Department of Biochemistry, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Soyeon Kim
- Department of Physiology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Seo Yun Lee
- Department of Physiology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
- Department of Life Science and Multidisciplinary Genome Institute, Hallym University, Chuncheon 24252, Republic of Korea
| | - Yeon-Ji Park
- Department of Biochemistry, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Yungyeong Heo
- Department of Biochemistry, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Soon Sang Park
- Department of Biochemistry, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Tae Jun Park
- Department of Biochemistry, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Jae-Ho Lee
- Department of Biochemistry, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Ho Chul Kang
- Department of Physiology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Jae-Hoon Ji
- Department of Biochemistry and Structural Biology, The University of Texas Health San Antonio, TX 78229-3000, USA
| | - Hyeseong Cho
- Department of Biochemistry, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| |
Collapse
|
6
|
He M, Jiang H, Li S, Xue M, Wang H, Zheng C, Tong J. The crosstalk between DNA-damage responses and innate immunity. Int Immunopharmacol 2024; 140:112768. [PMID: 39088918 DOI: 10.1016/j.intimp.2024.112768] [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: 05/20/2024] [Revised: 07/14/2024] [Accepted: 07/22/2024] [Indexed: 08/03/2024]
Abstract
DNA damage is typically caused during cell growth by DNA replication stress or exposure to endogenous or external toxins. The accumulation of damaged DNA causes genomic instability, which is the root cause of many serious disorders. Multiple cellular organisms utilize sophisticated signaling pathways against DNA damage, collectively known as DNA damage response (DDR) networks. Innate immune responses are activated following cellular abnormalities, including DNA damage. Interestingly, recent studies have indicated that there is an intimate relationship between the DDR network and innate immune responses. Diverse kinds of cytosolic DNA sensors, such as cGAS and STING, recognize damaged DNA and induce signals related to innate immune responses, which link defective DDR to innate immunity. Moreover, DDR components operate in immune signaling pathways to induce IFNs and/or a cascade of inflammatory cytokines via direct interactions with innate immune modulators. Consistently, defective DDR factors exacerbate the innate immune imbalance, resulting in severe diseases, including autoimmune disorders and tumorigenesis. Here, the latest progress in understanding crosstalk between the DDR network and innate immune responses is reviewed. Notably, the dual function of innate immune modulators in the DDR network may provide novel insights into understanding and developing targeted immunotherapies for DNA damage-related diseases, even carcinomas.
Collapse
Affiliation(s)
- Mei He
- College of Life Sciences, Hebei University, Baoding 071002, China; Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
| | - Hua Jiang
- Department of Hematology, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200000, China
| | - Shun Li
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu 610041, China
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450001, China.
| | - Huiqing Wang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China.
| | - Chunfu Zheng
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada.
| | - Jie Tong
- College of Life Sciences, Hebei University, Baoding 071002, China.
| |
Collapse
|
7
|
Zohourian N, Coll E, Dever M, Sheahan A, Burns-Lane P, Brown JAL. Evaluating the Cellular Roles of the Lysine Acetyltransferase Tip60 in Cancer: A Multi-Action Molecular Target for Precision Oncology. Cancers (Basel) 2024; 16:2677. [PMID: 39123405 PMCID: PMC11312108 DOI: 10.3390/cancers16152677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/04/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
Precision (individualized) medicine relies on the molecular profiling of tumors' dysregulated characteristics (genomic, epigenetic, transcriptomic) to identify the reliance on key pathways (including genome stability and epigenetic gene regulation) for viability or growth, and then utilises targeted therapeutics to disrupt these survival-dependent pathways. Non-mutational epigenetic changes alter cells' transcriptional profile and are a key feature found in many tumors. In contrast to genetic mutations, epigenetic changes are reversable, and restoring a normal epigenetic profile can inhibit tumor growth and progression. Lysine acetyltransferases (KATs or HATs) protect genome stability and integrity, and Tip60 is an essential acetyltransferase due to its roles as an epigenetic and transcriptional regulator, and as master regulator of the DNA double-strand break response. Tip60 is commonly downregulated and mislocalized in many cancers, and the roles that mislocalized Tip60 plays in cancer are not well understood. Here we categorize and discuss Tip60-regulated genes, evaluate Tip60-interacting proteins based on cellular localization, and explore the therapeutic potential of Tip60-targeting compounds as epigenetic inhibitors. Understanding the multiple roles Tip60 plays in tumorigenesis will improve our understanding of tumor progression and will inform therapeutic options, including informing potential combinatorial regimes with current chemotherapeutics, leading to improvements in patient outcomes.
Collapse
Affiliation(s)
- Nazanin Zohourian
- Department of Biological Science, University of Limerick, V94 T9PX Limerick, Ireland; (N.Z.)
| | - Erin Coll
- Department of Biological Science, University of Limerick, V94 T9PX Limerick, Ireland; (N.Z.)
| | - Muiread Dever
- Department of Biological Science, University of Limerick, V94 T9PX Limerick, Ireland; (N.Z.)
| | - Anna Sheahan
- Department of Biological Science, University of Limerick, V94 T9PX Limerick, Ireland; (N.Z.)
| | - Petra Burns-Lane
- Department of Biological Science, University of Limerick, V94 T9PX Limerick, Ireland; (N.Z.)
| | - James A. L. Brown
- Department of Biological Science, University of Limerick, V94 T9PX Limerick, Ireland; (N.Z.)
- Limerick Digital Cancer Research Centre (LDCRC), Health Research Institute (HRI), University of Limerick, V94 T9PX Limerick, Ireland
| |
Collapse
|
8
|
Tong J, Song J, Zhang W, Zhai J, Guan Q, Wang H, Liu G, Zheng C. When DNA-damage responses meet innate and adaptive immunity. Cell Mol Life Sci 2024; 81:185. [PMID: 38630271 PMCID: PMC11023972 DOI: 10.1007/s00018-024-05214-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 03/17/2024] [Accepted: 03/18/2024] [Indexed: 04/19/2024]
Abstract
When cells proliferate, stress on DNA replication or exposure to endogenous or external insults frequently results in DNA damage. DNA-Damage Response (DDR) networks are complex signaling pathways used by multicellular organisms to prevent DNA damage. Depending on the type of broken DNA, the various pathways, Base-Excision Repair (BER), Nucleotide Excision Repair (NER), Mismatch Repair (MMR), Homologous Recombination (HR), Non-Homologous End-Joining (NHEJ), Interstrand Crosslink (ICL) repair, and other direct repair pathways, can be activated separately or in combination to repair DNA damage. To preserve homeostasis, innate and adaptive immune responses are effective defenses against endogenous mutation or invasion by external pathogens. It is interesting to note that new research keeps showing how closely DDR components and the immune system are related. DDR and immunological response are linked by immune effectors such as the cyclic GMP-AMP synthase (cGAS)-Stimulator of Interferon Genes (STING) pathway. These effectors act as sensors of DNA damage-caused immune response. Furthermore, DDR components themselves function in immune responses to trigger the generation of inflammatory cytokines in a cascade or even trigger programmed cell death. Defective DDR components are known to disrupt genomic stability and compromise immunological responses, aggravating immune imbalance and leading to serious diseases such as cancer and autoimmune disorders. This study examines the most recent developments in the interaction between DDR elements and immunological responses. The DDR network's immune modulators' dual roles may offer new perspectives on treating infectious disorders linked to DNA damage, including cancer, and on the development of target immunotherapy.
Collapse
Affiliation(s)
- Jie Tong
- College of Life Science, Hebei University, Baoding, 071002, China
- Institute of Life Science and Green Development, Hebei University, Baoding, 071002, China
| | - Jiangwei Song
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, 100089, China
| | - Wuchao Zhang
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, 071000, China
| | - Jingbo Zhai
- Key Laboratory of Zoonose Prevention and Control at Universities of Inner Mongolia Autonomous Region, Medical College, Inner Mongolia Minzu University, Tongliao, 028000, China
| | - Qingli Guan
- The Affiliated Hospital of Chinese PLA 80th Group Army, Weifang, 261000, China
| | - Huiqing Wang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.
| | - Gentao Liu
- Department of Oncology, Tenth People's Hospital Affiliated to Tongji University & Cancer Center, Tongji University School of Medicine, Shanghai, 20000, China.
| | - Chunfu Zheng
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
9
|
Sultanov R, Mulyukina A, Zubkova O, Fedoseeva A, Bogomazova A, Klimina K, Larin A, Zatsepin T, Prikazchikova T, Lukina M, Bogomiakova M, Sharova E, Generozov E, Lagarkova M, Arapidi G. TP63-TRIM29 axis regulates enhancer methylation and chromosomal instability in prostate cancer. Epigenetics Chromatin 2024; 17:6. [PMID: 38481282 PMCID: PMC10938740 DOI: 10.1186/s13072-024-00529-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 02/09/2024] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND Prostate adenocarcinoma (PRAD) is the second leading cause of cancer-related deaths in men. High variability in DNA methylation and a high rate of large genomic rearrangements are often observed in PRAD. RESULTS To investigate the reasons for such high variance, we integrated DNA methylation, RNA-seq, and copy number alterations datasets from The Cancer Genome Atlas (TCGA), focusing on PRAD, and employed weighted gene co-expression network analysis (WGCNA). Our results show that only single cluster of co-expressed genes is associated with genomic and epigenomic instability. Within this cluster, TP63 and TRIM29 are key transcription regulators and are downregulated in PRAD. We discovered that TP63 regulates the level of enhancer methylation in prostate basal epithelial cells. TRIM29 forms a complex with TP63 and together regulates the expression of genes specific to the prostate basal epithelium. In addition, TRIM29 binds DNA repair proteins and prevents the formation of the TMPRSS2:ERG gene fusion typically observed in PRAD. CONCLUSION Our study demonstrates that TRIM29 and TP63 are important regulators in maintaining the identity of the basal epithelium under physiological conditions. Furthermore, we uncover the role of TRIM29 in PRAD development.
Collapse
Affiliation(s)
- R Sultanov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia.
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia.
| | - A Mulyukina
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - O Zubkova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - A Fedoseeva
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - A Bogomazova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - K Klimina
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - A Larin
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - T Zatsepin
- Department of Chemistry, Lomonosov Moscow State University, Moscow, Russia
| | - T Prikazchikova
- Department of Chemistry, Lomonosov Moscow State University, Moscow, Russia
| | - M Lukina
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - M Bogomiakova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - E Sharova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - E Generozov
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - M Lagarkova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - G Arapidi
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
10
|
Pratt HG, Ma L, Dziadowicz SA, Ott S, Whalley T, Szomolay B, Eubank TD, Hu G, Boone BA. Analysis of single nuclear chromatin accessibility reveals unique myeloid populations in human pancreatic ductal adenocarcinoma. Clin Transl Med 2024; 14:e1595. [PMID: 38426634 PMCID: PMC10905544 DOI: 10.1002/ctm2.1595] [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: 12/20/2023] [Revised: 01/30/2024] [Accepted: 02/06/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND A better understanding of the pancreatic ductal adenocarcinoma (PDAC) immune microenvironment is critical to developing new treatments and improving outcomes. Myeloid cells are of particular importance for PDAC progression; however, the presence of heterogenous subsets with different ontogeny and impact, along with some fluidity between them, (infiltrating monocytes vs. tissue-resident macrophages; M1 vs. M2) makes characterisation of myeloid populations challenging. Recent advances in single cell sequencing technology provide tools for characterisation of immune cell infiltrates, and open chromatin provides source and function data for myeloid cells to assist in more comprehensive characterisation. Thus, we explore single nuclear assay for transposase accessible chromatin (ATAC) sequencing (snATAC-Seq), a method to analyse open gene promoters and transcription factor binding, as an important means for discerning the myeloid composition in human PDAC tumours. METHODS Frozen pancreatic tissues (benign or PDAC) were prepared for snATAC-Seq using 10× Chromium technology. Signac was used for preliminary analysis, clustering and differentially accessible chromatin region identification. The genes annotated in promoter regions were used for Gene Ontology (GO) enrichment and cell type annotation. Gene signatures were used for survival analysis with The Cancer Genome Atlas (TCGA)-pancreatic adenocarcinoma (PAAD) dataset. RESULTS Myeloid cell transcription factor activities were higher in tumour than benign pancreatic samples, enabling us to further stratify tumour myeloid populations. Subcluster analysis revealed eight distinct myeloid populations. GO enrichment demonstrated unique functions for myeloid populations, including interleukin-1b signalling (recruited monocytes) and intracellular protein transport (dendritic cells). The identified gene signature for dendritic cells influenced survival (hazard ratio = .63, p = .03) in the TCGA-PAAD dataset, which was unique to PDAC. CONCLUSIONS These data suggest snATAC-Seq as a method for analysis of frozen human pancreatic tissues to distinguish myeloid populations. An improved understanding of myeloid cell heterogeneity and function is important for developing new treatment targets in PDAC.
Collapse
Affiliation(s)
- Hillary G. Pratt
- Cancer Cell BiologyWest Virginia UniversityMorgantownWest VirginiaUSA
- WVU Cancer InstituteWest Virginia UniversityMorgantownWest VirginiaUSA
| | - Li Ma
- Department of MicrobiologyImmunology and Cell BiologyWest Virginia UniversityMorgantownWest VirginiaUSA
| | - Sebastian A. Dziadowicz
- Department of MicrobiologyImmunology and Cell BiologyWest Virginia UniversityMorgantownWest VirginiaUSA
| | - Sascha Ott
- Warwick Medical SchoolUniversity of WarwickCoventryUK
| | | | - Barbara Szomolay
- Division of Infection and Immunity & Systems Immunity Research InstituteCardiff UniversityCardiffUK
| | - Timothy D. Eubank
- Cancer Cell BiologyWest Virginia UniversityMorgantownWest VirginiaUSA
- WVU Cancer InstituteWest Virginia UniversityMorgantownWest VirginiaUSA
- Department of MicrobiologyImmunology and Cell BiologyWest Virginia UniversityMorgantownWest VirginiaUSA
- In Vivo Multifunctional Magnetic Resonance CenterWest Virginia UniversityMorgantownWest VirginiaUSA
| | - Gangqing Hu
- WVU Cancer InstituteWest Virginia UniversityMorgantownWest VirginiaUSA
- Department of MicrobiologyImmunology and Cell BiologyWest Virginia UniversityMorgantownWest VirginiaUSA
| | - Brian A. Boone
- Cancer Cell BiologyWest Virginia UniversityMorgantownWest VirginiaUSA
- WVU Cancer InstituteWest Virginia UniversityMorgantownWest VirginiaUSA
- Department of MicrobiologyImmunology and Cell BiologyWest Virginia UniversityMorgantownWest VirginiaUSA
- Department of SurgeryWest Virginia UniversityMorgantownWest VirginiaUSA
| |
Collapse
|
11
|
Chaturvedi G, Sarusi-Portuguez A, Loza O, Shimoni-Sebag A, Yoron O, Lawrence YR, Zach L, Hakim O. Dose-Dependent Transcriptional Response to Ionizing Radiation Is Orchestrated with DNA Repair within the Nuclear Space. Int J Mol Sci 2024; 25:970. [PMID: 38256047 PMCID: PMC10815587 DOI: 10.3390/ijms25020970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 01/05/2024] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
Radiation therapy is commonly used to treat glioblastoma multiforme (GBM) brain tumors. Ionizing radiation (IR) induces dose-specific variations in transcriptional programs, implicating that they are tightly regulated and critical components in the tumor response and survival. Yet, our understanding of the downstream molecular events triggered by effective vs. non-effective IR doses is limited. Herein, we report that variations in the genetic programs are positively and functionally correlated with the exposure to effective or non-effective IR doses. Genome architecture analysis revealed that gene regulation is spatially and temporally coordinated with DNA repair kinetics. The radiation-activated genes were pre-positioned in active sub-nuclear compartments and were upregulated following the DNA damage response, while the DNA repair activity shifted to the inactive heterochromatic spatial compartments. The IR dose affected the levels of DNA damage repair and transcription modulation, but not the order of the events, which was linked to their spatial nuclear positioning. Thus, the distinct coordinated temporal dynamics of DNA damage repair and transcription reprogramming in the active and inactive sub-nuclear compartments highlight the importance of high-order genome organization in synchronizing the molecular events following IR.
Collapse
Affiliation(s)
- Garima Chaturvedi
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Building 206, Ramat Gan 5290002, Israel; (A.S.-P.)
| | - Avital Sarusi-Portuguez
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Building 206, Ramat Gan 5290002, Israel; (A.S.-P.)
| | - Olga Loza
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Building 206, Ramat Gan 5290002, Israel; (A.S.-P.)
| | - Ariel Shimoni-Sebag
- Institute of Oncology, Sheba Medical Center, Ramat Gan 5262000, Israel; (A.S.-S.)
| | - Orly Yoron
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Building 206, Ramat Gan 5290002, Israel; (A.S.-P.)
| | | | - Leor Zach
- Institute of Oncology, Tel Aviv Soraski Medical Center, Tel Aviv 6423906, Israel
| | - Ofir Hakim
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Building 206, Ramat Gan 5290002, Israel; (A.S.-P.)
| |
Collapse
|
12
|
Wu M, Jin MM, Cao XH, Zhao L, Li YH. Silencing TRIM29 Sensitizes Non-small Cell Lung Cancer Cells to Anlotinib by Promoting Apoptosis via Binding RAD50. Curr Cancer Drug Targets 2024; 24:445-454. [PMID: 37644752 DOI: 10.2174/1568009623666230829143148] [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: 03/08/2023] [Revised: 06/13/2023] [Accepted: 07/25/2023] [Indexed: 08/31/2023]
Abstract
BACKGROUND Previous studies have proposed that the transcriptional regulatory factor tripartite motif containing 29 (TRIM29) is involved in carcinogenesis via binding with nucleic acid. TRIM29 is confirmed to be highly expressed when the cancer cells acquire therapy-resistant properties. We noticed that TRIM29 levels were significantly increased in anlotinib-resistant NCIH1975 (NCI-H1975/AR) cells via mining data information from gene expression omnibus (GEO) gene microarray (GSE142031; log2 fold change > 1, p < 0.05). OBJECTIVE Our study aimed to investigate the function of TRIM29 on the resistance to anlotinib in non-small cell lung cancer (NSCLC) cells, including NCI-H1975 and A549 cells. METHODS Real-time RT-PCR and western blot were used to detect TRIM29 expression in anlotinib- resistant NSCLC (NSCLC/AR) cells. Apoptosis were determined through flow cytometry, acridine orange/ethidium bromide staining as well as western blot. ELISA was used to measure the content of C-X3-C motif chemokine ligand 1. Co-Immunoprecipitation assay was performed to verify the interaction between TRIM29 and RAD50 double-strand break repair protein (RAD50). RESULTS TRIM29 expression was shown to be elevated in the cytoplasm and nucleus of NSCLC/ AR cells compared to normal NSCLC cells. Next, we demonstrated that TRIM29 knockdown facilitated apoptosis and enhanced the sensitivity to anlotinib in NSCLC/AR cells. Based on the refined results citing from the database BioGRID, it was proved that TRIM29 interacted with RAD50. Herein, RAD50 overexpression diminished the pro-apoptotic effect induced by silencing TRIM29 in anlotinib-resistant A549 (A549/AR) cells. CONCLUSION Finally, we concluded that the increased sensitivity to anlotinib in NSCLC/AR cells was achieved by knocking down TRIM29, besides, the positive effects of TRIM29 knockdown were attributed to the promotion of apoptosis via binding to RAD50 in NSCLC/AR cell nucleus. Therefore, TRIM29 might become a potential target for overcoming anlotinib resistance in NSCLC treatment.
Collapse
Affiliation(s)
- Min Wu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, No. 100, Huaihai Avenue, Hefei, Anhui, People's Republic of China
- Department of Respiratory and Critical Care Medicine, Anhui Public Health Clinical Center, No. 100, Huaihai Avenue, Hefei, Anhui, People's Republic of China
| | - Meng-Meng Jin
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, No. 100, Huaihai Avenue, Hefei, Anhui, People's Republic of China
- Department of Respiratory and Critical Care Medicine, Anhui Public Health Clinical Center, No. 100, Huaihai Avenue, Hefei, Anhui, People's Republic of China
| | - Xiao-Hui Cao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, No. 100, Huaihai Avenue, Hefei, Anhui, People's Republic of China
- Department of Respiratory and Critical Care Medicine, Anhui Public Health Clinical Center, No. 100, Huaihai Avenue, Hefei, Anhui, People's Republic of China
| | - Lei Zhao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, No. 100, Huaihai Avenue, Hefei, Anhui, People's Republic of China
- Department of Respiratory and Critical Care Medicine, Anhui Public Health Clinical Center, No. 100, Huaihai Avenue, Hefei, Anhui, People's Republic of China
| | - Yong-Huai Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, No. 100, Huaihai Avenue, Hefei, Anhui, People's Republic of China
- Department of Respiratory and Critical Care Medicine, Anhui Public Health Clinical Center, No. 100, Huaihai Avenue, Hefei, Anhui, People's Republic of China
| |
Collapse
|
13
|
Bahreyni-Toossi MT, Zafari N, Azimian H, Mehrad-Majd H, Farhadi J, Vaziri Nezamdoust F. Alteration in Expression of Trim29, TRIM37, TRIM44, and β-Catenin Genes After Irradiation in Human Cells with Different Radiosensitivity. Cancer Biother Radiopharm 2023; 38:506-511. [PMID: 32833505 DOI: 10.1089/cbr.2020.3915] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Introduction: Radiotherapy is a crucial component of treatment for ∼70% of all cancer patients. The identification of effective biomarkers of radiosensitivity (RS) is a fundamental goal of radiobiology. The authors hypothesize that the RS of human normal and tumoral cells is correlated by the level of expression of TRIM29, TRIM37, TRIM44, and β-catenin genes. Materials and Methods: Clonogenic assay was performed and RS of four cell lines was determined by survival fraction at 2 Gy. To determine the level of gene expression 6 and 24 h after irradiation, RNA was extracted from each cell line, and expression of the above-mentioned genes in cell lines with different RS was determined by real-time polymerase chain reaction (PCR). Results: The clonogenic assay showed that human dermal fibroblasts (fibroblast) and HT-29 (colorectal) cells are radioresistant, while human foreskin fibroblasts (fibroblast) and QU-DB (lung) cells are radiosensitive. Analysis of the real-time PCR data, 6 h after irradiation, showed that the increase and decrease of the expression of TRIM29 and TRIM37 genes were directly correlated with the RS of normal and tumor cells. At 24 h postirradiation, a considerable difference was only observed in the expression of the β-catenin gene. Conclusion: This study showed that the TRIM29 and TRIM37 genes are involved in the cell response to radiation and proposed that these genes may be biomarkers for predicting RS in normal and tumoral cell lines.
Collapse
Affiliation(s)
| | - Navid Zafari
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hosein Azimian
- Medical Physics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hassan Mehrad-Majd
- Cancer Molecular Pathology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Javad Farhadi
- Department of Biochemistry and Molecular Biology, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
| | | |
Collapse
|
14
|
Heliez L, Ricordel C, Becuwe P, Pedeux R. Newly identified tumor suppressor functions of ING proteins. Curr Opin Pharmacol 2023; 68:102324. [PMID: 36521226 DOI: 10.1016/j.coph.2022.102324] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/27/2022] [Accepted: 11/01/2022] [Indexed: 12/15/2022]
Abstract
The INhibitor of Growth (ING) proteins (ING1, ING2, ING3, ING4 and ING5) are a family of epigenetic regulators. Their decreased expression in numerous cancers led to identifying the ING proteins as gatekeeper tumor suppressors as they regulate cell cycle progression, apoptosis and senescence. Subsequently, they were also described as caretaker tumor suppressors through their involvement in DNA replication and the DNA damage response (DDR). Recent studies have identified new interactions of the ING proteins with proteins or pathways implicated in cell proliferation, the maintenance of stem cells pluripotency or the DDR. Furthermore, the ING proteins have been identified as regulators of ribosomal RNA synthesis and of mRNA stability and as regulators of mitochondrial DNA transcription resulting in the regulation of metabolism. These new findings highlight new antitumorigenic activities of the ING proteins that are potential targets for cancer treatment.
Collapse
Affiliation(s)
- Léane Heliez
- Univ Rennes 1, INSERM, OSS (Oncogenesis Stress Signaling), UMR_S 1242, CLCC Eugene Marquis, F-35000, Rennes, France
| | - Charles Ricordel
- Univ Rennes 1, INSERM, OSS (Oncogenesis Stress Signaling), UMR_S 1242, CLCC Eugene Marquis, F-35000, Rennes, France; Service de Pneumologie, CHU de Rennes, Rennes, France
| | - Philippe Becuwe
- Univ Rennes 1, INSERM, OSS (Oncogenesis Stress Signaling), UMR_S 1242, CLCC Eugene Marquis, F-35000, Rennes, France; Faculté des Sciences et Technologies, Université de Lorraine, 54506 Vandoeuvre-lès-Nancy CEDEX, France
| | - Rémy Pedeux
- Univ Rennes 1, INSERM, OSS (Oncogenesis Stress Signaling), UMR_S 1242, CLCC Eugene Marquis, F-35000, Rennes, France.
| |
Collapse
|
15
|
Gao G, Hausmann S, Flores NM, Benitez AM, Shen J, Yang X, Person MD, Gayatri S, Cheng D, Lu Y, Liu B, Mazur PK, Bedford MT. The NFIB/CARM1 partnership is a driver in preclinical models of small cell lung cancer. Nat Commun 2023; 14:363. [PMID: 36690626 PMCID: PMC9870865 DOI: 10.1038/s41467-023-35864-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 01/04/2023] [Indexed: 01/24/2023] Open
Abstract
The coactivator associated arginine methyltransferase (CARM1) promotes transcription, as its name implies. It does so by modifying histones and chromatin bound proteins. We identified nuclear factor I B (NFIB) as a CARM1 substrate and show that this transcription factor utilizes CARM1 as a coactivator. Biochemical studies reveal that tripartite motif 29 (TRIM29) is an effector molecule for methylated NFIB. Importantly, NFIB harbors both oncogenic and metastatic activities, and is often overexpressed in small cell lung cancer (SCLC). Here, we explore the possibility that CARM1 methylation of NFIB is important for its transforming activity. Using a SCLC mouse model, we show that both CARM1 and the CARM1 methylation site on NFIB are critical for the rapid onset of SCLC. Furthermore, CARM1 and methylated NFIB are responsible for maintaining similar open chromatin states in tumors. Together, these findings suggest that CARM1 might be a therapeutic target for SCLC.
Collapse
Affiliation(s)
- Guozhen Gao
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Simone Hausmann
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Natasha M Flores
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Ana Morales Benitez
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jianjun Shen
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Xiaojie Yang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Maria D Person
- Center for Biomedical Research Support, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Sitaram Gayatri
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Evozyne Inc., Chicago, IL, 60614, USA
| | - Donghang Cheng
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yue Lu
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Bin Liu
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Pawel K Mazur
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Mark T Bedford
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
16
|
Yang X, Zhang Y, Xue Z, Hu Y, Zhou W, Xue Z, Liu X, Liu G, Li W, Liu X, Li X, Han M, Wang J. TRIM56 promotes malignant progression of glioblastoma by stabilizing cIAP1 protein. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:336. [PMID: 36471347 PMCID: PMC9724401 DOI: 10.1186/s13046-022-02534-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 11/07/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND The tripartite motif (TRIM) family of proteins plays a key role in the developmental growth and therapeutic resistance of many tumors. However, the regulatory mechanisms and biological functions of TRIM proteins in human glioblastoma (GBM) are not yet fully understood. In this study, we focused on TRIM56, which emerged as the most differentially expressed TRIM family member with increased expression in GBM. METHODS Western blot, real-time quantitative PCR (qRT-PCR), immunofluorescence (IF) and immunohistochemistry (IHC) were used to study the expression levels of TRIM56 and cIAP1 in GBM cell lines. Co-immunoprecipitation (co-IP) was used to explore the specific binding between target proteins and TRIM56. A xenograft animal model was used to verify the tumor promoting effect of TRIM56 on glioma in vivo. RESULTS We observed elevated expression of TRIM56 in malignant gliomas and revealed that TRIM56 promoted glioma progression in vitro and in a GBM xenograft model in nude mice. Analysis of the Human Ubiquitin Array and co-IPs showed that cIAP1 is a protein downstream of TRIM56. TRIM56 deubiquitinated cIAP1, mainly through the zinc finger domain (amino acids 21-205) of TRIM56, thereby reducing the degradation of cIAP1 and thus increasing its expression. TRIM56 also showed prognostic significance in overall survival of glioma patients. CONCLUSIONS TRIM56-regulated post-translational modifications may contribute to glioma development through stabilization of cIAP1. Furthermore, TRIM56 may serve as a novel prognostic indicator and therapeutic molecular target for GBM.
Collapse
Affiliation(s)
- Xu Yang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Xi Road, 250012 Jinan, Shandong People’s Republic of China ,Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250012 China ,Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012 China
| | - Yan Zhang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Xi Road, 250012 Jinan, Shandong People’s Republic of China ,Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250012 China ,Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012 China
| | - Zhiwei Xue
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Xi Road, 250012 Jinan, Shandong People’s Republic of China ,Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250012 China ,Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012 China
| | - Yaotian Hu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Xi Road, 250012 Jinan, Shandong People’s Republic of China ,Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250012 China ,Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012 China
| | - Wenjing Zhou
- grid.460018.b0000 0004 1769 9639Department of Blood Transfusion, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250022 China
| | - Zhiyi Xue
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Xi Road, 250012 Jinan, Shandong People’s Republic of China ,Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250012 China ,Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012 China
| | - Xuemeng Liu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Xi Road, 250012 Jinan, Shandong People’s Republic of China ,Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250012 China ,Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012 China
| | - Guowei Liu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Xi Road, 250012 Jinan, Shandong People’s Republic of China ,Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250012 China ,Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012 China
| | - Wenjie Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Xi Road, 250012 Jinan, Shandong People’s Republic of China ,Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250012 China ,Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012 China
| | - Xiaofei Liu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Xi Road, 250012 Jinan, Shandong People’s Republic of China ,Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250012 China ,Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012 China
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Xi Road, 250012 Jinan, Shandong People’s Republic of China ,Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250012 China ,Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012 China
| | - Mingzhi Han
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Xi Road, 250012 Jinan, Shandong People’s Republic of China ,Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250012 China ,Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012 China ,grid.27255.370000 0004 1761 1174Medical Integration and Practice Center, Cheeloo College of Medicine, Shandong University, Jinan, 250012 China
| | - Jian Wang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Xi Road, 250012 Jinan, Shandong People’s Republic of China ,Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250012 China ,Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012 China ,grid.7914.b0000 0004 1936 7443Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, 5009 Bergen, Norway
| |
Collapse
|
17
|
Lu T, Wu Y. Tripartite Motif Containing 26 is a Positive Predictor for Endometrial Carcinoma Patients and Regulates Cell Survival in Endometrial Carcinoma. Horm Metab Res 2022; 54:859-865. [PMID: 36108622 DOI: 10.1055/a-1926-7364] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Functioning as an E3 ubiquitin ligase, tripartite motif containing 26 (TRIM26) can regulate the tumor behavior and the relevant inflammatory immune response. Endometrial carcinoma is a major gynecological malignant tumor in the world, while no relevant research has been performed. KMplot, a web-based survival analysis tool, demonstrated that TRIM26 expression was positively correlated with the overall survival in gynecological tumors, such as ovarian, cervical, and endometrial cancer. The relatively low expression of TRIM26 was also found in endometrial cancer tissues and endometrial cancer cell lines. In the online Gene Expression Profiling Interactive Analysis (GEPIA) platform, TRIM26 was positively correlated with the pre-apoptosis genes of p53, BIM, BID, BAX, and BAK, and negatively correlated with the anti-apoptosis gene of BCLW. To further explore the function of TRIM26 in endometrial carcinoma, Ishikawa and KLE cells were infected with PLVX-TRIM26-derived lentivirus. TRIM26 overexpression suppressed the growth of endometrial cells, with downregulated p-AKT and upregulated BIM and BID expression. PLVX-TRIM26 overexpressed Ishikawa cells were injected subcutaneously into the side flanks of male BALB/C nude mice to construct a TRIM26-overexpression xenograft model. TRIM26 overexpression suppressed the growth of endometrial cancer as indicated by downregulated tumor volume and tumor weight with downregulated p-AKT expression. TRIM26 could regulate AKT pathway and apoptosis process to inhibit the growth of endometrial carcinoma, which can be utilized as a survival predictor.
Collapse
Affiliation(s)
- Tanmin Lu
- Department of Gynecology and Obstetrics, Liaocheng People's Hospital, Liaocheng City, China
| | - Yu Wu
- Department of Gynecology and Obstetrics, Liaocheng People's Hospital, Liaocheng City, China
| |
Collapse
|
18
|
Zhang Y, Zhang W, Zheng L, Guo Q. The roles and targeting options of TRIM family proteins in tumor. Front Pharmacol 2022; 13:999380. [PMID: 36249749 PMCID: PMC9561884 DOI: 10.3389/fphar.2022.999380] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Tripartite motif (TRIM) containing proteins are a class of E3 ubiquitin ligases, which are critically implicated in the occurrence and development of tumors. They can function through regulating various aspects of tumors, such as tumor proliferation, metastasis, apoptosis and the development of drug resistance during tumor therapy. Some members of TRIM family proteins can mediate protein ubiquitination and chromosome translocation via modulating several signaling pathways, like p53, NF-κB, AKT, MAPK, Wnt/β-catenin and other molecular regulatory mechanisms. The multi-domain nature/multi-functional biological role of TRIMs implies that blocking just one function or one domain might not be sufficient to obtain the desired therapeutic outcome, therefore, a detailed and systematic understanding of the biological functions of the individual domains of TRIMs is required. This review mainly described their roles and underlying mechanisms in tumorigenesis and progression, and it might shade light on a potential targeting strategy for TRIMs in tumor treatment, especially using PROTACs.
Collapse
Affiliation(s)
- Yuxin Zhang
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Wenzhou Zhang
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Lufeng Zheng
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Qianqian Guo
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
19
|
Luo M, Hou J, Mai H, Chen J, Chen H, Zhou B, Hou J, Jiang DK. TRIM26 inhibits hepatitis B virus replication by promoting HBx degradation and TRIM26 genetic polymorphism predicts PegIFNα treatment response of HBeAg-positive chronic hepatitis B Patients. Aliment Pharmacol Ther 2022; 56:878-889. [PMID: 35872575 DOI: 10.1111/apt.17124] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/11/2022] [Accepted: 06/23/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Hepatitis B virus (HBV) infection is a serious global health burden. TRIM26 has been reported to affect hepatitis C virus replication. AIMS To manifest the role of TRIM26 on HBV replication and explore if there are single-nucleotide polymorphisms (SNPs) in TRIM26 associated with response to pegylated interferon-alpha (PegIFNα) treatment in patients with chronic hepatitis B (CHB). METHODS We investigated the effect and mechanism of TRIM26 on HBV replication in vitro. The association between SNPs in TRIM26 and PegIFNα treatment response was evaluated in two independent cohorts including 238 and 707 patients with HBeAg-positive CHB. RESULTS Knockdown of TRIM26 increased, while overexpression of TRIM26 inhibited, HBV replication. Co-immunoprecipitation assays and immunofluorescence showed that TRIM26 interacted and co-localised with HBx. Co-transfection of HBx-HIS and TRIM26-FLAG plasmids in Huh7 cells showed that TRIM26 inhibited the expression of HBx. Furthermore, TRIM26 inhibited HBV replication by mediating HBx ubiquitination degradation, and TRIM26 SPRY domain was responsible for the interaction and degradation of HBx. Besides, IFN increased TRIM26 expression. TRIM26 rs116806878 was associated with response to PegIFNα in two CHB cohorts. Moreover, a polygenic score integrating TRIM26 rs116806878, STAT4 rs7574865 and CFB rs12614 (previously reported to be associated with response to PegIFNα) was related to response to PegIFNα in CHB. CONCLUSIONS TRIM26 inhibits HBV replication; IFN promotes TRIM26 expression. TRIM26 exerts an inhibitory effect on HBx by promoting ubiquitin-mediated degradation of HBx. Furthermore, TRIM26 rs116806878 is a potential predictive biomarker of response to PegIFNα in patients with CHB.
Collapse
Affiliation(s)
- Mengqi Luo
- State Key Laboratory of Organ Failure Research, Guangdong Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases and Hepatology Unit, Institutes of Liver Diseases Research of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
- The Key Laboratory of Molecular Pathology (Hepatic Diseases) of Guangxi, Department of Pathology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Jia Hou
- State Key Laboratory of Organ Failure Research, Guangdong Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases and Hepatology Unit, Institutes of Liver Diseases Research of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Haoming Mai
- State Key Laboratory of Organ Failure Research, Guangdong Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases and Hepatology Unit, Institutes of Liver Diseases Research of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiaxuan Chen
- State Key Laboratory of Organ Failure Research, Guangdong Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases and Hepatology Unit, Institutes of Liver Diseases Research of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Haitao Chen
- State Key Laboratory of Organ Failure Research, Guangdong Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases and Hepatology Unit, Institutes of Liver Diseases Research of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Bin Zhou
- State Key Laboratory of Organ Failure Research, Guangdong Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases and Hepatology Unit, Institutes of Liver Diseases Research of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jinlin Hou
- State Key Laboratory of Organ Failure Research, Guangdong Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases and Hepatology Unit, Institutes of Liver Diseases Research of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - De-Ke Jiang
- State Key Laboratory of Organ Failure Research, Guangdong Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases and Hepatology Unit, Institutes of Liver Diseases Research of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
- The Key Laboratory of Molecular Pathology (Hepatic Diseases) of Guangxi, Department of Pathology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| |
Collapse
|
20
|
Dai W, Wang J, Wang Z, Xiao Y, Li J, Hong L, Pei M, Zhang J, Yang P, Wu X, Tang W, Jiang X, Jiang P, Xiang L, Li A, Lin J, Liu S, Wang J. Comprehensive Analysis of the Prognostic Values of the TRIM Family in Hepatocellular Carcinoma. Front Oncol 2022; 11:767644. [PMID: 35004288 PMCID: PMC8733586 DOI: 10.3389/fonc.2021.767644] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 11/30/2021] [Indexed: 12/13/2022] Open
Abstract
Background Accumulating studies have demonstrated the abnormal expressions and prognostic values of certain members of the tripartite motif (TRIM) family in diverse cancers. However, comprehensive prognostic values of the TRIM family in hepatocellular carcinoma (HCC) are yet to be clearly defined. Methods The prognostic values of the TRIM family were evaluated by survival analysis and univariate Cox regression analysis based on gene expression data and clinical data of HCC from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. The expression profiles, protein–protein interaction among the TRIM family, prediction of transcription factors (TFs) or miRNAs, genetic alterations, correlations with the hallmarks of cancer and immune infiltrates, and pathway enrichment analysis were explored by multiple public databases. Further, a TRIM family gene-based signature for predicting overall survival (OS) in HCC was built by using the least absolute shrinkage and selection operator (LASSO) regression. TCGA–Liver Hepatocellular Carcinoma (LIHC) cohort was used as the training set, and GSE76427 was used for external validation. Time-dependent receiver operating characteristic (ROC) and survival analysis were used to estimate the signature. Finally, a nomogram combining the TRIM family risk score and clinical parameters was established. Results High expressions of TRIM family members including TRIM3, TRIM5, MID1, TRIM21, TRIM27, TRIM32, TRIM44, TRIM47, and TRIM72 were significantly associated with HCC patients’ poor OS. A novel TRIM family gene-based signature (including TRIM5, MID1, TRIM21, TRIM32, TRIM44, and TRIM47) was built for OS prediction in HCC. ROC curves suggested the signature’s good performance in OS prediction. HCC patients in the high-risk group had poorer OS than the low-risk patients based on the signature. A nomogram integrating the TRIM family risk score, age, and TNM stage was established. The ROC curves suggested that the signature presented better discrimination than the similar model without the TRIM family risk score. Conclusion Our study identified the potential application values of the TRIM family for outcome prediction in HCC.
Collapse
Affiliation(s)
- Weiyu Dai
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jing Wang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zhi Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yizhi Xiao
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiaying Li
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Linjie Hong
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Miaomiao Pei
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jieming Zhang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ping Yang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaosheng Wu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weimei Tang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoling Jiang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ping Jiang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Li Xiang
- Department of Gastroenterology, Longgang District People's Hospital, Shenzhen, China
| | - Aimin Li
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jianjiao Lin
- Department of Gastroenterology, Longgang District People's Hospital, Shenzhen, China
| | - Side Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Gastroenterology, Longgang District People's Hospital, Shenzhen, China
| | - Jide Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Gastroenterology, Longgang District People's Hospital, Shenzhen, China
| |
Collapse
|
21
|
Nagasawa S, Ikeda K, Shintani D, Yang C, Takeda S, Hasegawa K, Horie K, Inoue S. Identification of a Novel Oncogenic Fusion Gene SPON1-TRIM29 in Clinical Ovarian Cancer That Promotes Cell and Tumor Growth and Enhances Chemoresistance in A2780 Cells. Int J Mol Sci 2022; 23:689. [PMID: 35054873 PMCID: PMC8776205 DOI: 10.3390/ijms23020689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/04/2022] [Accepted: 01/06/2022] [Indexed: 02/04/2023] Open
Abstract
Gene structure alterations, such as chromosomal rearrangements that develop fusion genes, often contribute to tumorigenesis. It has been shown that the fusion genes identified in public RNA-sequencing datasets are mainly derived from intrachromosomal rearrangements. In this study, we explored fusion transcripts in clinical ovarian cancer specimens based on our RNA-sequencing data. We successfully identified an in-frame fusion transcript SPON1-TRIM29 in chromosome 11 from a recurrent tumor specimen of high-grade serous carcinoma (HGSC), which was not detected in the corresponding primary carcinoma, and validated the expression of the identical fusion transcript in another tumor from a distinct HGSC patient. Ovarian cancer A2780 cells stably expressing SPON1-TRIM29 exhibited an increase in cell growth, whereas a decrease in apoptosis was observed, even in the presence of anticancer drugs. The siRNA-mediated silencing of SPON1-TRIM29 fusion transcript substantially impaired the enhanced growth of A2780 cells expressing the chimeric gene treated with anticancer drugs. Moreover, a subcutaneous xenograft model using athymic mice indicated that SPON1-TRIM29-expressing A2780 cells rapidly generated tumors in vivo compared to control cells, whose growth was significantly repressed by the fusion-specific siRNA administration. Overall, the SPON1-TRIM29 fusion gene could be involved in carcinogenesis and chemotherapy resistance in ovarian cancer, and offers potential use as a diagnostic and therapeutic target for the disease with the fusion transcript.
Collapse
Affiliation(s)
- Saya Nagasawa
- Division of Systems Medicine & Gene Therapy, Saitama Medical University, Hidaka, Saitama 350-1241, Japan; (S.N.); (K.I.); (C.Y.)
- Department of Obstetrics and Gynecology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan;
| | - Kazuhiro Ikeda
- Division of Systems Medicine & Gene Therapy, Saitama Medical University, Hidaka, Saitama 350-1241, Japan; (S.N.); (K.I.); (C.Y.)
| | - Daisuke Shintani
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka, Saitama 350-1298, Japan; (D.S.); (K.H.)
| | - Chiujung Yang
- Division of Systems Medicine & Gene Therapy, Saitama Medical University, Hidaka, Saitama 350-1241, Japan; (S.N.); (K.I.); (C.Y.)
- Department of Obstetrics and Gynecology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan;
| | - Satoru Takeda
- Department of Obstetrics and Gynecology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan;
| | - Kosei Hasegawa
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka, Saitama 350-1298, Japan; (D.S.); (K.H.)
| | - Kuniko Horie
- Division of Systems Medicine & Gene Therapy, Saitama Medical University, Hidaka, Saitama 350-1241, Japan; (S.N.); (K.I.); (C.Y.)
| | - Satoshi Inoue
- Division of Systems Medicine & Gene Therapy, Saitama Medical University, Hidaka, Saitama 350-1241, Japan; (S.N.); (K.I.); (C.Y.)
- Department of Systems Aging Science and Medicine, Tokyo Metropolitan Institute of Gerontology, Itabashi-ku, Tokyo 173-0015, Japan
| |
Collapse
|
22
|
Hsu CY, Yanagi T, Ujiie H. TRIM29 in Cutaneous Squamous Cell Carcinoma. Front Med (Lausanne) 2022; 8:804166. [PMID: 34988104 PMCID: PMC8720877 DOI: 10.3389/fmed.2021.804166] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/02/2021] [Indexed: 12/13/2022] Open
Abstract
Tripartite motif (TRIM) proteins play important roles in a wide range of cell physiological processes, such as signal transduction, transcriptional regulation, innate immunity, and programmed cell death. TRIM29 protein, encoded by the ATDC gene, belongs to the RING-less group of TRIM protein family members. It consists of four zinc finger motifs in a B-box domain and a coiled-coil domain, and makes use of the B-box domain as E3 ubiquitin ligase in place of the RING. TRIM29 was found to be involved in the formation of homodimers and heterodimers in relation to DNA binding; additional studies have also demonstrated its role in carcinogenesis, DNA damage signaling, and the suppression of radiosensitivity. Recently, we reported that TRIM29 interacts with keratins and FAM83H to regulate keratin distribution. Further, in cutaneous SCC, the expression of TRIM29 is silenced by DNA methylation, leading to the loss of TRIM29 and promotion of keratinocyte migration. This paper reviews the role of TRIM family proteins in malignant tumors, especially the role of TRIM29 in cutaneous SCC.
Collapse
Affiliation(s)
- Che-Yuan Hsu
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Teruki Yanagi
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hideyuki Ujiie
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
23
|
GRWD1-WDR5-MLL2 Epigenetic Complex Mediates H3K4me3 Mark and Is Essential for Kaposi's Sarcoma-Associated Herpesvirus-Induced Cellular Transformation. mBio 2021; 12:e0343121. [PMID: 34933446 PMCID: PMC8689518 DOI: 10.1128/mbio.03431-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Infection by Kaposi's sarcoma-associated herpesvirus (KSHV) is causally associated with numerous cancers. The mechanism of KSHV-induced oncogenesis remains unclear. By performing a CRISPR-Cas9 screening in a model of KSHV-induced cellular transformation of primary cells, we identified epigenetic regulators that were essential for KSHV-induced cellular transformation. Examination of TCGA data sets of the top 9 genes, including glutamate-rich WD repeat containing 1 (GRWD1), a WD40 family protein upregulated by KSHV, that had positive effects on cell proliferation and survival of KSHV-transformed cells (KMM) but not the matched primary cells (MM), uncovered the predictive values of their expressions for patient survival in numerous types of cancer. We revealed global epigenetic remodeling including H3K4me3 epigenetic active mark in KMM cells compared to MM cells. Knockdown of GRWD1 inhibited cell proliferation, cellular transformation, and tumor formation and caused downregulation of global H3K4me3 mark in KMM cells. GRWD1 interacted with WD repeat domain 5 (WDR5), the core protein of H3K4 methyltransferase complex, and several H3K4me3 methyltransferases, including myeloid leukemia 2 (MLL2). Knockdown of WDR5 and MLL2 phenocopied GRWD1 knockdown, caused global reduction of H3K4me3 mark, and altered the expression of similar sets of genes. Transcriptome sequencing (RNA-seq) and chromatin immunoprecipitation sequencing (ChIP-seq) analyses further identified common and distinct cellular genes and pathways that were regulated by GRWD1, WDR5, and MLL2. These results indicate that KSHV hijacks the GRWD1-WDR5-MLL2 epigenetic complex to regulate H3K4me3 methylation of specific genes, which is essential for KSHV-induced cellular transformation. Our work has identified an epigenetic complex as a novel therapeutic target for KSHV-induced cancers. IMPORTANCE By performing a genome-wide CRISPR-Cas9 screening, we have identified cellular epigenetic regulators that are essential for KSHV-induced cellular transformation. Among them, GRWD1 regulates epigenetic active mark H3K4me3 by interacting with WDR5 and MLL2 and recruiting them to chromatin loci of specific genes in KSHV-transformed cells. Hence, KSHV hijacks the GRWD1-WDR5-MLL2 complex to remodel cellular epigenome and induce cellular transformation. Since the dysregulation of GRWD1 is associated with poor prognosis in several types of cancer, GRWD1 might also be a critical driver in other viral or nonviral cancers.
Collapse
|
24
|
Morgan D, Berggren KL, Spiess CD, Smith HM, Tejwani A, Weir SJ, Lominska CE, Thomas SM, Gan GN. Mitogen-activated protein kinase-activated protein kinase-2 (MK2) and its role in cell survival, inflammatory signaling, and migration in promoting cancer. Mol Carcinog 2021; 61:173-199. [PMID: 34559922 DOI: 10.1002/mc.23348] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 08/25/2021] [Accepted: 08/27/2021] [Indexed: 12/19/2022]
Abstract
Cancer and the immune system share an intimate relationship. Chronic inflammation increases the risk of cancer occurrence and can also drive inflammatory mediators into the tumor microenvironment enhancing tumor growth and survival. The p38 MAPK pathway is activated both acutely and chronically by stress, inflammatory chemokines, chronic inflammatory conditions, and cancer. These properties have led to extensive efforts to find effective drugs targeting p38, which have been unsuccessful. The immediate downstream serine/threonine kinase and substrate of p38 MAPK, mitogen-activated-protein-kinase-activated-protein-kinase-2 (MK2) protects cells against stressors by regulating the DNA damage response, transcription, protein and messenger RNA stability, and motility. The phosphorylation of downstream substrates by MK2 increases inflammatory cytokine production, drives an immune response, and contributes to wound healing. By binding directly to p38 MAPK, MK2 is responsible for the export of p38 MAPK from the nucleus which gives MK2 properties that make it unique among the large number of p38 MAPK substrates. Many of the substrates of both p38 MAPK and MK2 are separated between the cytosol and nucleus and interfering with MK2 and altering this intracellular translocation has implications for the actions of both p38 MAPK and MK2. The inhibition of MK2 has shown promise in combination with both chemotherapy and radiotherapy as a method for controlling cancer growth and metastasis in a variety of cancers. Whereas the current data are encouraging the field requires the development of selective and well tolerated drugs to target MK2 and a better understanding of its effects for effective clinical use.
Collapse
Affiliation(s)
- Deri Morgan
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Kiersten L Berggren
- Department of Internal Medicine, Division of Medical Oncology, Section of Radiation Oncology, UNM School of Medicine, The University of New Mexico, Albuquerque, New Mexico, USA
| | - Colby D Spiess
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Hannah M Smith
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Ajay Tejwani
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Scott J Weir
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Christopher E Lominska
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Sufi M Thomas
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas, USA.,Department of Otolaryngology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Gregory N Gan
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, Kansas, USA.,Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
25
|
Huang R, Fu Y, Deng Y. KLF4 transactivates TRIM29 expression and modulates keratin network. Biochem Biophys Rep 2021; 28:101117. [PMID: 34485714 PMCID: PMC8403550 DOI: 10.1016/j.bbrep.2021.101117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/23/2021] [Accepted: 08/24/2021] [Indexed: 11/16/2022] Open
Abstract
The Krüppel-like factor 4 (KLF4) is well known to be a conserved zinc-containing transcription factor that participates in diverse biological processes such as cell proliferation and differentiation. In this study, we found KLF4 can bind specific site in the promoter of TRIM29 to transactivate its transcription, and sumoylation modification on 278 lysine site was not essential for KLF4 to transactivate TRIM29 transcription. It also was showed that KLF4 promoted cell migration when overexpressed, and knockdown of TRIM29 abrogated the migration triggered by KLF4. In addition, overexpression of KLF4 reduced the phosphorylation level of keratin 8 at 432 amino acid site. Our study demonstrated that KLF4 is an important transcription factor on regulating TRIM29 expression and modulates the keratin network.
Collapse
Affiliation(s)
- Runqing Huang
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yang Fu
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yanhong Deng
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
26
|
Offermann A, Kang D, Watermann C, Weingart A, Hupe MC, Saraji A, Stegmann-Frehse J, Kruper R, Schüle R, Pantel K, Taubert H, Duensing S, Culig Z, Aigner A, Klapper W, Jonigk D, Philipp Kühnel M, Merseburger AS, Kirfel J, Sailer V, Perner S. Manuscript Title: Analysis of tripartite motif (TRIM) family gene expression in prostate cancer bone metastases. Carcinogenesis 2021; 42:1475-1484. [PMID: 34487169 DOI: 10.1093/carcin/bgab083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/29/2021] [Accepted: 09/04/2021] [Indexed: 12/27/2022] Open
Abstract
Tripartite motif (TRIM) family proteins are post-translational protein modifiers with E3-ubiquitin ligase activity, thereby involved in various biological processes. The molecular mechanisms driving prostate cancer (PCa) bone metastasis (BM) are incompletely understood, and targetable genetic alterations are lacking in the majority of cases. Therefore, we aimed to explore the expression and potential functional relevance of 71 TRIM members in bone metastatic PCa. We performed transcriptome analysis of all human TRIM family members and 770 cancer-related genes in 29 localized PCa and 30 PCa BM using Nanostring. KEGG, STRING and Ubibrowser were used for further bioinformatic gene correlation and pathway enrichment analyses. Compared to localized tumors, six TRIMs are under-expressed while nine TRIMs are over-expressed in BM. The differentially expressed TRIM proteins are linked to TNF-, TGFβ-, PI3K/AKT- and HIF-1-signaling, and to features such as proteoglycans, platelet activation, adhesion and ECM-interaction based on correlation to cancer-related genes. The identification of TRIM-specific E3-ligase-substrates revealed insight into functional connections to oncogenes, tumor suppressors and cancer-related pathways including androgen receptor- and TGFβ signaling, cell cycle regulation and splicing. In summary, this is the first study that comprehensively and systematically characterizes the expression of all TRIM members in PCa BM. Our results describe post-translational protein modification as an important regulatory mechanism of oncogenes, tumor suppressors, and pathway molecules in PCa progression. Therefore, this study may provide evidence for novel therapeutic targets, in particular for the treatment or prevention of BM.
Collapse
Affiliation(s)
- Anne Offermann
- Institute of Pathology, University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Duan Kang
- Institute of Pathology, University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Christian Watermann
- Institute of Pathology, University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Anika Weingart
- Institute of Pathology, University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Marie C Hupe
- Department of Urology, University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Alireza Saraji
- Institute of Pathology, University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Janine Stegmann-Frehse
- Institute of Pathology, University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | | | - Roland Schüle
- Klinik für Urologie und Zentrale Klinische Forschung, Klinikum der Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Klaus Pantel
- Institute for Tumor Biology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Helge Taubert
- Department of Urology and Paediatric Urology, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Stefan Duensing
- Molecular Urooncology, Department of Urology, University Hospital Heidelberg, Heidelberg, Germany
| | - Zoran Culig
- Experimental Urology, Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | - Achim Aigner
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, Faculty of Medicine, University of Leipzig, Germany
| | - Wolfram Klapper
- Institute of Pathology, Hematopathology Section and Lymph Node Registry, Universitätsklinikum Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Danny Jonigk
- Institute of Pathology, Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), German Center for Lung Research, Hannover, Germany
| | - Mark Philipp Kühnel
- Institute of Pathology, Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), German Center for Lung Research, Hannover, Germany
| | - Axel S Merseburger
- Department of Urology, University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Jutta Kirfel
- Institute of Pathology, University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Verena Sailer
- Institute of Pathology, University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Sven Perner
- Institute of Pathology, University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany.,Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| |
Collapse
|
27
|
Toptan T, Cantrell PS, Zeng X, Liu Y, Sun M, Yates NA, Chang Y, Moore PS. Proteomic approach to discover human cancer viruses from formalin-fixed tissues. JCI Insight 2020; 5:143003. [PMID: 33055416 PMCID: PMC7710300 DOI: 10.1172/jci.insight.143003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 10/07/2020] [Indexed: 12/11/2022] Open
Abstract
The challenge of discovering a completely new human tumor virus of unknown phylogeny or sequence depends on detecting viral molecules and differentiating them from host molecules in the virus-associated neoplasm. We developed differential peptide subtraction (DPS) using differential mass spectrometry (dMS) followed by targeted analysis to facilitate this discovery. We validated this approach by analyzing Merkel cell carcinoma (MCC), an aggressive human neoplasm, in which ~80% of cases are caused by the human Merkel cell polyomavirus (MCV). Approximately 20% of MCC have a high mutational burden and are negative for MCV, but are microscopically indistinguishable from virus positive cases. Using 23 (12 MCV+, 11 MCV-) formalin-fixed MCC, DPS identified both viral and human biomarkers (MCV large T antigen, CDKN2AIP, SERPINB5, and TRIM29) that discriminate MCV+ and MCV- MCC. Statistical analysis of 498,131 dMS features not matching the human proteome by DPS revealed 562 (0.11%) to be upregulated in virus-infected samples. Remarkably, 4 (20%) of the top 20 candidate MS spectra originated from MCV T oncoprotein peptides and confirmed by reverse translation degenerate oligonucleotide sequencing. DPS is a robust proteomic approach to identify potentially novel viral sequences in infectious tumors when nucleic acid-based methods are not feasible.
Collapse
Affiliation(s)
- Tuna Toptan
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Institute of Medical Virology, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | | | | | - Yang Liu
- Biomedical Mass Spectrometry Center and
| | - Mai Sun
- Biomedical Mass Spectrometry Center and
| | - Nathan A. Yates
- Biomedical Mass Spectrometry Center and
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yuan Chang
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Patrick S. Moore
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
28
|
Eberhardt W, Haeussler K, Nasrullah U, Pfeilschifter J. Multifaceted Roles of TRIM Proteins in Colorectal Carcinoma. Int J Mol Sci 2020; 21:ijms21207532. [PMID: 33066016 PMCID: PMC7590211 DOI: 10.3390/ijms21207532] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/06/2020] [Accepted: 10/07/2020] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most frequently diagnosed tumor in humans and one of the most common causes of cancer-related death worldwide. The pathogenesis of CRC follows a multistage process which together with somatic gene mutations is mainly attributed to the dysregulation of signaling pathways critically involved in the maintenance of homeostasis of epithelial integrity in the intestine. A growing number of studies has highlighted the critical impact of members of the tripartite motif (TRIM) protein family on most types of human malignancies including CRC. In accordance, abundant expression of many TRIM proteins has been observed in CRC tissues and is frequently correlating with poor survival of patients. Notably, some TRIM members can act as tumor suppressors depending on the context and the type of cancer which has been assessed. Mechanistically, most cancer-related TRIMs have a critical impact on cell cycle control, apoptosis, epithelial–mesenchymal transition (EMT), metastasis, and inflammation mainly through directly interfering with diverse oncogenic signaling pathways. In addition, some recent publications have emphasized the emerging role of some TRIM members to act as transcription factors and RNA-stabilizing factors thus adding a further level of complexity to the pleiotropic biological activities of TRIM proteins. The current review focuses on oncogenic signaling processes targeted by different TRIMs and their particular role in the development of CRC. A better understanding of the crosstalk of TRIMs with these signaling pathways relevant for CRC development is an important prerequisite for the validation of TRIM proteins as novel biomarkers and as potential targets of future therapies for CRC.
Collapse
|
29
|
Wikiniyadhanee R, Lerksuthirat T, Stitchantrakul W, Chitphuk S, Sura T, Dejsuphong D. TRIM29 is required for efficient recruitment of 53BP1 in response to DNA double-strand breaks in vertebrate cells. FEBS Open Bio 2020; 10:2055-2071. [PMID: 33017104 PMCID: PMC7530400 DOI: 10.1002/2211-5463.12954] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 07/18/2020] [Accepted: 08/14/2020] [Indexed: 12/17/2022] Open
Abstract
Tripartite motif‐containing protein 29 (TRIM29) is involved in DNA double‐strand break (DSB) repair. However, the specific roles of TRIM29 in DNA repair are not clearly understood. To investigate the involvement of TRIM29 in DNA DSB repair, we disrupted TRIM29 in DT40 cells by gene targeting with homologous recombination (HR). The roles of TRIM29 were investigated by clonogenic survival assays and immunofluorescence analyses. TRIM29 triallelic knockout (TRIM29−/−/−/+) cells were sensitive to etoposide, but resistant to camptothecin. Foci formation assays to assess DNA repair activities showed that the dissociation of etoposide‐induced phosphorylated H2A histone family member X (ɣ‐H2AX) foci was retained in TRIM29−/−/−/+ cells, and the formation of etoposide‐induced tumor suppressor p53‐binding protein 1 (53BP1) foci in TRIM29−/−/−/+ cells was slower compared with wild‐type (WT) cells. Interestingly, the kinetics of camptothecin‐induced RAD51 foci formation of TRIM29−/−/−/+ cells was higher than that of WT cells. These results indicate that TRIM29 is required for efficient recruitment of 53BP1 to facilitate the nonhomologous end‐joining (NHEJ) pathway and thereby suppress the HR pathway in response to DNA DSBs. TRIM29 regulates the choice of DNA DSB repair pathway by facilitating 53BP1 accumulation to promote NHEJ and may have potential for development into a therapeutic target to sensitize refractory cancers or as biomarker of personalized therapies.
Collapse
Affiliation(s)
- Rakkreat Wikiniyadhanee
- Section for Translational Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Tassanee Lerksuthirat
- Research Center, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Wasana Stitchantrakul
- Research Center, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Sermsiri Chitphuk
- Research Center, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Thanyachai Sura
- Department of Internal Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Donniphat Dejsuphong
- Section for Translational Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
30
|
Sun Q, Ye Z, Qin Y, Fan G, Ji S, Zhuo Q, Xu W, Liu W, Hu Q, Liu M, Zhang Z, Xu X, Yu X. Oncogenic function of TRIM2 in pancreatic cancer by activating ROS-related NRF2/ITGB7/FAK axis. Oncogene 2020; 39:6572-6588. [PMID: 32929153 DOI: 10.1038/s41388-020-01452-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 07/29/2020] [Accepted: 09/02/2020] [Indexed: 02/06/2023]
Abstract
Evidence suggests that tripartite motif-containing 2 (TRIM2) is associated with carcinogenic effects in several malignancies. However, the expression patterns and roles of TRIM2 in pancreatic cancer are rarely studied. Our study demonstrated that TRIM2 was expressed in a high percentage of pancreatic tumors. High TRIM2 expression was negatively correlated with the outcome of pancreatic cancer. TRIM2 silencing significantly inhibited the proliferation, migration, invasion, and in vivo tumorigenicity of pancreatic cancer cells. Regarding the mechanism involved, TRIM2 activated ROS-related E2-related factor 2 (NRF2)/antioxidant response element (ARE) signaling and the integrin/focal adhesion kinase (FAK) pathway. Treatment of pancreatic cancer cells with the antioxidant N-acetyl-L-cysteine decreased ROS activity and expression level of NRF2 and ITGB7. Increased translocation of NRF2 protein into nucleus further rescued the inhibited ITGB7 transcription. Moreover, NRF2 bound to the potential ARE on the promoter region and enhanced the transcriptional activity of ITGB7, indicating the bridging effect of NRF2 between the two signaling pathways. In summary, our study provides evidence that upregulated TRIM2 in pancreatic cancer predicts short survival for pancreatic cancer patients. TRIM2 accelerates pancreatic cancer progression via the ROS-related NRF2/ITGB7/FAK axis.
Collapse
Affiliation(s)
- Qiqing Sun
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, 200032, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, 200032, Shanghai, China
- Shanghai Pancreatic Cancer Institute, 200032, Shanghai, China
| | - Zeng Ye
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, 200032, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, 200032, Shanghai, China
- Shanghai Pancreatic Cancer Institute, 200032, Shanghai, China
| | - Yi Qin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, 200032, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, 200032, Shanghai, China
- Shanghai Pancreatic Cancer Institute, 200032, Shanghai, China
| | - Guixiong Fan
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, 200032, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, 200032, Shanghai, China
- Shanghai Pancreatic Cancer Institute, 200032, Shanghai, China
| | - Shunrong Ji
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, 200032, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, 200032, Shanghai, China
- Shanghai Pancreatic Cancer Institute, 200032, Shanghai, China
| | - Qifeng Zhuo
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, 200032, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, 200032, Shanghai, China
- Shanghai Pancreatic Cancer Institute, 200032, Shanghai, China
| | - Wenyan Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, 200032, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, 200032, Shanghai, China
- Shanghai Pancreatic Cancer Institute, 200032, Shanghai, China
| | - Wensheng Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, 200032, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, 200032, Shanghai, China
- Shanghai Pancreatic Cancer Institute, 200032, Shanghai, China
| | - Qiangsheng Hu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, 200032, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, 200032, Shanghai, China
- Shanghai Pancreatic Cancer Institute, 200032, Shanghai, China
| | - Mengqi Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, 200032, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, 200032, Shanghai, China
- Shanghai Pancreatic Cancer Institute, 200032, Shanghai, China
| | - Zheng Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, 200032, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, 200032, Shanghai, China
- Shanghai Pancreatic Cancer Institute, 200032, Shanghai, China
| | - Xiaowu Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 200032, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, 200032, Shanghai, China.
- Pancreatic Cancer Institute, Fudan University, 200032, Shanghai, China.
- Shanghai Pancreatic Cancer Institute, 200032, Shanghai, China.
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 200032, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, 200032, Shanghai, China.
- Pancreatic Cancer Institute, Fudan University, 200032, Shanghai, China.
- Shanghai Pancreatic Cancer Institute, 200032, Shanghai, China.
| |
Collapse
|
31
|
Luo S, Shen M, Chen H, Li W, Chen C. Long non‑coding RNA TP73‑AS1 accelerates the progression and cisplatin resistance of non‑small cell lung cancer by upregulating the expression of TRIM29 via competitively targeting microRNA‑34a‑5p. Mol Med Rep 2020; 22:3822-3832. [PMID: 32901838 PMCID: PMC7533438 DOI: 10.3892/mmr.2020.11473] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 05/29/2020] [Indexed: 12/22/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is a leading subtype of lung cancer, with high mortality rates. Recently, long non-coding RNAs (lncRNAs) have been associated with NSCLC. The present study aimed to examine the role of the TP73 antisense RNA 1 (TP73-AS1) lncRNA in NSCLC. TP73-AS1 and microRNA(miR)-34a-5p expression levels were measured using reverse transcription-quantitative PCR (RT-qPCR) and chromogenic in situ hybridization (CISH). Cell proliferation, apoptosis, migration and invasion was determined using Cell Counting Kit-8 (CCK-8), flow cytometry, Transwell and Matrigel assays, respectively. The median inhibitory concentration (IC50) value of cisplatin (cis-diamminedichloroplatinum; DDP) was assessed using a CCK-8 assay. The interaction between miR-34a-5p and TP73-AS1 or tripartite motif-containing 29 (TRIM29) was predicted using microRNA.org and Starbase, then verified using a dual-luciferase reporter assay. The expression of TRIM29 was quantified at the mRNA and protein level using RT-qPCR and western blot analysis, respectively. TP73-AS1 was significantly upregulated, while miR-34a-5p was downregulated in NSCLC tissues and cells. Functionally, TP73-AS1 knockdown inhibited proliferation, migration, invasion and DDP resistance, whilst inducing apoptosis in NSCLC cells. miR-34a-5p was identified as a target for TP73-AS1, and its inhibition reversed the effects of TP73-AS1 knockdown on NSCLC cells. In addition, TRIM29 was targeted by miR-34a-5p, and its overexpression reversed the effects of miR-34a-5p. Moreover, TP73-AS1 acted as a molecular sponge for miR-34a-5p, increasing the expression of TRIM29. In conclusion, TP73-AS1 contributed to proliferation, migration and DDP resistance but inhibited apoptosis of NSCLC cells by upregulating TRIM29 and sponging miR-34a-5p.
Collapse
Affiliation(s)
- Shunxiang Luo
- Department of Oncology, The First People's Hospital of Tianmen, Tianmen, Hubei 431700, P.R. China
| | - Ming Shen
- Department of Oncology, The First People's Hospital of Tianmen, Tianmen, Hubei 431700, P.R. China
| | - Hui Chen
- Department of Oncology, The First People's Hospital of Tianmen, Tianmen, Hubei 431700, P.R. China
| | - Weiwei Li
- Department of Oncology, The First People's Hospital of Tianmen, Tianmen, Hubei 431700, P.R. China
| | - Cong Chen
- Department of Oncology, The First People's Hospital of Tianmen, Tianmen, Hubei 431700, P.R. China
| |
Collapse
|
32
|
Akçimen F, Martins S, Liao C, Bourassa CV, Catoire H, Nicholson GA, Riess O, Raposo M, França MC, Vasconcelos J, Lima M, Lopes-Cendes I, Saraiva-Pereira ML, Jardim LB, Sequeiros J, Dion PA, Rouleau GA. Genome-wide association study identifies genetic factors that modify age at onset in Machado-Joseph disease. Aging (Albany NY) 2020; 12:4742-4756. [PMID: 32205469 PMCID: PMC7138549 DOI: 10.18632/aging.102825] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 01/27/2020] [Indexed: 02/07/2023]
Abstract
Machado-Joseph disease (MJD/SCA3) is the most common form of dominantly inherited ataxia worldwide. The disorder is caused by an expanded CAG repeat in the ATXN3 gene. Past studies have revealed that the length of the expansion partly explains the disease age at onset (AO) variability of MJD, which is confirmed in this study (Pearson’s correlation coefficient R2 = 0.62). Using a total of 786 MJD patients from five different geographical origins, a genome-wide association study (GWAS) was conducted to identify additional AO modifying factors that could explain some of the residual AO variability. We identified nine suggestively associated loci (P < 1 × 10−5). These loci were enriched for genes involved in vesicle transport, olfactory signaling, and synaptic pathways. Furthermore, associations between AO and the TRIM29 and RAG genes suggests that DNA repair mechanisms might be implicated in MJD pathogenesis. Our study demonstrates the existence of several additional genetic factors, along with CAG expansion, that may lead to a better understanding of the genotype-phenotype correlation in MJD.
Collapse
Affiliation(s)
- Fulya Akçimen
- Department of Human Genetics, McGill University, Montréal, Québec, Canada.,Montreal Neurological Institute and Hospital, McGill University, Montréal, Québec, Canada
| | - Sandra Martins
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
| | - Calwing Liao
- Department of Human Genetics, McGill University, Montréal, Québec, Canada.,Montreal Neurological Institute and Hospital, McGill University, Montréal, Québec, Canada
| | - Cynthia V Bourassa
- Montreal Neurological Institute and Hospital, McGill University, Montréal, Québec, Canada.,Department of Neurology and Neurosurgery, McGill University, Montréal, Québec, Canada
| | - Hélène Catoire
- Montreal Neurological Institute and Hospital, McGill University, Montréal, Québec, Canada.,Department of Neurology and Neurosurgery, McGill University, Montréal, Québec, Canada
| | - Garth A Nicholson
- University of Sydney, Department of Medicine, Concord Hospital, Concord, Australia
| | - Olaf Riess
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany
| | - Mafalda Raposo
- Faculdade de Ciências e Tecnologia, Universidade dos Açores e Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Marcondes C França
- Department of Neurology, Faculty of Medical Sciences, UNICAMP, São Paulo, Campinas, Brazil
| | - João Vasconcelos
- School of Medical Sciences, Department of Medical Genetics and Genomic Medicine, University of Campinas (UNICAMP), São Paulo, Campinas, Brazil
| | - Manuela Lima
- Faculdade de Ciências e Tecnologia, Universidade dos Açores e Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Iscia Lopes-Cendes
- The Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), São Paulo, Campinas, Brazil.,Departamento de Neurologia, Hospital do Divino Espírito Santo, Ponta Delgada, Portugal
| | - Maria Luiza Saraiva-Pereira
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil.,Depto. de Bioquímica - ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Laura B Jardim
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil.,Depto de Medicina Interna, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Jorge Sequeiros
- IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal.,Institute for Molecular and Cell Biology (IBMC), Universidade do Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Patrick A Dion
- Montreal Neurological Institute and Hospital, McGill University, Montréal, Québec, Canada.,Department of Neurology and Neurosurgery, McGill University, Montréal, Québec, Canada
| | - Guy A Rouleau
- Department of Human Genetics, McGill University, Montréal, Québec, Canada.,Montreal Neurological Institute and Hospital, McGill University, Montréal, Québec, Canada.,Department of Neurology and Neurosurgery, McGill University, Montréal, Québec, Canada
| |
Collapse
|
33
|
Yamada Y, Kimura N, Takayama KI, Sato Y, Suzuki T, Azuma K, Fujimura T, Ikeda K, Kume H, Inoue S. TRIM44 promotes cell proliferation and migration by inhibiting FRK in renal cell carcinoma. Cancer Sci 2020; 111:881-890. [PMID: 31883420 PMCID: PMC7060480 DOI: 10.1111/cas.14295] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 11/21/2019] [Accepted: 12/21/2019] [Indexed: 12/12/2022] Open
Abstract
TRIM44 has oncogenic roles in various cancers. However, TRIM44 expression and its function in renal cell carcinoma (RCC) are still unknown. Here in this study, we investigated the clinical significance of TRIM44 and its biological function in RCC. TRIM44 overexpression was significantly associated with clinical M stage, histologic type (clear cell) and presence of lymphatic invasion (P = .047, P = .005, and P = .028, respectively). Moreover, TRIM44 overexpression was significantly associated with poor prognosis in terms of cancer‐specific survival (P = .019). Gain‐of‐function and loss‐of‐function studies using TRIM44 and siTRIM44 transfection showed that TRIM44 promotes cell proliferation and cell migration in two RCC cell lines, Caki1 and 769P. To further investigate the role of TRIM44 in RCC, we performed integrated microarray analysis in Caki1 and 769P cells and explored the data in the Oncomine database. Interestingly, FRK was identified as a promising candidate target gene of TRIM44, which was downregulated in RCC compared with normal renal tissues. We found that cell proliferation was inhibited by TRIM44 knockdown and then recovered by siFRK treatment. Taken together, the present study revealed the association between high expression of TRIM44 and poor prognosis in RCC patients and that TRIM44 promotes cell proliferation by regulating FRK.
Collapse
Affiliation(s)
- Yuta Yamada
- Department of Urology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Naoki Kimura
- Department of Urology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Systems Aging Science and Medicine, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Ken-Ichi Takayama
- Department of Systems Aging Science and Medicine, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Yusuke Sato
- Department of Urology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takashi Suzuki
- Department of Pathology and Histotechnology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kotaro Azuma
- Department of Systems Aging Science and Medicine, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | | | - Kazuhiro Ikeda
- Division of Gene Regulation and Signal Transduction, Research Center of Genomic Medicine, Saitama Medical University, Saitama, Japan
| | - Haruki Kume
- Department of Urology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Satoshi Inoue
- Department of Systems Aging Science and Medicine, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan.,Division of Gene Regulation and Signal Transduction, Research Center of Genomic Medicine, Saitama Medical University, Saitama, Japan
| |
Collapse
|
34
|
Fink D, Yau T, Nabbi A, Wagner B, Wagner C, Hu SM, Lang V, Handschuh S, Riabowol K, Rülicke T. Loss of Ing3 Expression Results in Growth Retardation and Embryonic Death. Cancers (Basel) 2019; 12:cancers12010080. [PMID: 31905726 PMCID: PMC7017303 DOI: 10.3390/cancers12010080] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/19/2019] [Accepted: 12/24/2019] [Indexed: 12/29/2022] Open
Abstract
The ING3 candidate tumour suppressor belongs to a family of histone modifying proteins involved in regulating cell proliferation, senescence, apoptosis, chromatin remodeling, and DNA repair. It is a stoichiometric member of the minimal NuA4 histone acetyl transferase (HAT) complex consisting of EAF6, EPC1, ING3, and TIP60. This complex is responsible for the transcription of an essential cascade of genes involved in embryonic development and in tumour suppression. ING3 has been linked to head and neck and hepatocellular cancers, although its status as a tumour suppressor has not been well established. Recent studies suggest a pro-metastasis role in prostate cancer progression. Here, we describe a transgenic mouse strain with insertional mutation of an UbC-mCherry expression cassette into the endogenous Ing3 locus, resulting in the disruption of ING3 protein expression. Homozygous mutants are embryonically lethal, display growth retardation, and severe developmental disorders. At embryonic day (E) 10.5, the last time point viable homozygous embryos were found, they were approximately half the size of heterozygous mice that develop normally. µCT analysis revealed a developmental defect in neural tube closure, resulting in the failure of formation of closed primary brain vesicles in homozygous mid-gestation embryos. This is consistent with high ING3 expression levels in the embryonic brains of heterozygous and wild type mice and its lack in homozygous mutant embryos that show a lack of ectodermal differentiation. Our data provide direct evidence that ING3 is an essential factor for normal embryonic development and that it plays a fundamental role in prenatal brain formation.
Collapse
Affiliation(s)
- Dieter Fink
- Institute of Laboratory Animal Science, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (T.Y.); (B.W.); (S.M.H.); (V.L.); (T.R.)
- Correspondence: ; Tel.: +43-(0)-1-25077-2820
| | - Tienyin Yau
- Institute of Laboratory Animal Science, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (T.Y.); (B.W.); (S.M.H.); (V.L.); (T.R.)
| | - Arash Nabbi
- Departments of Biochemistry & Molecular Biology and Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; (A.N.); (K.R.)
| | - Bettina Wagner
- Institute of Laboratory Animal Science, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (T.Y.); (B.W.); (S.M.H.); (V.L.); (T.R.)
| | - Christine Wagner
- Division of Immunology, Allergy and Infectious Diseases (DIAID), Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria;
| | - Shiting Misaki Hu
- Institute of Laboratory Animal Science, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (T.Y.); (B.W.); (S.M.H.); (V.L.); (T.R.)
| | - Viktor Lang
- Institute of Laboratory Animal Science, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (T.Y.); (B.W.); (S.M.H.); (V.L.); (T.R.)
| | - Stephan Handschuh
- VetImaging, VetCore Facility for Research, University of Veterinary Medicine Vienna, 1210 Vienna, Austria;
| | - Karl Riabowol
- Departments of Biochemistry & Molecular Biology and Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; (A.N.); (K.R.)
| | - Thomas Rülicke
- Institute of Laboratory Animal Science, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (T.Y.); (B.W.); (S.M.H.); (V.L.); (T.R.)
| |
Collapse
|
35
|
Cao Y, Shi L, Wang M, Hou J, Wei Y, Du C. ATDC contributes to sustaining the growth and invasion of glioma cells through regulating Wnt/β-catenin signaling. Chem Biol Interact 2019; 305:148-155. [DOI: 10.1016/j.cbi.2019.03.033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 03/07/2019] [Accepted: 03/26/2019] [Indexed: 02/09/2023]
|
36
|
Chesnokova V, Zonis S, Barrett R, Kameda H, Wawrowsky K, Ben-Shlomo A, Yamamoto M, Gleeson J, Bresee C, Gorbunova V, Melmed S. Excess growth hormone suppresses DNA damage repair in epithelial cells. JCI Insight 2019; 4:e125762. [PMID: 30728323 PMCID: PMC6413789 DOI: 10.1172/jci.insight.125762] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 12/20/2018] [Indexed: 12/18/2022] Open
Abstract
Growth hormone (GH) decreases with age, and GH therapy has been advocated by some to sustain lean muscle mass and vigor in aging patients and advocated by athletes to enhance performance. Environmental insults and aging lead to DNA damage, which - if unrepaired - results in chromosomal instability and tumorigenesis. We show that GH suppresses epithelial DNA damage repair and blocks ataxia telangiectasia mutated (ATM) kinase autophosphorylation with decreased activity. Decreased phosphorylation of ATM target proteins p53, checkpoint kinase 2 (Chk2), and histone 2A variant led to decreased DNA repair by nonhomologous end-joining. In vivo, prolonged high GH levels resulted in a 60% increase in unrepaired colon epithelial DNA damage. GH suppression of ATM was mediated by induced tripartite motif containing protein 29 (TRIM29) and attenuated tat interacting protein 60 kDa (Tip60). By contrast, DNA repair was increased in human nontumorous colon cells (hNCC) where GH receptor (GHR) was stably suppressed and in colon tissue derived from GHR-/- mice. hNCC treated with etoposide and GH showed enhanced transformation, as evidenced by increased growth in soft agar. In mice bearing human colon GH-secreting xenografts, metastatic lesions were increased. The results elucidate a mechanism underlying GH-activated epithelial cell transformation and highlight an adverse risk for inappropriate adult GH treatment.
Collapse
Affiliation(s)
| | | | - Robert Barrett
- Board of Governors Regenerative Medicine Institute
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Department of Medicine, and
| | | | | | | | | | - John Gleeson
- Board of Governors Regenerative Medicine Institute
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Department of Medicine, and
| | - Catherine Bresee
- Biostatistics and Bioinformatics Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Vera Gorbunova
- Department of Biology, University of Rochester, Rochester, New York, USA
| | | |
Collapse
|
37
|
Yanagi T, Watanabe M, Hata H, Kitamura S, Imafuku K, Yanagi H, Homma A, Wang L, Takahashi H, Shimizu H, Hatakeyama S. Loss of TRIM29 Alters Keratin Distribution to Promote Cell Invasion in Squamous Cell Carcinoma. Cancer Res 2018; 78:6795-6806. [PMID: 30389700 DOI: 10.1158/0008-5472.can-18-1495] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 08/28/2018] [Accepted: 10/10/2018] [Indexed: 11/16/2022]
Abstract
: TRIM29 (tripartite motif-containing protein 29) is a TRIM family protein that has been implicated in breast, colorectal, and pancreatic cancers. However, its role in stratified squamous epithelial cells and tumors has not been elucidated. Here, we investigate the expression of TRIM29 in cutaneous head and neck squamous cell carcinomas (SCC) and its functions in the tumorigenesis of such cancers. TRIM29 expression was lower in malignant SCC lesions than in adjacent normal epithelial tissue or benign tumors. Lower expression of TRIM29 was associated with higher SCC invasiveness. Primary tumors of cutaneous SCC showed aberrant hypermethylation of TRIM29. Depletion of TRIM29 increased cancer cell migration and invasion; conversely, overexpression of TRIM29 suppressed these. Comprehensive proteomics and immunoprecipitation analyses identified keratins and keratin-interacting protein FAM83H as TRIM29 interactors. Knockdown of TRIM29 led to ectopic keratin localization of keratinocytes. In primary tumors, lower TRIM29 expression correlated with the altered expression of keratins. Our findings reveal an unexpected role for TRIM29 in regulating the distribution of keratins, as well as in the migration and invasion of SCC. They also suggest that the TRIM29-keratin axis could serve as a diagnostic and prognostic marker in stratified epithelial tumors and may provide a target for SCC therapeutics. SIGNIFICANCE: These findings identify TRIM29 as a novel diagnostic and prognostic marker in stratified epithelial tissues.
Collapse
Affiliation(s)
- Teruki Yanagi
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan.
| | - Masashi Watanabe
- Department of Biochemistry, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Hokkaido, Japan
| | - Hiroo Hata
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Shinya Kitamura
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Keisuke Imafuku
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroko Yanagi
- Department of Otolaryngology-Head and Neck Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Hokkaido, Japan
| | - Akihiro Homma
- Department of Otolaryngology-Head and Neck Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Hokkaido, Japan
| | - Lei Wang
- Department of Cancer Pathology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Hokkaido, Japan.,Global Station for Soft Matter, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Hokkaido, Japan
| | - Hidehisa Takahashi
- Department of Molecular Biology, Yokohama City University Graduate School of Medical Science, Yokohama, Japan
| | - Hiroshi Shimizu
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Shigetsugu Hatakeyama
- Department of Biochemistry, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Hokkaido, Japan.
| |
Collapse
|
38
|
Kimura N, Yamada Y, Takayama KI, Fujimura T, Takahashi S, Kume H, Inoue S. Androgen-responsive tripartite motif 36 enhances tumor-suppressive effect by regulating apoptosis-related pathway in prostate cancer. Cancer Sci 2018; 109:3840-3852. [PMID: 30238687 PMCID: PMC6272107 DOI: 10.1111/cas.13803] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 09/12/2018] [Accepted: 09/18/2018] [Indexed: 12/18/2022] Open
Abstract
Tripartite motif 36 (TRIM36) belongs to the TRIM family, most members of which are involved in ubiquitination and degradation of target proteins by functioning as E3 ubiquitin ligases. The function of TRIM36 has not been well documented, therefore, we investigated the clinical significance and function of TRIM36 in human prostate cancer (PC). Multivariate logistic regression analysis showed that TRIM36 immunoreactivity was an independent predictor of cancer‐specific survival of PC patients. Gain‐of‐function study revealed that overexpression of TRIM36 suppressed cell proliferation and migration of LNCaP, 22Rv1, and DU145 cells. Moreover, TRIM36 knockdown using siRNA suppressed apoptosis and promoted cell proliferation and migration in LNCaP and 22Rv1 cells. Furthermore, our microarray analysis revealed that the apoptosis‐related pathway was significantly upregulated by TRIM36 overexpression. The TUNEL assay showed that apoptosis promoted by docetaxel treatment was alleviated in siTRIM36‐treated LNCaP and 22Rv1 cells. Taken together, these results suggest that high expression of TRIM36 is associated with favorable prognosis and that TRIM36 plays a tumor‐suppressive role by inhibiting cell proliferation and migration as well as promoting apoptosis in PC.
Collapse
Affiliation(s)
- Naoki Kimura
- Department of Functional Biogerontology, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan.,Department of Urology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yuta Yamada
- Department of Urology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Urology, Chiba-Tokushukai Hospital, Chiba, Japan
| | - Ken-Ichi Takayama
- Department of Functional Biogerontology, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | | | - Satoru Takahashi
- Department of Urology, Nihon University School of Medicine, Tokyo, Japan
| | - Haruki Kume
- Department of Urology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Satoshi Inoue
- Department of Functional Biogerontology, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan.,Division of Gene Regulation and Signal Transduction, Research Center of Genomic Medicine, Saitama Medical University, Saitama, Japan
| |
Collapse
|
39
|
Xing J, Zhang A, Minze LJ, Li XC, Zhang Z. TRIM29 Negatively Regulates the Type I IFN Production in Response to RNA Virus. THE JOURNAL OF IMMUNOLOGY 2018; 201:183-192. [PMID: 29769269 DOI: 10.4049/jimmunol.1701569] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 04/30/2018] [Indexed: 12/25/2022]
Abstract
The innate immunity is critically important in protection against virus infections, and in the case of RNA viral infections, the signaling mechanisms that initiate robust protective innate immunity without triggering autoimmune inflammation remain incompletely defined. In this study, we found the E3 ligase TRIM29 was specifically expressed in poly I:C-stimulated human myeloid dendritic cells. The induced TRIM29 played a negative role in type I IFN production in response to poly I:C or dsRNA virus reovirus infection. Importantly, the challenge of wild-type mice with reovirus led to lethal infection. In contrast, deletion of TRIM29 protected the mice from this developing lethality. Additionally, TRIM29-/- mice have lower titers of reovirus in the heart, intestine, spleen, liver, and brain because of elevated production of type I IFN. Mechanistically, TRIM29 was shown to interact with MAVS and subsequently induce its K11-linked ubiquitination and degradation. Taken together, TRIM29 regulates negatively the host innate immune response to RNA virus, which could be employed by RNA viruses for viral pathogenesis.
Collapse
Affiliation(s)
- Junji Xing
- Department of Surgery, Houston Methodist, Houston, TX 77030.,Immunobiology and Transplant Science Center, Houston Methodist, Houston, TX 77030
| | - Ao Zhang
- Department of Surgery, Houston Methodist, Houston, TX 77030.,Immunobiology and Transplant Science Center, Houston Methodist, Houston, TX 77030.,State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; and
| | - Laurie J Minze
- Department of Surgery, Houston Methodist, Houston, TX 77030.,Immunobiology and Transplant Science Center, Houston Methodist, Houston, TX 77030
| | - Xian Chang Li
- Department of Surgery, Houston Methodist, Houston, TX 77030.,Immunobiology and Transplant Science Center, Houston Methodist, Houston, TX 77030.,Department of Surgery, Weill Cornell Medical College of Cornell University, New York, NY 10065
| | - Zhiqiang Zhang
- Department of Surgery, Houston Methodist, Houston, TX 77030; .,Immunobiology and Transplant Science Center, Houston Methodist, Houston, TX 77030.,Department of Surgery, Weill Cornell Medical College of Cornell University, New York, NY 10065
| |
Collapse
|
40
|
Liang C, Dong H, Miao C, Zhu J, Wang J, Li P, Li J, Wang Z. TRIM29 as a prognostic predictor for multiple human malignant neoplasms: a systematic review and meta-analysis. Oncotarget 2017; 9:12323-12332. [PMID: 29552313 PMCID: PMC5844749 DOI: 10.18632/oncotarget.23617] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 10/28/2017] [Indexed: 12/20/2022] Open
Abstract
Recent studies have shown that tripartite motif-containing protein 29 (TRIM29) had prognostic values in several cancers. However, different studies have been inconsistent. We conducted a meta-analysis to elucidate the precise predictive value of TRIM29 in various human malignant disease. Eleven eligible studies with 2046 patients were ultimately enrolled in this meta-analysis. Heterogeneity between studies was assessed using I2 statistics. Pooled Hazard ratios (HRs) with 95% confidence intervals (CIs) for patient survival and disease recurrence were calculated to investigate the correlation between TRIM29 expression and cancer prognosis. The results identified an important link between upregulated TRIM29 expression and poor prognosis in patients with multiple human malignant neoplasms in terms of recurrence-free survival (RFS)/disease-free survival (DFS) (HR = 1.66, 95% CI 1.36–2.04) but favorable progression-free survival (PFS)/metastasis-free survival (MFS) (HR = 0.37, 95% CI 0.16–0.85). We found that high TRIM29 expression predicted no significant impact on overall survival (OS) (HR = 1.32, 95% CI 0.90–1.93). Subgroup analyses showed that high TRIM29 expression predicted poor OS in Asians (HR = 2.21, 95% CI 1.78–2.74) but favorable OS in Caucasian (HR = 0.47, 95% CI 0.25–0.89). TRIM29 might play an essential role in carcinogenesis of multiple human malignant neoplasms and could serve as a biomarker for the prediction of patients’ prognosis.
Collapse
Affiliation(s)
- Chao Liang
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Huiyu Dong
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chenkui Miao
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jundong Zhu
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jie Wang
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Pu Li
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jie Li
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zengjun Wang
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
41
|
Choi SK, Pandiyan K, Eun JW, Yang X, Hong SH, Nam SW, Jones PA, Liang G, You JS. Epigenetic landscape change analysis during human EMT sheds light on a key EMT mediator TRIM29. Oncotarget 2017; 8:98322-98335. [PMID: 29228692 PMCID: PMC5716732 DOI: 10.18632/oncotarget.21681] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 09/22/2017] [Indexed: 11/25/2022] Open
Abstract
Epithelial to mesenchymal transition (EMT) is a key trans-differentiation process, which plays a critical role in physiology and pathology. Although gene expression changes in EMT have been scrutinized, study of epigenome is in its infancy. To understand epigenetic changes during TWIST-driven EMT, we used the AcceSssIble assay to study DNA methylation and chromatin accessibility in human mammary epithelial cells (HMECs). The DNA methylation changes were found to have functional significance in EMT - i.e. methylated genes were enriched for E-box motifs that can be recognized by TWIST, at the promoters suggesting a potential targeting phenomenon, whereas the demethylated regions were enriched for pro-metastatic genes, supporting the role of EMT in metastasis. TWIST-induced EMT triggers alterations in chromatin accessibility both independent of and dependent on DNA methylation changes, primarily resulting in closed chromatin conformation. By overlapping the genes, whose chromatin structure is changed during early EMT and a known "core EMT signature", we identified 18 driver candidate genes during EMT, 14 upregulated and 4 downregulated genes with corresponding chromatin structure changes. Among 18 genes, we focused on TRIM29 as a novel marker of EMT. Although loss of TRIM29 is insufficient to suppress CDH, it is enough to induce CDH2 and VIM. Gene functional annotation analysis shows the involvement of TRIM29 in epidermal development, cell differentiation and cell migration. Taken together, our results provide a robust snapshot of chromatin state during human EMT and identify TRIM29 as a core mediator of EMT.
Collapse
Affiliation(s)
- Sung Kyung Choi
- Department of Biochemistry, School of Medicine, Konkuk University, Seoul, Korea
| | - Kurinji Pandiyan
- Departments of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jung Woo Eun
- Department of Pathology, College of Medicine, The Catholic University, Seoul, Korea
| | - Xiaojing Yang
- Departments of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Seong Hwi Hong
- Department of Biochemistry, School of Medicine, Konkuk University, Seoul, Korea
| | - Suk Woo Nam
- Department of Pathology, College of Medicine, The Catholic University, Seoul, Korea
| | | | - Gangning Liang
- Departments of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jueng Soo You
- Department of Biochemistry, School of Medicine, Konkuk University, Seoul, Korea.,Research Institute of Medical Science, KonKuk University School of Medicine, Seoul, Korea
| |
Collapse
|
42
|
Ebner P, Versteeg GA, Ikeda F. Ubiquitin enzymes in the regulation of immune responses. Crit Rev Biochem Mol Biol 2017; 52:425-460. [PMID: 28524749 PMCID: PMC5490640 DOI: 10.1080/10409238.2017.1325829] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 04/06/2017] [Accepted: 04/28/2017] [Indexed: 12/25/2022]
Abstract
Ubiquitination plays a central role in the regulation of various biological functions including immune responses. Ubiquitination is induced by a cascade of enzymatic reactions by E1 ubiquitin activating enzyme, E2 ubiquitin conjugating enzyme, and E3 ubiquitin ligase, and reversed by deubiquitinases. Depending on the enzymes, specific linkage types of ubiquitin chains are generated or hydrolyzed. Because different linkage types of ubiquitin chains control the fate of the substrate, understanding the regulatory mechanisms of ubiquitin enzymes is central. In this review, we highlight the most recent knowledge of ubiquitination in the immune signaling cascades including the T cell and B cell signaling cascades as well as the TNF signaling cascade regulated by various ubiquitin enzymes. Furthermore, we highlight the TRIM ubiquitin ligase family as one of the examples of critical E3 ubiquitin ligases in the regulation of immune responses.
Collapse
|
43
|
Watanabe M, Hatakeyama S. TRIM proteins and diseases. J Biochem 2017; 161:135-144. [PMID: 28069866 DOI: 10.1093/jb/mvw087] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 09/20/2016] [Indexed: 12/20/2022] Open
Abstract
Ubiquitination is one of the posttranslational modifications that regulates a number of intracellular events including signal transduction, protein quality control, transcription, cell cycle, apoptosis and development. The ubiquitin system functions as a garbage machine to degrade target proteins and as a regulator for several signalling pathways. Biochemical reaction of ubiquitination requires several enzymes including E1, E2 and E3, and E3 ubiquitin ligases play roles as receptors for recognizing target proteins. Most of the tripartite motif (TRIM) proteins are E3 ubiquitin ligases. Recent studies have shown that some TRIM proteins function as important regulators for a variety of diseases including cancer, inflammatory diseases, infectious diseases, neuropsychiatric disorders, chromosomal abnormalities and developmental diseases. In this review, we summarize the involvement of TRIM proteins in the aetiology of various diseases.
Collapse
Affiliation(s)
- Masashi Watanabe
- Department of Biochemistry, Hokkaido University Graduate School of Medicine, Kita 15, Nishi 7, Kita-ku, Sapporo, Hokkaido 060-8638, Japan
| | - Shigetsugu Hatakeyama
- Department of Biochemistry, Hokkaido University Graduate School of Medicine, Kita 15, Nishi 7, Kita-ku, Sapporo, Hokkaido 060-8638, Japan
| |
Collapse
|
44
|
Hatakeyama S. TRIM Family Proteins: Roles in Autophagy, Immunity, and Carcinogenesis. Trends Biochem Sci 2017; 42:297-311. [DOI: 10.1016/j.tibs.2017.01.002] [Citation(s) in RCA: 635] [Impact Index Per Article: 79.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 12/28/2016] [Accepted: 01/02/2017] [Indexed: 01/19/2023]
|
45
|
Xu J, Li Z, Su Q, Zhao J, Ma J. TRIM29 promotes progression of thyroid carcinoma via activating P13K/AKT signaling pathway. Oncol Rep 2017; 37:1555-1564. [DOI: 10.3892/or.2017.5364] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 12/28/2016] [Indexed: 11/05/2022] Open
|
46
|
Xing J, Weng L, Yuan B, Wang Z, Jia L, Jin R, Lu H, Li XC, Liu YJ, Zhang Z. Identification of a role for TRIM29 in the control of innate immunity in the respiratory tract. Nat Immunol 2016; 17:1373-1380. [PMID: 27695001 PMCID: PMC5558830 DOI: 10.1038/ni.3580] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 09/02/2016] [Indexed: 12/13/2022]
Abstract
The respiratory tract is heavily populated with innate immune cells, but the mechanisms that control such cells are poorly defined. Here we found that the E3 ubiquitin ligase TRIM29 was a selective regulator of the activation of alveolar macrophages, the expression of type I interferons and the production of proinflammatory cytokines in the lungs. We found that deletion of TRIM29 enhanced macrophage production of type I interferons and protected mice from infection with influenza virus, while challenge of Trim29-/- mice with Haemophilus influenzae resulted in lethal lung inflammation due to massive production of proinflammatory cytokines by macrophages. Mechanistically, we demonstrated that TRIM29 inhibited interferon-regulatory factors and signaling via the transcription factor NF-κB by degrading the adaptor NEMO and that TRIM29 directly bound NEMO and subsequently induced its ubiquitination and proteolytic degradation. These data identify TRIM29 as a key negative regulator of alveolar macrophages and might have important clinical implications for local immunity and immunopathology.
Collapse
Affiliation(s)
- Junji Xing
- Immunobiology and Transplant Research Center, Houston Methodist Research Institute, Houston, Texas, USA
| | | | - Bin Yuan
- Immunobiology and Transplant Research Center, Houston Methodist Research Institute, Houston, Texas, USA
| | - Zhuo Wang
- Immunobiology and Transplant Research Center, Houston Methodist Research Institute, Houston, Texas, USA
| | - Li Jia
- Immunobiology and Transplant Research Center, Houston Methodist Research Institute, Houston, Texas, USA
| | - Rui Jin
- Immunobiology and Transplant Research Center, Houston Methodist Research Institute, Houston, Texas, USA
| | - Hongbo Lu
- Medimmune, Gaithersburg, Maryland, USA
| | - Xian Chang Li
- Immunobiology and Transplant Research Center, Houston Methodist Research Institute, Houston, Texas, USA
- Department of Surgery, Weill Cornell Medical College of Cornell University, New York, New York, USA
| | - Yong-Jun Liu
- Medimmune, Gaithersburg, Maryland, USA
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Zhiqiang Zhang
- Immunobiology and Transplant Research Center, Houston Methodist Research Institute, Houston, Texas, USA
- Department of Surgery, Weill Cornell Medical College of Cornell University, New York, New York, USA
| |
Collapse
|
47
|
Dükel M, Streitfeld WS, Tang TCC, Backman LRF, Ai L, May WS, Brown KD. The Breast Cancer Tumor Suppressor TRIM29 Is Expressed via ATM-dependent Signaling in Response to Hypoxia. J Biol Chem 2016; 291:21541-21552. [PMID: 27535224 DOI: 10.1074/jbc.m116.730960] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 08/17/2016] [Indexed: 02/01/2023] Open
Abstract
Reduced ATM function has been linked to breast cancer risk, and the TRIM29 protein is an emerging breast cancer tumor suppressor. Here we show that, in cultured breast tumor and non-tumorigenic mammary epithelial cells, TRIM29 is up-regulated in response to hypoxic stress but not DNA damage. Hypoxia-induced up-regulation of TRIM29 is dependent upon ATM and HIF1α and occurs through increased transcription of the TRIM29 gene. Basal expression of TRIM29 is also down-regulated in cells expressing diminished levels of ATM, and findings suggest that this occurs through basal NF-κB activity as knockdown of the NF-κB subunit RelA suppresses TRIM29 abundance. We have previously shown that the activity of the TWIST1 oncogene is antagonized by TRIM29 and now show that TRIM29 is necessary to block the up-regulation of TWIST1 that occurs in response to hypoxic stress. This study establishes TRIM29 as a hypoxia-induced tumor suppressor gene and provides a novel molecular mechanism for ATM-dependent breast cancer suppression.
Collapse
Affiliation(s)
- Muzaffer Dükel
- From the Departments of Biochemistry and Molecular Biology and
| | - W Scott Streitfeld
- Medicine, University of Florida College of Medicine, Gainesville, Florida 32610
| | | | | | - Lingbao Ai
- From the Departments of Biochemistry and Molecular Biology and
| | - W Stratford May
- Medicine, University of Florida College of Medicine, Gainesville, Florida 32610
| | - Kevin D Brown
- From the Departments of Biochemistry and Molecular Biology and
| |
Collapse
|
48
|
TRIMming p53's anticancer activity. Oncogene 2016; 35:5577-5584. [PMID: 26898759 DOI: 10.1038/onc.2016.33] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 01/11/2016] [Accepted: 01/12/2016] [Indexed: 12/11/2022]
Abstract
Several TRIM proteins control abundance and activity of p53. Along this route, TRIM proteins have a serious impact on carcinogenesis and prognosis for cancer patients. In the past years, a significant increase has been made in our understanding of how the TRIM protein family controls p53 activity.
Collapse
|
49
|
Hatakeyama S. Early evidence for the role of TRIM29 in multiple cancer models. Expert Opin Ther Targets 2016; 20:767-70. [DOI: 10.1517/14728222.2016.1148687] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Shigetsugu Hatakeyama
- Department of Biochemistry, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido 060-8638, Japan
| |
Collapse
|
50
|
Palmbos PL, Wang L, Yang H, Wang Y, Leflein J, Ahmet ML, Wilkinson JE, Kumar-Sinha C, Ney GM, Tomlins SA, Daignault S, Kunju LP, Wu XR, Lotan Y, Liebert M, Ljungman ME, Simeone DM. ATDC/TRIM29 Drives Invasive Bladder Cancer Formation through miRNA-Mediated and Epigenetic Mechanisms. Cancer Res 2015; 75:5155-66. [PMID: 26471361 DOI: 10.1158/0008-5472.can-15-0603] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 09/03/2015] [Indexed: 02/01/2023]
Abstract
Bladder cancer is a common and deadly malignancy but its treatment has advanced little due to poor understanding of the factors and pathways that promote disease. ATDC/TRIM29 is a highly expressed gene in several lethal tumor types, including bladder tumors, but its role as a pathogenic driver has not been established. Here we show that overexpression of ATDC in vivo is sufficient to drive both noninvasive and invasive bladder carcinoma development in transgenic mice. ATDC-driven bladder tumors were indistinguishable from human bladder cancers, which displayed similar gene expression signatures. Clinically, ATDC was highly expressed in bladder tumors in a manner associated with invasive growth behaviors. Mechanistically, ATDC exerted its oncogenic effects by suppressing miR-29 and subsequent upregulation of DNMT3A, leading to DNA methylation and silencing of the tumor suppressor PTEN. Taken together, our findings established a role for ATDC as a robust pathogenic driver of bladder cancer development, identified downstream effector pathways, and implicated ATDC as a candidate biomarker and therapeutic target.
Collapse
Affiliation(s)
- Phillip L Palmbos
- Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, Michigan. Translational Oncology Program, University of Michigan Medical Center, Ann Arbor, Michigan
| | - Lidong Wang
- Translational Oncology Program, University of Michigan Medical Center, Ann Arbor, Michigan. Department of Surgery, University of Michigan Medical Center, Ann Arbor, Michigan
| | - Huibin Yang
- Translational Oncology Program, University of Michigan Medical Center, Ann Arbor, Michigan. Department of Surgery, University of Michigan Medical Center, Ann Arbor, Michigan
| | - Yin Wang
- Translational Oncology Program, University of Michigan Medical Center, Ann Arbor, Michigan. Department of Surgery, University of Michigan Medical Center, Ann Arbor, Michigan
| | - Jacob Leflein
- Translational Oncology Program, University of Michigan Medical Center, Ann Arbor, Michigan. Department of Surgery, University of Michigan Medical Center, Ann Arbor, Michigan
| | - McKenzie L Ahmet
- Translational Oncology Program, University of Michigan Medical Center, Ann Arbor, Michigan. Department of Surgery, University of Michigan Medical Center, Ann Arbor, Michigan
| | - John E Wilkinson
- Department of Pathology, University of Michigan Medical Center, Ann Arbor, Michigan. Department of Laboratory Animal Medicine, University of Michigan Medical Center, Ann Arbor, Michigan
| | - Chandan Kumar-Sinha
- Department of Pathology, University of Michigan Medical Center, Ann Arbor, Michigan
| | - Gina M Ney
- Translational Oncology Program, University of Michigan Medical Center, Ann Arbor, Michigan. Department of Pediatrics, University of Michigan Medical Center, Ann Arbor, Michigan
| | - Scott A Tomlins
- Department of Pathology, University of Michigan Medical Center, Ann Arbor, Michigan
| | - Stephanie Daignault
- Department of Biostatistics, University of Michigan Medical Center, Ann Arbor, Michigan
| | - Lakshmi P Kunju
- Department of Pathology, University of Michigan Medical Center, Ann Arbor, Michigan
| | - Xue-Ru Wu
- Departments of Urology and Pathology and Veterans Affairs Medical Center in Manhattan, New York University School of Medicine, New York, New York
| | - Yair Lotan
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Monica Liebert
- Translational Oncology Program, University of Michigan Medical Center, Ann Arbor, Michigan. Department of Urology, University of Michigan Medical Center, Ann Arbor, Michigan
| | - Mats E Ljungman
- Translational Oncology Program, University of Michigan Medical Center, Ann Arbor, Michigan. Department of Radiation Oncology, University of Michigan Medical Center, Ann Arbor, Michigan
| | - Diane M Simeone
- Translational Oncology Program, University of Michigan Medical Center, Ann Arbor, Michigan. Department of Surgery, University of Michigan Medical Center, Ann Arbor, Michigan. Department of Molecular and Integrative Physiology, University of Michigan Medical Center, Ann Arbor, Michigan.
| |
Collapse
|