1
|
Chen W, Duan X, Zhu Z, Han Y, Li Y. Histone H1.4K75 acetylation promotes tumor growth and migration by regulating p53 and ERK1/2 pathway in non-small lung cancer. Biochem Biophys Res Commun 2025; 768:151880. [PMID: 40345010 DOI: 10.1016/j.bbrc.2025.151880] [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: 04/12/2025] [Revised: 04/15/2025] [Accepted: 04/22/2025] [Indexed: 05/11/2025]
Abstract
Histone H1.4 is a member of the linker histone H1 family, and its post-translational modifications (PTMs) are essential for its function. However, the role of H1.4 PTM in cancer development is not fully understood. Here, we report the discovery of a previously uncharacterized acetylation site at lysine 75 (K75) of H1.4 in non-small cell lung cancer (NSCLC). Point mutation of K75 with arginine (H1.4K75R) markedly suppressed cellular viability and migration in A549 and H1299 cells, and inhibit tumor growth in xenografts mouse models. Moreover, RNA-sequencing and Western Blot analyses revealed that H1.4K75 acetylation orchestrates dual regulatory effects, potentiating ERK1/2 signaling while repressing the p53 pathway. In summary, our studies show that H1.4K75 acetylation is essential for cell viability, migration and tumor growth in NSCLC, and may be a therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Wenqi Chen
- School of Life Sciences, Anhui University, Hefei, Anhui Province, China
| | - Xinyue Duan
- School of Life Sciences, Anhui University, Hefei, Anhui Province, China
| | - Zhebiao Zhu
- School of Life Sciences, Anhui University, Hefei, Anhui Province, China
| | - Yajing Han
- School of Life Sciences, Anhui University, Hefei, Anhui Province, China
| | - Yong Li
- School of Life Sciences, Anhui University, Hefei, Anhui Province, China.
| |
Collapse
|
2
|
Liu X, Feng J, Guo M, Chen C, Zhao T, Sun X, Zhang Y. Resetting the aging clock through epigenetic reprogramming: Insights from natural products. Pharmacol Ther 2025; 270:108850. [PMID: 40221101 DOI: 10.1016/j.pharmthera.2025.108850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 12/04/2024] [Accepted: 04/07/2025] [Indexed: 04/14/2025]
Abstract
Epigenetic modifications play a critical role in regulating gene expression under various physiological and pathological conditions. Epigenetic modifications reprogramming is a recognized hallmark of aging and a key component of the aging clock used to differentiate between chronological and biological age. The potential for prospective diagnosis and regulatory capabilities position epigenetic modifications as an emerging drug target to extend longevity and alleviate age-related organ dysfunctions. In the past few decades, numerous preclinical studies have demonstrated the therapeutic potential of natural products in various human diseases, including aging, with some advancing to clinical trials and clinical application. This review highlights the discovery and recent advancements in the aging clock, as well as the potential use of natural products as anti-aging therapeutics by correcting disordered epigenetic reprogramming. Specifically, the focus is on the imbalance of histone modifications, alterations in DNA methylation patterns, disrupted ATP-dependent chromatin remodeling, and changes in RNA modifications. By exploring these areas, new insights can be gained into aging prediction and anti-aging interventions.
Collapse
Affiliation(s)
- Xin Liu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin 150081, China; State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China
| | - Jing Feng
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin 150081, China; State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China
| | - Madi Guo
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin 150081, China; State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China
| | - Chen Chen
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin 150081, China; State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China
| | - Tong Zhao
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin 150081, China; State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China
| | - Xiuxiu Sun
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin 150081, China; State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China
| | - Yong Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin 150081, China; State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China.
| |
Collapse
|
3
|
Xu H, Gao J, Fu S, Liang Z, Zhang Y, Chen H, Zheng Q. Genkwanin impairs triple-negative breast cancer aggressiveness and metastasis by targeting Lysine Demethylase 4C. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156869. [PMID: 40412061 DOI: 10.1016/j.phymed.2025.156869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 05/02/2025] [Accepted: 05/15/2025] [Indexed: 05/27/2025]
Abstract
BACKGROUND Triple-negative breast cancer, the most aggressive breast cancer with highest metastatic capacity and the worst prognosis, accounts for 15 % of breast cancer. According to prior studies, the natural active component genkwanin suppressed cell growth in estrogen-receptor-positive breast cancer. Nonetheless, its impact on triple-negative breast cancer and its target remains poorly understood. PURPOSE To explore the effects of genkwanin and identify the direct targets and cellular mechanism of genkwanin in triple-negative breast cancer. STUDY DESIGN The effects of genkwanin were investigated in SUM-159PT and MDA-MB-231 cells. Direct target protein of genkwanin were identified by Quantitative proteomics. The in-vivo effect of genkwanin on triple-negative breast cancer was investigated using mouse models. METHODS The anti-metastasis effects of genkwanin were investigated by individual cell-tracking assay and 3D-on top assay. Quantitative proteomics was used to identify the target of genkwanin and microscale thermophoresis ligand-binding assay was performed to further confirmed it. In-vitro and in-vivo extreme limiting diluting assays were performed to investigate the inhibitory effect of stemness. Lung metastasis mouse model was used to evaluate the anti-metastasis effects of genkwanin. RESULTS This study demonstrated that genkwanin could significantly attenuate migration and invasion in triple-negative breast cancer cells. Notably, genkwanin could efficiently modulate the stemness of breast cancer cells and improve its sensitivity to paclitaxel (PTX). Lysine demethylase 4C (KDM4C) was identified as the target of genkwanin and the in-vivo therapeutic effect of genkwanin was also confirmed in 4T1-luc lung metastasis model. Mechanically, genkwanin reduced KDM4C expression, which regulated the lysine demethylation of histone H3, thereby suppressing KLF14 transcription. KLF14 acts as a switch of lipid synthesis and regulates intracellular cholesterol levels, inhibiting cancer metastasis. CONCLUSION These results demonstrate genkwanin's therapeutic potential in triple-negative breast cancer and its effect on targeting metastasis.
Collapse
Affiliation(s)
- Hanfeng Xu
- Department of Oncology, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine. Nanjing, 210003, PR China
| | - Jie Gao
- Department of Oncology, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine. Nanjing, 210003, PR China
| | - Shiyong Fu
- Department of Oncology, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine. Nanjing, 210003, PR China
| | - Zihao Liang
- Department of Clinical Research Center, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine. Nanjing, 210003, PR China
| | - Ying Zhang
- Department of Oncology, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine. Nanjing, 210003, PR China
| | - Huijuan Chen
- Department of Oncology, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine. Nanjing, 210003, PR China
| | - Qin Zheng
- Department of Oncology, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine. Nanjing, 210003, PR China.
| |
Collapse
|
4
|
Bueloni B, Garcia Fernandez de Barrena M, Avila MA, Bayo J, Mazzolini G. Epigenetic mechanisms involved in hepatocellular carcinoma development and progression. EGASTROENTEROLOGY 2025; 3:e100186. [PMID: 40432834 PMCID: PMC12107448 DOI: 10.1136/egastro-2025-100186] [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] [Received: 01/13/2025] [Accepted: 03/17/2025] [Indexed: 05/29/2025]
Abstract
Hepatocellular carcinoma (HCC) typically develops in the context of chronic liver disease, where prolonged hepatocyte exposure to inflammation drives the synergistic accumulation of genetic and epigenetic alterations. Epigenetic regulation encompasses multiple mechanisms that govern the transcription machinery accessibility to DNA. This process is regulated by the addition and removal of covalent marks on chromatin, which can either affect DNA-histone interactions or serve as scaffolds for other proteins, among other mechanisms. Recent research has revealed that epigenetic alterations can disrupt chromatin homeostasis, redirecting transcriptional regulation to favour cancer-promoting states. Consequently, these alterations play a pivotal role in the acquisition of cancer hallmarks and provide insights into several biological processes involved in hepatocarcinogenesis. This review highlights the key epigenetic mechanisms underlying the development, progression and dissemination of HCC, with a particular focus on DNA methylation and histone post-translational modifications. This knowledge is relevant for guiding the development of innovative therapeutic approaches based on epigenetic modulators.
Collapse
Affiliation(s)
- Barbara Bueloni
- Hepatology and Gene Therapy Program, Instituto de Investigaciones en Medicina Traslacional, Facultad de Ciencias Biomédicas, Universidad Austral, Pilar, Argentina
- HZ4 Liver Inc./Spectrum, Dover, Delaware, USA
| | - Maite Garcia Fernandez de Barrena
- Solid Tumor Program, Hepatology Laboratory, Applied Medical Research Center (CIMA), University of Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas, Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Matias Antonio Avila
- Solid Tumor Program, Hepatology Laboratory, Applied Medical Research Center (CIMA), University of Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas, Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Juan Bayo
- Hepatology and Gene Therapy Program, Instituto de Investigaciones en Medicina Traslacional, Facultad de Ciencias Biomédicas, Universidad Austral, Pilar, Argentina
- Solid Tumor Program, Hepatology Laboratory, Applied Medical Research Center (CIMA), University of Navarra, Pamplona, Spain
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Guillermo Mazzolini
- Hepatology and Gene Therapy Program, Instituto de Investigaciones en Medicina Traslacional, Facultad de Ciencias Biomédicas, Universidad Austral, Pilar, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Liver Unit, Hospital Universitario Austral, Pilar, Buenos Aires Province, Argentina
| |
Collapse
|
5
|
Liu KY, Leung D. Epigenetic Dysregulation of Retrotransposons in Cancer. Mol Cancer Res 2025; 23:369-378. [PMID: 39945628 DOI: 10.1158/1541-7786.mcr-24-0744] [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: 08/08/2024] [Revised: 01/24/2025] [Accepted: 02/10/2025] [Indexed: 05/03/2025]
Abstract
Approximately 97% of the human genome comprises noncoding sequences, with nearly half originating from transposable elements. Among these, retrotransposons represent a critical subclass that replicates via a "copy-and-paste" mechanism and significantly influences the regulation of host genomes. In both normal and pathologic contexts, retrotransposons contribute to a vast reservoir of regulatory elements that can modulate the expression of genes. If left unchecked, retrotransposons can substantially affect host transcriptional programs and genomic integrity. Therefore, various mechanisms, including epigenetic modifications, have been employed to mitigate their potentially deleterious effects. In diseases such as cancer, the epigenome is often significantly reprogrammed, which can lead to retrotransposon dysregulation. Drawing insights from recent studies conducted in human and murine cells, this review examines how retrotransposons expand the complexity of mammalian genomes, describes the impact of their epigenetic dysregulation on cancer development, and highlights the potential of targeting these sequences for therapeutic strategies.
Collapse
Affiliation(s)
- Kwok Yu Liu
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong SAR, China
| | - Danny Leung
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong SAR, China
- Center for Epigenomics Research, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong SAR, China
| |
Collapse
|
6
|
Jin M, Huang Y, Li B, Wang Y, Li Y, Chen Z, Tang Z, Liu C, Zhang L, Yuan X, Tian J, Liu B. Genetic Regulation of Alternative Polyadenylation Provides Novel Insights into Molecular Mechanisms Underlying Non-small Cell Lung Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2502008. [PMID: 40285671 DOI: 10.1002/advs.202502008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 03/30/2025] [Indexed: 04/29/2025]
Abstract
Emerging evidence emphasizes the critical role of alternative polyadenylation (APA) in posttranscriptional regulation of genes, and APA-associated genetic variants (apaQTLs) show particular relevance for multiple disease. However, genetic regulation of APA and its role in non-small cell lung cancer (NSCLC) risk have not been thoroughly studied. Here, by leveraging genotype and APA data from The Cancer Genome Atlas, the association between genetic variation and APA is determined in NSCLC samples. The identified apaQTLs are distinct from eQTLs and are preferentially enriched in functionally relevant characteristics, including poly(A) motifs, APA-associated RBP binding sites, functional elements, and known NSCLC risk loci. Moreover, genes associated with apaQTLs are broadly involved in cancer-related biological process. Of note, integration of apaQTL variants with traditional GWAS-derived PRS is proved as a potential screening tool for NSCLC. By integrating large-scale population and biological experiments, a functional apaQTL variant rs9606 in LYRM4 is identified. Mechanistically, rs9606 induces aberrant APA process of LYRM4 via allele-specific interacting with NUDT21, which lead to increased expression of oncogene LYRM4 and thus contribute to NSCLC risk. This study demonstrates the distinct contribution of APA-associated genetic variants in NSCLC risk, providing critical clues and potential targets for NSCLC etiology and clinical intervention.
Collapse
Affiliation(s)
- Meng Jin
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Yongbiao Huang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Bin Li
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Yuan Wang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Yan Li
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Zhirui Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Zhe Tang
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Chaofan Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Lei Zhang
- Department of Pulmonary and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Jianbo Tian
- Department of Epidemiology and Biostatistics, School of Public Health, Department of Gastrointestinal Oncology, Zhongnan Hospital of Wuhan University, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Bo Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| |
Collapse
|
7
|
Ratinho L, Meyer N, Greive S, Cressiot B, Pelta J. Nanopore sensing of protein and peptide conformation for point-of-care applications. Nat Commun 2025; 16:3211. [PMID: 40180898 PMCID: PMC11968944 DOI: 10.1038/s41467-025-58509-8] [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: 09/07/2024] [Accepted: 03/25/2025] [Indexed: 04/05/2025] Open
Abstract
The global population's aging and growth will likely result in an increase in chronic aging-related diseases. Early diagnosis could improve the medical care and quality of life. Many diseases are linked to misfolding or conformational changes in biomarker peptides and proteins, which affect their function and binding properties. Current clinical methods struggle to detect and quantify these changes. Therefore, there is a need for sensitive conformational sensors that can detect low-concentration analytes in biofluids. Nanopore electrical detection has shown potential in sensing subtle protein and peptide conformation changes. This technique can detect single molecules label-free while distinguishing shape or physicochemical property changes. Its proven sensitivity makes nanopore sensing technology promising for ultra-sensitive, personalized point-of-care devices. We focus on the capability of nanopore sensing for detecting and quantifying conformational modifications and enantiomers in biomarker proteins and peptides and discuss this technology as a solution to future societal health challenges.
Collapse
Affiliation(s)
- Laura Ratinho
- Université Paris-Saclay, Univ Evry, CY Cergy Paris Université, CNRS, LAMBE, Cergy, France
| | - Nathan Meyer
- Université Paris-Saclay, Univ Evry, CY Cergy Paris Université, CNRS, LAMBE, Cergy, France
| | | | - Benjamin Cressiot
- Université Paris-Saclay, Univ Evry, CY Cergy Paris Université, CNRS, LAMBE, Cergy, France.
| | - Juan Pelta
- Université Paris-Saclay, Univ Evry, CY Cergy Paris Université, CNRS, LAMBE, Evry-Courcouronnes, France.
| |
Collapse
|
8
|
Wu N, Sun Q, Yang L, Sun H, Zhou Z, Hu Q, Li C, Wang D, Zhang L, Hu Y, Cong X. HDAC3 and Snail2 complex promotes melanoma metastasis by epigenetic repression of IGFBP3. Int J Biol Macromol 2025; 300:140310. [PMID: 39864688 DOI: 10.1016/j.ijbiomac.2025.140310] [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: 09/11/2024] [Revised: 01/09/2025] [Accepted: 01/23/2025] [Indexed: 01/28/2025]
Abstract
The treatment of metastatic melanoma has long posed a complex challenge within clinical practice. Previous studies have found that EMT transcription factors are essential in the development of various cancers through their induction of EMT. Here, we demonstrate that Snail2 expression is dramatically increased in melanoma and is associated with an adverse prognosis. Elevated Snail2 in melanoma cells enhanced migratory and invasive capabilities in vitro and in vivo. Furthermore, RNA-Seq analysis revealed a significant reduction of IGFBP3 expression in melanoma cells overexpressing Snail2. IGFBP3 might mitigate the Snail2's ability to promote melanoma metastasis via the PI3K-AKT pathway. Moreover, Snail2 and HDAC3 collaborate to suppress IGFBP3 transcription through H3K4 deacetylation and H4K5 delactylation. Additionally, the combination of HDAC3 and p-GSK-3β inhibitors significantly improved the treatment outcomes for lung metastasis in melanoma in vivo. The results of our study indicate that Snail2, HDAC3, and IGFBP3 play significant roles in melanoma progression and represent promising therapeutic targets.
Collapse
Affiliation(s)
- Nan Wu
- Department of Biobank, China-Japan Union Hospital of Jilin University, Changchun 130033, China; Phase I Clinical Trial Research Laboratory, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Qian Sun
- Department of Biobank, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Liehao Yang
- Department of Biobank, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Hongyan Sun
- Department of Biobank, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Zilong Zhou
- Department of Biobank, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Qianying Hu
- Department of Biobank, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Chunyi Li
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun 130033, China
| | - Dongxu Wang
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun 130033, China
| | - Ling Zhang
- Key Laboratory of Pathobiology, Ministry of Education, and Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Yue Hu
- Department of Biobank, China-Japan Union Hospital of Jilin University, Changchun 130033, China.
| | - Xianling Cong
- Department of Biobank, China-Japan Union Hospital of Jilin University, Changchun 130033, China.
| |
Collapse
|
9
|
Chen Z, Peng C, Jin C, Wang Y, Wang T, Yang P, Peng W, Sun Q, Xu H, Nie H, Wang X, Tang J, Sun Y, Feng Y. PJA2 Suppresses Colorectal Cancer Progression by Controlling HDAC2 Degradation and Stability. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2401964. [PMID: 39928532 PMCID: PMC11967759 DOI: 10.1002/advs.202401964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 01/11/2025] [Indexed: 02/12/2025]
Abstract
PJA2 is documented to degrade various substrates. Nevertheless, the role of PJA2 as an E3 ubiquitin-protein ligase in colorectal cancer (CRC) progression remains unexplored. The correlation between PJA2 mRNA levels and clinical characteristics is investigated using data from The Cancer Genome Atlas (TCGA) database. Quantitative real-time polymerase chain reaction (qRT-PCR) and immunohistochemistry (IHC) are utilized to evaluate PJA2 expression levels in CRC tissues. The biological functions of PJA2 are confirmed through colony formation assays and azoxymethane/dextran sulfate sodium (AOM/DSS) mouse model of CRC, among other experimental approaches. The underlying molecular mechanisms of PJA2 action are elucidated using RNA sequencing (RNA-seq), co-immunoprecipitation (co-IP), proximity ligation assay (PLA), and chromatin immunoprecipitation (ChIP). Our research discovered that PJA2 is downregulated in CRC tissues and decreased PJA2 expression correlates with poor prognosis. Functionally, in vivo and in vitro experiments uncovered that PJA2 inhibits tumor cell proliferation and promotes apoptosis. Mechanistically, PJA2 recognized histone deacetylase 2 (HDAC2) via its RING-B-box domain (RBD) and bind to the N-terminal of HDAC2, facilitating ubiquitination at the lysine 90 (K90) residue. PJA2-mediated degradation of HDAC2 counteracts the transcriptional repression of the interferon-induced protein with the tetratricopeptide repeats (IFIT) family, thereby suppressing CRC progression. The data demonstrates that PJA2 suppresses CRC progression through the PJA2/HDAC2/IFIT axis, and its expression is regulated by HDAC2, thus constituting a positive feedback loop. Consequently, PJA2 may serve as a potential therapeutic target for CRC, and interrupting this feedback loop can represent a viable treatment strategy to restrain CRC progression.
Collapse
Affiliation(s)
- Zhihao Chen
- Department of General SurgeryColorectal Institute of Nanjing Medical UniversityThe First Affiliated Hospital of Nanjing Medical UniversityNanjing210029P. R. China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational MedicineNanjingP. R. China
- The First School of Clinical MedicineNanjing Medical UniversityNanjingP. R. China
| | - Chaofan Peng
- Department of General SurgeryColorectal Institute of Nanjing Medical UniversityThe First Affiliated Hospital of Nanjing Medical UniversityNanjing210029P. R. China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational MedicineNanjingP. R. China
- The First School of Clinical MedicineNanjing Medical UniversityNanjingP. R. China
| | - Chi Jin
- Department of General SurgeryColorectal Institute of Nanjing Medical UniversityThe First Affiliated Hospital of Nanjing Medical UniversityNanjing210029P. R. China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational MedicineNanjingP. R. China
- The First School of Clinical MedicineNanjing Medical UniversityNanjingP. R. China
| | - Ye Wang
- Department of General SurgeryColorectal Institute of Nanjing Medical UniversityThe First Affiliated Hospital of Nanjing Medical UniversityNanjing210029P. R. China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational MedicineNanjingP. R. China
- The First School of Clinical MedicineNanjing Medical UniversityNanjingP. R. China
| | - Tuo Wang
- Department of General SurgeryColorectal Institute of Nanjing Medical UniversityThe First Affiliated Hospital of Nanjing Medical UniversityNanjing210029P. R. China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational MedicineNanjingP. R. China
- The First School of Clinical MedicineNanjing Medical UniversityNanjingP. R. China
| | - Peng Yang
- Department of General SurgeryColorectal Institute of Nanjing Medical UniversityThe First Affiliated Hospital of Nanjing Medical UniversityNanjing210029P. R. China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational MedicineNanjingP. R. China
- The First School of Clinical MedicineNanjing Medical UniversityNanjingP. R. China
| | - Wen Peng
- Department of General SurgeryColorectal Institute of Nanjing Medical UniversityThe First Affiliated Hospital of Nanjing Medical UniversityNanjing210029P. R. China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational MedicineNanjingP. R. China
- The First School of Clinical MedicineNanjing Medical UniversityNanjingP. R. China
| | - Qingyang Sun
- Department of General SurgeryColorectal Institute of Nanjing Medical UniversityThe First Affiliated Hospital of Nanjing Medical UniversityNanjing210029P. R. China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational MedicineNanjingP. R. China
- The First School of Clinical MedicineNanjing Medical UniversityNanjingP. R. China
| | - Hengjie Xu
- Department of General SurgeryColorectal Institute of Nanjing Medical UniversityThe First Affiliated Hospital of Nanjing Medical UniversityNanjing210029P. R. China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational MedicineNanjingP. R. China
- The First School of Clinical MedicineNanjing Medical UniversityNanjingP. R. China
| | - Hongxu Nie
- Department of General SurgeryColorectal Institute of Nanjing Medical UniversityThe First Affiliated Hospital of Nanjing Medical UniversityNanjing210029P. R. China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational MedicineNanjingP. R. China
- The First School of Clinical MedicineNanjing Medical UniversityNanjingP. R. China
| | - Xiaowei Wang
- Department of General SurgeryColorectal Institute of Nanjing Medical UniversityThe First Affiliated Hospital of Nanjing Medical UniversityNanjing210029P. R. China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational MedicineNanjingP. R. China
- The First School of Clinical MedicineNanjing Medical UniversityNanjingP. R. China
| | - Junwei Tang
- Department of General SurgeryColorectal Institute of Nanjing Medical UniversityThe First Affiliated Hospital of Nanjing Medical UniversityNanjing210029P. R. China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational MedicineNanjingP. R. China
- The First School of Clinical MedicineNanjing Medical UniversityNanjingP. R. China
| | - Yueming Sun
- Department of General SurgeryColorectal Institute of Nanjing Medical UniversityThe First Affiliated Hospital of Nanjing Medical UniversityNanjing210029P. R. China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational MedicineNanjingP. R. China
- The First School of Clinical MedicineNanjing Medical UniversityNanjingP. R. China
| | - Yifei Feng
- Department of General SurgeryColorectal Institute of Nanjing Medical UniversityThe First Affiliated Hospital of Nanjing Medical UniversityNanjing210029P. R. China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational MedicineNanjingP. R. China
- The First School of Clinical MedicineNanjing Medical UniversityNanjingP. R. China
| |
Collapse
|
10
|
Lu C, Kang T, Zhang J, Yang K, Liu Y, Song K, Lin Q, Dixit D, Gimple RC, Zhang Q, Shi Z, Fan X, Wu Q, Li D, Shan D, Gao J, Gu D, You H, Li Y, Yang J, Zhao L, Qiu Z, Yang H, Zhao N, Gao W, Tao W, Lu Y, Chen Y, Ji J, Zhu Z, Kang C, Man J, Agnihotri S, Wang Q, Lin F, Qian X, Mack SC, Hu Z, Li C, Taylor MD, Liu N, Zhang N, Lu M, You Y, Rich JN, Zhang W, Wang X. Combined targeting of glioblastoma stem cells of different cellular states disrupts malignant progression. Nat Commun 2025; 16:2974. [PMID: 40140646 PMCID: PMC11947120 DOI: 10.1038/s41467-025-58366-5] [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: 03/26/2024] [Accepted: 03/19/2025] [Indexed: 03/28/2025] Open
Abstract
Glioblastoma (GBM) is the most lethal primary brain tumor with intra-tumoral hierarchy of glioblastoma stem cells (GSCs). The heterogeneity of GSCs within GBM inevitably leads to treatment resistance and tumor recurrence. Molecular mechanisms of different cellular state GSCs remain unclear. Here, we find that classical (CL) and mesenchymal (MES) GSCs are enriched in reactive immune region and high CL-MES signature informs poor prognosis in GBM. Through integrated analyses of GSCs RNA sequencing and single-cell RNA sequencing datasets, we identify specific GSCs targets, including MEOX2 for the CL GSCs and SRGN for the MES GSCs. MEOX2-NOTCH and SRGN-NFκB axes play important roles in promoting proliferation and maintaining stemness and subtype signatures of CL and MES GSCs, respectively. In the tumor microenvironment, MEOX2 and SRGN mediate the resistance of CL and MES GSCs to macrophage phagocytosis. Using genetic and pharmacologic approaches, we identify FDA-approved drugs targeting MEOX2 and SRGN. Combined CL and MES GSCs targeting demonstrates enhanced efficacy, both in vitro and in vivo. Our results highlighted a therapeutic strategy for the elimination of heterogeneous GSCs populations through combinatorial targeting of MEOX2 and SRGN in GSCs.
Collapse
Affiliation(s)
- Chenfei Lu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Tao Kang
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Junxia Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Kailin Yang
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA
| | - Yang Liu
- Department of Pharmacology, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Kefan Song
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qiankun Lin
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Deobrat Dixit
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA, USA
| | - Ryan C Gimple
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA
| | - Qian Zhang
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhumei Shi
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiao Fan
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qiulian Wu
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA, USA
| | - Daqi Li
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Danyang Shan
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiancheng Gao
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Danling Gu
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hao You
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yangqing Li
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Junlei Yang
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Linjie Zhao
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA, USA
| | - Zhixin Qiu
- Department of Anesthesiology, Zhongshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Hui Yang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Ningwei Zhao
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Wei Gao
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Weiwei Tao
- College of Biomedicine and Health & College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Yingmei Lu
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yun Chen
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jing Ji
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhe Zhu
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Chunsheng Kang
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Jianghong Man
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Sameer Agnihotri
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA, USA
| | - Qianghu Wang
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Fan Lin
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xu Qian
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Stephen C Mack
- Department of Developmental Neurobiology, Neurobiology and Brain Tumor Program, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Zhibin Hu
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chaojun Li
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Michael D Taylor
- Department of Pediatrics- Hematology/Oncology and Neurosurgery, Texas Children's Cancer Center, Hematology-Oncology Section, Baylor College of Medicine, Houston, Texas, USA
| | - Ning Liu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Nu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ming Lu
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yongping You
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Jeremy N Rich
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA, USA.
| | - Wei Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Xiuxing Wang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China.
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China.
- Jiangsu Cancer Hospital, Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
11
|
Pinzón Martín S, Mecinović J. Selenalysine as a Chemical Tool for Probing Histone Post-Translational Modifications. Bioconjug Chem 2025; 36:510-520. [PMID: 40040526 DOI: 10.1021/acs.bioconjchem.4c00567] [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: 03/06/2025]
Abstract
Post-translational modifications (PTMs) on histones play a crucial role in determining the structure and function of chromatin, thereby regulating the eukaryotic gene expression. Histone lysine methylation and acetylation are among the most widespread and biomedically important PTMs, with new chemical tools for their examination in high demand. Here, we report the first use of γ-selenalysine as an efficient lysine mimic for enzymatic methylation, acetylation, and deacetylation reactions catalyzed by histone lysine methyltransferases, acetyltransferases, and a deacetylase. We also show that easily accessible selenocysteine and cysteine residues can undergo chemo- and site-selective alkylation reactions to generate both unmodified and modified γ-selenalysine and related γ-thialysine residues in histone peptides. This dual-modification strategy enables the site-specific incorporation of two distinct functionalities into peptides, mimicking lysine post-translational modifications commonly found on histones. Our research presents a novel approach in which selenocysteine serves as a unique handle for the chemoselective introduction of selenalysine, along with its methylated and acetylated analogues. These tools are designed to facilitate the study of epigenetic proteins that are important for human health and disease.
Collapse
Affiliation(s)
- Sandra Pinzón Martín
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark
| | - Jasmin Mecinović
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark
| |
Collapse
|
12
|
Bashir K, Todaka D, Sako K, Ueda M, Aziz F, Seki M. Chemical application improves stress resilience in plants. PLANT MOLECULAR BIOLOGY 2025; 115:47. [PMID: 40105987 PMCID: PMC11922999 DOI: 10.1007/s11103-025-01566-w] [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] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 02/12/2025] [Indexed: 03/22/2025]
Abstract
In recent years, abiotic stresses, including droughts, floods, high temperatures, and salinity, have become increasingly frequent and severe. These stresses significantly hinder crop yields and product quality, posing substantial challenges to sustainable agriculture and global food security. Simultaneously, the rapidly growing global population exacerbates the need to enhance crop production under worsening environmental conditions. Consequently, the development of effective strategies to strengthen the resilience of crop plants against high temperatures, water scarcity, and extreme environmental conditions is critical for mitigating the impacts of abiotic stress. Plants respond to these environmental challenges by reprogramming their transcriptome and metabolome. Common strategies for developing stress-tolerant plants include screening germplasm, generating transgenic crop plants, and employing genome editing techniques. Recently, chemical treatment has emerged as a promising approach to enhance abiotic stress tolerance in crops. This technique involves the application of exogenous chemical compounds that induce molecular and physiological changes, thereby providing a protective shield against abiotic stress. Forward and reverse genetic approaches have facilitated the identification of chemicals capable of modulating plant responses to abiotic stresses. These priming agents function as epigenetic regulators, agonists, or antagonists, playing essential roles in regulating stomatal closure to conserve water, managing cellular signaling through reactive oxygen species and metabolites to sustain plant growth, and activating gluconeogenesis to enhance cellular metabolism. This review summarizes recent advancements in the field of chemical priming and explores strategies to improve stress tolerance and crop productivity, thereby contributing to the enhancement of global food security.
Collapse
Grants
- 18H04791 Ministry of Education, Culture, Sports, Science and Technology
- 18H04705 Ministry of Education, Culture, Sports, Science and Technology
- 23119522 Ministry of Education, Culture, Sports, Science and Technology
- 25119724 Ministry of Education, Culture, Sports, Science and Technology
- CREST (JPMJCR13B4) the Japan Science and Technology Agency (JST)
- A-STEP (JPMJTM19BS) the Japan Science and Technology Agency (JST)
- GteX (JPMJGX23B0) the Japan Science and Technology Agency (JST)
- ASPIRE (JPMJAP24A3) Japan Society for Technology of Plasticity
Collapse
Affiliation(s)
- Khurram Bashir
- Plant Genomic Network Research Team, RIKEN Center for Sustainable Resource Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan.
- Department of Life Sciences, SBA School of Science and Engineering, , Lahore University of Management Sciences, DHA Phase 5, Lahore, Pakistan.
| | - Daisuke Todaka
- Plant Genomic Network Research Team, RIKEN Center for Sustainable Resource Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
| | - Kaori Sako
- Plant Genomic Network Research Team, RIKEN Center for Sustainable Resource Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
- Department of Advanced Bioscience, Faculty of Agriculture, Kindai University, Nakamachi, Nara, 3327-204, Japan
| | - Minoru Ueda
- Plant Genomic Network Research Team, RIKEN Center for Sustainable Resource Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
- Plant Epigenome Regulation Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama, Japan
| | - Farhan Aziz
- Plant Genomic Network Research Team, RIKEN Center for Sustainable Resource Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
- Department of Life Sciences, SBA School of Science and Engineering, , Lahore University of Management Sciences, DHA Phase 5, Lahore, Pakistan
| | - Motoaki Seki
- Plant Genomic Network Research Team, RIKEN Center for Sustainable Resource Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan.
- Plant Epigenome Regulation Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama, Japan.
- Kihara Institute for Biological Research, Yokohama City University, Yokohama, Japan.
- Graduate School of Science and Engineering, Saitama University, Saitama, Saitama, Japan.
| |
Collapse
|
13
|
Stojchevski R, Sutanto EA, Sutanto R, Hadzi-Petrushev N, Mladenov M, Singh SR, Sinha JK, Ghosh S, Yarlagadda B, Singh KK, Verma P, Sengupta S, Bhaskar R, Avtanski D. Translational Advances in Oncogene and Tumor-Suppressor Gene Research. Cancers (Basel) 2025; 17:1008. [PMID: 40149342 PMCID: PMC11940485 DOI: 10.3390/cancers17061008] [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: 02/10/2025] [Revised: 03/10/2025] [Accepted: 03/15/2025] [Indexed: 03/29/2025] Open
Abstract
Cancer, characterized by the uncontrolled proliferation of cells, is one of the leading causes of death globally, with approximately one in five people developing the disease in their lifetime. While many driver genes were identified decades ago, and most cancers can be classified based on morphology and progression, there is still a significant gap in knowledge about genetic aberrations and nuclear DNA damage. The study of two critical groups of genes-tumor suppressors, which inhibit proliferation and promote apoptosis, and oncogenes, which regulate proliferation and survival-can help to understand the genomic causes behind tumorigenesis, leading to more personalized approaches to diagnosis and treatment. Aberration of tumor suppressors, which undergo two-hit and loss-of-function mutations, and oncogenes, activated forms of proto-oncogenes that experience one-hit and gain-of-function mutations, are responsible for the dysregulation of key signaling pathways that regulate cell division, such as p53, Rb, Ras/Raf/ERK/MAPK, PI3K/AKT, and Wnt/β-catenin. Modern breakthroughs in genomics research, like next-generation sequencing, have provided efficient strategies for mapping unique genomic changes that contribute to tumor heterogeneity. Novel therapeutic approaches have enabled personalized medicine, helping address genetic variability in tumor suppressors and oncogenes. This comprehensive review examines the molecular mechanisms behind tumor-suppressor genes and oncogenes, the key signaling pathways they regulate, epigenetic modifications, tumor heterogeneity, and the drug resistance mechanisms that drive carcinogenesis. Moreover, the review explores the clinical application of sequencing techniques, multiomics, diagnostic procedures, pharmacogenomics, and personalized treatment and prevention options, discussing future directions for emerging technologies.
Collapse
Affiliation(s)
- Radoslav Stojchevski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA;
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Edward Agus Sutanto
- CUNY School of Medicine, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA;
| | - Rinni Sutanto
- New York Institute of Technology College of Osteopathic Medicine, Glen Head, NY 11545, USA;
| | - Nikola Hadzi-Petrushev
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia; (N.H.-P.)
| | - Mitko Mladenov
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia; (N.H.-P.)
| | - Sajal Raj Singh
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | - Jitendra Kumar Sinha
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | - Shampa Ghosh
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | | | - Krishna Kumar Singh
- Symbiosis Centre for Information Technology (SCIT), Rajiv Gandhi InfoTech Park, Hinjawadi, Pune 411057, Maharashtra, India;
| | - Prashant Verma
- School of Management, BML Munjal University, NH8, Sidhrawali, Gurugram 122413, Haryana, India
| | - Sonali Sengupta
- Department of Gastroenterology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Dimiter Avtanski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA;
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| |
Collapse
|
14
|
Dai J, Lu X, Zhang C, Qu T, Li W, Su J, Guo R, Yin D, Wu P, Han L, Zhang E. NNMT promotes acquired EGFR-TKI resistance by forming EGR1 and lactate-mediated double positive feedback loops in non-small cell lung cancer. Mol Cancer 2025; 24:79. [PMID: 40089784 PMCID: PMC11909984 DOI: 10.1186/s12943-025-02285-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 02/26/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) are remarkably effective for treating EGFR-mutant non-small cell lung cancer (NSCLC). However, patients inevitably develop acquired drug resistance, resulting in recurrence or metastasis. It is important to identify novel effective therapeutic targets to reverse acquired TKI resistance. RESULTS Bioinformatics analysis revealed that nicotinamide N-methyltransferase (NNMT) was upregulated in EGFR-TKI resistant cells and tissues via EGR1-mediated transcriptional activation. High NNMT levels were correlated with poor prognosis in EGFR-mutated NSCLC patients, which could promote resistance to EGFR-TKIs in vitro and in vivo. Mechanistically, NNMT catalyzed the conversion of nicotinamide to 1-methyl nicotinamide by depleting S-adenosyl methionine (the methyl group donor), leading to a reduction in H3K9 trimethylation (H3K9me3) and H3K27 trimethylation (H3K27me3) and subsequent epigenetic activation of EGR1 and ALDH3A1. In addition, ALDH3A1 activation increased lactic acid levels, which further promoted NNMT expression via p300-mediated histone H3K18 lactylation on its promoter. Thus, NNMT mediates the formation of a double positive feedback loop via EGR1 and lactate, EGR1/NNMT/EGR1 and NNMT/ALDH3A1/lactate/NNMT. Moreover, the combination of a small-molecule inhibitor for NNMT (NNMTi) and osimertinib exhibited promising potential for the treatment of TKI resistance in an NSCLC osimertinib-resistant xenograft model. CONCLUSIONS The combined contribution of these two positive feedback loops promotes EGFR-TKI resistance in NSCLC. Our findings provide new insight into the role of histone methylation and histone lactylation in TKI resistance. The pivotal NNMT-mediated positive feedback loop may serve as a powerful therapeutic target for overcoming EGFR-TKI resistance in NSCLC.
Collapse
Affiliation(s)
- Jiali Dai
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Xiyi Lu
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Chang Zhang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Tianyu Qu
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Wei Li
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Jun Su
- Department of Oncology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - Renhua Guo
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Dandan Yin
- Clinical Research Center, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Zhong Fu Road, Gulou District, Nanjing, Jiangsu, 210003, PR China.
| | - Pingping Wu
- Department of Oncology, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institue of Cancer Research, Nanjing, Jiangsu, PR China.
| | - Liang Han
- Department of Oncology, Xuzhou Central Hospital, Xuzhou School of Clinical Medicine of Nanjing Medical University, Xuzhou, Jiangsu, PR China.
| | - Erbao Zhang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
15
|
Ulhe A, Sharma N, Mahajan A, Patil R, Hegde M, Bhalerao S, Mali A. Decoding the therapeutic landscape of alpha-linolenic acid: a network pharmacology and bioinformatics investigation against cancer-related epigenetic modifiers. J Biomol Struct Dyn 2025; 43:1929-1954. [PMID: 38088751 DOI: 10.1080/07391102.2023.2293267] [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: 10/13/2023] [Accepted: 11/29/2023] [Indexed: 02/01/2025]
Abstract
Omega-3 (n - 3) and omega-6 (n - 6) polyunsaturated fatty acids (PUFAs) are vital for human health, but an imbalance between these types is associated with chronic diseases, including cancer. Alpha-linolenic acid (ALA), a n - 3 PUFA, shows promise as an anticancer agent in both laboratory and animal studies. However, the precise molecular mechanisms underlying ALA's actions against cancer-related epigenetic modifiers (CaEpM) remain unclear. To understand this, we employed network pharmacology (NP) and molecular docking techniques. Our study identified 51 potential ALA targets and GO and KEGG pathway analysis revealed possible molecular targets and signaling pathways of ALA against CaEpM. From PPI analysis, EZH2, KAT2B, SIRT1, KAT2A, KDM6B, EHMT2, WDR5, SETD7, SIRT2, and HDAC3 emerged as the top 10 potential targets. Additionally, GeneMANIA functional association (GMFA) network analysis of these top 10 targets was performed to enhance NP insights and explore ALA's multi-target approach. After an exhaustive analysis of the core FGN subnetwork, it became evident that 9 out of the 15 targets-namely EZH2, SUZ12, EED, PARP1, HDAC3, DNMT1, NCOR2, KAT2B, and TRRAP-manifested evidently strong and abundant interconnections among each other. Molecular docking of both top 10 targets and core FGN targets confirmed strong binding affinity between ALA and SIRT2, WDR5, KDM6B, EHMT2, HDAC3, EZH2, PARP1, and KAT2B, underscoring their roles in ALA's anti-CaEpM mechanism. Our findings suggest that ALA may target key signaling pathways related to transcriptional regulation, microRNA involvement, stem cell pluripotency and cellular senescence in cancer epigenetics. These findings illuminate ALA's potential as a multi-target agent against CaEpM.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Amrita Ulhe
- Cancer Research Lab, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, Maharashtra, India
| | - Nidhi Sharma
- Cancer Research Lab, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, Maharashtra, India
| | - Akanksha Mahajan
- Cancer Research Lab, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, Maharashtra, India
| | - Rajesh Patil
- Sinhgad Technical Education Society's, Sinhgad College of Pharmacy, Department of Pharmaceutical Chemistry, Vadgaon (BK), Pune, Maharashtra, India
| | - Mahabaleshwar Hegde
- Center for Innovation in Nutrition, Health, Disease (CINHD), Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, Maharashtra, India
| | - Supriya Bhalerao
- Obesity and Diabetes Lab, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, Maharashtra, India
| | - Aniket Mali
- Cancer Research Lab, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, Maharashtra, India
| |
Collapse
|
16
|
Murphy AE, Askarova A, Lenhard B, Skene NG, Marzi S. Predicting gene expression from histone marks using chromatin deep learning models depends on histone mark function, regulatory distance and cellular states. Nucleic Acids Res 2025; 53:gkae1212. [PMID: 39660643 PMCID: PMC11879020 DOI: 10.1093/nar/gkae1212] [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: 03/29/2024] [Revised: 10/12/2024] [Accepted: 12/09/2024] [Indexed: 12/12/2024] Open
Abstract
To understand the complex relationship between histone mark activity and gene expression, recent advances have used in silico predictions based on large-scale machine learning models. However, these approaches have omitted key contributing factors like cell state, histone mark function or distal effects, which impact the relationship, limiting their findings. Moreover, downstream use of these models for new biological insight is lacking. Here, we present the most comprehensive study of this relationship to date - investigating seven histone marks in eleven cell types across a diverse range of cell states. We used convolutional and attention-based models to predict transcription from histone mark activity at promoters and distal regulatory elements. Our work shows that histone mark function, genomic distance and cellular states collectively influence a histone mark's relationship with transcription. We found that no individual histone mark is consistently the strongest predictor of gene expression across all genomic and cellular contexts. This highlights the need to consider all three factors when determining the effect of histone mark activity on transcriptional state. Furthermore, we conducted in silico histone mark perturbation assays, uncovering functional and disease related loci and highlighting frameworks for the use of chromatin deep learning models to uncover new biological insight.
Collapse
Affiliation(s)
- Alan E Murphy
- UK Dementia Research Institute at Imperial College London, 86 Wood Lane, London W12 0BZ, UK
- Department of Brain Sciences, Imperial College London, 86 Wood Lane, London W12 0BZ, UK
| | - Aydan Askarova
- UK Dementia Research Institute at Imperial College London, 86 Wood Lane, London W12 0BZ, UK
- Department of Brain Sciences, Imperial College London, 86 Wood Lane, London W12 0BZ, UK
| | - Boris Lenhard
- MRC London Institute of Medical Sciences, Imperial College London, Du Cane Road, London W12 0HS, UK
| | - Nathan G Skene
- UK Dementia Research Institute at Imperial College London, 86 Wood Lane, London W12 0BZ, UK
- Department of Brain Sciences, Imperial College London, 86 Wood Lane, London W12 0BZ, UK
| | - Sarah J Marzi
- Department of Brain Sciences, Imperial College London, 86 Wood Lane, London W12 0BZ, UK
- UK Dementia Research Institute at King’s College London, 338 Euston Road, London SE5 9RT, UK
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry Psychology & Neuroscience, King’s College London, 16 De Crespigny Park, London SE5 9RT, UK
| |
Collapse
|
17
|
Feng Q, Yang P, Lyu J, Liu X, Zhong S, Liang Y, Liu P, Huang L, Fan S, Zhang X. The overview of lactylation in the placenta of preeclampsia. Placenta 2025; 160:135-143. [PMID: 39799845 DOI: 10.1016/j.placenta.2025.01.003] [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: 09/29/2024] [Revised: 12/14/2024] [Accepted: 01/02/2025] [Indexed: 01/15/2025]
Abstract
BACKGROUND Preeclampsia is a major challenge for obstetricians due to its severe impacts on maternal and fetal health. Lysine lactylation (Kla) derived from lactate is a novel type of post-translational modification which has been confirmed to affect the malignant progression of diseases as an epigenetic modifier. However, the systemic lactylome profiling of preeclampsia is still unclear. MATERIAL AND METHODS Immunohistochemistry and protein immunoassay were performed on placenta tissues from preeclamptic patients and control pregnancies to compare lactylation levels between the groups. Then liquid chromatography-tandem mass spectrometry (LC-MS/MS) was utilized for quantitative lactylomic analysis and proteomic assessment for proteins with differentially lactated modification. Bioinformatics analyses were applied to reveal the conserved motif sequences and enrichment pathways. RESULTS Significant differences in protein lactylation levels were evident in the placenta between preeclamptic and control groups, with modifications observed in both histone and non-histone proteins. Lactylome analysis showed significant downregulation of 59 Kla proteins and 69 Kla sites in preeclamptic placentas, whereas 44 proteins and 60 sites were upregulated. These differentially lactylated proteins were primarily mitochondrial and associated with the citrate cycle (TCA cycle). Enriched metabolic pathways linked to lactylation included those important for vascular muscle contraction, platelet activation, and several signaling pathways like PI3K-Akt, PPAR, and cholesterol metabolism. CONCLUSIONS Preeclamptic placentas exhibit distinct lactylation profiles compared to normal pregnancies, primarily affecting mitochondrial and TCA cycle-related energy metabolism. These changes contribute to the pathophysiology of preeclampsia by involving metabolic pathways critical for angiogenesis and endothelial function.
Collapse
Affiliation(s)
- Qiaoli Feng
- Department of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Ping Yang
- Department of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, China; Shenzhen Key Laboratory on Technology for Early Diagnosis of Major Gynecological Diseases, Shenzhen, China; Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong province, China
| | - Jinli Lyu
- Department of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, China; Shenzhen Key Laboratory on Technology for Early Diagnosis of Major Gynecological Diseases, Shenzhen, China
| | - Xinyang Liu
- Department of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, China; Shenzhen Key Laboratory on Technology for Early Diagnosis of Major Gynecological Diseases, Shenzhen, China
| | - Shilin Zhong
- Department of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yiheng Liang
- Department of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Ping Liu
- Department of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Liting Huang
- Department of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, China; Shenzhen Key Laboratory on Technology for Early Diagnosis of Major Gynecological Diseases, Shenzhen, China
| | - Shangrong Fan
- Department of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, China; Shenzhen Key Laboratory on Technology for Early Diagnosis of Major Gynecological Diseases, Shenzhen, China.
| | - Xiaowei Zhang
- Department of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, China; Shenzhen Key Laboratory on Technology for Early Diagnosis of Major Gynecological Diseases, Shenzhen, China.
| |
Collapse
|
18
|
Lu T, Yang J, Cai Y, Ding M, Yu Z, Fang X, Zhou X, Wang X. NCAPD3 promotes diffuse large B-cell lymphoma progression through modulating SIRT1 expression in an H3K9 monomethylation-dependent manner. J Adv Res 2025; 68:163-178. [PMID: 38432395 PMCID: PMC11785590 DOI: 10.1016/j.jare.2024.02.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 01/31/2024] [Accepted: 02/29/2024] [Indexed: 03/05/2024] Open
Abstract
INTRODUCTION Condensin, a family of structural maintenance of chromosome complexes, has been shown to regulate chromosome compaction and segregation during mitosis. NCAPD3, a HEAT-repeat subunit of condensin II, plays a dominant role in condensin-mediated chromosome dynamics but remains unexplored in lymphoma. OBJECTIVES The study aims to unravel the molecular function and mechanism of NCAPD3 in diffuse large B-cell lymphoma (DLBCL). METHODS The expression and clinical significance of NCAPD3 were assessed in public database and clinical specimens. Chromosome spreads, co-immunoprecipitation (co-IP), mass spectrometry (MS), and chromatin immunoprecipitation (ChIP) assays were conducted to untangle the role and mechanism of NCAPD3 in DLBCL. RESULTS NCAPD3 was highly expressed in DLBCL, correlated with poor prognosis. NCAPD3 deficiency impeded cell proliferation, induced apoptosis and increased the chemosensitivity. Instead, NCAPD3 overexpression facilitated cell proliferation. In vivo experiments further indicated targeting NCAPD3 suppressed tumor growth. Noteworthily, NCAPD3 deficiency disturbed the mitosis, triggering the formation of aneuploids. To reveal the function of NCAPD3 in DLBCL, chromosome spreads were conducted, presenting that chromosomes became compact upon NCAPD3 overexpression, instead, loose, twisted and lacking axial rigidity upon NCAPD3 absence. Meanwhile, the classical transcription-activated marker, H3K4 trimethylation, was found globally upregulated after NCAPD3 knockout, suggesting that NCAPD3 might participate in chromatin remodeling and transcription regulation. MS revealed NCAPD3 could interact with transcription factor, TFII I. Further co-IP and ChIP assays verified NCAPD3 could be anchored at the promoter of SIRT1 by TFII I and then supported the transcription of SIRT1 via recognizing H3K9 monomethylation (H3K9me1) on SIRT1 promoter. Function reversion assay verified the oncogenic role of NCAPD3 in DLBCL was partially mediated by SIRT1. CONCLUSION This study demonstrated that dysregulation of NCAPD3 could disturb chromosome compaction and segregation and regulate the transcription activity of SIRT1 in an H3K9me1-dependent manner, which provided novel insights into targeted strategy for DLBCL.
Collapse
Affiliation(s)
- Tiange Lu
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China
| | - Juan Yang
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China; National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yiqing Cai
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China
| | - Mengfei Ding
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China
| | - Zhuoya Yu
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China
| | - Xiaosheng Fang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Taishan Scholars Program of Shandong Province, Jinan, Shandong 250021, China; Branch of National Clinical Research Center for Hematologic Diseases, Jinan, Shandong 250021, China; National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou 251006, China.
| | - Xiangxiang Zhou
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Taishan Scholars Program of Shandong Province, Jinan, Shandong 250021, China; Branch of National Clinical Research Center for Hematologic Diseases, Jinan, Shandong 250021, China; National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou 251006, China.
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China; Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Taishan Scholars Program of Shandong Province, Jinan, Shandong 250021, China; Branch of National Clinical Research Center for Hematologic Diseases, Jinan, Shandong 250021, China; National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou 251006, China.
| |
Collapse
|
19
|
Britto LS, Balasubramani D, Desai S, Phillips P, Trehan N, Cesarman E, Koff JL, Singh A. T Cells Spatially Regulate B Cell Receptor Signaling in Lymphomas through H3K9me3 Modifications. Adv Healthc Mater 2025; 14:e2401192. [PMID: 38837879 PMCID: PMC11617604 DOI: 10.1002/adhm.202401192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/27/2024] [Indexed: 06/07/2024]
Abstract
Activated B cell-like diffuse large B-cell lymphoma (ABC-DLBCL) is a subtype associated with poor survival outcomes. Despite identifying therapeutic targets through molecular characterization, targeted therapies have limited success. New strategies using immune-competent tissue models are needed to understand how DLBCL cells evade treatment. Here, synthetic hydrogel-based lymphoma organoids are used to demonstrate how signals in the lymphoid tumor microenvironment (Ly-TME) can alter B cell receptor (BCR) signaling and specific histone modifications, tri-methylation of histone 3 at lysine 9 (H3K9me3), dampening the effects of BCR pathway inhibition. Using imaging modalities, T cells increase DNA methyltransferase 3A expression and cytoskeleton formation in proximal ABC-DLBCL cells, regulated by H3K9me3. Expansion microscopy on lymphoma organoids reveals T cells increase the size and quantity of segregated H3K9me3 clusters in ABC-DLBCL cells. Findings suggest the re-organization of higher-order chromatin structures that may contribute to evasion or resistance to therapy via the emergence of novel transcriptional states. Treating ABC-DLBCL cells with a G9α histone methyltransferase inhibitor reverses T cell-mediated modulation of H3K9me3 and overcomes T cell-mediated attenuation of treatment response to BCR pathway inhibition. This study emphasizes the Ly-TME's role in altering DLBCL fate and suggests targeting aberrant signaling and microenvironmental cross-talk that can benefit high-risk patients.
Collapse
Affiliation(s)
- Lucy S. Britto
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory UniversityAtlantaGA30332USA
| | - Deepali Balasubramani
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory UniversityAtlantaGA30332USA
| | - Sona Desai
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory UniversityAtlantaGA30332USA
| | - Phunterion Phillips
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory UniversityAtlantaGA30332USA
| | - Neev Trehan
- St Richards HospitalUniversity Hospitals Sussex NHS Foundation TrustChichesterWest SussexPO19 6SEUK
| | - Ethel Cesarman
- Department of Pathology and Laboratory MedicineWeill Cornell MedicineNew YorkNY10065USA
| | - Jean L. Koff
- Winship Cancer CenterEmory University School of MedicineAtlantaGA30307USA
| | - Ankur Singh
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory UniversityAtlantaGA30332USA
- Woodruff School of Mechanical EngineeringGeorgia Institute of TechnologyAtlantaGA30318USA
- Petit Institute for Bioengineering and BiosciencesGeorgia Institute of TechnologyAtlantaGA30332USA
| |
Collapse
|
20
|
Tan X, Xun L, Yin Q, Chen C, Zhang T, Shen T. Epigenetic Modifications in HBV-Related Hepatocellular Carcinoma. J Viral Hepat 2025; 32:e14044. [PMID: 39868653 DOI: 10.1111/jvh.14044] [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: 07/31/2024] [Revised: 11/13/2024] [Accepted: 11/30/2024] [Indexed: 01/28/2025]
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver cancer. Hepatitis B virus (HBV) is the main pathogen for HCC development. HBV covalently closed circular DNA (cccDNA) forms extra-host chromatin-like minichromosomes in the nucleus of hepatocytes with host histones, non-histones, HBV X protein (HBx) and HBV core protein (HBc). Epigenetic alterations are dynamic and reversible, which regulate gene expression without altering the DNA sequence and play a pivotal role in the regulation of HCC onset and progression. The aim of this review is to elucidate the deregulation of epigenetic mechanisms involved in the pathogenesis of HBV-related HCC (HBV-HCC), including post-translational histone and non-histone modifications, DNA hypermethylation and hypomethylation, non-coding RNA modification on HBV cccDNA minichromosomes and host factors, effecting the replication/transcription of HBV cccDNA and transcription/translation of host genes, and thus HBV-HCC progression. It is expected that the epigenetic regulation perspective provides new ways for more in-depth development of therapeutic control of HBV-HCC.
Collapse
Affiliation(s)
- Xiaoqing Tan
- Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
- Department of Pulmonary and Critical Care Medicine, Yunnan Provincial Key Laboratory for Clinical Virology, Institute of Basic and Clinical Medicine, The First People's Hospital of Yunnan Province, Kunming, Peoples republic of China, China
| | - Linting Xun
- Department of Gastroenterology, the First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, People's Republic of China
| | - Qi Yin
- Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, People's Republic of China, China
| | - Chaohui Chen
- Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Tao Zhang
- Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
- Department of Pulmonary and Critical Care Medicine, Yunnan Provincial Key Laboratory for Clinical Virology, Institute of Basic and Clinical Medicine, The First People's Hospital of Yunnan Province, Kunming, Peoples republic of China, China
| | - Tao Shen
- Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
- Department of Pulmonary and Critical Care Medicine, Yunnan Provincial Key Laboratory for Clinical Virology, Institute of Basic and Clinical Medicine, The First People's Hospital of Yunnan Province, Kunming, Peoples republic of China, China
| |
Collapse
|
21
|
König L, Schmidts M. The role of chromatin-related epigenetic modulations in CAKUT. Curr Top Dev Biol 2025; 163:169-227. [PMID: 40254345 DOI: 10.1016/bs.ctdb.2024.11.006] [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] [Indexed: 04/22/2025]
Abstract
Congenital anomalies of the kidney and urinary tract (CAKUT) represent a major health burden in humans. Phenotypes range from renal hypoplasia or renal agenesis, cystic renal dysplasia, duplicated or horseshoe kidneys to obstruction of the ureteropelvic junction, megaureters, duplicated ureters, urethral valves or bladder malformations. Over the past decade, next-generation sequencing has identified numerous causative genes; however, the genetic basis of most cases remains unexplained. It is assumed that environmental factors have a significant impact on the phenotype, but, overall, the pathogenesis has remained poorly understood. Interestingly however, CAKUT is a common phenotypic feature in two human syndromes, Kabuki and Koolen-de Vries syndrome, caused by dysfunction of genes encoding for KMT2D and KANSL1, both members of protein complexes playing an important role in histone modifications. In this chapter, we discuss current knowledge regarding epigenetic modulation in renal development and a putatively under-recognized role of epigenetics in CAKUT.
Collapse
Affiliation(s)
- Luise König
- Center for Pediatrics and Adolescent Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Miriam Schmidts
- Center for Pediatrics and Adolescent Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; CIBSS-Center for Integrative Biological Signaling Studies, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
22
|
Bruno PS, Arshad A, Gogu MR, Waterman N, Flack R, Dunn K, Darie CC, Neagu AN. Post-Translational Modifications of Proteins Orchestrate All Hallmarks of Cancer. Life (Basel) 2025; 15:126. [PMID: 39860065 PMCID: PMC11766951 DOI: 10.3390/life15010126] [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: 12/23/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
Post-translational modifications (PTMs) of proteins dynamically build the buffering and adapting interface between oncogenic mutations and environmental stressors, on the one hand, and cancer cell structure, functioning, and behavior. Aberrant PTMs can be considered as enabling characteristics of cancer as long as they orchestrate all malignant modifications and variability in the proteome of cancer cells, cancer-associated cells, and tumor microenvironment (TME). On the other hand, PTMs of proteins can enhance anticancer mechanisms in the tumoral ecosystem or sustain the beneficial effects of oncologic therapies through degradation or inactivation of carcinogenic proteins or/and activation of tumor-suppressor proteins. In this review, we summarized and analyzed a wide spectrum of PTMs of proteins involved in all regulatory mechanisms that drive tumorigenesis, genetic instability, epigenetic reprogramming, all events of the metastatic cascade, cytoskeleton and extracellular matrix (ECM) remodeling, angiogenesis, immune response, tumor-associated microbiome, and metabolism rewiring as the most important hallmarks of cancer. All cancer hallmarks develop due to PTMs of proteins, which modulate gene transcription, intracellular and extracellular signaling, protein size, activity, stability and localization, trafficking, secretion, intracellular protein degradation or half-life, and protein-protein interactions (PPIs). PTMs associated with cancer can be exploited to better understand the underlying molecular mechanisms of this heterogeneous and chameleonic disease, find new biomarkers of cancer progression and prognosis, personalize oncotherapies, and discover new targets for drug development.
Collapse
Affiliation(s)
- Pathea Shawnae Bruno
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY 13699-5810, USA; (P.S.B.); (A.A.); (N.W.); (R.F.); (K.D.)
| | - Aneeta Arshad
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY 13699-5810, USA; (P.S.B.); (A.A.); (N.W.); (R.F.); (K.D.)
| | - Maria-Raluca Gogu
- Advanced Research and Development Center for Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, University Street No. 16, 700115 Iasi, Romania;
| | - Natalie Waterman
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY 13699-5810, USA; (P.S.B.); (A.A.); (N.W.); (R.F.); (K.D.)
| | - Rylie Flack
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY 13699-5810, USA; (P.S.B.); (A.A.); (N.W.); (R.F.); (K.D.)
| | - Kimberly Dunn
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY 13699-5810, USA; (P.S.B.); (A.A.); (N.W.); (R.F.); (K.D.)
| | - Costel C. Darie
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY 13699-5810, USA; (P.S.B.); (A.A.); (N.W.); (R.F.); (K.D.)
| | - Anca-Narcisa Neagu
- Laboratory of Animal Histology, Faculty of Biology, “Alexandru Ioan Cuza” University of Iași, Carol I bvd. 20A, 700505 Iasi, Romania
| |
Collapse
|
23
|
Shah V, Lam HY, Leong CHM, Sakaizawa R, Shah JS, Kumar AP. Epigenetic Control of Redox Pathways in Cancer Progression. Antioxid Redox Signal 2025. [PMID: 39815993 DOI: 10.1089/ars.2023.0465] [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] [Indexed: 01/18/2025]
Abstract
Significance: Growing evidence indicates the importance of redox reactions homeostasis, mediated predominantly by reactive oxygen species (ROS) in influencing the development, differentiation, progression, metastasis, programmed cell death, tumor microenvironment, and therapeutic resistance of cancer. Therefore, reviewing the ROS-linked epigenetic changes in cancer is fundamental to understanding the progression and prevention of cancer. Recent Advances: We review in depth the molecular mechanisms involved in ROS-mediated epigenetic changes that lead to alteration of gene expression by altering DNA, modifying histones, and remodeling chromatin and noncoding RNA. Critical Issues: In cancerous cells, alterations of the gene-expression regulatory elements could be generated by the virtue of imbalance in tumor microenvironment. Various oxidizing agents and mitochondrial electron transport chain are the major pathways that generate ROS. ROS plays a key role in carcinogenesis by activating pro-inflammatory signaling pathways and DNA damage. This loss of ROS-mediated epigenetic regulation of the signaling pathways may promote tumorigenesis. We address all such aspects in this review. Future Directions: Developments in this growing field of epigenetics are expected to contribute to further our understanding of human health and diseases such as cancer and to test the clinical applications of redox-based therapy. Recent studies of the cancer-epigenetic landscape have revealed pervasive deregulation of the epigenetic factors in cancer. Thus, the study of interaction between ROS and epigenetic factors in cancer holds a great promise in the development of effective and targeted treatment modalities. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Vandit Shah
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, India
| | - Hiu Yan Lam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Charlene Hoi-Mun Leong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Reo Sakaizawa
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jigna S Shah
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, India
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
24
|
Chen X, Xu H, Shu X, Song CX. Mapping epigenetic modifications by sequencing technologies. Cell Death Differ 2025; 32:56-65. [PMID: 37658169 PMCID: PMC11742697 DOI: 10.1038/s41418-023-01213-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 08/09/2023] [Accepted: 08/14/2023] [Indexed: 09/03/2023] Open
Abstract
The "epigenetics" concept was first described in 1942. Thus far, chemical modifications on histones, DNA, and RNA have emerged as three important building blocks of epigenetic modifications. Many epigenetic modifications have been intensively studied and found to be involved in most essential biological processes as well as human diseases, including cancer. Precisely and quantitatively mapping over 100 [1], 17 [2], and 160 [3] different known types of epigenetic modifications in histone, DNA, and RNA is the key to understanding the role of epigenetic modifications in gene regulation in diverse biological processes. With the rapid development of sequencing technologies, scientists are able to detect specific epigenetic modifications with various quantitative, high-resolution, whole-genome/transcriptome approaches. Here, we summarize recent advances in epigenetic modification sequencing technologies, focusing on major histone, DNA, and RNA modifications in mammalian cells.
Collapse
Affiliation(s)
- Xiufei Chen
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | - Haiqi Xu
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | - Xiao Shu
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | - Chun-Xiao Song
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK.
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK.
| |
Collapse
|
25
|
Kwon WA, Seo HK, Song G, Lee MK, Park WS. Advances in Therapy for Urothelial and Non-Urothelial Subtype Histologies of Advanced Bladder Cancer: From Etiology to Current Development. Biomedicines 2025; 13:86. [PMID: 39857670 PMCID: PMC11761267 DOI: 10.3390/biomedicines13010086] [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: 12/02/2024] [Revised: 12/30/2024] [Accepted: 12/30/2024] [Indexed: 01/27/2025] Open
Abstract
Urothelial carcinoma (UC) is the most common histological subtype of bladder tumors; however, bladder cancer represents a heterogeneous group of diseases with at least 40 distinct histological subtypes. Among these, the 2022 World Health Organization classification of urinary tract tumors identifies a range of less common subtypes of invasive UC, formerly known as variants, which are considered high-grade tumors, including squamous cell, small-cell, sarcomatoid urothelial, micropapillary, plasmacytoid, and urachal carcinomas, and adenocarcinoma. Their accurate histological diagnosis is critical for risk stratification and therapeutic decision-making, as most subtype histologies are associated with poorer outcomes than conventional UC. Despite the importance of a precise diagnosis, high-quality evidence on optimal treatments for subtype histologies remains limited owing to their rarity. In particular, neoadjuvant and adjuvant chemotherapy have not been well characterized, and prospective data are scarce. For advanced-stage diseases, clinical trial participation is strongly recommended to address the lack of robust evidence. Advances in molecular pathology and the development of targeted therapies and immunotherapies have reshaped our understanding and classification of bladder cancer subtypes, spurring efforts to identify predictive biomarkers to guide personalized treatment strategies. Nevertheless, the management of rare bladder cancer subgroups remains challenging because they are frequently excluded from clinical trials. For localized disease, curative options such as surgical resection or radiotherapy are available; however, treatment options become more limited in recurrence or metastasis, where systemic therapy is primarily used to control disease progression and palliate symptoms. Herein, we present recent advances in the management of urothelial and non-urothelial bladder cancer subtypes and also explore the current evidence guiding their treatment and emphasize the challenges and perspectives of future therapeutic strategies.
Collapse
Affiliation(s)
- Whi-An Kwon
- Department of Urology, Myongji Hospital, Hanyang University College of Medicine, Goyang-si 10475, Republic of Korea;
| | - Ho Kyung Seo
- Department of Urology, Center for Urologic Cancer, National Cancer Center, Goyang-si 10408, Republic of Korea; (H.K.S.); (G.S.)
| | - Geehyun Song
- Department of Urology, Center for Urologic Cancer, National Cancer Center, Goyang-si 10408, Republic of Korea; (H.K.S.); (G.S.)
| | - Min-Kyung Lee
- Department of Internal Medicine, Myongji Hospital, Hanyang University College of Medicine, Goyang-si 10475, Republic of Korea
| | - Weon Seo Park
- Department of Pathology, National Cancer Center, Goyang-si 10408, Republic of Korea
| |
Collapse
|
26
|
Gao Y, Siyu zhang, Zhang X, Du Y, Ni T, Hao S. Crosstalk between metabolic and epigenetic modifications during cell carcinogenesis. iScience 2024; 27:111359. [PMID: 39660050 PMCID: PMC11629229 DOI: 10.1016/j.isci.2024.111359] [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] [Indexed: 12/12/2024] Open
Abstract
Genetic mutations arising from various internal and external factors drive cells to become cancerous. Cancerous cells undergo numerous changes, including metabolic reprogramming and epigenetic modifications, to support their abnormal proliferation. This metabolic reprogramming leads to the altered expression of many metabolic enzymes and the accumulation of metabolites. Recent studies have shown that these enzymes and metabolites can serve as substrates or cofactors for chromatin-modifying enzymes, thereby participating in epigenetic modifications and promoting carcinogenesis. Additionally, epigenetic modifications play a role in the metabolic reprogramming and immune evasion of cancer cells, influencing cancer progression. This review focuses on the origins of cancer, particularly the metabolic reprogramming of cancer cells and changes in epigenetic modifications. We discuss how metabolites in cancer cells contribute to epigenetic remodeling, including lactylation, acetylation, succinylation, and crotonylation. Finally, we review the impact of epigenetic modifications on tumor immunity and the latest advancements in cancer therapies targeting these modifications.
Collapse
Affiliation(s)
- Yue Gao
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China
| | - Siyu zhang
- Key Lab of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, School of Life Sciences, Ningxia University, Yinchuan 750021, China
| | - Xianhong Zhang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China
| | - Yitian Du
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China
| | - Ting Ni
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China
| | - Shuailin Hao
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China
| |
Collapse
|
27
|
Lazo PA. Nuclear functions regulated by the VRK1 kinase. Nucleus 2024; 15:2353249. [PMID: 38753965 PMCID: PMC11734890 DOI: 10.1080/19491034.2024.2353249] [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: 04/02/2024] [Revised: 05/03/2024] [Accepted: 05/06/2024] [Indexed: 05/18/2024] Open
Abstract
In the nucleus, the VRK1 Ser-Thr kinase is distributed in nucleoplasm and chromatin, where it has different roles. VRK1 expression increases in response to mitogenic signals. VRK1 regulates cyclin D1 expression at G0 exit and facilitates chromosome condensation at the end of G2 and G2/M progression to mitosis. These effects are mediated by the phosphorylation of histone H3 at Thr3 by VRK1, and later in mitosis by haspin. VRK1 regulates the apigenetic patterns of histones in processes requiring chromating remodeling, such as transcription, replication and DNA repair. VRK1 is overexpressed in tumors, facilitating tumor progression and resistance to genotoxic treatments. VRK1 also regulates the organization of Cajal bodies assembled on coilin, which are necessary for the assembly of different types of RNP complexes. VRK1 pathogenic variants cuase defects in Cajal bodies, functionally altering neurons with long axons and leading to neurological diseases, such as amyotrophic laterla sclerosis, spinal muscular atrophy, distal hereditay motor neuropathies and Charcot-Marie-Tooth.
Collapse
Affiliation(s)
- Pedro A. Lazo
- Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC) - Universidad de Salamanca, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, Salamanca, Spain
| |
Collapse
|
28
|
Nadhan R, Isidoro C, Song YS, Dhanasekaran DN. LncRNAs and the cancer epigenome: Mechanisms and therapeutic potential. Cancer Lett 2024; 605:217297. [PMID: 39424260 DOI: 10.1016/j.canlet.2024.217297] [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: 08/23/2024] [Revised: 09/30/2024] [Accepted: 10/08/2024] [Indexed: 10/21/2024]
Abstract
Long non-coding RNAs (lncRNAs) have emerged as critical regulators of epigenome, modulating gene expression through DNA methylation, histone modification, and/or chromosome remodeling. Dysregulated lncRNAs act as oncogenes or tumor suppressors, driving tumor progression by shaping the cancer epigenome. By interacting with the writers, readers, and erasers of the epigenetic script, lncRNAs induce epigenetic modifications that bring about changes in cancer cell proliferation, apoptosis, epithelial-mesenchymal transition, migration, invasion, metastasis, cancer stemness and chemoresistance. This review analyzes and discusses the multifaceted role of lncRNAs in cancer pathobiology, from cancer genesis and progression through metastasis and therapy resistance. It also explores the therapeutic potential of targeting lncRNAs through innovative diagnostic, prognostic, and therapeutic strategies. Understanding the dynamic interplay between lncRNAs and epigenome is crucial for developing personalized therapeutic strategies, offering new avenues for precision cancer medicine.
Collapse
Affiliation(s)
- Revathy Nadhan
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | - Ciro Isidoro
- Laboratory of Molecular Pathology and NanoBioImaging, Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy.
| | - Yong Sang Song
- Department of Obstetrics and Gynecology, Cancer Research Institute, College of Medicine, Seoul National University, Seoul, 151-921, South Korea.
| | - Danny N Dhanasekaran
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Cell Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
29
|
Hontecillas-Prieto L, García-Domínguez DJ, Flores-Campos R, Flores JA, Pérez-Pérez A, de la Cruz-Merino L, Sánchez-Margalet V, Hajji N. Simplified acid extraction and quantification of histones in human tumor cells. Methods Cell Biol 2024; 191:1-14. [PMID: 39824551 DOI: 10.1016/bs.mcb.2024.10.002] [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] [Indexed: 01/20/2025]
Abstract
Histones are essential nuclear proteins that package eukaryotic DNA into chromosomes, play a vital role in gene regulation, DNA replication, DNA repair and chromosome condensation. Understanding histone modifications is crucial for grasping biological and disease-related processes. Specific alterations in histone modifications serve as sensitive and selective biomarkers for conditions like cancer, impacting both tumor and immune cells and affecting their interactions. Indeed, the interest in histone modifications is growing in the field of tumor immunology and immunotherapy. Different techniques have been developed to characterize histone proteins and their modifications. Here, we present a simple acid extraction protocol to identify and quantify histones. The workflow described here can be used to detect and measure histone proteins or specific residues of histone, even capturing changes resulting from treatment with epigenetic drugs (Epi-drugs) or other drugs in in different human cancer cell line models.
Collapse
Affiliation(s)
- Lourdes Hontecillas-Prieto
- Clinical Biochemistry Service, Hospital Universitario Virgen Macarena, University of Seville, Seville, Spain; Department of Medical Biochemistry and Molecular Biology and Immunology, Medical School, Virgen Macarena University Hospital, University of Seville, Seville, Spain; Institute of Biomedicine of Seville (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain; Department of Clinical Oncology, Hospital Universitario Virgen Macarena, University of Seville, Seville, Spain.
| | - Daniel J García-Domínguez
- Department of Medical Biochemistry and Molecular Biology and Immunology, Medical School, Virgen Macarena University Hospital, University of Seville, Seville, Spain; Institute of Biomedicine of Seville (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain
| | - Rocío Flores-Campos
- Department of Clinical Oncology, Hospital Universitario Virgen Macarena, University of Seville, Seville, Spain
| | - Juan Antonio Flores
- Department of Medical Biochemistry and Molecular Biology and Immunology, Medical School, Virgen Macarena University Hospital, University of Seville, Seville, Spain
| | - Antonio Pérez-Pérez
- Clinical Biochemistry Service, Hospital Universitario Virgen Macarena, University of Seville, Seville, Spain; Department of Medical Biochemistry and Molecular Biology and Immunology, Medical School, Virgen Macarena University Hospital, University of Seville, Seville, Spain
| | - Luis de la Cruz-Merino
- Institute of Biomedicine of Seville (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain; Department of Clinical Oncology, Hospital Universitario Virgen Macarena, University of Seville, Seville, Spain; Department of Medicine, University of Seville, Seville, Spain
| | - Víctor Sánchez-Margalet
- Clinical Biochemistry Service, Hospital Universitario Virgen Macarena, University of Seville, Seville, Spain; Department of Medical Biochemistry and Molecular Biology and Immunology, Medical School, Virgen Macarena University Hospital, University of Seville, Seville, Spain; Institute of Biomedicine of Seville (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain
| | - Nabil Hajji
- Department of Medical Biochemistry and Molecular Biology and Immunology, Medical School, Virgen Macarena University Hospital, University of Seville, Seville, Spain; Cancer Division, Faculty of medicine, Imperial college London, United Kingdom
| |
Collapse
|
30
|
Zhu R, Ni J, Ren J, Li D, Xu J, Yu X, Ma YJ, Kou L. Transcriptomic era of cancers in females: new epigenetic perspectives and therapeutic prospects. Front Oncol 2024; 14:1464125. [PMID: 39605897 PMCID: PMC11598703 DOI: 10.3389/fonc.2024.1464125] [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: 07/13/2024] [Accepted: 10/16/2024] [Indexed: 11/29/2024] Open
Abstract
In the era of transcriptomics, the role of epigenetics in the study of cancers in females has gained increasing recognition. This article explores the impact of epigenetic modifications, such as DNA methylation, histone modification, and non-coding RNA, on cancers in females, including breast, cervical, and ovarian cancers (1). Our findings suggest that these epigenetic markers not only influence tumor onset, progression, and metastasis but also present novel targets for therapeutic intervention. Detailed analyses of DNA methylation patterns have revealed aberrant events in cancer cells, particularly promoter region hypermethylation, which may lead to silencing of tumor suppressor genes. Furthermore, we examined the complex roles of histone modifications and long non-coding RNAs in regulating the expression of cancer-related genes, thereby providing a scientific basis for developing targeted epigenetic therapies. Our research emphasizes the importance of understanding the functions and mechanisms of epigenetics in cancers in females to develop effective treatment strategies. Future therapeutic approaches may include drugs targeting specific epigenetic markers, which could not only improve therapeutic outcomes but also enhance patient survival and quality of life. Through these efforts, we aim to offer new perspectives and hope for the prevention, diagnosis, and treatment of cancers in females.
Collapse
Affiliation(s)
- Runhe Zhu
- The Traditional Chinese Medicine College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiawei Ni
- The Traditional Chinese Medicine College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiayin Ren
- The Traditional Chinese Medicine College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Dongye Li
- The Traditional Chinese Medicine College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiawei Xu
- The Traditional Chinese Medicine College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xinru Yu
- The Pharmacy College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ying Jie Ma
- The First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Luan Kou
- Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, China
| |
Collapse
|
31
|
Zhang J, Wang Q, Liu J, Duan Y, Liu Z, Zhang Z, Li C. Active enhancers: recent research advances and insights into disease. Biol Direct 2024; 19:112. [PMID: 39533395 PMCID: PMC11556110 DOI: 10.1186/s13062-024-00559-x] [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: 07/22/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
Precise regulation of gene expression is crucial to development. Enhancers, the core of gene regulation, determine the spatiotemporal pattern of gene transcription. Since many disease-associated mutations are characterized in enhancers, the research on enhancer will provide clues to precise medicine. Rapid advances in high-throughput sequencing technology facilitate the characterization of enhancers at genome wide, but understanding the functional mechanisms of enhancers remains challenging. Herein, we provide a panorama of enhancer characteristics, including epigenetic modifications, enhancer transcripts, and enhancer-promoter interaction patterns. Furthermore, we outline the applications of high-throughput sequencing technology and functional genomics methods in enhancer research. Finally, we discuss the role of enhancers in human disease and their potential as targets for disease prevention and treatment strategies.
Collapse
Affiliation(s)
- Junyou Zhang
- School of Engineering Medicine, Beihang University, Beijing, 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
- Key Laboratory of Big Data-Based Precision Medicine (Ministry of Industry and Information Technology), Beihang University, Beijing, 100191, China
| | - Qilin Wang
- School of Engineering Medicine, Beihang University, Beijing, 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
- Key Laboratory of Big Data-Based Precision Medicine (Ministry of Industry and Information Technology), Beihang University, Beijing, 100191, China
| | - Jiaxin Liu
- School of Engineering Medicine, Beihang University, Beijing, 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
- Key Laboratory of Big Data-Based Precision Medicine (Ministry of Industry and Information Technology), Beihang University, Beijing, 100191, China
| | - Yingying Duan
- School of Engineering Medicine, Beihang University, Beijing, 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
- Key Laboratory of Big Data-Based Precision Medicine (Ministry of Industry and Information Technology), Beihang University, Beijing, 100191, China
| | - Zhaoshuo Liu
- School of Engineering Medicine, Beihang University, Beijing, 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
- Key Laboratory of Big Data-Based Precision Medicine (Ministry of Industry and Information Technology), Beihang University, Beijing, 100191, China
| | - Ziyi Zhang
- School of Engineering Medicine, Beihang University, Beijing, 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
- Key Laboratory of Big Data-Based Precision Medicine (Ministry of Industry and Information Technology), Beihang University, Beijing, 100191, China
| | - Chunyan Li
- School of Engineering Medicine, Beihang University, Beijing, 100191, China.
- School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China.
- Key Laboratory of Big Data-Based Precision Medicine (Ministry of Industry and Information Technology), Beihang University, Beijing, 100191, China.
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing, 100191, China.
| |
Collapse
|
32
|
X Zhang G, Yang B. Retained PAX2 expression associated with DNA mismatch repair deficiency in endometrial endometrioid adenocarcinoma. Histopathology 2024; 85:794-803. [PMID: 39075663 DOI: 10.1111/his.15281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/31/2024]
Abstract
AIMS Loss of expression of tumour suppressor PAX2 and MMR deficiency (dMMR) has been frequently seen in endometrial endometrioid adenocarcinoma (EEC). However, the relationship between PAX2 expression and MMR status is unknown. METHODS AND RESULTS We studied the PAX2 expression and examined its association with MMR status at the protein and genetic levels in 180 cases of EEC. Overall, total loss of PAX2 expression was found in about 70%, while retained PAX2 expression was seen in 30% of EEC. Among 125 cases with loss of PAX2, 68.8% were found in EECs with pMMR, while 31.2% were seen in those with dMMR. Among 55 cases of EECs with retained PAX2 expression, 92.7% were EECs with dMMR and 7.3% were those with pMMR (P < 0.001). While dMMR cases with MLH1 hypermethylation show almost equal retained or loss of PAX2 expression (52% versus 48%), dMMR with genetic alterations had significantly more retained PAX2 expression than loss of PAX2 (92.3% versus 7.7%), regardless of somatic or germline mutations. Loss of PAX2 was observed in 97.3% of dMMR with MLH1 hypermethylation compared to 2.7% of dMMR with genetic alterations (P < 0.001). Aggressive features such as higher tumour grades (FIGO 2-3) and advanced clinical stage (T2-T4) were significantly more frequently seen in dMMR with retained PAX2 expression, compared those to pMMR with loss of PAX2 expression. CONCLUSION Our study demonstrates a close correlation between retained PAX2 expression and dMMR in EEC. The molecular mechanism and clinical significance linking these two pathways in EEC remains to be unravelled.
Collapse
Affiliation(s)
- Gloria X Zhang
- Pathology and Laboratory Medicine Institute, The Cleveland Clinic, Cleveland, OH, USA
| | - Bin Yang
- Pathology and Laboratory Medicine Institute, The Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
33
|
Bhattacharjya D, Sivalingam N. Mechanism of 5-fluorouracil induced resistance and role of piperine and curcumin as chemo-sensitizers in colon cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:8445-8475. [PMID: 38878089 DOI: 10.1007/s00210-024-03189-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 05/27/2024] [Indexed: 10/30/2024]
Abstract
Among cancer-related deaths worldwide, colorectal cancer ranks second, accounting for 1.2% of deaths in those under 50 years and 0.6% of deaths in those between 50 and 54 years. The anticancer drug 5-fluorouracil is widely used to treat colorectal cancer. Due to a better understanding of the drug's mechanism of action, its anticancer activity has been increased through a variety of therapeutic alternatives. Clinical use of 5-FU has been severely restricted due to drug resistance. The chemoresistance mechanism of 5-FU is challenging to overcome because of the existence of several drug efflux transporters, DNA repair enzymes, signaling cascades, classical cellular processes, cancer stem cells, metastasis, and angiogenesis. Curcumin, a potent phytocompound derived from Curcuma longa, functions as a nuclear factor (NF)-κB inhibitor and sensitizer to numerous chemotherapeutic drugs. Piperine, an alkaloid found in Piper longum, inhibits cancer cell growth, causing cell cycle arrest and apoptosis. This review explores the mechanism of 5-FU-induced chemoresistance in colon cancer cells and the role of curcumin and piperine in enhancing the sensitivity of 5-FU-based chemotherapy. CLINICAL TRIAL REGISTRATION: Not applicable.
Collapse
Affiliation(s)
- Dorothy Bhattacharjya
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, Faculty of Engineering and Technology, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, 603 203, Chengalpattu District, Tamil Nadu, India
| | - Nageswaran Sivalingam
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, Faculty of Engineering and Technology, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, 603 203, Chengalpattu District, Tamil Nadu, India.
| |
Collapse
|
34
|
Flick KM, Demirci H, Demirci FY. Epigenetics of Conjunctival Melanoma: Current Knowledge and Future Directions. Cancers (Basel) 2024; 16:3687. [PMID: 39518125 PMCID: PMC11544918 DOI: 10.3390/cancers16213687] [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: 09/19/2024] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
The purpose of this article is to provide a literature review of the epigenetic understanding of conjunctival melanoma (CM), with a primary focus on current gaps in knowledge and future directions in research. CM is a rare aggressive cancer that predominantly affects older adults. Local recurrences and distant metastases commonly occur in CM patients; however, their prediction and management remain challenging. Hence, there is currently an unmet need for useful biomarkers and more effective treatments to improve the clinical outcomes of these patients. Like other cancers, CM occurrence and prognosis are believed to be influenced by multiple genetic and epigenetic factors that contribute to tumor development/progression/recurrence/spread, immune evasion, and primary/acquired resistance to therapies. Epigenetic alterations may involve changes in chromatin conformation/accessibility, post-translational histone modifications or the use of histone variants, changes in DNA methylation, alterations in levels/functions of short (small) or long non-coding RNAs (ncRNAs), or RNA modifications. While recent years have witnessed a rapid increase in available epigenetic technologies and epigenetic modulation-based treatment options, which has enabled the development/implementation of various epi-drugs in the cancer field, the epigenetic understanding of CM remains limited due to a relatively small number of epigenetic studies published to date. These studies primarily investigated DNA methylation, ncRNA (e.g., miRNA or circRNA) expression, or RNA methylation. While these initial epigenetic investigations have revealed some potential biomarkers and/or therapeutic targets, they had various limitations, and their findings warrant replication in independent and larger studies/samples. In summary, an in-depth understanding of CM epigenetics remains largely incomplete but essential for advancing our molecular knowledge and improving clinical management/outcomes of this aggressive disease.
Collapse
Affiliation(s)
- Kaylea M. Flick
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Hakan Demirci
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA
| | - F. Yesim Demirci
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
35
|
Prabhakaran R, Thamarai R, Sivasamy S, Dhandayuthapani S, Batra J, Kamaraj C, Karthik K, Shah MA, Mallik S. Epigenetic frontiers: miRNAs, long non-coding RNAs and nanomaterials are pioneering to cancer therapy. Epigenetics Chromatin 2024; 17:31. [PMID: 39415281 PMCID: PMC11484394 DOI: 10.1186/s13072-024-00554-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 09/25/2024] [Indexed: 10/18/2024] Open
Abstract
Cancer has arisen from both genetic mutations and epigenetic changes, making epigenetics a crucial area of research for innovative cancer prevention and treatment strategies. This dual perspective has propelled epigenetics into the forefront of cancer research. This review highlights the important roles of DNA methylation, histone modifications and non-coding RNAs (ncRNAs), particularly microRNAs (miRNAs) and long non-coding RNAs, which are key regulators of cancer-related gene expression. It explores the potential of epigenetic-based therapies to revolutionize patient outcomes by selectively modulating specific epigenetic markers involved in tumorigenesis. The review examines promising epigenetic biomarkers for early cancer detection and prognosis. It also highlights recent progress in oligonucleotide-based therapies, including antisense oligonucleotides (ASOs) and antimiRs, to precisely modulate epigenetic processes. Furthermore, the concept of epigenetic editing is discussed, providing insight into the future role of precision medicine for cancer patients. The integration of nanomedicine into cancer therapy has been explored and offers innovative approaches to improve therapeutic efficacy. This comprehensive review of recent advances in epigenetic-based cancer therapy seeks to advance the field of precision oncology, ultimately culminating in improved patient outcomes in the fight against cancer.
Collapse
Affiliation(s)
- Rajkumar Prabhakaran
- Central Research Facility, Santosh Deemed to be University, Ghaziabad, UP, India
| | - Rajkumar Thamarai
- UGC Dr. D.S. Kothari Postdoctoral Fellow, Department of Animal Science, Manonmaniam Sundaranar University, Tirunelveli, Tamil Nadu, 627012, India
| | - Sivabalan Sivasamy
- Central Research Facility, Santosh Deemed to be University, Ghaziabad, UP, India
| | | | - Jyoti Batra
- Central Research Facility, Santosh Deemed to be University, Ghaziabad, UP, India.
| | - Chinnaperumal Kamaraj
- Interdisciplinary Institute of Indian System of Medicine, Directorate of Research, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India.
| | - Krishnasamy Karthik
- Department of Mechanical Engineering, Vel Tech Rangarajan Dr. Sagunthala R&D Institute of Science and Technology, Chennai, India
| | - Mohd Asif Shah
- Department of Economics, Kardan University, Parwane Du, 1001, Kabul, Afghanistan.
- Division of Research and Development, Lovely Professional University, Phagwara, Punjab, 144001, India.
- Centre of Research Impact and Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, 140401, India.
| | - Saurav Mallik
- Department of Environmental Health, Harvard T H Chan School of Public Health, Boston, Massachusetts, 02115, United States.
- Department of Pharmacology & Toxicology, University of Arizona, Tucson, AZ, 85721, USA.
| |
Collapse
|
36
|
Gold S, Shilatifard A. Epigenetic therapies targeting histone lysine methylation: complex mechanisms and clinical challenges. J Clin Invest 2024; 134:e183391. [PMID: 39403928 PMCID: PMC11473148 DOI: 10.1172/jci183391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024] Open
Abstract
As epigenetic therapies continue to gain ground as potential treatment strategies for cancer and other diseases, compounds that target histone lysine methylation and the enzyme complexes represent a major frontier for therapeutic development. Clinically viable therapies targeting the activities of histone lysine methyltransferases (HKMT) and demethylases (HKDMs) have only recently begun to emerge following FDA approval of the EZH2 inhibitor tazemetostat in 2020 and remain limited to compounds targeting the well-studied SET domain-containing HKMTs and their opposing HKDMs. These include the H3K27 methyltransferases EZH2/EZH1, the singular H3K79 methyltransferase DOT1L, and the H3K4 methyltransferase MLL1/COMPASS as well as H3K9 and H3K36 methyltransferases. They additionally include the H3K4/9-preferential demethylase LSD1 and the H3K4-, H3K27-, and H3K36-preferential KDM5, KDM6, and KDM2 demethylase subfamilies, respectively. This Review discusses the results of recent clinical and preclinical studies relevant to all of these existing and potential therapies. It provides an update on advancements in therapeutic development, as well as more basic molecular understanding, within the past 5 years approximately. It also offers a perspective on histone lysine methylation that departs from the long-predominant "histone code" metaphor, emphasizing complex-disrupting inhibitors and proximity-based approaches rather than catalytic domain inhibitors in the outlook for future therapeutic development.
Collapse
|
37
|
Sun L, Meng H, Liu T, Zhao Q, Xia M, Zhao Z, Qian Y, Cui H, Zhong X, Chai K, Tian Y, Sun Y, Zhu B, Di J, Shui G, Zhang L, Zheng J, Guo S, Liu Y. Nucleolin malonylation as a nuclear-cytosol signal exchange mechanism to drive cell proliferation in Hepatocarcinoma by enhancing AKT translation. J Biol Chem 2024; 300:107785. [PMID: 39305961 PMCID: PMC11525140 DOI: 10.1016/j.jbc.2024.107785] [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: 06/25/2024] [Revised: 08/30/2024] [Accepted: 09/04/2024] [Indexed: 10/18/2024] Open
Abstract
Cancer cells undergo metabolic reprogramming that is intricately linked to malignancy. Protein acylations are especially responsive to metabolic changes, influencing signal transduction pathways and fostering cell proliferation. However, as a novel type of acylations, the involvement of malonylation in cancer remains poorly understood. In this study, we observed a significant reduction in malonyl-CoA levels in hepatocellular carcinoma (HCC), which correlated with a global decrease in malonylation. Subsequent nuclear malonylome analysis unveiled nucleolin (NCL) malonylation, which was notably enhanced in HCC biopsies. we demonstrated that NCL undergoes malonylation at lysine residues 124 and 398. This modification triggers the translocation of NCL from the nucleolus to nucleoplasm and cytoplasm, binding to AKT mRNA, and promoting AKT translation in HCC. Silencing AKT expression markedly attenuated HCC cell proliferation driven by NCL malonylation. These findings collectively highlight nuclear signaling in modulating AKT expression, suggesting NCL malonylation as a novel mechanism through which cancer cells drive cell proliferation.
Collapse
Affiliation(s)
- Liang Sun
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hanjing Meng
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Tao Liu
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, China
| | - Qiong Zhao
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Mingyi Xia
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zhongjun Zhao
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yuting Qian
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hao Cui
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xuefei Zhong
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Keli Chai
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yang Tian
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yang Sun
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Bao Zhu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jiehui Di
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Lianjun Zhang
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu, China; Key Laboratory of Synthetic Biology Regulatory Element, Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu, China
| | - Junnian Zheng
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Shutao Guo
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, China.
| | - Yong Liu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
38
|
Wang Q, Ma C, Mao H, Wang J. Epigenome editing by a CRISPR-Cas9-based acetyltransferase activates the ZNF334 gene to inhibit the growth of colorectal cancer. Int J Biol Macromol 2024; 277:134580. [PMID: 39122070 DOI: 10.1016/j.ijbiomac.2024.134580] [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: 04/28/2024] [Revised: 07/19/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024]
Abstract
Although therapeutic targets for colorectal cancer (CRC) treatment have been developed, the treatment outcomes are not ideal and survival rates for CRC patients remain low. It is critical to identify a specific target and develop an effective CRC treatment system. The ZNF334 gene is a newly identified member of Zinc-finger proteins (ZNFs), which is essential for key biological processes associated with tumorigenesis. Abnormal epigenetic reprogramming of the ZNF334 gene promoter region decreases its expression in CRC and further induces the occurrence of CRC. Here, we clarified that P300 in CRC can regulate the H3K9/27 ac in the ZNF334 promoter. Furthermore, histone acetylation of the ZNF334 promoter region was increased by dCas9-P300 to normalize the deficiency of ZNF334 expression, thereby inhibiting the growth of CRC. Collectively, our findings enable a facile way to affect gene expression using CRISPR/Cas9-based epigenome editing and further determine the causal link between histone acetylation and gene activation, providing a promising gene therapy strategy for the CRC treatment.
Collapse
Affiliation(s)
- Qin Wang
- School of Pharmacy, Southwest Minzu University, Chengdu, Sichuan 610225, China; BMI Center for Biomass Materials and Nanointerfaces, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan 610065, China.
| | - Chen Ma
- School of Pharmacy, Southwest Minzu University, Chengdu, Sichuan 610225, China
| | - Huixian Mao
- School of Pharmacy, Southwest Minzu University, Chengdu, Sichuan 610225, China
| | - Jin Wang
- Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 611731, China.
| |
Collapse
|
39
|
Tufail M, Jiang CH, Li N. Altered metabolism in cancer: insights into energy pathways and therapeutic targets. Mol Cancer 2024; 23:203. [PMID: 39294640 PMCID: PMC11409553 DOI: 10.1186/s12943-024-02119-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 09/09/2024] [Indexed: 09/21/2024] Open
Abstract
Cancer cells undergo significant metabolic reprogramming to support their rapid growth and survival. This study examines important metabolic pathways like glycolysis, oxidative phosphorylation, glutaminolysis, and lipid metabolism, focusing on how they are regulated and their contributions to the development of tumors. The interplay between oncogenes, tumor suppressors, epigenetic modifications, and the tumor microenvironment in modulating these pathways is examined. Furthermore, we discuss the therapeutic potential of targeting cancer metabolism, presenting inhibitors of glycolysis, glutaminolysis, the TCA cycle, fatty acid oxidation, LDH, and glucose transport, alongside emerging strategies targeting oxidative phosphorylation and lipid synthesis. Despite the promise, challenges such as metabolic plasticity and the need for combination therapies and robust biomarkers persist, underscoring the necessity for continued research in this dynamic field.
Collapse
Affiliation(s)
- Muhammad Tufail
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Can-Hua Jiang
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
- Institute of Oral Precancerous Lesions, Central South University, Changsha, China
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Ning Li
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China.
- Institute of Oral Precancerous Lesions, Central South University, Changsha, China.
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
40
|
Le M, Lu W, Tan X, Luo B, Yu T, Sun Y, Guo Z, Huang P, Zhu D, Wu Q, Ganesan A, Wen S. Design, Synthesis, and Biological Evaluation of Potent EZH2/LSD1 Dual Inhibitors for Prostate Cancer. J Med Chem 2024; 67:15586-15605. [PMID: 39196854 DOI: 10.1021/acs.jmedchem.4c01250] [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: 08/30/2024]
Abstract
As histone modification enzymes, EZH2 mediates H3K27 trimethylation (H3K27me3), whereas LSD1 removes methyl groups from H3K4me1/2 and H3K9me1/2. Synergistic anticancer effects of combining inhibitors of these two enzymes are observed in leukemia and prostate cancer. Thus, a series of EZH2/LSD1 dual inhibitors are designed and synthesized to evaluate their anticancer activity. After the structure-activity study, one of the best compounds, ML234, displayed excellent antiproliferative capacity against prostate cancer cell lines LNCAP, PC3, and 22RV1. Enzymatic assays ascertained that the anticancer potency of ML234 was mediated through coinhibition of EZH2 and LSD1. Moreover, the accumulation of H3K4me2 and H3K9me2 and the decrease of H3K27me3 induced by ML234 were verified by Western blot analysis. More importantly, the compound remarkably suppressed the tumor growth and enhanced the therapeutic efficacy of clinical drug enzalutamide in the 22RV1 xenograft mouse model, indicating that it may have potential as an anticancer agent in prostate cancer.
Collapse
Affiliation(s)
- Meiling Le
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Wenhua Lu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Xiaozhuo Tan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Bingling Luo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Tiantian Yu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Yameng Sun
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Zhirong Guo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Peng Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Daqian Zhu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Qiang Wu
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Taipa 999078, Macau
| | - A Ganesan
- School of Chemistry, Pharmacy & Pharmacology, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, United Kingdom
| | - Shijun Wen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| |
Collapse
|
41
|
Wu M, Li B, Shi L, Yang L, Liang C, Wang T, Sheng X. RNA sequencing and multiplexed immunohistochemistry reveal the factors for postoperative recurrence of stage IB-IIA cervical squamous cell carcinoma. Discov Oncol 2024; 15:422. [PMID: 39254825 PMCID: PMC11387578 DOI: 10.1007/s12672-024-01283-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 08/27/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND Stage IB-IIA Cervical Squamous Cell Carcinoma (CSCC) presents diverse clinical outcomes, the mechanisms that cause recurrence in CC patients remain unclear. The goal of this study was to identify predictive biomarkers leading to tumor recurrence in IB-IIA CSCC after surgical treatment by comparing the transcriptional and immune landscape between the recurrence and non-recurrence group. METHODS We performed mRNA sequencing and multiplexed immunohistochemistry (mIHC) analysis among stage IB-IIA patients with or without recurrence after surgical resection and were followed-up for a median of three years. RESULTS Integrated analysis indicates that the upregulated gene expression in zinc finger proteins, the activation of the PI3K/Akt pathway, and the low infiltration level of T follicular helper cells and B-cells may serve as potential recurrent biomarkers for CSCC. We also observed significant differences in the immune and genomic landscape between two groups. CONCLUSIONS These findings provide new insights into the relapse mechanisms of CSCC, which could potentially guide clinical exploration of drug targets.
Collapse
Affiliation(s)
- Meiyao Wu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China
- Department of Gynecologic Oncology Research Office, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China
- Guangzhou Key Laboratory of Targeted Therapy for Gynecologic Oncology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China
- Department of Obstetrics and Gynaecology, Queen Mary Hospital, The University of Hong Kong, Hong Kong, China
| | - Baixue Li
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China
- Department of Gynecologic Oncology Research Office, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China
- Guangzhou Key Laboratory of Targeted Therapy for Gynecologic Oncology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China
| | - Lina Shi
- Medical Department, Nanjing Geneseeq Technology Inc., Nanjing, 211899, China
| | - Lingling Yang
- Medical Department, Nanjing Geneseeq Technology Inc., Nanjing, 211899, China
| | - Chuqiao Liang
- Medical Department, Nanjing Geneseeq Technology Inc., Nanjing, 211899, China
| | - Tianhong Wang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China.
- Department of Gynecologic Oncology Research Office, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China.
- Guangzhou Key Laboratory of Targeted Therapy for Gynecologic Oncology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China.
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China.
| | - Xiujie Sheng
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China.
- Department of Gynecologic Oncology Research Office, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China.
- Guangzhou Key Laboratory of Targeted Therapy for Gynecologic Oncology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China.
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China.
| |
Collapse
|
42
|
Wahab MA, Del Gaudio N, Gargiulo B, Quagliariello V, Maurea N, Nebbioso A, Altucci L, Conte M. Exploring the Role of CBX3 as a Potential Therapeutic Target in Lung Cancer. Cancers (Basel) 2024; 16:3026. [PMID: 39272883 PMCID: PMC11394081 DOI: 10.3390/cancers16173026] [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: 07/29/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 09/15/2024] Open
Abstract
Epigenetic changes regulate gene expression through histone modifications, chromatin remodeling, and protein translation of these modifications. The PRC1 and PRC2 complexes shape gene repression via histone modifications. Specifically, the CBX protein family aids PRC1 recruitment to chromatin, impacting the progressive multistep process driving chromatin silencing. Among family members, CBX3 is a complex protein involved in aberrant epigenetic mechanisms that drive lung cancer progression. CBX3 promotes lung tumorigenesis by interacting with key pathways such as PI3K/AKT, Ras/KRAS, Wnt/β-catenin, MAPK, Notch, and p53, leading to increased proliferation, inhibition of apoptosis, and enhanced resistance to therapy. Given our current lack of knowledge, additional research is required to uncover the intricate mechanisms underlying CBX3 activity, as well as its involvement in molecular pathways and its potential biomarker evaluation. Specifically, the dissimilar roles of CBX3 could be reexamined to gain a greater insight into lung cancer pathogenesis. This review aims to provide a clear overview of the context-related molecular profile of CBX3, which could be useful for addressing clinical challenges and developing novel targeted therapies based on personalized medicine.
Collapse
Affiliation(s)
- Muhammad Aamir Wahab
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Nunzio Del Gaudio
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Biagio Gargiulo
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Vincenzo Quagliariello
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy
| | - Nicola Maurea
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy
| | - Angela Nebbioso
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
- Program of Medical Epigenetics, Vanvitelli Hospital, 80138 Naples, Italy
| | - Lucia Altucci
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
- Program of Medical Epigenetics, Vanvitelli Hospital, 80138 Naples, Italy
- Institute of Endocrinology and Oncology "Gaetano Salvatore" (IEOS), 80131 Naples, Italy
- Biogem Institute of Molecular and Genetic Biology, 83031 Ariano Irpino, Italy
| | - Mariarosaria Conte
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| |
Collapse
|
43
|
Tao J, Bian X, Zhou J, Zhang M. From microscopes to molecules: The evolution of prostate cancer diagnostics. Cytojournal 2024; 21:29. [PMID: 39391208 PMCID: PMC11464998 DOI: 10.25259/cytojournal_36_2024] [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: 03/28/2024] [Accepted: 05/27/2024] [Indexed: 10/12/2024] Open
Abstract
In the ever-evolving landscape of oncology, the battle against prostate cancer (PCa) stands at a transformative juncture, propelled by the integration of molecular diagnostics into traditional cytopathological frameworks. This synthesis not only heralds a new epoch of precision medicine but also significantly enhances our understanding of the disease's genetic intricacies. Our comprehensive review navigates through the latest advancements in molecular biomarkers and their detection technologies, illuminating the potential these innovations hold for the clinical realm. With PCa persisting as one of the most common malignancies among men globally, the quest for early and precise diagnostic methods has never been more critical. The spotlight in this endeavor shines on the molecular diagnostics that reveal the genetic underpinnings of PCa, offering insights into its onset, progression, and resistance to conventional therapies. Among the genetic aberrations, the TMPRSS2-ERG fusion and mutations in genes such as phosphatase and tensin homolog (PTEN) and myelocytomatosis viral oncogene homolog (MYC) are identified as significant players in the disease's pathology, providing not only diagnostic markers but also potential therapeutic targets. This review underscores a multimodal diagnostic approach, merging molecular diagnostics with cytopathology, as a cornerstone in managing PCa effectively. This strategy promises a future where treatment is not only tailored to the individual's genetic makeup but also anticipates the disease's trajectory, offering hope for improved prognosis and quality of life for patients.
Collapse
Affiliation(s)
- Junyue Tao
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xiaokang Bian
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jun Zhou
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Meng Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
44
|
Lee S, Lee S, Choi N, Kim J, Kweon J, Miller K, Kim J. PCAF promotes R-loop resolution via histone acetylation. Nucleic Acids Res 2024; 52:8643-8660. [PMID: 38936834 PMCID: PMC11347145 DOI: 10.1093/nar/gkae558] [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: 03/05/2024] [Revised: 06/04/2024] [Accepted: 06/19/2024] [Indexed: 06/29/2024] Open
Abstract
R-loops cause genome instability, disrupting normal cellular functions. Histone acetylation, particularly by p300/CBP-associated factor (PCAF), is essential for maintaining genome stability and regulating cellular processes. Understanding how R-loop formation and resolution are regulated is important because dysregulation of these processes can lead to multiple diseases, including cancer. This study explores the role of PCAF in maintaining genome stability, specifically for R-loop resolution. We found that PCAF depletion promotes the generation of R-loop structures, especially during ongoing transcription, thereby compromising genome stability. Mechanistically, we found that PCAF facilitates histone H4K8 acetylation, leading to recruitment of the a double-strand break repair protein (MRE11) and exonuclease 1 (EXO1) to R-loop sites. These in turn recruit Fanconi anemia (FA) proteins, including FANCM and BLM, to resolve the R-loop structure. Our findings suggest that PCAF, histone acetylation, and FA proteins collaborate to resolve R-loops and ensure genome stability. This study therefore provides novel mechanistic insights into the dynamics of R-loops as well as the role of PCAF in preserving genome stability. These results may help develop therapeutic strategies to target diseases associated with genome instability.
Collapse
Affiliation(s)
- Seo Yun Lee
- Department of Life Science and Multidisciplinary Genome Institute, Hallym University, Chuncheon 24252, Republic of Korea
| | - Soo Hyeon Lee
- Department of Life Science and Multidisciplinary Genome Institute, Hallym University, Chuncheon 24252, Republic of Korea
| | - Nak Hun Choi
- Department of Life Science and Multidisciplinary Genome Institute, Hallym University, Chuncheon 24252, Republic of Korea
| | - Ja Young Kim
- Department of Life Science and Multidisciplinary Genome Institute, Hallym University, Chuncheon 24252, Republic of Korea
| | - Jun Hee Kweon
- Department of Life Science and Multidisciplinary Genome Institute, Hallym University, Chuncheon 24252, Republic of Korea
| | - Kyle M Miller
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Jae Jin Kim
- Department of Life Science and Multidisciplinary Genome Institute, Hallym University, Chuncheon 24252, Republic of Korea
| |
Collapse
|
45
|
Porreca V, Barbagallo C, Corbella E, Peres M, Stella M, Mignogna G, Maras B, Ragusa M, Mancone C. Unveil Intrahepatic Cholangiocarcinoma Heterogeneity through the Lens of Omics and Multi-Omics Approaches. Cancers (Basel) 2024; 16:2889. [PMID: 39199659 PMCID: PMC11352949 DOI: 10.3390/cancers16162889] [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: 07/24/2024] [Revised: 08/12/2024] [Accepted: 08/16/2024] [Indexed: 09/01/2024] Open
Abstract
Intrahepatic cholangiocarcinoma (iCCA) is recognized worldwide as the second leading cause of morbidity and mortality among primary liver cancers, showing a continuously increasing incidence rate in recent years. iCCA aggressiveness is revealed through its rapid and silent intrahepatic expansion and spread through the lymphatic system leading to late diagnosis and poor prognoses. Multi-omics studies have aggregated information derived from single-omics data, providing a more comprehensive understanding of the phenomena being studied. These approaches are gradually becoming powerful tools for investigating the intricate pathobiology of iCCA, facilitating the correlation between molecular signature and phenotypic manifestation. Consequently, preliminary stratifications of iCCA patients have been proposed according to their "omics" features opening the possibility of identifying potential biomarkers for early diagnosis and developing new therapies based on personalized medicine (PM). The focus of this review is to provide new and advanced insight into the molecular pathobiology of the iCCA, starting from single- to the latest multi-omics approaches, paving the way for translating new basic research into therapeutic practices.
Collapse
Affiliation(s)
- Veronica Porreca
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (E.C.); (M.P.)
| | - Cristina Barbagallo
- Section of Biology and Genetics, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (C.B.); (M.S.); (M.R.)
| | - Eleonora Corbella
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (E.C.); (M.P.)
| | - Marco Peres
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (E.C.); (M.P.)
| | - Michele Stella
- Section of Biology and Genetics, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (C.B.); (M.S.); (M.R.)
| | - Giuseppina Mignogna
- Department of Biochemistry Science, Sapienza University of Rome, 00185 Rome, Italy; (G.M.); (B.M.)
| | - Bruno Maras
- Department of Biochemistry Science, Sapienza University of Rome, 00185 Rome, Italy; (G.M.); (B.M.)
| | - Marco Ragusa
- Section of Biology and Genetics, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (C.B.); (M.S.); (M.R.)
| | - Carmine Mancone
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (E.C.); (M.P.)
| |
Collapse
|
46
|
Huang B, Peng X, Zhai X, Hu J, Chen J, Yang S, Huang Q, Deng E, Li H, Barakat TS, Chen J, Pei D, Fan X, Chambers I, Zhang M. Inhibition of HDAC activity directly reprograms murine embryonic stem cells to trophoblast stem cells. Dev Cell 2024; 59:2101-2117.e8. [PMID: 38823394 DOI: 10.1016/j.devcel.2024.05.009] [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: 01/23/2024] [Accepted: 05/09/2024] [Indexed: 06/03/2024]
Abstract
Embryonic stem cells (ESCs) can differentiate into all cell types of the embryonic germ layers. ESCs can also generate totipotent 2C-like cells and trophectodermal cells. However, these latter transitions occur at low frequency due to epigenetic barriers, the nature of which is not fully understood. Here, we show that treating mouse ESCs with sodium butyrate (NaB) increases the population of 2C-like cells and enables direct reprogramming of ESCs into trophoblast stem cells (TSCs) without a transition through a 2C-like state. Mechanistically, NaB inhibits histone deacetylase activities in the LSD1-HDAC1/2 corepressor complex. This increases acetylation levels in the regulatory regions of both 2C- and TSC-specific genes, promoting their expression. In addition, NaB-treated cells acquire the capacity to generate blastocyst-like structures that can develop beyond the implantation stage in vitro and form deciduae in vivo. These results identify how epigenetics restrict the totipotent and trophectoderm fate in mouse ESCs.
Collapse
Affiliation(s)
- Boyan Huang
- GMU-GIBH Joint School of Life Sciences, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou National Laboratory, Guangzhou Medical University, Guangzhou 510005, China; Center for Cell Lineage and Atlas (CCLA), Bioland Laboratory, Guangzhou, China
| | - Xing Peng
- GMU-GIBH Joint School of Life Sciences, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou National Laboratory, Guangzhou Medical University, Guangzhou 510005, China
| | - Xuzhao Zhai
- GMU-GIBH Joint School of Life Sciences, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou National Laboratory, Guangzhou Medical University, Guangzhou 510005, China; Center for Cell Lineage and Atlas (CCLA), Bioland Laboratory, Guangzhou, China; Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Jie Hu
- GMU-GIBH Joint School of Life Sciences, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou National Laboratory, Guangzhou Medical University, Guangzhou 510005, China; Center for Cell Lineage and Atlas (CCLA), Bioland Laboratory, Guangzhou, China
| | - Junyu Chen
- Center for Cell Lineage and Atlas (CCLA), Bioland Laboratory, Guangzhou, China; School of Life Science, South China Normal University, Guangzhou 510005, China
| | - Suming Yang
- GMU-GIBH Joint School of Life Sciences, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou National Laboratory, Guangzhou Medical University, Guangzhou 510005, China; Center for Cell Lineage and Atlas (CCLA), Bioland Laboratory, Guangzhou, China
| | - Qingpei Huang
- GMU-GIBH Joint School of Life Sciences, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou National Laboratory, Guangzhou Medical University, Guangzhou 510005, China
| | - Enze Deng
- GMU-GIBH Joint School of Life Sciences, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou National Laboratory, Guangzhou Medical University, Guangzhou 510005, China
| | - Huanhuan Li
- GMU-GIBH Joint School of Life Sciences, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou National Laboratory, Guangzhou Medical University, Guangzhou 510005, China; Center for Cell Lineage and Atlas (CCLA), Bioland Laboratory, Guangzhou, China
| | - Tahsin Stefan Barakat
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Jiekai Chen
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510525, China; Center for Cell Lineage and Atlas (CCLA), Bioland Laboratory, Guangzhou, China
| | - Duanqing Pei
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510525, China
| | - Xiaoying Fan
- GMU-GIBH Joint School of Life Sciences, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou National Laboratory, Guangzhou Medical University, Guangzhou 510005, China.
| | - Ian Chambers
- Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh EH16 4UU, Scotland.
| | - Man Zhang
- GMU-GIBH Joint School of Life Sciences, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou National Laboratory, Guangzhou Medical University, Guangzhou 510005, China; Center for Cell Lineage and Atlas (CCLA), Bioland Laboratory, Guangzhou, China.
| |
Collapse
|
47
|
Chen T, Dumas M, Watson R, Vincoff S, Peng C, Zhao L, Hong L, Pertsemlidis S, Shaepers-Cheu M, Wang TZ, Srijay D, Monticello C, Vure P, Pulugurta R, Kholina K, Goel S, DeLisa MP, Truant R, Aguilar HC, Chatterjee P. PepMLM: Target Sequence-Conditioned Generation of Therapeutic Peptide Binders via Span Masked Language Modeling. ARXIV 2024:arXiv:2310.03842v3. [PMID: 37873004 PMCID: PMC10593082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Target proteins that lack accessible binding pockets and conformational stability have posed increasing challenges for drug development. Induced proximity strategies, such as PROTACs and molecular glues, have thus gained attention as pharmacological alternatives, but still require small molecule docking at binding pockets for targeted protein degradation. The computational design of protein-based binders presents unique opportunities to access "undruggable" targets, but have often relied on stable 3D structures or structure-influenced latent spaces for effective binder generation. In this work, we introduce PepMLM, a target sequence-conditioned generator of de novo linear peptide binders. By employing a novel span masking strategy that uniquely positions cognate peptide sequences at the C-terminus of target protein sequences, PepMLM fine-tunes the state-of-the-art ESM-2 pLM to fully reconstruct the binder region, achieving low perplexities matching or improving upon validated peptide-protein sequence pairs. After successful in silico benchmarking with AlphaFold-Multimer, outperforming RFDiffusion on structured targets, we experimentally verify PepMLM's efficacy via fusion of model-derived peptides to E3 ubiquitin ligase domains, demonstrating endogenous degradation of emergent viral phosphoproteins and Huntington's disease-driving proteins. In total, PepMLM enables the generative design of candidate binders to any target protein, without the requirement of target structure, empowering downstream therapeutic applications.
Collapse
Affiliation(s)
- Tianlai Chen
- Department of Biomedical Engineering, Duke University
| | - Madeleine Dumas
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University
- Department of Microbiology, College of Agriculture and Life Sciences, Cornell University
| | - Rio Watson
- Department of Biomedical Engineering, Duke University
| | | | - Christina Peng
- Department of Biochemistry and Biomedical Sciences, McMaster University
| | - Lin Zhao
- Department of Biomedical Engineering, Duke University
| | - Lauren Hong
- Department of Biomedical Engineering, Duke University
| | | | - Mayumi Shaepers-Cheu
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University
| | - Tian Zi Wang
- Department of Biomedical Engineering, Duke University
| | - Divya Srijay
- Department of Biomedical Engineering, Duke University
| | - Connor Monticello
- Department of Biochemistry and Biomedical Sciences, McMaster University
| | - Pranay Vure
- Department of Biomedical Engineering, Duke University
| | | | | | - Shrey Goel
- Department of Biomedical Engineering, Duke University
| | - Matthew P. DeLisa
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, USA
- Cornell Institute of Biotechnology, Cornell University, Ithaca, NY, USA
| | - Ray Truant
- Department of Biochemistry and Biomedical Sciences, McMaster University
| | - Hector C. Aguilar
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University
| | - Pranam Chatterjee
- Department of Biomedical Engineering, Duke University
- Department of Computer Science, Duke University
- Department of Biostatistics and Bioinformatics, Duke University
| |
Collapse
|
48
|
Silva-Carvalho AÉ, Filiú-Braga LDC, Bogéa GMR, de Assis AJB, Pittella-Silva F, Saldanha-Araujo F. GLP and G9a histone methyltransferases as potential therapeutic targets for lymphoid neoplasms. Cancer Cell Int 2024; 24:243. [PMID: 38997742 PMCID: PMC11249034 DOI: 10.1186/s12935-024-03441-y] [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: 01/09/2024] [Accepted: 07/08/2024] [Indexed: 07/14/2024] Open
Abstract
Histone methyltransferases (HMTs) are enzymes that regulate histone methylation and play an important role in controlling transcription by altering the chromatin structure. Aberrant activation of HMTs has been widely reported in certain types of neoplastic cells. Among them, G9a/EHMT2 and GLP/EHMT1 are crucial for H3K9 methylation, and their dysregulation has been associated with tumor initiation and progression in different types of cancer. More recently, it has been shown that G9a and GLP appear to play a critical role in several lymphoid hematologic malignancies. Importantly, the key roles played by both enzymes in various diseases made them attractive targets for drug development. In fact, in recent years, several groups have tried to develop small molecule inhibitors targeting their epigenetic activities as potential anticancer therapeutic tools. In this review, we discuss the physiological role of GLP and G9a, their oncogenic functions in hematologic malignancies of the lymphoid lineage, and the therapeutic potential of epigenetic drugs targeting G9a/GLP for cancer treatment.
Collapse
Affiliation(s)
| | | | | | - Alan Jhones Barbosa de Assis
- Laboratory of Molecular Pathology of Cancer, Faculty of Health Sciences and Medicine, University of Brasilia, Brasília, Brazil
| | - Fábio Pittella-Silva
- Laboratory of Molecular Pathology of Cancer, Faculty of Health Sciences and Medicine, University of Brasilia, Brasília, Brazil
| | - Felipe Saldanha-Araujo
- Hematology and Stem Cells Laboratory, Faculty of Health Sciences, University of Brasília, Brasilia, Brazil.
| |
Collapse
|
49
|
Huang G, Liu J, Yu A, Luo C, Zhu J, Wang Y, Dai Z, Zhang L, Feng Z, Lu J, Dong Z, Luo J, Chen W, Chen Z. Nuclear translocation of ISG15 regulated by PPP2R2B inhibits cisplatin resistance of bladder cancer. Cell Mol Life Sci 2024; 81:292. [PMID: 38976080 PMCID: PMC11335216 DOI: 10.1007/s00018-024-05320-1] [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: 02/13/2024] [Revised: 06/11/2024] [Accepted: 06/13/2024] [Indexed: 07/09/2024]
Abstract
Cisplatin resistance is a major challenge for systemic therapy against advanced bladder cancer (BC). Little information is available on the regulation of cisplatin resistance and the underlying mechanisms require elucidation. Here, we detected that downregulation of the tumor suppressor, PPP2R2B (a serine/threonine protein phosphatase 2 A regulatory subunit), in BC promoted cell proliferation and migration. What's more, low PPP2R2B expression was correlated with cisplatin resistance. In vitro and in vivo experiments verified that PPP2R2B could promote BC sensitivity to cisplatin. In terms of mechanism, we identified a novel function of PPP2R2B as a nucleocytoplasmic transport molecule. PPP2R2B promoted ISG15 entry into the nucleus by mediating binding of IPO5 with ISG15. Nuclear translocation of ISG15 inhibited DNA repair, further increasing ISG15 expression through activation of the STING pathway. Besides, PPP2R2B was down-regulated by SUV39H1-mediated histone 3 lysine 9 trimethylation, which could be restored by the SUV39H1-specific inhibitor, chaetocin. Our data suggest that PPP2R2B expression level is a potential biomarker for chemotherapy response and that chemotherapy in combination with chaetocin may be a feasible treatment strategy for patients with BC.
Collapse
Affiliation(s)
- Gaowei Huang
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Urology, Shenzhen People's Hospital (the Second Clinical Medical College, Jinan University, the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China
| | - Jinwen Liu
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Anze Yu
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Chenggong Luo
- Department of Urology, Guizhou Provincial People's Hospital, Guizhou University, Guiyang, 550002, China
| | - Jiangquan Zhu
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yinghan Wang
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Ziran Dai
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Lizhen Zhang
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zihao Feng
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jun Lu
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zhong Dong
- Department of Urology, Huizhou Central People's Hospital, Huizhou, 516001, China.
| | - Junhang Luo
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Wei Chen
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Zhenhua Chen
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
50
|
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
|