1
|
Ruan J, Li Q, Jin Y, Yin J, Ye C, Cheng F, Xu S, Chen R, Liu C, Rong X, Jiang M, Fu W, Zheng D, Chen J, Bao X, Wang H, Sheng J, Zhao P. Multiple-omics analysis reveals a dedifferentiation-immune loop in intrahepatic cholangiocarcinoma. Mol Ther 2025; 33:1803-1824. [PMID: 39943686 PMCID: PMC11997497 DOI: 10.1016/j.ymthe.2025.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 12/21/2024] [Accepted: 02/07/2025] [Indexed: 03/10/2025] Open
Abstract
Intrahepatic cholangiocarcinoma (ICC) is known for its diverse cell types and resistance to standard treatments, highlighting the importance of understanding its tumor microenvironment (TME) for improved prognostic accuracy and therapeutic innovation. Our study used a multi-omics approach to analyze the ICC TME in both human and mouse samples, linking survival outcomes to the complex cellular interactions within the TME. We discovered a dedifferentiation phenomenon in ICC cells driven by the Yes-associated protein (YAP) pathway, influenced by tumor-associated macrophages (TAMs). Conversely, ICC cells promoted an immunosuppressive environment in TAMs. Targeting TAMs in a transgenic mouse model disrupted this loop, enhancing T cell responses and suggesting a novel immunotherapy avenue for ICC. Our findings reveal a reciprocal dedifferentiation-immunosuppression loop between ICC cells and TAMs, advocating TAM targeting as a promising therapy and highlighting the potential of macrophage modulation in ICC treatment.
Collapse
Affiliation(s)
- Jian Ruan
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University and Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Hangzhou 310003, Zhejiang Province, People's Republic of China
| | - Qiong Li
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University and Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Hangzhou 310003, Zhejiang Province, People's Republic of China
| | - Yuzhi Jin
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University and Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Hangzhou 310003, Zhejiang Province, People's Republic of China
| | - Jie Yin
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Institute of Genetics, Zhejiang University and Department of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Chanqi Ye
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University and Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Hangzhou 310003, Zhejiang Province, People's Republic of China
| | - Fei Cheng
- Pathology Department, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, People's Republic of China
| | - Shuaishuai Xu
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University and Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Hangzhou 310003, Zhejiang Province, People's Republic of China
| | - Ruyin Chen
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University and Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Hangzhou 310003, Zhejiang Province, People's Republic of China
| | - Chuan Liu
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University and Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Hangzhou 310003, Zhejiang Province, People's Republic of China
| | - Xiaoxiang Rong
- Department of Oncology, Nanfang Hospital, Southern medical University, Guangzhou 510000, Guangdong Province, People's Republic of China
| | - Ming Jiang
- The Children's Hospital, Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou 310058, Zhejiang Province, People's Republic of China
| | - Wenguang Fu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, People's Republic of China
| | - Dayong Zheng
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, Guangdong Province, People's Republic of China
| | - Jinzhang Chen
- Department of Oncology, Nanfang Hospital, Southern medical University, Guangzhou 510000, Guangdong Province, People's Republic of China
| | - Xuanwen Bao
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University and Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Hangzhou 310003, Zhejiang Province, People's Republic of China
| | - Houhong Wang
- Department of General Surgery, The First Hospital Affiliated to Fuyang Normal University, Fuyang 236006, Anhui Province, People's Republic of China; Department of General Surgery, The Affiliated Bozhou Hospital of Anhui Medical University, Bozhou 236800, Anhui Province, People's Republic of China.
| | - Jianpeng Sheng
- College of Computer Science and Technology, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, Jiangsu Province, People's Republic of China; Chinese Institutes for Medical Research, Beijing 100000, People's Republic of China.
| | - Peng Zhao
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University and Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Hangzhou 310003, Zhejiang Province, People's Republic of China.
| |
Collapse
|
2
|
Zhi R, Li Q, Zhang H, Fan F. VPS45 Contributes to the Progression of Hepatocellular Carcinoma by Triggering the Wnt/β-Catenin Signaling Pathway. Mol Carcinog 2025; 64:744-755. [PMID: 39835603 PMCID: PMC11890426 DOI: 10.1002/mc.23884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 12/26/2024] [Accepted: 01/03/2025] [Indexed: 01/22/2025]
Abstract
Vacuolar protein sorting 45 (VPS45) has recently been implicated in the development of ovarian cancer and non-small cell lung cancer. However, its role in the onset and progression of hepatocellular carcinoma (HCC) remains unclear. This study aims to elucidate the function of VPS45 in HCC. Bioassays were conducted to assess the prognostic significance of VPS45 in HCC. Techniques such as western blotting and real-time quantitative polymerase chain reaction (qRT-PCR) were used to confirm the expression levels of VPS45 in HCC tissues and cell lines, as well as to evaluate the expression of downstream effectors in its potential tumorigenic pathways. The impact of VPS45 on HCC cell invasion, proliferation, and migration was assessed using the Cell Counting Kit-8 (CCK-8), wound healing, and transwell assays. Furthermore, the effect of VPS45 on HCC tumorigenesis in vivo was evaluated through subcutaneous tumor formation assays in BALB/c nude mice. VPS45 is markedly overexpressed in both HCC tissues and cell lines. Its expression escalates with advancing tumor grade and clinical stage, and high VPS45 levels are indicative of poor prognosis. In vitro experiments revealed that VPS45 overexpression significantly boosts HCC cell proliferation, migration, and invasion. Conversely, VPS45 knockdown hindered HCC progression in vivo. Investigation into pathway protein expression suggests that VPS45 facilitates HCC progression through its involvement in the Wnt/β-catenin signaling pathway. The overexpression of VPS45 contributes to the development of malignant phenotypes in HCC cells, resulting in a poor prognosis. Targeting VPS45 may offer a viable therapeutic strategy for managing HCC.
Collapse
Affiliation(s)
- Renhou Zhi
- Department of General SurgeryShanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi HospitalTaiyuanChina
| | - Qi Li
- Department of Anorectal SurgeryShanxi Provincial People's HospitalTaiyuanChina
| | - Huiqin Zhang
- Medical DepartmentShanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi HospitalTaiyuanChina
| | - Fan Fan
- Department of GastroenterologyShanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical UniversityTaiyuanChina
| |
Collapse
|
3
|
Jiang X, Ge X, Huang Y, Xie F, Chen C, Wang Z, Tao W, Zeng S, Lv L, Zhan Y, Bao L. Drug resistance in TKI therapy for hepatocellular carcinoma: Mechanisms and strategies. Cancer Lett 2025; 613:217472. [PMID: 39832650 DOI: 10.1016/j.canlet.2025.217472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
Tyrosine kinase inhibitors (TKIs) are such as sorafenib the first-line therapeutic drugs for patients with advanced hepatocellular carcinoma. However, patients with TKI-resistant advanced liver cancer are insensitive to TKI treatment, resulting in limited survival benefits. This paper comprehensively reviewed the mechanisms underlying TKI resistance in hepatocytes, investigating activation of tumor signaling pathways, epigenetic regulation, tumor microenvironment, and metabolic reprogramming. Based on resistance mechanisms, it also reviews preclinical and clinical studies of drug resistance strategies and summarizes targeted therapy combined with immunotherapy currently in investigational clinical trials. Understanding the interactions and clinical studies of these resistance mechanisms offers new hope for improving and prolonging patient survival.
Collapse
Affiliation(s)
- Xue Jiang
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Xiaoying Ge
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Yueying Huang
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Fangyuan Xie
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Chun Chen
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Zijun Wang
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Wanru Tao
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Sailiang Zeng
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Lei Lv
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Yangyang Zhan
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Leilei Bao
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| |
Collapse
|
4
|
Guo J, Chen L, Dai B, Sui C, Dong Z, Chen K, Duan K, Fang K, Li A, Wang K, Geng L. TM4SF1 overexpression in tumor-associated endothelial cells promotes microvascular invasion in hepatocellular carcinoma. Front Oncol 2025; 15:1526177. [PMID: 40123905 PMCID: PMC11925789 DOI: 10.3389/fonc.2025.1526177] [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: 11/11/2024] [Accepted: 01/20/2025] [Indexed: 03/25/2025] Open
Abstract
Background Microvascular invasion (MVI) is linked to poor prognosis, early recurrence and post-surgical intrahepatic metastasis of hepatocellular carcinoma (HCC) but roles of tumor-associated endothelial cells (TECs) remain unclear. The aim of the current study was to investigate the role of TECs in microvascular invasion in HCC. Methods Single-cell RNA sequencing (scRNA-seq) data from three patients with MVI and two patients with non-MVI HCC were used to identify TECs subpopulations via Seurat R package. Using bioinformatics analysis identified co-expression modules associated with MVI in TECs. Differential gene expression analysis, KME values and Gene Expression Profiling Interactive Analysis (GEPIA) survival were utilized to identify genes with significant involvement. TECs subgroup developmental trajectory was analyzed using monocle2. Five additional spatial transcriptomics (ST) datasets and four HCC postoperative pathological specimens were used to validate the differential expression of subgroups of TECs and hub genes between MVI and non-MVI groups. Results Distinct TECs subgroups had significant heterogeneity between datasets from MVI and non-MVI patients. MVI samples had TECs subgroups with increased levels of the epithelial-mesenchymal transition (EMT), endothelial cell migration and angiogenesis. Opposing EMT development was found in MVI TECs relative to non-MVI TECs. TM4SF1 was highly expressed in TECs undergoing the EMT and is thought to be linked to MVI. Conclusion TECs with elevated TM4SF1 expression facilitate MVI during HCC via an effect on the EMT, suggesting the potential of TM4SF1 as a therapeutic target.
Collapse
Affiliation(s)
- Junwu Guo
- Department of Special Treatment, Third Affiliated Hospital of Naval Medical University (Eastern Hepatobiliary Surgery Hospital), Shanghai, China
| | - Liangrui Chen
- Department of Hepatic Surgery II, Third Affiliated Hospital of Naval Medical University (Eastern Hepatobiliary Surgery Hospital), Shanghai, China
| | - Binghua Dai
- Department of Special Treatment, Third Affiliated Hospital of Naval Medical University (Eastern Hepatobiliary Surgery Hospital), Shanghai, China
| | - Chengjun Sui
- Department of Special Treatment, Third Affiliated Hospital of Naval Medical University (Eastern Hepatobiliary Surgery Hospital), Shanghai, China
| | - Zhitao Dong
- Department of Special Treatment, Third Affiliated Hospital of Naval Medical University (Eastern Hepatobiliary Surgery Hospital), Shanghai, China
| | - Keji Chen
- Department of Special Treatment, Third Affiliated Hospital of Naval Medical University (Eastern Hepatobiliary Surgery Hospital), Shanghai, China
| | - Kecai Duan
- Department of Special Treatment, Third Affiliated Hospital of Naval Medical University (Eastern Hepatobiliary Surgery Hospital), Shanghai, China
| | - Kunpeng Fang
- Department of Special Treatment, Third Affiliated Hospital of Naval Medical University (Eastern Hepatobiliary Surgery Hospital), Shanghai, China
| | - Aijun Li
- Department of Special Treatment, Third Affiliated Hospital of Naval Medical University (Eastern Hepatobiliary Surgery Hospital), Shanghai, China
| | - Kui Wang
- Department of Hepatic Surgery II, Third Affiliated Hospital of Naval Medical University (Eastern Hepatobiliary Surgery Hospital), Shanghai, China
| | - Li Geng
- Department of Special Treatment, Third Affiliated Hospital of Naval Medical University (Eastern Hepatobiliary Surgery Hospital), Shanghai, China
| |
Collapse
|
5
|
Yan B, Lu Q, Gao T, Xiao K, Zong Q, Lv H, Lv G, Wang L, Liu C, Yang W, Jiang G. CD146 regulates the stemness and chemoresistance of hepatocellular carcinoma via JAG2-NOTCH signaling. Cell Death Dis 2025; 16:150. [PMID: 40032820 DOI: 10.1038/s41419-025-07470-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 01/29/2025] [Accepted: 02/20/2025] [Indexed: 03/05/2025]
Abstract
CD146 plays a key role in cancer progression and metastasis. Cancer stem cells (CSCs) are responsible for tumor initiation, drug resistance, metastasis, and recurrence. In this study, we explored the role of CD146 in the regulation of liver CSCs. Here, we demonstrated that CD146 was highly expressed in liver CSCs. CD146 overexpression promoted the self-renewal ability and chemoresistance of Hepatocellular Carcinoma (HCC) cells in vitro and tumorigenicity in vivo. Inversely, knockdown of CD146 restrained these abilities. Mechanistically, CD146 activated the NF-κB signaling to up-regulate JAG2 expression and activated the Notch signaling, which resulted in increased stemness of HCC. Furthermore, JAG2 overexpression restored the Notch signaling activity, the stemness, and chemotherapeutic resistance caused by CD146 knockdown. These results demonstrated that CD146 positively regulates HCC stemness by activating the JAG2-NOTCH signaling. Combined targeting of CD146 and JAG2 may represent a novel therapeutic strategy for HCC treatment.
Collapse
MESH Headings
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/drug therapy
- Humans
- Liver Neoplasms/pathology
- Liver Neoplasms/metabolism
- Liver Neoplasms/genetics
- Liver Neoplasms/drug therapy
- Jagged-2 Protein/metabolism
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Signal Transduction
- Drug Resistance, Neoplasm
- Animals
- Receptors, Notch/metabolism
- CD146 Antigen/metabolism
- CD146 Antigen/genetics
- Cell Line, Tumor
- Mice, Nude
- Mice
- Gene Expression Regulation, Neoplastic
- NF-kappa B/metabolism
- Mice, Inbred BALB C
- Male
Collapse
Affiliation(s)
- Bing Yan
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital, Yangzhou, 225000, China
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225000, China
- Department of General Surgery, Pingxiang People's Hospital, Pingxiang, 337000, China
| | - QiuYu Lu
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Naval Medical University (Second Military Medical University), Shanghai, 200438, China
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China
| | - TianMing Gao
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital, Yangzhou, 225000, China
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225000, China
| | - KunQing Xiao
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital, Yangzhou, 225000, China
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225000, China
| | - QianNi Zong
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Naval Medical University (Second Military Medical University), Shanghai, 200438, China
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China
| | - HongWei Lv
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Naval Medical University (Second Military Medical University), Shanghai, 200438, China
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China
| | - GuiShuai Lv
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Naval Medical University (Second Military Medical University), Shanghai, 200438, China
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China
| | - Liang Wang
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Naval Medical University (Second Military Medical University), Shanghai, 200438, China
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China
| | - ChunYing Liu
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Naval Medical University (Second Military Medical University), Shanghai, 200438, China.
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China.
- Shanghai Key Laboratory of Hepatobiliary Tumor Biology, Shanghai, 200438, China.
- Key Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer, Ministry of Education, Shanghai, 200438, China.
| | - Wen Yang
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Naval Medical University (Second Military Medical University), Shanghai, 200438, China.
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China.
- Shanghai Key Laboratory of Hepatobiliary Tumor Biology, Shanghai, 200438, China.
- Key Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer, Ministry of Education, Shanghai, 200438, China.
| | - GuoQing Jiang
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital, Yangzhou, 225000, China.
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225000, China.
| |
Collapse
|
6
|
Yuan CR, Lee TKW. TM4SF1 - A new immune target for treatment of hepatocellular carcinoma: Editorial on "Targeting TM4SF1 promotes tumor senescence enhancing CD8+ T cell cytotoxic function in hepatocellular carcinoma". Clin Mol Hepatol 2025; 31:646-649. [PMID: 39924999 DOI: 10.3350/cmh.2025.0102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 02/06/2025] [Indexed: 02/11/2025] Open
Affiliation(s)
- Chen Rui Yuan
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong
| | - Terence Kin Wah Lee
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong
- State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Hong Kong
| |
Collapse
|
7
|
Li J, Bao X, Yu W, Chen X, Ni Y, Shi Y, Wang J, Sun Y, Chen A, Zhou W, Ye H. FOXA1 activates NOLC1 transcription through NOTCH pathway to promote cell stemness in lung adenocarcinoma. Kaohsiung J Med Sci 2025; 41:e12930. [PMID: 39789998 PMCID: PMC11827541 DOI: 10.1002/kjm2.12930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 12/13/2024] [Accepted: 12/15/2024] [Indexed: 01/12/2025] Open
Abstract
Tumor cell stemness plays a pivotal role in generating functional heterogeneity within tumors and is implicated in essential processes such as drug resistance, metastasis, and cell proliferation. Therefore, creating novel tumor diagnostic techniques and therapeutic plans requires a knowledge of the possible processes that preserve the stem cell-like qualities of cancers. Bioinformatics analysis of NOLC1 expression in lung adenocarcinoma (LUAD) and prediction of its upstream transcription factors and their binding sites were completed. RT-qPCR detection of NOLC1 and FOXA1 expression, colony formation assay of cell proliferation, Transwell assay of cell invasion, sphere formation assay of cell stemness, western blot detection of CD133, OCT4, GLI1, NOTCH1 and Hes1 expression, CCK-8 assay of IC50 value of cisplatin, and ChIP and dual-luciferase reporter validation of binding relationship between NOLC1 and FOXA1 were done. NOLC1 expression was elevated in LUAD cells and tissues. Decreased NOLC1 expression inhibited the proliferation and invasive capacity of LUAD cells, prevented LUAD cells from becoming stem cells, and suppressed cisplatin resistance in the cells. Rescue tests demonstrated that NOLC1 activated the NOTCH pathway to increase the stemness of LUAD cells and promoted cisplatin resistance in LUAD cells. The activation of NOLC1 transcription by FOXA1 was validated by bioinformatics prediction and molecular verification, and the FOXA1/NOLC1 axis enhanced the stemness of LUAD cells. Activation of NOLC1 transcription by FOXA1 through NOTCH pathway promoted stemness of LUAD. FOXA1/NOLC1 axis is expected to become a new target for inhibiting stemness of LUAD cells.
Collapse
Affiliation(s)
- Ji‐Fa Li
- Department of Respiratory and Critical Care Medicine of Affiliated Yueqing HospitalWenzhou Medical UniversityYueqingChina
| | - Xiao‐Qiong Bao
- Department of Respiratory and Critical Care Medicine of Affiliated Yueqing HospitalWenzhou Medical UniversityYueqingChina
| | - Wen‐Wen Yu
- Department of Respiratory and Critical Care Medicine of Affiliated Yueqing HospitalWenzhou Medical UniversityYueqingChina
| | - Xiang‐Xiang Chen
- Department of Respiratory and Critical Care Medicine of Affiliated Yueqing HospitalWenzhou Medical UniversityYueqingChina
| | - Yang‐Yang Ni
- Department of Respiratory and Critical Care Medicine of Affiliated Yueqing HospitalWenzhou Medical UniversityYueqingChina
| | - Yu‐Bo Shi
- Department of Respiratory and Critical Care Medicine of Affiliated Yueqing HospitalWenzhou Medical UniversityYueqingChina
| | - Jin‐Cong Wang
- Department of Respiratory and Critical Care Medicine of Affiliated Yueqing HospitalWenzhou Medical UniversityYueqingChina
| | - Yang‐Jie Sun
- Department of Respiratory and Critical Care Medicine of Affiliated Yueqing HospitalWenzhou Medical UniversityYueqingChina
| | - Ai‐Li Chen
- Department of Respiratory and Critical Care Medicine of Affiliated Yueqing HospitalWenzhou Medical UniversityYueqingChina
| | - Wei‐Long Zhou
- Department of Respiratory and Critical Care Medicine of Affiliated Yueqing HospitalWenzhou Medical UniversityYueqingChina
| | - Hua Ye
- Department of Respiratory and Critical Care Medicine of Affiliated Yueqing HospitalWenzhou Medical UniversityYueqingChina
| |
Collapse
|
8
|
Du Q, Zhang M, Gao A, He T, Guo M. Epigenetic silencing ZSCAN23 promotes pancreatic cancer growth by activating Wnt signaling. Cancer Biol Ther 2024; 25:2302924. [PMID: 38226836 PMCID: PMC10793710 DOI: 10.1080/15384047.2024.2302924] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 01/04/2024] [Indexed: 01/17/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most malignant tumor. Zinc finger and SCAN domain-containing protein 23 (ZSCAN23) is a new member of the SCAN domain family. The expression regulation and biological function remain to be elucidated. In this study, we explored the epigenetic regulation and the function of ZSCAN23 in PDAC. ZSCAN23 was methylated in 60.21% (171/284) of PDAC and its expression was regulated by promoter region methylation. The expression of ZSCAN23 inhibited cell proliferation, colony formation, migration, invasion, and induced apoptosis and G1/S phase arrest. ZSCAN23 suppressed Panc10.05 cell xenograft growth in mice. Mechanistically, ZSCAN23 inhibited Wnt signaling by interacting with myosin heavy chain 9 (MYH9) in pancreatic cancer cells. ZSCAN23 is frequently methylated in PDAC and may serve as a detective marker. ZSCAN23 suppresses PDAC cell growth both in vitro and in vivo.
Collapse
Affiliation(s)
- Qian Du
- Department of Gastroenterology and Hepatology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, People's Republic of China
- Department of Gastroenterology and Hepatology, the First Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Meiying Zhang
- Department of Gastroenterology and Hepatology, the First Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Aiai Gao
- Department of Gastroenterology and Hepatology, the First Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Tao He
- Department of Pathology, Characteristic Medical Center of the Chinese People's Armed Police Force, Tianjin, People's Republic of China
| | - Mingzhou Guo
- Department of Gastroenterology and Hepatology, the First Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
- National Key Laboratory of Kidney Diseases, the First Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
| |
Collapse
|
9
|
Chen S, He Z, Cai K, Zhang Y, Zhu H, Pang C, Zhang J, Wang D, Xu X. TRIM59/RBPJ positive feedback circuit confers gemcitabine resistance in pancreatic cancer by activating the Notch signaling pathway. Cell Death Dis 2024; 15:932. [PMID: 39725730 DOI: 10.1038/s41419-024-07324-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 12/07/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024]
Abstract
Pancreatic cancer (PC) is one of the most lethal malignant tumors that lacks effective treatment, and gemcitabine-based chemoresistance occurs frequently. Therefore, new therapeutic strategies for PC are urgently needed. Tripartite motif containing 59 (TRIM59) plays an important role in breast and lung cancer chemoresistance. However, the association between TRIM59 and gemcitabine resistance in PC remains unclear. We identified TRIM59 as an innovative E3 ubiquitin ligase that activated Notch signaling in PC. TRIM59 levels were increased in PC and positively correlated with poor prognosis and gemcitabine resistance in PC patients. TRIM59 facilitated gemcitabine resistance in PC cells in vitro and in vivo. TRIM59 interacted with recombination signal binding protein for immunoglobulin kappa J region (RBPJ) and stabilized it by promoting K63-linked ubiquitination. RBPJ transcriptionally upregulated TRIM59 expression, forming a positive feedback loop with TRIM59. We identified a novel TRIM59 inhibitor, catechin, and confirmed that it sensitized PC cells to gemcitabine. TRIM59 conferred gemcitabine resistance in PC by promoting RBPJ K63-linked ubiquitination, followed by activating Notch signaling. Therefore, our study provides a promising target for gemcitabine sensitization in PC treatment.
Collapse
Affiliation(s)
- Shiyu Chen
- Department of Hepatobiliary Pancreatic Surgery, South China Hospital, Medical School, Shenzhen University, Shenzhen, 518116, P. R. China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, China
| | - Zhiwei He
- Department of Hepatobiliary Surgery, Shenzhen University General Hospital & Shenzhen University Clinical Medical Academy Center, Shenzhen University, Shenzhen, 518000, China
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, 550001, China
| | - Kun Cai
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, China
- Department of Gastrointestinal Surgery, South China Hospital, Medical School, Shenzhen University, Shenzhen, 518116, P. R. China
| | - Yan Zhang
- Department of Hepatobiliary Pancreatic Surgery, South China Hospital, Medical School, Shenzhen University, Shenzhen, 518116, P. R. China
| | - Hongyan Zhu
- Department of Hepatobiliary Pancreatic Surgery, South China Hospital, Medical School, Shenzhen University, Shenzhen, 518116, P. R. China
| | - Chong Pang
- Department of Hepatobiliary Pancreatic Surgery, South China Hospital, Medical School, Shenzhen University, Shenzhen, 518116, P. R. China
| | - Jiaqi Zhang
- Department of Hepatobiliary Pancreatic Surgery, South China Hospital, Medical School, Shenzhen University, Shenzhen, 518116, P. R. China
| | - Dong Wang
- Department of Hepatobiliary Pancreatic Surgery, South China Hospital, Medical School, Shenzhen University, Shenzhen, 518116, P. R. China
| | - Xundi Xu
- Department of Hepatobiliary Pancreatic Surgery, South China Hospital, Medical School, Shenzhen University, Shenzhen, 518116, P. R. China.
| |
Collapse
|
10
|
Chen Y, Dai S, Cheng CS, Chen L. Lenvatinib and immune-checkpoint inhibitors in hepatocellular carcinoma: mechanistic insights, clinical efficacy, and future perspectives. J Hematol Oncol 2024; 17:130. [PMID: 39709431 DOI: 10.1186/s13045-024-01647-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 11/29/2024] [Indexed: 12/23/2024] Open
Abstract
Lenvatinib is a multi-target tyrosine kinase inhibitor widely used in the treatment of hepatocellular carcinoma (HCC). Its primary mechanism of action involves inhibiting signal pathways such as vascular endothelial growth factor receptors (VEGFR) and fibroblast growth factor receptors (FGFR), thereby reducing tumor cell proliferation and angiogenesis and affecting the tumor's immune microenvironment. In the treatment of liver cancer, although lenvatinib monotherapy has shown good clinical effect, the problem of drug resistance is becoming more and more serious. This resistance may be caused by a variety of factors, including genetic mutations, signaling pathway remodeling, and changes in the tumor microenvironment. In order to overcome drug resistance, the combination of lenvatinib and other therapeutic strategies has gradually become a research hotspot, and it is worth noting that the combination of lenvatinib and immune checkpoint inhibitors (ICIs) has shown a good application prospect. This combination not only enhances the anti-tumor immune response but also helps improve therapeutic efficacy. However, combination therapy also faces challenges regarding safety and tolerability. Therefore, studying the mechanisms of resistance and identifying relevant biomarkers is particularly important, as it aids in early diagnosis and personalized treatment. This article reviews the mechanisms of lenvatinib in treating liver cancer, the mechanisms and efficacy of its combination with immune checkpoint inhibitors, the causes of resistance, the exploration of biomarkers, and other novel combination therapy strategies for lenvatinib. We hope to provide insights into the use and research of lenvatinib in clinical and scientific settings, offering new strategies for the treatment of liver cancer.
Collapse
Affiliation(s)
- Yuhang Chen
- Department of Integrative Oncology, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong-An Road, Shanghai, 200032, China
| | - Suoyi Dai
- Department of Integrative Oncology, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong-An Road, Shanghai, 200032, China
| | - Chien-Shan Cheng
- Department of Integrative Oncology, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong-An Road, Shanghai, 200032, China.
| | - Lianyu Chen
- Department of Integrative Oncology, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong-An Road, Shanghai, 200032, China.
| |
Collapse
|
11
|
Liu Y, Gu X, Xuan M, Lou N, Fu L, Li J, Xue C. Notch signaling in digestive system cancers: Roles and therapeutic prospects. Cell Signal 2024; 124:111476. [PMID: 39428027 DOI: 10.1016/j.cellsig.2024.111476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/20/2024] [Accepted: 10/17/2024] [Indexed: 10/22/2024]
Abstract
Digestive system cancers rank among the most prevalent malignant tumors, maintaining persistently high incidence and mortality rates. Notch signaling activity, often aberrant in esophageal, gastric, hepatic, pancreatic, and colorectal cancers, plays a pivotal role in the initiation, progression, and therapy resistance of these malignancies. As a highly conserved pathway, Notch signaling is integral to cell differentiation, survival, proliferation, stem cell renewal, development, and morphogenesis. Its dysregulation has been increasingly linked to various diseases, particularly digestive system cancers. In these malignancies, altered Notch signaling influences multiple biological processes, including cell proliferation, invasion, cell cycle progression, immune evasion, drug resistance, and stemness maintenance. Understanding the mechanisms of Notch signaling in digestive system cancers is essential for the development of novel targeted therapies. Numerous Notch pathway-targeting drugs are currently in preclinical studies, demonstrating promising efficacy both as monotherapies and in combination with conventional anti-cancer treatments. This review summarizes recent high-quality findings on the involvement of Notch signaling in digestive system cancers, focusing on the expression changes and pathological mechanisms of its dysregulated components. Special emphasis is placed on the potential of translating Notch-targeted approaches into therapeutic strategies, which hold promise for overcoming the limitations of existing treatments and improving the poor prognosis associated with these cancers.
Collapse
Affiliation(s)
- Yingru Liu
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Xinyu Gu
- Department of Oncology, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471000, China
| | - Mengjuan Xuan
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Na Lou
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Leiya Fu
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Juan Li
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Chen Xue
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| |
Collapse
|
12
|
Li J, Cheng X, Huang D, Cui R. The regulatory role of mitotic catastrophe in hepatocellular carcinoma drug resistance mechanisms and its therapeutic potential. Biomed Pharmacother 2024; 180:117598. [PMID: 39461015 DOI: 10.1016/j.biopha.2024.117598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 10/29/2024] Open
Abstract
This review focuses on the role and underlying mechanisms of mitotic catastrophe (MC) in the regulation of drug resistance in hepatocellular carcinoma (HCC). HCC is one of the leading causes of cancer-related mortality worldwide, posing significant treatment challenges due to its high recurrence rates and drug resistance. Research suggests that MC, as a mechanism of cell death, plays a crucial role in enhancing the efficacy of HCC treatment by disrupting the replication and division mechanisms of tumor cells. The present review summarizes the molecular mechanisms of MC and its role in HCC drug resistance and explores the potential of combining MC with existing cancer therapies to improve treatment outcomes. Future research should focus on the in-depth elucidation of the molecular mechanisms of MC and its application in HCC therapy, providing new insights for the development of more effective treatments.
Collapse
Affiliation(s)
- Jianwang Li
- Department of Oncology, Xiangya School of Medicine Affiliated Haikou Hospital/Haikou People's Hospital, No.43, Renmin Avenue, Haikou, Hainan 570208, PR China.
| | - Xiaozhen Cheng
- Department of Oncology, Xiangya School of Medicine Affiliated Haikou Hospital/Haikou People's Hospital, No.43, Renmin Avenue, Haikou, Hainan 570208, PR China
| | - Denggao Huang
- Department of Central Laboratory, Xiangya School of Medicine Affiliated Haikou Hospital, No.43, Renmin Avenue, Haikou, Hainan 570208, PR China
| | - Ronghua Cui
- Department of Oncology, Xiangya School of Medicine Affiliated Haikou Hospital/Haikou People's Hospital, No.43, Renmin Avenue, Haikou, Hainan 570208, PR China
| |
Collapse
|
13
|
Zhang K, Zhu Z, Zhou J, Shi M, Wang N, Yu F, Xu L. Disulfidptosis-related gene expression reflects the prognosis of drug-resistant cancer patients and inhibition of MYH9 reverses sorafenib resistance. Transl Oncol 2024; 49:102091. [PMID: 39146597 PMCID: PMC11375144 DOI: 10.1016/j.tranon.2024.102091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 05/03/2024] [Accepted: 08/11/2024] [Indexed: 08/17/2024] Open
Abstract
The onset of drug resistance in advanced cancer patients markedly diminishes their prognosis. Recently, disulfidptosis, a novel form of cell death, has been identified, triggered by excessive disulfide formation leading to cell shrinkage and F-actin contraction. Previous studies have identified 15 essential genes (FLNA, FLNB, MYH9, TLN1, ACTB, MYL6, MYH10, CAPZB, DSTN, IQGAP1, ACTN4, PDLIM1, CD2AP, INF2, SLC7A11) associated with disulfidptosis. This study sourced pan-cancer mRNA expression data from Xena to thoroughly evaluate the molecular and clinical characteristics of disulfidptosis-related genes. Through unsupervised clustering, mRNA expression data identified the expression levels of disulfidptosis-related genes and potential clusters related to this form of cell death. Kaplan-Meier survival curves illustrated the correlation between different clusters and overall survival. The findings reveal that high expression of disulfidptosis-related genes is linked to poor survival in liver cancer. The GDSC database was utilized to analyze the relationship between disulfidptosis-related genes and the AUC of 198 drugs. The results demonstrate that 12 disulfidptosis-related genes influence sorafenib resistance, as revealed by the intersection of differential genes related to sorafenib resistance from the GSE109211 dataset. Among them, the MYH9 gene was found to play a crucial role in both. Finally, experimental evidence confirmed that MYH9 mitigates sorafenib resistance in hepatocellular carcinoma through disulfidptosis-like changes. This study identifies disulfidptosis as a promising avenue for enhancing the sensitivity of tumor cells to drugs, offering new therapeutic perspectives for future research on disulfidptosis and drug resistance in cancer patients.
Collapse
Affiliation(s)
- Kangnan Zhang
- Department of Gastroenterology, Shanghai Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200336, China
| | - Zhenhua Zhu
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200001, China
| | - Jingyi Zhou
- Department of Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Min Shi
- Department of Gastroenterology, Shanghai Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200336, China
| | - Na Wang
- Department of Gastroenterology, Shanghai Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200336, China.
| | - Fudong Yu
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Public Health School, Fudan University, Shanghai, 200030, China.
| | - Ling Xu
- Department of Gastroenterology, Shanghai Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200336, China.
| |
Collapse
|
14
|
Jia Q, Zhu Y, Yao H, Yin Y, Duan Z, Zheng J, Ma D, Yang M, Yang J, Zhang J, Liu D, Hua R, Huo Y, Fu X, Sun Y, Liu W. Oncogenic GALNT5 confers FOLFIRINOX resistance via activating the MYH9/ NOTCH/ DDR axis in pancreatic ductal adenocarcinoma. Cell Death Dis 2024; 15:767. [PMID: 39433745 PMCID: PMC11493973 DOI: 10.1038/s41419-024-07110-w] [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: 10/16/2023] [Revised: 09/16/2024] [Accepted: 09/24/2024] [Indexed: 10/23/2024]
Abstract
Chemotherapy resistance has been a great challenge in pancreatic ductal adenocarcinoma(PDAC) treatments. Current first-line chemotherapy regimens for PDAC include gemcitabine-based regimens such as AG regimen (albumin paclitaxel and gemcitabine), fluorouracil-based regiments such as FOLFIRINOX regimen ((5-fluorouracil5-FU), oxaliplatin, Irinotecan) and platinum-based regimens for patients with BRCA mutations. large amounts of work have been done on exploring the mechanism underlying resistance of gemcitabine-based and platinum-based regimens, while little research has been achieved on the mechanism of FOLFIRINOX regimens resistance. Hence, we identified Polypeptide N-Acetylgalactosaminyltransferase 5, (GALNT5) as a vital regulator and a potential therapeutic target in FOLFIRINOX regimens resistance. Colony formation assays and flow cytometry assays were performed to explore the roles of GALNT5 in cell proliferation and apoptosis in PDAC treated with FOLFIRINOX. IC50 alterations were calculated in GALNT5 knockdown and overexpressed cell lines. RNA-seq followed by GSEA (gene set enrichment analysis) was displayed to explore the potential mechanism. WB (western blotting), real-time PCR, and IF (immunofluorescence) were performed to validate relative pathways. The mouse orthotopic xenograft PDAC model was established to examine GALNT5 functions in vivo. GALNT5 was highly expressed in PDAC tissues and predicted poor prognosis in PDAC. Upregulation of GALNT5 in PDAC cells conferred FOLFIRINOX resistance on PDAC by inhibiting DNA damage. Moreover, GALNT5 interacted with MYH9, thus participating in the activation of the NOTCH pathways, resulting in hampering FOI-induced DNA damage. Functions of GALNT5 promoting FOLFIRINOX resistance were validated in vivo. In this study, we found that aberrantly overexpressed GALNT5 in PDAC took part in the activation of the NOTCH pathway by interacting with MYH9, thus inhibiting the DDR to achieve FOLFIRINOX resistance and causing poor prognosis. We identified GALNT5 as a potential therapeutic target for PDAC patients resistant to FOLFIRINOX chemotherapy.
Collapse
MESH Headings
- Humans
- Carcinoma, Pancreatic Ductal/drug therapy
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/pathology
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- N-Acetylgalactosaminyltransferases/metabolism
- N-Acetylgalactosaminyltransferases/genetics
- Oxaliplatin/pharmacology
- Oxaliplatin/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Animals
- Fluorouracil/pharmacology
- Fluorouracil/therapeutic use
- Leucovorin/pharmacology
- Leucovorin/therapeutic use
- Mice
- Cell Line, Tumor
- Polypeptide N-acetylgalactosaminyltransferase
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/metabolism
- Receptors, Notch/metabolism
- Irinotecan/pharmacology
- Irinotecan/therapeutic use
- Mice, Nude
- Cell Proliferation/drug effects
- Apoptosis/drug effects
- Signal Transduction/drug effects
- Gene Expression Regulation, Neoplastic/drug effects
- Female
- Xenograft Model Antitumor Assays
- Myosin Heavy Chains
Collapse
Affiliation(s)
- Qinyuan Jia
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P.R. China
| | - Yuheng Zhu
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P.R. China
| | - Hongfei Yao
- Department of Hepato-Biliary-Pancreatic Surgery, General Surgery, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, P.R. China
| | - Yifan Yin
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P.R. China
| | - Zonghao Duan
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P.R. China
| | - Jiahao Zheng
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, P.R. China
| | - Ding Ma
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P.R. China
| | - Minwei Yang
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P.R. China
| | - Jianyu Yang
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P.R. China
| | - Junfeng Zhang
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P.R. China
| | - Dejun Liu
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P.R. China
| | - Rong Hua
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P.R. China
| | - Yanmiao Huo
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P.R. China.
| | - Xueliang Fu
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P.R. China.
| | - Yongwei Sun
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P.R. China.
| | - Wei Liu
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P.R. China.
| |
Collapse
|
15
|
Li W, Yang LJ, Xiong YY, Li ZS, Li X, Wen Y. 4,5-Dimethoxycanthin-6-one Inhibits Glioblastoma Stem Cell and Tumor Growth by Inhibiting TSPAN1 Interaction with TM4SF1. Neurochem Res 2024; 49:2897-2909. [PMID: 39060768 DOI: 10.1007/s11064-024-04211-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/05/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024]
Abstract
Glioblastoma stem cells (GSCs) have been implicated in the self-renewal and treatment resistance of glioblastoma (GBM). Our previous study found that 4,5-dimethoxycanthin-6-one has the potential to inhibit GBM cell proliferation. This current study aims to elucidate the molecular mechanism underlying the effects of 4,5-dimethoxycanthin-6-one in GBM development. The effect of 4,5-dimethoxycanthin-6-one on GSC formation and differentiation was explored in human GBM cell lines U251 and U87. Subsequently, 4,5-dimethoxycanthin-6-one binding to tetraspanin 1 (TSPAN1) / transmembrane 4 L six family member 1 (TM4SF1) was analyzed by molecular simulation docking. Co-immunoprecipitation (Co-IP) and immunofluorescence (IF) were used to assess the interactions between TSPAN1 and TM4SF1 in GSCs. Cell proliferation was detected by cell counting kit-8 (CCK-8) and colony formation assay. To evaluate cell migration, invasion and apoptosis, we employed wound healing assay, transwell and flow cytometry, respectively. Furthermore, subcutaneous xenograft tumor models in nude mice were constructed to evaluate the impact of 4,5-dimethoxycanthin-6-one on GSCs in vivo by examining tumor growth and histological characteristics. 4,5-Dimethoxycanthin-6-one inhibited GSC formation and promoted stem cell differentiation in a concentration-dependent manner. Molecular docking models of 4,5-dimethoxycanthin-6-one with TM4SF1 and TSPAN1 were constructed. Then, the interaction between TSPAN1 and TM4SF1 in GSC was clarified. Moreover, 4,5-dimethoxycanthin-6-one significantly inhibited the expressions of TM4SF1 and TSPAN1 in vitro and in vivo. Overexpression of TSPAN1 partially reversed the inhibitory effects of 4,5-dimethoxycanthin-6-one on GSC formation, proliferation, migration and invasion. 4,5-Dimethoxycanthin-6-one inhibited GBM progression by inhibiting TSPAN1/TM4SF1 axis. 4,5-Dimethoxycanthin-6-one might be a novel and effective drug for the treatment of GBM.
Collapse
Affiliation(s)
- Wei Li
- Department of Neurosurgery, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Li-Jian Yang
- Department of Neurosurgery, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yuan-Yuan Xiong
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zeng-Shi Li
- Department of Neurosurgery, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xi Li
- Department of Neurosurgery, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yi Wen
- Department of Imaging, Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, Hunan, China.
- Department of Imaging, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China.
| |
Collapse
|
16
|
Ye G, Ye M, Jin X. Roles of clinical application of lenvatinib and its resistance mechanism in advanced hepatocellular carcinoma (Review). Am J Cancer Res 2024; 14:4113-4171. [PMID: 39417171 PMCID: PMC11477829 DOI: 10.62347/ujvp4361] [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: 05/09/2024] [Accepted: 09/03/2024] [Indexed: 10/19/2024] Open
Abstract
Lenvatinib (LEN) is a multi-target TKI, which plays a pivotal role in the treatment of advanced hepatocellular carcinoma (HCC). The inevitable occurrence of drug resistance still prevents curative potential and is deleterious for the prognosis, and a growing body of studies is accumulating, which have devoted themselves to unveiling its underlying resistance mechanism and made some progress. The dysregulation of crucial signaling pathways, non-coding RNA and RNA modifications were proven to be associated with LEN resistance. A range of drugs were found to influence LEN therapeutic efficacy. In addition, the superiority of LEN combination therapy has been shown to potentially overcome the limitations of LEN monotherapy in a series of research, and a range of promising indicators for predicting treatment response and prognosis have been discovered in recent years. In this review, we summarize the latest developments in LEN resistance, the efficacy and safety of LEN combination therapy as well as associated indicators, which may provide new insight into its resistance as well as ideas in the treatment of advanced HCC.
Collapse
Affiliation(s)
- Ganghui Ye
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo UniversityNingbo 315211, Zhejiang, P. R. China
- Department of Oncology, The First Hospital of Ningbo UniversityNingbo 315020, Zhejiang, P. R. China
- Department of Radiation Oncology, Taizhou Central Hospital (Taizhou University Hospital)Taizhou 318000, Zhejiang, P. R. China
| | - Meng Ye
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo UniversityNingbo 315211, Zhejiang, P. R. China
- Department of Oncology, The First Hospital of Ningbo UniversityNingbo 315020, Zhejiang, P. R. China
| | - Xiaofeng Jin
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo UniversityNingbo 315211, Zhejiang, P. R. China
- Department of Oncology, The First Hospital of Ningbo UniversityNingbo 315020, Zhejiang, P. R. China
| |
Collapse
|
17
|
Li Y, Pan Y, Yang X, Wang Y, Liu B, Zhang Y, Gao X, Wang Y, Zhou H, Li F. Unveiling the enigmatic role of MYH9 in tumor biology: a comprehensive review. Cell Commun Signal 2024; 22:417. [PMID: 39192336 PMCID: PMC11351104 DOI: 10.1186/s12964-024-01781-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 08/06/2024] [Indexed: 08/29/2024] Open
Abstract
Non-muscle myosin heavy chain IIA (MYH9), a member of the non-muscle myosin II (NM II) family, is widely expressed in cells. The interaction of MYH9 with actin in the cytoplasm can hydrolyze ATP, completing the conversion of chemical energy to mechanical motion. MYH9 participates in various cellular processes, such as cell adhesion, migration, movement, and even signal transduction. Mutations in MYH9 are often associated with autosomal dominant platelet disorders and kidney diseases. Over the past decade, tumor-related research has gradually revealed a close relationship between MYH9 and the occurrence and development of tumors. This article provides a review of the research progress on the role of MYH9 in cancer regulation. We also discussed the anti-cancer effects of MYH9 under special circumstances, as well as its regulation of T cell function. In addition, given the importance of MYH9 as a key hub in oncogenic signal transduction, we summarize the current therapeutic strategies targeting MYH9 as well as the ongoing challenges.
Collapse
Affiliation(s)
- Yunkuo Li
- Department of Urology Il, The First Hospital of Jilin University, Changchun, 130021, China
| | - Yujie Pan
- Department of Urology Il, The First Hospital of Jilin University, Changchun, 130021, China
| | - Xiangzhe Yang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Yuxiong Wang
- Department of Urology Il, The First Hospital of Jilin University, Changchun, 130021, China
| | - Bin Liu
- Department of Urology Il, The First Hospital of Jilin University, Changchun, 130021, China
| | - Yanghe Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Xin Gao
- Department of Urology Il, The First Hospital of Jilin University, Changchun, 130021, China
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Honglan Zhou
- Department of Urology Il, The First Hospital of Jilin University, Changchun, 130021, China
| | - Faping Li
- Department of Urology Il, The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
18
|
Gou Z, Zhang D, Cao H, Li Y, Li Y, Zhao Z, Wang Y, Wang Y, Zhou H. Exploring the nexus between MYH9 and tumors: novel insights and new therapeutic opportunities. Front Cell Dev Biol 2024; 12:1421763. [PMID: 39149512 PMCID: PMC11325155 DOI: 10.3389/fcell.2024.1421763] [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: 04/23/2024] [Accepted: 07/19/2024] [Indexed: 08/17/2024] Open
Abstract
The myosin heavy chain 9 (MYH9) gene, located on human chromosome 22, encodes non-muscle myosin heavy chain IIA (NM IIA). This protein is essential to various cellular events, such as generating intracellular chemomechanical force and facilitating the movement of the actin cytoskeleton. Mutations associated with thrombocytopenia in autosomal dominant diseases first highlighted the significance of the MYH9 gene. In recent years, numerous studies have demonstrated the pivotal roles of MYH9 in various cancers. However, its effects on cancer are intricate and not fully comprehended. Furthermore, the elevated expression of MYH9 in certain malignancies suggests its potential as a target for tumor therapy. Nonetheless, there is a paucity of literature summarizing MYH9's role in tumors and the therapeutic strategies centered on it, necessitating a systematic analysis. This paper comprehensively reviews and analyzes the pertinent literature in this domain, elucidating the fundamental structural characteristics, biological functions, and the nexus between MYH9 and tumors. The mechanisms through which MYH9 contributes to tumor development and its multifaceted roles in the tumorigenic process are also explored. Additionally, we discuss the relationship between MYH9-related diseases (MYH9-RD) and tumors and also summarize tumor therapeutic approaches targeting MYH9. The potential clinical applications of studying the MYH9 gene include improving early diagnosis, clinical staging, and prognosis of tumors. This paper is anticipated to provide novel insights for tumor therapy.
Collapse
Affiliation(s)
- Zixuan Gou
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Difei Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Hongliang Cao
- Department of Urology II, The First Hospital of Jilin University, Changchun, China
| | - Yao Li
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Yunkuo Li
- Department of Urology II, The First Hospital of Jilin University, Changchun, China
| | - Zijian Zhao
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Ye Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Honglan Zhou
- Department of Urology II, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
19
|
Fan FM, Fleishman JS, Chen J, Chen ZS, Dong HH. New insights into the mechanism of resistance to lenvatinib and strategies for lenvatinib sensitization in hepatocellular carcinoma. Drug Discov Today 2024; 29:104069. [PMID: 38936692 DOI: 10.1016/j.drudis.2024.104069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/04/2024] [Accepted: 06/21/2024] [Indexed: 06/29/2024]
Abstract
Lenvatinib is a multikinase inhibitor that suppresses vascular endothelial growth factor receptor (VEGFR), fibroblast growth factor receptor (FGFR), platelet-derived growth factor receptor α (PDGFRα), as well as the proto-oncogenes RET and KIT. Lenvatinib has been approved by the US Food and Drug Administration (FDA) for the first-line treatment of hepatocellular carcinoma (HCC) due to its superior efficacy when compared to sorafenib. Unfortunately, the development of drug resistance to lenvatinib is becoming increasingly common. Thus, there is an urgent need to identify the factors that lead to drug resistance and ways to mitigate it. We summarize the molecular mechanisms that lead to lenvatinib resistance (LR) in HCC, which involve programmed cell death (PCD), translocation processes, and changes in the tumor microenvironment (TME), and provide strategies to reverse resistance.
Collapse
Affiliation(s)
- Fei-Mu Fan
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Wuhan 430000, China
| | - Joshua S Fleishman
- College of Pharmacy and Health Sciences, St John's University, Queens, NY 11439, USA
| | - Jin Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Wuhan 430000, China.
| | - Zhe-Sheng Chen
- College of Pharmacy and Health Sciences, St John's University, Queens, NY 11439, USA.
| | - Han-Hua Dong
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Wuhan 430000, China.
| |
Collapse
|
20
|
Li K, Li T, Niu Y, Gao Y, Shi Y, He Y, Zhang X, Wang Y, Cao J, Hu X, Chen M, Shi R. Decreased NMIIA heavy chain phosphorylation at S1943 promotes mitoxantrone resistance by upregulating BCRP and N-cadherin expression in breast cancer cells. Biochem Cell Biol 2024; 102:213-225. [PMID: 38190650 DOI: 10.1139/bcb-2023-0232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2024] Open
Abstract
Mitoxantrone (MX) is an effective treatment for breast cancer; however, high efflux of MX that is accomplished by breast cancer resistance protein (BCRP) leads to acquired multidrug resistance (MDR), reducing MX's therapeutic efficacy in breast cancer. Non-muscle myosin IIA (NMIIA) and its heavy phosphorylation at S1943 have been revealed to play key roles in tumor metastasis and progression, including in breast cancer; however, their molecular function in BCRP-mediated MDR in breast cancer remains unknown. In this study, we revealed that the expression of NMIIA heavy chain phosphorylation at S1943 was downregulated in BCRP-overexpressing breast cancer MCF-7/MX cells, and stable expression of NMIIA-S1943A mutant increased BCRP expression and promoted the resistance of MCF-7/MX cells to MX. Meanwhile, NMIIA S1943 phosphorylation induced by epidermal growth factor (EGF) was accompanied by the downregulation of BCRP in MCF-7/MX cells. Furthermore, stable expression of NMIIA-S1943A in MCF-7/MX cells resulted in upregulation of N-cadherin and the accumulation of β-catenin on the cell surface, which inhibited the nucleus translocation of β-catenin and Wnt/β-catenin-based proliferative signaling. EGF stimulation of MCF-7/MX cells showed the downregulation of N-cadherin and β-catenin. Our results suggest that decreased NMIIA heavy phosphorylation at S1943 increases BCRP expression and promotes MX resistance in breast cancer cells via upregulating N-cadherin expression.
Collapse
Affiliation(s)
- Kemin Li
- Department of Pharmacology, Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Tian Li
- Department of Pharmacology, Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Yanan Niu
- Department of Pharmacology, Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Yu Gao
- Department of Pharmacology, Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Yifan Shi
- Department of Pharmacology, Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Yifan He
- Department of Pharmacology, Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Xuanping Zhang
- Department of Pharmacology, Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Yan Wang
- Department of Pharmacology, Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Jing Cao
- Department of Critical Care Medicine, the First Hospital of Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Xiaoling Hu
- Department of Pharmacology, Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Min Chen
- Department of Pharmacology, Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Ruizan Shi
- Department of Pharmacology, Shanxi Medical University, Taiyuan 030001, People's Republic of China
| |
Collapse
|
21
|
Jin Y, Eum DY, Lee C, Park SY, Shim JW, Choi YJ, Choi SH, Kim JG, Heo K, Park SJ. Breast cancer malignancy is governed by regulation of the macroH2A2/TM4SF1 axis, the AKT/NF-κB pathway, and elevated MMP13 expression. Mol Carcinog 2024; 63:714-727. [PMID: 38251858 DOI: 10.1002/mc.23683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/21/2023] [Accepted: 01/08/2024] [Indexed: 01/23/2024]
Abstract
The histone variant, macroH2A (mH2A) influences gene expression through epigenetic regulation. Tumor suppressive function of mH2A isoforms has been reported in various cancer types, but few studies have investigated the functional role of mH2A2 in breast cancer pathophysiology. This study aimed to determine the significance of mH2A2 in breast cancer development and progression by exploring its downstream regulatory mechanisms. Knockdown of mH2A2 facilitated the migration and invasion of breast cancer cells, whereas its overexpression exhibited the opposite effect. In vivo experiments revealed that augmenting mH2A2 expression reduced tumor growth and lung metastasis. Microarray analysis showed that TM4SF1 emerged as a likely target linked to mH2A2 owing to its significant suppression in breast cancer cell lines where mH2A2 was overexpressed among the genes that exhibited over twofold upregulation upon mH2A2 knockdown. Suppressing TM4SF1 reduced the migration, invasion, tumor growth, and metastasis of breast cancer cells in vitro and in vivo. TM4SF1 depletion reversed the increased aggressiveness triggered by mH2A2 knockdown, suggesting a close interplay between mH2A2 and TM4SF1. Our findings also highlight the role of the mH2A2/TM4SF1 axis in activating the AKT/NF-κB pathway. Consequently, activated NF-κB signaling leads to increased expression and secretion of MMP13, a potent promoter of metastasis. In summary, we propose that the orchestrated regulation of the mH2A2/TM4SF1 axis in conjunction with the AKT/NF-κB pathway and the subsequent elevation in MMP13 expression constitute pivotal factors governing the malignancy of breast cancer.
Collapse
Affiliation(s)
- Yunho Jin
- Research Center, Dongnam Institute of Radiological & Medical Sciences (DIRAMS), Busan, Republic of Korea
| | - Da-Young Eum
- Research Center, Dongnam Institute of Radiological & Medical Sciences (DIRAMS), Busan, Republic of Korea
| | - Chaeyoung Lee
- Research Center, Dongnam Institute of Radiological & Medical Sciences (DIRAMS), Busan, Republic of Korea
| | - Soon Yong Park
- Research Center, Dongnam Institute of Radiological & Medical Sciences (DIRAMS), Busan, Republic of Korea
| | - Jae Woong Shim
- Research Center, Dongnam Institute of Radiological & Medical Sciences (DIRAMS), Busan, Republic of Korea
| | - Yoo Jin Choi
- Research Center, Dongnam Institute of Radiological & Medical Sciences (DIRAMS), Busan, Republic of Korea
| | - Si Ho Choi
- Research Center, Dongnam Institute of Radiological & Medical Sciences (DIRAMS), Busan, Republic of Korea
| | - Joong-Gook Kim
- Research Center, Dongnam Institute of Radiological & Medical Sciences (DIRAMS), Busan, Republic of Korea
| | - Kyu Heo
- Research Center, Dongnam Institute of Radiological & Medical Sciences (DIRAMS), Busan, Republic of Korea
| | - Seong-Joon Park
- Research Center, Dongnam Institute of Radiological & Medical Sciences (DIRAMS), Busan, Republic of Korea
| |
Collapse
|
22
|
Wang X, Wang Y, Liu Z, Zhao H, Yao GD, Liu Q, Song SJ. New daphnane diterpenoidal 1,3,4-oxdiazole derivatives as potential anti-hepatoma agents: Synthesis, biological evaluation and molecular modeling studies. Bioorg Chem 2024; 145:107208. [PMID: 38354501 DOI: 10.1016/j.bioorg.2024.107208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 02/03/2024] [Accepted: 02/11/2024] [Indexed: 02/16/2024]
Abstract
Hepatocellular carcinoma (HCC) is a major challenge for human healthy. Daphnane-type diterpenes have attracted increasingly attention due to remarkable pharmaceutical potential including anti-HCC activity. To further develop this class of compounds as inhibitors of HCC, the daphnane diterpenoids 12-O-debenzoyl-Yuanhuacine (YHC) and 12-hydroxydaphnetoxin (YHE) were prepared by a standard chemical transformation from dried flower buds of the Daphne genkwa plant. Subsequently, 22 daphnane diterpenoidal 1,3,4-oxdiazole derivatives were rationally designed and synthesized based on YHC and YHE. The assessment of the target compound's anti-hepatocellular carcinoma activity revealed that YHC1 exhibited comparable activity to sorafenib in the Hep3B cell line, while demonstrating higher selectivity. The mechanistic investigation demonstrates that compound YHC1 induces cell cycle arrest at the G0/G1 phase, cellular senescence, apoptosis, and elevates cellular reactive oxygen species levels. Moreover, molecular docking and CETSA results confirm the interaction between YHC1 and YAP1 as well as TEAD1. Co-IP experiments further validated that YHC1 can effectively inhibit the binding of YAP1 and TEAD1. In conclusion, YHC1 selectively targets YAP1 and TEAD1, exhibiting its anti-hepatocellular carcinoma effects through the inhibition of their interaction.
Collapse
Affiliation(s)
- Xinyi Wang
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province, PR China; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province, PR China; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China
| | - Yujue Wang
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province, PR China; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province, PR China; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China
| | - Zijian Liu
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province, PR China; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province, PR China; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China
| | - Hongwei Zhao
- Jilin Yizheng Pharmaceutical Group Co., Ltd., Jilin Province, Siping 136001, PR China
| | - Guo-Dong Yao
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province, PR China; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province, PR China; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China.
| | - Qingbo Liu
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province, PR China; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province, PR China; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China; Jilin Yizheng Pharmaceutical Group Co., Ltd., Jilin Province, Siping 136001, PR China.
| | - Shao-Jiang Song
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province, PR China; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province, PR China; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China.
| |
Collapse
|
23
|
Zhang Y, Cui J, Cang Z, Pei J, Zhang X, Song B, Fan X, Ma X, Li Y. Hair follicle stem cells promote epidermal regeneration under expanded condition. Front Physiol 2024; 15:1306011. [PMID: 38455843 PMCID: PMC10917960 DOI: 10.3389/fphys.2024.1306011] [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: 10/03/2023] [Accepted: 02/09/2024] [Indexed: 03/09/2024] Open
Abstract
Skin soft tissue expansion is the process of obtaining excess skin mixed with skin development, wound healing, and mechanical stretching. Previous studies have reported that tissue expansion significantly induces epidermal proliferation throughout the skin. However, the mechanisms underlying epidermal regeneration during skin soft tissue expansion are yet to be clarified. Hair follicle stem cells (HFSCs) have been recognized as a promising approach for epidermal regeneration. This study examines HFSC-related epidermal regeneration mechanisms under expanded condition and proposes a potential method for its cellular and molecular regulation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xing Fan
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Xianjie Ma
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Yang Li
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| |
Collapse
|