1
|
Zhang BW, Huang T, Yang YF, Li MY, Shao GB. Lysine methyltransferase SETD7 in cancer: functions, molecular mechanisms and therapeutic implications. Mol Biol Rep 2025; 52:389. [PMID: 40232640 DOI: 10.1007/s11033-025-10494-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 04/07/2025] [Indexed: 04/16/2025]
Abstract
Since its discovery as a histone methyltransferase, SETD7 has been implicated in many signaling pathways and carcinogenesis. SETD7 catalyzes the methylation of histone H3 and non-histone proteins, regulating their translation, stability and activity. SETD7 is frequently abnormally expressed and has a significant influence on cell proliferation, invasion, autophagy and immune response. As cancer is a complex disease, an outstanding concept in cancer biology is the "hallmarks of cancer". In this review, we focus on the involvement of SETD7 in the hallmarks of cancer, describing its functions and underlying mechanisms in detail. Additionally, we discuss non-coding RNAs and chemical inhibitors targeting SETD7, highlighting the potential and importance of SETD7 in cancer therapy.
Collapse
Affiliation(s)
- Bo-Wen Zhang
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Ting Huang
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Yi-Fan Yang
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Ming-Yang Li
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Gen-Bao Shao
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China.
- Present Address: Jiangsu University, No.301 Xuefu Road, Jingkou District, Zhenjiang, Jiangsu province, China.
| |
Collapse
|
2
|
Priyamvada P, Ashok G, Joshi T, Anbarasu S, Anbarasu A, Ramaiah S. Unravelling the molecular mechanistic pathway underlying the anticancer effects of kaempferol in colorectal cancer: a reverse pharmacology network approach. Mol Divers 2025; 29:1049-1067. [PMID: 38795259 DOI: 10.1007/s11030-024-10890-0] [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: 02/05/2024] [Accepted: 04/30/2024] [Indexed: 05/27/2024]
Abstract
Colorectal cancer (CRC) is the third most diagnosed and highly fatal malignancy, presenting serious health concerns worldwide. The search for an effective cure for CRC is challenging and poses a serious concern. Kaempferol is a potent anti-cancerous bioactive compound often suggested for treating various cancers, including CRC. However, its underlying molecular mechanism against CRC remains unclear. The present study delves into kaempferol's molecular pathways and underlying molecular mechanisms against CRC targets. The target protein-coding genes for kaempferol were retrieved, and the CRC-associated genes were curated. Twelve common targets with a disease specificity index of > 0.6 were validated for their protein expression at different stages of CRC. Over-expressed USP1, SETD7, POLH, TDP1 and RACGAP1 were selected for further studies. The binding affinities of kaempferol to the corresponding proteins were evaluated using molecular docking and Molecular Dynamics (MD) simulations. SETD7 exhibited the highest binding affinity with the lowest binding energy (- 8.06 kcal/mol). Additionally, the MD simulation, and MM-PBSA conferred SETD7-kaempferol complex had the least root-mean-square deviation with lower interaction energy and higher conformational stability. The protein-protein interaction of SETD7 constructed revealed direct interactors, namely, DNMT1, FOXO1, FOXO3, FOXO4, H3-3B, H3-4, H3C12, H3C13, SETD7, SIRT1 and TP53, have a potential role in cancer progression through FOXO signalling. In summary, our study revealed kaempferol's multi-target and synergistic effect on multiple CRC targets and its underlying mechanisms. Finally, the study recommends in-vitro and in-vivo trials for validation of anti-cancerous drugs for CRC.
Collapse
Affiliation(s)
- P Priyamvada
- Medical and Biological Computing Laboratory, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Tamil Nadu, Vellore, 632014, India
- Department of Bio-Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Tamil Nadu, Vellore, 632014, India
| | - Gayathri Ashok
- Department of Bio-Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Tamil Nadu, Vellore, 632014, India
| | - Tushar Joshi
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Tamil Nadu, Vellore, 632014, India
| | - Suvitha Anbarasu
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Tamil Nadu, Vellore, 632014, India
| | - Anand Anbarasu
- Medical and Biological Computing Laboratory, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Tamil Nadu, Vellore, 632014, India
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Tamil Nadu, Vellore, 632014, India
| | - Sudha Ramaiah
- Medical and Biological Computing Laboratory, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Tamil Nadu, Vellore, 632014, India.
- Department of Bio-Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Tamil Nadu, Vellore, 632014, India.
| |
Collapse
|
3
|
Zhang Z, Li M, Hou Y, Huang T, Zhang B, Lin Q, Shao G. SETD7 promotes LC3B methylation and degradation in ovarian cancer. J Biol Chem 2025; 301:108134. [PMID: 39725038 PMCID: PMC11791264 DOI: 10.1016/j.jbc.2024.108134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 12/08/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024] Open
Abstract
Microtubule-associated protein 1 light chain 3 (LC3) is a key autophagy-related protein involved in regulating autophagosome formation and autophagy activity. Post-translational modifications of LC3 are necessary to modulate its function. However, LC3 protein methylation and its physiological significance have not yet been elucidated. Here, we show that SET domain containing lysine methyltransferase 7 (SETD7) interacts with LC3B, a common isoform of LC3, and methylates LC3B at lysine 51 (K51). SETD7-mediated methylation of LC3B promotes ubiquitination and degradation of LC3B, resulting in reduced autophagosome formation. Furthermore, SETD7 exerts a tumor-promotive function in ovarian cancer (OC) cells in a K51 methylation-dependent manner. Collectively, our data define a novel modification of LC3B and highlight the oncogenic effect of SETD7 via mediating LC3B methylation and degradation.
Collapse
Affiliation(s)
- Ziwei Zhang
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Mingyang Li
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yanan Hou
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Ting Huang
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Bowen Zhang
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Qiong Lin
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Genbao Shao
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China.
| |
Collapse
|
4
|
Das S, Parigi SM, Luo X, Fransson J, Kern BC, Okhovat A, Diaz OE, Sorini C, Czarnewski P, Webb AT, Morales RA, Lebon S, Monasterio G, Castillo F, Tripathi KP, He N, Pelczar P, Schaltenberg N, De la Fuente M, López-Köstner F, Nylén S, Larsen HL, Kuiper R, Antonson P, Hermoso MA, Huber S, Biton M, Scharaw S, Gustafsson JÅ, Katajisto P, Villablanca EJ. Liver X receptor unlinks intestinal regeneration and tumorigenesis. Nature 2025; 637:1198-1206. [PMID: 39567700 PMCID: PMC11779645 DOI: 10.1038/s41586-024-08247-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 10/17/2024] [Indexed: 11/22/2024]
Abstract
Uncontrolled regeneration leads to neoplastic transformation1-3. The intestinal epithelium requires precise regulation during continuous homeostatic and damage-induced tissue renewal to prevent neoplastic transformation, suggesting that pathways unlinking tumour growth from regenerative processes must exist. Here, by mining RNA-sequencing datasets from two intestinal damage models4,5 and using pharmacological, transcriptomics and genetic tools, we identified liver X receptor (LXR) pathway activation as a tissue adaptation to damage that reciprocally regulates intestinal regeneration and tumorigenesis. Using single-cell RNA sequencing, intestinal organoids, and gain- and loss-of-function experiments, we demonstrate that LXR activation in intestinal epithelial cells induces amphiregulin (Areg), enhancing regenerative responses. This response is coordinated by the LXR-ligand-producing enzyme CYP27A1, which was upregulated in damaged intestinal crypt niches. Deletion of Cyp27a1 impaired intestinal regeneration, which was rescued by exogenous LXR agonists. Notably, in tumour models, Cyp27a1 deficiency led to increased tumour growth, whereas LXR activation elicited anti-tumour responses dependent on adaptive immunity. Consistently, human colorectal cancer specimens exhibited reduced levels of CYP27A1, LXR target genes, and B and CD8 T cell gene signatures. We therefore identify an epithelial adaptation mechanism to damage, whereby LXR functions as a rheostat, promoting tissue repair while limiting tumorigenesis.
Collapse
Affiliation(s)
- Srustidhar Das
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden.
- Center of Molecular Medicine, Stockholm, Sweden.
| | - S Martina Parigi
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
| | - Xinxin Luo
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
| | - Jennifer Fransson
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
| | - Bianca C Kern
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
| | - Ali Okhovat
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
- Structural Genomics Consortium, Division of Rheumatology, Department of Medicine Solna, Karolinska Institute and University Hospital, Stockholm, Sweden
| | - Oscar E Diaz
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
| | - Chiara Sorini
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
| | - Paulo Czarnewski
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
- Science for Life Laboratory, Department of Biochemistry and Biophysics, National Bioinformatics Infrastructure Sweden, Stockholm University, Solna, Sweden
| | - Anna T Webb
- Department of Cell and Molecular Biology, Karolinska Institutet, Solna, Sweden
| | - Rodrigo A Morales
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
| | - Sacha Lebon
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Gustavo Monasterio
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
| | - Francisca Castillo
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
| | - Kumar P Tripathi
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
| | - Ning He
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
| | - Penelope Pelczar
- I. Medizinische Klinik, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Nicola Schaltenberg
- I. Medizinische Klinik, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Marjorie De la Fuente
- Center of Biomedical Research (CIBMED), School of Medicine, Faculty of Medicine-Clinica Las Condes, Universidad Finis Terrae, Santiago, Chile
- Laboratory of Innate Immunity, Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Francisco López-Köstner
- Centro de Enfermedades Digestivas, Programa Enfermedad Inflamatoria Intestinal, Clínica Universidad de Los Andes, Universidad de Los Andes, Santiago, Chile
| | - Susanne Nylén
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Hjalte List Larsen
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), University of Copenhagen, Copenhagen, Denmark
| | - Raoul Kuiper
- Section for Aquatic Biosecurity Research, Norwegian Veterinary Institute, Ås, Norway
- Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Per Antonson
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Marcela A Hermoso
- Laboratory of Innate Immunity, Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Samuel Huber
- I. Medizinische Klinik, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Moshe Biton
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Sandra Scharaw
- Department of Cell and Molecular Biology, Karolinska Institutet, Solna, Sweden
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Jan-Åke Gustafsson
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Pekka Katajisto
- Department of Cell and Molecular Biology, Karolinska Institutet, Solna, Sweden
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Eduardo J Villablanca
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden.
- Center of Molecular Medicine, Stockholm, Sweden.
- Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
5
|
Wu X, Wang S, Pan Y, Li M, Song M, Zhang H, Deng M, Yang X, Xu J, Zhang S, Zhang J, Wang F, Plikus MV, Lv C, Yu L, Yu Z. m 6A Reader PRRC2A Promotes Colorectal Cancer Progression via CK1ε-Mediated Activation of WNT and YAP Signaling Pathways. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2406935. [PMID: 39582289 PMCID: PMC11744581 DOI: 10.1002/advs.202406935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 10/12/2024] [Indexed: 11/26/2024]
Abstract
Colorectal cancer (CRC) is the third most common cancer type and the second highest mortality rate among cancers. However, the mechanisms underlying CRC progression remain to be fully understood. In this work, a recently identified m6A-modified RNA reader protein Proline-rich Coiled-coil 2a (PRRC2A) is markedly upregulated in CRC, and intestinal epithelium-specific deletion of Prrc2a significantly suppressed tumor cell growth, stemness, and migratory capacity, while its overexpression promoted these behaviors. Through multiomics analysis, PRRC2A directly targeted CSNK1E (encoding CK1ε), maintaining its RNA stability in an m6A-dependent manner, and that elevated CK1ε can concomitantly result in activation of the WNT and YAP signaling pathways. Interestingly, PRRC2A is directly regulated by the transcription factor ATF1 in its promoter. In summary, the work reveals a novel mechanism by which m6A reader PRRC2A promotes colorectal cancer progression via CK1ε and aberrant upregulation of WNT and YAP signaling. Therefore, PRRC2A and CK1ε can be potential therapeutic targets for treating CRC.
Collapse
Affiliation(s)
- Xi Wu
- The First Affiliated Hospital of Zhengzhou UniversityTianjian Laboratory of Advanced Biomedical SciencesAcademy of Medical SciencesZhengzhou UniversityZhengzhouHenan450052China
- State Key Laboratory of Animal Biotech BreedingCollege of Biological SciencesChina Agricultural UniversityBeijing100193China
| | - Shiyang Wang
- State Key Laboratory of Animal Biotech BreedingCollege of Biological SciencesChina Agricultural UniversityBeijing100193China
| | - Yuwei Pan
- State Key Laboratory of Animal Biotech BreedingCollege of Biological SciencesChina Agricultural UniversityBeijing100193China
| | - Mengzhen Li
- State Key Laboratory of Animal Biotech BreedingCollege of Biological SciencesChina Agricultural UniversityBeijing100193China
| | - Manyu Song
- Key Laboratory of Precision Nutrition and Food QualityMinistry of EducationDepartment of Nutrition and HealthChina Agricultural UniversityBeijing100193China
| | - Hanfu Zhang
- State Key Laboratory of Animal Biotech BreedingCollege of Biological SciencesChina Agricultural UniversityBeijing100193China
| | - Min Deng
- State Key Laboratory of Animal Biotech BreedingCollege of Biological SciencesChina Agricultural UniversityBeijing100193China
| | - Xu Yang
- State Key Laboratory of Animal Biotech BreedingCollege of Biological SciencesChina Agricultural UniversityBeijing100193China
| | - Jiuzhi Xu
- State Key Laboratory of Animal Biotech BreedingCollege of Biological SciencesChina Agricultural UniversityBeijing100193China
| | - Shuo Zhang
- State Key Laboratory of Animal Biotech BreedingCollege of Biological SciencesChina Agricultural UniversityBeijing100193China
| | - Jinhua Zhang
- The college of Life Science and BioengineeringBeijing Jiaotong UniversityBeijing100044China
| | - Fengchao Wang
- National Institute of Biological ScienceBeijing102206China
| | - Maksim V. Plikus
- Department of Developmental and Cell BiologySue and Bill Gross Stem Cell Research CenterCenter for Complex Biological SystemsUniversity of CaliforniaIrvineCA92697USA
| | - Cong Lv
- Key Laboratory of Precision Nutrition and Food QualityMinistry of EducationDepartment of Nutrition and HealthChina Agricultural UniversityBeijing100193China
| | - Lu Yu
- State Key Laboratory of Animal Biotech BreedingCollege of Biological SciencesChina Agricultural UniversityBeijing100193China
| | - Zhengquan Yu
- The First Affiliated Hospital of Zhengzhou UniversityTianjian Laboratory of Advanced Biomedical SciencesAcademy of Medical SciencesZhengzhou UniversityZhengzhouHenan450052China
- State Key Laboratory of Animal Biotech BreedingCollege of Biological SciencesChina Agricultural UniversityBeijing100193China
| |
Collapse
|
6
|
Ren Z, Su R, Liu D, Wang Q, Liu S, Kong D, Qiu Y. Yes-associated protein indispensably mediates hirsutine-induced inhibition on cell growth and Wnt/β-catenin signaling in colorectal cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156156. [PMID: 39437684 DOI: 10.1016/j.phymed.2024.156156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/26/2024] [Accepted: 10/12/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND AND PURPOSE Targeting Wnt/β-catenin signaling emerges as one of the promising strategies for colorectal cancer (CRC) treatment, as this signaling is highly activated in CRC progression. Despite reports on the cytotoxic effects of hirsutine (HT), an indole alkaloid found in herbal medicines from the genus Uncaria, its therapeutic potential for CRC and the involved mechanisms are poorly understood. This study investigates the anticancer efficacy and the probable mechanisms of HT against CRC. METHODS To evaluate in vitro anticancer activity of HT, cell growth examined by MTT and colony formation assay, and apoptosis examined by flow cytometry were analyzed. To explore the mechanisms, RNA-sequencing, western blotting, dual-luciferase reporter assays, immunofluorescence, and co-immunoprecipitation were performed. Mouse model of azoxymethane/dextran sodium sulfate (AOM/DSS)-induced colon cancer was utilized to assess HT's in vivo anticancer efficacy. RESULTS HT significantly inhibited CRC cell proliferation with IC50 values of 22.25 ± 3.27 μM for SW620 cells and 22.24 ± 2.36 μM for HCT116 cells, and induced apoptosis. HT decreased protein levels of Wnt3a and β-catenin dose- and time-dependently, and inhibited TOP/FOP FLASH reporter activity, nuclear travel of β-catenin, and downstream targets like c-Myc, Cyclin D1, VEGF. HT reduced β-catenin protein half-life, and the reversal of this effect by MG132 indicated that HT facilitated proteasome-dependent degradation of β-catenin in these two cell lines. HT also increased β-catenin ubiquitination without affecting Axin and β-TrCP levels. HT treatment for 24 h induced YAP cytoplasmic retention, enhanced YAP interacting with β-catenin and β-TrCP, triggering destruction complex formation and β-catenin ubiquitination and degradation, while YAP siRNA impaired these effects. Additionally, β-catenin overexpression and LiCl treatment counteracted HT-induced inhibition on cell growth and Wnt/β-catenin cascade. In model of AOM/DSS-induced mouse colon cancer, compared with AOM/DSS treatment group, HT recovered colon length, reduced tumor numbers and radius, and downregulated β-catenin and Ki-67, while upregulated cleaved PARP in the colorectal tissue with tumors. CONCLUSION HT exhibits anticancer activity against CRC probably by inhibiting Wnt/β-catenin signaling, with YAP playing an indispensible role during the process, highlighting HT as a potential novel candidate drug for CRC therapy.
Collapse
Affiliation(s)
- Zehao Ren
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Ruixin Su
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Donghui Liu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Qian Wang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Shanshan Liu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Dexin Kong
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China.
| | - Yuling Qiu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
7
|
Gadewal N, Patidar D, Natu A, Gupta S, Bastikar V. In silico screening of phytochemicals against chromatin modifier, SETD7 for remodeling of the immunosuppressive tumor microenvironment in renal cancer. Mol Divers 2024:10.1007/s11030-024-11038-w. [PMID: 39602041 DOI: 10.1007/s11030-024-11038-w] [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: 06/04/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024]
Abstract
The tumor microenvironment and immune evasion function in a complex cellular network profoundly challenge the clinical outcome of promising therapies. Our recently published study reported that the subset of genes upregulated in ccRCC due to H3K4me1 and DNA demethylation potentially leads to an immunosuppressive environment. Thus, modulating H3K4me1 chromatin modifier SETD7 with a natural inhibitor in combination with immunotherapy might improve the immune landscape for a better therapeutic outcome. The present study was conducted via virtual screening and MD simulation using compounds from the literature, IMPPAT, and SuperNatural database. The phytochemical IMPHY002979 showed better binding affinity and lower energy than the reported R-PFI-2 and cyproheptadine inhibitors. The phytochemicals interact with the SET domain through H-bonding, as confirmed by MD simulation and molecular interaction analysis. Further, the compound was assessed using ADME parameters and free energy estimation, showing better pharmacokinetic properties. Therefore, the non-accessibility of the histone methyltransferase activity domain of SET7 with IMPHY002979 can downregulate H3K4me1 and, thereby, the expression of genes potentially responsible for immunosuppressive TME. Thus, patient stratification based on molecular markers for immunotherapy and combining epigenetic modulators with therapeutic drugs will improve the efficacy of immunotherapy in ccRCC.
Collapse
Affiliation(s)
- Nikhil Gadewal
- Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Cancer Research Institute, Kharghar, Navi Mumbai, MH, 410210, India
- Center for Computational Biology & Translational Research, Amity Institute of Biotechnology, Amity University, Mumbai, MH, India
| | - Diya Patidar
- Center for Computational Biology & Translational Research, Amity Institute of Biotechnology, Amity University, Mumbai, MH, India
| | - Abhiram Natu
- Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Cancer Research Institute, Kharghar, Navi Mumbai, MH, 410210, India
- Training School Complex, Homi Bhabha National Institute, Anushakti Nagar, Mumbai, MH, 400094, India
| | - Sanjay Gupta
- Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Cancer Research Institute, Kharghar, Navi Mumbai, MH, 410210, India.
- Training School Complex, Homi Bhabha National Institute, Anushakti Nagar, Mumbai, MH, 400094, India.
| | - Virupaksha Bastikar
- Center for Computational Biology & Translational Research, Amity Institute of Biotechnology, Amity University, Mumbai, MH, India.
| |
Collapse
|
8
|
Ramalho S, Dopler A, Faller W. Ribosome specialization in cancer: a spotlight on ribosomal proteins. NAR Cancer 2024; 6:zcae029. [PMID: 38989007 PMCID: PMC11231584 DOI: 10.1093/narcan/zcae029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 06/11/2024] [Accepted: 06/19/2024] [Indexed: 07/12/2024] Open
Abstract
In the past few decades, our view of ribosomes has changed substantially. Rather than passive machines without significant variability, it is now acknowledged that they are heterogeneous, and have direct regulatory capacity. This 'ribosome heterogeneity' comes in many flavors, including in both the RNA and protein components of ribosomes, so there are many paths through which ribosome specialization could arise. It is easy to imagine that specialized ribosomes could have wide physiological roles, through the translation of specific mRNA populations, and there is now evidence for this in several contexts. Translation is highly dysregulated in cancer, needed to support oncogenic phenotypes and to overcome cellular stress. However, the role of ribosome specialization in this is not clear. In this review we focus on specialized ribosomes in cancer. Specifically, we assess the impact that post-translational modifications and differential ribosome incorporation of ribosomal proteins (RPs) have in this disease. We focus on studies that have shown a ribosome-mediated change in translation of specific mRNA populations, and hypothesize how such a process could be driving other phenotypes. We review the impact of RP-mediated heterogeneity in both intrinsic and extrinsic oncogenic processes, and consider how this knowledge could be leveraged to benefit patients.
Collapse
Affiliation(s)
- Sofia Ramalho
- Division of Oncogenomics, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Anna Dopler
- Division of Oncogenomics, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - William James Faller
- Division of Oncogenomics, The Netherlands Cancer Institute, Amsterdam, Netherlands
| |
Collapse
|
9
|
Daks A, Parfenyev S, Shuvalov O, Fedorova O, Nazarov A, Melino G, Barlev NA. Lysine-specific methyltransferase Set7/9 in stemness, differentiation, and development. Biol Direct 2024; 19:41. [PMID: 38812048 PMCID: PMC11137904 DOI: 10.1186/s13062-024-00484-z] [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: 04/17/2024] [Accepted: 05/21/2024] [Indexed: 05/31/2024] Open
Abstract
The enzymes performing protein post-translational modifications (PTMs) form a critical post-translational regulatory circuitry that orchestrates literally all cellular processes in the organism. In particular, the balance between cellular stemness and differentiation is crucial for the development of multicellular organisms. Importantly, the fine-tuning of this balance on the genetic level is largely mediated by specific PTMs of histones including lysine methylation. Lysine methylation is carried out by special enzymes (lysine methyltransferases) that transfer the methyl group from S-adenosyl-L-methionine to the lysine residues of protein substrates. Set7/9 is one of the exemplary protein methyltransferases that however, has not been fully studied yet. It was originally discovered as histone H3 lysine 4-specific methyltransferase, which later was shown to methylate a number of non-histone proteins that are crucial regulators of stemness and differentiation, including p53, pRb, YAP, DNMT1, SOX2, FOXO3, and others. In this review we summarize the information available to date on the role of Set7/9 in cellular differentiation and tissue development during embryogenesis and in adult organisms. Finally, we highlight and discuss the role of Set7/9 in pathological processes associated with aberrant cellular differentiation and self-renewal, including the formation of cancer stem cells.
Collapse
Affiliation(s)
- Alexandra Daks
- Institute of Cytology, Russian Academy of Sciences, St Petersburg, Russian Federation, 194064.
| | - Sergey Parfenyev
- Institute of Cytology, Russian Academy of Sciences, St Petersburg, Russian Federation, 194064
| | - Oleg Shuvalov
- Institute of Cytology, Russian Academy of Sciences, St Petersburg, Russian Federation, 194064
| | - Olga Fedorova
- Institute of Cytology, Russian Academy of Sciences, St Petersburg, Russian Federation, 194064
| | - Alexander Nazarov
- Institute of Cytology, Russian Academy of Sciences, St Petersburg, Russian Federation, 194064
| | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Nickolai A Barlev
- Institute of Cytology, Russian Academy of Sciences, St Petersburg, Russian Federation, 194064.
- Department of Biomedical Sciences, School of Medicine, Nazarbayev University, 001000, Astana, Kazakhstan.
| |
Collapse
|
10
|
Wang J, Tan S, Zhang Y, Xu J, Li Y, Cheng Q, Ding C, Liu X, Chang J. Set7/9 aggravates ischemic brain injury via enhancing glutamine metabolism in a blocking Sirt5 manner. Cell Death Differ 2024; 31:511-523. [PMID: 38365969 PMCID: PMC11043079 DOI: 10.1038/s41418-024-01264-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 01/30/2024] [Accepted: 01/30/2024] [Indexed: 02/18/2024] Open
Abstract
The aberrant expression of methyltransferase Set7/9 plays a role in various diseases. However, the contribution of Set7/9 in ischemic stroke remains unclear. Here, we show ischemic injury results in a rapid elevation of Set7/9, which is accompanied by the downregulation of Sirt5, a deacetylase reported to protect against injury. Proteomic analysis identifies the decrease of chromobox homolog 1 (Cbx1) in knockdown Set7/9 neurons. Mechanistically, Set7/9 promotes the binding of Cbx1 to H3K9me2/3 and forms a transcription repressor complex at the Sirt5 promoter, ultimately repressing Sirt5 transcription. Thus, the deacetylation of Sirt5 substrate, glutaminase, which catalyzes the hydrolysis of glutamine to glutamate and ammonia, is decreased, promoting glutaminase expression and triggering excitotoxicity. Blocking Set7/9 eliminates H3K9me2/3 from the Sirt5 promoter and normalizes Sirt5 expression and Set7/9 knockout efficiently ameliorates brain ischemic injury by reducing the accumulation of ammonia and glutamate in a Sirt5-dependent manner. Collectively, the Set7/9-Sirt5 axis may be a promising epigenetic therapeutic target.
Collapse
Affiliation(s)
- Jinghuan Wang
- Shanghai Key Labortary of Vascular Lesions Regulation and Remodeling, Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, Shanghai, 201203, China
| | - Subei Tan
- Shanghai Key Labortary of Vascular Lesions Regulation and Remodeling, Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, Shanghai, 201203, China
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, Zhongshan Hospital, Fudan University, Shanghai, 201203, China
| | - Yuyu Zhang
- Shanghai Key Labortary of Vascular Lesions Regulation and Remodeling, Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, Shanghai, 201203, China
| | - Jie Xu
- Shanghai Key Labortary of Vascular Lesions Regulation and Remodeling, Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, Shanghai, 201203, China
| | - Yuhui Li
- Shanghai Key Labortary of Vascular Lesions Regulation and Remodeling, Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, Shanghai, 201203, China
| | - Qianwen Cheng
- Shanghai Key Labortary of Vascular Lesions Regulation and Remodeling, Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, Shanghai, 201203, China
| | - Chen Ding
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, Zhongshan Hospital, Fudan University, Shanghai, 201203, China.
| | - Xinhua Liu
- Shanghai Key Labortary of Vascular Lesions Regulation and Remodeling, Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, Shanghai, 201203, China.
| | - Jun Chang
- Shanghai Key Labortary of Vascular Lesions Regulation and Remodeling, Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, Shanghai, 201203, China.
| |
Collapse
|
11
|
Alasaadi DN, Alvizi L, Hartmann J, Stillman N, Moghe P, Hiiragi T, Mayor R. Competence for neural crest induction is controlled by hydrostatic pressure through Yap. Nat Cell Biol 2024; 26:530-541. [PMID: 38499770 PMCID: PMC11021196 DOI: 10.1038/s41556-024-01378-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 02/15/2024] [Indexed: 03/20/2024]
Abstract
Embryonic induction is a key mechanism in development that corresponds to an interaction between a signalling and a responding tissue, causing a change in the direction of differentiation by the responding tissue. Considerable progress has been achieved in identifying inductive signals, yet how tissues control their responsiveness to these signals, known as competence, remains poorly understood. While the role of molecular signals in competence has been studied, how tissue mechanics influence competence remains unexplored. Here we investigate the role of hydrostatic pressure in controlling competence in neural crest cells, an embryonic cell population. We show that neural crest competence decreases concomitantly with an increase in the hydrostatic pressure of the blastocoel, an embryonic cavity in contact with the prospective neural crest. By manipulating hydrostatic pressure in vivo, we show that this increase leads to the inhibition of Yap signalling and impairs Wnt activation in the responding tissue, which would be required for neural crest induction. We further show that hydrostatic pressure controls neural crest induction in amphibian and mouse embryos and in human cells, suggesting a conserved mechanism across vertebrates. Our work sets out how tissue mechanics can interplay with signalling pathways to regulate embryonic competence.
Collapse
Affiliation(s)
- Delan N Alasaadi
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Lucas Alvizi
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Jonas Hartmann
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Namid Stillman
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Prachiti Moghe
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands
- Collaboration for joint PhD degree between the European Molecular Biology Laboratory (EMBL) and Heidelberg University, Faculty of Biosciences, Heidelberg, Germany
| | - Takashi Hiiragi
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands
- Institute for the Advanced Study of Human Biology, Kyoto University, Kyoto, Japan
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, London, UK.
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile.
| |
Collapse
|
12
|
Aziz N, Hong YH, Kim HG, Kim JH, Cho JY. Tumor-suppressive functions of protein lysine methyltransferases. Exp Mol Med 2023; 55:2475-2497. [PMID: 38036730 PMCID: PMC10766653 DOI: 10.1038/s12276-023-01117-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 08/30/2023] [Accepted: 09/05/2023] [Indexed: 12/02/2023] Open
Abstract
Protein lysine methyltransferases (PKMTs) play crucial roles in histone and nonhistone modifications, and their dysregulation has been linked to the development and progression of cancer. While the majority of studies have focused on the oncogenic functions of PKMTs, extensive evidence has indicated that these enzymes also play roles in tumor suppression by regulating the stability of p53 and β-catenin, promoting α-tubulin-mediated genomic stability, and regulating the transcription of oncogenes and tumor suppressors. Despite their contradictory roles in tumorigenesis, many PKMTs have been identified as potential therapeutic targets for cancer treatment. However, PKMT inhibitors may have unintended negative effects depending on the specific cancer type and target enzyme. Therefore, this review aims to comprehensively summarize the tumor-suppressive effects of PKMTs and to provide new insights into the development of anticancer drugs targeting PKMTs.
Collapse
Affiliation(s)
- Nur Aziz
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Yo Han Hong
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Han Gyung Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Ji Hye Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
13
|
Porzberg MRB, Lenstra DC, Damen E, Blaauw RH, Rutjes FPJT, Wegert A, Mecinović J. (R)-PFI-2 Analogues as Substrates and Inhibitors of Histone Lysine Methyltransferase SETD7. ChemMedChem 2023; 18:e202300457. [PMID: 37872124 DOI: 10.1002/cmdc.202300457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/22/2023] [Accepted: 10/23/2023] [Indexed: 10/25/2023]
Abstract
(R)-PFI-2 is a histone substrate-competitive inhibitor of the human histone lysine monomethyltransferase SETD7. Aimed at developing potent inhibitors of SETD7 that can also act as small molecule substrates, we replaced the pyrrolidine ring of (R)-PFI-2 with several side chains bearing nucleophilic functional groups. We explored the inhibitory activity of 20 novel (R)-PFI-2 analogues, and found that the most potent analogue has a hydroxyethyl side chain (7). SETD7's ability to catalyse methylation of (R)-PFI-2-based small molecules was evaluated by mass spectrometric assays, and we observed efficient methylation of analogues bearing lysine mimicking nucleophilic amines. The optimal side chain was found to be an aminoethyl group (1), which was surprisingly also dimethylated by SETD7. The work demonstrates that small molecules can act as both substrates and inhibitors of biomedically important SETD7.
Collapse
Affiliation(s)
- Miriam R B Porzberg
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230, Odense, Denmark
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ, Nijmegen (The, Netherlands
| | - Danny C Lenstra
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ, Nijmegen (The, Netherlands
| | - Eddy Damen
- Symeres Netherlands B.V., Kerkenbos 1013, 6546 BB, Nijmegen (The, Netherlands
| | - Richard H Blaauw
- Symeres Netherlands B.V., Kerkenbos 1013, 6546 BB, Nijmegen (The, Netherlands
| | - Floris P J T Rutjes
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ, Nijmegen (The, Netherlands
| | - Anita Wegert
- Symeres Netherlands B.V., Kerkenbos 1013, 6546 BB, Nijmegen (The, Netherlands
| | - Jasmin Mecinović
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230, Odense, Denmark
| |
Collapse
|
14
|
Monteiro FL, Góis A, Direito I, Melo T, Neves B, Alves MI, Batista I, Domingues MDR, Helguero LA. Inhibiting SETD7 methyl-transferase activity impairs differentiation, lipid metabolism and lactogenesis in mammary epithelial cells. FEBS Lett 2023; 597:2656-2671. [PMID: 37723127 DOI: 10.1002/1873-3468.14737] [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: 02/08/2023] [Revised: 06/18/2023] [Accepted: 07/21/2023] [Indexed: 09/20/2023]
Abstract
SETD7 (SET7/9, KMT7) is a lysine methyltransferase that targets master regulators of cell proliferation and differentiation. Here, the impact of inhibiting SETD7 catalytic activity on mammary epithelial cell differentiation was studied by focusing on genes associated with epithelial differentiation, lactogenesis, and lipid metabolism in HC11 and EpH4 cell lines. Setd7 mRNA and protein levels were induced upon lactogenic differentiation in both cell lines. Inhibition of SETD7 activity by the compound (R)-PFI-2 increased cell proliferation and downregulated E-cadherin, beta-catenin, lactoferrin, insulin-like growth factor binding protein 5, and beta-casein levels. In addition, inhibition of SETD7 activity affected the lipid profile and altered the mRNA expression of the phospholipid biosynthesis-related genes choline phosphotransferase 1, and ethanolamine-phosphate cytidylyltransferase. Altogether, the results suggest that inhibiting SETD7 catalytic activity impairs mammary epithelial and lactogenic differentiation.
Collapse
Affiliation(s)
- Fátima Liliana Monteiro
- Institute of Biomedicine (IBIMED), Department of Medical Sciences (DCM), Universidade de Aveiro, Portugal
| | - André Góis
- Institute of Biomedicine (IBIMED), Department of Medical Sciences (DCM), Universidade de Aveiro, Portugal
| | - Inês Direito
- Institute of Biomedicine (IBIMED), Department of Medical Sciences (DCM), Universidade de Aveiro, Portugal
| | - Tânia Melo
- Department of Chemistry, Mass Spectrometry Centre & LAQV-REQUIMTE, University of Aveiro, Portugal
| | - Bruna Neves
- Department of Chemistry, Mass Spectrometry Centre & LAQV-REQUIMTE, University of Aveiro, Portugal
- Department of Chemistry, CESAM-Centre for Environmental and Marine Studies, University of Aveiro, Portugal
| | - Mariana I Alves
- Institute of Biomedicine (IBIMED), Department of Medical Sciences (DCM), Universidade de Aveiro, Portugal
| | - Inês Batista
- Institute of Biomedicine (IBIMED), Department of Medical Sciences (DCM), Universidade de Aveiro, Portugal
| | | | - Luisa A Helguero
- Institute of Biomedicine (IBIMED), Department of Medical Sciences (DCM), Universidade de Aveiro, Portugal
| |
Collapse
|
15
|
Chiang CY, Fan S, Zheng H, Guo W, Zheng Z, Sun Y, Zhong C, Zeng J, Li S, Zhang M, Xiao T, Zheng D. Methylation of KRAS by SETD7 promotes KRAS degradation in non-small cell lung cancer. Cell Rep 2023; 42:113003. [PMID: 37682707 DOI: 10.1016/j.celrep.2023.113003] [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: 07/19/2021] [Revised: 06/18/2023] [Accepted: 08/02/2023] [Indexed: 09/10/2023] Open
Abstract
Oncogenic KRAS mutations are a key driver for initiation and progression in non-small cell lung cancer (NSCLC). However, how post-translational modifications (PTMs) of KRAS, especially methylation, modify KRAS activity remain largely unclear. Here, we show that SET domain containing histone lysine methyltransferase 7 (SETD7) interacts with KRAS and methylates KRAS at lysines 182 and 184. SETD7-mediated methylation of KRAS leads to degradation of KRAS and attenuation of the RAS/MEK/ERK signaling cascade, endowing SETD7 with a potent tumor-suppressive role in NSCLC, both in vitro and in vivo. Mechanistically, RABGEF1, a ubiquitin E3 ligase of KRAS, is recruited and promotes KRAS degradation in a K182/K184 methylation-dependent manner. Notably, SETD7 is inversely correlated with KRAS at the protein level in clinical NSCLC tissues. Low SETD7 or RABGEF1 expression is associated with poor prognosis in lung adenocarcinoma patients. Altogether, our results define a tumor-suppressive function of SETD7 that operates via modulating KRAS methylation and degradation.
Collapse
Affiliation(s)
- Cheng-Yao Chiang
- Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, International Cancer Center, Department of Cell Biology and Genetics, Shenzhen University Medical School, Thoracic Surgery Department of the First Affiliated Hospital, Shenzhen University, Shenzhen 518055, China
| | - Songqing Fan
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Hongmei Zheng
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Wenjun Guo
- Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, International Cancer Center, Department of Cell Biology and Genetics, Shenzhen University Medical School, Thoracic Surgery Department of the First Affiliated Hospital, Shenzhen University, Shenzhen 518055, China
| | - Zehan Zheng
- Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, International Cancer Center, Department of Cell Biology and Genetics, Shenzhen University Medical School, Thoracic Surgery Department of the First Affiliated Hospital, Shenzhen University, Shenzhen 518055, China
| | - Yihua Sun
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Chuanqi Zhong
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Juan Zeng
- School of Biomedical Engineering, Guangdong Medical University, Dongguan, Guangdong 523808, China
| | - Shuaihu Li
- Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, International Cancer Center, Department of Cell Biology and Genetics, Shenzhen University Medical School, Thoracic Surgery Department of the First Affiliated Hospital, Shenzhen University, Shenzhen 518055, China
| | - Min Zhang
- Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, International Cancer Center, Department of Cell Biology and Genetics, Shenzhen University Medical School, Thoracic Surgery Department of the First Affiliated Hospital, Shenzhen University, Shenzhen 518055, China
| | - Tian Xiao
- Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, International Cancer Center, Department of Cell Biology and Genetics, Shenzhen University Medical School, Thoracic Surgery Department of the First Affiliated Hospital, Shenzhen University, Shenzhen 518055, China.
| | - Duo Zheng
- Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, International Cancer Center, Department of Cell Biology and Genetics, Shenzhen University Medical School, Thoracic Surgery Department of the First Affiliated Hospital, Shenzhen University, Shenzhen 518055, China.
| |
Collapse
|
16
|
Kovács SA, Fekete JT, Győrffy B. Predictive biomarkers of immunotherapy response with pharmacological applications in solid tumors. Acta Pharmacol Sin 2023; 44:1879-1889. [PMID: 37055532 PMCID: PMC10462766 DOI: 10.1038/s41401-023-01079-6] [Citation(s) in RCA: 76] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 03/14/2023] [Indexed: 04/15/2023]
Abstract
Immune-checkpoint inhibitors show promising effects in the treatment of multiple tumor types. Biomarkers are biological indicators used to select patients for a systemic anticancer treatment, but there are only a few clinically useful biomarkers such as PD-L1 expression and tumor mutational burden, which can be used to predict immunotherapy response. In this study, we established a database consisting of both gene expression and clinical data to identify biomarkers of response to anti-PD-1, anti-PD-L1, and anti-CTLA-4 immunotherapies. A GEO screening was executed to identify datasets with simultaneously available clinical response and transcriptomic data regardless of cancer type. The screening was restricted to the studies involving administration of anti-PD-1 (nivolumab, pembrolizumab), anti-PD-L1 (atezolizumab, durvalumab) or anti-CTLA-4 (ipilimumab) agents. Receiver operating characteristic (ROC) analysis and Mann-Whitney test were executed across all genes to identify features related to therapy response. The database consisted of 1434 tumor tissue samples from 19 datasets with esophageal, gastric, head and neck, lung, and urothelial cancers, plus melanoma. The strongest druggable gene candidates linked to anti-PD-1 resistance were SPIN1 (AUC = 0.682, P = 9.1E-12), SRC (AUC = 0.667, P = 5.9E-10), SETD7 (AUC = 0.663, P = 1.0E-09), FGFR3 (AUC = 0.657, P = 3.7E-09), YAP1 (AUC = 0.655, P = 6.0E-09), TEAD3 (AUC = 0.649, P = 4.1E-08) and BCL2 (AUC = 0.634, P = 9.7E-08). In the anti-CTLA-4 treatment cohort, BLCAP (AUC = 0.735, P = 2.1E-06) was the most promising gene candidate. No therapeutically relevant target was found to be predictive in the anti-PD-L1 cohort. In the anti-PD-1 group, we were able to confirm the significant correlation with survival for the mismatch-repair genes MLH1 and MSH6. A web platform for further analysis and validation of new biomarker candidates was set up and available at https://www.rocplot.com/immune . In summary, a database and a web platform were established to investigate biomarkers of immunotherapy response in a large cohort of solid tumor samples. Our results could help to identify new patient cohorts eligible for immunotherapy.
Collapse
Affiliation(s)
- Szonja Anna Kovács
- Department of Bioinformatics, Semmelweis University, Tűzoltó utca 7-9, 1094, Budapest, Hungary
- Doctoral School of Pathological Sciences, Semmelweis University, Üllői út 26, 1085, Budapest, Hungary
- National Laboratory for Drug Research and Development, Magyar tudósok körútja 2 1117, Budapest, Hungary
| | - János Tibor Fekete
- National Laboratory for Drug Research and Development, Magyar tudósok körútja 2 1117, Budapest, Hungary
- Research Centre for Natural Sciences, Oncology Biomarker Research Group, Institute of Enzymology, Eötvös Loránd Research Network, Magyar Tudósok körútja 2, 1117, Budapest, Hungary
| | - Balázs Győrffy
- Department of Bioinformatics, Semmelweis University, Tűzoltó utca 7-9, 1094, Budapest, Hungary.
- Department of Pediatrics, Semmelweis University, Tűzoltó utca 7-9, 1094, Budapest, Hungary.
| |
Collapse
|
17
|
Wang D, Li Y, Xu C, Wang H, Huang X, Jin X, Ren S, Gao J, Tong J, Liu J, Zhou J, Shi L. SETD7 promotes lateral plate mesoderm formation by modulating the Wnt/β-catenin signaling pathway. iScience 2023; 26:106917. [PMID: 37378343 PMCID: PMC10291335 DOI: 10.1016/j.isci.2023.106917] [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: 07/24/2022] [Revised: 02/16/2023] [Accepted: 05/14/2023] [Indexed: 06/29/2023] Open
Abstract
The role of SET domain containing 7 (SETD7) during human hematopoietic development remains elusive. Here, we found that deletion of SETD7 attenuated the generation of hematopoietic progenitor cells (HPCs) during the induction of hematopoietic differentiation from human embryonic stem cells (hESCs). Further analysis specified that SETD7 was required for lateral plate mesoderm (LPM) specification but dispensable for the generation of endothelial progenitor cells (EPCs) and HPCs. Mechanistically, rather than depending on its histone methyltransferase activity, SETD7 interacted with β-catenin at lysine residue 180 facilitated its degradation. Diminished SETD7 expression led to the accumulation of β-catenin and the consequent activation of the Wnt signaling pathway, which altered LPM patterning and facilitated the production of paraxial mesoderm (PM). Taken together, the findings indicate that SETD7 is related to LPM and PM patterning via posttranslational regulation of the Wnt/β-catenin signaling pathway, providing novel insights into mesoderm specification during hematopoietic differentiation from hESCs.
Collapse
Affiliation(s)
- Ding Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
- CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin 300020, China
| | - Yapu Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
- CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin 300020, China
| | - Changlu Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
- CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin 300020, China
| | - Hongtao Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
- CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin 300020, China
| | - Xin Huang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
- CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin 300020, China
| | - Xu Jin
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
- CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin 300020, China
| | - Sirui Ren
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
- CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin 300020, China
| | - Jie Gao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
- CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin 300020, China
| | - Jingyuan Tong
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
- CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin 300020, China
| | - Jinhua Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
- CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin 300020, China
| | - Jiaxi Zhou
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
- CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin 300020, China
| | - Lihong Shi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
- Tianjin Institutes of Health Science, Tianjin 301600, China
- CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin 300020, China
| |
Collapse
|
18
|
Guan Z, Liang Y, Zhu Z, Yang A, Li S, Guo J, Wang F, Yang H, Zhang N, Wang X, Wang J. Cytosine arabinoside exposure induced cytotoxic effects and neural tube defects in mice and embryo stem cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 262:115141. [PMID: 37320917 DOI: 10.1016/j.ecoenv.2023.115141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 03/16/2023] [Accepted: 06/12/2023] [Indexed: 06/17/2023]
Abstract
Cytosine arabinoside (Ara-C) is one of the most widely used chemotherapeutic agents for hematological malignancies. The residues of Ara-C have been detected in wastewater and river water with increased usage and discharge. As the ability to cross the placenta and the teratogenicity at low ng/L levels, the toxic effects on pregnant women and infants have been concerned. The toxicity of Ara-C exposure on early embryonic neurodevelopment has not been fully elucidated. In this study, pregnant C57BL/6 mice were injected with different doses of Ara-C on Gestation day (GD) 7.5 and assessed on GD11.5 and GD13.5 to explore the neural developmental effects of Ara-C. HE staining, immunofluorescence, western blot, EdU assay, and flow cytometry were utilized to determine the toxic effects of Ara-C in vivo and in vitro. Our results showed that Ara-C (15-22.5 mg/kg body weight) induced the occurrence of neural tube defects (NTDs). The expression of PH3 was markedly reduced in embryos with Ara-C-induced NTDs, compared to the control group (P < 0.05). In contrast, cell apoptosis was markedly increased. Increased expression levels of GFAP and decreased Nestin were observed in the embryonic brain tissues in Ara-C induced NTDs. The level of β-catenin was also decreased on both GD11.5 and GD13.5. These results were confirmed in vitro using mouse Sv129 embryonic stem cells (mESC). Ara-C at a dose comparable to the environment level (0.05 nM) had cytotoxicity. Impaired Wnt/β-catenin signaling pathway is involved in Ara-C exposure induced imbalance between cell proliferation, apoptosis, and differentiation, which might contribute to Ara-C-induced occurrence of NTDs. Our data indicated the environmental concentration of Ara-C had cytotoxicity and that maternal exposure to Ara-C induced NTDs. These results might provide more information to understand the environmental toxic impact of Ara-C on neurodevelopment.
Collapse
Affiliation(s)
- Zhen Guan
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Translational Medicine Laboratory, Capital Institute of Pediatrics, Beijing 100020, China
| | - Yingchao Liang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Translational Medicine Laboratory, Capital Institute of Pediatrics, Beijing 100020, China
| | - Zhiqiang Zhu
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Translational Medicine Laboratory, Capital Institute of Pediatrics, Beijing 100020, China
| | - Aiyun Yang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Translational Medicine Laboratory, Capital Institute of Pediatrics, Beijing 100020, China
| | - Shen Li
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Translational Medicine Laboratory, Capital Institute of Pediatrics, Beijing 100020, China
| | - Jin Guo
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing 100020, China
| | - Fang Wang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing 100020, China
| | - Huimin Yang
- Growth and Development Department, Capital Institute of Pediatrics, Beijing 100020, China
| | - Na Zhang
- Key Laboratory of Environmental and Viral Oncology, College of Life Science and Chemistry, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China.
| | - Xiuwei Wang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Translational Medicine Laboratory, Capital Institute of Pediatrics, Beijing 100020, China.
| | - Jianhua Wang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Translational Medicine Laboratory, Capital Institute of Pediatrics, Beijing 100020, China.
| |
Collapse
|
19
|
Zhang F, Mao D, He Z, Li W, Zhang X, Li L. SLCO4A1-AS1 regulates laryngeal squamous cell carcinoma cell phenotypes via the Wnt pathway. Oral Dis 2023; 29:390-401. [PMID: 33914996 DOI: 10.1111/odi.13893] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 04/14/2021] [Accepted: 04/19/2021] [Indexed: 02/07/2023]
Abstract
AIM Long non-coding RNAs were widely reported to regulate laryngeal squamous cell carcinoma (LSCC), a prevalent tumor in the head and neck. We aimed to investigate the role of solute carrier organic anion transporter family member 4A1 antisense RNA 1 (SLCO4A1-AS1) in LSCC. MATERIALS & METHODS CCK-8 and colony formation assays were conducted to examine the viability and proliferation of LSCC cells. The apoptosis of LSCC cells was evaluated using flow cytometry and TUNEL assays. The distribution of SLCO4A1-AS1 in LSCC cells was detected by subcellular fractionation assay. The interaction between molecules was confirmed using luciferase reporter assay. RESULTS SLCO4A1-AS1 was overexpressed in LSCC tissues and cells. Furthermore, silenced SLCO4A1-AS1 repressed the proliferation and facilitated apoptosis of LSCC cells. Mechanistical investigation revealed that SLCO4A1-AS1 was a competing endogenous RNA (ceRNA) to upregulate SETD7 by binding with miR-7855-p. Additionally, SLCO4A1-AS1 positively regulated the Wnt/β-catenin signaling pathway by upregulating SETD7. Rescue experiments demonstrated that SLCO4A1-AS1 promoted LSCC proliferation and inhibited LSCC apoptosis by upregulation of SETD7 and activation of the Wnt/β-catenin pathway. CONCLUSION SLCO4A1-AS1 promotes proliferation and inhibits apoptosis of LSCC cells by upregulation of SETD7 in a miR-7855-5p dependent way to activate the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Otolaryngology, Yongchuan Hospital (T.C.M) of Chongqing Medical University, Chongqing, China
| | - Dehong Mao
- Department of Otolaryngology, Yongchuan Hospital (T.C.M) of Chongqing Medical University, Chongqing, China
| | - Zhongmei He
- Department of Otolaryngology, Yongchuan Hospital (T.C.M) of Chongqing Medical University, Chongqing, China
| | - Weichun Li
- Department of Otolaryngology, Yongchuan Hospital (T.C.M) of Chongqing Medical University, Chongqing, China
| | - Xu Zhang
- Department of Otolaryngology, Yongchuan Hospital (T.C.M) of Chongqing Medical University, Chongqing, China
| | - Linglong Li
- Department of Otolaryngology, Yongchuan Hospital (T.C.M) of Chongqing Medical University, Chongqing, China
| |
Collapse
|
20
|
Yang S, Wang X, Bai J, Duan B. The role of SET domain containing lysine methyltransferase 7 in tumorigenesis and development. Cell Cycle 2023; 22:269-275. [PMID: 36101480 PMCID: PMC9851238 DOI: 10.1080/15384101.2022.2122257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 04/22/2022] [Accepted: 09/05/2022] [Indexed: 01/22/2023] Open
Abstract
SET domain containing lysine methyltransferase 7 (SETD7) belongs to the protein lysine methyltransferase family and can catalyze the monomethylation of histone H3K4, which plays a vital role in the regulation of cell cycle, cell differentiation, DNA damage response and chromatin remodeling through K/R-S/T-K (K is lysine residue) sites and the recognition of substrates mediated by SET, i-SET, and n-SET domains and electrostatic action. SETD7 also can regulate the transcription of several genes including β-catenin, Cullin l and lin-28 homolog A (LIN28A), etc. In addition, the abnormal expression of SETD7 can promote the proliferation, migration, invasion of tumor cells, predict the poor prognosis of tumor patients, and may be a potential target for tumor therapy. This paper reviews the structure of SETD7, its role in tumor genesis and development, and the current research progress of relevant targeted drugs to explore its regulatory mechanism in tumor genesis and development and the prospect of targeted therapy.
Collapse
Affiliation(s)
- Shangzhen Yang
- Department of Medical Oncology of Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi, China
- Xi’an Medical University, Xi’an, Shaanxi, China
| | - Xi Wang
- Department of Medical Oncology of Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi, China
| | - Jun Bai
- Department of Medical Oncology of Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi, China
| | - Baojun Duan
- Department of Medical Oncology of Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi, China
- Department of Medical Oncology of Baoji Central Hospital, Baoji Central Hospital, Baoji, Shaanxi, China
| |
Collapse
|
21
|
Ambrosini S, Montecucco F, Kolijn D, Pedicino D, Akhmedov A, Mohammed SA, Herwig M, Gorica E, Szabó PL, Weber L, Russo G, Vinci R, Matter CM, Liuzzo G, Brown PJ, Rossi FMV, Camici GG, Sciarretta S, Beltrami AP, Crea F, Podesser B, Lüscher TF, Kiss A, Ruschitzka F, Hamdani N, Costantino S, Paneni F. Methylation of the Hippo effector YAP by the methyltransferase SETD7 drives myocardial ischaemic injury: a translational study. Cardiovasc Res 2023; 118:3374-3385. [PMID: 35709329 DOI: 10.1093/cvr/cvac102] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 05/12/2022] [Accepted: 06/02/2022] [Indexed: 01/25/2023] Open
Abstract
AIMS Methylation of non-histone proteins is emerging as a central regulatory mechanism in health and disease. The methyltransferase SETD7 has shown to methylate and alter the function of a variety of proteins in vitro; however, its function in the heart is poorly understood. The present study investigates the role of SETD7 in myocardial ischaemic injury. METHODS AND RESULTS Experiments were performed in neonatal rat ventricular myocytes (NRVMs), SETD7 knockout mice (SETD7-/-) undergoing myocardial ischaemia/reperfusion (I/R) injury, left ventricular (LV) myocardial samples from patients with ischaemic cardiomyopathy (ICM), and peripheral blood mononuclear cells (PBMCs) from patients with ST-elevation MI (STEMI). We show that SETD7 is activated upon energy deprivation in cultured NRVMs and methylates the Hippo pathway effector YAP, leading to its cytosolic retention and impaired transcription of antioxidant genes manganese superoxide dismutase (MnSOD) and catalase (CAT). Such impairment of antioxidant defence was associated with mitochondrial reactive oxygen species (mtROS), organelle swelling, and apoptosis. Selective pharmacological inhibition of SETD7 by (R)-PFI-2 restored YAP nuclear localization, thus preventing mtROS, mitochondrial damage, and apoptosis in NRVMs. In mice, genetic deletion of SETD7 attenuated myocardial I/R injury, mtROS, and LV dysfunction by restoring YAP-dependent transcription of MnSOD and CAT. Moreover, in cardiomyocytes isolated from I/R mice and ICM patients, (R)-PFI-2 prevented mtROS accumulation, while improving Ca2+-activated tension. Finally, SETD7 was up-regulated in PBMCs from STEMI patients and negatively correlated with MnSOD and CAT. CONCLUSION We show a methylation-dependent checkpoint regulating oxidative stress during myocardial ischaemia. SETD7 inhibition may represent a valid therapeutic strategy in this setting.
Collapse
Affiliation(s)
- Samuele Ambrosini
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, viale Benedetto XV, 16132, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genova-Italian Cardiovascular Network, Largo Rosanna Benzi, 10, 16132 Genova, Italy
| | - Detmar Kolijn
- Institute of Physiology, Ruhr University, Universitätsstraße 150, 44801 Bochum, Germany
- Molecular and Experimental Cardiology, Ruhr University, Universitätsstraße 150, 44801 Bochum, Germany
- Department of Cardiology, St-Josef Hospital, Ruhr University, Gudrunstraße 56, 44791 Bochum, Germany
| | - Daniela Pedicino
- Dipartimento di Scienze Cardiovascolari e Toraciche, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Via Giuseppe Moscati, 31, 00168 Rome, Italy
| | - Alexander Akhmedov
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Shafeeq A Mohammed
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Melissa Herwig
- Institute of Physiology, Ruhr University, Universitätsstraße 150, 44801 Bochum, Germany
- Molecular and Experimental Cardiology, Ruhr University, Universitätsstraße 150, 44801 Bochum, Germany
- Department of Cardiology, St-Josef Hospital, Ruhr University, Gudrunstraße 56, 44791 Bochum, Germany
| | - Era Gorica
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, 8952 Schlieren, Switzerland
- Department of Pharmacy, University of Pisa, via Bonanno, 6, I-56126 Pisa, Italy
| | - Petra L Szabó
- Ludwig-Boltzmann-Institute for Cardiovascular Research at the Center for Biomedical Research, Medical University of Vienna, Währinger Gürtel 18-20A-1090 Wien, Austria
| | - Lukas Weber
- Ludwig-Boltzmann-Institute for Cardiovascular Research at the Center for Biomedical Research, Medical University of Vienna, Währinger Gürtel 18-20A-1090 Wien, Austria
| | - Giulio Russo
- Dipartimento di Scienze Cardiovascolari e Toraciche, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Via Giuseppe Moscati, 31, 00168 Rome, Italy
| | - Ramona Vinci
- Dipartimento di Scienze Cardiovascolari e Toraciche, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Via Giuseppe Moscati, 31, 00168 Rome, Italy
| | - Christian M Matter
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, 8952 Schlieren, Switzerland
- University Heart Center, Cardiology, University Hospital Zurich, Rämistrasse 100, 8091 Zürich, Switzerland
| | - Giovanna Liuzzo
- Dipartimento di Scienze Cardiovascolari e Toraciche, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Via Giuseppe Moscati, 31, 00168 Rome, Italy
| | - Peter J Brown
- Structural Genomics Consortium, Univerity of Toronto, MaRS South Tower, Suite 700101 College Street, Toronto, ON M5G 1L7, Canada
| | - Fabio M V Rossi
- Biomedical Research Centre, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, 8952 Schlieren, Switzerland
- University Heart Center, Cardiology, University Hospital Zurich, Rämistrasse 100, 8091 Zürich, Switzerland
- Department of Research and Education, University Hospital Zurich, Rämistrasse 100, 8091 Zürich, Switzerland
| | - Sebastiano Sciarretta
- Dipartimento di Scienze e Biotecnologie Medico-Chirurgiche, Sapienza Università di Roma, C.so della Repubblica, 79, 04100 Latina LT, Italy
- Department of AngioCardioNeurology, IRCCS Neuromed, Via Atinense, 18, 86077 Pozzilli IS, Italy
| | - Antonio P Beltrami
- University of Udine, Piazzale Massimiliano Kolbe, 4, 33100 Udine, Italy
- Institute of Clinical Pathology, Academic Hospital "Santa Maria della Misericordia", ASUFC, 33100 Udine, Italy
| | - Filippo Crea
- Dipartimento di Scienze Cardiovascolari e Toraciche, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Via Giuseppe Moscati, 31, 00168 Rome, Italy
| | - Bruno Podesser
- Ludwig-Boltzmann-Institute for Cardiovascular Research at the Center for Biomedical Research, Medical University of Vienna, Währinger Gürtel 18-20A-1090 Wien, Austria
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, 8952 Schlieren, Switzerland
- Royal Brompton & Harefield Hospitals, Imperial College and King's College, Sydney Street, London SW3 6NP, UK
| | - Attila Kiss
- Ludwig-Boltzmann-Institute for Cardiovascular Research at the Center for Biomedical Research, Medical University of Vienna, Währinger Gürtel 18-20A-1090 Wien, Austria
| | - Frank Ruschitzka
- University Heart Center, Cardiology, University Hospital Zurich, Rämistrasse 100, 8091 Zürich, Switzerland
| | - Nazha Hamdani
- Institute of Physiology, Ruhr University, Universitätsstraße 150, 44801 Bochum, Germany
- Molecular and Experimental Cardiology, Ruhr University, Universitätsstraße 150, 44801 Bochum, Germany
- Department of Cardiology, St-Josef Hospital, Ruhr University, Gudrunstraße 56, 44791 Bochum, Germany
| | - Sarah Costantino
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, 8952 Schlieren, Switzerland
- University Heart Center, Cardiology, University Hospital Zurich, Rämistrasse 100, 8091 Zürich, Switzerland
| | - Francesco Paneni
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, 8952 Schlieren, Switzerland
- University Heart Center, Cardiology, University Hospital Zurich, Rämistrasse 100, 8091 Zürich, Switzerland
- Department of Research and Education, University Hospital Zurich, Rämistrasse 100, 8091 Zürich, Switzerland
| |
Collapse
|
22
|
Yan S, Wang P, Wei H, Jia R, Zhen M, Li Q, Xue C, Li J. Treatment of ulcerative colitis with Wu-Mei-Wan by inhibiting intestinal inflammatory response and repairing damaged intestinal mucosa. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 105:154362. [PMID: 35947900 DOI: 10.1016/j.phymed.2022.154362] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 06/17/2022] [Accepted: 07/26/2022] [Indexed: 05/23/2023]
Abstract
BACKGROUND Wu-Mei-Wan (WMW), a traditional Chinese medicine, has been applied in the treatment of gastrointestinal diseases with long-term diarrhea and mucopurulent bloody stool as the main symptoms since ancient times. Studies have shown that WMW inhibits intestinal inflammation, repairs damaged intestinal mucosa, resists colon necrosis, and resists intestinal fibrosis. However, the specific mechanism of action is not yet clear. OBJECTIVE Ulcerative colitis (UC), an intestinal disease with intestinal inflammation and injury as the main pathological manifestations, is one of the high-risk factors for colon cancer. Inhibiting the inflammatory response and promoting colonic epithelial repair are critical to the treatment of UC. However, there is still a lack of remedies with satisfactory curative effects. In this study, the role of WMW in dextran sulfate sodium (DSS)-induced colitis in mice and its related mechanisms are discussed from two aspects: intestinal inflammation and tissue repair. METHODS DSS was used to induce colitis in mice and the therapeutic effect of WMW was analyzed by disease activity score, histopathological score, colon length measurement, serum cytokine detection, and flow cytometry. Macrophage activation and colonic stem cell proliferation were observed by immunohistochemistry. The expression of critical molecules in macrophage activation and colonic stem cell proliferation signaling pathways in colon tissue was detected with immunohistochemistry, immunofluorescence staining, RT-qPCR, and Western blot. RESULTS WMW could significantly alleviate DSS-induced colitis. We showed that WMW could reduce disease activity, reduce pathological scores, limit weight loss, inhibit colon shortening, inhibit inflammatory factor secretion, attenuate inflammatory response, and promote the repair of damaged colonic epithelium. WMW inhibited the activation of colonic macrophages, and its mechanism might be inhibiting the Notch/NF-κB/NLRP3 pathway; WMW promoted the proliferation of colonic stem cells, and its mechanism was associated with the regulation of the Hippo/YAP signaling pathway. CONCLUSION The results of this study suggested that WMW could treat UC via a mechanism that inhibited the intestinal inflammatory response and repaired damaged intestinal mucosa.
Collapse
Affiliation(s)
- Shuguang Yan
- College of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, China; Key Laboratory of Gastrointestinal Diseases and Prescriptions in Shaanxi Province, Xianyang 712046, China
| | - Ping Wang
- College of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Hailiang Wei
- Department of General Surgery, The Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang 712000, China
| | - Rui Jia
- College of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, China; Key Laboratory of Gastrointestinal Diseases and Prescriptions in Shaanxi Province, Xianyang 712046, China
| | - Meijia Zhen
- College of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, China; Key Laboratory of Gastrointestinal Diseases and Prescriptions in Shaanxi Province, Xianyang 712046, China
| | - Qian Li
- Medical Research and Experiment Center, Shaanxi University of Chinese Medicine, Xianyang 712046
| | - Chang Xue
- Department of Chinese Medicine, Ankang Central Hospital, Ankang 725099, China.
| | - Jingtao Li
- Key Laboratory of Gastrointestinal Diseases and Prescriptions in Shaanxi Province, Xianyang 712046, China; Department of infectious disease, The Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang 712000, China.
| |
Collapse
|
23
|
Villablanca EJ, Selin K, Hedin CRH. Mechanisms of mucosal healing: treating inflammatory bowel disease without immunosuppression? NATURE REVIEWS. GASTROENTEROLOGY & HEPATOLOGY 2022. [PMID: 35440774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/08/2022]
Abstract
Almost all currently available treatments for inflammatory bowel disease (IBD) act by inhibiting inflammation, often blocking specific inflammatory molecules. However, given the infectious and neoplastic disease burden associated with chronic immunosuppressive therapy, the goal of attaining mucosal healing without immunosuppression is attractive. The absence of treatments that directly promote mucosal healing and regeneration in IBD could be linked to the lack of understanding of the underlying pathways. The range of potential strategies to achieve mucosal healing is diverse. However, the targeting of regenerative mechanisms has not yet been achieved for IBD. Stem cells provide hope as a regenerative treatment and are used in limited clinical situations. Growth factors are available for the treatment of short bowel syndrome but have not yet been applied in IBD. The therapeutic application of organoid culture and stem cell therapy to generate new intestinal tissue could provide a novel mechanism to restore barrier function in IBD. Furthermore, blocking key effectors of barrier dysfunction (such as MLCK or damage-associated molecular pattern molecules) has shown promise in experimental IBD. Here, we review the diversity of molecular targets available to directly promote mucosal healing, experimental models to identify new potential pathways and some of the anticipated potential therapies for IBD.
Collapse
Affiliation(s)
- Eduardo J Villablanca
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden.
| | - Katja Selin
- Gastroenterology unit, Department of Gastroenterology, Dermatovenereology and Rheumatology, Karolinska University Hospital, Stockholm, Sweden.,Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Charlotte R H Hedin
- Gastroenterology unit, Department of Gastroenterology, Dermatovenereology and Rheumatology, Karolinska University Hospital, Stockholm, Sweden. .,Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
24
|
Villablanca EJ, Selin K, Hedin CRH. Mechanisms of mucosal healing: treating inflammatory bowel disease without immunosuppression? Nat Rev Gastroenterol Hepatol 2022; 19:493-507. [PMID: 35440774 DOI: 10.1038/s41575-022-00604-y] [Citation(s) in RCA: 111] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/08/2022] [Indexed: 12/12/2022]
Abstract
Almost all currently available treatments for inflammatory bowel disease (IBD) act by inhibiting inflammation, often blocking specific inflammatory molecules. However, given the infectious and neoplastic disease burden associated with chronic immunosuppressive therapy, the goal of attaining mucosal healing without immunosuppression is attractive. The absence of treatments that directly promote mucosal healing and regeneration in IBD could be linked to the lack of understanding of the underlying pathways. The range of potential strategies to achieve mucosal healing is diverse. However, the targeting of regenerative mechanisms has not yet been achieved for IBD. Stem cells provide hope as a regenerative treatment and are used in limited clinical situations. Growth factors are available for the treatment of short bowel syndrome but have not yet been applied in IBD. The therapeutic application of organoid culture and stem cell therapy to generate new intestinal tissue could provide a novel mechanism to restore barrier function in IBD. Furthermore, blocking key effectors of barrier dysfunction (such as MLCK or damage-associated molecular pattern molecules) has shown promise in experimental IBD. Here, we review the diversity of molecular targets available to directly promote mucosal healing, experimental models to identify new potential pathways and some of the anticipated potential therapies for IBD.
Collapse
Affiliation(s)
- Eduardo J Villablanca
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden.
| | - Katja Selin
- Gastroenterology unit, Department of Gastroenterology, Dermatovenereology and Rheumatology, Karolinska University Hospital, Stockholm, Sweden.,Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Charlotte R H Hedin
- Gastroenterology unit, Department of Gastroenterology, Dermatovenereology and Rheumatology, Karolinska University Hospital, Stockholm, Sweden. .,Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
25
|
Horiguchi H, Kadomatsu T, Yumoto S, Masuda T, Miyata K, Yamamura S, Sato M, Morinaga J, Ohtsuki S, Baba H, Moroishi T, Oike Y. Tumor cell-derived ANGPTL2 promotes β-catenin-driven intestinal tumorigenesis. Oncogene 2022; 41:4028-4041. [PMID: 35831580 DOI: 10.1038/s41388-022-02405-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 06/24/2022] [Accepted: 06/29/2022] [Indexed: 11/09/2022]
Abstract
Uncontrolled proliferation of intestinal epithelial cells caused by mutations in genes of the WNT/β-catenin pathway is associated with development of intestinal cancers. We previously reported that intestinal stromal cell-derived angiopoietin-like protein 2 (ANGPTL2) controls epithelial regeneration and intestinal immune responses. However, the role of tumor cell-derived ANGPTL2 in intestinal tumorigenesis remained unclear. Here, we show that tumor cell-derived ANGPTL2 promotes β-catenin-driven intestinal tumorigenesis. ANGPTL2 deficiency suppressed intestinal tumor development in an experimental mouse model of sporadic colon cancer. We also found that increased ANGPTL2 expression in colorectal cancer (CRC) cells augments β-catenin pathway signaling and promotes tumor cell proliferation. Relevant to mechanism, our findings suggest that tumor cell-derived ANGPTL2 upregulates expression of OB-cadherin, which then interacts with β-catenin, blocking destruction complex-independent proteasomal degradation of β-catenin proteins. Moreover, our observations support a model whereby ANGPTL2-induced OB-cadherin expression in CRC cells is accompanied by decreased cell surface integrin α5β1 expression. These findings overall provide novel insight into mechanisms of β-catenin-driven intestinal tumorigenesis.
Collapse
Affiliation(s)
- Haruki Horiguchi
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan.,Department of Aging and Geriatric Medicine, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Tsuyoshi Kadomatsu
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan. .,Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860-8556, Japan.
| | - Shinsei Yumoto
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan.,Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Takeshi Masuda
- Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Keishi Miyata
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan.,Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Shuji Yamamura
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Michio Sato
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Jun Morinaga
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Sumio Ohtsuki
- Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Toshiro Moroishi
- Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860-8556, Japan.,Department of Cell Signaling and Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Yuichi Oike
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan. .,Department of Aging and Geriatric Medicine, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan. .,Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860-8556, Japan.
| |
Collapse
|
26
|
Chiang C, Yang H, Zhu L, Chen C, Chen C, Zuo Y, Zheng D. The Epigenetic Regulation of Nonhistone Proteins by SETD7: New Targets in Cancer. Front Genet 2022; 13:918509. [PMID: 35812730 PMCID: PMC9256981 DOI: 10.3389/fgene.2022.918509] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/27/2022] [Indexed: 11/23/2022] Open
Abstract
Epigenetic modifications are essential mechanism by which to ensure cell homeostasis. One such modification is lysine methylation of nonhistone proteins by SETD7, a mono-methyltransferase containing SET domains. SETD7 methylates over 30 proteins and is thus involved in various classical pathways. As such, SETD7 has been implicated in both the basic functions of normal tissues but also in several pathologies, such as cancers. In this review, we summarize the current knowledge of SETD7 substrates, especially transcriptional-related proteins and enzymes, and their putative roles upon SETD7-mediated methylation. We focus on the role of SETD7 in cancers, and speculate on the possible points of intervention and areas for future research.
Collapse
Affiliation(s)
- Chengyao Chiang
- Southern University of Science and Technology, Yantian Hospital, Shenzhen, China
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Cell Biology and Genetics, Department of Pharmacy, Shenzhen University International Cancer Center, School of Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital (Shenzhen Institute of Translational Medicine), Shenzhen University, Shenzhen, China
| | - Heng Yang
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Cell Biology and Genetics, Department of Pharmacy, Shenzhen University International Cancer Center, School of Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital (Shenzhen Institute of Translational Medicine), Shenzhen University, Shenzhen, China
| | - Lizhi Zhu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Cell Biology and Genetics, Department of Pharmacy, Shenzhen University International Cancer Center, School of Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital (Shenzhen Institute of Translational Medicine), Shenzhen University, Shenzhen, China
| | - Chunlan Chen
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Cell Biology and Genetics, Department of Pharmacy, Shenzhen University International Cancer Center, School of Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital (Shenzhen Institute of Translational Medicine), Shenzhen University, Shenzhen, China
| | - Cheng Chen
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Cell Biology and Genetics, Department of Pharmacy, Shenzhen University International Cancer Center, School of Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital (Shenzhen Institute of Translational Medicine), Shenzhen University, Shenzhen, China
| | - You Zuo
- Southern University of Science and Technology, Yantian Hospital, Shenzhen, China
- *Correspondence: You Zuo, ; Duo Zheng,
| | - Duo Zheng
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Cell Biology and Genetics, Department of Pharmacy, Shenzhen University International Cancer Center, School of Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital (Shenzhen Institute of Translational Medicine), Shenzhen University, Shenzhen, China
- *Correspondence: You Zuo, ; Duo Zheng,
| |
Collapse
|
27
|
A Systematic Review to Define the Multi-Faceted Role of Lysine Methyltransferase SETD7 in Cancer. Cancers (Basel) 2022; 14:cancers14061414. [PMID: 35326563 PMCID: PMC8946661 DOI: 10.3390/cancers14061414] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/02/2022] [Accepted: 03/04/2022] [Indexed: 01/27/2023] Open
Abstract
Histone–lysine N-methyltransferase SETD7 regulates a variety of cancer-related processes, in a tissue-type and signalling context-dependent manner. To date, there is no consensus regarding SETD7´s biological functions, or potential for cancer diagnostics and therapeutics. In this work, we summarised the literature on SETD7 expression and function in cancer, to identify the contexts where SETD7 expression and targeting can lead to improvements in cancer diagnosis and therapy. The most studied cancers were found to be lung and osteosarcoma followed by colorectal and breast cancers. SETD7 mRNA and/or protein expression in human cancer tissue was evaluated using public databases and/or in-house cohorts, but its prognostic significance remains inconclusive. The most studied cancer-related processes regulated by SETD7 were cell proliferation, apoptosis, epithelial-mesenchymal transition, migration and invasion with special relevance to the pRb/E2F-1 pathway. SETD7 consistently prevented epithelial to mesenchymal transition in different cancer types, and inhibition of its function appears to be associated with improved response to DNA-damaging agents in most of the analysed studies. Stabilising mutations in SETD7 target proteins prevent their methylation or promote other competing post-translational modifications that can override the SETD7 effect. This indicates that a clear discrimination of these mutations and competing signalling pathways must be considered in future functional studies.
Collapse
|
28
|
Daks A, Vasileva E, Fedorova O, Shuvalov O, Barlev NA. The Role of Lysine Methyltransferase SET7/9 in Proliferation and Cell Stress Response. Life (Basel) 2022; 12:life12030362. [PMID: 35330113 PMCID: PMC8949485 DOI: 10.3390/life12030362] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/25/2022] [Accepted: 02/26/2022] [Indexed: 12/14/2022] Open
Abstract
Lysine-specific methyltransferase 7 (KMT7) SET7/9, aka Set7, Set9, or SetD7, or KMT5 was discovered 20 years ago, yet its biological role remains rather enigmatic. In this review, we analyze the particularities of SET7/9 enzymatic activity and substrate specificity with respect to its biological importance, mostly focusing on its two well-characterized biological functions: cellular proliferation and stress response.
Collapse
Affiliation(s)
- Alexandra Daks
- Institute of Cytology RAS, 194064 St. Petersburg, Russia; (A.D.); (E.V.); (O.F.); (O.S.)
| | - Elena Vasileva
- Institute of Cytology RAS, 194064 St. Petersburg, Russia; (A.D.); (E.V.); (O.F.); (O.S.)
- Children’s Hospital Los Angeles, University of Southern California, Los Angeles, CA 90027, USA
| | - Olga Fedorova
- Institute of Cytology RAS, 194064 St. Petersburg, Russia; (A.D.); (E.V.); (O.F.); (O.S.)
| | - Oleg Shuvalov
- Institute of Cytology RAS, 194064 St. Petersburg, Russia; (A.D.); (E.V.); (O.F.); (O.S.)
| | - Nickolai A. Barlev
- Institute of Cytology RAS, 194064 St. Petersburg, Russia; (A.D.); (E.V.); (O.F.); (O.S.)
- Correspondence:
| |
Collapse
|
29
|
Ma C, Yu X, Li D, Fan Y, Tang Y, Tao Q, Zheng L. Inhibition of SET domain-containing (lysine methyltransferase) 7 alleviates cognitive impairment through suppressing the activation of NOD-like receptor protein 3 inflammasome in isoflurane-induced aged mice. Hum Exp Toxicol 2022; 41:9603271211061497. [PMID: 35187972 DOI: 10.1177/09603271211061497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND As a common postoperative complication to elderly patients, postoperative cognitive dysfunction (POCD) is a central nervous system complication, often taking place after anesthesia and surgery. (Su(var)3-9, enhancer-of-zeste, and trithorax) domain-containing protein 7 (SETD7) plays important roles in metabolic-related diseases, viral infections, tumor formation, and some inflammatory reactions. However, the role and mechanism of SETD7 in POCD have not been previously studied. METHODS RT-PCR and Western blot were performed to evaluate the efficiency of knockdown of SETD7. The pathological changes of hippocampal neurons in isoflurane-anesthetized mice were detected by HE staining, and the Morris water maze experiment was performed to evaluate the learning and memory abilities of mice. The effect of SETD7 on the hippocampus in isoflurane-induced aged mice was examined by Western blot and TUNEL assay. Then ELISA assay was applied to determine the expression of some inflammatory cytokines, followed by the detection of expression of NOD-like receptor protein 3 (NLRP3) inflammasome through Western blot. RESULTS The data of this research revealed that SETD7 knockdown improved cognitive impairment in isoflurane-anesthetized mice, ameliorated cell pyroptosis, inhibited the release of inflammatory cytokines, and suppressed the activation of NLRP3 inflammasome in the hippocampus in isoflurane-induced aged mice. CONCLUSION Collectively, these results provided evidence that the inhibition of SETD7 could alleviate neuroinflammation, pyroptosis, and cognitive impairment by suppressing the activation of the NLRP3 inflammasome in isoflurane-induced aged mice.
Collapse
Affiliation(s)
- Chao Ma
- Department of Anesthesiology, 543160the Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Xianjun Yu
- Department of Anesthesiology, 543160the Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Dong Li
- Department of General Surgery, 543160the Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Youwen Fan
- Department of General Surgery, 543160the Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Yajun Tang
- Department of General Surgery, 543160the Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Qiang Tao
- Department of General Surgery, 543160the Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Lei Zheng
- Department of Anesthesiology, 543160the Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| |
Collapse
|
30
|
Meriwether D, Jones AE, Ashby JW, Solorzano-Vargas RS, Dorreh N, Noori S, Grijalva V, Ball AB, Semis M, Divakaruni AS, Mack JJ, Herschman HR, Martin MG, Fogelman AM, Reddy ST. Macrophage COX2 Mediates Efferocytosis, Resolution Reprogramming, and Intestinal Epithelial Repair. Cell Mol Gastroenterol Hepatol 2022; 13:1095-1120. [PMID: 35017061 PMCID: PMC8873959 DOI: 10.1016/j.jcmgh.2022.01.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 12/30/2021] [Accepted: 01/03/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Phagocytosis (efferocytosis) of apoptotic neutrophils by macrophages anchors the resolution of intestinal inflammation. Efferocytosis prevents secondary necrosis and inhibits further inflammation, and also reprograms macrophages to facilitate tissue repair and promote resolution function. Macrophage efferocytosis and efferocytosis-dependent reprogramming are implicated in the pathogenesis of inflammatory bowel disease. We previously reported that absence of macrophage cyclooxygenase 2 (COX2) exacerbates inflammatory bowel disease-like intestinal inflammation. To elucidate the underlying pathogenic mechanism, we investigated here whether COX2 mediates macrophage efferocytosis and efferocytosis-dependent reprogramming, including intestinal epithelial repair capacity. METHODS Using apoptotic neutrophils and synthetic apoptotic targets, we determined the effects of macrophage specific Cox2 knockout and pharmacological COX2 inhibition on the efferocytosis capacity of mouse primary macrophages. COX2-mediated efferocytosis-dependent eicosanoid lipidomics was determined by liquid chromatography tandem mass spectrometry. Small intestinal epithelial organoids were employed to assay the effects of COX2 on efferocytosis-dependent intestinal epithelial repair. RESULTS Loss of COX2 impaired efferocytosis in mouse primary macrophages, in part, by affecting the binding capacity of macrophages for apoptotic cells. This effect was comparable to that of high-dose lipopolysaccharide and was accompanied by both dysregulation of macrophage polarization and the inhibited expression of genes involved in apoptotic cell binding. COX2 modulated the production of efferocytosis-dependent lipid inflammatory mediators that include the eicosanoids prostaglandin I2, prostaglandin E2, lipoxin A4, and 15d-PGJ2; and further affected secondary efferocytosis. Finally, macrophage efferocytosis induced, in a macrophage COX2-dependent manner, a tissue restitution and repair phenotype in intestinal epithelial organoids. CONCLUSIONS Macrophage COX2 potentiates efferocytosis capacity and efferocytosis-dependent reprogramming, facilitating macrophage intestinal epithelial repair capacity.
Collapse
Affiliation(s)
- David Meriwether
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California,Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California,Correspondence Address correspondence to: David Meriwether, PhD, Department of Medicine, Division of Digestive Diseases, David Geffen School of Medicine at UCLA, University of California Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095-5347. fax: 310-206-3605.
| | - Anthony E. Jones
- Department of Medical and Molecular Pharmacology, University of California, Los Angeles, Los Angeles, California
| | - Julianne W. Ashby
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - R. Sergio Solorzano-Vargas
- Division of Gastroenterology, Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Nasrin Dorreh
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Shoreh Noori
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Victor Grijalva
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Andréa B. Ball
- Department of Medical and Molecular Pharmacology, University of California, Los Angeles, Los Angeles, California
| | - Margarita Semis
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Ajit S. Divakaruni
- Department of Medical and Molecular Pharmacology, University of California, Los Angeles, Los Angeles, California
| | - Julia J. Mack
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Harvey R. Herschman
- Department of Medical and Molecular Pharmacology, University of California, Los Angeles, Los Angeles, California
| | - Martin G. Martin
- Division of Gastroenterology, Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Alan M. Fogelman
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Srinivasa T. Reddy
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California,Department of Medical and Molecular Pharmacology, University of California, Los Angeles, Los Angeles, California,Srinivasa T. Reddy, PhD, Department of Medicine, David Geffen School of Medicine at UCLA, 10833 Le Conte Avenue, Room 43-144 CHS, Los Angeles, CA 90095-1679. fax: 310-206-3605.
| |
Collapse
|
31
|
Chen Q, Wang H, Li Z, Li F, Liang L, Zou Y, Shen H, Li J, Xia Y, Cheng Z, Yang T, Wang K, Shen F. Circular RNA ACTN4 promotes intrahepatic cholangiocarcinoma progression by recruiting YBX1 to initiate FZD7 transcription. J Hepatol 2022; 76:135-147. [PMID: 34509526 DOI: 10.1016/j.jhep.2021.08.027] [Citation(s) in RCA: 138] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 08/24/2021] [Accepted: 08/26/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Intrahepatic cholangiocarcinoma (ICC) is a primary liver cancer with high aggressiveness and extremely poor prognosis. The role of circular RNAs (circRNAs) in ICC carcinogenesis and progression remains to be determined. METHODS CircRNA microarray was performed to screen significantly upregulated circRNAs in paired ICC and non-tumor tissues. Colony formation, transwell, and xenograft models were used to examine the role of circRNAs in ICC proliferation and metastasis. RNA pulldown, mass spectrometry, chromatin immunoprecipitation, RNA-binding protein immunoprecipitation, chromatin isolation by RNA purification, electrophoretic mobility shift assay, and luciferase reporter assays were used to explore the molecular sponge role of the circRNA (via miRNA binding), and the interaction between circRNA and RNA-binding proteins. RESULTS Hsa_circ_0050898, which originated from exon 1 to exon 20 of the ACTN4 gene (named circACTN4), was significantly upregulated in ICC. High circACTN4 expression was associated with enhanced tumor proliferation and metastasis in vitro and in vivo, as well as a worse prognosis following ICC resection. In addition, circACTN4 upregulated Yes-associated protein 1 (YAP1) expression by sponging miR-424-5p. More importantly, circACTN4 also recruited Y-box binding protein 1 (YBX1) to stimulate Frizzled-7 (FZD7) transcription. Furthermore, circACTN4 overexpression in ICC cells enhanced the interaction between YAP1 and β-catenin, which are the core components of the Hippo and Wnt signaling pathways, respectively. CONCLUSIONS CircACTN4 was upregulated in ICC and promoted ICC proliferation and metastasis by acting as a molecular sponge of miR-424-5p, as well as by interacting with YBX1 to transcriptionally activate FZD7. These results suggest that circACTN4 is a potential prognostic marker and therapeutic target for ICC. LAY SUMMARY Intrahepatic cholangiocarcinoma is a primary liver cancer associated with aggressiveness and extremely poor prognosis. It is essential for therapeutic development that we uncover relevant pathogenic pathways. Herein, we showed that a circular RNA (circACTN4) was highly expressed in intrahepatic cholangiocarcinoma and was positively associated with tumor growth and metastasis through key developmental signaling pathways. Thus, circACTN4 could be a prognostic biomarker and therapeutic target for intrahepatic cholangiocarcinoma.
Collapse
Affiliation(s)
- Qinjunjie Chen
- Department of Hepatic Surgery IV, the Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Haibo Wang
- Department of Hepatic Surgery IV, the Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Zheng Li
- Department of Hepatic Surgery IV, the Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Fengwei Li
- Department of Hepatic Surgery IV, the Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China; Department of Hepatic Surgery II, the Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Leilei Liang
- Department of Gynecological Oncology, Chinese Academy of Medical Sciences Cancer Institute and Hospital: Cancer Hospital Chinese Academy of Medical Sciences, Beijing, China
| | - Yiran Zou
- Department of Hepatic Surgery IV, the Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Hao Shen
- Department of Hepatobiliary Surgery, the Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Jun Li
- Department of Hepatic Surgery IV, the Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Yong Xia
- Department of Hepatic Surgery IV, the Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Zhangjun Cheng
- Hepato-Pancreato-Biliary Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Tian Yang
- Department of Hepatobiliary Surgery, the Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Kui Wang
- Department of Hepatic Surgery II, the Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Feng Shen
- Department of Hepatic Surgery IV, the Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
32
|
Zhang L, Cao R, Li D, Sun Y, Zhang J, Wang X, Khan A, Liu Z, Niu B, Xu J, Xie J. Ethionine-mediated reduction of S-adenosylmethionine is responsible for the neural tube defects in the developing mouse embryo-mediated m6A modification and is involved in neural tube defects via modulating Wnt/β-catenin signaling pathway. Epigenetics Chromatin 2021; 14:52. [PMID: 34863249 PMCID: PMC8645112 DOI: 10.1186/s13072-021-00426-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 11/16/2021] [Indexed: 11/16/2022] Open
Abstract
Neural tube defects (NTDs) remain one of the most life-threatening birth defects affecting infants. Most patients with NTDs eventually develop lifelong disability, which cause significant morbidity and mortality and seriously reduce the quality of life. Our previous study has found that ethionine inhibits cell viability by disrupting the balance between proliferation and apoptosis, and preventing neural stem cells from differentiating into neurons and astrocytes. However, how ethionine participates in the pathogenesis of neural tube development through N6-methyladenosine (m6A) modification remains unknown. This study aims to investigate METTL3- and ALKBH5-mediated m6A modification function and mechanism in NTDs. Herein, our results demonstrate that SAM play not only a compensatory role, it also leads to changes of m6A modification in neural tube development and regulation. Additionally, these data implicate that METTL3 is enriched in HT-22 cells, and METTL3 knockdown reduces cell proliferation and increases apoptosis through suppressing Wnt/β-catenin signaling pathway. Significantly, overexpression of ALKBH5 can only inhibit cell proliferation, but cannot promote cell apoptosis. This research reveals an important role of SAM in development of NTDs, providing a good theoretical basis for further research on NTDs. This finding represents a novel epigenetic mechanism underlying that the m6A modification has profound and lasting implications for neural tube development.
Collapse
Affiliation(s)
- Li Zhang
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,Department of Hepatobiliary and Pancreatic Surgery, First Clinical Medical College, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,Institute of Liver Diseases and Organ Transplantation, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Rui Cao
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,Biology Institute of Shanxi, Taiyuan, 030001, Shanxi, China
| | - Dandan Li
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Yuqing Sun
- Department of Hepatobiliary and Pancreatic Surgery, First Clinical Medical College, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.,Institute of Liver Diseases and Organ Transplantation, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Juan Zhang
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Xiuwei Wang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Ajab Khan
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Zhizhen Liu
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Bo Niu
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| | - Jun Xu
- Department of Hepatobiliary and Pancreatic Surgery, First Clinical Medical College, Shanxi Medical University, Taiyuan, 030001, Shanxi, China. .,Institute of Liver Diseases and Organ Transplantation, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| | - Jun Xie
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
33
|
Astudillo P. An emergent Wnt5a/YAP/TAZ regulatory circuit and its possible role in cancer. Semin Cell Dev Biol 2021; 125:45-54. [PMID: 34764023 DOI: 10.1016/j.semcdb.2021.10.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/04/2021] [Accepted: 10/07/2021] [Indexed: 12/29/2022]
Abstract
Wnt5a is a ligand that plays several roles in development, homeostasis, and disease. A growing body of evidence indicates that Wnt5a is involved in cancer progression. Despite extensive research in this field, our knowledge about how Wnt5a is precisely involved in cancer is still incomplete. It is usually thought that certain combinations of Frizzled receptors and co-receptors might explain the observed effects of Wnt5a either as a tumor suppressor or by promoting migration and invasion. While accepting this 'receptor context' model, this review proposes that Wnt5a is integrated within a larger regulatory circuit involving β-catenin, YAP/TAZ, and LATS1/2. Remarkably, WNT5A and YAP1 are transcriptionally regulated by the Hippo and Wnt pathways, respectively, and might form a regulatory circuit acting through LATS kinases and secreted Wnt/β-catenin inhibitors, including Wnt5a itself. Therefore, understanding the precise role of Wnt5a and YAP in cancer requires a systems biology perspective.
Collapse
Affiliation(s)
- Pablo Astudillo
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile.
| |
Collapse
|
34
|
Daks A, Shuvalov O, Fedorova O, Petukhov A, Lezina L, Zharova A, Baidyuk E, Khudiakov A, Barlev NA. p53-Independent Effects of Set7/9 Lysine Methyltransferase on Metabolism of Non-Small Cell Lung Cancer Cells. Front Oncol 2021; 11:706668. [PMID: 34692483 PMCID: PMC8528242 DOI: 10.3389/fonc.2021.706668] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 09/14/2021] [Indexed: 11/13/2022] Open
Abstract
Set7/9 is a lysine-specific methyltransferase, which regulates the functioning of both the histone and non-histone substrates, thereby significantly affecting the global gene expression landscape. Using microarray expression profiling, we have identified several key master regulators of metabolic networks, including c-Myc, that were affected by Set7/9 status. Consistent with this observation, c-Myc transcriptional targets-genes encoding the glycolytic enzymes hexokinase (HK2), aldolase (ALDOB), and lactate dehydrogenase (LDHA)-were upregulated upon Set7/9 knockdown (Set7/9KD). Importantly, we showed the short hairpin RNA (shRNA)-mediated attenuation of Set7/9 augmented c-Myc, GLUT1, HK2, ALDOA, and LDHA expression in non-small cell lung cancer (NSCLC) cell lines, not only at the transcriptional but also at the protein level. In line with this observation, Set7/9KD significantly augmented the membrane mitochondrial potential (MMP), glycolysis, respiration, and the proliferation rate of NSCLC cells. Importantly, all these effects of Set7/9 on cell metabolism were p53-independent. Bioinformatic analysis has shown a synergistic impact of Set7/9 together with either GLUT1, HIF1A, HK2, or LDHA on the survival of lung cancer patients. Based on these evidence, we hypothesize that Set7/9 can be an important regulator of energy metabolism in NSCLC.
Collapse
Affiliation(s)
- Alexandra Daks
- Institute of Cytology, Russian Academy of Sciences, St Petersburg, Russia
| | - Oleg Shuvalov
- Institute of Cytology, Russian Academy of Sciences, St Petersburg, Russia
| | - Olga Fedorova
- Institute of Cytology, Russian Academy of Sciences, St Petersburg, Russia
| | - Alexey Petukhov
- Institute of Cytology, Russian Academy of Sciences, St Petersburg, Russia.,Institute of Molecular Biology and Genetics, Almazov National Medical Research Centre, St Petersburg, Russia
| | - Larissa Lezina
- Regulation of Cell Signaling Laboratory, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Arsenia Zharova
- Institute of Cytology, Russian Academy of Sciences, St Petersburg, Russia
| | - Ekaterina Baidyuk
- Institute of Cytology, Russian Academy of Sciences, St Petersburg, Russia
| | - Alexander Khudiakov
- Institute of Molecular Biology and Genetics, Almazov National Medical Research Centre, St Petersburg, Russia
| | - Nickolai A Barlev
- Institute of Cytology, Russian Academy of Sciences, St Petersburg, Russia.,Regulation of Cell Signaling Laboratory, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| |
Collapse
|
35
|
Li M, Ning J, Wang J, Yan Q, Zhao K, Jia X. SETD7 regulates chondrocyte differentiation and glycolysis via the Hippo signaling pathway and HIF‑1α. Int J Mol Med 2021; 48:210. [PMID: 34617577 PMCID: PMC8510680 DOI: 10.3892/ijmm.2021.5043] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 08/30/2021] [Indexed: 12/24/2022] Open
Abstract
Chondrocytes are well adapted to hypoxia and produce more functional extracellular matrix in low oxygen environments in vitro. In our previous study, methyltransferase SET domain containing (SETD)7 regulated chondrocyte activity in hypoxic conditions. However, the precise association between SETD7 and chondrocyte differentiation under low oxygen partial pressure remains unclear. The association between SETD7 and chondrocyte differentiation was studied by silencing SETD7 in chondrocytes in vitro. The results showed that the silencing of SETD7 in ATDC5 cells inhibited the Hippo signaling pathway, decreased Yes-associated protein (YAP) phosphorylation and increased the levels of YAP and hypoxia inducible factor-1α (HIF-1α) in the nucleus. YAP combined with HIF-1α to form a complex that promoted the expression of genes involved in chondrogenic differentiation and the glycolytic pathway. Thus, SETD7 inhibited chondrocyte differentiation and glycolysis via the Hippo signaling pathway. The present study demonstrated that SETD7 was a potential molecular target that maintained the chondrocyte phenotype during cartilage tissue engineering and cartilage-associated disease.
Collapse
Affiliation(s)
- Maoquan Li
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat‑sen University, Guangzhou, Guangdong 510055, P.R. China
| | - Jinqiu Ning
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat‑sen University, Guangzhou, Guangdong 510055, P.R. China
| | - Jiwei Wang
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510140, P.R. China
| | - Qiqian Yan
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat‑sen University, Guangzhou, Guangdong 510055, P.R. China
| | - Ke Zhao
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat‑sen University, Guangzhou, Guangdong 510055, P.R. China
| | - Xiaoshi Jia
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat‑sen University, Guangzhou, Guangdong 510055, P.R. China
| |
Collapse
|
36
|
Zhang C, Niu Y, Wang Z, Xu X, Li Y, Ma L, Wang J, Yu Y. Corosolic acid inhibits cancer progression by decreasing the level of CDK19-mediated O-GlcNAcylation in liver cancer cells. Cell Death Dis 2021; 12:889. [PMID: 34588426 PMCID: PMC8481254 DOI: 10.1038/s41419-021-04164-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 08/19/2021] [Accepted: 09/09/2021] [Indexed: 02/08/2023]
Abstract
Diabetes is an important risk factor for liver cancer, but its mechanism is unknown. Corosolic acid (CA) has been proven to have both hypoglycemic and antitumor effects, so revealing the function of CA can help us understand the relationship between diabetes and liver cancer. In previous studies, we confirmed that CA can effectively inhibit the expression of YAP, an important oncoprotein in HCC cells, and the proliferation of HCC cells. In addition, we also found that O-GlcNAcylation plays an indispensable role in HCC tumorigenesis. However, it is not clear whether CA can inhibit the effect of O-GlcNAcylation on HCC cells. In this study, the antitumor ability of CA was investigated by inhibiting the O-GlcNAcylation level and its corresponding mechanism. The results showed that HG (high glucose) could promote the proliferation of liver cancer cells, while CA could inhibit cell growth under HG conditions and tumor growth in a xenotransplantation model. CA can inhibit the activation of the HBP pathway and reduce the expression of YAP and OGT under HG conditions. Importantly, we found that CA can reduce YAP expression and O-GlcNAcylation by inhibiting the activity of CDK19. Overexpression of CDK19 partially reversed the CA-induced decrease in YAP and O-GlcNAcylation. This is the first evidence that CA can reduce the proliferative capacity of cells with high glucose levels and further inhibit tumor growth by inactivating the CDK19/YAP/O-GlcNAcylation pathway, suggesting that CA is a candidate drug for the development of treatments against diabetes-associated liver cancer.
Collapse
Affiliation(s)
- Congcong Zhang
- Institute for Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, 200030, Shanghai, P.R. China
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 200071, Shanghai, P.R. China
- Henan University of Chinese Medicine, 450046, Zhengzhou, P.R. China
| | - Yongjie Niu
- Institute for Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, 200030, Shanghai, P.R. China
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 200071, Shanghai, P.R. China
| | - Zhixian Wang
- Institute for Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, 200030, Shanghai, P.R. China
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 200071, Shanghai, P.R. China
| | - Xin Xu
- Institute for Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, 200030, Shanghai, P.R. China
| | - Yan Li
- Department of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 200071, Shanghai, P.R. China
| | - Lifang Ma
- Institute for Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, 200030, Shanghai, P.R. China.
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, 200030, Shanghai, China.
| | - Jiayi Wang
- Institute for Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, 200030, Shanghai, P.R. China.
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, 200030, Shanghai, China.
| | - Yongchun Yu
- Institute for Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, 200030, Shanghai, P.R. China.
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 200071, Shanghai, P.R. China.
| |
Collapse
|
37
|
Norrie disease protein is essential for cochlear hair cell maturation. Proc Natl Acad Sci U S A 2021; 118:2106369118. [PMID: 34544869 DOI: 10.1073/pnas.2106369118] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/13/2021] [Indexed: 11/18/2022] Open
Abstract
Mutations in the gene for Norrie disease protein (Ndp) cause syndromic deafness and blindness. We show here that cochlear function in an Ndp knockout mouse deteriorated with age: At P3-P4, hair cells (HCs) showed progressive loss of Pou4f3 and Gfi1, key transcription factors for HC maturation, and Myo7a, a specialized myosin required for normal function of HC stereocilia. Loss of expression of these genes correlated to increasing HC loss and profound hearing loss by 2 mo. We show that overexpression of the Ndp gene in neonatal supporting cells or, remarkably, up-regulation of canonical Wnt signaling in HCs rescued HCs and cochlear function. We conclude that Ndp secreted from supporting cells orchestrates a transcriptional network for the maintenance and survival of HCs and that increasing the level of β-catenin, the intracellular effector of Wnt signaling, is sufficient to replace the functional requirement for Ndp in the cochlea.
Collapse
|
38
|
Sharma A, Mir R, Galande S. Epigenetic Regulation of the Wnt/β-Catenin Signaling Pathway in Cancer. Front Genet 2021; 12:681053. [PMID: 34552611 PMCID: PMC8450413 DOI: 10.3389/fgene.2021.681053] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 06/30/2021] [Indexed: 12/12/2022] Open
Abstract
Studies over the past four decades have elucidated the role of Wnt/β-catenin mediated regulation in cell proliferation, differentiation and migration. These processes are fundamental to embryonic development, regeneration potential of tissues, as well as cancer initiation and progression. In this review, we focus on the epigenetic players which influence the Wnt/β-catenin pathway via modulation of its components and coordinated regulation of the Wnt target genes. The role played by crosstalk with other signaling pathways mediating tumorigenesis is also elaborated. The Hippo/YAP pathway is particularly emphasized due to its extensive crosstalk via the Wnt destruction complex. Further, we highlight the recent advances in developing potential therapeutic interventions targeting the epigenetic machinery based on the characterization of these regulatory networks for effective treatment of various cancers and also for regenerative therapies.
Collapse
Affiliation(s)
- Ankita Sharma
- Centre of Excellence in Epigenetics, Department of Biology, Indian Institute of Science Education and Research, Pune, India
| | - Rafeeq Mir
- Centre for Interdisciplinary Research and Innovations, University of Kashmir, Srinagar, India
| | - Sanjeev Galande
- Centre of Excellence in Epigenetics, Department of Biology, Indian Institute of Science Education and Research, Pune, India.,Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Greater Noida, India
| |
Collapse
|
39
|
Sun C, He B, Sun M, Lv X, Wang F, Chen J, Zhang J, Ye Z, Wen J, Liu P. Yes-Associated Protein in Atherosclerosis and Related Complications: A Potential Therapeutic Target That Requires Further Exploration. Front Cardiovasc Med 2021; 8:704208. [PMID: 34513949 PMCID: PMC8430249 DOI: 10.3389/fcvm.2021.704208] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 08/06/2021] [Indexed: 11/13/2022] Open
Abstract
Atherosclerosis and its complications diseases remain leading causes of cardiovascular morbidity and mortality, bringing a massive burden on public health worldwide. Atherosclerosis is recognized as chronic inflammation, and involves several highly correlated processes, including lipid metabolism dysfunction, endothelial cell dysfunction, inflammation, oxidative stress, vascular smooth muscle cell activation, platelet activation, thrombosis, altered matrix metabolism, and vascular remodeling. Within the past few decades, accumulating evidence has shown that the Yes-associated protein (YAP), the major effector of the Hippo pathway, can play a crucial role in pathogenesis and development of atherosclerosis. Activation of YAP-related pathways, which are induced by alerting flow pattern and matrix stiffness among others, can regulate processes including vascular endothelial cell dysfunction, monocyte infiltration, and smooth muscle cell migration, which contribute to atherosclerotic lesion formation. Further, YAP potentially modulates atherosclerotic complications such as vascular calcification and intraplaque hemorrhage, which require further investigation. Here, we summarized the relevant literature to outline current findings detailing the relationship between of YAP and atherosclerosis and highlight areas for future research.
Collapse
Affiliation(s)
- Congrui Sun
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Bin He
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Mingsheng Sun
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Xiaoshuo Lv
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Feng Wang
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Jie Chen
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Jianbin Zhang
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Zhidong Ye
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Jianyan Wen
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China.,Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Peng Liu
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China.,Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
40
|
Wu MH, Padilla-Rodriguez M, Blum I, Camenisch A, Figliuolo da Paz V, Ollerton M, Muller J, Momtaz S, Mitchell SAT, Kiela P, Thorne C, Wilson JM, Cox CM. Proliferation in the developing intestine is regulated by the endosomal protein Endotubin. Dev Biol 2021; 480:50-61. [PMID: 34411593 DOI: 10.1016/j.ydbio.2021.08.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 08/05/2021] [Accepted: 08/14/2021] [Indexed: 11/19/2022]
Abstract
During postnatal intestinal development, the intestinal epithelium is highly proliferative, and this proliferation is regulated by signaling in the intervillous and crypt regions. This signaling is primarily mediated by Wnt, and requires membrane trafficking. However, the mechanisms by which membrane trafficking regulates signaling during this developmental phase are largely unknown. Endotubin (EDTB, MAMDC4) is an endosomal protein that is highly expressed in the apical endocytic complex (AEC) of villus enterocytes during fetal and postnatal development, and knockout of EDTB results in defective formation of the AEC and giant lysosome. Further, knockout of EDTB in cell lines results in decreased proliferation. However, the role of EDTB in proliferation during the development of the intestine is unknown. Using Villin-CreERT2 in EDTBfl/fl mice, we deleted EDTB in the intestine in the early postnatal period, or in enteroids in vitro after isolation of intervillous cells. Loss of EDTB results in decreased proliferation in the developing intestinal epithelium and decreased ability to form enteroids. EDTB is present in cells that contain the stem cell markers LGR5 and OLFM4, indicating that it is expressed in the proliferative compartment. Further, using immunoblot analysis and TCF/LEF-GFP mice as a reporter of Wnt activity, we find that knockout of EDTB results in decreased Wnt signaling. Our results show that EDTB is essential for normal proliferation during the early stages of intestinal development and suggest that this effect is through modulation of Wnt signaling.
Collapse
Affiliation(s)
- Meng-Han Wu
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA.
| | | | - Isabella Blum
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA.
| | - Abigail Camenisch
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA.
| | | | | | - John Muller
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA.
| | - Samina Momtaz
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA.
| | - Stefanie A T Mitchell
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA.
| | - Pawel Kiela
- Departments of Pediatrics and Immunobiology, University of Arizona, Tucson, AZ, USA; Steele Children's Research Center, University of Arizona, Tucson, AZ, USA.
| | - Curtis Thorne
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA; The University of Arizona Cancer Center, University of Arizona, Tucson, AZ, USA; Bio5 Institute, University of Arizona, Tucson, AZ, USA; Steele Children's Research Center, University of Arizona, Tucson, AZ, USA.
| | - Jean M Wilson
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA; The University of Arizona Cancer Center, University of Arizona, Tucson, AZ, USA; Bio5 Institute, University of Arizona, Tucson, AZ, USA.
| | - Christopher M Cox
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
41
|
Li Y, Yang G, Yang C, Tang P, Chen J, Zhang J, Liu J, Ouyang L. Targeting Autophagy-Related Epigenetic Regulators for Cancer Drug Discovery. J Med Chem 2021; 64:11798-11815. [PMID: 34378389 DOI: 10.1021/acs.jmedchem.1c00579] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Existing evidence has demonstrated that epigenetic modifications (including DNA methylation, histone modifications, and microRNAs), which are associated with the occurrence and development of tumors, can directly or indirectly regulate autophagy. In particular, nuclear events induced by several epigenetic regulators can regulate the autophagic process and expression levels of tumor-associated genes, thereby promoting tumor progression. Tumor-associated microRNAs, including oncogenic and tumor-suppressive microRNAs, are of great significance to autophagy during tumor progression. Targeting autophagy with emerging epigenetic drugs is expected to be a promising therapeutic strategy for human tumors. From this perspective, we aim to summarize the role of epigenetic modification in the autophagic process and the underlying molecular mechanisms of tumorigenesis. Furthermore, the regulatory efficacy of epigenetic drugs on the autophagic process in tumors is also summarized. This perspective may provide a theoretical basis for the combined treatment of epigenetic drugs/autophagy mediators in tumors.
Collapse
Affiliation(s)
- Yang Li
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Gaoxia Yang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Chengcan Yang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Pan Tang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Juncheng Chen
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Jifa Zhang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Jie Liu
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Liang Ouyang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
42
|
Wang H, Ma P, Liu P, Guo D, Liu Z, Zhang Z. lncRNA SNHG6 promotes hepatocellular carcinoma progression by interacting with HNRNPL/PTBP1 to facilitate SETD7/LZTFL1 mRNA destabilization. Cancer Lett 2021; 520:121-131. [PMID: 34252487 DOI: 10.1016/j.canlet.2021.07.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 07/01/2021] [Accepted: 07/06/2021] [Indexed: 01/03/2023]
Abstract
The lncRNA SNHG6 (small nucleolar RNA host gene 6) plays vital roles in tumorigenesis and the progression of hepatocellular carcinoma (HCC). However, the regulatory mechanisms of SNHG6 are largely unknown. In this study, we identified, via quantitative proteomics, specific cytoskeleton-associated proteins and enzyme modulators to be potential targets of SNHG6. SNHG6 reduced the mRNA levels of lysine methyltransferase, SET domain containing 7 (SETD7) and leucine zipper transcription factor-like 1 (LZTFL1) by posttranscriptional destabilization. Silencing of SETD7 or LZTFL1 reversed the suppressive effects of SNHG6 knockdown on HCC progression. Heterogeneous nuclear ribonucleoprotein L (HNRNPL) and polypyrimidine tract binding protein 1 (PTBP1) were identified as SNHG6-interacting proteins that bind to SETD7 or LZTFL1 mRNA. Forced expression of SNHG6 led to HNRNPL being competitively adsorbed by SNHG6, thereby removing its stabilizing effect on SETD7. Concurrently, the functional SNHG6-PTBP1 complex facilitated the degradation of LZTFL1 mRNA in hepatoma cells. These results indicated that SNHG6 promotes HCC progression by functioning as a "decoy plus guide" for HNRNPL and PTBP1 to facilitate mRNA decay of SETD7 and LZTFL1, thereby serving as a novel therapeutic target for HCC.
Collapse
Affiliation(s)
- Haitao Wang
- Department of Hepatobiliary and Pancreas, Research Center of Digestive Diseases, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Pei Ma
- Center for Gene Diagnosis, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China; Department of Forensic Medicine, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Pengpeng Liu
- Department of Hepatobiliary and Pancreas, Research Center of Digestive Diseases, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Deliang Guo
- Department of Hepatobiliary and Pancreas, Research Center of Digestive Diseases, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Zhisu Liu
- Department of Hepatobiliary and Pancreas, Research Center of Digestive Diseases, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China.
| | - Zhonglin Zhang
- Department of Hepatobiliary and Pancreas, Research Center of Digestive Diseases, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
43
|
Yang L, Jin M, Jeong KW. Histone H3K4 Methyltransferases as Targets for Drug-Resistant Cancers. BIOLOGY 2021; 10:581. [PMID: 34201935 PMCID: PMC8301125 DOI: 10.3390/biology10070581] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 06/16/2021] [Accepted: 06/22/2021] [Indexed: 12/30/2022]
Abstract
The KMT2 (MLL) family of proteins, including the major histone H3K4 methyltransferase found in mammals, exists as large complexes with common subunit proteins and exhibits enzymatic activity. SMYD, another H3K4 methyltransferase, and SET7/9 proteins catalyze the methylation of several non-histone targets, in addition to histone H3K4 residues. Despite these structural and functional commonalities, H3K4 methyltransferase proteins have specificity for their target genes and play a role in the development of various cancers as well as in drug resistance. In this review, we examine the overall role of histone H3K4 methyltransferase in the development of various cancers and in the progression of drug resistance. Compounds that inhibit protein-protein interactions between KMT2 family proteins and their common subunits or the activity of SMYD and SET7/9 are continuously being developed for the treatment of acute leukemia, triple-negative breast cancer, and castration-resistant prostate cancer. These H3K4 methyltransferase inhibitors, either alone or in combination with other drugs, are expected to play a role in overcoming drug resistance in leukemia and various solid cancers.
Collapse
Affiliation(s)
- Liu Yang
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, China;
| | - Mingli Jin
- Gachon Research Institute of Pharmaceutical Sciences, College of Pharmacy, Gachon University, 191 Hambakmoero, Yeonsu-gu, Incheon 21936, Korea;
| | - Kwang Won Jeong
- Gachon Research Institute of Pharmaceutical Sciences, College of Pharmacy, Gachon University, 191 Hambakmoero, Yeonsu-gu, Incheon 21936, Korea;
| |
Collapse
|
44
|
Xing J, Jie W. Methyltransferase SET domain family and its relationship with cardiovascular development and diseases. Zhejiang Da Xue Xue Bao Yi Xue Ban 2021; 51:251-260. [PMID: 35462466 DOI: 10.3724/zdxbyxb-2021-0192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Abnormal epigenetic modification is closely related to the occurrence and development of cardiovascular diseases. The SET domain (SETD) family is an important epigenetic modifying enzyme containing SETD. They mainly affect gene expression by methylating H3K4, H3K9, H3K36 and H4K20. Additionally, the SETD family catalyzes the methylation of non-histone proteins, thereby affects the signal transduction of signal transduction and activator of transcription (STAT) 1, Wnt/β-catenin, hypoxia-inducible factor (HIF)-1α and Hippo/YAP pathways. The SETD family has the following regulatory effects on cardiovascular development and diseases: regulating coronary artery formation and cardiac development; protecting cardiac tissue from ischemia reperfusion injury; regulating inflammation, oxidative stress and apoptosis in cardiovascular complications of diabetes; participating in the formation of pulmonary hypertension; regulating thrombosis, cardiac hypertrophy and arrhythmia. This article summarizes the basic structures, expression regulation mechanisms and the role of existing SETD family members in cardiovascular development and diseases, in order to provide a basis for understanding the molecular mechanism of cardiovascular disease and exploring the therapeutic targets.
Collapse
Affiliation(s)
- Jingci Xing
- 1. Department of Pathology, School of Basic Medicine Sciences, Guangdong Medical University, Zhanjiang 524023, Guangdong Province, China
| | - Wei Jie
- 1. Department of Pathology, School of Basic Medicine Sciences, Guangdong Medical University, Zhanjiang 524023, Guangdong Province, China.,Medical University, Key Laboratory of Emergency and Trauma, Ministry of Education, Hainan Provincial Key Laboratory of Tropical Cardiovascular Diseases Research, Haikou 571199, China
| |
Collapse
|
45
|
Wang L, Chennupati R, Jin YJ, Li R, Wang S, Günther S, Offermanns S. YAP/TAZ Are Required to Suppress Osteogenic Differentiation of Vascular Smooth Muscle Cells. iScience 2020; 23:101860. [PMID: 33319178 PMCID: PMC7726335 DOI: 10.1016/j.isci.2020.101860] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 09/10/2020] [Accepted: 11/20/2020] [Indexed: 12/22/2022] Open
Abstract
Vascular smooth muscle cells (VSMCs) represent the prevailing cell type of arterial vessels and are essential for blood vessel structure and homeostasis. They have substantial potential for phenotypic plasticity when exposed to various stimuli in their local microenvironment. How VSMCs maintain their differentiated contractile phenotype is still poorly understood. Here we demonstrate that the Hippo pathway effectors YAP and TAZ play a critical role in maintaining the differentiated contractile phenotype of VSMCs. In the absence of YAP/TAZ, VSMCs lose their differentiated phenotype and undergo osteogenic differentiation, which results in vascular calcification. Osteogenic transdifferentiation was accompanied by the upregulation of Wnt target genes. The absence of YAP/TAZ in VSMCs led to Disheveled 3 (DVL3) nuclear translocation and upregulation of osteogenesis-associated genes independent of canonical Wnt/β-catenin signaling activation. Our data indicate that cytoplasmic YAP/TAZ interact with DVL3 to avoid its nuclear translocation and osteogenic differentiation, thereby maintaining the differentiated phenotype of VSMCs. YAP/TAZ play an important role in maintaining vascular SMCs contractile phenotype Loss of YAP/TAZ in vSMCs leads to reduced expression of smooth muscle marker genes Loss of YAP/TAZ in vSMCs results in reduced artery contractility Deficiency of YAP/TAZ in vSMCs leads to osteogenic transdifferentiation
Collapse
Affiliation(s)
- Lei Wang
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim 61231, Germany
| | - Ramesh Chennupati
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim 61231, Germany
| | - Young-June Jin
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim 61231, Germany
| | - Rui Li
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim 61231, Germany
| | - ShengPeng Wang
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim 61231, Germany.,Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Yanta District, Xi'an 710061, China
| | - Stefan Günther
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Stefan Offermanns
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim 61231, Germany.,Center for Molecular Medicine, Medical Faculty, Goethe University, Frankfurt am Main 60590, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Frankfurt Rhine-Main, 13347 Berlin, Germany
| |
Collapse
|
46
|
Sun L, Rollins D, Qi Y, Fredericks J, Mansell TJ, Jergens A, Phillips GJ, Wannemuehler M, Wang Q. TNFα regulates intestinal organoids from mice with both defined and conventional microbiota. Int J Biol Macromol 2020; 164:548-556. [PMID: 32693143 PMCID: PMC7657954 DOI: 10.1016/j.ijbiomac.2020.07.176] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 07/07/2020] [Accepted: 07/14/2020] [Indexed: 02/07/2023]
Abstract
Cytokines are key factors affecting the fate of intestinal stem cells (ISCs) and effective reagents to manipulate ISCs for research purpose. Tumor necrosis factor alpha (TNFα) is a cytokine produced primarily by monocytes and macrophages. It can induce apoptotic cell death and inflammation, and to inhibit tumorigenesis and viral replication. Additionally, TNFα has been shown to play a critical role in the pathogenesis of inflammatory bowel disease (IBD). It is therefore important to identify the mechanism by which individual cytokines affect particular cell types. For this purpose, we used both conventional (CONV) and altered Schaedler flora (ASF) C3H/HeN mice to elucidate the effect of different microbial populations (complex versus defined) on growth of miniguts derived from two different intestinal environments. Furthermore, we studied the effects of different concentrations of TNFα extracted from the lymph and spleen on the growth and viability of ISCs recovered from mice bearing the ASF or CONV microbiota. The effect of TNFα on miniguts growth depends not only on the source and concentration, but also on the intestinal microenvironment from which the ISCs were derived. The findings suggest that TNFα influences the proliferation of miniguts derived from ISCs and, therefore, modulates mucosal homeostasis of the host.
Collapse
Affiliation(s)
- Liping Sun
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, United States; School of Environmental & Resource Sciences, Zhejiang A&F University, Lin'an, Zhejiang 311300, China
| | - Derrick Rollins
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, United States; Department of Statistics, Iowa State University, Ames, IA, United States
| | - Yijun Qi
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, United States
| | - Jorrell Fredericks
- Department of Veterinary Microbiology and Preventative Medicine, Iowa State University, Ames, IA, United States
| | - Thomas J Mansell
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, United States
| | - Albert Jergens
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, IA, United States
| | - Gregory J Phillips
- Department of Veterinary Microbiology and Preventative Medicine, Iowa State University, Ames, IA, United States
| | - Michael Wannemuehler
- Department of Veterinary Microbiology and Preventative Medicine, Iowa State University, Ames, IA, United States
| | - Qun Wang
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, United States.
| |
Collapse
|
47
|
YAP/TAZ Signalling in Colorectal Cancer: Lessons from Consensus Molecular Subtypes. Cancers (Basel) 2020; 12:cancers12113160. [PMID: 33126419 PMCID: PMC7692643 DOI: 10.3390/cancers12113160] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/23/2020] [Accepted: 10/26/2020] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Colorectal cancer (CRC) is a heterogeneous disease that can be divided into 4 consensus molecular subtypes (CMS) according to molecular profiling. The CMS classification is now considered as a reference framework for understanding the heterogeneity of CRC and for the implementation of precision medicine. Although the contribution of YAP/TAZ signalling to CRC has been intensively studied, there is little information on its role within each CMS subtype. This article aims to provide an overview of our knowledge of YAP/TAZ in CRC through the lens of the CMS classification. Abstract Recent advance in the characterization of the heterogeneity of colorectal cancer has led to the definition of a consensus molecular classification within four CMS subgroups, each associated with specific molecular and clinical features. Investigating the signalling pathways that drive colorectal cancer progression in relation to the CMS classification may help design therapeutic strategies tailored for each CMS subtype. The two main effectors of the Hippo pathway YAP and its paralogue TAZ have been intensively scrutinized for their contribution to colon carcinogenesis. Here, we review the knowledge of YAP/TAZ implication in colorectal cancer from the perspective of the CMS framework. We identify gaps in our current understanding and delineate research avenues for future work.
Collapse
|
48
|
Reggiani F, Gobbi G, Ciarrocchi A, Sancisi V. YAP and TAZ Are Not Identical Twins. Trends Biochem Sci 2020; 46:154-168. [PMID: 32981815 DOI: 10.1016/j.tibs.2020.08.012] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 08/13/2020] [Accepted: 08/27/2020] [Indexed: 12/12/2022]
Abstract
Yes-associated protein (YAP) and TAZ (WW domain containing transcription regulator 1, or WWTR1) are paralog transcriptional regulators, able to integrate mechanical, metabolic, and signaling inputs to regulate cell growth and differentiation during development and neoplastic progression. YAP and TAZ hold common and distinctive structural features, reflecting only partially overlapping regulatory mechanisms. The two paralogs interact with both shared and specific transcriptional partners and control nonidentical transcriptional programs. Although most of the available literature considers YAP and TAZ as functionally redundant, they play distinctive or even contrasting roles in different contexts. The issue of their divergent roles is currently underexplored but holds fundamental implications for mechanistic and translational studies. Here, we aim to review the available literature on the biological functions of YAP and TAZ, highlighting differential roles that distinguish these two paralogues.
Collapse
Affiliation(s)
- Francesca Reggiani
- Laboratory of Translational Research, Azienda USL- IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Giulia Gobbi
- Laboratory of Translational Research, Azienda USL- IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Alessia Ciarrocchi
- Laboratory of Translational Research, Azienda USL- IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Valentina Sancisi
- Laboratory of Translational Research, Azienda USL- IRCCS di Reggio Emilia, Reggio Emilia, Italy.
| |
Collapse
|
49
|
Yu Y, Su X, Qin Q, Hou Y, Zhang X, Zhang H, Jia M, Chen Y. Yes-associated protein and transcriptional coactivator with PDZ-binding motif as new targets in cardiovascular diseases. Pharmacol Res 2020; 159:105009. [DOI: 10.1016/j.phrs.2020.105009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 05/14/2020] [Accepted: 06/05/2020] [Indexed: 12/12/2022]
|
50
|
Rugo HS, Jacobs I, Sharma S, Scappaticci F, Paul TA, Jensen-Pergakes K, Malouf GG. The Promise for Histone Methyltransferase Inhibitors for Epigenetic Therapy in Clinical Oncology: A Narrative Review. Adv Ther 2020; 37:3059-3082. [PMID: 32445185 PMCID: PMC7467409 DOI: 10.1007/s12325-020-01379-x] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Indexed: 12/21/2022]
Abstract
Epigenetic processes are essential for normal development and the maintenance of tissue-specific gene expression in mammals. Changes in gene expression and malignant cellular transformation can result from disruption of epigenetic mechanisms, and global disruption in the epigenetic landscape is a key feature of cancer. The study of epigenetics in cancer has revealed that human cancer cells harbor both genetic alterations and epigenetic abnormalities that interplay at all stages of cancer development. Unlike genetic mutations, epigenetic aberrations are potentially reversible through epigenetic therapy, providing a therapeutically relevant treatment option. Histone methyltransferase inhibitors are emerging as an epigenetic therapy approach with great promise in the field of clinical oncology. The recent accelerated approval of the enhancer of zeste homolog 2 (EZH2; also known as histone-lysine N-methyltransferase EZH2) inhibitor tazemetostat for metastatic or locally advanced epithelioid sarcoma marks the first approval of such a compound for the treatment of cancer. Many other histone methyltransferase inhibitors are currently in development, some of which are being tested in clinical studies. This review focuses on histone methyltransferase inhibitors, highlighting their potential in the treatment of cancer. We also discuss the role for such epigenetic drugs in overcoming epigenetically driven drug resistance mechanisms, and their value in combination with other therapeutic approaches such as immunotherapy.
Collapse
|