1
|
Zhou C, Qiu Q, Liu X, Zhang T, Liang L, Yuan Y, Chen Y, Sun W. Novel exosome-associated LncRNA model predicts colorectal cancer prognosis and drug response. Hereditas 2025; 162:79. [PMID: 40380258 DOI: 10.1186/s41065-025-00445-0] [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: 03/09/2025] [Accepted: 05/03/2025] [Indexed: 05/19/2025] Open
Abstract
BACKGROUND Exosomes are extracellular vesicles that carry various biological substances and have potential as functional mediators in cancers. However, little is known about special molecules in colorectal cancer (CRC) exosomes and their immunological functions. AIMS Using genomic data from the TCGA-CRC cohort, we constructed a prognostic model based on exosome-related lncRNA for the first time, and the biological role of MIR4713HG in CRC was deeply analyzed. METHOD In this study, we downloaded the gene expression data and clinical data of CRC from the TCGA database. The limma package, SVM-REF and univariate Cox analysis were used to screen out core ERG (CERG) in CRC. LASSO and multivariate Cox regression analyses were used to filter out CERG-related LncRNA and construct a risk score. We explored the distribution and expression levels of ERG in immune cell types by scRNA-seq data. xCell was used to calculate the infiltration levels of stromal cells and immune cells in CRC. KM plotter was used for immunotherapy evaluation of core ERG. Next, we further provide colony formation assay, Transwell assay and xenograft models to understand the carcinogenic effect of MIR4713HG. RESULT First, 43 differentially expressed ERG and 7 CERG were obtained. We explored the expression and distribution levels of CERG in 9 types of cells by scRNA-seq data. In addition, two key exosome-associated LncRNA (MIR4713HG and ZEB1-AS1) were obtained, and a risk score (EALncRI) was constructed. EALncRI could accurately predict the prognosis of CRC. Based on the EALncRI, we constructed a nomogram that is easy to use in clinical practice, which can more accurately and stably predict the prognosis of CRC patients. Furthermore, EALncRI was significantly correlated with the expression of 5 HLA molecules and 13 immune checkpoint molecules. MIR4713HG showed a good predictive effect in the overall survival of patients with immunotherapy evaluation. Knocking down the expression of MIR4713HG significantly inhibited proliferation and migration, and also impaired subcutaneous tumor growth in nude mice. CONCLUSION In this study, a variety of machine learning algorithms were used to construct the EALncRI based on ERG, which can effectively predict the prognosis and distinguish the immune landscape of CRC. More importantly, we conducted an in-depth study on MIR4713HG, which may become an important therapeutic target in CRC.
Collapse
Affiliation(s)
- Chi Zhou
- Department of General Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Qian Qiu
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xinyu Liu
- Bengbu Medical University, Bengbu, China
| | - Tiantian Zhang
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Leilei Liang
- Department of Gynecologic Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Yihang Yuan
- Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School Nanjing University, Nanjing, China
| | - Yufo Chen
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical University, Bengbu, China.
| | - Weijie Sun
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical University, Bengbu, China.
| |
Collapse
|
2
|
Nihira NT, Kudo R, Ohta T. Inflammation and tumor immune escape in response to DNA damage. Semin Cancer Biol 2025; 110:36-45. [PMID: 39938581 DOI: 10.1016/j.semcancer.2025.02.005] [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: 07/30/2024] [Revised: 01/08/2025] [Accepted: 02/03/2025] [Indexed: 02/14/2025]
Abstract
Senescent and cancer cells share common inflammatory characteristics, including factors of the senescence-associated secretory phenotype (SASP) and the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway. Inflammation in the tumor microenvironment not only provides an opportunity for immune cells to attack cancer cells, but also promotes cancer invasion and metastasis. Immune checkpoint molecule PD-L1 is transcriptionally induced by inflammation, and the immunological state of PD-L1-positive tumors influences the efficacy of Immune checkpoint inhibitors (ICIs). ICIs are effective against the PD-L1-positive "hot" tumors; however, the non-immunoactive "cold" tumors that express PD-L1 rarely respond to ICIs, suggesting that converting PD-L1-positive "cold" tumors into "hot" tumors would improve the efficacy of ICIs. To eliminate cancer via the innate immune system, a therapeutic strategy for manipulating inflammatory responses must be established. To date, the molecular mechanisms of inflammation-induced tumorigenesis are not yet fully understood. However, it is becoming clear that the regulatory mechanisms of inflammation in cancer via the cGAS-STING pathway play an important role in both cancer and sensescent cells. In this review, we focus on inflammation and immune escape triggered by DNA damage in cancer and senescent cells.
Collapse
Affiliation(s)
- Naoe Taira Nihira
- Department of Translational Oncology, St. Marianna University Graduate School of Medicine, Kawasaki, Japan
| | - Rei Kudo
- Division of Cancer RNA Research, National Cancer Center Research Institute, Tokyo, Japan
| | - Tomohiko Ohta
- Department of Translational Oncology, St. Marianna University Graduate School of Medicine, Kawasaki, Japan.
| |
Collapse
|
3
|
Huang J, Mao H, Wu W, Jiang S, Chen L, Ma T, Zhai C, Meng Y. Identifying Antihepatocellular Carcinoma Compounds in Gansui Banxia Decoction Using Live Cell Adsorption. Drug Des Devel Ther 2025; 19:3437-3457. [PMID: 40322038 PMCID: PMC12049131 DOI: 10.2147/dddt.s513086] [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: 12/24/2024] [Accepted: 04/04/2025] [Indexed: 05/08/2025] Open
Abstract
Purpose Gansui Banxia Decoction (GSBXD) is a traditional Chinese medicine formula for the treatment of hepatocellular carcinoma (HCC). Preliminary studies have identified the constituents and anticancer efficacy of GSBXD, but there is a gap in the screening of its lead compounds. Using live cell affinity combined with solid-phase extraction (LCA-SPE) and virtual screening and in vitro activity assays, we obtained 14 reliable potential lead compounds. Methods Coculturing H22 mouse HCC cells with GSBXD ethanol extract, isolating and purifying the bioactive fractions using LCA-SPE, and identifying the unknown bioactive components by ultraperformance liquid chromatography coupled with quadrupole time-of-flight full information tandem mass spectrometry (UPLC-QTOF-MSE) with the UNIFI information processing platform were performed. Network pharmacology and molecular docking techniques predicted the potential mechanisms of these compounds against HCC. The enzyme-linked immunosorbent assay was used to examine the effects of core compounds on the expression of p53 and Bcl-2 in vitro. Results Fourteen compounds screened from GSBXD using SPE-LCA-UPLC-QTOF-MSE may be the main bioactive components. Network pharmacology predictions suggested that protein kinases regulate Bcl-2 and p53 expression to trigger the apoptosis in cancer cells. Molecular docking identified three core compounds, namely, lactiflorin, schaftoside, and violanthin, which showed high affinities for the relevant proteins. Experimental verification confirmed their superior anticancer activity in vitro. Their anti-HCC effects likely involved in the upregulation of p53 and downregulation of Bcl-2 expression at the cellular level. Conclusion We developed a stable and accurate SPE-LCA method, which successfully isolated and characterized 14 potentially active compounds from GSBXD. They may improve HCC by promoting p53 expression and reducing Bcl-2 expression. This work lays a foundation for discovering lead compounds and exploring potential mechanisms in traditional Chinese medicine formulas.
Collapse
MESH Headings
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/metabolism
- Mice
- Animals
- Liver Neoplasms/drug therapy
- Liver Neoplasms/pathology
- Liver Neoplasms/metabolism
- Drugs, Chinese Herbal/pharmacology
- Drugs, Chinese Herbal/chemistry
- Drugs, Chinese Herbal/isolation & purification
- Antineoplastic Agents, Phytogenic/pharmacology
- Antineoplastic Agents, Phytogenic/isolation & purification
- Antineoplastic Agents, Phytogenic/chemistry
- Drug Screening Assays, Antitumor
- Molecular Docking Simulation
- Cell Proliferation/drug effects
- Humans
- Dose-Response Relationship, Drug
- Solid Phase Extraction
- Adsorption
- Structure-Activity Relationship
- Tumor Cells, Cultured
- Cell Line, Tumor
- Medicine, Chinese Traditional
- Tumor Suppressor Protein p53/metabolism
- Apoptosis/drug effects
- Proto-Oncogene Proteins c-bcl-2/metabolism
- Tandem Mass Spectrometry
Collapse
Affiliation(s)
- Jiahao Huang
- Key Laboratory of Basic and Application Research of Beiyao, Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
| | - Hongyu Mao
- Key Laboratory of Basic and Application Research of Beiyao, Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
- Sinopharm Harbin General Hospital, Harbin, Heilongjiang, People’s Republic of China
| | - Weidong Wu
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
| | - Siliang Jiang
- Key Laboratory of Basic and Application Research of Beiyao, Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
| | - Lanru Chen
- Key Laboratory of Basic and Application Research of Beiyao, Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
| | - Tianyu Ma
- Key Laboratory of Basic and Application Research of Beiyao, Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
| | - Chunmei Zhai
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
| | - Yonghai Meng
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
| |
Collapse
|
4
|
Zhang G, Zhang G, Zhao Y, Wan Y, Jiang B, Wang H. Unveiling the nexus of p53 and PD-L1: insights into immunotherapy resistance mechanisms in hepatocellular carcinoma. Am J Cancer Res 2025; 15:1410-1435. [PMID: 40371157 PMCID: PMC12070102 DOI: 10.62347/brto3272] [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: 01/26/2025] [Accepted: 03/25/2025] [Indexed: 05/16/2025] Open
Abstract
Hepatocellular carcinoma (HCC), the predominant form of primary liver cancer worldwide, continues to pose a substantial health challenge with limited treatment options for advanced stages. Despite progress in therapies such as surgery, transplantation, and targeted treatments, prognosis remains bleak for many patients. The advent of immunotherapy has revolutionized the landscape of advanced HCC treatment, offering hope for improved outcomes. However, its efficacy is limited, with a modest response rate of approximately 20% as a single-agent therapy, underscoring the urgent need to decipher mechanisms of immunotherapy resistance. Tumor protein 53 gene (TP53), a pivotal tumor suppressor gene, and Programmed death ligand 1 (PD-L1), a crucial immune checkpoint ligand, play central roles in HCC's evasion of immune responses. Understanding how tumor protein 53 (p53) influences PD-L1 expression and immune system interactions is essential for unraveling the complexities of immunotherapy resistance mechanisms. Elucidating these molecular interactions not only enhances our understanding of HCC's underlying mechanisms but also lays the foundation for developing targeted treatments that may improve outcomes for patients with advanced-stage liver cancer. Ultimately, deciphering the nexus of p53 and PD-L1 in immunotherapy resistance promises to advance treatment strategies and outcomes in the challenging landscape of HCC. This review delves into the intricate relationship between p53 and PD-L1 concerning immunotherapy resistance in HCC, offering insights that could pave the way for novel therapeutic strategies aimed at enhancing treatment efficacy and overcoming resistance in advanced stages of the disease.
Collapse
Affiliation(s)
- Guoyuan Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Taihe Hospital, Hubei University of MedicineShiyan 442000, Hubei Province, China
- Department of Hepatobiliary and Pancreatic Surgery, Hubei Provincial Clinical Research Center for Precision Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of MedicineShiyan 442000, Hubei, China
| | - Gan Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Taihe Hospital, Hubei University of MedicineShiyan 442000, Hubei Province, China
- Department of Hepatobiliary and Pancreatic Surgery, Hubei Provincial Clinical Research Center for Precision Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of MedicineShiyan 442000, Hubei, China
| | - Yixuan Zhao
- Department of Hepatobiliary and Pancreatic Surgery, Taihe Hospital, Hubei University of MedicineShiyan 442000, Hubei Province, China
- Department of Hepatobiliary and Pancreatic Surgery, Hubei Provincial Clinical Research Center for Precision Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of MedicineShiyan 442000, Hubei, China
| | - Yunyan Wan
- Department of Hepatobiliary and Pancreatic Surgery, Taihe Hospital, Hubei University of MedicineShiyan 442000, Hubei Province, China
- Department of Hepatobiliary and Pancreatic Surgery, Hubei Provincial Clinical Research Center for Precision Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of MedicineShiyan 442000, Hubei, China
| | - Bin Jiang
- Department of Hepatobiliary and Pancreatic Surgery, Taihe Hospital, Hubei University of MedicineShiyan 442000, Hubei Province, China
- Department of Hepatobiliary and Pancreatic Surgery, Hubei Provincial Clinical Research Center for Precision Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of MedicineShiyan 442000, Hubei, China
| | - Huaxiang Wang
- Department of Hepatobiliary and Pancreatic Surgery, Taihe Hospital, Hubei University of MedicineShiyan 442000, Hubei Province, China
- Department of Hepatobiliary and Pancreatic Surgery, Hubei Provincial Clinical Research Center for Precision Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of MedicineShiyan 442000, Hubei, China
| |
Collapse
|
5
|
Isermann T, Schneider KL, Wegwitz F, De Oliveira T, Conradi LC, Volk V, Feuerhake F, Papke B, Stintzing S, Mundt B, Kühnel F, Moll UM, Schulz-Heddergott R. Enhancement of colorectal cancer therapy through interruption of the HSF1-HSP90 axis by p53 activation or cell cycle inhibition. Cell Death Differ 2025:10.1038/s41418-025-01502-x. [PMID: 40204953 DOI: 10.1038/s41418-025-01502-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 03/04/2025] [Accepted: 03/21/2025] [Indexed: 04/11/2025] Open
Abstract
The stress-associated chaperone system is an actionable target in cancer therapies. It is ubiquitously upregulated in cancer tissues and enables tumorigenicity by stabilizing oncoproteins. Most inhibitors target the key component, heat-shock protein 90 (HSP90). Although HSP90 inhibitors are highly tumor-selective, they fail in clinical trials. These failures are partly due to interference with a negative regulatory feedback loop in the heat-shock response (HSR): in response to HSP90 inhibition, there is compensatory synthesis of stress-inducible chaperones, mediated by the transcription factor heat-shock-factor 1 (HSF1). We recently identified that wild-type p53 reduces the HSR by repressing HSF1 via a p21-CDK4/6-MAPK-HSF1 axis. Here, we test whether in HSP90-based therapies, simultaneous p53 activation or direct cell cycle inhibition interrupts the deleterious HSF1-HSR axis and improves the efficiency of HSP90 inhibitors. We found that the clinically relevant p53 activator Idasanutlin suppresses the HSF1-HSR activity in HSP90 inhibitor-based therapies. This combination synergistically reduces cell viability and accelerates cell death in p53-proficient colorectal cancer (CRC) cells, murine tumor-derived organoids, and patient-derived organoids (PDOs). Mechanistically, upon combination therapy, CRC cells upregulate p53-associated pathways, apoptosis, and inflammatory pathways. Likewise, in a CRC mouse model, dual HSF1-HSP90 inhibition represses tumor growth and remodels immune cell composition. Importantly, inhibition of the cyclin-dependent kinases 4/6 (CDK4/6) under HSP90 inhibition phenocopies synergistic repression of the HSR in p53-proficient CRC cells. Moreover, in p53-deficient CRC cells, HSP90 inhibition in combination with CDK4/6 inhibitors similarly suppresses the HSF1-HSR and reduces cancer growth. Likewise, p53-mutated PDOs respond to dual HSF1-HSP90 inhibition, providing a strategy to target CRC independent of the p53 status. In sum, we provide new options to improve HSP90-based therapies to enhance CRC therapies.
Collapse
Affiliation(s)
- Tamara Isermann
- Department of Molecular Oncology, University Medical Center Göttingen, Göttingen, Germany
- Laboratory of Molecular Tumor Pathology and Systems Biology, Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (DKTK); Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kim Lucia Schneider
- Department of Molecular Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Florian Wegwitz
- Department of Gynecology and Obstetrics, University Medical Center Göttingen, Göttingen, Germany
| | - Tiago De Oliveira
- Department of General, Visceral, and Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
| | - Lena-Christin Conradi
- Department of General, Visceral, and Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
| | - Valery Volk
- Institute for Pathology, Hannover Medical School, Hannover, Germany
| | | | - Björn Papke
- Laboratory of Molecular Tumor Pathology and Systems Biology, Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (DKTK); Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sebastian Stintzing
- German Cancer Consortium (DKTK); Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Hematology, Oncology and Cancer Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Bettina Mundt
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Florian Kühnel
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Ute M Moll
- Department of Pathology, Stony Brook University, Stony Brook, NY, USA
| | | |
Collapse
|
6
|
Singh SR, Bhaskar R, Ghosh S, Yarlagadda B, Singh KK, Verma P, Sengupta S, Mladenov M, Hadzi-Petrushev N, Stojchevski R, Sinha JK, Avtanski D. Exploring the Genetic Orchestra of Cancer: The Interplay Between Oncogenes and Tumor-Suppressor Genes. Cancers (Basel) 2025; 17:1082. [PMID: 40227591 PMCID: PMC11988167 DOI: 10.3390/cancers17071082] [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: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025] Open
Abstract
Cancer is complex because of the critical imbalance in genetic regulation as characterized by both the overexpression of oncogenes (OGs), mainly through mutations, amplifications, and translocations, and the inactivation of tumor-suppressor genes (TSGs), which entail the preservation of genomic integrity by inducing apoptosis to counter the malignant growth. Reviewing the intricate molecular interplay between OGs and TSGs draws attention to their cell cycle, apoptosis, and cancer metabolism regulation. In the present review, we discuss seminal discoveries, such as Knudson's two-hit hypothesis, which framed the field's understanding of cancer genetics, leading to the next breakthroughs with next-generation sequencing and epigenetic profiling, revealing novel insights into OG and TSG dysregulation with opportunities for targeted therapy. The key pathways, such as MAPK/ERK, PI3K/AKT/mTOR, and Wnt/β-catenin, are presented in the context of tumor progression. Importantly, we further highlighted the advances in therapeutic strategies, including inhibitors of KRAS and MYC and restoration of TSG function, despite which mechanisms of resistance and tumor heterogeneity pose daunting challenges. A high-level understanding of interactions between OG-TSGs forms the basis for effective, personalized cancer treatment-something to strive for in better clinical outcomes. This synthesis should integrate foundational biology with translation and, in this case, contribute to the ongoing effort against cancer.
Collapse
Affiliation(s)
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeongsan-si 38541, Republic of Korea;
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan-si 38541, Republic of Korea
| | - Shampa Ghosh
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, India
| | | | - Krishna Kumar Singh
- Symbiosis Centre for Information Technology (SCIT), Symbiosis International (Deemed University), Rajiv Gandhi InfoTech Park, Hinjawadi, Pune 411057, India
| | - Prashant Verma
- School of Management, BML Munjal University, NH8, Sidhrawali, Gurugram 122413, India
| | - Sonali Sengupta
- Department of Gastroenterology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Mitko Mladenov
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia
| | - Nikola Hadzi-Petrushev
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia
| | - 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
| | | | - 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
|
7
|
Huang L, Wang W, Zhu Z, Li Q, Li M, Zhou H, Xu Q, Wen W, Wang Q, Yu F. Novel starting points for fragment-based drug design against human heat-shock protein 90 identified using crystallographic fragment screening. IUCRJ 2025; 12:177-187. [PMID: 39819741 PMCID: PMC11878448 DOI: 10.1107/s2052252524012247] [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: 07/26/2024] [Accepted: 12/18/2024] [Indexed: 01/19/2025]
Abstract
Heat-shock protein 90 (HSP90) is a highly active molecular chaperone that plays a crucial role in cellular function. It facilitates the folding, assembly and stability of various oncogenic proteins, particularly kinases and transcription factors involved in regulating tumor growth and maintenance signaling pathways. Consequently, HSP90 inhibitors are being explored as drugs for cancer therapy. Crystallographic fragment screening is a novel screening method that has been developed in recent years for fragment-based drug discovery and is known for its high hit rate and its ability to provide direct insights into the complex structures of proteins and compounds. In this paper, high-diffraction-resolution crystals of the N-terminal domain of human HSP90α were employed in crystallographic fragment screening to discover binding fragments and binding sites. A diverse library of 800 structurally distinct fragments was screened, yielding 91 starting points for the fragment-based drug design of new HSP90α N-terminal inhibitors. Nearly a thousand crystals were measured, with 738 being processed and phased using a highly automated data-processing pipeline including data reduction and phasing, refinement and hit identification via PanDDA multi-data-set analysis. The 91 identified compounds bind to eight distinct regions of the HSP90α N-terminus, with 63 fragments located in the ATP-binding pocket and its surroundings, thus demonstrating the potential for the development of HSP90α- and ATP-binding inhibitors. This study emphasizes crystallographic fragment screening as a powerful method that can effectively identify fragment molecules and inhibitors that bind to HSP90α, contributing to ongoing efforts in cancer drug discovery.
Collapse
Affiliation(s)
- Liqing Huang
- Shanghai Institute of Applied Physics, Chinese Academy of SciencesShanghai201800People’s Republic of China
- University of Chinese Academy of SciencesBeijing100049People’s Republic of China
| | - Weiwei Wang
- Shanghai Synchrotron Radiation FacilityShanghai Advanced Research Institute, Chinese Academy of SciencesShanghai201204People’s Republic of China
| | - Zhimin Zhu
- Shanghai Institute of Applied Physics, Chinese Academy of SciencesShanghai201800People’s Republic of China
- University of Chinese Academy of SciencesBeijing100049People’s Republic of China
| | - Qianhui Li
- Shanghai Institute of Applied Physics, Chinese Academy of SciencesShanghai201800People’s Republic of China
- University of Chinese Academy of SciencesBeijing100049People’s Republic of China
| | - Minjun Li
- Shanghai Synchrotron Radiation FacilityShanghai Advanced Research Institute, Chinese Academy of SciencesShanghai201204People’s Republic of China
| | - Huan Zhou
- Shanghai Institute of Applied Physics, Chinese Academy of SciencesShanghai201800People’s Republic of China
- Shanghai Synchrotron Radiation FacilityShanghai Advanced Research Institute, Chinese Academy of SciencesShanghai201204People’s Republic of China
| | - Qin Xu
- Shanghai Institute of Applied Physics, Chinese Academy of SciencesShanghai201800People’s Republic of China
- Shanghai Synchrotron Radiation FacilityShanghai Advanced Research Institute, Chinese Academy of SciencesShanghai201204People’s Republic of China
| | - Wen Wen
- Shanghai Institute of Applied Physics, Chinese Academy of SciencesShanghai201800People’s Republic of China
- Shanghai Synchrotron Radiation FacilityShanghai Advanced Research Institute, Chinese Academy of SciencesShanghai201204People’s Republic of China
| | - Qisheng Wang
- Shanghai Institute of Applied Physics, Chinese Academy of SciencesShanghai201800People’s Republic of China
- Shanghai Synchrotron Radiation FacilityShanghai Advanced Research Institute, Chinese Academy of SciencesShanghai201204People’s Republic of China
| | - Feng Yu
- Shanghai Institute of Applied Physics, Chinese Academy of SciencesShanghai201800People’s Republic of China
- Shanghai Synchrotron Radiation FacilityShanghai Advanced Research Institute, Chinese Academy of SciencesShanghai201204People’s Republic of China
| |
Collapse
|
8
|
Butera A, Amelio I. Deciphering the significance of p53 mutant proteins. Trends Cell Biol 2025; 35:258-268. [PMID: 38960851 DOI: 10.1016/j.tcb.2024.06.003] [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: 04/29/2024] [Revised: 06/07/2024] [Accepted: 06/10/2024] [Indexed: 07/05/2024]
Abstract
Mutations in the p53 gene compromise its role as guardian of genomic integrity, yielding predominantly missense p53 mutant proteins. The gain-of-function hypothesis has long suggested that these mutant proteins acquire new oncogenic properties; however, recent studies challenge this notion, indicating that targeting these mutants may not impact the fitness of cancer cells. Mounting evidence indicates that tumorigenesis involves a cooperative interplay between driver mutations and cellular state, influenced by developmental stage, external insults, and tissue damage. Consistently, the behavior and properties of p53 mutants are altered by the context. This article aims to provide a balanced summary of the evolving evidence regarding the contribution of p53 mutants in the biology of cancer while contemplating alternative frameworks to decipher the complexity of p53 mutants within their physiological contexts.
Collapse
Affiliation(s)
- Alessio Butera
- Chair of Systems Toxicology, University of Konstanz, Konstanz, Germany
| | - Ivano Amelio
- Chair of Systems Toxicology, University of Konstanz, Konstanz, Germany.
| |
Collapse
|
9
|
Zhang XF, Chen Q, Jiang Q, Hu QY. Targeting STAT3 with SH-4-54 suppresses stemness and chemoresistance in cancer stem-like cells derived from colorectal cancer. World J Clin Oncol 2025; 16:97296. [PMID: 39995553 PMCID: PMC11686562 DOI: 10.5306/wjco.v16.i2.97296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/26/2024] [Accepted: 11/19/2024] [Indexed: 12/11/2024] Open
Abstract
BACKGROUND Over the years, the numbers of treatment options for colorectal cancer (CRC) have increased, leading to notable improvements in the overall survival of CRC patients. Although therapy may initially yield positive results, the development of drug resistance can result in treatment failure and cancer recurrence. This resistance is often attributed to the presence of cancer stem cells (CSCs). These CSCs not only contribute to therapeutic resistance but also play crucial roles in the initiation and development of tumor metastasis. AIM To investigate the antitumor effects of SH-4-54, which are mediated by targeting CSCs relative to treatment outcomes. METHODS CSCs were enriched by culturing CRC cells in serum-free medium. Hallmarks of stemness and IL-6/JAK2/STAT3 signaling were detected by Western blotting. Indicators of CSC malignancy, including proliferation, invasion, and tumor formation, were measured. RESULTS In this study, we employed SH-4-54, which exhibits anticancer activity in solid tumors through targeting the SH2 domain of both the signal transducer and activator of transcription (STAT)3 and the STAT5, and evaluated its effects on stemness and chemoresistance in colorectal CSCs. As expected, SH-4-54 treatment inhibited the phosphorylation of STAT3 (p-STAT3) and decreased the percentage of ALDH1A1-positive CRC cells. The addition of SH-4-54 dissociated colorectal spheroids and decreased the expression of stemness markers, including ALDH1A1, CD44 and Nanog. SH-4-54 treatment decreased IL-6/JAK2/STAT3 signaling by inhibiting p-STAT3 and thus inhibited spheroid formation by SW480 and LoVo cells. Moreover, SH-4-54 treatment inhibited indicators of malignancy, including cell proliferation, invasion, and tumor formation, in CSCs in vitro and in vivo. Notably, SH-4-54 treatment significantly increased chemosensitivity to oxaplatin. CONCLUSION Taken together, these results indicate that SH-4-54 is a promising molecule that exerts antitumor effects on colorectal CSCs by inhibiting STAT3 signaling.
Collapse
Affiliation(s)
- Xu-Fan Zhang
- Department of Nuclear Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 641000, Sichuan Province, China
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan Province, China
| | - Qian Chen
- Department of Nuclear Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 641000, Sichuan Province, China
| | - Qin Jiang
- Department of Laboratory Medicine, Hospital of Mianyang Traditional Chinese Medicine, Chengdu 641000, Sichuan Province, China
| | - Qiong-Ying Hu
- Department of Laboratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan Province, China
| |
Collapse
|
10
|
Moniruzzaman M, Wong KY, Janjua TI, Martin JH, Begun J, Popat A. Cannabidiol Targets Colorectal Cancer Cells via Cannabinoid Receptor 2, Independent of Common Mutations. ACS Pharmacol Transl Sci 2025; 8:543-556. [PMID: 39974647 PMCID: PMC11833734 DOI: 10.1021/acsptsci.4c00644] [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: 11/05/2024] [Revised: 12/05/2024] [Accepted: 12/11/2024] [Indexed: 02/21/2025]
Abstract
Cannabidiol (CBD) is a non-neurotoxic, phytocannabinoid from cannabis with reported medicinal properties, including antiepileptic and anti-inflammatory activity. Several in vitro and in vivo studies have shown that CBD has antitumor potential against colorectal cancer (CRC), the third deadliest cancer in the world. However, as different mutations influence the antitumor effects and CBD can bind a variety of receptors, it is yet to be determined whether specific CRC mutations affect CBD's efficacy in treatment of CRC. To investigate this, we selected four CRC cell lines, including HCT116, HT-29, LS174T, and LS153, which harbor distinct mutations. Cells were treated with a range of concentrations of CBD to evaluate its cytotoxic effects and impact on cell proliferation, migration, and invasion by using a live-cell imaging system. IC50 values were then calculated for each parameter. The level of endoplasmic reticulum (ER) stress pathway markers was also measured using qRTPCR. The requirements for CB1 or CB2 receptor-medicated signaling were investigated using the selective inhibitors AM251 and SR144528, respectively. Our results demonstrate that CBD induces apoptosis and halts proliferation, migration, and invasion of CRC cell lines in a concentration-dependent manner. CBD showed potent antitumor effects in the tested cell lines with no obvious effect from different mutations such as KRAS, BRAF, APC, PTEN, etc. CBD also induced ER stress in CRC cells but not in healthy intestinal organoids. Cotreatment with SR144528 inhibited the effects of indicating involvement of CB2 receptor activation in the anticancer effects of CBD. Together, these results demonstrated that CBD could be effective for CRC regardless of the underlying mutation through CB2 receptor activation.
Collapse
Affiliation(s)
- Md Moniruzzaman
- School
of Pharmacy, The University of Queensland, Brisbane 4102, Australia
- Inflammatory
Bowel Diseases Group, Mater Research Institute—UQ at Translational
Research Institute, The University of Queensland, Brisbane 4102, Australia
- Faculty
of Medicine, The University of Queensland, Brisbane 4102, Australia
| | - Kuan Yau Wong
- Immunopathology
Group, Mater Research Institute—UQ at Translational Research
Institute, The University of Queensland, Brisbane 4102, Australia
| | | | - Jennifer H. Martin
- Clinical
Pharmacology, School of Medicine and Public Health, University of Newcastle, Hunter Medical Research Institute, Kookaburra Circuit, Newcastle 2308, Australia
| | - Jakob Begun
- Inflammatory
Bowel Diseases Group, Mater Research Institute—UQ at Translational
Research Institute, The University of Queensland, Brisbane 4102, Australia
- Faculty
of Medicine, The University of Queensland, Brisbane 4102, Australia
| | - Amirali Popat
- School
of Pharmacy, The University of Queensland, Brisbane 4102, Australia
| |
Collapse
|
11
|
Jin Y, Liu H, Wang Y, Zhang R, Wang Q, Wang Y, Cui H, Wang X, Bian Y. Pathogenesis and treatment of colitis-associated colorectal cancer: Insights from Traditional Chinese Medicine. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119096. [PMID: 39532222 DOI: 10.1016/j.jep.2024.119096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 10/11/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Inflammatory Bowel Disease (IBD) is an inflammatory intestinal disease, and with prolonged illness duration, the annual risk of IBD progressing to colitis-associated colorectal cancer (CAC) gradually increases. In recent years, there has been a noticeable trend towards the application of traditional Chinese medicine (TCM) in the treatment of CAC. AIM OF THIS REVIEW This comprehensive review summarizes the pathogenesis of CAC and details the therapeutic benefits of TCM in treating CAC, including various TCM prescriptions and ingredients, establishing the theoretical foundation for the application of TCM in CAC treatment. METHODS We assessed literature published before March 24, 2024, from several databases, including Web of Science, PubMed, Scopus and Google Scholar. The keywords used include "traditional Chinese medicine", "traditional Chinese medicine prescriptions", "traditional Chinese medicine ingredients", "herbal medicine", "colitis-associated colorectal cancer", "inflammatory bowel disease", "colorectal cancer" and "colitis-cancer transformation". We conducted a comprehensive collection and collation of pertinent scientific articles from various databases, focusing on the efficacy of TCM in the prevention and treatment of "colitis-cancer transformation". RESULTS This paper provides a concise summary and thorough analysis of twenty-eight prescriptions and ingredients of TCM for the prevention and treatment of CAC, based on existing experimental and clinical research. There are positive signs that TCM can effectively prevent and treat the "colitis-cancer transformation" through repairing the intestinal mucosal barrier, correcting intestinal flora imbalance, and regulating intestinal immune responses. CONCLUSION TCM possesses comprehensive regulatory advantages that are multifaceted, multilevel, and multitarget. It has a definite curative effect in the prevention and treatment of CAC. It is essential to enhance the clinical efficacy of TCM in the prevention and treatment of CAC based on syndrome differentiation and treatment, with the assistance of modern medicine.
Collapse
Affiliation(s)
- Yutong Jin
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Haizhao Liu
- Department of Integrated Traditional Chinese and Western Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, 300192, China
| | - Yuhui Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Ruixuan Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Qiaochu Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300073, China
| | - Yao Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Huantian Cui
- First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, China.
| | - Xiangling Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Yuhong Bian
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
12
|
Li M, Chen H, Yang X, Zhang W, Ma C, Wang Q, Wang X, Gao R. Conditional knockout of the NSD2 gene in mouse intestinal epithelial cells inhibits colorectal cancer progression. Animal Model Exp Med 2025; 8:322-331. [PMID: 38400589 PMCID: PMC11871126 DOI: 10.1002/ame2.12392] [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: 08/18/2023] [Accepted: 01/16/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND Nuclear receptor-binding SET domain 2 (NSD2) is a histone methyltransferase, that catalyzes dimethylation of lysine 36 of histone 3 (H3K36me2) and is associated with active transcription of a series of genes. NSD2 is overexpressed in multiple types of solid human tumors and has been proven to be related to unfavorable prognosis in several types of tumors. METHODS We established a mouse model in which the NSD2 gene was conditionally knocked out in intestinal epithelial cells. We used azoxymethane and dextran sodium sulfate to chemically induce murine colorectal cancer. The development of colorectal tumors were investigated using post-necropsy quantification, immunohistochemistry, and enzyme-linked immunosorbent assay (ELISA). RESULTS Compared with wild-type (WT) control mice, NSD2fl/fl-Vil1-Cre mice exhibited significantly decreased tumor numbers, histopathological changes, and cytokine expression in colorectal tumors. CONCLUSIONS Conditional knockout of NSD2 in intestinal epithelial cells significantly inhibits colorectal cancer progression.
Collapse
Affiliation(s)
- Mengyuan Li
- National Human Diseases Animal Model Resource CenterInstitute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
| | - Hanxue Chen
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
| | - Xingjiu Yang
- National Human Diseases Animal Model Resource CenterInstitute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
| | - Wenlong Zhang
- National Human Diseases Animal Model Resource CenterInstitute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
| | - Chengyan Ma
- National Human Diseases Animal Model Resource CenterInstitute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
| | - Qinghong Wang
- National Human Diseases Animal Model Resource CenterInstitute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
| | - Xinpei Wang
- National Human Diseases Animal Model Resource CenterInstitute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
| | - Ran Gao
- National Human Diseases Animal Model Resource CenterInstitute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
| |
Collapse
|
13
|
Wang C, Lu M, Chen C, Chen J, Cai Y, Wang H, Tao L, Yin W, Chen J. Integrating scRNA-seq and Visium HD for the analysis of the tumor microenvironment in the progression of colorectal cancer. Int Immunopharmacol 2025; 145:113752. [PMID: 39642568 DOI: 10.1016/j.intimp.2024.113752] [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: 10/29/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Colorectal cancer (CRC) development is a complex, multi-stage process, transitioning from normal to adenomatous tissue, and then to invasive carcinoma. Despite research, there's a knowledge gap on using high-resolution spatial omics to understand CRC's tumor microenvironment dynamics. METHODS We used single-cell transcriptomics to study major biological changes and cell interactions in CRC progression. Additionally, high-resolution spatial transcriptomics helped us examine the spatial distribution of cells with significant pathway changes, offering insights into the tumor microenvironment's development throughout CRC stages. RESULTS In the progression of CRC, plasma cells, neutrophils, and fibroblasts exhibit the most significant changes in hallmark pathways, while epithelial cells show the most pronounced alterations in metabolic pathways. We also identified a population of NOTUM + epithelial cells and IGHG1/3 + plasma cells that are concentrated at the boundary between normal tissue and adenomas. Pathway analysis further suggests that these NOTUM + cells activate numerous cancer-related pathways, despite the absence of significant pathological morphological changes. Additionally, we conducted a targeted drug prediction analysis to identify potential therapeutic agents for NOTUM-expressing epithelial cells. CONCLUSIONS Analyzing scRNA-seq and Visium HD data, we found that IGHG1/3 + plasma cells and tumor-associated neutrophil (TANs) may significantly affect colorectal tissue transformation from normal to adenoma and carcinoma. These cells are concentrated at the transition between normal and adenomatous tissue. We also found NOTUM-expressing cells at the edge of normal and adenomatous areas, possibly indicating a morphological transition as normal cells evolve into adenoma cells.
Collapse
Affiliation(s)
- Chun Wang
- Department of Pathology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Mengying Lu
- Department of Pathology, Peking University Shenzhen Hospital, Shenzhen, China; School of Medicine,Southern University of Science and Technology, Shenzhen, China
| | - Cuimin Chen
- Department of Pathology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jiajun Chen
- Department of Pathology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Yusi Cai
- Department of Pathology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Hao Wang
- Department of Pathology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Lili Tao
- Department of Pathology, Peking University Shenzhen Hospital, Shenzhen, China; School of Medicine,Southern University of Science and Technology, Shenzhen, China
| | - Weihua Yin
- Department of Pathology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jiakang Chen
- Department of Pathology, Peking University Shenzhen Hospital, Shenzhen, China.
| |
Collapse
|
14
|
Su J, Wang H, Wang Z. The Multiple Roles of Heat Shock Proteins in the Development of Inflammatory Bowel Disease. Curr Mol Med 2025; 25:132-145. [PMID: 38465431 DOI: 10.2174/0115665240286793240306053111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/31/2024] [Accepted: 02/22/2024] [Indexed: 03/12/2024]
Abstract
Inflammatory bowel disease (IBD), a chronic inflammatory condition of the human intestine, comprises Crohn's disease (CD) and ulcerative colitis (UC). IBD causes severe gastrointestinal symptoms and increases the risk of developing colorectal carcinoma. Although the etiology of IBD remains ambiguous, complex interactions between genetic predisposition, microbiota, epithelial barrier, and immune factors have been implicated. The disruption of intestinal homeostasis is a cardinal characteristic of IBD. Patients with IBD exhibit intestinal microbiota dysbiosis, impaired epithelial tight junctions, and immune dysregulation; however, the relationship between them is not completely understood. As the largest body surface is exposed to the external environment, the gastrointestinal tract epithelium is continuously subjected to environmental and endogenous stressors that can disrupt cellular homeostasis and survival. Heat shock proteins (HSPs) are endogenous factors that play crucial roles in various physiological processes, such as maintaining intestinal homeostasis and influencing IBD progression. Specifically, HSPs share an intricate association with microbes, intestinal epithelium, and the immune system. In this review, we aim to elucidate the impact of HSPs on IBD development by examining their involvement in the interactions between the intestinal microbiota, epithelial barrier, and immune system. The recent clinical and animal models and cellular research delineating the relationship between HSPs and IBD are summarized. Additionally, new perspectives on IBD treatment approaches have been proposed.
Collapse
Affiliation(s)
- Jinfeng Su
- Department of Neonatology, Shenzhen Baoan Women's and Children's Hospital, Jinan University, Shenzhen, 518100, China
| | - Haiyan Wang
- Department of Obstetrics and Gynecology, Shenzhen Baoan Women's and Children's Hospital, Jinan University, Shenzhen, 518100, China
| | - Zun Wang
- Department of Breast and Thyroid Surgery, Shenzhen Baoan Women's and Children's Hospital, Jinan University, Shenzhen, 518100, China
| |
Collapse
|
15
|
Nakayama M, Saito H, Murakami K, Oshima H, Oshima M. Missense Mutant p53 Transactivates Wnt/β-Catenin Signaling in Neighboring p53-Destabilized Cells through the COX-2/PGE2 Pathway. CANCER RESEARCH COMMUNICATIONS 2025; 5:13-23. [PMID: 39641656 PMCID: PMC11695814 DOI: 10.1158/2767-9764.crc-24-0471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/08/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024]
Abstract
SIGNIFICANCE There is intratumor heterogeneity in the stabilization of missense mutant p53, and it has been thought that only cells with nuclear accumulation of mutant p53 have oncogenic function. However, using mouse intestinal tumor-derived organoids, we show that mutant p53-stabilized cells transactivate Wnt/β-catenin signaling in neighboring p53-destabilized cells through activating the COX-2/PGE2 pathway. These results suggest that both p53-stabilized cells and p53-destabilized cells contribute to malignant progression through interaction within the intratumor microenvironment.
Collapse
Affiliation(s)
- Mizuho Nakayama
- Division of Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
- WPI Nano-Life Science Institute (NanoLSI), Kanazawa University, Kanazawa, Japan
| | - Hiroshi Saito
- Division of Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
- Department of Gastrointestinal Surgery, Kanazawa University, Kanazawa, Japan
| | - Kazuhiro Murakami
- Division of Epithelial Stem Cell Biology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Hiroko Oshima
- Division of Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
- WPI Nano-Life Science Institute (NanoLSI), Kanazawa University, Kanazawa, Japan
| | - Masanobu Oshima
- Division of Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
- WPI Nano-Life Science Institute (NanoLSI), Kanazawa University, Kanazawa, Japan
| |
Collapse
|
16
|
Xiang Z, Wang Y, Ma X, Song S, He Y, Zhou J, Feng L, Yang S, Wu Y, Yu B, Xia G, Xu W, Zhao Y, Wang L. Targeting the NOTCH2/ADAM10/TCF7L2 Axis-Mediated Transcriptional Regulation of Wnt Pathway Suppresses Tumor Growth and Enhances Chemosensitivity in Colorectal Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2405758. [PMID: 39601111 PMCID: PMC11744699 DOI: 10.1002/advs.202405758] [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: 05/26/2024] [Revised: 09/12/2024] [Indexed: 11/29/2024]
Abstract
Wnt/β-catenin/transcription factor (TCF) transcriptional activity plays an integral role in colorectal cancer (CRC) carcinogenesis. However, to date, no drugs targeting this pathway are used in clinical practice owing to the undesirable and serious side effects. In this study, it is found that the transcriptional regulation of Wnt pathway is activated and associated with liver metastasis in CRC. Through high-throughput screening of 24 inhibitors on 12 CRC and three colorectal organoids in this organoid living biobank, adavivint is found to exhibit anti-tumor activity and low toxicity in colorectal organoids, independent of the canonical Wnt/β-catenin signaling. Mechanistically, ADAM10 is screened as a target of adavivint to specifically regulate the protein expression of NOTCH2, which mediates the transcriptional regulation of the Wnt pathway. NOTCH2 not directly interact with TCF7-like 2 (TCF7L2), a key downstream transcriptional factor of canonical Wnt/β-catenin signaling, but directly activated the transcription of TCF7L2 and Wnt target genes, such as MYC, JUN and CCND1/2. Furthermore, use of adavivint or blockage of ADAM10/NOTCH2/TCF7L2 signaling enhances the chemosensitivity of CRC cells. Overall, this study provides a promising candidate for the development of small-molecule inhibitors and reveals a potential therapeutic target for CRC.
Collapse
Affiliation(s)
- Zhen Xiang
- Department of Hepatic SurgeryFudan University Shanghai Cancer Center270 Dong‐An RoadShanghai200032China
| | - Yiwei Wang
- Department of general surgeryShanghai Jiao Tong University Affiliated Sixth People's Hospital600 Yishan RdShanghai200233P. R. China
| | - Xiao Ma
- Fudan University Shanghai Cancer Center270 Dong‐An RoadShanghai200032P. R. China
| | - Shuzheng Song
- Department of Colorectal SurgeryDepartment of General SurgeryShanghai East HospitalTongji University School of Medicine150 Jimo RoadShanghai200120P. R. China
| | - Yuanqiao He
- Center of Laboratory Animal ScienceNanchang UniversityNo.999, Xuefu RoadNanchang330031P. R. China
| | - Jiamin Zhou
- Department of Hepatic SurgeryFudan University Shanghai Cancer Center270 Dong‐An RoadShanghai200032China
| | - Longhai Feng
- Department of Colorectal SurgeryThe Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital)1 Banshan East RoadHangzhou310022P. R. China
| | - Su Yang
- Department of Thoracic SurgeryRuijin HospitalShanghai Jiaotong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Yibin Wu
- Department of Hepatic SurgeryFudan University Shanghai Cancer Center270 Dong‐An RoadShanghai200032China
| | - Bingran Yu
- Department of Hepatic SurgeryFudan University Shanghai Cancer Center270 Dong‐An RoadShanghai200032China
| | - Guangkai Xia
- Department of general surgeryShanghai Jiao Tong University Affiliated Sixth People's Hospital600 Yishan RdShanghai200233P. R. China
| | - Weiqi Xu
- Department of Hepatic SurgeryFudan University Shanghai Cancer Center270 Dong‐An RoadShanghai200032China
| | - Yiming Zhao
- Department of Hepatic SurgeryFudan University Shanghai Cancer Center270 Dong‐An RoadShanghai200032China
| | - Lu Wang
- Department of Hepatic SurgeryFudan University Shanghai Cancer Center270 Dong‐An RoadShanghai200032China
| |
Collapse
|
17
|
Nakamoto-Matsubara R, Nardi V, Horick N, Fukushima T, Han RS, Shome R, Ochi K, Panaroni C, Fulzele K, Rexha F, Branagan AR, Cirstea D, Yee AJ, Scadden DT, Raje NS. Integration of clinical outcomes and molecular features in extramedullary disease in multiple myeloma. Blood Cancer J 2024; 14:224. [PMID: 39715752 DOI: 10.1038/s41408-024-01190-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 10/31/2024] [Accepted: 11/13/2024] [Indexed: 12/25/2024] Open
Abstract
Multiple myeloma (MM) remains incurable despite novel therapeutics. A major contributor to the development of relapsed/refractory and resistant MM is extraosseous extramedullary disease (EMD), whose molecular biology is still not fully understood. We analyzed 528 MM patients who presented to our institution between 2014 and 2021 and who had undergone molecular testing. We defined EMD as organ plasmacytoma distinct from bones and evaluated patients for the development of EMD with the goal of defining their molecular characteristics. Here, we show that RAS/BRAF mutations are likely essential for the development of EMD. Our results also indicate that the underlying reason for the negative outcomes in patients with poor prognostic factors such as duplication 1q and deletion 17p is largely due to the development of EMD. However, the presence of TP53 mutation remains a poor prognostic factor regardless of EMD development. Furthermore, mutation sites of TP53 were different between EMD versus non-EMD patients, with gain-of-function mutations enriched in patients with EMD. Our data highlights distinct molecular abnormalities in patients with EMD and provides potential mechanistic insights for novel therapeutic targets for the future.
Collapse
Affiliation(s)
- Rie Nakamoto-Matsubara
- Center for Multiple Myeloma, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Valentina Nardi
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Nora Horick
- Biostatistics Center, Massachusetts General Hospital, Boston, MA, USA
| | - Tsuyoshi Fukushima
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ryan S Han
- Center for Multiple Myeloma, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Rajib Shome
- Center for Multiple Myeloma, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Kiyosumi Ochi
- Center for Multiple Myeloma, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Cristina Panaroni
- Center for Multiple Myeloma, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Keertik Fulzele
- Center for Multiple Myeloma, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Farah Rexha
- Center for Multiple Myeloma, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Andrew R Branagan
- Center for Multiple Myeloma, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Diana Cirstea
- Center for Multiple Myeloma, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Andrew J Yee
- Center for Multiple Myeloma, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - David T Scadden
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Noopur S Raje
- Center for Multiple Myeloma, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
18
|
Shi M, Yang Y, Huang N, Zeng D, Mo Z, Wang J, Zhang X, Liu R, Wang C, Rong X, Wu Z, Huang Q, Shang H, Tang J, Wang Z, Cai J, Huang G, Guan Y, Guo J, Mu Q, Wang J, Liao W. Genetic and microenvironmental evolution of colorectal liver metastases under chemotherapy. Cell Rep Med 2024; 5:101838. [PMID: 39631402 PMCID: PMC11722126 DOI: 10.1016/j.xcrm.2024.101838] [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: 05/17/2024] [Revised: 09/16/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024]
Abstract
Drug resistance limits the efficacy of chemotherapy for colorectal cancer liver metastasis (CRLM). However, the evolution of CRLM during drug treatment remains poorly elucidated. Multi-omics and treatment response data from 115 samples of 49 patients with CRLM undergoing bevacizumab (BVZ)-based chemotherapy show little difference in genomic alterations in 92% of cases, while remarkable differences are observed at the transcriptomic level. By decoupling intrinsic and acquired resistance, we find that hepatocyte and myeloid cell infiltration contribute to 38.5% and 23.1% of acquired resistance, respectively. Importantly, SMAD4 mutations and chr20q copy-number gain are associated with intrinsic chemoresistance. Gene interference experiments suggest that SMAD4R361H/C mutations confer BVZ and 5-fluorouracil (5-FU) resistance through STAT3 signaling. Notably, supplementing BVZ and 5-FU with the STAT3 inhibitor GB201 restores therapeutic efficacy in SMAD4R361H/C cancer cells. Our study uncovers the evolutionary dynamics of CRLM and its microenvironment during treatment and offers strategies to overcome drug resistance.
Collapse
Affiliation(s)
- Min Shi
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China; Cancer Center, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China; Foshan Key Laboratory of Translational Medicine in Oncology, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China
| | - Yingxi Yang
- Department of Chemical and Biological Engineering, Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Na Huang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Dongqiang Zeng
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China; Cancer Center, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China; Foshan Key Laboratory of Translational Medicine in Oncology, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China
| | - Zongchao Mo
- Department of Chemical and Biological Engineering, Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Jiao Wang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Xiaomeng Zhang
- Department of Chemical and Biological Engineering, Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Ran Liu
- Department of Chemical and Biological Engineering, Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Chunlin Wang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Xiaoxiang Rong
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Zhenzhen Wu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Qiong Huang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Haixia Shang
- Department of Chemical and Biological Engineering, Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Jihong Tang
- Department of Chemical and Biological Engineering, Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Zhaojun Wang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Jianan Cai
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Genjie Huang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Yijin Guan
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Jian Guo
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Quanhua Mu
- Department of Chemical and Biological Engineering, Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Jiguang Wang
- Department of Chemical and Biological Engineering, Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China; SIAT-HKUST Joint Laboratory of Cell Evolution and Digital Health, HKUST Shenzhen-Hong Kong Collaborative Innovation Research Institute, Futian, Shenzhen 518000, P.R. China.
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China; Cancer Center, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China; Foshan Key Laboratory of Translational Medicine in Oncology, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China.
| |
Collapse
|
19
|
Gilardini Montani MS, Benedetti R, Cirone M. Targeting EZH2 in Cancer: Mechanisms, Pathways, and Therapeutic Potential. Molecules 2024; 29:5817. [PMID: 39769907 PMCID: PMC11678268 DOI: 10.3390/molecules29245817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
Enhancer of zeste homolog 2 (EZH2) is a methyltransferase involved in cell cycle regulation, cell differentiation, and cell death and plays a role in modulating the immune response. Although it mainly functions by catalyzing the tri-methylation of H3 histone on K27 (H3K27), to inhibit the transcription of target genes, EZH2 can directly methylate several transcription factors or form complexes with them, regulating their functions. EZH2 expression/activity is often dysregulated in cancer, contributing to carcinogenesis and immune escape, thereby representing an important target in anti-cancer therapy. This review summarizes some of the mechanisms through which EZH2 regulates the expression and function of tumor suppressor genes and oncogenic molecules such as STAT3, mutant p53, and c-Myc and how it modulates the anti-cancer immune response. The influence of posttranslational modifications on EZH2 activity and stability and the possible strategies leading to its inhibition are also reviewed.
Collapse
Affiliation(s)
| | | | - Mara Cirone
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy;
| |
Collapse
|
20
|
Ahmadi SE, Rahimian E, Rahimi S, Zarandi B, Bahraini M, Soleymani M, Safdari SM, Shabannezhad A, Jaafari N, Safa M. From regulation to deregulation of p53 in hematologic malignancies: implications for diagnosis, prognosis and therapy. Biomark Res 2024; 12:137. [PMID: 39538363 PMCID: PMC11565275 DOI: 10.1186/s40364-024-00676-9] [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: 07/16/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
The p53 protein, encoded by the TP53 gene, serves as a critical tumor suppressor, playing a vital role in maintaining genomic stability and regulating cellular responses to stress. Dysregulation of p53 is frequently observed in hematological malignancies, significantly impacting disease progression and patient outcomes. This review aims to examine the regulatory mechanisms of p53, the implications of TP53 mutations in various hematological cancers, and emerging therapeutic strategies targeting p53. We conducted a comprehensive literature review to synthesize recent findings related to p53's multifaceted role in hematologic cancers, focusing on its regulatory pathways and therapeutic potential. TP53 mutations in hematological malignancies often lead to treatment resistance and poor prognosis. Current therapeutic strategies, including p53 reactivation and gene therapy, show promise in improving treatment outcomes. Understanding the intricacies of p53 regulation and the consequences of its mutations is essential for developing effective diagnostic and therapeutic strategies in hematological malignancies, ultimately enhancing patient care and survival.
Collapse
Affiliation(s)
- Seyed Esmaeil Ahmadi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Elahe Rahimian
- Department of Medical Translational Oncology, National Center for Tumor Diseases (NCT) Dresden, Dresden, Germany
| | - Samira Rahimi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Bahman Zarandi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mehran Bahraini
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maral Soleymani
- Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyed Mehrab Safdari
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ashkan Shabannezhad
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Niloofar Jaafari
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Majid Safa
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
21
|
Peng Y, Bai J, Li W, Su Z, Cheng X. Advancements in p53-Based Anti-Tumor Gene Therapy Research. Molecules 2024; 29:5315. [PMID: 39598704 PMCID: PMC11596491 DOI: 10.3390/molecules29225315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024] Open
Abstract
The p53 gene is one of the genes most closely associated with human tumors and has become a popular target for tumor drug design. Currently, p53-based gene therapy techniques have been developed, but these therapies face challenges such as immaturity, high safety hazards, limited efficacy, and low patient acceptance. However, researchers are no less enthusiastic about the treatment because of its theoretical potential to treat cancer. In this paper, the advances in p53-based gene therapy and related nucleic acid delivery technologies were reviewed and prospected in order to support further development in this field.
Collapse
Affiliation(s)
- Yuanwan Peng
- Institute of Modern Fermentation Engineering and Future Foods, School of Light Industry and Food Engineering, Guangxi University, No. 100, Daxuedong Road, Nanning 530004, China; (Y.P.); (J.B.); (W.L.)
| | - Jinping Bai
- Institute of Modern Fermentation Engineering and Future Foods, School of Light Industry and Food Engineering, Guangxi University, No. 100, Daxuedong Road, Nanning 530004, China; (Y.P.); (J.B.); (W.L.)
| | - Wang Li
- Institute of Modern Fermentation Engineering and Future Foods, School of Light Industry and Food Engineering, Guangxi University, No. 100, Daxuedong Road, Nanning 530004, China; (Y.P.); (J.B.); (W.L.)
| | - Zhengding Su
- School of Pharmaceutical Sciences and Institute of Materia Medica, Xinjiang University, Urumqi 830017, China
| | - Xiyao Cheng
- Institute of Modern Fermentation Engineering and Future Foods, School of Light Industry and Food Engineering, Guangxi University, No. 100, Daxuedong Road, Nanning 530004, China; (Y.P.); (J.B.); (W.L.)
| |
Collapse
|
22
|
Chen Y, Zheng Z, Wang L, Chen R, He M, Zhao X, Jin L, Yao J. Deciphering STAT3's negative regulation of LHPP in ESCC progression through single-cell transcriptomics analysis. Mol Med 2024; 30:192. [PMID: 39468431 PMCID: PMC11520558 DOI: 10.1186/s10020-024-00962-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 10/17/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Esophageal Squamous Cell Carcinoma (ESCC) remains a predominant health concern in the world, characterized by high prevalence and mortality rates. Advances in single-cell transcriptomics have revolutionized cancer research by enabling a precise dissection of cellular and molecular diversity within tumors. OBJECTIVE This study aims to elucidate the cellular dynamics and molecular mechanisms in ESCC, focusing on the transcriptional influence of STAT3 (Signal Transducer and Activator of Transcription 3) and its interaction with LHPP, thereby uncovering potential therapeutic targets. METHODS Single-cell RNA sequencing was employed to analyze 44,206 cells from tumor and adjacent normal tissues of ESCC patients, identifying distinct cell types and their transcriptional shifts. We conducted differential gene expression analysis to assess changes within the tumor microenvironment (TME). Validation of key regulatory interactions was performed using qPCR in a cohort of 21 ESCC patients and further substantiated through experimental assays in ESCC cell lines. RESULTS The study revealed critical alterations in cell composition and gene expression across identified cell populations, with a notable shift towards pro-tumorigenic states. A significant regulatory influence of STAT3 on LHPP was discovered, establishing a novel aspect of ESCC pathogenesis. Elevated levels of STAT3 and suppressed LHPP expression were validated in clinical samples. Functional assays confirmed that STAT3 directly represses LHPP at the promoter level, and disruption of this interaction by promoter mutations diminished STAT3's repressive effect. CONCLUSION This investigation underscores the central role of STAT3 as a regulator in ESCC, directly impacting LHPP expression and suggesting a regulatory loop crucial for tumor behavior. The insights gained from our comprehensive cellular and molecular analysis offer a deeper understanding of the dynamics within the ESCC microenvironment. These findings pave the way for targeted therapeutic interventions focusing on the STAT3-LHPP axis, providing a strategic approach to improve ESCC management and prognosis.
Collapse
Affiliation(s)
- Yurao Chen
- Department of Radiation Oncology, Huaian Hospital of Huaian City, Huaian, 223299, Jiangsu, China
- Department of Radiation Oncology, Huaian Cancer Hospital, Huaian, 223299, Jiangsu, China
| | - Zemao Zheng
- Department of Radiation Oncology, Huaian Hospital of Huaian City, Huaian, 223299, Jiangsu, China
- Department of Radiation Oncology, Huaian Cancer Hospital, Huaian, 223299, Jiangsu, China
| | - Luoshai Wang
- Department of Thoracic Surgery, Huaian Hospital of Huaian City, Huaian, 223299, Jiangsu, China
| | - Ronghuai Chen
- Department of Radiation Oncology, Huaian Hospital of Huaian City, Huaian, 223299, Jiangsu, China
- Department of Radiation Oncology, Huaian Cancer Hospital, Huaian, 223299, Jiangsu, China
| | - Ming He
- Department of Radiation Oncology, Huaian Hospital of Huaian City, Huaian, 223299, Jiangsu, China
- Department of Radiation Oncology, Huaian Cancer Hospital, Huaian, 223299, Jiangsu, China
| | - Xiang Zhao
- Department of Radiation Oncology, Huaian Hospital of Huaian City, Huaian, 223299, Jiangsu, China
- Department of Radiation Oncology, Huaian Cancer Hospital, Huaian, 223299, Jiangsu, China
| | - Liyan Jin
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, 213000, Jiangsu, China.
- Department of Oncology, The Wujin Clinical college of Xuzhou Medical University, Changzhou, 213000, Jiangsu, China.
| | - Juan Yao
- Department of Radiation Oncology, Huaian Hospital of Huaian City, Huaian, 223299, Jiangsu, China.
- Department of Radiation Oncology, Huaian Cancer Hospital, Huaian, 223299, Jiangsu, China.
| |
Collapse
|
23
|
Fan L, Liu B, Wang Y, Tang B, Xu T, Fu J, Wang C, Liu Y, Ge L, Wei H, Ren W. Intestinal Lactobacillus murinus-derived small RNAs target porcine polyamine metabolism. Proc Natl Acad Sci U S A 2024; 121:e2413241121. [PMID: 39361652 PMCID: PMC11474053 DOI: 10.1073/pnas.2413241121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 08/21/2024] [Indexed: 10/05/2024] Open
Abstract
Gut microbiota plays a vital role in host metabolism; however, the influence of gut microbes on polyamine metabolism is unknown. Here, we found germ-free models possess elevated polyamine levels in the colon. Mechanistically, intestinal Lactobacillus murinus-derived small RNAs in extracellular vesicles down-regulate host polyamine metabolism by targeting the expression of enzymes in polyamine metabolism. In addition, Lactobacillus murinus delays recovery of dextran sodium sulfate-induced colitis by reducing polyamine levels in mice. Notably, a decline in the abundance of small RNAs was observed in the colon of mice with colorectal cancer (CRC) and human CRC specimens, accompanied by elevated polyamine levels. Collectively, our study identifies a specific underlying mechanism used by intestinal microbiota to modulate host polyamine metabolism, which provides potential intervention for the treatment of polyamine-associated diseases.
Collapse
Affiliation(s)
- Lijuan Fan
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou510642, China
- National Center of Technology Innovation for Pigs, Chongqing402460, China
| | - Bingnan Liu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou510642, China
- National Center of Technology Innovation for Pigs, Chongqing402460, China
| | - Youxia Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou510642, China
- National Center of Technology Innovation for Pigs, Chongqing402460, China
| | - Bin Tang
- Department of Clinical Laboratory, Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, Jiangjin, Chongqing402260, China
| | - Tianqi Xu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou225009, China
| | - Jian Fu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou510642, China
- National Center of Technology Innovation for Pigs, Chongqing402460, China
| | - Chuanlong Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou510642, China
- National Center of Technology Innovation for Pigs, Chongqing402460, China
| | - Yuan Liu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou225009, China
| | - Liangpeng Ge
- National Center of Technology Innovation for Pigs, Chongqing Academy of Animal Sciences, Key Laboratory of Pig Industry Science, Ministry of Agriculture, Chongqing402460, China
| | - Hong Wei
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Technology, Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan430070, China
| | - Wenkai Ren
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou510642, China
- National Center of Technology Innovation for Pigs, Chongqing402460, China
| |
Collapse
|
24
|
Du Y, Chen Y, Yan Z, Yang J, Da M. Zinc finger protein 263 promotes colorectal cancer cell progression by activating STAT3 and enhancing chemoradiotherapy resistance. Sci Rep 2024; 14:21827. [PMID: 39294234 PMCID: PMC11410824 DOI: 10.1038/s41598-024-72636-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 09/09/2024] [Indexed: 09/20/2024] Open
Abstract
Zinc finger protein 263 (ZNF263) is frequently upregulated in various tumor types; however, its function and regulatory mechanism in colorectal cancer (CRC) have not yet been elucidated. In this study, the expression of ZNF263 was systematically examined using data from The Cancer Genome Atlas database and samples from patients with CRC. The results indicated that high expression of ZNF263 in CRC tissues is significantly associated with tumor grade, lymph node metastasis and disant metastasis. Additionally, overexpression of ZNF263 significantly promoted the proliferation, invasion, migration, and epithelial-mesenchymal transition of CRC cells, while also increasing signal transducer and activator of transcription 3 (STAT3) expression and mRNA stability. Conversely, knockdown of ZNF263 inhibited the malignant behavior of CRC cells and decreased STAT3 expression and mRNA stability. Further mechanism studies using chromatin immunoprecipitation (CHIP) and luciferase assays verified that ZNF263 directly binds to the STAT3 promoter. Rescue experiments demonstrated that the knockdown or overexpression of STAT3 could significantly reverse the effects of ZNF263 on CRC cells. Additionally, our study found that overexpression of ZNF263 enhanced the resistance of CRC cells to the chemoradiotherapy. In summary, this study not only elucidated the significant role of ZNF263 in CRC but also proposed novel approaches and methodologies for the diagnosis and treatment of this malignancy.
Collapse
Affiliation(s)
- Yadan Du
- The First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China
- Department of Surgical Oncology, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Yawen Chen
- The First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China
- Department of Surgical Oncology, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Zaihua Yan
- The Second Department of Gastrointestinal Surgery, The Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, China
| | - Jian Yang
- Department of Surgical Oncology, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Mingxu Da
- The First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China.
- Department of Surgical Oncology, Gansu Provincial Hospital, Lanzhou, 730000, China.
| |
Collapse
|
25
|
Huber A, Allam AH, Dijkstra C, Thiem S, Huynh J, Poh AR, Konecnik J, Jacob SP, Busuttil R, Liao Y, Chisanga D, Shi W, Alorro MG, Forrow S, Tauriello DVF, Batlle E, Boussioutas A, Williams DS, Buchert M, Ernst M, Eissmann MF. Mutant TP53 switches therapeutic vulnerability during gastric cancer progression within interleukin-6 family cytokines. Cell Rep 2024; 43:114616. [PMID: 39128004 PMCID: PMC11372443 DOI: 10.1016/j.celrep.2024.114616] [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: 02/01/2024] [Revised: 06/17/2024] [Accepted: 07/25/2024] [Indexed: 08/13/2024] Open
Abstract
Although aberrant activation of the KRAS and PI3K pathway alongside TP53 mutations account for frequent aberrations in human gastric cancers, neither the sequence nor the individual contributions of these mutations have been clarified. Here, we establish an allelic series of mice to afford conditional expression in the glandular epithelium of KrasG12D;Pik3caH1047R or Trp53R172H and/or ablation of Pten or Trp53. We find that KrasG12D;Pik3caH1047R is sufficient to induce adenomas and that lesions progress to carcinoma when also harboring Pten deletions. An additional challenge with either Trp53 loss- or gain-of-function alleles further accelerated tumor progression and triggered metastatic disease. While tumor-intrinsic STAT3 signaling in response to gp130 family cytokines remained as a gatekeeper for all stages of tumor development, metastatic progression required a mutant Trp53-induced interleukin (IL)-11 to IL-6 dependency switch. Consistent with the poorer survival of patients with high IL-6 expression, we identify IL-6/STAT3 signaling as a therapeutic vulnerability for TP53-mutant gastric cancer.
Collapse
Affiliation(s)
- Anne Huber
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia
| | - Amr H Allam
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia
| | - Christine Dijkstra
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia
| | - Stefan Thiem
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia
| | - Jennifer Huynh
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia
| | - Ashleigh R Poh
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia
| | - Joshua Konecnik
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia
| | - Saumya P Jacob
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia
| | - Rita Busuttil
- Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; Department of Gastroenterology, The Alfred Hospital, Melbourne, VIC 3004, Australia
| | - Yang Liao
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia
| | - David Chisanga
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia
| | - Wei Shi
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia
| | - Mariah G Alorro
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia
| | - Stephen Forrow
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Daniele V F Tauriello
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Eduard Batlle
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Alex Boussioutas
- Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; Department of Gastroenterology, The Alfred Hospital, Melbourne, VIC 3004, Australia
| | - David S Williams
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia; Department of Anatomical Pathology, Austin Health, Heidelberg, VIC 3084, Australia
| | - Michael Buchert
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia
| | - Matthias Ernst
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia.
| | - Moritz F Eissmann
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia.
| |
Collapse
|
26
|
Park KH, Kim HC, Won YS, Yoon WK, Choi I, Han SB, Kang JS. Vitamin D 3 Upregulated Protein 1 Deficiency Promotes Azoxymethane/Dextran Sulfate Sodium-Induced Colorectal Carcinogenesis in Mice. Cancers (Basel) 2024; 16:2934. [PMID: 39272794 PMCID: PMC11394134 DOI: 10.3390/cancers16172934] [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: 07/11/2024] [Revised: 07/24/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024] Open
Abstract
VDUP1 acts as a tumor suppressor gene in various cancers. VDUP1 is expressed at low levels in sporadic and ulcerative-colitis-associated colorectal cancer. However, the effects of VDUP1 deficiency on CAC remain unclear. In this study, we found that VDUP1 deficiency promoted CAC development in mice. Wild-type (WT) and VDUP1 KO mice were used to investigate the role of VDUP1 in the development of azoxymethane (AOM)- and dextran sulfate sodium (DSS)-induced CAC. VDUP1 levels significantly decreased in the colonic tumor and adjacent nontumoral tissues of WT mice after AOM/DSS treatment. Moreover, AOM/DSS-treated VDUP1 KO mice exhibited a worse survival rate, disease activity index, and tumor burden than WT mice. VDUP1 deficiency significantly induced cell proliferation and anti-apoptosis in tumor tissues of VDUP1 KO mice compared to WT littermates. Additionally, mRNA levels of interleukin-6 and tumor necrosis factor-alpha and active forms of signal transducer and activator of transcription 3 and nuclear factor-kappa B p65 were significantly increased in the tumor tissues of VDUP1 KO mice. Overall, this study demonstrated that the loss of VDUP1 promoted AOM/DSS-induced colon tumorigenesis in mice, highlighting the potential of VDUP1-targeting strategies for colon cancer prevention and treatment.
Collapse
Affiliation(s)
- Ki Hwan Park
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Cheongwon-gu, Cheongju-si 28116, Chungcheongbuk-do, Republic of Korea
| | - Hyoung-Chin Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Cheongwon-gu, Cheongju-si 28116, Chungcheongbuk-do, Republic of Korea
| | - Young-Suk Won
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Cheongwon-gu, Cheongju-si 28116, Chungcheongbuk-do, Republic of Korea
| | - Won Kee Yoon
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Cheongwon-gu, Cheongju-si 28116, Chungcheongbuk-do, Republic of Korea
| | - Inpyo Choi
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseoung-gu, Daejeon-si 34141, Republic of Korea
| | - Sang-Bae Han
- College of Pharmacy, Chungbuk National University, 194-21 Osongsaemgmyung-1-ro, Heungdeok-gu, Cheongju-si 28160, Chungcheongbuk-do, Republic of Korea
| | - Jong Soon Kang
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Cheongwon-gu, Cheongju-si 28116, Chungcheongbuk-do, Republic of Korea
| |
Collapse
|
27
|
Yuan L, Meng Y, Xiang J. KLF4 Induces Colorectal Cancer by Promoting EMT via STAT3 Activation. Dig Dis Sci 2024; 69:2841-2855. [PMID: 38816600 DOI: 10.1007/s10620-024-08473-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 05/01/2024] [Indexed: 06/01/2024]
Abstract
OBJECTIVE Krüppel-like factor 4 (KLF4) has been demonstrated to exert a pro-carcinogenic effect in solid tissues. However, the precise biological function and underlying mechanisms in colorectal cancer (CRC) remains elucidated. AIMS To investigate whether KLF4 participates in the proliferation and invasion of CRC. METHODS The expression of KLF4 was investigated using immunohistochemistry and immunoblotting. The clinical significance of KLF4 was evaluated. Furthermore, the effect of inhibiting or overexpressing KLF4 on tumor was examined. Immunoblotting and qPCR were used to detect Epithelial-mesenchymal transition-related proteins levels. Additionally, the molecular function of KLF4 is related to the STAT3 signaling pathway and was determined through JASPAR, GSEA analysis, and in vitro experiments. RESULTS KLF4 exhibits down-regulated expression in CRC and is part of the vessel invasion, TNM stage, and worse prognosis. In vitro studies have shown that KLF4 promotes cellular proliferation and invasion, as well as EMT processes. Xenograft tumor models confirmed the oncogenic role of KLF4 in nude mice. Furthermore, GSEA and JASPAR databases analysis reveal that the binding of KLF4 to the signal transducer and activator of transcription 3 (STAT3) promoter site induces activation of p-STAT3 signaling. Subsequent targeting of STAT3 confirmed its pivotal role in mediating the oncogenic effects exerted by KLF4. CONCLUSION The study suggests that KLF4 activates STAT3 signaling, inducing epithelial-mesenchymal transition, thereby promoting CRC progression.
Collapse
Affiliation(s)
- Lebin Yuan
- Department of Nail and Breast Surgery, Affiliated Xiangyang Central Hospital of Hubei University of Arts and Science, Xiangyang Center Hospital, Xiangyang, Hubei, China
| | - Yanqiu Meng
- Oncology Department, First Affiliated Hospital of Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Jiajia Xiang
- Molecular Centre Laboratory, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
28
|
Benedetti R, Di Crosta M, D’Orazi G, Cirone M. Post-Translational Modifications (PTMs) of mutp53 and Epigenetic Changes Induced by mutp53. BIOLOGY 2024; 13:508. [PMID: 39056701 PMCID: PMC11273943 DOI: 10.3390/biology13070508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/05/2024] [Accepted: 07/06/2024] [Indexed: 07/28/2024]
Abstract
Wild-type (wt) p53 and mutant forms (mutp53) play a key but opposite role in carcinogenesis. wtP53 acts as an oncosuppressor, preventing oncogenic transformation, while mutp53, which loses this property, may instead favor this process. This suggests that a better understanding of the mechanisms activating wtp53 while inhibiting mutp53 may help to design more effective anti-cancer treatments. In this review, we examine possible PTMs with which both wt- and mutp53 can be decorated and discuss how their manipulation could represent a possible strategy to control the stability and function of these proteins, focusing in particular on mutp53. The impact of ubiquitination, phosphorylation, acetylation, and methylation of p53, in the context of several solid and hematologic cancers, will be discussed. Finally, we will describe some of the recent studies reporting that wt- and mutp53 may influence the expression and activity of enzymes responsible for epigenetic changes such as acetylation, methylation, and microRNA regulation and the possible consequences of such changes.
Collapse
Affiliation(s)
- Rossella Benedetti
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (R.B.); (M.D.C.)
| | - Michele Di Crosta
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (R.B.); (M.D.C.)
| | - Gabriella D’Orazi
- Department of Neurosciences, Imaging and Clinical Sciences, University “G. D’Annunzio”, 66013 Chieti, Italy
| | - Mara Cirone
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (R.B.); (M.D.C.)
| |
Collapse
|
29
|
Wang H, Li Y, Qiu M, Wang J. Thrombocytopenia and hyperprogression after radiotherapy and camrelizumab treatment in an esophageal cancer patient with increased JAK2 gene copies: a case report. Front Oncol 2024; 14:1283428. [PMID: 38974233 PMCID: PMC11224440 DOI: 10.3389/fonc.2024.1283428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 05/30/2024] [Indexed: 07/09/2024] Open
Abstract
Radiotherapy (RT) and immune checkpoint inhibitor (ICI) are important treatments for esophageal cancer. Some studies have confirmed the safety and effectiveness of using RT in combination with ICI, while serious side effects have been exhibited by some patients. We report a patient with metastatic esophageal cancer who received RT combined with ICI. The patient experienced severe thrombocytopenia, and treatment with thrombopoietin and corticosteroids were ineffective. Finally, the patient developed abscopal hyperprogression outside the radiation field. Interestingly, next-generation sequencing revealed increased JAK2 gene copies in the surgical slices. The JAK2/STAT3 pathway is involved in the regulation of megakaryocyte development. Recurrent thrombocytopenia may activate the JAK2/STAT3 pathway, leading to megakaryocyte differentiation and platelet biogenesis. However, persistent activation of the JAK2/STAT3 pathway has been associated with immune ICI resistance and tumor progression. This case indicates that thrombocytopenia and increased JAK2 gene copies may be risk factors for poor prognosis after ICI and RT treatment.
Collapse
Affiliation(s)
- Hang Wang
- Department of Endocrinology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
- College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, China
- Department of Cardiology, Hospital of Traditional Chinese Medicine in Qijiang District, Chongqing, China
| | - Yun Li
- Department of Respiratory and Critical Care Medicine, The First People’s Hospital of Neijiang, Neijiang, Sichuan, China
| | - Min Qiu
- Department of Oncology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Jianmin Wang
- Department of Oncology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| |
Collapse
|
30
|
O’Reilly M, Krstic A, Iglesias-Martinez LF, Ryan ÉJ, Moran B, Winter D, Sheahan K, McDermott R, Kolch W. Genomic and Transcriptomic Analysis of a Patient with Early-Onset Colorectal Cancer and Therapy-Induced Focal Nodular Hyperplasia: A Case Report. J Pers Med 2024; 14:639. [PMID: 38929861 PMCID: PMC11204579 DOI: 10.3390/jpm14060639] [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: 05/01/2024] [Revised: 06/04/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
Early-onset colorectal cancer (EOCRC), defined as colorectal cancer in individuals under 50 years of age, has shown an alarming increase in incidence worldwide. We report a case of a twenty-four-year-old female with a strong family history of colorectal cancer (CRC) but without an identified underlying genetic predisposition syndrome. Two years after primary surgery and adjuvant chemotherapy, the patient developed new liver lesions. Extensive diagnostic imaging was conducted to investigate suspected liver metastases, ultimately leading to a diagnosis of focal nodular hyperplasia. The young age of the patient has prompted comprehensive genomic and transcriptomic profiling in order to identify potential oncogenic drivers and inform further clinical management of the patient. Besides a number of oncogenic mutations identified in the patient's tumour sample, including KRAS G12D, TP53 R248W and TTN L28470V, we have also identified a homozygous deletion of 24.5 MB on chromosome 8. A multivariate Cox regression analysis of this patient's mutation profile conferred a favourable prognosis when compared with the TCGA COADREAD database. Notably, the identified deletion on chromosome 8 includes the WRN gene, which could contribute to the patient's overall positive response to chemotherapy. The complex clinical presentation, including the need for emergency surgery, early age at diagnosis, strong family history, and unexpected findings on surveillance imaging, necessitated a multidisciplinary approach involving medical, radiation, and surgical oncologists, along with psychological support and reproductive medicine specialists. Molecular profiling of the tumour strongly indicates that patients with complex mutational profile and rare genomic rearrangements require a prolonged surveillance and personalised informed interventions.
Collapse
Affiliation(s)
- Mary O’Reilly
- Systems Biology Ireland, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland; (A.K.); (L.F.I.-M.); (W.K.)
- St. Vincent’s University Hospital, University College Dublin, D04 V1W8 Dublin, Ireland; (É.J.R.); (B.M.); (D.W.); (K.S.); (R.M.)
| | - Aleksandar Krstic
- Systems Biology Ireland, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland; (A.K.); (L.F.I.-M.); (W.K.)
| | - Luis F. Iglesias-Martinez
- Systems Biology Ireland, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland; (A.K.); (L.F.I.-M.); (W.K.)
| | - Éanna J. Ryan
- St. Vincent’s University Hospital, University College Dublin, D04 V1W8 Dublin, Ireland; (É.J.R.); (B.M.); (D.W.); (K.S.); (R.M.)
| | - Bruce Moran
- St. Vincent’s University Hospital, University College Dublin, D04 V1W8 Dublin, Ireland; (É.J.R.); (B.M.); (D.W.); (K.S.); (R.M.)
| | - Des Winter
- St. Vincent’s University Hospital, University College Dublin, D04 V1W8 Dublin, Ireland; (É.J.R.); (B.M.); (D.W.); (K.S.); (R.M.)
| | - Kieran Sheahan
- St. Vincent’s University Hospital, University College Dublin, D04 V1W8 Dublin, Ireland; (É.J.R.); (B.M.); (D.W.); (K.S.); (R.M.)
| | - Ray McDermott
- St. Vincent’s University Hospital, University College Dublin, D04 V1W8 Dublin, Ireland; (É.J.R.); (B.M.); (D.W.); (K.S.); (R.M.)
| | - Walter Kolch
- Systems Biology Ireland, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland; (A.K.); (L.F.I.-M.); (W.K.)
- Conway Institute, University College Dublin, D04 V1W8 Dublin, Ireland
| |
Collapse
|
31
|
Ma Z, Zhou F, Jin H, Wu X. Crosstalk between CXCL12/CXCR4/ACKR3 and the STAT3 Pathway. Cells 2024; 13:1027. [PMID: 38920657 PMCID: PMC11201928 DOI: 10.3390/cells13121027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/09/2024] [Accepted: 06/10/2024] [Indexed: 06/27/2024] Open
Abstract
The reciprocal modulation between the CXCL12/CXCR4/ACKR3 axis and the STAT3 signaling pathway plays a crucial role in the progression of various diseases and neoplasms. Activation of the CXCL12/CXCR4/ACKR3 axis triggers the STAT3 pathway through multiple mechanisms, while the STAT3 pathway also regulates the expression of CXCL12. This review offers a thorough and systematic analysis of the reciprocal regulatory mechanisms between the CXCL12/CXCR4/ACKR3 signaling axis and the STAT3 signaling pathway in the context of diseases, particularly tumors. It explores the potential clinical applications in tumor treatment, highlighting possible therapeutic targets and novel strategies for targeted tumor therapy.
Collapse
Affiliation(s)
| | | | | | - Xiaoming Wu
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, Chenggong Campus, 727 South Jingming Road, Kunming 650500, China; (Z.M.); (F.Z.); (H.J.)
| |
Collapse
|
32
|
Liu Y, Su Z, Tavana O, Gu W. Understanding the complexity of p53 in a new era of tumor suppression. Cancer Cell 2024; 42:946-967. [PMID: 38729160 PMCID: PMC11190820 DOI: 10.1016/j.ccell.2024.04.009] [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: 02/01/2024] [Revised: 03/15/2024] [Accepted: 04/16/2024] [Indexed: 05/12/2024]
Abstract
p53 was discovered 45 years ago as an SV40 large T antigen binding protein, coded by the most frequently mutated TP53 gene in human cancers. As a transcription factor, p53 is tightly regulated by a rich network of post-translational modifications to execute its diverse functions in tumor suppression. Although early studies established p53-mediated cell-cycle arrest, apoptosis, and senescence as the classic barriers in cancer development, a growing number of new functions of p53 have been discovered and the scope of p53-mediated anti-tumor activity is largely expanded. Here, we review the complexity of different layers of p53 regulation, and the recent advance of the p53 pathway in metabolism, ferroptosis, immunity, and others that contribute to tumor suppression. We also discuss the challenge regarding how to activate p53 function specifically effective in inhibiting tumor growth without harming normal homeostasis for cancer therapy.
Collapse
Affiliation(s)
- Yanqing Liu
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Zhenyi Su
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Omid Tavana
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Wei Gu
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA; Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA.
| |
Collapse
|
33
|
Xiong H, Zhang W, Xie M, Chen R, Chen H, Lin Q. Long non-coding RNA JPX promotes endometrial carcinoma progression via janus kinase 2/signal transducer and activator of transcription 3. Front Oncol 2024; 14:1340050. [PMID: 38784043 PMCID: PMC11112342 DOI: 10.3389/fonc.2024.1340050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 04/03/2024] [Indexed: 05/25/2024] Open
Abstract
Introduction Although LncRNA JPX has been linked to a number of malignancies, it is yet unknown how it relates to endometrial carcinoma (EC). Investigating the expression, functional activities, and underlying molecular processes of lncRNA JPX in EC was the goal of this work. Methods RT-qPCR was used to examine the differences in lncRNA/microRNA (miRNA, miR)/mRNA expression between normal cervical and EC tissues or cells. Cell Counting Kit-8, flow cytometry, and transwell were used to evaluate the association between lncRNA JPX/miR-140-3p/phosphoinositide-3-kinase catalytic subunit α (PIK3CA) in Ishikawa and JEC cell lines. The impact of JPX on the downstream janus kinase (JAK)2/signal transducer and activator of transcription (STAT)3 signaling pathway was investigated using Western blot analysis. Results When comparing EC tissues to nearby normal tissues, JPX expression is markedly increased in EC tissues, with greater expression in advanced-stage EC. Furthermore, compared to normal epithelial cells, EC cell lines have higher levels of JPX expression. In Ishikawa and JEC endometrial cancer cell lines, we used siRNA-mediated suppression of JPX to find lower cell viability, increased apoptosis, cell cycle arrest, and reduced migration and invasion. We next verified that miR-140-3p binds to downstream target cells to impede the transcription and translation of PIK3CA, which in turn prevents the growth of Ishikawa and JEC cells. JPX functions as a ceRNA to adsorb miR-140-3p. This procedure required controlling JAK2/STAT3, a downstream signal. Conclusion JPX enhances the development of Ishikawa and JEC cells and activates downstream JAK2/STAT3 signal transduction via the miR-140-3p/PIK3CA axis, offering a possible therapeutic target for the treatment of EC.
Collapse
Affiliation(s)
- Hanzhen Xiong
- Department of Pathology, Guangdong Provincial Key Laboratory of Major Obstetric Disease, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wei Zhang
- Department of Pathology, Guangdong Provincial Key Laboratory of Major Obstetric Disease, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Mingyu Xie
- Department of Pathology, Guangdong Provincial Key Laboratory of Major Obstetric Disease, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Pathology, Central People’s Hospital of Zhanjiang, Zhanjiang, Guangdong, China
| | - Ruichao Chen
- Department of Pathology, Guangdong Provincial Key Laboratory of Major Obstetric Disease, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Hui Chen
- Department of Pathology, Guangdong Provincial Key Laboratory of Major Obstetric Disease, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Qiongyan Lin
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Gynecologic Oncology Research Office, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
34
|
Huang X, Cao Z, Qian J, Ding T, Wu Y, Zhang H, Zhong S, Wang X, Ren X, Zhang W, Xu Y, Yao G, Wang X, Yang X, Wen L, Zhang Y. Nanoreceptors promote mutant p53 protein degradation by mimicking selective autophagy receptors. NATURE NANOTECHNOLOGY 2024; 19:545-553. [PMID: 38216684 DOI: 10.1038/s41565-023-01562-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 11/01/2023] [Indexed: 01/14/2024]
Abstract
In some cancers mutant p53 promotes the occurrence, development, metastasis and drug resistance of tumours, with targeted protein degradation seen as an effective therapeutic strategy. However, a lack of specific autophagy receptors limits this. Here, we propose the synthesis of biomimetic nanoreceptors (NRs) that mimic selective autophagy receptors. The NRs have both a component for targeting the desired protein, mutant-p53-binding peptide, and a component for enhancing degradation, cationic lipid. The peptide can bind to mutant p53 while the cationic lipid simultaneously targets autophagosomes and elevates the levels of autophagosome formation, increasing mutant p53 degradation. The NRs are demonstrated in vitro and in a patient-derived xenograft ovarian cancer model in vivo. The work highlights a possible direction for treating diseases by protein degradation.
Collapse
Affiliation(s)
- Xiaowan Huang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, People's Republic of China
- School of Medicine, South China University of Technology, Guangzhou, People's Republic of China
| | - Ziyang Cao
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, People's Republic of China
| | - Jieying Qian
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, People's Republic of China
| | - Tao Ding
- School of Medicine, South China University of Technology, Guangzhou, People's Republic of China
| | - Yanxia Wu
- Molecular Cancer Research Center, School of Medicine, Sun Yat-Sen University, Shenzhen, People's Republic of China
| | - Hao Zhang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, People's Republic of China
| | - Suqin Zhong
- School of Medicine, South China University of Technology, Guangzhou, People's Republic of China
| | - Xiaoli Wang
- School of Medicine, South China University of Technology, Guangzhou, People's Republic of China
| | - Xiaoguang Ren
- School of Medicine, South China University of Technology, Guangzhou, People's Republic of China
| | - Wang Zhang
- School of Medicine, South China University of Technology, Guangzhou, People's Republic of China
| | - Youcui Xu
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
| | - Guangyu Yao
- Breast Center, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Xingwu Wang
- Molecular Cancer Research Center, School of Medicine, Sun Yat-Sen University, Shenzhen, People's Republic of China
| | - Xianzhu Yang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, People's Republic of China.
- National Engineering Research Centre for Tissue Restoration and Reconstruction and Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, People's Republic of China.
| | - Longping Wen
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China.
| | - Yunjiao Zhang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, People's Republic of China.
- School of Medicine, South China University of Technology, Guangzhou, People's Republic of China.
- National Engineering Research Centre for Tissue Restoration and Reconstruction and Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, People's Republic of China.
| |
Collapse
|
35
|
Gagliano T, Kerschbamer E, Baccarani U, Minisini M, Di Giorgio E, Dalla E, Weichenberger CX, Cherchi V, Terrosu G, Brancolini C. Changes in chromatin accessibility and transcriptional landscape induced by HDAC inhibitors in TP53 mutated patient-derived colon cancer organoids. Biomed Pharmacother 2024; 173:116374. [PMID: 38447451 DOI: 10.1016/j.biopha.2024.116374] [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: 01/08/2024] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 03/08/2024] Open
Abstract
Here we present the generation and characterization of patient-derived organoids (PDOs) from colorectal cancer patients. PDOs derived from two patients with TP53 mutations were tested with two different HDAC inhibitors (SAHA and NKL54). Cell death induction, transcriptome, and chromatin accessibility changes were analyzed. HDACIs promote the upregulation of low expressed genes and the downregulation of highly expressed genes. A similar differential effect is observed at the level of chromatin accessibility. Only SAHA is a potent inducer of cell death, which is characterized by the upregulation of BH3-only genes BIK and BMF. Up-regulation of BIK is associated with increased accessibility in an intronic region that has enhancer properties. SAHA, but not NKL54, also causes downregulation of BCL2L1 and decreases chromatin accessibility in three distinct regions of the BCL2L1 locus. Both inhibitors upregulate the expression of innate immunity genes and members of the MHC family. In summary, our exploratory study indicates a mechanism of action for SAHA and demonstrate the low efficacy of NKL54 as a single agent for apoptosis induction, using two PDOs. These observations need to be validated in a larger cohort of PDOs.
Collapse
Affiliation(s)
- Teresa Gagliano
- Department of Medicine, Università degli Studi di Udine, Institute for Biomedicine, P.le Kolbe 4, Udine 33100, Italy
| | - Emanuela Kerschbamer
- Department of Medicine, Università degli Studi di Udine, Institute for Biomedicine, P.le Kolbe 4, Udine 33100, Italy
| | - Umberto Baccarani
- Department of Medicine, Università degli Studi di Udine, Institute for Biomedicine, P.le Kolbe 4, Udine 33100, Italy
| | - Martina Minisini
- Department of Medicine, Università degli Studi di Udine, Institute for Biomedicine, P.le Kolbe 4, Udine 33100, Italy
| | - Eros Di Giorgio
- Department of Medicine, Università degli Studi di Udine, Institute for Biomedicine, P.le Kolbe 4, Udine 33100, Italy
| | - Emiliano Dalla
- Department of Medicine, Università degli Studi di Udine, Institute for Biomedicine, P.le Kolbe 4, Udine 33100, Italy
| | | | - Vittorio Cherchi
- General Surgery Clinic and Liver Transplant Center, University-Hospital of Udine, Udine, Italy
| | - Giovanni Terrosu
- Department of Medicine, Università degli Studi di Udine, Institute for Biomedicine, P.le Kolbe 4, Udine 33100, Italy
| | - Claudio Brancolini
- Department of Medicine, Università degli Studi di Udine, Institute for Biomedicine, P.le Kolbe 4, Udine 33100, Italy.
| |
Collapse
|
36
|
Li J, Zong Y, Tuo Z, Liu J, Liu J. The role of RASA2 in predicting radioresistance in lung cancer through regulation of p53. Transl Lung Cancer Res 2024; 13:587-602. [PMID: 38601440 PMCID: PMC11002505 DOI: 10.21037/tlcr-24-160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 03/20/2024] [Indexed: 04/12/2024]
Abstract
Background One of the most common causes of lung cancer relapse after clinical treatment is radioresistance. However, the mechanism underlying radioresistance remains unclear. In this study, we investigated the role of Ras p21 protein activator (RASA2) in non-small cell lung cancer (NSCLC). Methods The messenger RNA (mRNA) of RASA2 was tested via reverse-transcription quantitative polymerase chain reaction (RT-qPCR) of cancer tissues from patients with NSCLC. Computed tomography (CT) and bioluminescent imaging (BLI) were used to monitor the tumor growth of patients and orthotopic mice, respectively. Protein-protein interaction was quantified via immunoprecipitation and glutathione S transferase (GST) pulldown assay. Western blotting was used to evaluate the phosphorylation and ubiquitination level of p53. Results The results indicated a negative correlation between the mRNA expression levels of RASA2 in tumor tissues with patients' response to radiotherapy. Patients with a high expression of RASA2 had a lower objective response rate (ORR) after 1 month of radiotherapy than patients with low expression of RASA2 after 1 month of radiotherapy. In terms of mechanism, we proved that RASA2 can directly bind to p53 to promote the phosphorylation of p53, which inhibits its transcriptional activity and further promotes its degradation through the ubiquitin/proteasome pathway. In this process, the apoptosis of tumor cells is inhibited due to impaired p53 surveillance, which leads to radioresistance. Conclusions Our results demonstrate that RASA2 negatively regulates p53 in cancer cells and therefore promotes radioresistance, providing a new predictive biomarker and a potential therapeutic target for radioresistance.
Collapse
Affiliation(s)
- Jie Li
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Zong
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhan Tuo
- Department of Radiology, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Junwei Liu
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jun Liu
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
37
|
Guo X, Li Y, Chen X, Sun B, Guo X. Urocortin-1 promotes colorectal cancer cell migration and proliferation and inhibits apoptosis via inhibition of the p53 signaling pathway. J Cancer Res Clin Oncol 2024; 150:163. [PMID: 38546882 PMCID: PMC10978644 DOI: 10.1007/s00432-024-05693-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 03/08/2024] [Indexed: 04/01/2024]
Abstract
PURPOSE To investigate the effect of urocortin-1 (UCN-1) on growth, migration, and apoptosis in colorectal cancer (CRC) in vivo and vitro and the mechanism by which UCN-1 modulates CRC cells in vitro. METHODS The correlation between UCN-1 and CRC was evaluated using The Cancer Genome Atlas (TCGA) database and a tissue microarray. The expression of UCN-1 in CRC cells was assessed using quantitative real-time polymerase chain reaction (RT-qPCR) and western blotting. In vitro, the influence of UCN-1 on the proliferation, apoptosis, and migration of HT-29, HCT-116, and RKO cells was explored using the celigo cell counting assay or cell counting kit-8 (CCK8), flow cytometry, and wound healing or Transwell assays, respectively. In vivo, the effect of UCN-1 on CRC growth and progression was evaluated in nude mice. The downstream pathway underlying UCN-1-mediated regulation of CRC was determined using the phospho-kinase profiler array in RKO cells. Lentiviruses were used to knockdown or upregulate UCN-1 expression in cells. RESULTS Both the TCGA and tissue microarray results showed that UCN-1 was strongly expressed in the tissues of patients with CRC. Furthermore, the tissue microarray results showed that the expression of UCN-1 was higher in male than in female patients, and high expression of UCN-1 was associated with higher risk of lymphatic metastasis and later pathological stage. UCN-1 knockdown caused a reduction in CRC cell proliferation, migration, and colony formation, as well as an increase in apoptosis. In xenograft experiments, tumors generated from RKO cells with UCN-1 knockdown exhibited reduced volumes and weights. A reduction in the expression of Ki-67 in xenograft tumors indicated that UCN-1 knockdown curbed tumor growth. The human phospho-kinase array showed that the p53 signaling pathway participated in UCN-1-mediated CRC development. The suppression in migration and proliferation caused by UCN-1 knockdown was reversed by inhibitors of p53 signal pathway, while the increase in cell apoptosis was suppressed. On the other hand, overexpression of UCN-1 promoted proliferation and migration and inhibited apoptosis in CRC cells. Overexpression of p53 reversed the effect of UCN-1 overexpression on CRC development. CONCLUSION UCN-1 promotes migration and proliferation and inhibits apoptosis via inhibition of the p53 signaling pathway.
Collapse
Affiliation(s)
- Xiaolan Guo
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ya Li
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiangyu Chen
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Binghua Sun
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaolan Guo
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
38
|
Di Crosta M, Arena A, Benedetti R, Gilardini Montani MS, Cirone M. 5-AZA Upregulates SOCS3 and PTPN6/SHP1, Inhibiting STAT3 and Potentiating the Effects of AG490 against Primary Effusion Lymphoma Cells. Curr Issues Mol Biol 2024; 46:2468-2479. [PMID: 38534772 DOI: 10.3390/cimb46030156] [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: 01/31/2024] [Revised: 03/07/2024] [Accepted: 03/12/2024] [Indexed: 03/28/2024] Open
Abstract
Epigenetic modifications, including aberrant DNA methylation occurring at the promoters of oncogenes and oncosuppressor genes and histone modifications, can contribute to carcinogenesis. Aberrant methylation mediated by histone methylatransferases, alongside histones, can affect methylation of proteins involved in the regulation of pro-survival pathways such as JAK/STAT and contribute to their activation. In this study, we used DNA or histone demethylating agents, 5-Azacytidine (5-AZA) or DS-3201 (valemetostat), respectively, to treat primary effusion lymphoma (PEL) cells, alone or in combination with AG490, a Signal transducer and activator of transcription 3 (STAT3) inhibitor. Cell viability was investigated by trypan blue assay and FACS analysis. The molecular changes induced by 5-AZA and/or AG490 treatments were investigated by Western blot analysis, while cytokine release by PEL cells treated by these drugs was evaluated by Luminex. Statistical analyses were performed with Graphpad Prism® software (version 9) and analyzed by Student's t test or a nonparametric one-way ANOVA test. The results obtained in this study suggest that 5-AZA upregulated molecules that inhibit STAT3 tyrosine phosphorylation, namely Suppressor of Cytokine Signaling 3 (SOCS3) and tyrosine-protein phosphatase non-receptor type (PTPN) 6/Src homology region 2 domain-containing phosphatase-1 (SHP-1), reducing STAT3 activation and downregulating several STAT3 pro-survival targets in PEL cells. As this lymphoma is highly dependent on the constitutive activation of STAT3, 5-AZA impaired PEL cell survival, and when used in combination with AG490 JAK2/STAT3 inhibitor, it potentiated its cytotoxic effect. Differently from 5-AZA, the inhibition of the EZH1/2 histone methyltransferase by DS-3201, reported to contribute to STAT3 activation in other cancers, slightly affected STAT3 phosphorylation or survival in PEL cells, either alone or in combination with AG490. This study suggests that 5-AZA, by upregulating the expression level of SOCS3 and PTPN6/SHP1, reduced STAT3 activation and improved the outcome of treatment targeting this transcription factor in PEL cells.
Collapse
Affiliation(s)
- Michele Di Crosta
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Andrea Arena
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Rossella Benedetti
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | | | - Mara Cirone
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| |
Collapse
|
39
|
Gould SI, Wuest AN, Dong K, Johnson GA, Hsu A, Narendra VK, Atwa O, Levine SS, Liu DR, Sánchez Rivera FJ. High-throughput evaluation of genetic variants with prime editing sensor libraries. Nat Biotechnol 2024:10.1038/s41587-024-02172-9. [PMID: 38472508 DOI: 10.1038/s41587-024-02172-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 02/09/2024] [Indexed: 03/14/2024]
Abstract
Tumor genomes often harbor a complex spectrum of single nucleotide alterations and chromosomal rearrangements that can perturb protein function. Prime editing has been applied to install and evaluate genetic variants, but previous approaches have been limited by the variable efficiency of prime editing guide RNAs. Here we present a high-throughput prime editing sensor strategy that couples prime editing guide RNAs with synthetic versions of their cognate target sites to quantitatively assess the functional impact of endogenous genetic variants. We screen over 1,000 endogenous cancer-associated variants of TP53-the most frequently mutated gene in cancer-to identify alleles that impact p53 function in mechanistically diverse ways. We find that certain endogenous TP53 variants, particularly those in the p53 oligomerization domain, display opposite phenotypes in exogenous overexpression systems. Our results emphasize the physiological importance of gene dosage in shaping native protein stoichiometry and protein-protein interactions, and establish a framework for studying genetic variants in their endogenous sequence context at scale.
Collapse
Affiliation(s)
- Samuel I Gould
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Alexandra N Wuest
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kexin Dong
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- University of Chinese Academy of Sciences, Beijing, China
| | - Grace A Johnson
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Alvin Hsu
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Varun K Narendra
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ondine Atwa
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Stuart S Levine
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - David R Liu
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Francisco J Sánchez Rivera
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
40
|
Ibusuki R, Iwama E, Shimauchi A, Tsutsumi H, Yoneshima Y, Tanaka K, Okamoto I. TP53 gain-of-function mutations promote osimertinib resistance via TNF-α-NF-κB signaling in EGFR-mutated lung cancer. NPJ Precis Oncol 2024; 8:60. [PMID: 38431700 PMCID: PMC10908812 DOI: 10.1038/s41698-024-00557-2] [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: 09/11/2023] [Accepted: 02/20/2024] [Indexed: 03/05/2024] Open
Abstract
EGFR tyrosine kinase inhibitors (TKIs) are effective against EGFR-mutated lung cancer, but tumors eventually develop resistance to these drugs. Although TP53 gain-of-function (GOF) mutations promote carcinogenesis, their effect on EGFR-TKI efficacy has remained unclear. We here established EGFR-mutated lung cancer cell lines that express wild-type (WT) or various mutant p53 proteins with CRISPR-Cas9 technology and found that TP53-GOF mutations promote early development of resistance to the EGFR-TKI osimertinib associated with sustained activation of ERK and expression of c-Myc. Gene expression analysis revealed that osimertinib activates TNF-α-NF-κB signaling specifically in TP53-GOF mutant cells. In such cells, osimertinib promoted interaction of p53 with the NF-κB subunit p65, translocation of the resulting complex to the nucleus and its binding to the TNF promoter, and TNF-α production. Concurrent treatment of TP53-GOF mutant cells with the TNF-α inhibitor infliximab suppressed acquisition of osimertinib resistance as well as restored osimertinib sensitivity in resistant cells in association with attenuation of ERK activation and c-Myc expression. Our findings indicate that induction of TNF-α expression by osimertinib in TP53-GOF mutant cells contributes to the early development of osimertinib resistance, and that TNF-α inhibition may therefore be an effective strategy to overcome such resistance in EGFR-mutant lung cancer with TP53-GOF mutations.
Collapse
Affiliation(s)
- Ritsu Ibusuki
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Eiji Iwama
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Atsushi Shimauchi
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hirono Tsutsumi
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yasuto Yoneshima
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kentaro Tanaka
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Isamu Okamoto
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
41
|
Ely ZA, Mathey-Andrews N, Naranjo S, Gould SI, Mercer KL, Newby GA, Cabana CM, Rideout WM, Jaramillo GC, Khirallah JM, Holland K, Randolph PB, Freed-Pastor WA, Davis JR, Kulstad Z, Westcott PMK, Lin L, Anzalone AV, Horton BL, Pattada NB, Shanahan SL, Ye Z, Spranger S, Xu Q, Sánchez-Rivera FJ, Liu DR, Jacks T. A prime editor mouse to model a broad spectrum of somatic mutations in vivo. Nat Biotechnol 2024; 42:424-436. [PMID: 37169967 PMCID: PMC11120832 DOI: 10.1038/s41587-023-01783-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 04/05/2023] [Indexed: 05/13/2023]
Abstract
Genetically engineered mouse models only capture a small fraction of the genetic lesions that drive human cancer. Current CRISPR-Cas9 models can expand this fraction but are limited by their reliance on error-prone DNA repair. Here we develop a system for in vivo prime editing by encoding a Cre-inducible prime editor in the mouse germline. This model allows rapid, precise engineering of a wide range of mutations in cell lines and organoids derived from primary tissues, including a clinically relevant Kras mutation associated with drug resistance and Trp53 hotspot mutations commonly observed in pancreatic cancer. With this system, we demonstrate somatic prime editing in vivo using lipid nanoparticles, and we model lung and pancreatic cancer through viral delivery of prime editing guide RNAs or orthotopic transplantation of prime-edited organoids. We believe that this approach will accelerate functional studies of cancer-associated mutations and complex genetic combinations that are challenging to construct with traditional models.
Collapse
Affiliation(s)
- Zackery A Ely
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Nicolas Mathey-Andrews
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Santiago Naranjo
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Samuel I Gould
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kim L Mercer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Gregory A Newby
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Christina M Cabana
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - William M Rideout
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Grissel Cervantes Jaramillo
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard-MIT Division of Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Katie Holland
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Angelo State University, San Angelo, TX, USA
| | - Peyton B Randolph
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - William A Freed-Pastor
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jessie R Davis
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Zachary Kulstad
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Peter M K Westcott
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Cold Spring Harbor Laboratory, Huntington, NY, USA
| | - Lin Lin
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Andrew V Anzalone
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Brendan L Horton
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Nimisha B Pattada
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sean-Luc Shanahan
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Zhongfeng Ye
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - Stefani Spranger
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Qiaobing Xu
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - Francisco J Sánchez-Rivera
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - David R Liu
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Tyler Jacks
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
42
|
Song B, Yang P, Zhang S. Cell fate regulation governed by p53: Friends or reversible foes in cancer therapy. Cancer Commun (Lond) 2024; 44:297-360. [PMID: 38311377 PMCID: PMC10958678 DOI: 10.1002/cac2.12520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 01/03/2024] [Accepted: 01/11/2024] [Indexed: 02/10/2024] Open
Abstract
Cancer is a leading cause of death worldwide. Targeted therapies aimed at key oncogenic driver mutations in combination with chemotherapy and radiotherapy as well as immunotherapy have benefited cancer patients considerably. Tumor protein p53 (TP53), a crucial tumor suppressor gene encoding p53, regulates numerous downstream genes and cellular phenotypes in response to various stressors. The affected genes are involved in diverse processes, including cell cycle arrest, DNA repair, cellular senescence, metabolic homeostasis, apoptosis, and autophagy. However, accumulating recent studies have continued to reveal novel and unexpected functions of p53 in governing the fate of tumors, for example, functions in ferroptosis, immunity, the tumor microenvironment and microbiome metabolism. Among the possibilities, the evolutionary plasticity of p53 is the most controversial, partially due to the dizzying array of biological functions that have been attributed to different regulatory mechanisms of p53 signaling. Nearly 40 years after its discovery, this key tumor suppressor remains somewhat enigmatic. The intricate and diverse functions of p53 in regulating cell fate during cancer treatment are only the tip of the iceberg with respect to its equally complicated structural biology, which has been painstakingly revealed. Additionally, TP53 mutation is one of the most significant genetic alterations in cancer, contributing to rapid cancer cell growth and tumor progression. Here, we summarized recent advances that implicate altered p53 in modulating the response to various cancer therapies, including chemotherapy, radiotherapy, and immunotherapy. Furthermore, we also discussed potential strategies for targeting p53 as a therapeutic option for cancer.
Collapse
Affiliation(s)
- Bin Song
- Laboratory of Radiation MedicineWest China Second University HospitalSichuan UniversityChengduSichuanP. R. China
| | - Ping Yang
- Laboratory of Radiation MedicineWest China Second University HospitalSichuan UniversityChengduSichuanP. R. China
| | - Shuyu Zhang
- Laboratory of Radiation MedicineWest China Second University HospitalSichuan UniversityChengduSichuanP. R. China
- The Second Affiliated Hospital of Chengdu Medical CollegeChina National Nuclear Corporation 416 HospitalChengduSichuanP. R. China
- Laboratory of Radiation MedicineNHC Key Laboratory of Nuclear Technology Medical TransformationWest China School of Basic Medical Sciences & Forensic MedicineSichuan UniversityChengduSichuanP. R. China
| |
Collapse
|
43
|
Isermann T, Schneider KL, Wegwitz F, De Oliveira T, Conradi LC, Volk V, Feuerhake F, Papke B, Stintzing S, Mundt B, Kühnel F, Moll UM, Schulz-Heddergott R. Enhancement of colorectal cancer therapy through interruption of the HSF1-HSP90 axis by p53 activation or cell cycle inhibition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.22.581507. [PMID: 38464125 PMCID: PMC10925225 DOI: 10.1101/2024.02.22.581507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
The stress-associated molecular chaperone system is an actionable target in cancer therapies. It is ubiquitously upregulated in cancer tissues and enables tumorigenicity by stabilizing hundreds of oncoproteins and disturbing the stoichiometry of protein complexes. Most inhibitors target the key component heat-shock protein 90 (HSP90). However, although classical HSP90 inhibitors are highly tumor-selective, they fail in phase 3 clinical oncology trials. These failures are at least partly due to an interference with a negative feedback loop by HSP90 inhibition, known as heat-shock response (HSR): in response to HSP90 inhibition there is compensatory synthesis of stress-inducible chaperones, mediated by the transcription factor heat-shock factor 1 (HSF1). We recently identified that wildtype p53 (p53) actively reduces the HSR by repressing HSF1 via a p21-CDK4/6-MAPK-HSF1 axis. Here we test the hypothesis that in HSP90-based therapies simultaneous p53 activation or direct cell cycle inhibition interrupts the deleterious HSF1-HSR axis and improves the efficiency of HSP90 inhibitors. Indeed, we find that the clinically relevant p53 activator Idasanutlin suppresses the HSF1-HSR activity in HSP90 inhibitor-based therapies. This combination synergistically reduces cell viability and accelerates cell death in p53-proficient colorectal cancer (CRC) cells, murine tumor-derived organoids and patient-derived organoids (PDOs). Mechanistically, upon combination therapy human CRC cells strongly upregulate p53-associated pathways, apoptosis, and inflammatory immune pathways. Likewise, in the chemical AOM/DSS CRC model in mice, dual HSF1-HSP90 inhibition strongly represses tumor growth and remodels immune cell composition, yet displays only minor toxicities in mice and normal mucosa-derived organoids. Importantly, inhibition of the cyclin dependent kinases 4 and 6 (CDK4/6) under HSP90 inhibition phenocopies synergistic repression of the HSR in p53-proficient CRC cells. Even more important, in p53-deficient (mutp53-harboring) CRC cells, an HSP90 inhibition in combination with CDK4/6 inhibitors similarly suppresses the HSF1-HSR system and reduces cancer growth. Likewise, p53-mutated PDOs strongly respond to dual HSF1-HSP90 pathway inhibition and thus, providing a strategy to target CRC independent of the p53 status. In sum, activating p53 (in p53-proficient cancer cells) or inhibiting CDK4/6 (independent of the p53 status) provide new options to improve the clinical outcome of HSP90-based therapies and to enhance colorectal cancer therapy.
Collapse
Affiliation(s)
- Tamara Isermann
- Department of Molecular Oncology, University Medical Center Göttingen, Göttingen, Germany
- Charité – Universitätsmedizin Berlin, Institute of Pathology, Laboratory of Molecular Tumor Pathology and Systems Biology, Berlin, Germany
- German Cancer Consortium (DKTK); Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kim Lucia Schneider
- Department of Molecular Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Florian Wegwitz
- Department of Gynecology and Obstetrics, University Medical Center Göttingen, Göttingen, Germany
| | - Tiago De Oliveira
- Department of General, Visceral, and Pediatric Surgery, University Medical Center Göttingen, Germany
| | - Lena-Christin Conradi
- Department of General, Visceral, and Pediatric Surgery, University Medical Center Göttingen, Germany
| | - Valery Volk
- Institute for Pathology, Hannover Medical School, Hannover, Germany
| | | | - Björn Papke
- Charité – Universitätsmedizin Berlin, Institute of Pathology, Laboratory of Molecular Tumor Pathology and Systems Biology, Berlin, Germany
- German Cancer Consortium (DKTK); Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sebastian Stintzing
- Charité – Universitätsmedizin Berlin, Department of Hematology, Oncology, and Cancer Immunology, Berlin, Germany
| | - Bettina Mundt
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Florian Kühnel
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Ute M. Moll
- Department of Pathology, Stony Brook University, Stony Brook, NY
| | | |
Collapse
|
44
|
Dibra D, Xiong S, Moyer SM, El-Naggar AK, Qi Y, Su X, Kong EK, Korkut A, Lozano G. Mutant p53 protects triple-negative breast adenocarcinomas from ferroptosis in vivo. SCIENCE ADVANCES 2024; 10:eadk1835. [PMID: 38354236 PMCID: PMC10866549 DOI: 10.1126/sciadv.adk1835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 01/12/2024] [Indexed: 02/16/2024]
Abstract
The TP53 tumor suppressor gene is mutated early in most of the patients with triple-negative breast cancer (TNBC). The most frequent TP53 alterations are missense mutations that contribute to tumor aggressiveness. Here, we used an autochthonous somatic TNBC mouse model, in which mutant p53 can be toggled on and off genetically while leaving the tumor microenvironment intact and wild-type for p53 to identify physiological dependencies on mutant p53. In TNBCs that develop in this model, deletion of two different hotspot p53R172H and p53R245W mutants triggers ferroptosis in vivo, a cell death mechanism involving iron-dependent lipid peroxidation. Mutant p53 protects cells from ferroptosis inducers, and ferroptosis inhibitors reverse the effects of mutant p53 loss in vivo. Single-cell transcriptomic data revealed that mutant p53 protects cells from undergoing ferroptosis through NRF2-dependent regulation of Mgst3 and Prdx6, which encode two glutathione-dependent peroxidases that detoxify lipid peroxides. Thus, mutant p53 protects TNBCs from ferroptotic death.
Collapse
Affiliation(s)
- Denada Dibra
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shunbin Xiong
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sydney M. Moyer
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Genetics and Epigenetics Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Adel K. El-Naggar
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yuan Qi
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xiaoping Su
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Elisabeth K. Kong
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Anil Korkut
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Guillermina Lozano
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Genetics and Epigenetics Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| |
Collapse
|
45
|
Zhao M, Wang T, Gleber-Netto FO, Chen Z, McGrail DJ, Gomez JA, Ju W, Gadhikar MA, Ma W, Shen L, Wang Q, Tang X, Pathak S, Raso MG, Burks JK, Lin SY, Wang J, Multani AS, Pickering CR, Chen J, Myers JN, Zhou G. Mutant p53 gains oncogenic functions through a chromosomal instability-induced cytosolic DNA response. Nat Commun 2024; 15:180. [PMID: 38167338 PMCID: PMC10761733 DOI: 10.1038/s41467-023-44239-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 12/05/2023] [Indexed: 01/05/2024] Open
Abstract
Inactivating TP53 mutations leads to a loss of function of p53, but can also often result in oncogenic gain-of-function (GOF) of mutant p53 (mutp53) proteins which promotes tumor development and progression. The GOF activities of TP53 mutations are well documented, but the mechanisms involved remain poorly understood. Here, we study the mutp53 interactome and find that by targeting minichromosome maintenance complex components (MCMs), GOF mutp53 predisposes cells to replication stress and chromosomal instability (CIN), leading to a tumor cell-autonomous and cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING)-dependent cytosolic DNA response that activates downstream non-canonical nuclear factor kappa light chain enhancer of activated B cell (NC-NF-κB) signaling. Consequently, GOF mutp53-MCMs-CIN-cytosolic DNA-cGAS-STING-NC-NF-κB signaling promotes tumor cell metastasis and an immunosuppressive tumor microenvironment through antagonizing interferon signaling and regulating genes associated with pro-tumorigenic inflammation. Our findings have important implications for understanding not only the GOF activities of TP53 mutations but also the genome-guardian role of p53 and its inactivation during tumor development and progression.
Collapse
Affiliation(s)
- Mei Zhao
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Tianxiao Wang
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Head and Neck Surgery, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Frederico O Gleber-Netto
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Zhen Chen
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Daniel J McGrail
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, 44195, USA
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Javier A Gomez
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Wutong Ju
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Mayur A Gadhikar
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Wencai Ma
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Li Shen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Qi Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Ximing Tang
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Sen Pathak
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Maria Gabriela Raso
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jared K Burks
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Shiaw-Yih Lin
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jing Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Asha S Multani
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Curtis R Pickering
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Surgery-Otolaryngology, Yale School of Medicine, New Haven, CT, 06250, USA
| | - Junjie Chen
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jeffrey N Myers
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Ge Zhou
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
46
|
Efe G, Dunbar KJ, Sugiura K, Cunningham K, Carcamo S, Karaiskos S, Tang Q, Cruz-Acuña R, Resnick-Silverman L, Peura J, Lu C, Hasson D, Klein-Szanto AJ, Taylor AM, Manfredi JJ, Prives C, Rustgi AK. p53 Gain-of-Function Mutation Induces Metastasis via BRD4-Dependent CSF-1 Expression. Cancer Discov 2023; 13:2632-2651. [PMID: 37676642 PMCID: PMC10841313 DOI: 10.1158/2159-8290.cd-23-0601] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/15/2023] [Accepted: 09/05/2023] [Indexed: 09/08/2023]
Abstract
TP53 mutations are frequent in esophageal squamous cell carcinoma (ESCC) and other SCCs and are associated with a proclivity for metastasis. Here, we report that colony-stimulating factor-1 (CSF-1) expression is upregulated significantly in a p53-R172H-dependent manner in metastatic lung lesions of ESCC. The p53-R172H-dependent CSF-1 signaling, through its cognate receptor CSF-1R, increases tumor cell invasion and lung metastasis, which in turn is mediated in part through Stat3 phosphorylation and epithelial-to-mesenchymal transition (EMT). In Trp53R172H tumor cells, p53 occupies the Csf-1 promoter. The Csf-1 locus is enriched with histone 3 lysine 27 acetylation (H3K27ac), which is likely permissive for fostering an interaction between bromodomain-containing domain 4 (BRD4) and p53-R172H to regulate Csf-1 transcription. Inhibition of BRD4 not only reduces tumor invasion and lung metastasis but also reduces circulating CSF-1 levels. Overall, our results establish a novel p53-R172H-dependent BRD4-CSF-1 axis that promotes ESCC lung metastasis and suggest avenues for therapeutic strategies for this difficult-to-treat disease. SIGNIFICANCE The invasion-metastasis cascade is a recalcitrant barrier to effective cancer therapy. We establish that the p53-R172H-dependent BRD4-CSF-1 axis is a mediator of prometastatic properties, correlates with patient survival and tumor stages, and its inhibition significantly reduces tumor cell invasion and lung metastasis. This axis can be exploited for therapeutic advantage. This article is featured in Selected Articles from This Issue, p. 2489.
Collapse
Affiliation(s)
- Gizem Efe
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, 10032, USA
- Department of Genetics and Development, Columbia University, New York, NY, 10032, USA
| | - Karen J. Dunbar
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, 10032, USA
| | - Kensuke Sugiura
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, 10032, USA
| | - Katherine Cunningham
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, 10032, USA
| | - Saul Carcamo
- Tisch Cancer Institute Bioinformatics for Next Generation Sequencing (BiNGS) core, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Spyros Karaiskos
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, 10032, USA
| | - Qiaosi Tang
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, 10032, USA
| | - Ricardo Cruz-Acuña
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, 10032, USA
| | - Lois Resnick-Silverman
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jessica Peura
- Division of Hematology-Oncology, University of Massachusetts Medical School, Worcester, MA, 01655, USA
| | - Chao Lu
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, 10032, USA
- Department of Genetics and Development, Columbia University, New York, NY, 10032, USA
| | - Dan Hasson
- Tisch Cancer Institute Bioinformatics for Next Generation Sequencing (BiNGS) core, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | | | - Alison M. Taylor
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, 10032, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY, 10032, USA
| | - James J. Manfredi
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Carol Prives
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, 10032, USA
- Department of Biological Sciences, Columbia University, Columbia University, New York, NY, 10032, USA
| | - Anil K. Rustgi
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, 10032, USA
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY, 10032, USA
| |
Collapse
|
47
|
Dong Y, Xu W, Qi D, Qu H, Jin Q, Sun M, Wang X, Quan C. CLDN6 inhibits colorectal cancer proliferation dependent on restraining p53 ubiquitination via ZO-1/PTEN axis. Cell Signal 2023; 112:110930. [PMID: 37852424 DOI: 10.1016/j.cellsig.2023.110930] [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: 09/04/2023] [Revised: 10/03/2023] [Accepted: 10/15/2023] [Indexed: 10/20/2023]
Abstract
Colorectal cancer (CRC) is one of the most common cancers in the world. Abnormal proliferation is a chief characteristic of cancer and is the initiation of CRC progression. As an important component of tight junctions, CLDN6 regulates the proliferation of multiple tumors. Our previous study showed that CLDN6 was low expressed in CRC, and CLDN6 overexpression inhibited CRC proliferation. However, the specific mechanism of how CLDN6 works remains unclear. This research aimed to reveal the relationship between CLDN6 and clinical features, as well as the molecular mechanism by which CLDN6 inhibited CRC proliferation. We found that low expression of CLDN6 was associated with pathological grade and prognosis of CRC patients, and confirmed that CLDN6 inhibited CRC proliferation dependent on p53. Mechanically, we elucidated that CLDN6 regulated ubiquitination to enhance p53 stability and nuclear import by PTEN/AKT/MDM2 pathway. Through the PDZ-binding motif (PBM), CLDN6 bound to ZO-1 to interact with PTEN, and regulate AKT/MDM2 pathway. Collectively, our data enriched the theoretical basis for CLDN6 as a potential biomarker for diagnosis, therapy and prognosis of CRC.
Collapse
Affiliation(s)
- Yuan Dong
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun 130021, Jilin, China
| | - Wenhong Xu
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun 130021, Jilin, China
| | - Da Qi
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun 130021, Jilin, China
| | - Huinan Qu
- Department of Histology and Embryology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun 130021, Jilin, China
| | - Qiu Jin
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun 130021, Jilin, China
| | - Minghao Sun
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun 130021, Jilin, China
| | - Xudong Wang
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun 130000, Jilin, China.
| | - Chengshi Quan
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun 130021, Jilin, China.
| |
Collapse
|
48
|
Deng H, Gong X, Ji G, Li C, Cheng S. KIF2C promotes clear cell renal cell carcinoma progression via activating JAK2/STAT3 signaling pathway. Mol Cell Probes 2023; 72:101938. [PMID: 37863123 DOI: 10.1016/j.mcp.2023.101938] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/14/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023]
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) is one of the most common malignant tumors that can be highly aggressive. Despite advances in the exploration of its underlying molecular biology, the clinical outcome for advanced ccRCC is still unsatisfied. Recently, more attention was paid to the functions of Kinesin family member 2C (KIF2C) in cancer progression, while the specific function of KIF2C in ccRCC has not been sufficiently elucidated. The present study aims to investigate the role of KIF2C in the progression of ccRCC and reveal potential mechanisms. METHODS Expression of KIF2C in ccRCC tissues and adjacent normal tissue was compared and the association of KIF2C expression level with tumor grade, stage, and metastasis were analyzed using online web tool. Kaplan-Meier survival was performed to detect the association of KIF2C expression and patient' prognosis. Stably cell lines with KIF2C knockdown or overexpression were constructed by lentivirus infection. CCK-8, colony formation, scratch healing, and transwell invasion assays were carried out to explore the effect of KIF2C knockdown or overexpression on the proliferation, migration, and invasion of ccRCC cells. Gene set enrichment analysis (GSEA) was conducted to reveal signaling pathways associated with KIF2C expression. The effect of KIF2C on JAK2/STAT3 signaling pathway were explored by western blot assay. RESULTS KIF2C expression was significantly upregulated in ccRCC tissues and was higher with the increase of tumor grade, stage, and metastasis. Higher expression of KIF2C was correlated with worse overall survival and diseases free survival in ccRCC patients. Silence of KIF2C inhibited proliferation, migration, and invasion in ccRCC cells. Conversely, overexpression of KIF2C had the opposite effect. GSEA results showed that JAK/STAT signaling pathway was markedly enriched in KIF2Chigh group. Pearson' correlation revealed that KIF2C expression was significantly associated with genes in JAK2/STAT3 signaling. Western blot results showed that KIF2C knockdown decreased protein expression of p-JAK2 and p-STAT3, and KIF2C overexpression increased the phosphorylation of JAK2 and STAT3. AG490, a JAK2/STAT3 signaling inhibitor, could partly impair the tumor-promoting effects of KIF2C in ccRCC. CONCLUSION KIF2C expression was significantly upregulated in ccRCC and correlated with tumor grade, stage, metastasis, and patients' prognosis. KIF2C promoted ccRCC progression via activating JAK2/STAT3 signaling pathway, and KIF2C might be a novel target in ccRCC therapy.
Collapse
Affiliation(s)
- Hao Deng
- Department of Urology, The First People's Hospital of Jingzhou, Jingzhou, 434000, PR China
| | - Xiaobo Gong
- Department of Urology, The First People's Hospital of Jingzhou, Jingzhou, 434000, PR China
| | - Guanghai Ji
- Department of Urology, The First People's Hospital of Jingzhou, Jingzhou, 434000, PR China
| | - Chenglong Li
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, PR China.
| | - Shaoping Cheng
- Department of Urology, The First People's Hospital of Jingzhou, Jingzhou, 434000, PR China.
| |
Collapse
|
49
|
Čižmáriková M, Michalková R, Mirossay L, Mojžišová G, Zigová M, Bardelčíková A, Mojžiš J. Ellagic Acid and Cancer Hallmarks: Insights from Experimental Evidence. Biomolecules 2023; 13:1653. [PMID: 38002335 PMCID: PMC10669545 DOI: 10.3390/biom13111653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/10/2023] [Accepted: 11/12/2023] [Indexed: 11/26/2023] Open
Abstract
Cancer is a complex and multifaceted disease with a high global incidence and mortality rate. Although cancer therapy has evolved significantly over the years, numerous challenges persist on the path to effectively combating this multifaceted disease. Natural compounds derived from plants, fungi, or marine organisms have garnered considerable attention as potential therapeutic agents in the field of cancer research. Ellagic acid (EA), a natural polyphenolic compound found in various fruits and nuts, has emerged as a potential cancer prevention and treatment agent. This review summarizes the experimental evidence supporting the role of EA in targeting key hallmarks of cancer, including proliferation, angiogenesis, apoptosis evasion, immune evasion, inflammation, genomic instability, and more. We discuss the molecular mechanisms by which EA modulates signaling pathways and molecular targets involved in these cancer hallmarks, based on in vitro and in vivo studies. The multifaceted actions of EA make it a promising candidate for cancer prevention and therapy. Understanding its impact on cancer biology can pave the way for developing novel strategies to combat this complex disease.
Collapse
Affiliation(s)
- Martina Čižmáriková
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (M.Č.); (R.M.); (M.Z.); (A.B.)
| | - Radka Michalková
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (M.Č.); (R.M.); (M.Z.); (A.B.)
| | - Ladislav Mirossay
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (M.Č.); (R.M.); (M.Z.); (A.B.)
| | - Gabriela Mojžišová
- Center of Clinical and Preclinical Research MEDIPARK, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia;
| | - Martina Zigová
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (M.Č.); (R.M.); (M.Z.); (A.B.)
| | - Annamária Bardelčíková
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (M.Č.); (R.M.); (M.Z.); (A.B.)
| | - Ján Mojžiš
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (M.Č.); (R.M.); (M.Z.); (A.B.)
| |
Collapse
|
50
|
Zhou RW, Harpaz N, Itzkowitz SH, Parsons RE. Molecular mechanisms in colitis-associated colorectal cancer. Oncogenesis 2023; 12:48. [PMID: 37884500 PMCID: PMC10603140 DOI: 10.1038/s41389-023-00492-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/30/2023] [Accepted: 09/11/2023] [Indexed: 10/28/2023] Open
Abstract
Sustained chronic inflammation of the large intestine leads to tissue damage and repair, which is associated with an increased incidence of colitis-associated colorectal cancer (CAC). The genetic makeup of CAC is somewhat similar to sporadic colorectal carcinoma (sCRC), but there are differences in the sequence and timing of alterations in the carcinogenesis process. Several models have been developed to explain the development of CAC, particularly the "field cancerization" model, which proposes that chronic inflammation accelerates mutagenesis and selects for the clonal expansion of phenotypically normal, pro-tumorigenic cells. In contrast, the "Big Bang" model posits that tumorigenic clones with multiple driver gene mutations emerge spontaneously. The details of CAC tumorigenesis-and how they differ from sCRC-are not yet fully understood. In this Review, we discuss recent genetic, epigenetic, and environmental findings related to CAC pathogenesis in the past five years, with a focus on unbiased, high-resolution genetic profiling of non-dysplastic field cancerization in the context of inflammatory bowel disease (IBD).
Collapse
Affiliation(s)
- Royce W Zhou
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Molecular Medicine Program, Internal Medicine Residency Program, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Noam Harpaz
- The Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Steven H Itzkowitz
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- The Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Ramon E Parsons
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|