1
|
Zhao Y, Zhu L, Lin X, Li B, Miu B, Qiu J, Gao S, Liu J. JS-K induces ferroptosis in renal carcinoma cells by regulating the c-Myc-GSTP1 Axis. Sci Rep 2025; 15:15987. [PMID: 40341677 PMCID: PMC12062407 DOI: 10.1038/s41598-025-97887-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 04/08/2025] [Indexed: 05/10/2025] Open
Abstract
JS-K is a precursor drug of nitric oxide (NO) and inhibits tumor growth through various mechanisms. Ferroptosis, a form of cell death closely related to lipid peroxidation, is increasingly being recognized for its role in cancer biology. However, the relevance of ferroptosis in the anti-tumor effects of JS-K is yet to be defined. The cytotoxic effects of erastin and JS-K were evaluated in various renal cell carcinoma (RCC) cell lines and normal human renal epithelial cells. Cell viability and the intracellular levels of ferrous ions, glutathione (GSH), lipid peroxides, and malondialdehyde (MDA) were measured using standard in vitro assays. The expression levels of specific proteins were analyzed by western blotting. Subcutaneous xenografts of RCC were established in a nude mouse model, and the anti-tumor effects of JS-K were assessed by histological and immunohistochemical methods. Erastin selectively inhibited the growth of RCC cells without affecting normal renal cells. In addition, JS-K induced ferroptosis in RCC cells by reducing cellular GSH levels, increasing lipid peroxidation, and elevating ferrous ion levels, and the effects of JS-K were neutralized by N-acetylcysteine (NAC). At the molecular level, JS-K downregulated GSTP1 by blocking the transcription factor c-Myc. Finally, JS-K inhibited tumor growth in a mouse model by inducing ferroptosis. JS-K induces ferroptosis in RCC cells by depleting glutathione through the inhibition of the c-Myc-GSTP1 axis.
Collapse
Affiliation(s)
- Yuwan Zhao
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, 57 Renmin Street South, Zhanjiang, 524001, Guangdong, China
| | - LuGang Zhu
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, 57 Renmin Street South, Zhanjiang, 524001, Guangdong, China
| | - Xinghua Lin
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, 57 Renmin Street South, Zhanjiang, 524001, Guangdong, China
| | - Bin Li
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, 57 Renmin Street South, Zhanjiang, 524001, Guangdong, China
| | - Bailiang Miu
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, 57 Renmin Street South, Zhanjiang, 524001, Guangdong, China
| | - Jingping Qiu
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, 57 Renmin Street South, Zhanjiang, 524001, Guangdong, China
| | - Sheng Gao
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, 57 Renmin Street South, Zhanjiang, 524001, Guangdong, China
| | - Jianjun Liu
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, 57 Renmin Street South, Zhanjiang, 524001, Guangdong, China.
| |
Collapse
|
2
|
Iyer AR, Gurumurthy A, Chu SCA, Kodgule R, Aguilar AR, Saari T, Ramzan A, Rosa J, Gupta J, Emmanuel A, Hall CN, Runge JS, Owczarczyk AB, Cho JW, Weiss MB, Anyoha R, Sikkink K, Gemus S, Fulco CP, Perry AM, Schmitt AD, Engreitz JM, Brown NA, Cieslik MP, Ryan RJ. Selective Enhancer Dependencies in MYC-Intact and MYC-Rearranged Germinal Center B-cell Diffuse Large B-cell Lymphoma. Blood Cancer Discov 2025; 6:233-253. [PMID: 40067173 PMCID: PMC12050968 DOI: 10.1158/2643-3230.bcd-24-0126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 12/28/2024] [Accepted: 03/10/2025] [Indexed: 03/15/2025] Open
Abstract
SIGNIFICANCE Aberrant MYC activity defines the most aggressive GCB-DLBCLs. We characterized a mechanism of MYC transcriptional activation via a native enhancer that is active in MYC-intact GCB-DLBCL, establishing fitness-sustaining cis- and trans-regulatory circuitry in GCB-DLBCL models that lack MYC enhancer-hijacking rearrangement. See related commentary by Mulet-Lazaro and Delwel, p. 149.
Collapse
Affiliation(s)
- Ashwin R. Iyer
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Aishwarya Gurumurthy
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Shih-Chun A. Chu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Rohan Kodgule
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Athalee R. Aguilar
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Travis Saari
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Abdullah Ramzan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Jan Rosa
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Juhi Gupta
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Arvind Emmanuel
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Cody N. Hall
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - John S. Runge
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Anna B. Owczarczyk
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Jang W. Cho
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Matthew B. Weiss
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Rockwell Anyoha
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts
| | | | | | - Charles P. Fulco
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts
| | - Anamarija M. Perry
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | | | - Jesse M. Engreitz
- Department of Genetics, Stanford University School of Medicine, Stanford, California
- BASE Initiative, Betty Irene Moore Children’s Heart Center, Lucile Packard Children’s Hospital, Stanford, California
- Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Noah A. Brown
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Marcin P. Cieslik
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Russell J.H. Ryan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
3
|
Ysebaert L, Mouchel PL, Laurent C, Quillet-Mary A. The multi-faceted roles of MYC in the prognosis of chronic lymphocytic leukemia. Leuk Lymphoma 2025; 66:805-817. [PMID: 39743868 DOI: 10.1080/10428194.2024.2447362] [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: 08/03/2024] [Revised: 12/19/2024] [Accepted: 12/21/2024] [Indexed: 01/04/2025]
Abstract
In this review, we focus on the pro-oncogene MYC, the modes of deregulation in mouse and human B-cells, its undisputable importance in the evaluation of biological prognostication of patients, but also how it impacts on response to modern therapeutics, and how it should be targeted to improve the overall survival of chronic lymphocytic lymphoma (CLL) patients. After an overview of the current understanding of the molecular dysregulation of c-MYC, we will show how CLL, both in its indolent and transformed phases, has developed among other B-cell lymphomas a tight regulation of its expression through the chronic activation of B-Cell Receptors (among others). This is particularly important if one desires to understand the mechanisms at stake in the over-expression of c-MYC especially in the lymph nodes compartment. So doing, we will show how this oncogene orchestrates pivotal cellular functions such as metabolism, drug resistance, proliferation and histologic transformation (Richter syndrome).
Collapse
MESH Headings
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Lymphocytic, Chronic, B-Cell/mortality
- Prognosis
- Proto-Oncogene Proteins c-myc/genetics
- Proto-Oncogene Proteins c-myc/metabolism
- Animals
- Gene Expression Regulation, Leukemic
Collapse
Affiliation(s)
- Loic Ysebaert
- Centre de Recherches en Cancérologie de Toulouse, INSERM UMR1037, CNRS UMR5071, Université Toulouse III-Paul Sabatier, Toulouse, France
- Laboratoire d'Excellence 'TOUCAN-2', Toulouse, France
- Department of Hematology, IUC Toulouse-Oncopole, Toulouse, France
| | | | - Camille Laurent
- Centre de Recherches en Cancérologie de Toulouse, INSERM UMR1037, CNRS UMR5071, Université Toulouse III-Paul Sabatier, Toulouse, France
- Laboratoire d'Excellence 'TOUCAN-2', Toulouse, France
- Department of Hematology, IUC Toulouse-Oncopole, Toulouse, France
| | - Anne Quillet-Mary
- Centre de Recherches en Cancérologie de Toulouse, INSERM UMR1037, CNRS UMR5071, Université Toulouse III-Paul Sabatier, Toulouse, France
- Laboratoire d'Excellence 'TOUCAN-2', Toulouse, France
| |
Collapse
|
4
|
Linstra R, Stappenbelt C, Bakker FJ, Everts M, Bhattacharya A, Yu S, van Bergen SD, van der Vegt B, Wisman GBA, Fehrmann RSN, de Bruyn M, van Vugt MATM. MYC controls STING levels to downregulate inflammatory signaling in breast cancer cells upon DNA damage. J Biol Chem 2025:108560. [PMID: 40311680 DOI: 10.1016/j.jbc.2025.108560] [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: 10/11/2024] [Revised: 04/22/2025] [Accepted: 04/24/2025] [Indexed: 05/03/2025] Open
Abstract
Amplification of the MYC proto-oncogene is frequently observed in various cancer types, including triple negative breast cancer (TNBC). Emerging evidence suggests that suppression of local anti-tumor immune responses by MYC, at least in part, explains the tumor-promoting effects of MYC. Specifically, MYC upregulation was demonstrated to suppress the tumor-cell intrinsic activation of a type I IFN response and thereby hamper innate inflammatory signaling, which may contribute to the disappointing response to immunotherapy in patients with TNBC. In this study, we show that MYC interferes with protein expression and functionality of the STING pathway. MYC-mediated STING downregulation in BT-549 and MDA-MB-231 triple-negative breast cancer cell lines require the DNA binding ability of MYC, and is independent of binding of MYC to its co-repressor MIZ1. Both STAT1 and STAT3 promote the steady-state expression levels of STING, and STAT3 cooperates with MYC in regulating STING. Conversely, MYC-mediated downregulation of STING affects protein levels of STAT1 and downstream chemokine production. Furthermore, we show that MYC overexpression hampers immune cell activation triggered by DNA damage through etoposide or irradiation treatment, and specifically impedes the activation of natural killer cells. Collectively, these results show that MYC controls STING levels and thereby regulates tumor cell-intrinsic inflammatory signaling. These results contribute to our understanding of how MYC suppresses inflammatory signaling in TNBC, and may explain why a large fraction of patients with TNBC do not benefit from immunotherapy.
Collapse
Affiliation(s)
- Renske Linstra
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713GZ, Groningen, the Netherlands
| | - Chantal Stappenbelt
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713GZ, Groningen, the Netherlands
| | - Femke J Bakker
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713GZ, Groningen, the Netherlands
| | - Marieke Everts
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713GZ, Groningen, the Netherlands
| | - Arkajyoti Bhattacharya
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713GZ, Groningen, the Netherlands
| | - Shibo Yu
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Stella D van Bergen
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713GZ, Groningen, the Netherlands
| | - Bert van der Vegt
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - G Bea A Wisman
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Rudolf S N Fehrmann
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713GZ, Groningen, the Netherlands
| | - Marco de Bruyn
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Marcel A T M van Vugt
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713GZ, Groningen, the Netherlands.
| |
Collapse
|
5
|
Wang S, Han P, Mi P, Wang C, Lu M, Li X, Xu B, Wang H, Gao Y, Hou Y, Tan X, Liang J, Ding X, Zhang Y, Zhang T, Yuan D, Gao L, Zhang C. The Role of the Hexosamine-Sialic Acid Metabolic Pathway Mediated by GFPT1/NANS in c-Myc-Driven Hepatocellular Carcinoma. Cell Mol Gastroenterol Hepatol 2025:101523. [PMID: 40280277 DOI: 10.1016/j.jcmgh.2025.101523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 04/10/2025] [Accepted: 04/16/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND & AIMS Hepatocellular carcinoma (HCC) frequently involves metabolic reprogramming, which promotes oncogenesis and metastasis. However, the underlying molecular mechanisms remain insufficiently explored. In this study, we aim to investigate the metabolic abnormalities in c-Myc-driven HCC development and their potential therapeutic implications. METHODS RNA sequencing and metabolomics were performed on HCC and adjacent tissues in a murine HCC model established by hydrodynamic tail-vein injection of c-Myc and sgTrp53/Cas9 plasmids. Key catalytic enzyme gene knockout was used to assess tumor formation and murine survival. Gene expression was analyzed using quantitative polymerase chain reaction, immunohistochemistry, and Western blot. Chromatin immunoprecipitation followed by quantitative polymerase chain reaction and luciferase assays verified c-Myc regulation. RESULTS RNA sequencing data revealed that the hexosamine biosynthetic pathway was significantly activated in c-Myc-driven HCC. The rate-limiting enzyme GFPT1 (rather than GFPT2) was up-regulated in the first step of this pathway. Knocking out GFPT1 reduces tumor growth and prolongs murine survival. Human specimens showed that GFPT1 was overexpressed in HCC tissues and was associated with advanced Edmondson-Steiner grades and short patient survival. Further luciferase reporter assays confirmed that c-Myc binds directly to the promoter region of GFPT1 and activates its transcription. Subsequent examination of the downstream pathways of the hexosamine biosynthetic pathway showed that the sialic acid synthesis (but not O-GlcNac glycosylation) pathway was enhanced, which was mediated by a key enzyme, N-acetylneuraminic acid synthase. Knockout of N-acetylneuraminic acid synthase also inhibits tumor growth and extends murine survival in c-Myc-driven HCC models. CONCLUSIONS These findings indicate that the activation of the hexosamine biosynthetic pathway/sialic acid pathway is an important mechanism underlying the development of c-Myc-driven HCC. Inhibitors of GFPT1, along with anti- N-acetylneuraminic acid synthase may offer a promising therapeutic strategy.
Collapse
Affiliation(s)
- Shiguan Wang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, China; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Pan Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China; State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Ping Mi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chunxue Wang
- Department of Pathology, School of Basic Medical Sciences and Qilu Hospital, Shandong University, Jinan, China; Department of Pathology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Miao Lu
- Hepato-Pancreato-Biliary Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Xinying Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bowen Xu
- Advanced Medical Research Institute, Shandong University, Jinan, China
| | - Haoran Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yingchen Gao
- Department of Pathology, School of Basic Medical Sciences and Qilu Hospital, Shandong University, Jinan, China
| | - Yanlei Hou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xueying Tan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jinyuan Liang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xue Ding
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yan Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tingguo Zhang
- Department of Pathology, School of Basic Medical Sciences and Qilu Hospital, Shandong University, Jinan, China
| | - Detian Yuan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lei Gao
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China.
| | - Cuijuan Zhang
- Department of Pathology, School of Basic Medical Sciences and Qilu Hospital, Shandong University, Jinan, China.
| |
Collapse
|
6
|
Sberna S, Filipuzzi M, Bianchi N, Croci O, Fardella F, Soriani C, Rohban S, Carnevali S, Albertini AA, Crosetto N, Rodighiero S, Chiesa A, Curti L, Campaner S. Senataxin prevents replicative stress induced by the Myc oncogene. Cell Death Dis 2025; 16:187. [PMID: 40108134 PMCID: PMC11923212 DOI: 10.1038/s41419-025-07485-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 02/14/2025] [Accepted: 02/26/2025] [Indexed: 03/22/2025]
Abstract
Replicative stress (RS) is emerging as a promising therapeutic target in oncology, yet full exploitation of its potential requires a detailed understanding of the mechanisms and genes involved. Here, we investigated the RNA helicase Senataxin (SETX), an enzyme that resolves RNA-DNA hybrids and R-loops, to address its role in preventing RS by oncogenic Myc. Upon Myc activation, silencing of SETX led to selective engagement of the DNA damage response (DDR) and robust cytotoxicity. Pharmacological dissection of the upstream kinases regulating the DDR uncovered a protective role of the ATR pathway, that once inhibited, boosted SETX driven-DDR. While SETX loss did not lead to a genome-wide increase of R-loops, mechanistic analyses revealed enhanced R-loops localized at DDR-foci and newly replicated genomic loci, compatible with a selective role of SETX in resolving RNA-DNA hybrids to alleviate Myc-induced RS. Genome-wide mapping of DNA double-strand breaks confirmed that SETX silencing exacerbated DNA damage at transcription-replication conflict (TRC) regions at early replicated sites. We propose that SETX prevents Myc-induced TRCs by resolving transcription-associated R-loops that encounter the replisome. The identification of SETX as a genetic liability of oncogenic Myc opens up new therapeutic options against aggressive Myc-driven tumors.
Collapse
Affiliation(s)
- Silvia Sberna
- Center for Genomic Science of IIT, CGS@SEMM (Istituto Italiano di Tecnologia at European School of Molecular Medicine), Fondazione Istituto Italiano di Tecnologia (IIT), 20139, Milan, Italy
| | - Marco Filipuzzi
- Center for Genomic Science of IIT, CGS@SEMM (Istituto Italiano di Tecnologia at European School of Molecular Medicine), Fondazione Istituto Italiano di Tecnologia (IIT), 20139, Milan, Italy
| | - Nicola Bianchi
- Center for Genomic Science of IIT, CGS@SEMM (Istituto Italiano di Tecnologia at European School of Molecular Medicine), Fondazione Istituto Italiano di Tecnologia (IIT), 20139, Milan, Italy
| | - Ottavio Croci
- Center for Genomic Science of IIT, CGS@SEMM (Istituto Italiano di Tecnologia at European School of Molecular Medicine), Fondazione Istituto Italiano di Tecnologia (IIT), 20139, Milan, Italy
| | - Federica Fardella
- Center for Genomic Science of IIT, CGS@SEMM (Istituto Italiano di Tecnologia at European School of Molecular Medicine), Fondazione Istituto Italiano di Tecnologia (IIT), 20139, Milan, Italy
| | - Chiara Soriani
- Imaging Unit, Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Sara Rohban
- Center for Genomic Science of IIT, CGS@SEMM (Istituto Italiano di Tecnologia at European School of Molecular Medicine), Fondazione Istituto Italiano di Tecnologia (IIT), 20139, Milan, Italy
| | - Sara Carnevali
- Center for Genomic Science of IIT, CGS@SEMM (Istituto Italiano di Tecnologia at European School of Molecular Medicine), Fondazione Istituto Italiano di Tecnologia (IIT), 20139, Milan, Italy
| | | | - Nicola Crosetto
- Human Technopole, Viale Rita Levi-Montalcini 1, 20157, Milan, Italy
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, SE, 17165, Sweden
- Science for Life Laboratory, Tomtebodavägen 23A, Solna, SE, 17165, Sweden
| | - Simona Rodighiero
- Imaging Unit, Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Arianna Chiesa
- Center for Genomic Science of IIT, CGS@SEMM (Istituto Italiano di Tecnologia at European School of Molecular Medicine), Fondazione Istituto Italiano di Tecnologia (IIT), 20139, Milan, Italy
| | - Laura Curti
- Center for Genomic Science of IIT, CGS@SEMM (Istituto Italiano di Tecnologia at European School of Molecular Medicine), Fondazione Istituto Italiano di Tecnologia (IIT), 20139, Milan, Italy
| | - Stefano Campaner
- Center for Genomic Science of IIT, CGS@SEMM (Istituto Italiano di Tecnologia at European School of Molecular Medicine), Fondazione Istituto Italiano di Tecnologia (IIT), 20139, Milan, Italy.
- Department of Molecular Medicine, University of Padua, Padua, Italy.
| |
Collapse
|
7
|
Martin Sobral L, Walker FM, Madhavan K, Janko E, Donthula S, Balakrishnan I, Wang D, Pierce A, Haag MM, Carstens BJ, Serkova NJ, Foreman NK, Venkataraman S, Veo B, Vibhakar R, Dahl NA. Targeting processive transcription for Myc-driven circuitry in medulloblastoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.14.643337. [PMID: 40166273 PMCID: PMC11956955 DOI: 10.1101/2025.03.14.643337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Background Medulloblastoma is the most common malignant brain tumor of childhood. The highest-risk tumors are driven by recurrent Myc amplifications (Myc-MB) and experience poorer outcomes despite intensive multimodal therapy. The Myc transcription factor defines core regulatory circuitry for these tumors and acts to broadly amplify downstream pro-survival transcriptional programs. Therapeutic targeting of Myc directly has proven elusive, but inhibiting transcriptional cofactors may present an indirect means of drugging the oncogenic transcriptional circuitry sustaining Myc-MB. Methods Independent CRISPR-Cas9 screens were pooled to identify conserved dependencies in Myc-MB. We performed chromatin conformation capture (Hi-C) from primary patient Myc-MB samples to map enhancer-promoter interactions. We then treated in vitro and xenograft models with CDK9/7 inhibitors to evaluate effect on Myc-driven programs and tumor growth. Results Eight CRISPR-Cas9 screens performed across three independent labs identify CDK9 as a conserved dependency in Myc-MB. Myc-MB cells are susceptible to CDK9 inhibition, which is synergistic with concurrent inhibition of CDK7. Inhibition of transcriptional CDKs disrupts enhancer-promoter activity in Myc-MB and downregulates Myc-driven transcriptional programs, exerting potent anti-tumor effect. Conclusions Our findings identify CDK9 inhibition as a translationally promising strategy for the treatment of Myc-MB. K ey P oints CDK9 is an intrinsic dependency in Myc-driven medulloblastomaDual CDK9/7 inhibition disrupts Myc-driven transcriptional circuitryCDK9 inhibitors should be developed as pharmaceutical agents for Myc-MB. I mportance of the S tudy Medulloblastoma is the most common malignant brain tumor of childhood, and outcomes for high-risk subgroups remain unsatisfactory despite intensive multimodal therapy. In this study, we pool multiple independent CRISPR-Cas9 screens to identify transcriptional cofactors such as CDK9 as conserved dependencies in Myc-MB. Using Hi-C from primary patient samples, we map Myc enhancer-promoter interactions and show that they can be disrupted using inhibition of transcriptional CDKs. CDK9 inhibitor treatment depletes Myc-driven transcriptional programs, leading to potent anti-tumor effect in vitro and prolongation of xenograft survival in vivo . With a large number of CDK9 inhibitory compounds now in clinical development, this study highlights the opportunity for clinical translation of these for children diagnosed with Myc-MB.
Collapse
|
8
|
Watanabe H, Inoue Y, Tsuchiya K, Asada K, Suzuki M, Ogawa H, Tanahashi M, Watanabe T, Matsuura S, Yasuda K, Ohnishi I, Imokawa S, Yasui H, Karayama M, Suzuki Y, Hozumi H, Furuhashi K, Enomoto N, Fujisawa T, Funai K, Shinmura K, Sugimura H, Inui N, Suda T. Lethal co-expression intolerance underlies the mutually exclusive expression of ASCL1 and NEUROD1 in SCLC cells. NPJ Precis Oncol 2025; 9:74. [PMID: 40082639 PMCID: PMC11906894 DOI: 10.1038/s41698-025-00860-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 02/28/2025] [Indexed: 03/16/2025] Open
Abstract
Small cell lung cancer (SCLC) subtypes, defined by the expression of lineage-specific transcription factors (TFs), are thought to be mutually exclusive, with intra-tumoral heterogeneities. This study investigated the mechanism underlying this phenomenon with the aim of identifying a novel vulnerability of SCLC. We profiled the expression status of ASCL1, NEUROD1, POU2F3, and YAP1 in 151 surgically obtained human SCLC samples. On subtyping, a high degree of mutual exclusivity was observed between ASCL1 and NEUROD1 expression at the cell, but not tissue, level. Inducible co-expression models of all combinations of ASCL1, NEUROD1, POU2F3, YAP1, and ATOH1 using SCLC cell lines showed that some expression combinations, such as ASCL1 and NEUROD1, exhibited mutual repression and caused growth inhibition and apoptosis. Gene expression and ATAC-seq analyses of the ASCL1 and NEUROD1 co-expression models revealed that co-expression of ASCL1 in NEUROD1-driven cells, and of NEUROD1 in ASCL1-driven cells, both (although more efficiently by the former) reprogrammed the cell lineage to favor the ectopically expressed factor, with rewiring of chromatin accessibility. Mechanistically, co-expressed NEUROD1 in ASCL1-driven SCLC cells caused apoptosis by downregulating BCL2, likely in a MYC-independent manner. In conclusion, lethal co-expression intolerance underlies the mutual exclusivity between these pioneer TFs, ASCL1 and NEUROD1, in an SCLC cell. Further investigation is warranted to enable therapeutic targeting of this vulnerability.
Collapse
Affiliation(s)
- Hirofumi Watanabe
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yusuke Inoue
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| | - Kazuo Tsuchiya
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kazuhiro Asada
- Department of Respiratory Medicine, Shizuoka General Hospital, Shizuoka, Japan
| | - Makoto Suzuki
- Department of Pathology, Shizuoka General Hospital, Shizuoka, Japan
| | - Hiroshi Ogawa
- Department of Pathology, Seirei Mikatahara General Hospital, Hamamatsu, Japan
| | - Masayuki Tanahashi
- Division of Thoracic Surgery, Respiratory Disease Center, Seirei Mikatahara General Hospital, Hamamatsu, Japan
| | - Takuya Watanabe
- Division of Thoracic Surgery, Respiratory Disease Center, Seirei Mikatahara General Hospital, Hamamatsu, Japan
| | - Shun Matsuura
- Department of Respiratory Medicine, Fujieda Municipal General Hospital, Fujieda, Japan
| | - Kazuyo Yasuda
- Department of Pathology, Shizuoka General Hospital, Shizuoka, Japan
- Department of Pathology, Fujieda Municipal General Hospital, Fujieda, Japan
| | - Ippei Ohnishi
- Division of Pathology, Iwata City Hospital, Iwata, Japan
| | - Shiro Imokawa
- Department of Respiratory Medicine, Iwata City Hospital, Iwata, Japan
| | - Hideki Yasui
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Masato Karayama
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Department of Chemotherapy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yuzo Suzuki
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hironao Hozumi
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kazuki Furuhashi
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Noriyuki Enomoto
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Tomoyuki Fujisawa
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kazuhito Funai
- First Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kazuya Shinmura
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Haruhiko Sugimura
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Sasaki Institute, Sasaki Foundation, Tokyo, Japan
| | - Naoki Inui
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Department of Clinical Pharmacology and Therapeutics, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takafumi Suda
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
9
|
Avolio E, Bassani B, Campanile M, Mohammed KA, Muti P, Bruno A, Spinetti G, Madeddu P. Shared molecular, cellular, and environmental hallmarks in cardiovascular disease and cancer: Any place for drug repurposing? Pharmacol Rev 2025; 77:100033. [PMID: 40148035 DOI: 10.1016/j.pharmr.2024.100033] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 12/17/2024] [Indexed: 03/29/2025] Open
Abstract
Cancer and cardiovascular disease (CVD) are the 2 biggest killers worldwide. Specific treatments have been developed for the 2 diseases. However, mutual therapeutic targets should be considered because of the overlap of cellular and molecular mechanisms. Cancer research has grown at a fast pace, leading to an increasing number of new mechanistic treatments. Some of these drugs could prove useful for treating CVD, which realizes the concept of cancer drug repurposing. This review provides a comprehensive outline of the shared hallmarks of cancer and CVD, primarily ischemic heart disease and heart failure. We focus on chronic inflammation, altered immune response, stromal and vascular cell activation, and underlying signaling pathways causing pathological tissue remodeling. There is an obvious scope for targeting those shared mechanisms, thereby achieving reciprocal preventive and therapeutic benefits. Major attention is devoted to illustrating the logic, advantages, challenges, and viable examples of drug repurposing and discussing the potential influence of sex, gender, age, and ethnicity in realizing this approach. Artificial intelligence will help to refine the personalized application of drug repurposing for patients with CVD. SIGNIFICANCE STATEMENT: Cancer and cardiovascular disease (CVD), the 2 biggest killers worldwide, share several underlying cellular and molecular mechanisms. So far, specific therapies have been developed to tackle the 2 diseases. However, the development of new cardiovascular drugs has been slow compared with cancer drugs. Understanding the intersection between pathological mechanisms of the 2 diseases provides the basis for repurposing cancer therapeutics for CVD treatment. This approach could allow the rapid development of new drugs for patients with CVDs.
Collapse
Affiliation(s)
- Elisa Avolio
- Bristol Heart Institute, Laboratory of Experimental Cardiovascular Medicine, Translational Health Sciences, Bristol Medical School, University of Bristol, United Kingdom.
| | - Barbara Bassani
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, IRCCS MultiMedica, Milan, Italy
| | - Marzia Campanile
- Laboratory of Cardiovascular Pathophysiology - Regenerative Medicine, IRCCS MultiMedica, Milan, Italy; Department of Biosciences, University of Milan, Milan, Italy
| | - Khaled Ak Mohammed
- Bristol Heart Institute, Laboratory of Experimental Cardiovascular Medicine, Translational Health Sciences, Bristol Medical School, University of Bristol, United Kingdom; Department of Cardiothoracic Surgery, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Paola Muti
- IRCCS MultiMedica, Milan, Italy; Department of Biomedical, Surgical and Dental Health Sciences, University of Milan, Italy
| | - Antonino Bruno
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, IRCCS MultiMedica, Milan, Italy; Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy.
| | - Gaia Spinetti
- Laboratory of Cardiovascular Pathophysiology - Regenerative Medicine, IRCCS MultiMedica, Milan, Italy.
| | - Paolo Madeddu
- Bristol Heart Institute, Laboratory of Experimental Cardiovascular Medicine, Translational Health Sciences, Bristol Medical School, University of Bristol, United Kingdom.
| |
Collapse
|
10
|
Popovic M, Isermann L, Geißen S, Senft K, Georgomanolis T, Baldus S, Frezza C, Trifunovic A. Tissue-specific adaptations to cytochrome c oxidase deficiency shape physiological outcomes. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167567. [PMID: 39613003 DOI: 10.1016/j.bbadis.2024.167567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/05/2024] [Accepted: 11/05/2024] [Indexed: 12/01/2024]
Abstract
It becomes increasingly clear that the tissue specificity of mitochondrial diseases might in part rely on their ability to compensate for mitochondrial defects, contributing to the heterogeneous nature of mitochondrial diseases. Here, we investigated tissue-specific responses to cytochrome c oxidase (CIV or COX) deficiency using a mouse model with heart and skeletal muscle-specific depletion of the COX assembly factor COX10. At three weeks of age, both tissues exhibit pronounced CIV depletion but respond differently to oxidative phosphorylation (OXPHOS) impairment. Heart-specific COX10 depletion caused severe dilated cardiomyopathy, while skeletal muscle experiences less damage. Cardiac CIV deficiency triggered extensive metabolic remodelling and stress response activation, potentially worsening cardiomyopathy, whereas skeletal muscle showed no stress response or significant metabolic changes. Our findings highlight distinct tissue capacities for managing CIV deficiency, explaining how identical primary defects can lead to different phenotypic outcomes and contribute to the heterogeneous progression of mitochondrial diseases.
Collapse
Affiliation(s)
- Milica Popovic
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Institute for Mitochondrial Diseases and Aging, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50931 Cologne, Germany
| | - Lea Isermann
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Institute for Mitochondrial Diseases and Aging, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50931 Cologne, Germany
| | - Simon Geißen
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, 50937, Germany; Clinic III for Internal Medicine, University Hospital Cologne, 50937, Germany; Center for Molecular Medicine (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Katharina Senft
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Institute for Mitochondrial Diseases and Aging, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50931 Cologne, Germany
| | - Theodoros Georgomanolis
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Institute for Metabolomics in Ageing, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50931 Cologne, Germany
| | - Stephan Baldus
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, 50937, Germany; Clinic III for Internal Medicine, University Hospital Cologne, 50937, Germany; Center for Molecular Medicine (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Christian Frezza
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Institute for Metabolomics in Ageing, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50931 Cologne, Germany; Institute of Genetics, Faculty of Mathematics and Natural Sciences, University of Cologne, D-50931 Cologne, Germany
| | - Aleksandra Trifunovic
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Institute for Mitochondrial Diseases and Aging, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50931 Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, 50931 Cologne, Germany.
| |
Collapse
|
11
|
Huang YC, Yuan TM, Liu BH, Liang RY, Liu KL, Chuang SM. GCIP and SIRT6 cooperatively suppress ITGAV gene expression by modulating c-myc transcription ability. J Biol Chem 2025; 301:108314. [PMID: 39955062 PMCID: PMC11930424 DOI: 10.1016/j.jbc.2025.108314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 01/23/2025] [Accepted: 01/25/2025] [Indexed: 02/17/2025] Open
Abstract
Grap2 and CyclinD1 interacting protein (GCIP) has been suggested to function as a tumor suppressor and acts as a transcriptional regulator that negatively controls cancer cell growth, invasion, and migration. Knockdown of GCIP reportedly enhances cancer cell migration and invasion, but no previous study has examined the mechanism(s) by which GCIP suppresses migration/invasion in cancer cells. Here, we report that cDNA microarray-based expression profiling of A549 cells without and with knockdown of GCIP reveals that the expression levels of ITGAV and ICAM-1 are negatively regulated by GCIP. In vitro co-immunoprecipitation and in vivo proximity ligation assays reveal that GCIP interacts with c-Myc. Sequence analyses reveal the presence of two c-Myc regulatory motifs (E-boxes) within the ITGAV promoter. Luciferase reporter and ChIP assays indicate that GCIP represses ITGAV transcription by interacting with c-Myc on the E-box binding sites of the ITGAV promoter region. Furthermore, GCIP interacts with SIRT6 in vitro and in vivo and cooperates with SIRT6, thereby linking its activity, to negatively regulate transcription at the E-box by modulating c-Myc transcription ability. Taken together, these findings contribute to our understanding of GCIP in tumorigenesis and identify a previously unrecognized function of GCIP: It can interact with c-Myc and SIRT6 at E-box binding sites of the ITGAV promoter region. Our data collectively reveal a regulatory network involving GCIP, SIRT6, c-Myc, and ITGAV, and suggest that the SIRT6-GCIP complex negatively regulates the oncogenic function of c-Myc in cell proliferation and migration.
Collapse
Affiliation(s)
- Yi-Ching Huang
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Tien-Ming Yuan
- Department of Surgery, Feng Yuan Hospital, Ministry of Health and Welfare, Taichung, Taiwan; Department of Dental Technology and Materials Science, Central Taiwan University of Science and Technology, Taichung, Taiwan
| | - Bang-Hung Liu
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Ruei-Yue Liang
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Kai-Li Liu
- Department of Nutrition, Chung Shan Medical University, Taichung, Taiwan; Department of Nutrition, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Show-Mei Chuang
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan; Department of Law, National Chung Hsing University, Taichung, Taiwan.
| |
Collapse
|
12
|
Rosen HT, Li K, Li EL, Currier B, Brittain SM, Garcia FJ, Beard DC, Haenni-Holzinger S, Dovala D, McKenna JM, Schirle M, Maimone TJ, Nomura DK. Sulfinyl Aziridines as Stereoselective Covalent Destabilizing Degraders of the Oncogenic Transcription Factor MYC. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.24.639755. [PMID: 40060528 PMCID: PMC11888305 DOI: 10.1101/2025.02.24.639755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/18/2025]
Abstract
While MYC is a significant oncogenic transcription factor driver of cancer, directly targeting MYC has remained challenging due to its intrinsic disorder and poorly defined structure, deeming it "undruggable." Whether transient pockets formed within intrinsically disordered and unstructured regions of proteins can be selectively targeted with small molecules remains an outstanding challenge. Here, we developed a bespoke stereochemically-paired spirocyclic oxindole aziridine covalent library and screened this library for degradation of MYC. Through this screen, we identified a hit covalent ligand KL2-236, bearing a unique sulfinyl aziridine warhead, that engaged MYC in vitro as pure MYC/MAX protein complex and in situ in cancer cells to destabilize MYC, inhibit MYC transcriptional activity and degrade MYC in a proteasome-dependent manner through targeting intrinsically disordered C203 and D205 residues. Notably, this reactivity was most pronounced for specific stereoisomers of KL2-236 with a diastereomer KL4-019 that was largely inactive. Mutagenesis of both C203 and D205 completely attenuated KL2-236-mediated MYC degradation. We have also optimized our initial KL2-236 hit compound to generate a more durable MYC degrader KL4-219A in cancer cells. Our results reveal a novel ligandable site within MYC and indicate that certain intrinsically disordered regions within high-value protein targets, such as MYC, can be interrogated by isomerically unique chiral small molecules, leading to destabilization and degradation.
Collapse
Affiliation(s)
- Hannah T. Rosen
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720 USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720 USA
- Innovative Genomics Institute, Berkeley, CA 94720 USA
- Novartis-Berkeley Translational Chemical Biology Institute, Berkeley, CA 94720 USA
| | - Kelvin Li
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720 USA
- Novartis-Berkeley Translational Chemical Biology Institute, Berkeley, CA 94720 USA
| | - Erin L. Li
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720 USA
- Novartis-Berkeley Translational Chemical Biology Institute, Berkeley, CA 94720 USA
| | - Brynne Currier
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720 USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720 USA
- Innovative Genomics Institute, Berkeley, CA 94720 USA
- Novartis-Berkeley Translational Chemical Biology Institute, Berkeley, CA 94720 USA
| | - Scott M. Brittain
- Novartis-Berkeley Translational Chemical Biology Institute, Berkeley, CA 94720 USA
- Novartis Biomedical Research, Emeryville, CA USA; Cambridge, MA USA; Basel, Switzerland
| | - Francisco J. Garcia
- Novartis-Berkeley Translational Chemical Biology Institute, Berkeley, CA 94720 USA
- Novartis Biomedical Research, Emeryville, CA USA; Cambridge, MA USA; Basel, Switzerland
| | - Diana C. Beard
- Novartis-Berkeley Translational Chemical Biology Institute, Berkeley, CA 94720 USA
- Novartis Biomedical Research, Emeryville, CA USA; Cambridge, MA USA; Basel, Switzerland
| | - Sandra Haenni-Holzinger
- Novartis-Berkeley Translational Chemical Biology Institute, Berkeley, CA 94720 USA
- Novartis Biomedical Research, Emeryville, CA USA; Cambridge, MA USA; Basel, Switzerland
| | - Dustin Dovala
- Novartis-Berkeley Translational Chemical Biology Institute, Berkeley, CA 94720 USA
- Novartis Biomedical Research, Emeryville, CA USA; Cambridge, MA USA; Basel, Switzerland
| | - Jeffrey M. McKenna
- Novartis-Berkeley Translational Chemical Biology Institute, Berkeley, CA 94720 USA
- Novartis Biomedical Research, Emeryville, CA USA; Cambridge, MA USA; Basel, Switzerland
| | - Markus Schirle
- Novartis-Berkeley Translational Chemical Biology Institute, Berkeley, CA 94720 USA
- Novartis Biomedical Research, Emeryville, CA USA; Cambridge, MA USA; Basel, Switzerland
| | - Thomas J. Maimone
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720 USA
- Novartis-Berkeley Translational Chemical Biology Institute, Berkeley, CA 94720 USA
| | - Daniel K. Nomura
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720 USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720 USA
- Innovative Genomics Institute, Berkeley, CA 94720 USA
- Novartis-Berkeley Translational Chemical Biology Institute, Berkeley, CA 94720 USA
| |
Collapse
|
13
|
Grunewald L, Andersch L, Helmsauer K, Schwiebert S, Klaus A, Henssen AG, Straka T, Lodrini M, Wicha SG, Fuchs S, Hertwig F, Westermann F, Vitali A, Caramel C, Büchel G, Eilers M, Astrahantseff K, Eggert A, Höpken UE, Schulte JH, Blankenstein T, Anders K, Künkele A. Targeting MYCN upregulates L1CAM tumor antigen in MYCN-dysregulated neuroblastoma to increase CAR T cell efficacy. Pharmacol Res 2025; 212:107608. [PMID: 39828101 DOI: 10.1016/j.phrs.2025.107608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 12/18/2024] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
Current treatment protocols have limited success against MYCN-amplified neuroblastoma. Adoptive T cell therapy presents an innovative strategy to improve cure rates. However, L1CAM-targeting CAR T cells achieved only limited response against refractory/relapsed neuroblastoma so far. We investigated how oncogenic MYCN levels influence tumor cell response to CAR T cells, as one possible factor limiting clinical success. A MYCN-inducible neuroblastoma cell model was created. L1CAM-CAR T cell effector function was assessed (activation markers, cytokine release, tumor cytotoxicity) after coculture with the model or MYCN-amplified neuroblastoma cell lines. RNA sequencing datasets characterizing the model were compared to publicly available RNA/proteomic datasets. MYCN-directed L1CAM regulation was explored using public ChIP-sequencing datasets. Synergism between CAR T cells and the indirect MYCN inhibitor, MLN8237, was assessed in vitro using the Bliss model and in vivo in an immunocompromised mouse model. Inducing high MYCN levels in the neuroblastoma cell model reduced L1CAM expression and, consequently, L1CAM-CAR T cell effector function in vitro. Primary neuroblastomas possessing high MYCN levels expressed lower levels of both the L1CAM transcript and L1CAM tumor antigen. MLN8237 treatment restored L1CAM tumor expression and L1CAM-CAR T cell effector function. Combining MLN8237 and L1CAM-CAR T cell treatment synergistically enhanced MYCN-overexpressing tumor cytotoxicity in vitro and in vivo concomitant with severe in vivo toxicity. We identify target antigen downregulation as source of resistance against L1CAM-CAR T cells in MYCN-driven neuroblastoma cells. These data suggest that L1CAM-CAR T cell therapy combined with pharmacological MYCN inhibition may benefit patients with MYCN-amplified neuroblastoma.
Collapse
Affiliation(s)
- Laura Grunewald
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Pediatric Oncology and Hematology, Augustenburger Platz 1, Berlin 13353, Germany; German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Lena Andersch
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Pediatric Oncology and Hematology, Augustenburger Platz 1, Berlin 13353, Germany; German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany; Freie Universität Berlin, Kaiserswerther Str. 16-18, Berlin 14195, Germany
| | - Konstantin Helmsauer
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Pediatric Oncology and Hematology, Augustenburger Platz 1, Berlin 13353, Germany; Neuroblastoma Research Group, Experimental and Clinical Research Center (ECRC) of the Charité and the Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Lindenberger Weg 80, Berlin 13125, Germany
| | - Silke Schwiebert
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Pediatric Oncology and Hematology, Augustenburger Platz 1, Berlin 13353, Germany
| | - Anika Klaus
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Pediatric Oncology and Hematology, Augustenburger Platz 1, Berlin 13353, Germany
| | - Anton G Henssen
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Pediatric Oncology and Hematology, Augustenburger Platz 1, Berlin 13353, Germany; Neuroblastoma Research Group, Experimental and Clinical Research Center (ECRC) of the Charité and the Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Lindenberger Weg 80, Berlin 13125, Germany
| | - Teresa Straka
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Pediatric Oncology and Hematology, Augustenburger Platz 1, Berlin 13353, Germany
| | - Marco Lodrini
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Pediatric Oncology and Hematology, Augustenburger Platz 1, Berlin 13353, Germany
| | - Sebastian G Wicha
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, Bundesstrasse 45, Hamburg 20146, Germany
| | - Steffen Fuchs
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Pediatric Oncology and Hematology, Augustenburger Platz 1, Berlin 13353, Germany; German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany; German Cancer Consortium (DKTK), Partner Site Berlin, Virchowweg 23, Berlin 10117, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Anna-Louisa-Karsch-Strasse 2, Berlin 10178, Germany
| | - Falk Hertwig
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Pediatric Oncology and Hematology, Augustenburger Platz 1, Berlin 13353, Germany
| | - Frank Westermann
- German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Alice Vitali
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Pediatric Oncology and Hematology, Augustenburger Platz 1, Berlin 13353, Germany
| | - Carlotta Caramel
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Pediatric Oncology and Hematology, Augustenburger Platz 1, Berlin 13353, Germany
| | - Gabriele Büchel
- Department of Biochemistry and Molecular Biology, Theodor Boveri Institute, Biocenter, University of Würzburg, Am Hubland, Würzburg 97074, Germany; Mildred Scheel Early Career Center, University Hospital Würzburg, Josef-Schneider-Str. 6, Würzburg 97080, Germany
| | - Martin Eilers
- Department of Biochemistry and Molecular Biology, Theodor Boveri Institute, Biocenter, University of Würzburg, Am Hubland, Würzburg 97074, Germany
| | - Kathy Astrahantseff
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Pediatric Oncology and Hematology, Augustenburger Platz 1, Berlin 13353, Germany
| | - Angelika Eggert
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Pediatric Oncology and Hematology, Augustenburger Platz 1, Berlin 13353, Germany; German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany; German Cancer Consortium (DKTK), Partner Site Berlin, Virchowweg 23, Berlin 10117, Germany
| | - Uta E Höpken
- Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Robert-Rössle Str. 10, Berlin 13125, Germany
| | - Johannes H Schulte
- Universitätsklinik für Kinder, und Jugendmedizin, Department of Pediatric Hematology and Oncology, Hoppe-Seyler-Straße 1, Tübingen 72076, Germany
| | - Thomas Blankenstein
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Pediatric Oncology and Hematology, Augustenburger Platz 1, Berlin 13353, Germany; Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Robert-Rössle Str. 10, Berlin 13125, Germany
| | - Kathleen Anders
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Pediatric Oncology and Hematology, Augustenburger Platz 1, Berlin 13353, Germany
| | - Annette Künkele
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Pediatric Oncology and Hematology, Augustenburger Platz 1, Berlin 13353, Germany; German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany; German Cancer Consortium (DKTK), Partner Site Berlin, Virchowweg 23, Berlin 10117, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Anna-Louisa-Karsch-Strasse 2, Berlin 10178, Germany.
| |
Collapse
|
14
|
Tregub PP, Komleva YK, Kukla MV, Averchuk AS, Vetchinova AS, Rozanova NA, Illarioshkin SN, Salmina AB. Brain Plasticity and Cell Competition: Immediate Early Genes Are the Focus. Cells 2025; 14:143. [PMID: 39851571 PMCID: PMC11763428 DOI: 10.3390/cells14020143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 01/13/2025] [Accepted: 01/17/2025] [Indexed: 01/26/2025] Open
Abstract
Brain plasticity is at the basis of many cognitive functions, including learning and memory. It includes several mechanisms of synaptic and extrasynaptic changes, neurogenesis, and the formation and elimination of synapses. The plasticity of synaptic transmission involves the expression of immediate early genes (IEGs) that regulate neuronal activity, thereby supporting learning and memory. In addition, IEGs are involved in the regulation of brain cells' metabolism, proliferation, and survival, in the establishment of multicellular ensembles, and, presumably, in cell competition in the tissue. In this review, we analyze the current understanding of the role of IEGs (c-Fos, c-Myc, Arg3.1/Arc) in controlling brain plasticity in physiological and pathological conditions, including brain aging and neurodegeneration. This work might inspire new gene therapy strategies targeting IEGs to regulate synaptic plasticity, and potentially prevent or mitigate neurodegenerative diseases.
Collapse
Affiliation(s)
- Pavel P. Tregub
- Research Center of Neurology, 125367 Moscow, Russia
- I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | | | | | | | - Anna S. Vetchinova
- I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | | | | | | |
Collapse
|
15
|
Yang C, Pang X, Teng S, Wilson S, Gu X, Xie G. MYC Overexpression Enhances Sensitivity to MEK Inhibition in Head and Neck Squamous Cell Carcinoma. Int J Mol Sci 2025; 26:588. [PMID: 39859304 PMCID: PMC11766173 DOI: 10.3390/ijms26020588] [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: 12/09/2024] [Revised: 01/05/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
MEK inhibitors, such as trametinib, have shown therapeutic potential in head and neck squamous cell carcinoma (HNSCC). However, the factors influencing cancer cell sensitivity and resistance to MEK inhibition remain poorly understood. In our study, we observed that MEK inhibition significantly reduced the expression of MYC, a transcription factor critical for the therapeutic response. MYC overexpression markedly enhanced the sensitivity of HNSCC cells to trametinib, as evidenced by delayed wound healing and reduced colony formation. Cell cycle analysis revealed that trametinib induced a G1 phase arrest, whereas MYC overexpression accelerated cell cycle progression, with a reduced induction of p27 and p21 and diminished decreases in E2F1 and phospho-Ser2/5 levels. Flow cytometry and protein analyses demonstrated that MYC overexpression amplified trametinib-induced apoptosis and DNA damage, as evidenced by elevated levels of pro-apoptotic markers (p53, cleaved PARP, and BIM) and γH2AX. In vivo xenograft models confirmed these findings, showing increased sensitivity to trametinib in MYC-overexpressing tumors. Moreover, MEK inhibition increased autophagy in HNSCC cells, a factor critical for therapeutic resistance. Inhibiting trametinib-induced autophagy further enhanced apoptotic cell death. These findings suggest that MYC expression and autophagy play crucial roles in HNSCC's response to MEK inhibition. Combining trametinib with autophagy inhibition may improve therapeutic outcomes in HNSCC.
Collapse
Affiliation(s)
- Cuicui Yang
- Department of Oral Pathology, Howard University, 600 W Street NW, Washington, DC 20059, USA; (C.Y.); (X.P.); (S.W.); (X.G.)
- Cancer Center, Howard University, 2041 Georgia Avenue NW, Washington, DC 20059, USA
| | - Xiaowu Pang
- Department of Oral Pathology, Howard University, 600 W Street NW, Washington, DC 20059, USA; (C.Y.); (X.P.); (S.W.); (X.G.)
| | - Shaolei Teng
- Department of Biology, Howard University, 415 College St. NW, Washington, DC 20059, USA;
| | - Shamel Wilson
- Department of Oral Pathology, Howard University, 600 W Street NW, Washington, DC 20059, USA; (C.Y.); (X.P.); (S.W.); (X.G.)
| | - Xinbin Gu
- Department of Oral Pathology, Howard University, 600 W Street NW, Washington, DC 20059, USA; (C.Y.); (X.P.); (S.W.); (X.G.)
- Cancer Center, Howard University, 2041 Georgia Avenue NW, Washington, DC 20059, USA
| | - Guiqin Xie
- Department of Oral Pathology, Howard University, 600 W Street NW, Washington, DC 20059, USA; (C.Y.); (X.P.); (S.W.); (X.G.)
- Cancer Center, Howard University, 2041 Georgia Avenue NW, Washington, DC 20059, USA
| |
Collapse
|
16
|
Solares S, León J, García-Gutiérrez L. The Functional Interaction Between Epstein-Barr Virus and MYC in the Pathogenesis of Burkitt Lymphoma. Cancers (Basel) 2024; 16:4212. [PMID: 39766110 PMCID: PMC11674381 DOI: 10.3390/cancers16244212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/13/2024] [Accepted: 12/14/2024] [Indexed: 01/11/2025] Open
Abstract
The Epstein-Barr virus (EBV) is associated with a wide range of diseases, malignant and non-malignant. EBV was, in fact, the first virus described with cell transformation capacity, discovered by Epstein in 1964 in lymphoma samples from African children. Since then, EBV has been associated with several human tumors including nasopharyngeal carcinoma, gastric carcinoma, T-cell lymphoma, Hodgkin lymphoma, diffuse large B cell lymphoma, and Burkitt lymphoma among others. The molecular hallmark of Burkitt lymphoma (BL) is a chromosomal translocation that involves the MYC gene and immunoglobulin loci, resulting in the deregulated expression of MYC, an oncogenic transcription factor that appears deregulated in about half of human tumors. The role of MYC in lymphoma is well established, as MYC overexpression drives B cell proliferation through multiple mechanisms, foremost, the stimulation of the cell cycle. Indeed, MYC is found overexpressed or deregulated in several non-Hodgkin lymphomas. Most endemic and many sporadic BLs are associated with EBV infection. While some mechanisms by which EBV can contribute to BL have been reported, the mechanism that links MYC translocation and EBV infection in BL is still under debate. Here, we review the main EBV-associated diseases, with a special focus on BL, and we discuss the interaction of EBV and MYC translocation during B cell malignant transformation in BL.
Collapse
Affiliation(s)
| | | | - Lucía García-Gutiérrez
- Instituto de Biomedicina y Biotecnología de Cantabria, Departamento de Biología Molecular, Universidad de Cantabria-CSIC, Albert Einstein 22, 39011 Cantabria, Spain; (S.S.); (J.L.)
| |
Collapse
|
17
|
Ai M, Ma H, He J, Xu F, Ming Y, Ye Z, Zheng Q, Luo D, Yang K, Li J, Nie C, Pu W, Peng Y. Targeting oncogenic transcriptional factor c-myc by oligonucleotide PROTAC for the treatment of hepatocellular carcinoma. Eur J Med Chem 2024; 280:116978. [PMID: 39447458 DOI: 10.1016/j.ejmech.2024.116978] [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: 09/02/2024] [Revised: 10/08/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024]
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related death, but effective therapeutic strategies are limited. Transcriptional factor c-Myc plays an oncogenic role in tumorigenesis and is an attractive target for HCC treatment. However, targeted therapy against c-Myc remains challenging. Herein, by conjugating VH032 with an optimized DNA sequence that recognized c-Myc complex, we discovered oligonucleotide-based proteolysis targeting chimeras (PROTACs), termed as MP-16 and MP-17, which effectively induced degradation of c-Myc. Mechanically, MP-16 or MP-17 directly interacted with c-Myc complex to form VHL/PROTAC/c-Myc ternary complex, and triggered c-Myc degradation by recruiting ubiquitin-proteasome system, suppressing cell proliferation of HCC cells. In mice model, MP-16 or MP-17 significantly inhibited HCC tumor growth and exhibited promising drug safety. This work provided novel oligonucleotide PROTACs that degraded c-Myc, giving a new lead structure for HCC therapy.
Collapse
Affiliation(s)
- Min Ai
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610064, China
| | - Hulin Ma
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610064, China
| | - Jianhua He
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610064, China
| | - Fuyan Xu
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610064, China
| | - Yue Ming
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610064, China
| | - Zixia Ye
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610064, China
| | - Qingquan Zheng
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610064, China
| | - Dongdong Luo
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610064, China
| | - Kaichuan Yang
- Engineering Research Center for Pharmaceuticals and Equipments of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610052, China
| | - Jiao Li
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610064, China
| | - Chunlai Nie
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610064, China.
| | - Wenchen Pu
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610064, China.
| | - Yong Peng
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610064, China; Frontier Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, China.
| |
Collapse
|
18
|
Willemsen M, Bulgarelli J, Chauhan S, Lereim R, Angeli D, Grisendi G, Krebbers G, Davidson I, Kyte J, Guidoboni M, Luiten R, Bakker W. Changes in AXL and/or MITF melanoma subpopulations in patients receiving immunotherapy. IMMUNO-ONCOLOGY TECHNOLOGY 2024; 24:101009. [PMID: 39697983 PMCID: PMC11652950 DOI: 10.1016/j.iotech.2024.101009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
Background Tumor heterogeneity is a hurdle to effective therapy, as illustrated by the 'mixed responses' frequently seen in immunotherapy-treated patients. Previously, AXL+ tumor cells were identified to be highly resistant to targeted therapy, whereas more differentiated MITF+ tumor cells do respond to RAF and MEK inhibitors. Patients and methods In this study, we analyzed tumor heterogeneity and explored the presence of the previously described AXL+ or MITF+ melanoma subpopulations in metastatic tissues by NanoString gene expression analysis, single-cell RNA sequencing and in situ multiplex immunofluorescence. Furthermore, we analyzed how these subpopulations correlate with immunological pressure and response to immunotherapy by immunomodulating antibodies or autologous tumor lysate-loaded dendritic cell vaccination. Results Our data demonstrate large interpatient variability and variable therapy-induced changes independent of the type of therapy. We identify the presence of previously described AXL+ and MITF+ subpopulations in metastatic tissues both at the mRNA level and in situ at the protein level, and demonstrate that MITF+ melanoma cells are significantly decreased upon immunotherapy, while AXL+ melanoma cell numbers are stable. MITF+ tumor cells showed the most significant inverse correlation with CD8+ T cells. Our patient cohort also shows that immunotherapy-induced changes in the abundance of AXL+ or MITF+ tumor cells did not correlate with improved survival. Conclusions Overall, this study suggests that more differentiated MITF+ tumors are efficiently targeted by immunotherapy, while AXL+ tumor cells may be more resistant, analogous to their response to targeted therapy.
Collapse
Affiliation(s)
- M. Willemsen
- Department of Dermatology and Netherlands Institute for Pigment Disorders, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - J. Bulgarelli
- Immunotherapy Cell Therapy and Biobank (ITCB) Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - S.K. Chauhan
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - R.R. Lereim
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - D. Angeli
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - G. Grisendi
- Laboratory of Cellular Therapy, Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - G. Krebbers
- Department of Dermatology and Netherlands Institute for Pigment Disorders, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - I. Davidson
- Department of Functional Genomics and Cancer, IGBMC, CNRS/INSERM, Illkirch, France
| | - J.A. Kyte
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Department of Clinical Cancer Research, Oslo University Hospital, Oslo, Norway
| | - M. Guidoboni
- Department of Oncology, Ferrara University Hospital, University of Ferrara, Ferrara, Italy
| | - R.M. Luiten
- Department of Dermatology and Netherlands Institute for Pigment Disorders, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - W.J. Bakker
- Department of Dermatology and Netherlands Institute for Pigment Disorders, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| |
Collapse
|
19
|
Knight A, Houser J, Otasevic T, Juran V, Vybihal V, Smrcka M, Piskacek M. Myc 9aaTAD activation domain binds to mediator of transcription with superior high affinity. Mol Med 2024; 30:211. [PMID: 39538178 PMCID: PMC11558822 DOI: 10.1186/s10020-024-00896-7] [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: 04/18/2024] [Accepted: 08/12/2024] [Indexed: 11/16/2024] Open
Abstract
The overexpression of MYC genes is frequently found in many human cancers, including adult and pediatric malignant brain tumors. Targeting MYC genes continues to be challenging due to their undruggable nature. Using our prediction algorithm, the nine-amino-acid activation domain (9aaTAD) has been identified in all four Yamanaka factors, including c-Myc. The predicted activation function was experimentally demonstrated for all these short peptides in transactivation assay. We generated a set of c-Myc constructs (1-108, 69-108 and 98-108) in the N-terminal regions and tested their ability to initiate transcription in one hybrid assay. The presence and absence of 9aaTAD (region 100-108) in the constructs strongly correlated with their activation functions (5-, 3- and 67-times respectively). Surprisingly, we observed co-activation function of the myc region 69-103, called here acetyl-TAD, previously described by Faiola et al. (Mol Cell Biol 25:10220-10234, 2005) and characterized in this study as a new domain collaborating with the 9aaTAD. We discovered strong interactions on a nanomolar scale between the Myc-9aaTAD activation domains and the KIX domain of CBP coactivator. We showed conservation of the 9aaTADs in the MYC family. In summary for the c-Myc oncogene, the acetyl-TAD and the 9aaTAD domains jointly mediated activation function. The c-Myc protein is largely intrinsically disordered and therefore difficult to target with small-molecule inhibitors. For the c-Myc driven tumors, the strong c-Myc interaction with the KIX domain represents a promising druggable target.
Collapse
Affiliation(s)
- Andrea Knight
- School of Life Science, Faculty of Science and Engineering, Anglia Ruskin University, East Road, Cambridge, CB1 1PT, UK.
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University Brno, Masaryk University, Kamenice 753/5, Brno, 625 00, Czech Republic.
- Department of Neurosurgery, University Hospital Brno, and Faculty of Medicine, Masaryk University, Brno, Czech Republic.
| | - Josef Houser
- Central European Institute of Technology (CEITEC), Masaryk University Brno, Brno, Czech Republic
- National Centre for Biomolecular Research (NCBR), Faculty of Science, Masaryk University Brno, Brno, Czech Republic
- Core Facility Biomolecular Interactions and Crystallization (CF BIC), Masaryk University Brno, Brno, Czech Republic
| | - Tomas Otasevic
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University Brno, Masaryk University, Kamenice 753/5, Brno, 625 00, Czech Republic
| | - Vilem Juran
- Department of Neurosurgery, University Hospital Brno, and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Vaclav Vybihal
- Department of Neurosurgery, University Hospital Brno, and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Martin Smrcka
- Department of Neurosurgery, University Hospital Brno, and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Martin Piskacek
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University Brno, Masaryk University, Kamenice 753/5, Brno, 625 00, Czech Republic.
| |
Collapse
|
20
|
Guerrieri AN, Hattinger CM, Marchesini F, Melloni M, Serra M, Ibrahim T, Penzo M. The Interplay Between the MYC Oncogene and Ribosomal Proteins in Osteosarcoma Onset and Progression: Potential Mechanisms and Indication of Candidate Therapeutic Targets. Int J Mol Sci 2024; 25:12031. [PMID: 39596100 PMCID: PMC11593864 DOI: 10.3390/ijms252212031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/05/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
High-grade osteosarcoma (OS) is the most common primary bone tumor mainly affecting children and young adults. First-line treatment consists of neo-adjuvant chemotherapy with doxorubicin, cisplatin, and methotrexate and surgery. The mean long-term survival rate for localized disease at diagnosis is 65-70%, dropping down to 20% when metastases are present at diagnosis. Therefore, curing OS is a clinical challenge, particularly for patients that do not respond to standard treatments. MYC has frequently been reported to be involved in the pathogenesis of OS and its high expression may be associated with drug resistance and patients' worse prognosis. Moreover, MYC is a master regulator of ribosomal proteins (RPs) synthesis and ribosome biogenesis (RiBi), which is often up-regulated in human tumors. In recent years, RPs have been recognized not only for their traditional role in ribosome assembly but also for their extra-ribosomal functions, many of which are linked to the onset and progression of cancer. In this review we focus on the role and possible interplay of MYC and RPs expression in association with drug resistance and worse prognosis in OS and discuss therapeutic options that target de-regulated MYC, RiBi, or RPs, which are already clinically available or under evaluation in clinical trials.
Collapse
Affiliation(s)
- Ania Naila Guerrieri
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (A.N.G.); (M.S.); (T.I.)
| | - Claudia Maria Hattinger
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (A.N.G.); (M.S.); (T.I.)
| | - Federica Marchesini
- Center for Applied Biomedical Research (CRBA), Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy; (F.M.); (M.M.)
| | - Martina Melloni
- Center for Applied Biomedical Research (CRBA), Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy; (F.M.); (M.M.)
| | - Massimo Serra
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (A.N.G.); (M.S.); (T.I.)
| | - Toni Ibrahim
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (A.N.G.); (M.S.); (T.I.)
| | - Marianna Penzo
- Center for Applied Biomedical Research (CRBA), Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy; (F.M.); (M.M.)
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| |
Collapse
|
21
|
Tuo Y, Ye Y. Sterile Alpha Motif Domain-Containing 5 Suppresses Malignant Phenotypes and Tumor Growth in Breast Cancer: Regulation of Polo-Like Kinase 1 and c-Myc Signaling in a Xenograft Model. Cureus 2024; 16:e73259. [PMID: 39524172 PMCID: PMC11550111 DOI: 10.7759/cureus.73259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2024] [Indexed: 11/16/2024] Open
Abstract
Background Breast cancer, particularly the triple-negative breast cancer (TNBC) subtype, remains a significant clinical challenge due to its resistance to standard chemotherapy and high recurrence rate. In this study, we explored the role of Sterile Alpha Motif Domain-Containing 5 (SAMD5) as a potential regulatory partner with the c-Myc oncogenic signaling pathway in breast cancer. Materials and methods Functional assays were conducted to investigate the effects of SAMD5 overexpression on cell viability, colony formation, and invasive behavior in TNBC cell lines. This study further assessed the expression levels of proliferation and invasion markers, including Ki67 (a marker for cell proliferation), Matrix Metalloproteinase-2 (MMP2), and Matrix Metalloproteinase-9 (MMP9). Mechanistic analyses identified a negative correlation between SAMD5 and Polo-like Kinase 1 (PLK1), a gene frequently overexpressed in breast cancer, particularly in TNBC. The effects of PLK1 knockdown on cell viability, colony formation, and invasion were observed, along with the impact of PLK1 overexpression on SAMD5's inhibitory activity. In vivo studies were performed using a xenograft tumor model in nude mice to evaluate the impact of SAMD5 overexpression on tumor weight and volume. Results SAMD5 overexpression significantly reduced cell viability, colony formation, and invasion in TNBC cells, and downregulated key proteins in the c-Myc signaling pathway, including c-Myc itself, β-catenin, Cyclin-Dependent Kinase 4 (CDK4), Cyclin-Dependent Kinase 6 (CDK6), and Cyclin D1. PLK1 overexpression was found to counteract SAMD5's inhibitory effects. In vivo experiments demonstrated that SAMD5 overexpression led to a marked reduction in tumor weight and volume, effects that were partially reversed by PLK1 overexpression. Conclusions SAMD5 acts as a tumor suppressor in breast cancer, particularly in TNBC, by inhibiting critical cellular processes and downregulating the c-Myc signaling pathway. This effect appears to be mediated, in part, through its negative association with PLK1. Targeting the SAMD5/PLK1 axis offers a promising therapeutic strategy for addressing aggressive breast cancers.
Collapse
Affiliation(s)
- YouLin Tuo
- Department of Breast Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, CHN
| | - YiFeng Ye
- Department of Breast Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, CHN
| |
Collapse
|
22
|
Zhai W, Yang W, Ge J, Xiao X, Wu K, She K, Zhou Y, Kong Y, Wu L, Luo S, Pu X. ADAMTS4 exacerbates lung cancer progression via regulating c-Myc protein stability and activating MAPK signaling pathway. Biol Direct 2024; 19:94. [PMID: 39415271 PMCID: PMC11483991 DOI: 10.1186/s13062-024-00512-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: 05/31/2023] [Accepted: 08/08/2024] [Indexed: 10/18/2024] Open
Abstract
BACKGROUND Lung cancer is one of the most frequent cancers and the leading cause of cancer-related deaths worldwide with poor prognosis. A disintegrin and metalloproteinase with thrombospondin motifs 4 (ADAMTS4) is crucial in the regulation of the extracellular matrix (ECM), impacting its formation, homeostasis and remodeling, and has been linked to cancer progression. However, the specific involvement of ADAMTS4 in the development of lung cancer remains unclear. METHODS ADAMTS4 expression was identified in human lung cancer samples by immunohistochemical (IHC) staining and the correlation of ADAMTS4 with clinical outcome was determined. The functional impact of ADAMTS4 on malignant phenotypes of lung cancer cells was explored both in vitro and in vivo. The mechanisms underlying ADAMTS4-mediated lung cancer progression were explored by ubiquitination-related assays. RESULTS Our study revealed a significant upregulation of ADAMTS4 at the protein level in lung cancer tissues compared to para-carcinoma normal tissues. High ADAMTS4 expression inversely correlated with the prognosis of lung cancer patients. Knockdown of ADAMTS4 inhibited the proliferation and migration of lung cancer cells, as well as the tubule formation of HUVECs, while ADAMTS4 overexpression exerted opposite effects. Mechanistically, we found that ADAMTS4 stabilized c-Myc by inhibiting its ubiquitination, thereby promoting the in vitro and in vivo growth of lung cancer cells and inducing HUVECs tubule formation in lung cancer. In addition, our results suggested that ADAMTS4 overexpression activated MAPK signaling pathway. CONCLUSIONS We highlighted the promoting role of ADAMTS4 in lung cancer progression and proposed ADAMTS4 as a promising therapeutic target for lung cancer patients.
Collapse
Affiliation(s)
- Wei Zhai
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Road, Wuhan, 430030, Hubei, China
| | - Wensheng Yang
- Department of Thoracic Surgery, The Affiliated Shaoyang Hospital, Hengyang Medical School, University of South China, No. 36, Hongqi Road, Daxiang District, Shaoyang, 422000, Hunan, China
| | - Jing Ge
- Department of Geriatrics and Institute of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277, Jiefang Road, Wuhan, 430030, Hubei, China
| | - Xuelian Xiao
- Department of Medical Administration, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, No. 283 Tongzipo Road, Yuelu District, Changsha, 410013, Hunan, China
| | - Kang Wu
- Sansure Biotech Inc.,, No. 680, Lusong Road, Yuelu District, Changsha, 410205, Hunan, China
| | - Kelin She
- Department of Thoracic Surgery, Hunan Provincial Pecople's Hospital, The First Affiliated Hospital of Huan Nomal University, No. 61, Jiefang West Road, Furong District, Changsha, 410013, Hunan, China
| | - Yu Zhou
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, No. 283, Tongzipo Road, Yuelu District, Changsha, 410013, Hunan, China
| | - Yi Kong
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, No. 283, Tongzipo Road, Yuelu District, Changsha, 410013, Hunan, China
| | - Lin Wu
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, No. 283, Tongzipo Road, Yuelu District, Changsha, 410013, Hunan, China
| | - Shiya Luo
- Sansure Biotech Inc.,, No. 680, Lusong Road, Yuelu District, Changsha, 410205, Hunan, China
| | - Xingxiang Pu
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, No. 283, Tongzipo Road, Yuelu District, Changsha, 410013, Hunan, China.
| |
Collapse
|
23
|
Seabrook LJ, Franco CN, Loy CA, Osman J, Fredlender C, Zimak J, Campos M, Nguyen ST, Watson RL, Levine SR, Khalil MF, Sumigray K, Trader DJ, Albrecht LV. Methylarginine targeting chimeras for lysosomal degradation of intracellular proteins. Nat Chem Biol 2024:10.1038/s41589-024-01741-y. [PMID: 39414979 DOI: 10.1038/s41589-024-01741-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 09/05/2024] [Indexed: 10/18/2024]
Abstract
A paradigm shift in drug development is the discovery of small molecules that harness the ubiquitin-proteasomal pathway to eliminate pathogenic proteins. Here we provide a modality for targeted protein degradation in lysosomes. We exploit an endogenous lysosomal pathway whereby protein arginine methyltransferases (PRMTs) initiate substrate degradation via arginine methylation. We developed a heterobifunctional small molecule, methylarginine targeting chimera (MrTAC), that recruits PRMT1 to a target protein for induced degradation in lysosomes. MrTAC compounds degraded substrates across cell lines, timescales and doses. MrTAC degradation required target protein methylation for subsequent lysosomal delivery via microautophagy. A library of MrTAC molecules exemplified the generality of MrTAC to degrade known targets and neo-substrates-glycogen synthase kinase 3β, MYC, bromodomain-containing protein 4 and histone deacetylase 6. MrTAC selectively degraded target proteins and drove biological loss-of-function phenotypes in survival, transcription and proliferation. Collectively, MrTAC demonstrates the utility of endogenous lysosomal proteolysis in the generation of a new class of small molecule degraders.
Collapse
Affiliation(s)
- Laurence J Seabrook
- Department of Developmental & Cell Biology, School of Biological Sciences, University of California, Irvine, Irvine, CA, USA
| | - Carolina N Franco
- Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA
| | - Cody A Loy
- Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA
| | - Jaida Osman
- Department of Chemistry, School of Physical Sciences, University of California, Irvine, Irvine, CA, USA
| | - Callie Fredlender
- Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA
| | - Jan Zimak
- Center for Neurotherapeutics, University of California, Irvine, Irvine, CA, USA
| | - Melissa Campos
- Department of Developmental & Cell Biology, School of Biological Sciences, University of California, Irvine, Irvine, CA, USA
| | - Steven T Nguyen
- Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA
| | - Richard L Watson
- Department of Medicine, Division of Pulmonary & Critical Care, University of California, Los Angeles, Los Angeles, CA, USA
| | - Samantha R Levine
- Center for Neurotherapeutics, University of California, Irvine, Irvine, CA, USA
| | - Marian F Khalil
- Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA
| | - Kaelyn Sumigray
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Darci J Trader
- Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA
- Department of Chemistry, School of Physical Sciences, University of California, Irvine, Irvine, CA, USA
| | - Lauren V Albrecht
- Department of Developmental & Cell Biology, School of Biological Sciences, University of California, Irvine, Irvine, CA, USA.
- Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
24
|
Straube J, Janardhanan Y, Haldar R, Bywater MJ. Immune control in acute myeloid leukemia. Exp Hematol 2024; 138:104256. [PMID: 38876254 DOI: 10.1016/j.exphem.2024.104256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/09/2024] [Accepted: 06/10/2024] [Indexed: 06/16/2024]
Abstract
Acute myeloid leukemia (AML) is a genetically heterogeneous disease, in that a multitude of oncogenic drivers and chromosomal abnormalities have been identified and associated with the leukemic transformation of myeloid blasts. However, little is known as to how individual mutations influence the interaction between the immune system and AML cells and the efficacy of the immune system in AML disease control. In this review, we will discuss how AML cells potentially activate the immune system and what evidence there is to support the role of the immune system in controlling this disease. We will specifically examine the importance of antigen presentation in fostering an effective anti-AML immune response, explore the disruption of immune responses during AML disease progression, and discuss the emerging role of the oncoprotein MYC in driving immune suppression in AML.
Collapse
Affiliation(s)
- Jasmin Straube
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia; The University of Queensland, Brisbane, Queensland, Australia
| | | | - Rohit Haldar
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Megan J Bywater
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia; The University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
25
|
Mulè P, Fernandez-Perez D, Amato S, Manganaro D, Oldani P, Brandini S, Diaferia G, Cuomo A, Recordati C, Soriani C, Dondi A, Zanotti M, Rustichelli S, Bisso A, Pece S, Rodighiero S, Natoli G, Amati B, Ferrari KJ, Chiacchiera F, Pasini D. WNT Oncogenic Transcription Requires MYC Suppression of Lysosomal Activity and EPCAM Stabilization in Gastric Tumors. Gastroenterology 2024; 167:903-918. [PMID: 38971196 DOI: 10.1053/j.gastro.2024.06.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 07/08/2024]
Abstract
BACKGROUND & AIMS WNT signaling is central to spatial tissue arrangement and regulating stem cell activity, and it represents the hallmark of gastrointestinal cancers. Although its role in driving intestinal tumors is well characterized, WNT's role in gastric tumorigenesis remains elusive. METHODS We have developed mouse models to control the specific expression of an oncogenic form of β-catenin (CTNNB1) in combination with MYC activation in Lgr5+ cells of the gastric antrum. We used multiomics approaches applied in vivo and in organoid models to characterize their cooperation in driving gastric tumorigenesis. RESULTS We report that constitutive β-catenin stabilization in the stomach has negligible oncogenic effects and requires MYC activation to induce gastric tumor formation. Although physiologically low MYC levels in gastric glands limit β-catenin transcriptional activity, increased MYC expression unleashes the WNT oncogenic transcriptional program, promoting β-catenin enhancer invasion without a direct transcriptional cooperation. MYC activation induces a metabolic rewiring that suppresses lysosomal biogenesis through mTOR and ERK activation and MiT/TFE inhibition. This prevents EPCAM degradation by macropinocytosis, promoting β-catenin chromatin accumulation and activation of WNT oncogenic transcription. CONCLUSION Our results uncovered a new signaling framework with important implications for the control of gastric epithelial architecture and WNT-dependent oncogenic transformation.
Collapse
Affiliation(s)
- Patrizia Mulè
- European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Department of Experimental Oncology, Milan, Italy
| | - Daniel Fernandez-Perez
- European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Department of Experimental Oncology, Milan, Italy
| | - Simona Amato
- European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Department of Experimental Oncology, Milan, Italy
| | - Daria Manganaro
- European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Department of Experimental Oncology, Milan, Italy
| | - Paola Oldani
- European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Department of Experimental Oncology, Milan, Italy
| | - Stefania Brandini
- European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Department of Experimental Oncology, Milan, Italy
| | - Giuseppe Diaferia
- European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Department of Experimental Oncology, Milan, Italy
| | - Alessandro Cuomo
- European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Department of Experimental Oncology, Milan, Italy
| | | | - Chiara Soriani
- European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Department of Experimental Oncology, Milan, Italy
| | - Ambra Dondi
- European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Department of Experimental Oncology, Milan, Italy
| | - Marika Zanotti
- European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Department of Experimental Oncology, Milan, Italy
| | - Samantha Rustichelli
- European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Department of Experimental Oncology, Milan, Italy
| | - Andrea Bisso
- European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Department of Experimental Oncology, Milan, Italy
| | - Salvatore Pece
- European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Department of Experimental Oncology, Milan, Italy
| | - Simona Rodighiero
- European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Department of Experimental Oncology, Milan, Italy
| | - Gioacchino Natoli
- European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Department of Experimental Oncology, Milan, Italy
| | - Bruno Amati
- European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Department of Experimental Oncology, Milan, Italy
| | - Karin Johanna Ferrari
- European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Department of Experimental Oncology, Milan, Italy
| | - Fulvio Chiacchiera
- University of Trento, Department of Cellular, Computational and Integrative Biology, Trento, Italy
| | - Diego Pasini
- European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Department of Experimental Oncology, Milan, Italy; University of Milan, Department of Health Sciences, Milan, Italy.
| |
Collapse
|
26
|
Biffo S, Ruggero D, Santoro MM. The crosstalk between metabolism and translation. Cell Metab 2024; 36:1945-1962. [PMID: 39232280 PMCID: PMC11586076 DOI: 10.1016/j.cmet.2024.07.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/24/2024] [Accepted: 07/31/2024] [Indexed: 09/06/2024]
Abstract
Metabolism and mRNA translation represent critical steps involved in modulating gene expression and cellular physiology. Being the most energy-consuming process in the cell, mRNA translation is strictly linked to cellular metabolism and in synchrony with it. Indeed, several mRNAs for metabolic pathways are regulated at the translational level, resulting in translation being a coordinator of metabolism. On the other hand, there is a growing appreciation for how metabolism impacts several aspects of RNA biology. For example, metabolic pathways and metabolites directly control the selectivity and efficiency of the translational machinery, as well as post-transcriptional modifications of RNA to fine-tune protein synthesis. Consistently, alterations in the intricate interplay between translational control and cellular metabolism have emerged as a critical axis underlying human diseases. A better understanding of such events will foresee innovative therapeutic strategies in human disease states.
Collapse
Affiliation(s)
- Stefano Biffo
- National Institute of Molecular Genetics and Biosciences Department, University of Milan, Milan, Italy.
| | - Davide Ruggero
- Department of Cellular and Molecular Pharmacology, Department of Urology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA.
| | - Massimo Mattia Santoro
- Laboratory of Angiogenesis and Cancer Metabolism, Department of Biology, University of Padua, Padua, Italy.
| |
Collapse
|
27
|
Wang X, Zhang L, Si H. Combining luteolin and curcumin synergistically suppresses triple-negative breast cancer by regulating IFN and TGF-β signaling pathways. Biomed Pharmacother 2024; 178:117221. [PMID: 39111078 DOI: 10.1016/j.biopha.2024.117221] [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: 02/22/2024] [Revised: 07/15/2024] [Accepted: 07/26/2024] [Indexed: 08/25/2024] Open
Abstract
Combining two or more chemicals in chemotherapy is rapidly increasing because of its higher efficacy, lower toxicity, lower dosages, and lower drug resistance. Here, we identified a novel combination of luteolin (LUT) and curcumin (CUR), two bioactive compounds from foods, synergistically suppressed triple-negative breast cancer (TNBC) cell proliferation (LUT 30 µM + CUR 20 µM), colony formation (LUT 1 µM + CUR 2 µM), and tumor growth in xenograft mice (LUT 10 mg/kg body weight/day + CUR 20 mg/kg body weight/day, i.p. injection every other day, 5 weeks), while the individual chemical alone did not show these inhibitory effects significantly at the selected concentrations/dosages. Our total RNA transcriptome analysis in xenograft tumors revealed that combining LUT and CUR synergistically activated type I interferon (IFN) signaling and suppressed transforming growth factor-beta (TGF-β) signaling pathways, which was further confirmed by the expression/activity of several proteins of the pathways in tumors. In addition, this combination of LUT and CUR also synergistically decreased oncoprotein levels of c-Myc and Notch1, the critical molecules required to maintain stem cell properties, tumor clonal evolution, and drug resistance. These results suggest that the combination of LUT and CUR synergistically inhibits TNBC by suppressing multiple cellular mechanisms, such as proliferation, colony formation, and transformation, as well as tumor migration, invasion, and metastasis, via regulating IFN and TGF-β signaling pathways. Therefore, combining LUT and CUR may be an effective therapeutic agent to treat highly aggressive, drug-resistant TNBC patients after clinical trials.
Collapse
Affiliation(s)
- Xiaoyong Wang
- Department of Food and Animal Sciences, Tennessee State University, Nashville, TN 37209, USA; Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Lijuan Zhang
- Department of Food and Animal Sciences, Tennessee State University, Nashville, TN 37209, USA
| | - Hongwei Si
- Department of Food and Animal Sciences, Tennessee State University, Nashville, TN 37209, USA.
| |
Collapse
|
28
|
Qi Y, Rezaeian AH, Wang J, Huang D, Chen H, Inuzuka H, Wei W. Molecular insights and clinical implications for the tumor suppressor role of SCF FBXW7 E3 ubiquitin ligase. Biochim Biophys Acta Rev Cancer 2024; 1879:189140. [PMID: 38909632 PMCID: PMC11390337 DOI: 10.1016/j.bbcan.2024.189140] [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: 04/12/2024] [Revised: 06/04/2024] [Accepted: 06/17/2024] [Indexed: 06/25/2024]
Abstract
FBXW7 is one of the most well-characterized F-box proteins, serving as substrate receptor subunit of SKP1-CUL1-F-box (SCF) E3 ligase complexes. SCFFBXW7 is responsible for the degradation of various oncogenic proteins such as cyclin E, c-MYC, c-JUN, NOTCH, and MCL1. Therefore, FBXW7 functions largely as a major tumor suppressor. In keeping with this notion, FBXW7 gene mutations or downregulations have been found and reported in many types of malignant tumors, such as endometrial, colorectal, lung, and breast cancers, which facilitate the proliferation, invasion, migration, and drug resistance of cancer cells. Therefore, it is critical to review newly identified FBXW7 regulation and tumor suppressor function under physiological and pathological conditions to develop effective strategies for the treatment of FBXW7-altered cancers. Since a growing body of evidence has revealed the tumor-suppressive activity and role of FBXW7, here, we updated FBXW7 upstream and downstream signaling including FBXW7 ubiquitin substrates, the multi-level FBXW7 regulatory mechanisms, and dysregulation of FBXW7 in cancer, and discussed promising cancer therapies targeting FBXW7 regulators and downstream effectors, to provide a comprehensive picture of FBXW7 and facilitate the study in this field.
Collapse
Affiliation(s)
- Yihang Qi
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Abdol-Hossein Rezaeian
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Jingchao Wang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Daoyuan Huang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Hong Chen
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Hiroyuki Inuzuka
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
29
|
Vidal R, Leen E, Herold S, Müller M, Fleischhauer D, Schülein-Völk C, Papadopoulos D, Röschert I, Uhl L, Ade CP, Gallant P, Bayliss R, Eilers M, Büchel G. Association with TFIIIC limits MYCN localisation in hubs of active promoters and chromatin accumulation of non-phosphorylated RNA polymerase II. eLife 2024; 13:RP94407. [PMID: 39177021 PMCID: PMC11343564 DOI: 10.7554/elife.94407] [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] [Indexed: 08/24/2024] Open
Abstract
MYC family oncoproteins regulate the expression of a large number of genes and broadly stimulate elongation by RNA polymerase II (RNAPII). While the factors that control the chromatin association of MYC proteins are well understood, much less is known about how interacting proteins mediate MYC's effects on transcription. Here, we show that TFIIIC, an architectural protein complex that controls the three-dimensional chromatin organisation at its target sites, binds directly to the amino-terminal transcriptional regulatory domain of MYCN. Surprisingly, TFIIIC has no discernible role in MYCN-dependent gene expression and transcription elongation. Instead, MYCN and TFIIIC preferentially bind to promoters with paused RNAPII and globally limit the accumulation of non-phosphorylated RNAPII at promoters. Consistent with its ubiquitous role in transcription, MYCN broadly participates in hubs of active promoters. Depletion of TFIIIC further increases MYCN localisation to these hubs. This increase correlates with a failure of the nuclear exosome and BRCA1, both of which are involved in nascent RNA degradation, to localise to active promoters. Our data suggest that MYCN and TFIIIC exert an censoring function in early transcription that limits promoter accumulation of inactive RNAPII and facilitates promoter-proximal degradation of nascent RNA.
Collapse
Affiliation(s)
- Raphael Vidal
- Theodor Boveri Institute, Department of Biochemistry and Molecular Biology, Biocenter, University of WürzburgWürzburgGermany
- Comprehensive Cancer Center MainfrankenWürzburgGermany
| | - Eoin Leen
- Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of LeedsLeedsUnited Kingdom
| | - Steffi Herold
- Theodor Boveri Institute, Department of Biochemistry and Molecular Biology, Biocenter, University of WürzburgWürzburgGermany
| | - Mareike Müller
- Theodor Boveri Institute, Department of Biochemistry and Molecular Biology, Biocenter, University of WürzburgWürzburgGermany
- Mildred Scheel Early Career Center, University Hospital WürzburgWürzburgGermany
| | - Daniel Fleischhauer
- Theodor Boveri Institute, Department of Biochemistry and Molecular Biology, Biocenter, University of WürzburgWürzburgGermany
| | - Christina Schülein-Völk
- Theodor Boveri Institute, Core Unit High-Content Microscopy, Biocenter, University of WürzburgWürzburgGermany
| | - Dimitrios Papadopoulos
- Theodor Boveri Institute, Department of Biochemistry and Molecular Biology, Biocenter, University of WürzburgWürzburgGermany
- Mildred Scheel Early Career Center, University Hospital WürzburgWürzburgGermany
| | - Isabelle Röschert
- Theodor Boveri Institute, Department of Biochemistry and Molecular Biology, Biocenter, University of WürzburgWürzburgGermany
| | - Leonie Uhl
- Theodor Boveri Institute, Department of Biochemistry and Molecular Biology, Biocenter, University of WürzburgWürzburgGermany
| | - Carsten P Ade
- Theodor Boveri Institute, Department of Biochemistry and Molecular Biology, Biocenter, University of WürzburgWürzburgGermany
| | - Peter Gallant
- Theodor Boveri Institute, Department of Biochemistry and Molecular Biology, Biocenter, University of WürzburgWürzburgGermany
| | - Richard Bayliss
- Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of LeedsLeedsUnited Kingdom
| | - Martin Eilers
- Theodor Boveri Institute, Department of Biochemistry and Molecular Biology, Biocenter, University of WürzburgWürzburgGermany
- Comprehensive Cancer Center MainfrankenWürzburgGermany
| | - Gabriele Büchel
- Theodor Boveri Institute, Department of Biochemistry and Molecular Biology, Biocenter, University of WürzburgWürzburgGermany
- Comprehensive Cancer Center MainfrankenWürzburgGermany
- Mildred Scheel Early Career Center, University Hospital WürzburgWürzburgGermany
| |
Collapse
|
30
|
Casacuberta-Serra S, González-Larreategui Í, Capitán-Leo D, Soucek L. MYC and KRAS cooperation: from historical challenges to therapeutic opportunities in cancer. Signal Transduct Target Ther 2024; 9:205. [PMID: 39164274 PMCID: PMC11336233 DOI: 10.1038/s41392-024-01907-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: 01/12/2024] [Revised: 06/05/2024] [Accepted: 06/24/2024] [Indexed: 08/22/2024] Open
Abstract
RAS and MYC rank amongst the most commonly altered oncogenes in cancer, with RAS being the most frequently mutated and MYC the most amplified. The cooperative interplay between RAS and MYC constitutes a complex and multifaceted phenomenon, profoundly influencing tumor development. Together and individually, these two oncogenes regulate most, if not all, hallmarks of cancer, including cell death escape, replicative immortality, tumor-associated angiogenesis, cell invasion and metastasis, metabolic adaptation, and immune evasion. Due to their frequent alteration and role in tumorigenesis, MYC and RAS emerge as highly appealing targets in cancer therapy. However, due to their complex nature, both oncogenes have been long considered "undruggable" and, until recently, no drugs directly targeting them had reached the clinic. This review aims to shed light on their complex partnership, with special attention to their active collaboration in fostering an immunosuppressive milieu and driving immunotherapeutic resistance in cancer. Within this review, we also present an update on the different inhibitors targeting RAS and MYC currently undergoing clinical trials, along with their clinical outcomes and the different combination strategies being explored to overcome drug resistance. This recent clinical development suggests a paradigm shift in the long-standing belief of RAS and MYC "undruggability", hinting at a new era in their therapeutic targeting.
Collapse
Affiliation(s)
| | - Íñigo González-Larreategui
- Models of cancer therapies Laboratory, Vall d'Hebron Institute of Oncology, Cellex Centre, Hospital University Vall d'Hebron Campus, Barcelona, Spain
| | - Daniel Capitán-Leo
- Models of cancer therapies Laboratory, Vall d'Hebron Institute of Oncology, Cellex Centre, Hospital University Vall d'Hebron Campus, Barcelona, Spain
| | - Laura Soucek
- Peptomyc S.L., Barcelona, Spain.
- Models of cancer therapies Laboratory, Vall d'Hebron Institute of Oncology, Cellex Centre, Hospital University Vall d'Hebron Campus, Barcelona, Spain.
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain.
- Department of Biochemistry and Molecular Biology, Universitat Autonoma de Barcelona, Bellaterra, Spain.
| |
Collapse
|
31
|
Curti L, Rohban S, Bianchi N, Croci O, Andronache A, Barozzi S, Mattioli M, Ricci F, Pastori E, Sberna S, Bellotti S, Accialini A, Ballarino R, Crosetto N, Wade M, Parazzoli D, Campaner S. CDK12 controls transcription at damaged genes and prevents MYC-induced transcription-replication conflicts. Nat Commun 2024; 15:7100. [PMID: 39155303 PMCID: PMC11330984 DOI: 10.1038/s41467-024-51229-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 08/01/2024] [Indexed: 08/20/2024] Open
Abstract
The identification of genes involved in replicative stress is key to understanding cancer evolution and to identify therapeutic targets. Here, we show that CDK12 prevents transcription-replication conflicts (TRCs) and the activation of cytotoxic replicative stress upon deregulation of the MYC oncogene. CDK12 was recruited at damaged genes by PARP-dependent DDR-signaling and elongation-competent RNAPII, to repress transcription. Either loss or chemical inhibition of CDK12 led to DDR-resistant transcription of damaged genes. Loss of CDK12 exacerbated TRCs in MYC-overexpressing cells and led to the accumulation of double-strand DNA breaks, occurring between co-directional early-replicating regions and transcribed genes. Overall, our data demonstrate that CDK12 protects genome integrity by repressing transcription of damaged genes, which is required for proper resolution of DSBs at oncogene-induced TRCs. This provides a rationale that explains both how CDK12 deficiency can promote tandem duplications of early-replicated regions during tumor evolution, and how CDK12 targeting can exacerbate replicative-stress in tumors.
Collapse
Affiliation(s)
- Laura Curti
- Center for Genomic Science of IIT, CGS@SEMM (Istituto Italiano di Tecnologia at European School of Molecular Medicine), Fondazione Istituto Italiano di Tecnologia (IIT), 20139, Milan, Italy
| | - Sara Rohban
- Center for Genomic Science of IIT, CGS@SEMM (Istituto Italiano di Tecnologia at European School of Molecular Medicine), Fondazione Istituto Italiano di Tecnologia (IIT), 20139, Milan, Italy
| | - Nicola Bianchi
- Center for Genomic Science of IIT, CGS@SEMM (Istituto Italiano di Tecnologia at European School of Molecular Medicine), Fondazione Istituto Italiano di Tecnologia (IIT), 20139, Milan, Italy
| | - Ottavio Croci
- Center for Genomic Science of IIT, CGS@SEMM (Istituto Italiano di Tecnologia at European School of Molecular Medicine), Fondazione Istituto Italiano di Tecnologia (IIT), 20139, Milan, Italy
| | - Adrian Andronache
- Center for Genomic Science of IIT, CGS@SEMM (Istituto Italiano di Tecnologia at European School of Molecular Medicine), Fondazione Istituto Italiano di Tecnologia (IIT), 20139, Milan, Italy
| | - Sara Barozzi
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Michela Mattioli
- Center for Genomic Science of IIT, CGS@SEMM (Istituto Italiano di Tecnologia at European School of Molecular Medicine), Fondazione Istituto Italiano di Tecnologia (IIT), 20139, Milan, Italy
| | - Fernanda Ricci
- Center for Genomic Science of IIT, CGS@SEMM (Istituto Italiano di Tecnologia at European School of Molecular Medicine), Fondazione Istituto Italiano di Tecnologia (IIT), 20139, Milan, Italy
| | - Elena Pastori
- Center for Genomic Science of IIT, CGS@SEMM (Istituto Italiano di Tecnologia at European School of Molecular Medicine), Fondazione Istituto Italiano di Tecnologia (IIT), 20139, Milan, Italy
| | - Silvia Sberna
- Center for Genomic Science of IIT, CGS@SEMM (Istituto Italiano di Tecnologia at European School of Molecular Medicine), Fondazione Istituto Italiano di Tecnologia (IIT), 20139, Milan, Italy
| | - Simone Bellotti
- Center for Genomic Science of IIT, CGS@SEMM (Istituto Italiano di Tecnologia at European School of Molecular Medicine), Fondazione Istituto Italiano di Tecnologia (IIT), 20139, Milan, Italy
| | - Anna Accialini
- Center for Genomic Science of IIT, CGS@SEMM (Istituto Italiano di Tecnologia at European School of Molecular Medicine), Fondazione Istituto Italiano di Tecnologia (IIT), 20139, Milan, Italy
| | - Roberto Ballarino
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, SE-17165, Stockholm, Sweden
- Science for Life Laboratory, Tomtebodavägen 23A, SE-17165, Solna, Sweden
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Nicola Crosetto
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, SE-17165, Stockholm, Sweden
- Science for Life Laboratory, Tomtebodavägen 23A, SE-17165, Solna, Sweden
- Human Technopole, Viale Rita Levi-Montalcini 1, 20157, Milan, Italy
| | - Mark Wade
- Center for Genomic Science of IIT, CGS@SEMM (Istituto Italiano di Tecnologia at European School of Molecular Medicine), Fondazione Istituto Italiano di Tecnologia (IIT), 20139, Milan, Italy
- Astex Pharmaceuticals, 436 Cambridge Science Park, CB4 0QA, Cambridge, UK
| | - Dario Parazzoli
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Stefano Campaner
- Center for Genomic Science of IIT, CGS@SEMM (Istituto Italiano di Tecnologia at European School of Molecular Medicine), Fondazione Istituto Italiano di Tecnologia (IIT), 20139, Milan, Italy.
| |
Collapse
|
32
|
Miyahira AK, Kamran SC, Jamaspishvili T, Marshall CH, Maxwell KN, Parolia A, Zorko NA, Pienta KJ, Soule HR. Disrupting prostate cancer research: Challenge accepted; report from the 2023 Coffey-Holden Prostate Cancer Academy Meeting. Prostate 2024; 84:993-1015. [PMID: 38682886 DOI: 10.1002/pros.24721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 04/16/2024] [Indexed: 05/01/2024]
Abstract
INTRODUCTION The 2023 Coffey-Holden Prostate Cancer Academy (CHPCA) Meeting, themed "Disrupting Prostate Cancer Research: Challenge Accepted," was convened at the University of California, Los Angeles, Luskin Conference Center, in Los Angeles, CA, from June 22 to 25, 2023. METHODS The 2023 marked the 10th Annual CHPCA Meeting, a discussion-oriented scientific think-tank conference convened annually by the Prostate Cancer Foundation, which centers on innovative and emerging research topics deemed pivotal for advancing critical unmet needs in prostate cancer research and clinical care. The 2023 CHPCA Meeting was attended by 81 academic investigators and included 40 talks across 8 sessions. RESULTS The central topic areas covered at the meeting included: targeting transcription factor neo-enhancesomes in cancer, AR as a pro-differentiation and oncogenic transcription factor, why few are cured with androgen deprivation therapy and how to change dogma to cure metastatic prostate cancer without castration, reducing prostate cancer morbidity and mortality with genetics, opportunities for radiation to enhance therapeutic benefit in oligometastatic prostate cancer, novel immunotherapeutic approaches, and the new era of artificial intelligence-driven precision medicine. DISCUSSION This article provides an overview of the scientific presentations delivered at the 2023 CHPCA Meeting, such that this knowledge can help in facilitating the advancement of prostate cancer research worldwide.
Collapse
Affiliation(s)
- Andrea K Miyahira
- Science Department, Prostate Cancer Foundation, Santa Monica, California, USA
| | - Sophia C Kamran
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Tamara Jamaspishvili
- Department of Pathology and Laboratory Medicine, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Catherine H Marshall
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kara N Maxwell
- Department of Medicine-Hematology/Oncology and Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Medicine Service, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania, USA
| | - Abhijit Parolia
- Department of Pathology, Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Nicholas A Zorko
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
- University of Minnesota Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Kenneth J Pienta
- The James Buchanan Brady Urological Institute, The Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Howard R Soule
- Science Department, Prostate Cancer Foundation, Santa Monica, California, USA
| |
Collapse
|
33
|
Hushmandi K, Saadat SH, Raei M, Daneshi S, Aref AR, Nabavi N, Taheriazam A, Hashemi M. Implications of c-Myc in the pathogenesis and treatment efficacy of urological cancers. Pathol Res Pract 2024; 259:155381. [PMID: 38833803 DOI: 10.1016/j.prp.2024.155381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/08/2024] [Accepted: 05/28/2024] [Indexed: 06/06/2024]
Abstract
Urological cancers, including prostate, bladder, and renal cancers, are significant causes of death and negatively impact the quality of life for patients. The development and progression of these cancers are linked to the dysregulation of molecular pathways. c-Myc, recognized as an oncogene, exhibits abnormal levels in various types of tumors, and current evidence supports the therapeutic targeting of c-Myc in cancer treatment. This review aims to elucidate the role of c-Myc in driving the progression of urological cancers. c-Myc functions to enhance tumorigenesis and has been documented to increase growth and metastasis in prostate, bladder, and renal cancers. Furthermore, the dysregulation of c-Myc can result in a diminished response to therapy in these cancers. Non-coding RNAs, β-catenin, and XIAP are among the regulators of c-Myc in urological cancers. Targeting and suppressing c-Myc therapeutically for the treatment of these cancers has been explored. Additionally, the expression level of c-Myc may serve as a prognostic factor in clinical settings.
Collapse
Affiliation(s)
- Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Seyed Hassan Saadat
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mehdi Raei
- Health Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran; Department of Epidemiology and Biostatistics, School of Health, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Salman Daneshi
- Department of Public Health,School of Health,Jiroft University Of Medical Sciences, Jiroft, Iran
| | - Amir Reza Aref
- Department of Translational Sciences, Xsphera Biosciences Inc. Boston, MA, USA; Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Noushin Nabavi
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, V6H3Z6, Vancouver, BC, Canada
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
34
|
Papadopoulos D, Ha SA, Fleischhauer D, Uhl L, Russell TJ, Mikicic I, Schneider K, Brem A, Valanju OR, Cossa G, Gallant P, Schuelein-Voelk C, Maric HM, Beli P, Büchel G, Vos SM, Eilers M. The MYCN oncoprotein is an RNA-binding accessory factor of the nuclear exosome targeting complex. Mol Cell 2024; 84:2070-2086.e20. [PMID: 38703770 DOI: 10.1016/j.molcel.2024.04.007] [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: 09/05/2023] [Revised: 02/28/2024] [Accepted: 04/10/2024] [Indexed: 05/06/2024]
Abstract
The MYCN oncoprotein binds active promoters in a heterodimer with its partner protein MAX. MYCN also interacts with the nuclear exosome, a 3'-5' exoribonuclease complex, suggesting a function in RNA metabolism. Here, we show that MYCN forms stable high-molecular-weight complexes with the exosome and multiple RNA-binding proteins. MYCN binds RNA in vitro and in cells via a conserved sequence termed MYCBoxI. In cells, MYCN associates with thousands of intronic transcripts together with the ZCCHC8 subunit of the nuclear exosome targeting complex and enhances their processing. Perturbing exosome function results in global re-localization of MYCN from promoters to intronic RNAs. On chromatin, MYCN is then replaced by the MNT(MXD6) repressor protein, inhibiting MYCN-dependent transcription. RNA-binding-deficient alleles show that RNA-binding limits MYCN's ability to activate cell growth-related genes but is required for MYCN's ability to promote progression through S phase and enhance the stress resilience of neuroblastoma cells.
Collapse
Affiliation(s)
- Dimitrios Papadopoulos
- Theodor Boveri Institute, Department of Biochemistry and Molecular Biology, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany; Mildred Scheel Early Career Center, University Hospital Würzburg, Josef-Schneider-Str. 6, 97080 Würzburg, Germany
| | - Stefanie Anh Ha
- Theodor Boveri Institute, Department of Biochemistry and Molecular Biology, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Daniel Fleischhauer
- Theodor Boveri Institute, Department of Biochemistry and Molecular Biology, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Leonie Uhl
- Theodor Boveri Institute, Department of Biochemistry and Molecular Biology, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Timothy J Russell
- Massachusetts Institute of Technology, Department of Biology, 31 Ames Street, Cambridge, MA 02142, USA
| | - Ivan Mikicic
- Institute of Developmental Biology and Neurobiology (IDN), Johannes Gutenberg University, Ackermannweg 4, 55128 Mainz, Germany; Institute of Molecular Biology (IMB), Johannes Gutenberg University, Ackermannweg 4, 55128 Mainz, Germany
| | - Katharina Schneider
- Massachusetts Institute of Technology, Department of Biology, 31 Ames Street, Cambridge, MA 02142, USA
| | - Annika Brem
- Massachusetts Institute of Technology, Department of Biology, 31 Ames Street, Cambridge, MA 02142, USA
| | - Omkar Rajendra Valanju
- Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Josef-Schneider-Str. 2, Building D15, 97080 Würzburg, Germany
| | - Giacomo Cossa
- Theodor Boveri Institute, Department of Biochemistry and Molecular Biology, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Peter Gallant
- Theodor Boveri Institute, Department of Biochemistry and Molecular Biology, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Christina Schuelein-Voelk
- Theodor Boveri Institute, Core Unit High-Content Microscopy, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Hans Michael Maric
- Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Josef-Schneider-Str. 2, Building D15, 97080 Würzburg, Germany
| | - Petra Beli
- Institute of Developmental Biology and Neurobiology (IDN), Johannes Gutenberg University, Ackermannweg 4, 55128 Mainz, Germany; Institute of Molecular Biology (IMB), Johannes Gutenberg University, Ackermannweg 4, 55128 Mainz, Germany
| | - Gabriele Büchel
- Theodor Boveri Institute, Department of Biochemistry and Molecular Biology, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany; Mildred Scheel Early Career Center, University Hospital Würzburg, Josef-Schneider-Str. 6, 97080 Würzburg, Germany
| | - Seychelle M Vos
- Massachusetts Institute of Technology, Department of Biology, 31 Ames Street, Cambridge, MA 02142, USA.
| | - Martin Eilers
- Theodor Boveri Institute, Department of Biochemistry and Molecular Biology, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany.
| |
Collapse
|
35
|
Liu X, Shi Q, Qi P, Wang Z, Zhang T, Zhang S, Wu J, Guo Z, Chen J, Zhang Q. Recent advances in living cell nucleic acid probes based on nanomaterials for early cancer diagnosis. Asian J Pharm Sci 2024; 19:100910. [PMID: 38948397 PMCID: PMC11214190 DOI: 10.1016/j.ajps.2024.100910] [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: 06/30/2023] [Revised: 11/16/2023] [Accepted: 02/05/2024] [Indexed: 07/02/2024] Open
Abstract
The early diagnosis of cancer is vital for effective treatment and improved prognosis. Tumor biomarkers, which can be used for the early diagnosis, treatment, and prognostic evaluation of cancer, have emerged as a topic of intense research interest in recent years. Nucleic acid, as a type of tumor biomarker, contains vital genetic information, which is of great significance for the occurrence and development of cancer. Currently, living cell nucleic acid probes, which enable the in situ imaging and dynamic monitoring of nucleic acids, have become a rapidly developing field. This review focuses on living cell nucleic acid probes that can be used for the early diagnosis of tumors. We describe the fundamental design of the probe in terms of three units and focus on the roles of different nanomaterials in probe delivery.
Collapse
Affiliation(s)
- Xuyao Liu
- Department of Thyroid Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun 130021, China
| | - Qi Shi
- Department of Thyroid Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun 130021, China
| | - Peng Qi
- Department of Thyroid Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun 130021, China
| | - Ziming Wang
- Department of Thyroid Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun 130021, China
| | - Tongyue Zhang
- Department of Thyroid Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun 130021, China
| | - Sijia Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Jiayan Wu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Zhaopei Guo
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Jie Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Qiang Zhang
- Department of Thyroid Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun 130021, China
| |
Collapse
|
36
|
Li S, Feng T, Liu Y, Yang Q, Song A, Wang S, Xie J, Zhang J, Yuan B, Sun Z. m 1A inhibition fuels oncolytic virus-elicited antitumor immunity via downregulating MYC/PD-L1 signaling. Int J Oral Sci 2024; 16:36. [PMID: 38730256 PMCID: PMC11087574 DOI: 10.1038/s41368-024-00304-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 03/13/2024] [Accepted: 04/15/2024] [Indexed: 05/12/2024] Open
Abstract
N1-methyladenosine (m1A) RNA methylation is critical for regulating mRNA translation; however, its role in the development, progression, and immunotherapy response of head and neck squamous cell carcinoma (HNSCC) remains largely unknown. Using Tgfbr1 and Pten conditional knockout (2cKO) mice, we found the neoplastic transformation of oral mucosa was accompanied by increased m1A modification levels. Analysis of m1A-associated genes identified TRMT61A as a key m1A writer linked to cancer progression and poor prognosis. Mechanistically, TRMT61A-mediated tRNA-m1A modification promotes MYC protein synthesis, upregulating programmed death-ligand 1 (PD-L1) expression. Moreover, m1A modification levels were also elevated in tumors treated with oncolytic herpes simplex virus (oHSV), contributing to reactive PD-L1 upregulation. Therapeutic m1A inhibition sustained oHSV-induced antitumor immunity and reduced tumor growth, representing a promising strategy to alleviate resistance. These findings indicate that m1A inhibition can prevent immune escape after oHSV therapy by reducing PD-L1 expression, providing a mutually reinforcing combination immunotherapy approach.
Collapse
Affiliation(s)
- Shujin Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Tian Feng
- School of Public Health, Wuhan University, Wuhan, China
| | - Yuantong Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Qichao Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - An Song
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Shuo Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Jun Xie
- State Key Laboratory of Virology, Medical Research Institute, Wuhan University, Wuhan, China
| | - Junjie Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- State Key Laboratory of Virology, Medical Research Institute, Wuhan University, Wuhan, China
| | - Bifeng Yuan
- School of Public Health, Wuhan University, Wuhan, China
| | - Zhijun Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China.
- Department of Oral Maxillofacial-Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
37
|
Ya A, Deng C, Godek KM. Cell Competition Eliminates Aneuploid Human Pluripotent Stem Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.08.593217. [PMID: 38766106 PMCID: PMC11100710 DOI: 10.1101/2024.05.08.593217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Human pluripotent stem cells (hPSCs) maintain diploid populations for generations despite a persistently high rate of mitotic errors that cause aneuploidy, or chromosome imbalances. Consequently, to maintain genome stability, aneuploidy must inhibit hPSC proliferation, but the mechanisms are unknown. Here, we surprisingly find that homogeneous aneuploid populations of hPSCs proliferate unlike aneuploid non-transformed somatic cells. Instead, in mosaic populations, cell non-autonomous competition between neighboring diploid and aneuploid hPSCs eliminates less fit aneuploid cells. Aneuploid hPSCs with lower Myc or higher p53 levels relative to diploid neighbors are outcompeted but conversely gain a selective advantage when Myc and p53 relative abundance switches. Thus, although hPSCs frequently missegregate chromosomes and inherently tolerate aneuploidy, Myc- and p53-driven cell competition preserves their genome integrity. These findings have important implications for the use of hPSCs in regenerative medicine and for how diploid human embryos are established despite the prevalence of aneuploidy during early development.
Collapse
Affiliation(s)
- Amanda Ya
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Chenhui Deng
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Kristina M. Godek
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
- Lead contact
| |
Collapse
|
38
|
Peng M, Keppeke GD, Tsai LK, Chang CC, Liu JL, Sung LY. The IMPDH cytoophidium couples metabolism and fetal development in mice. Cell Mol Life Sci 2024; 81:210. [PMID: 38717553 PMCID: PMC11078715 DOI: 10.1007/s00018-024-05233-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/04/2024] [Accepted: 04/05/2024] [Indexed: 05/12/2024]
Abstract
The cytoophidium is an evolutionarily conserved subcellular structure formed by filamentous polymers of metabolic enzymes. In vertebrates, inosine monophosphate dehydrogenase (IMPDH), which catalyses the rate-limiting step in guanosine triphosphate (GTP) biosynthesis, is one of the best-known cytoophidium-forming enzymes. Formation of the cytoophidium has been proposed to alleviate the inhibition of IMPDH, thereby facilitating GTP production to support the rapid proliferation of certain cell types such as lymphocytes, cancer cells and pluripotent stem cells (PSCs). However, past studies lacked appropriate models to elucidate the significance of IMPDH cytoophidium under normal physiological conditions. In this study, we demonstrate that the presence of IMPDH cytoophidium in mouse PSCs correlates with their metabolic status rather than pluripotency. By introducing IMPDH2 Y12C point mutation through genome editing, we established mouse embryonic stem cell (ESC) lines incapable of forming IMPDH polymers and the cytoophidium. Our data indicate an important role of IMPDH cytoophidium in sustaining a positive feedback loop that couples nucleotide biosynthesis with upstream metabolic pathways. Additionally, we find that IMPDH2 Y12C mutation leads to decreased cell proliferation and increased DNA damage in teratomas, as well as impaired embryo development following blastocoel injection. Further analysis shows that IMPDH cytoophidium assembly in mouse embryonic development begins after implantation and gradually increases throughout fetal development. These findings provide insights into the regulation of IMPDH polymerisation in embryogenesis and its significance in coordinating cell metabolism and development.
Collapse
Affiliation(s)
- Min Peng
- Institute of Biotechnology, National Taiwan University, Taipei, 106, Taiwan
| | - Gerson D Keppeke
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
| | - Li-Kuang Tsai
- Institute of Biotechnology, National Taiwan University, Taipei, 106, Taiwan
| | - Chia-Chun Chang
- Institute of Biotechnology, National Taiwan University, Taipei, 106, Taiwan.
| | - Ji-Long Liu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK.
| | - Li-Ying Sung
- Institute of Biotechnology, National Taiwan University, Taipei, 106, Taiwan.
- Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, 106, Taiwan.
- Center for Biotechnology, National Taiwan University, Taipei, 106, Taiwan.
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, 115, Taiwan.
| |
Collapse
|
39
|
Wang J, Yang Y, Shao F, Meng Y, Guo D, He J, Lu Z. Acetate reprogrammes tumour metabolism and promotes PD-L1 expression and immune evasion by upregulating c-Myc. Nat Metab 2024; 6:914-932. [PMID: 38702440 DOI: 10.1038/s42255-024-01037-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 03/21/2024] [Indexed: 05/06/2024]
Abstract
Acetate, a precursor of acetyl-CoA, is instrumental in energy production, lipid synthesis and protein acetylation. However, whether acetate reprogrammes tumour metabolism and plays a role in tumour immune evasion remains unclear. Here, we show that acetate is the most abundant short-chain fatty acid in human non-small cell lung cancer tissues, with increased tumour-enriched acetate uptake. Acetate-derived acetyl-CoA induces c-Myc acetylation, which is mediated by the moonlighting function of the metabolic enzyme dihydrolipoamide S-acetyltransferase. Acetylated c-Myc increases its stability and subsequent transcription of the genes encoding programmed death-ligand 1, glycolytic enzymes, monocarboxylate transporter 1 and cell cycle accelerators. Dietary acetate supplementation promotes tumour growth and inhibits CD8+ T cell infiltration, whereas disruption of acetate uptake inhibits immune evasion, which increases the efficacy of anti-PD-1-based therapy. These findings highlight a critical role of acetate promoting tumour growth beyond its metabolic role as a carbon source by reprogramming tumour metabolism and immune evasion, and underscore the potential of controlling acetate metabolism to curb tumour growth and improve the response to immune checkpoint blockade therapy.
Collapse
Affiliation(s)
- Juhong Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yannan Yang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fei Shao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Laboratory of Translational Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ying Meng
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Dong Guo
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Zhimin Lu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.
- Cancer Center, Zhejiang University, Hangzhou, China.
| |
Collapse
|
40
|
Guarducci C, Nardone A, Russo D, Nagy Z, Heraud C, Grinshpun A, Zhang Q, Freelander A, Leventhal MJ, Feit A, Cohen Feit G, Feiglin A, Liu W, Hermida-Prado F, Kesten N, Ma W, De Angelis C, Morlando A, O'Donnell M, Naumenko S, Huang S, Nguyen QD, Huang Y, Malorni L, Bergholz JS, Zhao JJ, Fraenkel E, Lim E, Schiff R, Shapiro GI, Jeselsohn R. Selective CDK7 Inhibition Suppresses Cell Cycle Progression and MYC Signaling While Enhancing Apoptosis in Therapy-resistant Estrogen Receptor-positive Breast Cancer. Clin Cancer Res 2024; 30:1889-1905. [PMID: 38381406 PMCID: PMC11061603 DOI: 10.1158/1078-0432.ccr-23-2975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 01/09/2024] [Accepted: 02/16/2024] [Indexed: 02/22/2024]
Abstract
PURPOSE Resistance to endocrine therapy (ET) and CDK4/6 inhibitors (CDK4/6i) is a clinical challenge in estrogen receptor (ER)-positive (ER+) breast cancer. Cyclin-dependent kinase 7 (CDK7) is a candidate target in endocrine-resistant ER+ breast cancer models and selective CDK7 inhibitors (CDK7i) are in clinical development for the treatment of ER+ breast cancer. Nonetheless, the precise mechanisms responsible for the activity of CDK7i in ER+ breast cancer remain elusive. Herein, we sought to unravel these mechanisms. EXPERIMENTAL DESIGN We conducted multi-omic analyses in ER+ breast cancer models in vitro and in vivo, including models with different genetic backgrounds. We also performed genome-wide CRISPR/Cas9 knockout screens to identify potential therapeutic vulnerabilities in CDK4/6i-resistant models. RESULTS We found that the on-target antitumor effects of CDK7 inhibition in ER+ breast cancer are in part p53 dependent, and involve cell cycle inhibition and suppression of c-Myc. Moreover, CDK7 inhibition exhibited cytotoxic effects, distinctive from the cytostatic nature of ET and CDK4/6i. CDK7 inhibition resulted in suppression of ER phosphorylation at S118; however, long-term CDK7 inhibition resulted in increased ER signaling, supporting the combination of ET with a CDK7i. Finally, genome-wide CRISPR/Cas9 knockout screens identified CDK7 and MYC signaling as putative vulnerabilities in CDK4/6i resistance, and CDK7 inhibition effectively inhibited CDK4/6i-resistant models. CONCLUSIONS Taken together, these findings support the clinical investigation of selective CDK7 inhibition combined with ET to overcome treatment resistance in ER+ breast cancer. In addition, our study highlights the potential of increased c-Myc activity and intact p53 as predictors of sensitivity to CDK7i-based treatments.
Collapse
Affiliation(s)
- Cristina Guarducci
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Agostina Nardone
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Douglas Russo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Zsuzsanna Nagy
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Capucine Heraud
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Albert Grinshpun
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Qi Zhang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Allegra Freelander
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Mathew Joseph Leventhal
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
- Computational and Systems Biology PhD program, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Avery Feit
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Gabriella Cohen Feit
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ariel Feiglin
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts
| | - Weihan Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Francisco Hermida-Prado
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Nikolas Kesten
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Wen Ma
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Carmine De Angelis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, Naples, Italy
| | - Antonio Morlando
- Bioinformatics Unit, Department of Oncology, Hospital of Prato, Azienda USL Toscana Centro, Prato, Italy
| | - Madison O'Donnell
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Sergey Naumenko
- Department of Biostatistics, Harvard Chan School of Public Health, Boston, Massachusetts
| | - Shixia Huang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Quang-Dé Nguyen
- Lurie Family Imaging Center, Center for Biomedical Imaging in Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ying Huang
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Luca Malorni
- Translational Research Unit, Department of Oncology, Hospital of Prato, Azienda USL Toscana Centro, Prato, Italy
| | - Johann S. Bergholz
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts
| | - Jean J. Zhao
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts
| | - Ernest Fraenkel
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Elgene Lim
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Rachel Schiff
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Geoffrey I. Shapiro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Rinath Jeselsohn
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
41
|
Khan AJ, Man S, Abbas M, Liu S, Zhang F. FBXO8 is a novel prognostic biomarker in different molecular subtypes of breast cancer and suppresses breast cancer progression by targeting c-MYC. Biochim Biophys Acta Gen Subj 2024; 1868:130577. [PMID: 38301858 DOI: 10.1016/j.bbagen.2024.130577] [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: 12/07/2023] [Revised: 01/09/2024] [Accepted: 01/21/2024] [Indexed: 02/03/2024]
Abstract
F-box only protein 8 (FBXO8) is a recently identified member of the F-box proteins, showcasing its novelty in this protein family. Extensive research has established FBXO8's role as a tumor suppressor in various cancers, including hepatocellular carcinoma, and colorectal cancer, Nevertheless, its functional, mechanistic, and prognostic roles in primary and metastatic breast cancer, particularly in different molecular subtypes of breast cancer, various stages, as well as its potential implications in immunotherapy, tumor microenvironment, and prognostic survival among breast cancer patients, remain unexplored. In this article, we employed a multi-dimensional investigation leveraging TCGA, TIMER, TISIDB, STRING, MEXPRESS, UALCAN, and cBioPortal databases to explore the underlying suppression mechanism of FBXO8 in breast cancer. FBXO8 negatively correlates with MYC, NOTCH, WNT and inflammatory signaling pathways in breast tumor microenvironment. Furthermore we conducted RT-PCR, western blot, cell proliferation, cell migration, and mRNA target gene RT-PCR analyses to elucidate the role of FBXO8 in breast cancer progression. Mechanistically, PTEN and FBXW7 expression were down-regulated and MYC, IL10, IL6, NOTCH1, WNT6 mRNA expressions were up-regulated in FBXO8 knockdown cell lines. c-MYC silenced cells showed an increase in FBXO8 protein level, which suggests a negative feedback loop between FBXO8 and c-MYC to control breast cancer metastasis. These findings illuminate the novel role of FBXO8 as a prognostic and therapeutic target across different molecular subtypes of breast cancer. Finally, through the utilization of virtual screening and Molecular Dynamics simulations, we successfully identified two FDA-approved medications, Ledipasvir and Paritaprevir, that demonstrated robust binding capabilities and interactions with FBXO8.
Collapse
Affiliation(s)
- Abdul Jamil Khan
- Biomedical Nanocenter, School of Life Science, Inner Mongolia Agricultural University, Hohhot 010011, China
| | - Shad Man
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot 010020, China
| | - Manzar Abbas
- Inner Mongolia Saikexing Institute of Breeding and Reproductive Biotechnology in Domestic Animal, Hohhot 011517, China
| | - Shihao Liu
- Department of Informatics and Computer Engineering, Simon Kuznets Kharkiv National University of Economics, Nauky аve., 9-А, Kharkiv 61166, Ukraine
| | - Feng Zhang
- Key Laboratory of Optical Technology and Instrument for Medicine, Ministry of Education, University of Shanghai for Science and Technology, Shanghai 200093, China; Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China.
| |
Collapse
|
42
|
Liu Z, Zhang X, Xu M, Hong JJ, Ciardiello A, Lei H, Shern JF, Thiele CJ. MYCN drives oncogenesis by cooperating with the histone methyltransferase G9a and the WDR5 adaptor to orchestrate global gene transcription. PLoS Biol 2024; 22:e3002240. [PMID: 38547242 PMCID: PMC11003700 DOI: 10.1371/journal.pbio.3002240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 04/09/2024] [Accepted: 02/28/2024] [Indexed: 04/11/2024] Open
Abstract
MYCN activates canonical MYC targets involved in ribosome biogenesis, protein synthesis, and represses neuronal differentiation genes to drive oncogenesis in neuroblastoma (NB). How MYCN orchestrates global gene expression remains incompletely understood. Our study finds that MYCN binds promoters to up-regulate canonical MYC targets but binds to both enhancers and promoters to repress differentiation genes. MYCN binding also increases H3K4me3 and H3K27ac on canonical MYC target promoters and decreases H3K27ac on neuronal differentiation gene enhancers and promoters. WDR5 facilitates MYCN promoter binding to activate canonical MYC target genes, whereas MYCN recruits G9a to enhancers to repress neuronal differentiation genes. Targeting both MYCN's active and repressive transcriptional activities using both WDR5 and G9a inhibitors synergistically suppresses NB growth. We demonstrate that MYCN cooperates with WDR5 and G9a to orchestrate global gene transcription. The targeting of both these cofactors is a novel therapeutic strategy to indirectly target the oncogenic activity of MYCN.
Collapse
Affiliation(s)
- Zhihui Liu
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Xiyuan Zhang
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Man Xu
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Jason J. Hong
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Amanda Ciardiello
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Haiyan Lei
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Jack F. Shern
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Carol J. Thiele
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, United States of America
| |
Collapse
|
43
|
Gaballa A, Gebhardt-Wolf A, Krenz B, Mattavelli G, John M, Cossa G, Andreani S, Schülein-Völk C, Montesinos F, Vidal R, Kastner C, Ade CP, Kneitz B, Gasteiger G, Gallant P, Rosenfeldt M, Riedel A, Eilers M. PAF1c links S-phase progression to immune evasion and MYC function in pancreatic carcinoma. Nat Commun 2024; 15:1446. [PMID: 38365788 PMCID: PMC10873513 DOI: 10.1038/s41467-024-45760-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 02/05/2024] [Indexed: 02/18/2024] Open
Abstract
In pancreatic ductal adenocarcinoma (PDAC), endogenous MYC is required for S-phase progression and escape from immune surveillance. Here we show that MYC in PDAC cells is needed for the recruitment of the PAF1c transcription elongation complex to RNA polymerase and that depletion of CTR9, a PAF1c subunit, enables long-term survival of PDAC-bearing mice. PAF1c is largely dispensable for normal proliferation and regulation of MYC target genes. Instead, PAF1c limits DNA damage associated with S-phase progression by being essential for the expression of long genes involved in replication and DNA repair. Surprisingly, the survival benefit conferred by CTR9 depletion is not due to DNA damage, but to T-cell activation and restoration of immune surveillance. This is because CTR9 depletion releases RNA polymerase and elongation factors from the body of long genes and promotes the transcription of short genes, including MHC class I genes. The data argue that functionally distinct gene sets compete for elongation factors and directly link MYC-driven S-phase progression to tumor immune evasion.
Collapse
Affiliation(s)
- Abdallah Gaballa
- Department of Biochemistry and Molecular Biologyy, Theodor Boveri Institute, Biocenter, Julius Maximilian University Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Anneli Gebhardt-Wolf
- Department of Biochemistry and Molecular Biologyy, Theodor Boveri Institute, Biocenter, Julius Maximilian University Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Bastian Krenz
- Department of Biochemistry and Molecular Biologyy, Theodor Boveri Institute, Biocenter, Julius Maximilian University Würzburg, Am Hubland, 97074, Würzburg, Germany
- Mildred Scheel Early Career Center, University Hospital Würzburg, Josef-Schneider-Str. 2, 97080, Würzburg, Germany
| | - Greta Mattavelli
- Mildred Scheel Early Career Center, University Hospital Würzburg, Josef-Schneider-Str. 2, 97080, Würzburg, Germany
| | - Mara John
- Mildred Scheel Early Career Center, University Hospital Würzburg, Josef-Schneider-Str. 2, 97080, Würzburg, Germany
| | - Giacomo Cossa
- Department of Biochemistry and Molecular Biologyy, Theodor Boveri Institute, Biocenter, Julius Maximilian University Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Silvia Andreani
- Department of Biochemistry and Molecular Biologyy, Theodor Boveri Institute, Biocenter, Julius Maximilian University Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Christina Schülein-Völk
- Core Unit High-Content Microscopy, Theodor Boveri Institute, Biocenter, Julius Maximilian University Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Francisco Montesinos
- Department of Biochemistry and Molecular Biologyy, Theodor Boveri Institute, Biocenter, Julius Maximilian University Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Raphael Vidal
- Department of Biochemistry and Molecular Biologyy, Theodor Boveri Institute, Biocenter, Julius Maximilian University Würzburg, Am Hubland, 97074, Würzburg, Germany
- Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, Josef-Schneider-Str. 2, 97080, Würzburg, Germany
| | - Carolin Kastner
- Mildred Scheel Early Career Center, University Hospital Würzburg, Josef-Schneider-Str. 2, 97080, Würzburg, Germany
| | - Carsten P Ade
- Department of Biochemistry and Molecular Biologyy, Theodor Boveri Institute, Biocenter, Julius Maximilian University Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Burkhard Kneitz
- Department of Urology and Pediatric Urology, University Hospital Würzburg, Josef-Schneider-Str. 2, 97080, Würzburg, Germany
| | - Georg Gasteiger
- Würzburg Institute of Systems Immunology, Max Planck Research Group, Julius Maximilian University Würzburg, Versbacher Str. 9, 97078, Würzburg, Germany
| | - Peter Gallant
- Department of Biochemistry and Molecular Biologyy, Theodor Boveri Institute, Biocenter, Julius Maximilian University Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Mathias Rosenfeldt
- Institute of Pathology, Julius Maximilian University Würzburg, Josef-Schneider-Str. 2, 97080, Würzburg, Germany
| | - Angela Riedel
- Mildred Scheel Early Career Center, University Hospital Würzburg, Josef-Schneider-Str. 2, 97080, Würzburg, Germany
| | - Martin Eilers
- Department of Biochemistry and Molecular Biologyy, Theodor Boveri Institute, Biocenter, Julius Maximilian University Würzburg, Am Hubland, 97074, Würzburg, Germany.
- Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, Josef-Schneider-Str. 2, 97080, Würzburg, Germany.
| |
Collapse
|
44
|
Sepulveda GP, Gushchanskaia ES, Mora-Martin A, Esse R, Nikorich I, Ceballos A, Kwan J, Blum BC, Dholiya P, Emili A, Perissi V, Cardamone MD, Grishok A. DOT1L stimulates MYC/Mondo transcription factor activity by promoting its degradation cycle on chromatin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.06.579191. [PMID: 38370658 PMCID: PMC10871221 DOI: 10.1101/2024.02.06.579191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
The proto-oncogene c-MYC is a key representative of the MYC transcription factor network regulating growth and metabolism. MML-1 (Myc- and Mondo-like) is its homolog in C. elegans. The functional and molecular cooperation between c-MYC and H3 lysine 79 methyltransferase DOT1L was demonstrated in several human cancer types, and we have earlier discovered the connection between C. elegans MML-1 and DOT-1.1. Here, we demonstrate the critical role of DOT1L/DOT-1.1 in regulating c-MYC/MML-1 target genes genome-wide by ensuring the removal of "spent" transcription factors from chromatin by the nuclear proteasome. Moreover, we uncover a previously unrecognized proteolytic activity of DOT1L, which may facilitate c-MYC turnover. This new mechanism of c-MYC regulation by DOT1L may lead to the development of new approaches for cancer treatment.
Collapse
Affiliation(s)
- Gian P. Sepulveda
- Department of Biochemistry & Cell Biology, Boston University School of Medicine, Boston, MA, 02118, USA
- Graduate Program in Genetics and Genomics, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Ekaterina S. Gushchanskaia
- Department of Biochemistry & Cell Biology, Boston University School of Medicine, Boston, MA, 02118, USA
- Present address: Tessera Therapeutics, Somerville, MA, 02143, USA
| | - Alexandra Mora-Martin
- Department of Biochemistry & Cell Biology, Boston University School of Medicine, Boston, MA, 02118, USA
- Present address: Spanish National Cancer Research Center (CNIO), 28029, Madrid, Spain
| | - Ruben Esse
- Department of Biochemistry & Cell Biology, Boston University School of Medicine, Boston, MA, 02118, USA
- Present address: Cell and Gene Therapy Catapult, Guy’s Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Iana Nikorich
- Department of Biochemistry & Cell Biology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Ainhoa Ceballos
- Department of Biochemistry and Molecular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
- Present address: Research Unit, Diagnostica Longwood S.L. 50011 Zaragoza, Spain
| | - Julian Kwan
- Center for Network Systems Biology, Boston University, Boston, MA, 02118, USA
| | - Benjamin C. Blum
- Center for Network Systems Biology, Boston University, Boston, MA, 02118, USA
| | - Prakruti Dholiya
- Department of Biochemistry & Cell Biology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Andrew Emili
- Department of Biochemistry & Cell Biology, Boston University School of Medicine, Boston, MA, 02118, USA
- Center for Network Systems Biology, Boston University, Boston, MA, 02118, USA
- Division of Computational Biology, Boston University School of Medicine, Boston, MA, 02118, USA
- Present address: OHSU Knight Cancer Institute, School of Medicine, Portland, OR, 97239, USA
| | - Valentina Perissi
- Department of Biochemistry & Cell Biology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Maria D. Cardamone
- Department of Biochemistry & Cell Biology, Boston University School of Medicine, Boston, MA, 02118, USA
- Present address: Korro Bio Inc., Cambridge, MA, 02139, USA
| | - Alla Grishok
- Department of Biochemistry & Cell Biology, Boston University School of Medicine, Boston, MA, 02118, USA
- Genome Science Institute, Boston University, Boston, MA, 02118, USA
| |
Collapse
|
45
|
Zhang D, Zhao F, Liu H, Guo P, Li Z, Li S. FABP6 serves as a new therapeutic target in esophageal tumor. Aging (Albany NY) 2024; 16:1640-1662. [PMID: 38277205 PMCID: PMC10866426 DOI: 10.18632/aging.205448] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 12/04/2023] [Indexed: 01/27/2024]
Abstract
BACKGROUND Esophageal cancer is one of the most common malignant tumors with high incidence and mortality rates. Despite the continuous development of treatment options, the prognosis for esophageal cancer patients remains poor. Therefore, there is an urgent need for new diagnostic and therapeutic targets in clinical practice to improve the survival of patients with esophageal cancer. METHODS In this study, we conducted a comprehensive scRNA-seq analysis of the tumor microenvironment in primary esophageal tumors to elucidate cell composition and heterogeneity. Using Seurat, we identified eight clusters, encompassing non-immune cells (fibroblasts, myofibroblasts, endothelial cells, and epithelial cells) and immunocytes (myeloid-derived cells, T cells, B cells, and plasma cells). Compared to normal tissues, tumors exhibited an increased proportion of epithelial cells and alterations in immune cell infiltration. Analysis of epithelial cells revealed a cluster (cluster 0) with a high differentiation score and early distribution, suggesting its importance as a precursor cell. RESULTS Cluster 0 was characterized by high expression of FABP6, indicating a potential role in fatty acid metabolism and tumor growth. T cell analysis revealed shifts in the balance between Treg and CD8+ effector T cells in tumor tissues. Cellular communication analysis identified increased interactions between FABP6+ tumor cells and T cells, with the involvement of the MIF-related pathway and the CD74-CD44 interaction. This study provides insights into the cellular landscape and immune interactions within esophageal tumors, contributing to a better understanding of tumor heterogeneity and potential therapeutic targets.
Collapse
Affiliation(s)
- Dengfeng Zhang
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Fangchao Zhao
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Haitao Liu
- College of Life Science, Inner Mongolia University, Hohhot, Inner Mongolia 010031, China
| | - Pengfei Guo
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Zhirong Li
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Shujun Li
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| |
Collapse
|
46
|
Li Z, Huang Y, Hung TI, Sun J, Aispuro D, Chen B, Guevara N, Ji F, Cong X, Zhu L, Wang S, Guo Z, Chang CE, Xue M. MYC-Targeting Inhibitors Generated from a Stereodiversified Bicyclic Peptide Library. J Am Chem Soc 2024; 146:1356-1363. [PMID: 38170904 PMCID: PMC10797614 DOI: 10.1021/jacs.3c09615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 01/05/2024]
Abstract
Here, we present the second generation of our bicyclic peptide library (NTB), featuring a stereodiversified structure and a simplified construction strategy. We utilized a tandem ring-opening metathesis and ring-closing metathesis reaction (ROM-RCM) to cyclize the linear peptide library in a single step, representing the first reported instance of this reaction being applied to the preparation of macrocyclic peptides. Moreover, the resulting bicyclic peptide can be easily linearized for MS/MS sequencing with a one-step deallylation process. We employed this library to screen against the E363-R378 epitope of MYC and identified several MYC-targeting bicyclic peptides. Subsequent in vitro cell studies demonstrated that one candidate, NT-B2R, effectively suppressed MYC transcription activities and cell proliferation.
Collapse
Affiliation(s)
- Zhonghan Li
- Department
of Chemistry, University of California,
Riverside, Riverside, California 92521, United States
| | - Yi Huang
- Department
of Chemistry, University of California,
Riverside, Riverside, California 92521, United States
| | - Ta I Hung
- Department
of Chemistry, University of California,
Riverside, Riverside, California 92521, United States
| | - Jianan Sun
- Environmental
Toxicology Graduate Program, University
of California, Riverside, Riverside, California 92521, United States
| | - Desiree Aispuro
- Environmental
Toxicology Graduate Program, University
of California, Riverside, Riverside, California 92521, United States
| | - Boxi Chen
- Department
of Chemistry, University of California,
Riverside, Riverside, California 92521, United States
| | - Nathan Guevara
- Department
of Chemistry, University of California,
Riverside, Riverside, California 92521, United States
| | - Fei Ji
- Department
of Chemistry, University of California,
Riverside, Riverside, California 92521, United States
| | - Xu Cong
- Department
of Chemistry, University of California,
Riverside, Riverside, California 92521, United States
| | - Lingchao Zhu
- Department
of Chemistry, University of California,
Riverside, Riverside, California 92521, United States
| | - Siwen Wang
- Environmental
Toxicology Graduate Program, University
of California, Riverside, Riverside, California 92521, United States
| | - Zhili Guo
- Department
of Chemistry, University of California,
Riverside, Riverside, California 92521, United States
| | - Chia-en Chang
- Department
of Chemistry, University of California,
Riverside, Riverside, California 92521, United States
- Environmental
Toxicology Graduate Program, University
of California, Riverside, Riverside, California 92521, United States
| | - Min Xue
- Department
of Chemistry, University of California,
Riverside, Riverside, California 92521, United States
- Environmental
Toxicology Graduate Program, University
of California, Riverside, Riverside, California 92521, United States
| |
Collapse
|
47
|
Park YR, Jee W, Park SM, Kim SW, Jung JH, Kim H, Kim KI, Jang HJ. Acetylcorynoline Induces Apoptosis and G2/M Phase Arrest through the c-Myc Signaling Pathway in Colon Cancer Cells. Int J Mol Sci 2023; 24:17589. [PMID: 38139419 PMCID: PMC10744070 DOI: 10.3390/ijms242417589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
Colorectal cancer (CRC) is the third most commonly diagnosed cancer worldwide, and despite advances in treatment, survival rates are still low; therefore, the development of novel drugs is imperative. Acetylcorynoline (ACN) is derived from Corydalis ambigua Cham. et Schltdl tubers. The effect of ACN on colon cancer is still unknown. Therefore, we investigated its potential effects. Our data showed that ACN inhibited cell viability and proliferation. Moreover, ACN induced apoptosis and cell cycle arrest by inhibiting cell growth. In the present study, we hypothesized that ACN regulates c-Myc through CNOT2 or MID1IP1. ACN reduced the protein expression of oncogenic genes, decreased c-Myc half-life, and rapidly inhibited the serum stimulation response. Moreover, knockdown of CNOT2 and MID1IP1 with ACN increased apoptosis and further reduced the expression of oncogenes. In addition, ACN exhibited a synergistic effect with low-dose 5-fluorouracil (5-FU) and doxorubicin (Dox). Collectively, our data demonstrate that ACN inhibited c-Myc expression through CNOT2 and MID1IP1, and induced apoptosis. These findings indicate the potential of ACN as a therapeutic agent against colon cancer.
Collapse
Affiliation(s)
- Ye-Rin Park
- College of Korean Medicine, Kyung Hee University, 24, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; (Y.-R.P.); (W.J.); (S.-M.P.); (S.-W.K.); (J.-H.J.); (H.K.); (K.-I.K.)
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Wona Jee
- College of Korean Medicine, Kyung Hee University, 24, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; (Y.-R.P.); (W.J.); (S.-M.P.); (S.-W.K.); (J.-H.J.); (H.K.); (K.-I.K.)
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - So-Mi Park
- College of Korean Medicine, Kyung Hee University, 24, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; (Y.-R.P.); (W.J.); (S.-M.P.); (S.-W.K.); (J.-H.J.); (H.K.); (K.-I.K.)
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Seok-Woo Kim
- College of Korean Medicine, Kyung Hee University, 24, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; (Y.-R.P.); (W.J.); (S.-M.P.); (S.-W.K.); (J.-H.J.); (H.K.); (K.-I.K.)
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Ji-Hoon Jung
- College of Korean Medicine, Kyung Hee University, 24, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; (Y.-R.P.); (W.J.); (S.-M.P.); (S.-W.K.); (J.-H.J.); (H.K.); (K.-I.K.)
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Hyungsuk Kim
- College of Korean Medicine, Kyung Hee University, 24, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; (Y.-R.P.); (W.J.); (S.-M.P.); (S.-W.K.); (J.-H.J.); (H.K.); (K.-I.K.)
- Department of Korean Rehabilitation Medicine, Kyung Hee University Medical Center, Seoul 02447, Republic of Korea
| | - Kwan-Il Kim
- College of Korean Medicine, Kyung Hee University, 24, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; (Y.-R.P.); (W.J.); (S.-M.P.); (S.-W.K.); (J.-H.J.); (H.K.); (K.-I.K.)
- Division of Allergy, Immune and Respiratory System, Department of Internal Medicine, College of Korean Medicine, Kyung Hee Medical Center, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Hyeung-Jin Jang
- College of Korean Medicine, Kyung Hee University, 24, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; (Y.-R.P.); (W.J.); (S.-M.P.); (S.-W.K.); (J.-H.J.); (H.K.); (K.-I.K.)
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
48
|
Zhang Y, Zhang X, Zhang Y, Xu H, Wei Z, Wang X, Li Y, Guo J, Wu F, Fang X, Pang L, Deng B, Yu D. c-Myc inhibits LAPTM5 expression in B-cell lymphomas. Ann Hematol 2023; 102:3499-3513. [PMID: 37713124 DOI: 10.1007/s00277-023-05434-9] [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: 04/27/2023] [Accepted: 08/30/2023] [Indexed: 09/16/2023]
Abstract
Myc is a pivotal protooncogenic transcription factor that contributes to the development of almost all Burkitt's lymphomas and about one-third of diffuse large B-cell lymphomas. How B-cells sustain their uncontrolled proliferation due to high Myc is not yet well defined. Here, we found that Myc trans-represses the expression of murine LAPTM5, a gene coding a lysosome-associated protein, by binding to two E-boxes in the LAPTM5 promoter. While the product of intact mRNA (CDS+3'UTR) of LAPTM5 failed to suppress the growth of B-lymphomas, either the protein coded by coding sequence (CDS) itself or the non-coding 3'-untranslated region (3'UTR) mRNA was able to inhibit the growth of B-lymphomas. Moreover, Myc trans-activated miR-17-3p, which promoted tumor growth. Strikingly, LAPTM5 3'UTR contains 11 miR-17-3p-binding sites through which the LAPTM5 protein synthesis was inhibited. The functional interplay between low LAPTM5 mRNA and high miR-17-3p due to high Myc in B-lymphomas leads to further dampening of tumor-suppressive LAPTM5 protein, which promotes tumor progression. Our results indicate that Myc inhibits LAPTM5 expression in B-lymphoma cells by transcriptional and post-transcriptional modifications.
Collapse
Affiliation(s)
- Yanqing Zhang
- Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University Medical College, 136 Jiangyang Road, Yangzhou, Jiangsu Province, 225009, China
| | - Xin Zhang
- Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University Medical College, 136 Jiangyang Road, Yangzhou, Jiangsu Province, 225009, China
- Department of Pathology, Sir Run Run Shaw Hospital, Institute of Clinical Science, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi Zhang
- Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University Medical College, 136 Jiangyang Road, Yangzhou, Jiangsu Province, 225009, China
| | - Han Xu
- Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University Medical College, 136 Jiangyang Road, Yangzhou, Jiangsu Province, 225009, China
| | - Zichen Wei
- Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University Medical College, 136 Jiangyang Road, Yangzhou, Jiangsu Province, 225009, China
| | - Xin Wang
- Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University Medical College, 136 Jiangyang Road, Yangzhou, Jiangsu Province, 225009, China
| | - Yan Li
- Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University Medical College, 136 Jiangyang Road, Yangzhou, Jiangsu Province, 225009, China
| | - Junrong Guo
- Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University Medical College, 136 Jiangyang Road, Yangzhou, Jiangsu Province, 225009, China
| | - Fan Wu
- Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University Medical College, 136 Jiangyang Road, Yangzhou, Jiangsu Province, 225009, China
| | - Xiao Fang
- Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University Medical College, 136 Jiangyang Road, Yangzhou, Jiangsu Province, 225009, China
| | - Lei Pang
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Bin Deng
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Duonan Yu
- Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou, China.
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University Medical College, 136 Jiangyang Road, Yangzhou, Jiangsu Province, 225009, China.
| |
Collapse
|
49
|
Molina E, García-Gutiérrez L, Junco V, Perez-Olivares M, de Yébenes VG, Blanco R, Quevedo L, Acosta JC, Marín AV, Ulgiati D, Merino R, Delgado MD, Varela I, Regueiro JR, Moreno de Alborán I, Ramiro AR, León J. MYC directly transactivates CR2/CD21, the receptor of the Epstein-Barr virus, enhancing the viral infection of Burkitt lymphoma cells. Oncogene 2023; 42:3358-3370. [PMID: 37773203 DOI: 10.1038/s41388-023-02846-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 09/12/2023] [Accepted: 09/18/2023] [Indexed: 10/01/2023]
Abstract
MYC is an oncogenic transcription factor dysregulated in about half of total human tumors. While transcriptomic studies reveal more than 1000 genes regulated by MYC, a much smaller fraction of genes is directly transactivated by MYC. Virtually all Burkitt lymphoma (BL) carry chromosomal translocations involving MYC oncogene. Most endemic BL and a fraction of sporadic BL are associated with Epstein-Barr virus (EBV) infection. The currently accepted mechanism is that EBV is the BL-causing agent inducing MYC translocation. Herein we show that the EBV receptor, CR2 (also called CD21), is a direct MYC target gene. This is based on several pieces of evidence: MYC induces CR2 expression in both proliferating and arrested cells and in the absence of protein synthesis, binds the CR2 promoter and transactivates CR2 in an E-box-dependent manner. Moreover, using mice with conditional MYC ablation we show that MYC induces CR2 in primary B cells. Importantly, modulation of MYC levels directly correlates with EBV's ability of infection in BL cells. Altogether, in contrast to the widely accepted hypothesis for the correlation between EBV and BL, we propose an alternative hypothesis in which MYC dysregulation could be the first event leading to the subsequent EBV infection.
Collapse
Affiliation(s)
- Ester Molina
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Universidad de Cantabria-CSIC, Santander, Spain
- Departamento de Biología Molecular, Universidad de Cantabria, Santander, Spain
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Lucía García-Gutiérrez
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Universidad de Cantabria-CSIC, Santander, Spain
- Departamento de Biología Molecular, Universidad de Cantabria, Santander, Spain
| | - Vanessa Junco
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Universidad de Cantabria-CSIC, Santander, Spain
- Departamento de Biología Molecular, Universidad de Cantabria, Santander, Spain
| | - Mercedes Perez-Olivares
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB)-CSIC, Madrid, Spain
| | - Virginia G de Yébenes
- Centro Nacional de Investigaciones Cardiovasculares-CNIC Carlos III, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Universidad Complutense, School of Medicine and 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Rosa Blanco
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Universidad de Cantabria-CSIC, Santander, Spain
- Departamento de Biología Molecular, Universidad de Cantabria, Santander, Spain
| | - Laura Quevedo
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Universidad de Cantabria-CSIC, Santander, Spain
- Departamento de Biología Molecular, Universidad de Cantabria, Santander, Spain
| | - Juan C Acosta
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Universidad de Cantabria-CSIC, Santander, Spain
- Departamento de Biología Molecular, Universidad de Cantabria, Santander, Spain
| | - Ana V Marín
- Department of Immunology, Ophthalmology and ENT, Universidad Complutense, School of Medicine and 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Daniela Ulgiati
- School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia
| | - Ramon Merino
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Universidad de Cantabria-CSIC, Santander, Spain
- Departamento de Biología Molecular, Universidad de Cantabria, Santander, Spain
| | - M Dolores Delgado
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Universidad de Cantabria-CSIC, Santander, Spain
- Departamento de Biología Molecular, Universidad de Cantabria, Santander, Spain
| | - Ignacio Varela
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Universidad de Cantabria-CSIC, Santander, Spain
- Departamento de Biología Molecular, Universidad de Cantabria, Santander, Spain
| | - José R Regueiro
- Department of Immunology, Ophthalmology and ENT, Universidad Complutense, School of Medicine and 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | | | - Almudena R Ramiro
- Centro Nacional de Investigaciones Cardiovasculares-CNIC Carlos III, Madrid, Spain
| | - Javier León
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Universidad de Cantabria-CSIC, Santander, Spain.
- Departamento de Biología Molecular, Universidad de Cantabria, Santander, Spain.
| |
Collapse
|
50
|
Hamilton G, Stickler S, Rath B. Integration of signaling pathway and bromodomain and extra-terminal domain inhibition for the treatment of mutant Kirsten rat sarcoma viral oncogene homolog cancer. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:1027-1038. [PMID: 38023987 PMCID: PMC10651355 DOI: 10.37349/etat.2023.00178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 09/04/2023] [Indexed: 12/01/2023] Open
Abstract
Mutant Kirsten rat sarcoma viral oncogene homolog (KRAS) is now a drugable oncogenic driver and the KRAS G12C variant responds clinically to sotorasib and adagrasib that covalently block the cysteine of the active center and inhibit downstream signaling and proliferation. Unfortunately, progression-free survival (PFS) of lung cancer patients is only 5-6 months and no survival advantage has been found for sotorasib in comparison to docetaxel chemotherapy. Increased responses to KRAS inhibitors are tested in combination with the son of sevenless 1 (SOS1) inhibitors, upstream and downstream signaling modulators as well as chemotherapeutics. Some of these approaches are limited by toxicity to normal tissues and by diverse mechanisms of resistance. In essence, most of these attempts are directed to the inhibition of proliferation by impairment of the signal transduction pathways. The final target of KRAS-mediated growth stimulation is MYC in the cell nucleus that stimulates transcription of a host of genes. In detail, MYC alters genomic enhancer and super-enhancers of transcription that are frequently deregulated in cancer. Such enhancers can be targeted by bromodomain and extra-terminal (BET) inhibitors (BETi) or degraders and this review discusses whether integrated SOS1 inhibition and BET targeting of MYC synergizes against mutant KRAS tumor growth. BET degraders in the form of proteolysis-targeting chimeras (PROTACs) combined with BAY-293-mediated SOS1 inhibition revealed marked cytotoxic synergy against mutant KRAS cancer cells and may constitute a promising option for clinical treatment.
Collapse
Affiliation(s)
- Gerhard Hamilton
- Department of Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Sandra Stickler
- Department of Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Barbara Rath
- Department of Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| |
Collapse
|