1
|
Liu Y, Stockwell BR, Jiang X, Gu W. p53-regulated non-apoptotic cell death pathways and their relevance in cancer and other diseases. Nat Rev Mol Cell Biol 2025:10.1038/s41580-025-00842-3. [PMID: 40204927 DOI: 10.1038/s41580-025-00842-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2025] [Indexed: 04/11/2025]
Abstract
Programmed cell death is a mechanism that is crucial for numerous physiological and pathological processes. Whereas p53-mediated apoptosis is a major cell death pathway in cancer, accumulating evidence indicates that p53 also has crucial roles in controlling different non-apoptotic cell death (NACD) pathways, including ferroptosis, necroptosis, pyroptosis, autophagy-dependent cell death, entotic cell death, parthanatos and paraptosis, and may regulate PANoptosis, cuproptosis and disulfidptosis. Notably, the function of p53 in these NACDs substantially contributes to its biological effects, particularly in cancer development and other pathological processes. In this Review, we discuss recent advances in understanding the roles and underlying mechanisms of p53-mediated NACDs, focusing on ferroptosis, necroptosis and pyroptosis. We discuss the complex and distinct physiological settings in which NACDs are regulated by p53, and potential targeting of p53-regulated NACDs for the treatment of cancer and other human diseases. Finally, we highlight several important questions concerning p53-regulated NACDs that warrant further investigation.
Collapse
Affiliation(s)
- Yanqing Liu
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Brent R Stockwell
- Department of Chemistry, Columbia University, New York, NY, USA
- Department of Biological Sciences, Columbia University, New York, NY, USA
- Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Xuejun Jiang
- Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Wei Gu
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA.
- Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA.
| |
Collapse
|
2
|
He H, Li X, Su F, Jin H, Zhang J, Wang Y. Current and Emerging Approaches Targeting G9a for the Treatment of Various Diseases. J Med Chem 2025; 68:1068-1089. [PMID: 39740072 DOI: 10.1021/acs.jmedchem.4c02781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
Abstract
G9a, a histone lysine methyltransferase, is instrumental in regulating gene expression through epigenetic modifications. Its overexpression is closely linked to the progression of various human diseases, including cancers. Therefore, targeting G9a enzyme is a promising strategy for treating various diseases. Although no G9a inhibitors have yet reached clinical trials, several small molecule inhibitors have demonstrated strong preclinical efficacy. For instance, the orally available inhibitor 16 (DS79932728) shows significant potential for treating sickle cell disease, while 34 (compound 15h) has shown promising treatment of rhabdomyosarcoma. This Perspective summarizes the protein structure and biological functions of G9a, along with its association with various diseases. We highlight the design strategies, structure-activity relationships, and biological activity assessments of G9a inhibitors. Additionally, we discuss the unique advantages of the mechanisms of novel G9a inhibitors, offering insights for the future development of more effective drugs targeting G9a.
Collapse
Affiliation(s)
- Hua He
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Center and Laboratory of Neuro-system and Multimorbidity, Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiaoxue Li
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Center and Laboratory of Neuro-system and Multimorbidity, Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Feijing Su
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Center and Laboratory of Neuro-system and Multimorbidity, Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hong Jin
- College of Life Sciences, Sichuan University, Chengdu, Sichuan 610065, China
| | - Jifa Zhang
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Center and Laboratory of Neuro-system and Multimorbidity, Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yuxi Wang
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Center and Laboratory of Neuro-system and Multimorbidity, Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, Sichuan 610212, China
| |
Collapse
|
3
|
Huo Z, Zhang S, Su G, Cai Y, Chen R, Jiang M, Yang D, Zhang S, Xiong Y, Zhang X. Immunohistochemical Profiling of Histone Modification Biomarkers Identifies Subtype-Specific Epigenetic Signatures and Potential Drug Targets in Breast Cancer. Int J Mol Sci 2025; 26:770. [PMID: 39859484 PMCID: PMC11765579 DOI: 10.3390/ijms26020770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/10/2025] [Accepted: 01/14/2025] [Indexed: 01/30/2025] Open
Abstract
Breast cancer (BC) subtypes exhibit distinct epigenetic landscapes, with triple-negative breast cancer (TNBC) lacking effective targeted therapies. This study investigates histone biomarkers and therapeutic vulnerabilities across BC subtypes. The immunohistochemical profiling of >20 histone biomarkers, including histone modifications, modifiers, and oncohistone mutations, was conducted on a discovery cohort and a validation cohort of BC tissues, healthy controls, and cell line models. Transcriptomic and cell growth analyses were conducted to evaluate the effects of the small-molecule G9a inhibitor in diverse BC models. Key histone biomarkers, including H3K9me2, H3K36me2, and H3K79me, were differentially expressed across BC subtypes. H3K9me2 emerged as an independent predictor for distinguishing TNBC from other less-aggressive BC subtypes, with elevated expression correlating with higher tumor grade and stage. G9a inhibition impaired cell proliferation and modulated epithelial-mesenchymal transition pathways, with the strongest impact in basal-like TNBC. The disruption of the oncogene and tumor suppressor regulation (e.g., TP53, SATB1) was observed in TNBC. This study highlights G9a's context-dependent roles in BC, supporting its potential as a therapeutic target. The findings provide a foundation for subtype-specific epigenetic therapies to improve outcomes in aggressive BC subtypes.
Collapse
Affiliation(s)
- Zirong Huo
- School of Life Science, Northwest University, Xi’an 710069, China; (Z.H.); (S.Z.); (G.S.); (Y.C.); (R.C.); (M.J.); (D.Y.); (S.Z.); (Y.X.)
| | - Sitong Zhang
- School of Life Science, Northwest University, Xi’an 710069, China; (Z.H.); (S.Z.); (G.S.); (Y.C.); (R.C.); (M.J.); (D.Y.); (S.Z.); (Y.X.)
| | - Guodong Su
- School of Life Science, Northwest University, Xi’an 710069, China; (Z.H.); (S.Z.); (G.S.); (Y.C.); (R.C.); (M.J.); (D.Y.); (S.Z.); (Y.X.)
| | - Yu Cai
- School of Life Science, Northwest University, Xi’an 710069, China; (Z.H.); (S.Z.); (G.S.); (Y.C.); (R.C.); (M.J.); (D.Y.); (S.Z.); (Y.X.)
| | - Rui Chen
- School of Life Science, Northwest University, Xi’an 710069, China; (Z.H.); (S.Z.); (G.S.); (Y.C.); (R.C.); (M.J.); (D.Y.); (S.Z.); (Y.X.)
| | - Mengju Jiang
- School of Life Science, Northwest University, Xi’an 710069, China; (Z.H.); (S.Z.); (G.S.); (Y.C.); (R.C.); (M.J.); (D.Y.); (S.Z.); (Y.X.)
| | - Dongyan Yang
- School of Life Science, Northwest University, Xi’an 710069, China; (Z.H.); (S.Z.); (G.S.); (Y.C.); (R.C.); (M.J.); (D.Y.); (S.Z.); (Y.X.)
| | - Shengchao Zhang
- School of Life Science, Northwest University, Xi’an 710069, China; (Z.H.); (S.Z.); (G.S.); (Y.C.); (R.C.); (M.J.); (D.Y.); (S.Z.); (Y.X.)
| | - Yuyan Xiong
- School of Life Science, Northwest University, Xi’an 710069, China; (Z.H.); (S.Z.); (G.S.); (Y.C.); (R.C.); (M.J.); (D.Y.); (S.Z.); (Y.X.)
| | - Xi Zhang
- School of Life Science, Northwest University, Xi’an 710069, China; (Z.H.); (S.Z.); (G.S.); (Y.C.); (R.C.); (M.J.); (D.Y.); (S.Z.); (Y.X.)
- School of Professional Studies, Northwestern University, Evanston, IL 60201, USA
| |
Collapse
|
4
|
Galassi C, Esteller M, Vitale I, Galluzzi L. Epigenetic control of immunoevasion in cancer stem cells. Trends Cancer 2024; 10:1052-1071. [PMID: 39244477 DOI: 10.1016/j.trecan.2024.08.004] [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: 03/30/2024] [Revised: 07/24/2024] [Accepted: 08/12/2024] [Indexed: 09/09/2024]
Abstract
Cancer stem cells (CSCs) are a poorly differentiated population of malignant cells that (at least in some neoplasms) is responsible for tumor progression, resistance to therapy, and disease relapse. According to a widely accepted model, all stages of cancer progression involve the ability of neoplastic cells to evade recognition or elimination by the host immune system. In line with this notion, CSCs are not only able to cope with environmental and therapy-elicited stress better than their more differentiated counterparts but also appear to better evade tumor-targeting immune responses. We summarize epigenetic modifications of DNA and histones through which CSCs evade immune recognition or elimination, and propose that such alterations constitute promising therapeutic targets to increase the sensitivity of some malignancies to immunotherapy.
Collapse
Affiliation(s)
- Claudia Galassi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Manel Esteller
- Cancer Epigenetics Group, Josep Carreras Leukaemia Research Institute (IJC), Barcelona, Spain; Centro de Investigacion Biomedica en Red Cancer (CIBERONC), Madrid, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain; Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona (UB), Barcelona, Catalonia, Spain
| | - Ilio Vitale
- Italian Institute for Genomic Medicine, Istituto di Ricovero e Cura a Carattere Scientifico (IRCSS) Candiolo, Torino, Italy; Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Italy.
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA; Sandra and Edward Meyer Cancer Center, New York, NY, USA; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.
| |
Collapse
|
5
|
Saqirile, Deng Y, Li K, Yan W, Li K, Wang C. Gene Expression Regulation and the Signal Transduction of Programmed Cell Death. Curr Issues Mol Biol 2024; 46:10264-10298. [PMID: 39329964 DOI: 10.3390/cimb46090612] [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: 07/30/2024] [Revised: 09/13/2024] [Accepted: 09/14/2024] [Indexed: 09/28/2024] Open
Abstract
Cell death is of great significance in maintaining tissue homeostasis and bodily functions. With considerable research coming to the fore, it has been found that programmed cell death presents in multiple modalities in the body, which is not only limited to apoptosis, but also can be divided into autophagy, pyroptosis, ferroptosis, mitotic catastrophe, entosis, netosis, and other ways. Different forms of programmed cell death have disparate or analogous characteristics with each other, and their occurrence is accompanied by multiple signal transduction and the role of a myriad of regulatory factors. In recent years, scholars across the world have carried out considerable in-depth research on programmed cell death, and new forms of cell death are being discovered continually. Concomitantly, the mechanisms of intricate signaling pathways and regulators have been discovered. More critically, cancer cells tend to choose distinct ways to evade cell death, and different tumors adapt to different manners of death. Therefore, targeting the cell death network has been regarded as an effective tumor treatment strategy for a long time. The objective of our paper is to review the signaling pathways and gene regulation in several typical types of programmed cell death and their correlation with cancer.
Collapse
Affiliation(s)
- Saqirile
- Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, 49 Xilingol South Road, Yu Quan District, Hohhot 010020, China
| | - Yuxin Deng
- Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, 49 Xilingol South Road, Yu Quan District, Hohhot 010020, China
| | - Kexin Li
- Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, 49 Xilingol South Road, Yu Quan District, Hohhot 010020, China
| | - Wenxin Yan
- Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, 49 Xilingol South Road, Yu Quan District, Hohhot 010020, China
| | - Ke Li
- Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, 49 Xilingol South Road, Yu Quan District, Hohhot 010020, China
| | - Changshan Wang
- Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, 49 Xilingol South Road, Yu Quan District, Hohhot 010020, China
| |
Collapse
|
6
|
Al-Ruwishan A, Amer B, Salem A, Abdi A, Chimpandu N, Esa A, Melemenis A, Saleem MZ, Mathew R, Gamallat Y. Advancements in Understanding the Hide-and-Seek Strategy of Hibernating Breast Cancer Cells and Their Implications in Oncology from a Broader Perspective: A Comprehensive Overview. Curr Issues Mol Biol 2024; 46:8340-8367. [PMID: 39194709 DOI: 10.3390/cimb46080492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/27/2024] [Accepted: 07/29/2024] [Indexed: 08/29/2024] Open
Abstract
Despite recent advancements in technology, breast cancer still poses a significant threat, often resulting in fatal consequences. While early detection and treatments have shown some promise, many breast cancer patients continue to struggle with the persistent fear of the disease returning. This fear is valid, as breast cancer cells can lay dormant for years before remerging, evading traditional treatments like a game of hide and seek. The biology of these dormant breast cancer cells presents a crucial yet poorly understood challenge in clinical settings. In this review, we aim to explore the mysterious world of dormant breast cancer cells and their significant impact on patient outcomes and prognosis. We shed light on the elusive role of the G9a enzyme and many other epigenetic factors in breast cancer recurrence, highlighting its potential as a target for eliminating dormant cancer cells and preventing disease relapse. Through this comprehensive review, we not only emphasise the urgency of unravelling the dynamics of dormant breast cancer cells to improve patient outcomes and advance personalised oncology but also provide a guide for fellow researchers. By clearly outlining the clinical and research gaps surrounding dormant breast cancer cells from a molecular perspective, we aim to inspire further exploration of this critical area, ultimately leading to improved patient care and treatment strategies.
Collapse
Affiliation(s)
- Aiman Al-Ruwishan
- Space for Research Initiative, Research Horizons, London NW10 2PU, UK
| | - Bushra Amer
- Department of Family Medicine, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Ahmed Salem
- Department of Biological and Biochemical Sciences, Faculty of Chemical Technology, University of Pardubice, 53210 Pardubice, Czech Republic
| | - Ahmed Abdi
- Independent Researcher, Uxbridge UB9 6JH, UK
| | | | | | | | - Muhammad Zubair Saleem
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Roselit Mathew
- Department of Oncology, Biochemistry and Molecular Biology, and Laboratory Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Yaser Gamallat
- Department of Oncology, Biochemistry and Molecular Biology, and Laboratory Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| |
Collapse
|
7
|
Wang X, Chen Z, Tang J, Cao J. Identification and Validation of a Necroptosis-Related Prognostic Model in Tumor Recurrence and Tumor Immune Microenvironment in Breast Cancer Management. J Inflamm Res 2024; 17:5057-5076. [PMID: 39081870 PMCID: PMC11288355 DOI: 10.2147/jir.s460551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 07/02/2024] [Indexed: 08/02/2024] Open
Abstract
Background Breast cancer is the leading cause of cancer-related death in women. Necroptosis, a form of programmed necrotic cell death, occurs in many solid tumors, including breast cancer, and influences anti-tumor immunity. The role of necroptosis in managing breast cancer recurrence remains unclear. Methods Gene expression profiles and clinical data of breast cancer patients were obtained from the GEO (GSE20685, GSE21653, GSE25055) and TCGA databases. Data analysis and visualization were performed using R. Unsupervised Consensus Clustering and LASSO-COX regression stratified breast cancer patients. GO, KEGG, GSVA, ESTIMATE, and ROC analyses were used to investigate necroptotic signatures. In vitro and in vivo experiments validated necroptosis's role in breast cancer immunity. Results The potential function of necroptotic signature in immunity was first indicated with GO analysis in BRCA cohort. Next, two prognostic models based on the necroptotic profiles both suggested a link between low-risk group with a particular necroptotic immune signature. And a variety of immune cells and immune pathways were shown to be positively associated with a patient's risk score. As an altered immune checkpoint pattern was observed after regulating necroptotic genes, where TIM-3 and LAGLS9 elevated significantly in low-risk group, further validation in vitro and in vivo demonstrated that manipulating a subset of necroptotic gene set could sensitize tumor response to the co-blockade immunotherapy of anti-TIM-3 and anti-PD-1. Conclusion We demonstrated two strategies to stratify breast cancer patients based on their necroptotic profiles and showed that necroptotic signature could assign patients with different tumor immune microenvironment patterns and different recurrence-related prognosis. A subset of necroptotic gene set, composed of TLR3, RIPK3, NLRP3, CASP1, ALDH2 and EZH2, was identified as a biomarker set for predicting immunotherapy-response and recurrence-related prognosis. Targeting necroptosis could helpfacilitate the development of novel breast cancer treatments and tailor personalized medical treatment.
Collapse
Affiliation(s)
- Xiaobo Wang
- Department of General Surgery, the Second Xiangya Hospital of Central South University, Changsha, Hunan, People’s Republic of China
| | - Zongyao Chen
- Department of General Surgery, the Second Xiangya Hospital of Central South University, Changsha, Hunan, People’s Republic of China
| | - Jianing Tang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Department of Liver Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Jing Cao
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Multidisciplinary Breast Cancer Center, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Clinical Research Center for Breast Cancer in Hunan Province, Changsha, Hunan, People’s Republic of China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, People’s Republic of China
| |
Collapse
|
8
|
Pollin G, Mathison AJ, de Assuncao TM, Thomas A, Zeighami A, Salmonson A, Liu H, Urrutia G, Vankayala P, Pandol SJ, Hong JC, Zimmermann MT, Iovanna J, Jin VX, Urrutia R, Lomberk G. Ehmt2 inactivation in pancreatic epithelial cells shapes the transcriptional landscape and inflammation response of the whole pancreas. Front Genet 2024; 15:1412767. [PMID: 38948355 PMCID: PMC11211573 DOI: 10.3389/fgene.2024.1412767] [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: 04/05/2024] [Accepted: 05/17/2024] [Indexed: 07/02/2024] Open
Abstract
Introduction: The Euchromatic Histone Methyl Transferase Protein 2 (EHMT2), also known as G9a, deposits transcriptionally repressive chromatin marks that play pivotal roles in the maturation and homeostasis of multiple organs. Recently, we have shown that Ehmt2 inactivation in the mouse pancreas alters growth and immune gene expression networks, antagonizing Kras-mediated pancreatic cancer initiation and promotion. Here, we elucidate the essential role of Ehmt2 in maintaining a transcriptional landscape that protects organs from inflammation. Methods: Comparative RNA-seq studies between normal postnatal and young adult pancreatic tissue from Ehmt2 conditional knockout animals (Ehmt2 fl/fl ) targeted to the exocrine pancreatic epithelial cells (Pdx1-Cre and P48 Cre/+ ), reveal alterations in gene expression networks in the whole organ related to injury-inflammation-repair, suggesting an increased predisposition to damage. Thus, we induced an inflammation repair response in the Ehmt2 fl/fl pancreas and used a data science-based approach to integrate RNA-seq-derived pathways and networks, deconvolution digital cytology, and spatial transcriptomics. We also analyzed the tissue response to damage at the morphological, biochemical, and molecular pathology levels. Results and discussion: The Ehmt2 fl/fl pancreas displays an enhanced injury-inflammation-repair response, offering insights into fundamental molecular and cellular mechanisms involved in this process. More importantly, these data show that conditional Ehmt2 inactivation in exocrine cells reprograms the local environment to recruit mesenchymal and immunological cells needed to mount an increased inflammatory response. Mechanistically, this response is an enhanced injury-inflammation-repair reaction with a small contribution of specific Ehmt2-regulated transcripts. Thus, this new knowledge extends the mechanisms underlying the role of the Ehmt2-mediated pathway in suppressing pancreatic cancer initiation and modulating inflammatory pancreatic diseases.
Collapse
Affiliation(s)
- Gareth Pollin
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
- Division of Research, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Angela J. Mathison
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
- Division of Research, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Thiago M. de Assuncao
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
- Division of Research, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Anju Thomas
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
- Division of Research, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Atefeh Zeighami
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Ann Salmonson
- Division of Research, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Hongfei Liu
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Guillermo Urrutia
- Division of Research, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Pallavi Vankayala
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Stephen J. Pandol
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Johnny C. Hong
- Division of Transplantation, Department of Surgery, College of Medicine, Pennsylvania State University, Hershey, PA, United States
| | - Michael T. Zimmermann
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, United States
- Clinical and Translational Sciences Institute, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Juan Iovanna
- Centre de Recherche en Cancérologie de Marseille (CRCM), Institut National de la Santé et de la Recherche médicale (INSERM) U1068, CNRS UMR 7258, Parc Scientifique et Technologique de Luminy, Aix-Marseille Université and Institut Paoli-Calmettes, Marseille, France
| | - Victor X. Jin
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
- Division of Biostatistics, Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Raul Urrutia
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
- Division of Research, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Gwen Lomberk
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
- Division of Research, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
9
|
Bel’skaya LV, Dyachenko EI. Oxidative Stress in Breast Cancer: A Biochemical Map of Reactive Oxygen Species Production. Curr Issues Mol Biol 2024; 46:4646-4687. [PMID: 38785550 PMCID: PMC11120394 DOI: 10.3390/cimb46050282] [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: 04/05/2024] [Revised: 05/08/2024] [Accepted: 05/11/2024] [Indexed: 05/25/2024] Open
Abstract
This review systematizes information about the metabolic features of breast cancer directly related to oxidative stress. It has been shown those redox changes occur at all levels and affect many regulatory systems in the human body. The features of the biochemical processes occurring in breast cancer are described, ranging from nonspecific, at first glance, and strictly biochemical to hormone-induced reactions, genetic and epigenetic regulation, which allows for a broader and deeper understanding of the principles of oncogenesis, as well as maintaining the viability of cancer cells in the mammary gland. Specific pathways of the activation of oxidative stress have been studied as a response to the overproduction of stress hormones and estrogens, and specific ways to reduce its negative impact have been described. The diversity of participants that trigger redox reactions from different sides is considered more fully: glycolytic activity in breast cancer, and the nature of consumption of amino acids and metals. The role of metals in oxidative stress is discussed in detail. They can act as both co-factors and direct participants in oxidative stress, since they are either a trigger mechanism for lipid peroxidation or capable of activating signaling pathways that affect tumorigenesis. Special attention has been paid to the genetic and epigenetic regulation of breast tumors. A complex cascade of mechanisms of epigenetic regulation is explained, which made it possible to reconsider the existing opinion about the triggers and pathways for launching the oncological process, the survival of cancer cells and their ability to localize.
Collapse
Affiliation(s)
- Lyudmila V. Bel’skaya
- Biochemistry Research Laboratory, Omsk State Pedagogical University, 644099 Omsk, Russia;
| | | |
Collapse
|
10
|
Pollin G, Mathison AJ, de Assuncao TM, Thomas A, Zeighami L, Salmonson A, Liu H, Urrutia G, Vankayala P, Pandol SJ, Zimmermann MT, Iovanna J, Jin VX, Urrutia R, Lomberk G. EHMT2 Inactivation in Pancreatic Epithelial Cells Shapes the Transcriptional Landscape and Inflammation Response of the Whole Pancreas. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.14.584700. [PMID: 38529489 PMCID: PMC10962735 DOI: 10.1101/2024.03.14.584700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
The Euchromatic Histone Methyl Transferase Protein 2 (EHMT2), also known as G9a, deposits transcriptionally repressive chromatin marks that play pivotal roles in the maturation and homeostasis of multiple organs. Recently, we have shown that EHMT2 inactivation alters growth and immune gene expression networks, antagonizing KRAS-mediated pancreatic cancer initiation and promotion. Here, we elucidate the essential role of EHMT2 in maintaining a transcriptional landscape that protects organs from inflammation. Comparative RNA-seq studies between normal postnatal and young adult pancreatic tissue from EHMT2 conditional knockout animals ( EHMT2 fl/fl ) targeted to the exocrine pancreatic epithelial cells ( Pdx1-Cre and P48 Cre/+ ), reveal alterations in gene expression networks in the whole organ related to injury-inflammation-repair, suggesting an increased predisposition to damage. Thus, we induced an inflammation repair response in the EHMT2 fl/fl pancreas and used a data science-based approach to integrate RNA-seq-derived pathways and networks, deconvolution digital cytology, and spatial transcriptomics. We also analyzed the tissue response to damage at the morphological, biochemical, and molecular pathology levels. The EHMT2 fl/fl pancreas displays an enhanced injury-inflammation-repair response, offering insights into fundamental molecular and cellular mechanisms involved in this process. More importantly, these data show that conditional EHMT2 inactivation in exocrine cells reprograms the local environment to recruit mesenchymal and immunological cells needed to mount an increased inflammatory response. Mechanistically, this response is an enhanced injury-inflammation-repair reaction with a small contribution of specific EHMT2-regulated transcripts. Thus, this new knowledge extends the mechanisms underlying the role of the EHMT2-mediated pathway in suppressing pancreatic cancer initiation and modulating inflammatory pancreatic diseases.
Collapse
|
11
|
Singh J, Sah B, Shen Y, Liu L. Histone methyltransferase inhibitor UNC0642 promotes breast cancer cell death by upregulating TXNIP-dependent oxidative stress. Chem Biol Interact 2023; 385:110720. [PMID: 37748637 DOI: 10.1016/j.cbi.2023.110720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 09/04/2023] [Accepted: 09/18/2023] [Indexed: 09/27/2023]
Abstract
Breast cancer (BC) is one of the most frequent type of cancer in women worldwide. Current therapeutic strategies for BC are not always effective. In this study, we investigated the anticancer activity of an epigenetic compound UNC0642 and its mechanism of action in suppressing BC cell growth and survival. UNC0642 was developed as a selective inhibitor of G9a that is responsible for histone H3K9 methylation. After screening different BC cell lines, we found UNC0642 had the lowest IC-50 against MDA-MB-231 cells, a triple-negative BC cell line. To identify additional UNC0642 targets, we performed RNA-seq analyses in BC cells following UNC0642 treatment. UNC0642 significantly upregulated mRNA expression of thioredoxin-interacting protein (TXNIP), which was also validated by western blotting. We further showed that TXNIP upregulation was associated with dose-dependent elevation of reactive oxygen species, concurrent with loss of mitochondrial membrane potential and activation of caspase-3-dependent apoptosis. Finally, we demonstrated that UNC0642 treatment induced BC cell apoptosis in vitro and suppression of tumor growth in xenograft mouse models that was coupled with TXNIP activation. Taken together, our results show that UNC0642 exerts its antitumor function via upregulating TXNIP expression and oxidative stress to impair mitochondrial function and induce caspase-dependent cell death. This observation could inform future breast cancer therapies by targeting TXNIP-dependent ROS signaling.
Collapse
Affiliation(s)
- Jasvinder Singh
- The Hormel Institute, University of Minnesota, Austin, MN, 55912, USA.
| | - Bindeshwar Sah
- The Hormel Institute, University of Minnesota, Austin, MN, 55912, USA
| | - Yao Shen
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Liang Liu
- The Hormel Institute, University of Minnesota, Austin, MN, 55912, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 55455, USA
| |
Collapse
|
12
|
Chi L, Zhong L, Lee D, Yu X, Caballero A, Nieman B, Delgado-Olguin P. G9a inactivation in progenitor cells with Isl1-Cre with reduced recombinase activity models aspects of Dandy-Walker complex. Biol Open 2023; 12:bio059894. [PMID: 37470706 PMCID: PMC10399207 DOI: 10.1242/bio.059894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 07/10/2023] [Indexed: 07/21/2023] Open
Abstract
G9a, also known as EHMT2, is essential for embryogenesis and has specific functions in multiple developmental processes. G9a inactivation affects development of the nervous system, which is formed with contribution of descendants of progenitor cells expressing the transcription factor Isl1. However, the function of G9a in Isl1-expressing progenitors is unknown. Here, we show that G9a is required for proper development of multiple structures formed with contribution of Isl1-expressing progenitors. A Cre-dependent GFP reporter revealed that the recombinase activity of the Isl1-Cre used in this study to inactivate G9a was reduced to a subset of Isl1-expressing progenitor cells. G9a mutants reached endpoint by 7 weeks of age with cardiac hypertrophy, hydrocephalus, underdeveloped cerebellum and hind limb paralysis, modeling aspects of Dandy-Walker complex. Moreover, neuroepithelium of the lateral ventricle derived from Isl1-expressing progenitors was thinner and disorganized, potentially compromising cerebrospinal fluid dynamics in G9a mutants. Micro-computed tomography after iodine staining revealed increased volume of the heart, eye lens and brain structures in G9a mutant fetuses. Thus, altered development of descendants of the second heart field and the neural crest could contribute to multicomponent malformation like Dandy-Walker.
Collapse
Affiliation(s)
- Lijun Chi
- Translational Medicine, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada
| | - Ling Zhong
- Translational Medicine, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada
- Department of Endocrinology, National Health Committee Key Laboratory of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Dorothy Lee
- Translational Medicine, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada
- Department of Physiology, University of Toronto, Toronto, ON M5S1A8, Canada
| | - Xinwen Yu
- Translational Medicine, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada
| | - Amalia Caballero
- Translational Medicine, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S1A8, Canada
| | - Brian Nieman
- Translational Medicine, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada
- Mouse Imaging Centre (MICe), The Hospital for Sick Children, Toronto, ON M5T3H7, Canada
- Ontario Institute for Cancer Research, Toronto, ON M5G0A3, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G1L7, Canada
| | - Paul Delgado-Olguin
- Translational Medicine, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S1A8, Canada
- Heart and Stroke Richard Lewar Centre of Excellence, Toronto, ON M5S3H2, Canada
| |
Collapse
|
13
|
In vivo metabolic imaging identifies lipid vulnerability in a preclinical model of Her2+/Neu breast cancer residual disease and recurrence. NPJ Breast Cancer 2022; 8:111. [PMID: 36163365 PMCID: PMC9512922 DOI: 10.1038/s41523-022-00481-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 09/16/2022] [Indexed: 11/08/2022] Open
Abstract
Recurrent cancer cells that evade therapy is a leading cause of death in breast cancer patients. This risk is high for women showing an overexpression of human epidermal growth factor receptor 2 (Her2). Cells that persist can rely on different substrates for energy production relative to their primary tumor counterpart. Here, we characterize metabolic reprogramming related to tumor dormancy and recurrence in a doxycycline-induced Her2+/Neu model of breast cancer with varying times to recurrence using longitudinal fluorescence microscopy. Glucose uptake (2-NBDG) and mitochondrial membrane potential (TMRE) imaging metabolically phenotype mammary tumors as they transition to regression, dormancy, and recurrence. “Fast-recurrence” tumors (time to recurrence ~55 days), transition from glycolysis to mitochondrial metabolism during regression and this persists upon recurrence. “Slow-recurrence” tumors (time to recurrence ~100 days) rely on both glycolysis and mitochondrial metabolism during recurrence. The increase in mitochondrial activity in fast-recurrence tumors is attributed to a switch from glucose to fatty acids as the primary energy source for mitochondrial metabolism. Consequently, when fast-recurrence tumors receive treatment with a fatty acid inhibitor, Etomoxir, tumors report an increase in glucose uptake and lipid synthesis during regression. Treatment with Etomoxir ultimately prolongs survival. We show that metabolic reprogramming reports on tumor recurrence characteristics, particularly at time points that are essential for actionable targets. The temporal characteristics of metabolic reprogramming will be critical in determining the use of an appropriate timing for potential therapies; namely, the notion that metabolic-targeted inhibition during regression reports long-term therapeutic benefit.
Collapse
|
14
|
Shen Y, Singh J, Sah B, Chen Z, Ha W, Henzler C, Su T, Xie L, Deng Y, Li G, Guo H, Hibshoosh H, Liu L. The Histone Demethylase HR Suppresses Breast Cancer Development through Enhanced CELF2 Tumor Suppressor Activity. Cancers (Basel) 2022; 14:4648. [PMID: 36230572 PMCID: PMC9564370 DOI: 10.3390/cancers14194648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/21/2022] [Accepted: 09/22/2022] [Indexed: 11/17/2022] Open
Abstract
The hairless (HR) gene encodes a transcription factor with histone demethylase activity that is essential for development and tissue homeostasis. Previous studies suggest that mutational inactivation of HR promotes tumorigenesis. To investigate HR mutations in breast cancer, we performed targeted next-generation sequencing using DNA isolated from primary breast cancer tissues. We identified HR somatic mutations in approximately 15% of the patient cohort (n = 85), compared with 23% for BRCA2, 13% for GATA3, 7% for BRCA1, and 3% for PTEN in the same patient cohort. We also found an average 23% HR copy number loss in breast cancers. In support of HR's antitumor functions, HR reconstitution in HR-deficient human breast cancer cells significantly suppressed tumor growth in orthotopic xenograft mouse models. We further demonstrated that HR's antitumor activity was at least partly mediated by transcriptional activation of CELF2, a tumor suppressor with RNA-binding activity. Consistent with HR's histone demethylase activity, pharmacologic inhibition of histone methylation suppressed HR-deficient breast cancer cell proliferation, migration and tumor growth. Taken together, we identified HR as a novel tumor suppressor that is frequently mutated in breast cancer. We also showed that pharmacologic inhibition of histone methylation is effective in suppressing HR-deficient breast tumor growth and progression.
Collapse
Affiliation(s)
- Yao Shen
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Jasvinder Singh
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Bindeshwar Sah
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Zhongming Chen
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Wootae Ha
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Christine Henzler
- Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Tao Su
- Department of Pathology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Lillian Xie
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Yibin Deng
- Department of Urology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Gen Li
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Hua Guo
- Department of Pathology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Hanina Hibshoosh
- Department of Pathology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Liang Liu
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
15
|
Ang GCK, Gupta A, Surana U, Yap SXL, Taneja R. Potential Therapeutics Targeting Upstream Regulators and Interactors of EHMT1/2. Cancers (Basel) 2022; 14:2855. [PMID: 35740522 PMCID: PMC9221123 DOI: 10.3390/cancers14122855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/07/2022] [Accepted: 06/07/2022] [Indexed: 11/16/2022] Open
Abstract
Euchromatin histone lysine methyltransferases (EHMTs) are epigenetic regulators responsible for silencing gene transcription by catalyzing H3K9 dimethylation. Dysregulation of EHMT1/2 has been reported in multiple cancers and is associated with poor clinical outcomes. Although substantial insights have been gleaned into the downstream targets and pathways regulated by EHMT1/2, few studies have uncovered mechanisms responsible for their dysregulated expression. Moreover, EHMT1/2 interacting partners, which can influence their function and, therefore, the expression of target genes, have not been extensively explored. As none of the currently available EHMT inhibitors have made it past clinical trials, understanding upstream regulators and EHMT protein complexes may provide unique insights into novel therapeutic avenues in EHMT-overexpressing cancers. Here, we review our current understanding of the regulators and interacting partners of EHMTs. We also discuss available therapeutic drugs that target the upstream regulators and binding partners of EHMTs and could potentially modulate EHMT function in cancer progression.
Collapse
Affiliation(s)
- Gareth Chin Khye Ang
- Healthy Longevity Translational Research Program, Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (G.C.K.A.); (A.G.)
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore;
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research A*STAR, 61 Biopolis Drive, Singapore 138673, Singapore
| | - Amogh Gupta
- Healthy Longevity Translational Research Program, Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (G.C.K.A.); (A.G.)
| | - Uttam Surana
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore;
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research A*STAR, 61 Biopolis Drive, Singapore 138673, Singapore
| | - Shirlyn Xue Ling Yap
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore;
| | - Reshma Taneja
- Healthy Longevity Translational Research Program, Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (G.C.K.A.); (A.G.)
| |
Collapse
|
16
|
Huang JL, Chen SY, Lin CS. Targeting Cancer Stem Cells through Epigenetic Modulation of Interferon Response. J Pers Med 2022; 12:jpm12040556. [PMID: 35455671 PMCID: PMC9027081 DOI: 10.3390/jpm12040556] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 03/26/2022] [Accepted: 03/30/2022] [Indexed: 12/12/2022] Open
Abstract
Cancer stem cells (CSCs) are a small subset of cancer cells and are thought to play a critical role in the initiation and maintenance of tumor mass. CSCs exhibit similar hallmarks to normal stem cells, such as self-renewal, differentiation, and homeostasis. In addition, CSCs are equipped with several features so as to evade anticancer mechanisms. Therefore, it is hard to eliminate CSCs by conventional anticancer therapeutics that are effective at clearing bulk cancer cells. Interferons are innate cytokines and are the key players in immune surveillance to respond to invaded pathogens. Interferons are also crucial for adaptive immunity for the killing of specific aliens including cancer cells. However, CSCs usually evolve to escape from interferon-mediated immune surveillance and to shape the niche as a “cold” tumor microenvironment (TME). These CSC characteristics are related to their unique epigenetic regulations that are different from those of normal and bulk cancer cells. In this review, we introduce the roles of epigenetic modifiers, focusing on LSD1, BMI1, G9a, and SETDB1, in contributing to CSC characteristics and discussing the interplay between CSCs and interferon response. We also discuss the emerging strategy for eradicating CSCs by targeting these epigenetic modifiers, which can elevate cytosolic nuclei acids, trigger interferon response, and reshape a “hot” TME for improving cancer immunotherapy. The key epigenetic and immune genes involved in this crosstalk can be used as biomarkers for precision oncology.
Collapse
Affiliation(s)
- Jau-Ling Huang
- Department of Bioscience Technology, College of Health Science, Chang Jung Christian University, Tainan 711, Taiwan;
| | - Si-Yun Chen
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Chang-Shen Lin
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan
- Correspondence:
| |
Collapse
|
17
|
Selective histone methyltransferase G9a inhibition reduces metastatic development of Ewing sarcoma through the epigenetic regulation of NEU1. Oncogene 2022; 41:2638-2650. [PMID: 35354905 PMCID: PMC9054661 DOI: 10.1038/s41388-022-02279-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 03/02/2022] [Accepted: 03/15/2022] [Indexed: 11/08/2022]
Abstract
Ewing sarcoma (EWS) is an aggressive bone and soft tissue tumor with high susceptibility to metastasize. The underlying molecular mechanisms leading to EWS metastases remain poorly understood. Epigenetic changes have been implicated in EWS tumor growth and progression. Linking epigenetics and metastases may provide insight into novel molecular targets in EWS and improve its treatment. Here, we evaluated the effects of a selective G9a histone methyltransferase inhibitor (BIX01294) on EWS metastatic process. Our results showed that overexpression of G9a in tumors from EWS patients correlates with poor prognosis. Moreover, we observe a significantly higher expression of G9a in metastatic EWS tumor as compared to either primary or recurrent tumor. Using functional assays, we demonstrate that pharmacological G9a inhibition using BIX01294 disrupts several metastatic steps in vitro, such as migration, invasion, adhesion, colony formation and vasculogenic mimicry. Moreover, BIX01294 reduces tumor growth and metastases in two spontaneous metastases mouse models. We further identified the sialidase NEU1 as a direct target and effector of G9a in the metastatic process in EWS. NEU1 overexpression impairs migration, invasion and clonogenic capacity of EWS cell lines. Overall, G9a inhibition impairs metastases in vitro and in vivo through the overexpression of NEU1. G9a has strong potential as a prognostic marker and may be a promising therapeutic target for EWS patients.
Collapse
|
18
|
Lin HY, Wu HJ, Chen SY, Hou MF, Lin CS, Chu PY. Epigenetic therapy combination of UNC0638 and CI-994 suppresses breast cancer via epigenetic remodeling of BIRC5 and GADD45A. Biomed Pharmacother 2022; 145:112431. [PMID: 34798471 DOI: 10.1016/j.biopha.2021.112431] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 11/08/2021] [Accepted: 11/12/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND There is currently a growing interest in the roles of epigenetic mechanisms in the diagnosis, prognosis, and therapies associated with precision oncology for breast cancer (BC). This study aimed to demonstrate the clinical significance of euchromatic histone lysine methyltransferase 2 (EHMT2), histone deacetylase 1 (HDAC1) and HDAC2 in BC, to evaluate the antitumor effectiveness of a combination of the selective inhibitors UNC0638 and CI-994 (U+C), and to clarify the underlying mechanisms. METHODS Multi-omic analysis was used to study the clinical significance of the biomarkers of interest. The effects of U+C treatment were evaluated by detecting cell viability, cell cycle, apoptosis, and representative gene expressions. RNA-Seq and Gene Set Enrichment Analysis (GSEA) were employed to identify over-represented genes associated with the treatment. Chromatin immunoprecipitation and qPCR (ChIP-qPCR) assay were applied to verify epigenetic profiling on the identified promoters. RESULTS The significance of elevated expressions of EHMT2, HDAC1, and HDAC2 in tumor tissue and BC basal-like subtype in predicting a poor prognosis was noted. The U+C combined treatment showed an enhanced suppressive effect as compared to single agent treatment, perturbed the cell cycle, induced apoptosis, reduced expressions of the genes representing anti-apoptosis, stemness, drug resistance and basal-like state, while increasing luminal-like state genes. In addition, the combined U+C treatment suppressed xenograft tumor growth. The epigenetic reprogramming of histones was identified in the down-regulated BIRC5 and upregulated GADD45A. CONCLUSION These findings demonstrate that selectively targeting EHMT2, HDAC1, and HDAC2 by concurrent U+C treatment suppresses BC tumor progression via epigenetic remodeling of BIRC5 and GADD45A.
Collapse
Affiliation(s)
- Hung-Yu Lin
- College of Medicine, National Chung Hsing University, Taichung 402, Taiwan; Research Assistant Center, Show Chwan Memorial Hospital, Changhua 500, Taiwan.
| | - Hsing-Ju Wu
- College of Medicine, National Chung Hsing University, Taichung 402, Taiwan; Research Assistant Center, Show Chwan Memorial Hospital, Changhua 500, Taiwan; Department of Biology, National Changhua University of Education, Changhua 500, Taiwan.
| | - Si-Yun Chen
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Department of Pathology, Show Chwan Memorial Hospital, Changhua 500, Taiwan.
| | - Ming-Feng Hou
- Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; Division of Breast Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
| | - Chang-Shen Lin
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; Department of Biological Sciences, National Sun Yet-sen University, Kaohsiung 804, Taiwan.
| | - Pei-Yi Chu
- College of Medicine, National Chung Hsing University, Taichung 402, Taiwan; Department of Pathology, Show Chwan Memorial Hospital, Changhua 500, Taiwan; School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan; Department of Health Food, Chung Chou University of Science and Technology, Changhua 510, Taiwan; National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan.
| |
Collapse
|
19
|
Nachiyappan A, Gupta N, Taneja R. EHMT1/EHMT2 in EMT, Cancer Stemness and Drug Resistance: Emerging Evidence and Mechanisms. FEBS J 2021; 289:1329-1351. [PMID: 34954891 DOI: 10.1111/febs.16334] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 11/25/2021] [Accepted: 12/23/2021] [Indexed: 11/29/2022]
Abstract
Metastasis, therapy failure and tumor recurrence are major clinical challenges in cancer. The interplay between tumor initiating cells (TICs) and Epithelial-Mesenchymal transition (EMT) drives tumor progression and spread. Recent advances have highlighted the involvement of epigenetic deregulation in these processes. The Euchromatin Histone Lysine Methyltransferase 1 (EHMT1) and Euchromatin Histone Lysine Methyltransferase 2 (EHMT2) that primarily mediate histone 3 lysine 9 di-methylation (H3K9me2), as well as methylation of non-histone proteins, are now recognized to be aberrantly expressed in many cancers. Their deregulated expression is associated with EMT, cellular plasticity and therapy resistance. In this review, we summarize evidence of their myriad roles in cancer metastasis, stemness and drug resistance. We discuss cancer-type specific molecular targets, context-dependent mechanisms and future directions of research in targeting EHMT1/EHMT2 for the treatment of cancer.
Collapse
Affiliation(s)
- Alamelu Nachiyappan
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 117593
| | - Neelima Gupta
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 117593
| | - Reshma Taneja
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 117593.,Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, 117593
| |
Collapse
|
20
|
Das D, Karthik N, Taneja R. Crosstalk Between Inflammatory Signaling and Methylation in Cancer. Front Cell Dev Biol 2021; 9:756458. [PMID: 34901003 PMCID: PMC8652226 DOI: 10.3389/fcell.2021.756458] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 10/11/2021] [Indexed: 01/08/2023] Open
Abstract
Inflammation is an intricate immune response against infection and tissue damage. While the initial immune response is important for preventing tumorigenesis, chronic inflammation is implicated in cancer pathogenesis. It has been linked to various stages of tumor development including transformation, proliferation, angiogenesis, and metastasis. Immune cells, through the production of inflammatory mediators such as cytokines, chemokines, transforming growth factors, and adhesion molecules contribute to the survival, growth, and progression of the tumor in its microenvironment. The aberrant expression and secretion of pro-inflammatory and growth factors by the tumor cells result in the recruitment of immune cells, thus creating a mutual crosstalk. The reciprocal signaling between the tumor cells and the immune cells creates and maintains a successful tumor niche. Many inflammatory factors are regulated by epigenetic mechanisms including DNA methylation and histone modifications. In particular, DNA and histone methylation are crucial forms of transcriptional regulation and aberrant methylation has been associated with deregulated gene expression in oncogenesis. Such deregulations have been reported in both solid tumors and hematological malignancies. With technological advancements to study genome-wide epigenetic landscapes, it is now possible to identify molecular mechanisms underlying altered inflammatory profiles in cancer. In this review, we discuss the role of DNA and histone methylation in regulation of inflammatory pathways in human cancers and review the merits and challenges of targeting inflammatory mediators as well as epigenetic regulators in cancer.
Collapse
Affiliation(s)
- Dipanwita Das
- Department of Physiology, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Nandini Karthik
- Department of Physiology, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Reshma Taneja
- Department of Physiology, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
21
|
Liu S, Joshi K, Denning MF, Zhang J. RIPK3 signaling and its role in the pathogenesis of cancers. Cell Mol Life Sci 2021; 78:7199-7217. [PMID: 34654937 PMCID: PMC9044760 DOI: 10.1007/s00018-021-03947-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/04/2021] [Accepted: 09/20/2021] [Indexed: 02/07/2023]
Abstract
RIPK3 (receptor-interacting protein kinase 3) is a serine/threonine-protein kinase. As a key component of necrosomes, RIPK3 is an essential mediator of inflammatory factors (such as TNFα-tumor necrosis factor α) and infection-induced necroptosis, a programmed necrosis. In addition, RIPK3 signaling is also involved in the regulation of apoptosis, cytokine/chemokine production, mitochondrial metabolism, autophagy, and cell proliferation by interacting with and/or phosphorylating the critical regulators of the corresponding signaling pathways. Similar to apoptosis, RIPK3-signaling-mediated necroptosis is inactivated in most types of cancers, suggesting RIPK3 might play a critical suppressive role in the pathogenesis of cancers. However, in some inflammatory types of cancers, such as pancreatic cancers and colorectal cancers, RIPK3 signaling might promote cancer development by stimulating proliferation signaling in tumor cells and inducing an immunosuppressive response in the tumor environment. In this review, we summarize recent research progress in the regulators of RIPK3 signaling, and discuss the function of this pathway in the regulation of mixed lineage kinase domain-like (MLKL)-mediated necroptosis and MLKL-independent cellular behaviors. In addition, we deliberate the potential roles of RIPK3 signaling in the pathogenesis of different types of cancers and discuss the potential strategies for targeting this pathway in cancer therapy.
Collapse
Affiliation(s)
- Shanhui Liu
- Department of Cancer Biology, Oncology Institute, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Maywood, IL, 60153, USA
| | - Kanak Joshi
- Department of Cancer Biology, Oncology Institute, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Maywood, IL, 60153, USA
| | - Mitchell F Denning
- Department of Cancer Biology, Oncology Institute, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Maywood, IL, 60153, USA
| | - Jiwang Zhang
- Department of Cancer Biology, Oncology Institute, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Maywood, IL, 60153, USA.
- Department of Pathology and Department of Radiation Oncology, Loyola University Medical Center, Maywood, IL, 60153, USA.
| |
Collapse
|
22
|
Emerging role of G9a in cancer stemness and promises as a therapeutic target. Oncogenesis 2021; 10:76. [PMID: 34775469 PMCID: PMC8590690 DOI: 10.1038/s41389-021-00370-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 12/13/2022] Open
Abstract
The histone methyltransferase G9a is well-documented for its implication in neoplastic growth. However, recent investigations have demonstrated a key involvement of this chromatin writer in maintaining the self-renewal and tumor-initiating capacities of cancer stem cells (CSCs). Direct inhibition of G9a’s catalytic activity was reported as a promising therapeutic target in multiple preclinical studies. Yet, none of the available pharmacological inhibitors of G9a activity have shown success at the early stages of clinical testing. Here, we discuss central findings of oncogenic expression and activation of G9a in CSCs from different origins, as well as the impact of the suppression of G9a histone methyltransferase activity in such contexts. We will explore the challenges posed by direct and systemic inhibition of G9a activity in the perspective of clinical translation of documented small molecules. Finally, we will discuss recent advances in drug discovery as viable strategies to develop context-specific drugs, selectively targeting G9a in CSC populations.
Collapse
|
23
|
Poulard C, Noureddine LM, Pruvost L, Le Romancer M. Structure, Activity, and Function of the Protein Lysine Methyltransferase G9a. Life (Basel) 2021; 11:life11101082. [PMID: 34685453 PMCID: PMC8541646 DOI: 10.3390/life11101082] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/08/2021] [Accepted: 10/08/2021] [Indexed: 12/17/2022] Open
Abstract
G9a is a lysine methyltransferase catalyzing the majority of histone H3 mono- and dimethylation at Lys-9 (H3K9), responsible for transcriptional repression events in euchromatin. G9a has been shown to methylate various lysine residues of non-histone proteins and acts as a coactivator for several transcription factors. This review will provide an overview of the structural features of G9a and its paralog called G9a-like protein (GLP), explore the biochemical features of G9a, and describe its post-translational modifications and the specific inhibitors available to target its catalytic activity. Aside from its role on histone substrates, the review will highlight some non-histone targets of G9a, in order gain insight into their role in specific cellular mechanisms. Indeed, G9a was largely described to be involved in embryonic development, hypoxia, and DNA repair. Finally, the involvement of G9a in cancer biology will be presented.
Collapse
Affiliation(s)
- Coralie Poulard
- Cancer Research Cancer of Lyon, Université de Lyon, F-69000 Lyon, France; (L.M.N.); (L.P.); (M.L.R.)
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
- Correspondence:
| | - Lara M. Noureddine
- Cancer Research Cancer of Lyon, Université de Lyon, F-69000 Lyon, France; (L.M.N.); (L.P.); (M.L.R.)
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences, Lebanese University, Hadat-Beirut 90565, Lebanon
| | - Ludivine Pruvost
- Cancer Research Cancer of Lyon, Université de Lyon, F-69000 Lyon, France; (L.M.N.); (L.P.); (M.L.R.)
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
| | - Muriel Le Romancer
- Cancer Research Cancer of Lyon, Université de Lyon, F-69000 Lyon, France; (L.M.N.); (L.P.); (M.L.R.)
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
| |
Collapse
|
24
|
Newcomb R, Dean E, McKinney BJ, Alvarez JV. Context-dependent effects of whole-genome duplication during mammary tumor recurrence. Sci Rep 2021; 11:14932. [PMID: 34294755 PMCID: PMC8298634 DOI: 10.1038/s41598-021-94332-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 07/06/2021] [Indexed: 12/24/2022] Open
Abstract
Whole-genome duplication (WGD) generates polyploid cells possessing more than two copies of the genome and is among the most common genetic abnormalities in cancer. The frequency of WGD increases in advanced and metastatic tumors, and WGD is associated with poor prognosis in diverse tumor types, suggesting a functional role for polyploidy in tumor progression. Experimental evidence suggests that polyploidy has both tumor-promoting and suppressing effects, but how polyploidy regulates tumor progression remains unclear. Using a genetically engineered mouse model of Her2-driven breast cancer, we explored the prevalence and consequences of whole-genome duplication during tumor growth and recurrence. While primary tumors in this model are invariably diploid, nearly 40% of recurrent tumors undergo WGD. WGD in recurrent tumors was associated with increased chromosomal instability, decreased proliferation and increased survival in stress conditions. The effects of WGD on tumor growth were dependent on tumor stage. Surprisingly, in recurrent tumor cells WGD slowed tumor formation, growth rate and opposed the process of recurrence, while WGD promoted the growth of primary tumors. These findings highlight the importance of identifying conditions that promote the growth of polyploid tumors, including the cooperating genetic mutations that allow cells to overcome the barriers to WGD tumor cell growth and proliferation.
Collapse
Affiliation(s)
- Rachel Newcomb
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, 27710, USA
| | - Emily Dean
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, 27710, USA
| | - Brock J McKinney
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, 27710, USA
| | - James V Alvarez
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, 27710, USA.
| |
Collapse
|