1
|
Pi J, Wang Y, Zhao Y, Yang J. FBXL18 promotes endometrial carcinoma progression via destabilizing DUSP16 and thus activating JNK signaling pathway. Cancer Cell Int 2025; 25:180. [PMID: 40382593 DOI: 10.1186/s12935-025-03808-9] [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: 07/24/2024] [Accepted: 05/04/2025] [Indexed: 05/20/2025] Open
Abstract
OBJECTIVE The therapeutic options for patients with advanced endometrial carcinoma (EC) were still limited and the prognosis remained unfavorable. F-box and leucine-rich repeat protein 18 (FBXL18), belonging to the F-box protein family, was frequently altered in human cancer, while its functional role and underlying mechanisms in EC were largely unexplored. METHODS The expression of FBXL18 in EC tissues and cells were explored using data mining strategies and further experiments. Multiple in vitro assays, including CCK-8, colony formation, wound healing, and Transwell invasion assays, were performed to assess the function of FBXL18 on cell proliferation, migration, and invasion. Bioinformatic analyses, western blot, qRT-PCR, Co-immunoprecipitation and ubiquitination assays were employed to identify the downstream pathway and direct substrate of FBXL18. RESULTS FBXL18 was highly expressed in EC tissues and cell lines, and EC patients with high FBXL18 expression had poor clinical outcome. Loss- and gain-of-function assays showed that silencing FBXL18 suppressed EC cell proliferation, migration, and invasion, while overexpressing FBXL18 caused the opposite effects. Mechanistically, FBXL18 could physically interacted with DUSP16, a dual specificity phosphatase, leading to its ubiquitination and degradation, and thus activating JNK signaling pathway. Upregulation of DUSP16 in EC cells alleviated FBXL18 overexpression-induced activation of JNK signaling pathway, and reversed FBXL18 overexpression-mediated enhanced cell capacities of proliferation, migration, and invasion. CONCLUSION In summary, our study had showcased the elevated expression, prognostic prediction performance, and the malignant tumor-promoting role of FBXL18 in EC. The novel mechanisms underlying this phenotype are that FBXL18 promotes the ubiquitination and degradation of DUSP16, and thus activates JNK/c-JUN signaling to facilitate EC progression.
Collapse
Affiliation(s)
- Jie Pi
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yong Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yuzi Zhao
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jing Yang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
2
|
Enomoto A, Fukasawa T. JNK signaling dominance in hyperthermia. Cell Stress Chaperones 2025; 30:100080. [PMID: 40339685 DOI: 10.1016/j.cstres.2025.100080] [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: 03/13/2025] [Revised: 04/25/2025] [Accepted: 04/29/2025] [Indexed: 05/10/2025] Open
Abstract
Hyperthermia is a promising anticancer treatment that induces heat stress, stimulating various signal transduction pathways to maintain cellular homeostasis. Mitogen-activated protein kinases (MAPKs) link various extracellular stimuli with cytoplasmic and nuclear mediators through a three-tiered cascade of kinases, including MAPKs, MAP2Ks, and MAP3Ks. In mammals, three major groups of MAPKs have been characterized: extracellular signal-regulated protein kinases (ERK), p38 MAPKs, and c-Jun NH2-terminal kinases (JNK). Each group of MAPKs is heat-activated and exhibits distinct biological functions. However, the differences and advantages of the regulation of each MAPK with temperature changes remain unknown. Our results demonstrated that JNK was activated in a temperature-dependent manner, with degradation of the JNK phosphatases despite transient phosphorylation of ERK with induction of the ERK phosphatases. This brief insight deepens our current understanding of the deregulation of the ERK and JNK cascades in hyperthermia.
Collapse
Affiliation(s)
- Atsushi Enomoto
- Laboratory of Molecular Radiology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan.
| | - Takemichi Fukasawa
- Department of Dermatology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| |
Collapse
|
3
|
Xing L, Wu S, Xue S, Li X. A Novel Neutrophil Extracellular Trap Signature Predicts Patient Chemotherapy Resistance and Prognosis in Lung Adenocarcinoma. Mol Biotechnol 2025; 67:1939-1957. [PMID: 38734842 DOI: 10.1007/s12033-024-01170-1] [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: 01/09/2024] [Accepted: 04/02/2024] [Indexed: 05/13/2024]
Abstract
Chemoresistance is a key obstacle in the long-term survival of patients with locally and advanced lung adenocarcinoma (LUAD). This study used bioinformatic analysis to reveal the chemoresistance of gene-neutrophil extracellular traps (NETs) associated with LUAD. RNA sequencing data and LUAD expression patterns were obtained from the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases, respectively. The GeneCards database was used to identify NETosis-related genes (NRGs). To identify hub genes with significant and consistent expression, differential analysis was performed using the TCGA-LUAD and GEO datasets. LUAD subtypes were determined based on these hub genes, followed by prognostic analysis. Immunological scoring and infiltration analysis were conducted using NETosis scores (N-scores) derived from the TCGA-LUAD dataset. A clinical prognostic model was established and analyzed, and its clinical applications explored. Twenty-two hub genes were identified, and consensus clustering was used to identify two subgroups based on their expression levels. The Kaplan-Meier (KM) curves demonstrated statistically significant differences in prognosis between the two LUAD subtypes. Based on the median score, patients were further divided into high and low N-score groups, and KM curves showed that the N-scores were more precise at predicting the prognosis of patients with LUAD for overall survival (OS). Immunological infiltration analysis revealed significant differences in the abundances of 10 immune cell infiltrates between the high and low N-score groups. Risk scores indicated significant differences in prognosis between the two extreme score groups. The risk scores for the prognostic model also indicated significant differences between the two groups. The results provide new insights into NETosis-related differentially expressed genes (NRDEGs) associated with chemotherapy resistance in patients with LUAD. The established prognostic model is promising and could help with clinical applications to evaluate patient survival and therapeutic efficiency.
Collapse
Affiliation(s)
- Long Xing
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450000, Henan, China
- Department of Oncology, Affiliated Hospital of Qingdao Binhai University, Qingdao, Shandong, China
| | - Shuangli Wu
- Department of Special Examination, Affiliated Hospital of Qingdao Binhai University, Qingdao, Shandong, China
| | - Shiyue Xue
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Xingya Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450000, Henan, China.
| |
Collapse
|
4
|
Liu P, Zhou S, Zhou Z, Jin Z, Chen W, Li Z, Xu J, Chen F, Li Y, Wen Y, Zhang S, Zhang C, Li B, Zhao J, Chen H. Discovery and antitumor evaluation of a mitochondria-targeting ruthenium complex for effective cancer therapy. Cancer Lett 2025; 616:217582. [PMID: 40021041 DOI: 10.1016/j.canlet.2025.217582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 03/03/2025]
Abstract
Ruthenium-based metallodrugs have garnered attention as a promising alternative for anticancer therapy, aiming to overcome chemoresistance and severe side effects linked to platinum-based drugs. However, ruthenium complexes tested in clinical trials to date have yielded unsatisfactory results. This study synthesized a positively charged ruthenium complex (Ru-2) that effectively penetrated cancer cells and exhibited superior cytotoxicity to cisplatin in vitro against cancer cell lines and organoids. Ru-2 selectively targeted mitochondria, disrupting their function by depolarizing mitochondrial membrane potential, elevating reactive oxygen species production, and impairing both oxidative phosphorylation and the tricarboxylic acid cycle. Furthermore, Ru-2 triggered endoplasmic reticulum (ER) stress and apoptosis. Integrative transcriptomic and proteomic analyses, performed using RNA sequencing and mass spectrometry, identified key molecular changes in cancer cells treated with Ru-2. For enhanced in vivo application, we developed a transferrin-based nanomedicine formulation, TF/Ru-2, incorporating Ru-2 into transferrin. In vivo studies demonstrated that both Ru-2 and TF/Ru-2 exhibited superior antitumor efficacy and improved biosafety compared to cisplatin. This study presents a novel ruthenium complex and a transferrin-based drug delivery platform with significant potential for future cancer therapies.
Collapse
Affiliation(s)
- Peng Liu
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Shangbo Zhou
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Zhijun Zhou
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Zihan Jin
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Wei Chen
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Zihang Li
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Jiaqi Xu
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Feng Chen
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - You Li
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Yingfei Wen
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Shiqiang Zhang
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Changhua Zhang
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China; Clinical Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Binbin Li
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China.
| | - Jing Zhao
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China.
| | - Hengxing Chen
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China; Clinical Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China.
| |
Collapse
|
5
|
Chen H, Na X, Hu P, Ma Q, Yu R. WWP1 mediates the ubiquitination and degradation of HIPK3 in bladder cancer cells. J Biol Chem 2025:108528. [PMID: 40280416 DOI: 10.1016/j.jbc.2025.108528] [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: 09/08/2024] [Revised: 03/20/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025] Open
Abstract
Protein homeostasis is primarily regulated by post-translational modifications (PTMs). HIPK3 has been recognized as a tumor suppressor across various cancers. However, the impact of PTMs on HIPK3 remains insufficiently explored. This study identified WWP1 as an E3 ubiquitin ligase targeting HIPK3, demonstrating that WWP1 downregulates HIPK3 protein levels by facilitating its ubiquitination. Mechanistically, WWP1 directly interacts with HIPK3, promoting K48-linked polyubiquitination at the K1187 site. The WWP1/HIPK3 axis modulates cancer cell chemosensitivity through the regulation of the JNK signaling pathway. Additionally, Myc was found to act as a transcription factor, enhancing WWP1 expression. These findings offer novel insights into the regulation of HIPK3 at the PTM level.
Collapse
Affiliation(s)
- Haichao Chen
- Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Xinyu Na
- Department of Urology, Zhenhai People's Hospital, Ningbo, China
| | - Pengcheng Hu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University
| | - Qi Ma
- Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Rui Yu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University.
| |
Collapse
|
6
|
Hou J, Yi J, Wang Y, Cui L, Xia W, Liang Z, Ye L, Wang Z, Gao S, Wang Z. Quantification of β-Elemene by GC-MS and Preliminary Evaluation of Its Relationship With Antitumor Efficacy in Cancer Patients. JOURNAL OF ANALYTICAL METHODS IN CHEMISTRY 2025; 2025:6694947. [PMID: 40201224 PMCID: PMC11972858 DOI: 10.1155/jamc/6694947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 02/24/2025] [Indexed: 04/10/2025]
Abstract
Objectives: To establish and validate a sensitive and robust gas chromatography-mass spectrometry (GC-MS) method for the quantification of β-elemene in human plasma and assess the correlation between antitumor effect and β-elemene concentration in vivo. Methods: The chromatographic column was HP-5 ms (30 m × 0.25 mm, 0.25 μm, Agilent, United States of America). The carrier gas was helium (purity > 99.5%). The flow rate was 1.0 mL/min and the total run time was 11.0 min. The plasma sample was pretreated with protein precipitation plus liquid-liquid extraction. Cancer patients were enrolled and their samples were collected for analysis. Results: Calibration range of β-elemene was 200.0-20,000.0 ng/mL, with correlation coefficients > 0.99. The intra- and interday precision and accuracy were less than 5.8% and within the range of -10.4%-6.6%. The exposure level of β-elemene in the responder group ranged from 278.13 to 11,886.27 ng/mL, with a median of 3568.91 ng/mL, while in the nonresponder group, the range was from 675.92 to 9716.52 ng/mL, with a median of 3351.94 ng/mL. No difference was found in the β-elemene exposure level between the two groups (p > 0.05). Conclusions: This method was effectively developed, validated, and utilized to quantify β-elemene in cancer patients. The initial findings indicated no significant relationship between therapeutic efficacy and the concentration of β-elemene.
Collapse
Affiliation(s)
- Juanjuan Hou
- College of Traditional Chinese Medicine, Yunnan University of Traditional Chinese Medicine, Kunming 650500, Yunnan, China
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Jia Yi
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Yan Wang
- Department of Oncology, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Lili Cui
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Wenwen Xia
- College of Traditional Chinese Medicine, Yunnan University of Traditional Chinese Medicine, Kunming 650500, Yunnan, China
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Zhengyan Liang
- College of Traditional Chinese Medicine, Yunnan University of Traditional Chinese Medicine, Kunming 650500, Yunnan, China
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Liya Ye
- College of Traditional Chinese Medicine, Yunnan University of Traditional Chinese Medicine, Kunming 650500, Yunnan, China
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Zhipeng Wang
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Shouhong Gao
- College of Traditional Chinese Medicine, Yunnan University of Traditional Chinese Medicine, Kunming 650500, Yunnan, China
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Zhan Wang
- Department of Oncology, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| |
Collapse
|
7
|
Jiang Y, Yang H, Ye Z, Huang Y, Li P, Jiang Z, Han S, Ma L. Multi-omic analyses reveal aberrant DNA methylation patterns and the associated biomarkers of nasopharyngeal carcinoma and its cancer stem cells. Sci Rep 2025; 15:9733. [PMID: 40118861 PMCID: PMC11928619 DOI: 10.1038/s41598-025-87038-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 01/15/2025] [Indexed: 03/24/2025] Open
Abstract
Aberrant DNA methylation is a hallmark of nasopharyngeal carcinoma (NPC) pathogenesis. The aberrant DNA methylation patterns in NPC, particularly in its cancer stem cells (CSCs), and their underlying significance require further elucidation. We integratively performed DNA methylome and transcriptome combined with single-nucleus RNA sequencing to investigate DNA methylation and gene expression patterns of NPC and CSCs. Unlike Epstein-Barr virus (EBV)-negative cells, NPC and CSCs harboring EBV displayed global DNA hypermethylation and they were more oncogenic and immunosuppressive. By correlating DNA methylation and gene expression profiles, we disclosed potential relationships between aberrant DNA methylation, tumorigenesis, metastasis, immunotherapy response, and radiotherapy resistance of NPC. After validating with datasets from GEO and TCGA, we identified aberrant DNA methylation-associated biomarkers including 9 NPC-specific diagnostic markers that had significantly higher DNA methylation levels in NPC than in normal tissues and 8 types of cancers, and 12 potential prognostic markers that were highly correlated to cell cycle dysregulation. Notably, 2 of these potential biomarkers highly expressed in CSCs were validated at the single-cell level. Our study not only identified new potential diagnostic and prognostic biomarkers but also provided new insight into aberrant DNA methylation-associated pathogenesis of NPC, which is beneficial for the development of precision diagnosis and treatment schemes.
Collapse
Affiliation(s)
- Yike Jiang
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Hongtian Yang
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
| | - Zilu Ye
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
- Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Yunchuanxiang Huang
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, Shenzhen, 518055, China
| | - Ping Li
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
| | - Ziyi Jiang
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
| | - Sanyang Han
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China.
- Tsinghua Shenzhen International Graduate School, Shenzhen, 518055, China.
| | - Lan Ma
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China.
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China.
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, Shenzhen, 518055, China.
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China.
- Tsinghua Shenzhen International Graduate School, Shenzhen, 518055, China.
| |
Collapse
|
8
|
Kim M, Hong WC, Kang HW, Kim JH, Lee D, Cheong JH, Jung HS, Kwon W, Jang JY, Kim HJ, Park JS. SLC5A3 depletion promotes apoptosis by inducing mitochondrial dysfunction and mitophagy in gemcitabine-resistant pancreatic cancer cells. Cell Death Dis 2025; 16:161. [PMID: 40055335 PMCID: PMC11889219 DOI: 10.1038/s41419-025-07476-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 02/07/2025] [Accepted: 02/24/2025] [Indexed: 05/13/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer with poor prognosis, largely due to the rapid development of chemoresistance in patients. Mitochondrial dynamics play a crucial role in cancer cell survival. Currently, the specific mechanisms underlying gemcitabine resistance in PDAC remain unknown. In this study, we identified the sodium/myo-inositol co-transporter solute carrier family 5 member 3 (SLC5A3) as a key modulator promoting chemoresistance in PDAC. SLC5A3 levels were significantly upregulated in gemcitabine-resistant PDAC cells, enhancing their cell survival by stabilizing the mitochondrial functions and inhibiting apoptosis. Mitochondrial analysis showed that SLC5A3 inhibition disrupted the mitochondrial dynamics, leading to increased reactive oxygen species production, mitochondrial fission, and impaired oxidative phosphorylation. Moreover, SLC5A3 inhibition activated the PTEN-induced kinase 1/Parkin-mediated mitophagy pathway, resulting in the excessive removal of damaged and healthy mitochondria, thereby depleting the mitochondrial reserves and sensitizing the cells to apoptosis. In vivo studies revealed that targeting SLC5A3 enhanced the efficacy of gemcitabine and significantly reduced the tumor growth. Collectively, these results suggest SLC5A3-mediated mitochondrial regulation as a promising therapeutic strategy to overcome gemcitabine resistance in PDAC.
Collapse
Affiliation(s)
- Minsoo Kim
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, College of Medicine, Seoul, Republic of Korea
| | - Woosol Chris Hong
- Department of Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyeon Woong Kang
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, College of Medicine, Seoul, Republic of Korea
| | - Ju Hyun Kim
- Department of Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Dongyong Lee
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jae-Ho Cheong
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, College of Medicine, Seoul, Republic of Korea
| | - Hye-Sol Jung
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Wooil Kwon
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jin-Young Jang
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyo Jung Kim
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea.
| | - Joon Seong Park
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
9
|
Zhou C, Wu K, Gu M, Yang Y, Tu J, Huang X. Reversal of chemotherapy resistance in gastric cancer with traditional Chinese medicine as sensitizer: potential mechanism of action. Front Oncol 2025; 15:1524182. [PMID: 40052129 PMCID: PMC11882405 DOI: 10.3389/fonc.2025.1524182] [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: 11/07/2024] [Accepted: 02/03/2025] [Indexed: 03/09/2025] Open
Abstract
Gastric cancer (GC) remains one of the most common types of cancer, ranking fifth among cancer-related deaths worldwide. Chemotherapy is an effective treatment for advanced GC. However, the development of chemotherapy resistance, which involves the malfunction of several signaling pathways and is the consequence of numerous variables interacting, seriously affects patient treatment and leads to poor clinical outcomes. Therefore, in order to treat GC, it is imperative to find novel medications that will increase chemotherapy sensitivity and reverse chemotherapy resistance. Traditional Chinese medicine (TCM) has been extensively researched as an adjuvant medication in recent years. It has been shown to have anticancer benefits and to be crucial in enhancing chemotherapy sensitivity and reducing chemotherapy resistance. Given this, the mechanism of treatment resistance in GC is summed up in this work. The theoretical foundation for TCM as a sensitizer in adjuvant treatment of GC is established by introducing the primary signal pathways and possible targets implicated in improving chemotherapy sensitivity and reversing chemotherapy resistance of GC by TCM and active ingredients.
Collapse
Affiliation(s)
| | | | | | | | | | - Xuan Huang
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese
Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
10
|
Wu X, Liu B, Deng SZ, Xiong T, Dai L, Cheng B. Disulfidptosis-related immune patterns predict prognosis and characterize the tumor microenvironment in oral squamous cell carcinoma. BMC Oral Health 2025; 25:180. [PMID: 39894803 PMCID: PMC11789412 DOI: 10.1186/s12903-024-05279-2] [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: 10/24/2023] [Accepted: 11/29/2024] [Indexed: 02/04/2025] Open
Abstract
BACKGROUND Establishing a prognostic risk model based on immunological and disulfidptosis signatures enables precise prognosis prediction of oral squamous cell carcinoma (OSCC). METHODS Differentially expressed immune and disulfidptosis genes were identified in OSCC and normal tissues. We examined the model's clinical applicability and its relationship to immune cell infiltration. Additionally, the risk score, ssGSEA, ESTIMATE, and CIBERSORT were used to evaluate the intrinsic molecular subtypes, immunological checkpoints, abundances of tumor-infiltrating immune cell types and proportions between the two risk groups. GO-KEGG and GSVA analyses were performed to identify enriched pathways. RESULTS We analyzed the correlation immune genes based on the 14 disulfidptosis-related genes, and found 379 disulfidptosis-related immune genes (DRIGs). After univariate Cox regression we obtained 30 DRIGs and least absolute shrinkage and selection operator (LASSO) regression to reduce the number of genes to 16. Finally we created a nine-DRIGs risk model, of which four were upregulated and five were downregulated. The analysis results showed that disulfidptosis was tightly related to immune cells, immunological-related pathways, the tumor microenvironment (TME), immune checkpoints, human leukocyte antigen (HLA), and tumor mutational burden (TMB). The nomogram, integrating the risk score and clinical factors, accurately predicted overall survival. CONCLUSIONS This novel risk model highlights the role of disulfidptosis-related immune genes in OSCC prognosis. With this model, we can more accurately predict the prognosis of patients with OSCC, as well as assess the potential effects of their TME and immunotherapy.
Collapse
Affiliation(s)
- Xuechen Wu
- Department of Stomatology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China
| | - Boxin Liu
- Department of Blood Transfusion, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, China
| | - Shi-Zhou Deng
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Tengteng Xiong
- Department of Stomatology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China
| | - Lin Dai
- Department of Stomatology, Wuhan No.1 Hospital, Wuhan, China.
| | - Bo Cheng
- Department of Stomatology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China.
| |
Collapse
|
11
|
Zheng S, Piao Y, Jung SN, Oh C, Lim MA, Nguyen Q, Shen S, Park SH, Cui S, Piao S, Kim YI, Kim JW, Won HR, Chang JW, Shan Y, Liu L, Koo BS. Gene Expression Alteration by Non-thermal Plasma-Activated Media Treatment in Radioresistant Head and Neck Squamous Cell Carcinoma. Clin Exp Otorhinolaryngol 2025; 18:73-87. [PMID: 39757757 PMCID: PMC11917201 DOI: 10.21053/ceo.2024.00238] [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: 08/14/2024] [Accepted: 01/05/2025] [Indexed: 01/07/2025] Open
Abstract
OBJECTIVES Head and neck squamous cell carcinoma (HNSCC) exhibits high recurrence rates, particularly in cases of radioresistant HNSCC (RR-HNSCC). Non-thermal plasma (NTP) therapy effectively suppresses the progression of HNSCC. However, the therapeutic mechanisms of NTP therapy in treating RR-HNSCC are not well understood. In this study, we explored the regulatory role of NTP in the RR-HNSCC signaling pathway and identified its signature genes. METHODS After constructing two RR-HNSCC cell lines, we prepared cell lysates from cells treated or not treated with NTP-activated media (NTPAM) and performed RNA sequencing to determine their mRNA expression profiles. Based on the RNA sequencing results, we identified differentially expressed genes (DEGs), followed by a bioinformatics analysis to identify candidate molecules potentially associated with NTPAM therapy for RR-HNSCC. RESULTS NTPAM reduced RR-HNSCC cell viability in vitro. RNA sequencing results indicated that NTPAM treatment activated the reactive oxygen species (ROS) pathway and induced ferroptosis in RR-HNSCC cell lines. Among the 1,924 genes correlated with radiation treatment, eight showed statistical significance in both the cell lines and The Cancer Genome Atlas (TCGA) cohort. Only five genes-ABCC3, DUSP16, PDGFB, RAF1, and THBS1-showed consistent results between the NTPAM data sequencing and TCGA data. LASSO regression analysis revealed that five genes were associated with cancer prognosis, with a hazard ratio of 2.26. In RR-HNSCC cells, NTPAM affected DUSP16, PDGFB, and THBS1 as activated markers within 6 hours, and this effect persisted for 12 hours. Furthermore, enrichment analysis indicated that these three DEGs were associated with the extracellular matrix, transforming growth factor-beta, phosphoinositide 3-kinase/protein kinase B, and mesenchymal-epithelial transition factor pathways. CONCLUSION NTPAM therapy exerts cytotoxic effects in RR-HNSCC cell lines by inducing specific ROS-mediated ferroptosis. DUSP16, PDGFB, and THBS1 were identified as crucial targets for reversing the radiation resistance induced by NTPAM therapy, providing insights into the mechanisms and clinical applications of NTPAM treatment in RR-HNSCC.
Collapse
Affiliation(s)
- Sicong Zheng
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
| | - Yudan Piao
- Dental Department, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Otolaryngology-Head and Neck Surgery, Chungnam National University College of Medicine, Daejeon, Korea
| | - Seung-Nam Jung
- Department of Otolaryngology-Head and Neck Surgery, Chungnam National University College of Medicine, Daejeon, Korea
| | - Chan Oh
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
| | - Mi Ae Lim
- Department of Otolaryngology-Head and Neck Surgery, Chungnam National University College of Medicine, Daejeon, Korea
| | - QuocKhanh Nguyen
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
| | - Shan Shen
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
| | - Se-Hee Park
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
| | - Shengzhe Cui
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
| | - Shuyu Piao
- Department of Otolaryngology-Head and Neck Surgery, Chungnam National University College of Medicine, Daejeon, Korea
| | - Young Il Kim
- Department of Radiation Oncology, Chungnam National University Sejong Hospital, Sejong, Korea
| | - Ji Won Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Chungnam National University Sejong Hospital, Sejong, Korea
| | - Ho-Ryun Won
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Otorhinolaryngology-Head and Neck Surgery, Chungnam National University Sejong Hospital, Sejong, Korea
| | - Jae Won Chang
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Otolaryngology-Head and Neck Surgery, Chungnam National University College of Medicine, Daejeon, Korea
| | - Yujuan Shan
- Department of Nutrition, Public Health and Management College, Wenzhou Medical University, Wenzhou, China
| | - Lihua Liu
- Department of Nutrition, Public Health and Management College, Wenzhou Medical University, Wenzhou, China
| | - Bon Seok Koo
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Otolaryngology-Head and Neck Surgery, Chungnam National University College of Medicine, Daejeon, Korea
| |
Collapse
|
12
|
He H, Zheng S, Jin S, Huang W, Wei E, Guan S, Yang C. Nucleotide metabolism-associated drug resistance gene NDUFA4L2 promotes colon cancer progression and 5-FU resistance. Sci Rep 2025; 15:570. [PMID: 39747340 PMCID: PMC11695588 DOI: 10.1038/s41598-024-84353-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025] Open
Abstract
Chemotherapy is an effective way to improve the prognosis of colorectal cancer patients, but patient resistance to chemotherapeutic agents is becoming a major obstacle to treatment. Nucleotide metabolism correlates with the progression of colorectal cancer and chemotherapy resistance, but the mechanisms involved need to be further investigated. We calculated the half-maximal inhibitory concentrations (IC50) of 5-Fluorouracil (5-FU) in colorectal cancer patients using the "oncopredict" package, screened nucleotide metabolism-related drug resistance genes, and constructed a risk score model. According to the Kaplan-Meier(KM) analysis, the overall survival (OS) and disease-free survival (PFS) of the high-risk group were significantly lower than those of the low-risk group. In addition, the nomogram we constructed had good performance in predicting OS in colon adenocarcinoma (COAD) patients. We validated NDUFA4L2 by cellular functionality experiments, animal tumorigenesis experiments and drug resistance experiments. It was demonstrated that NDUFA4L2 promoted the proliferation and migration of colon cancer cells, while the abnormal regulation of NDUFA4L2 affected the 5-FU resistance of colon cancer cells. In conclusion, we found that NDUFA4L2 promotes the progression and metastasis of colon cancer, as well as resistance to 5-FU chemotherapy.
Collapse
Affiliation(s)
- Hongxin He
- Department of Colorectal Surgery, Clinical Oncology School of Fujian Medical University, Fuzhou, 350004, Fujian, Fujian, P.R. China
| | - Shiyao Zheng
- Department of Colorectal Surgery, Clinical Oncology School of Fujian Medical University, Fuzhou, 350004, Fujian, Fujian, P.R. China
| | - Shangkun Jin
- Department of Colorectal Surgery, Clinical Oncology School of Fujian Medical University, Fuzhou, 350004, Fujian, Fujian, P.R. China
| | - Weijie Huang
- Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, China
| | - Enhao Wei
- Department of Colorectal Surgery, Clinical Oncology School of Fujian Medical University, Fuzhou, 350004, Fujian, Fujian, P.R. China
| | - Shen Guan
- Department of Colorectal Surgery, Clinical Oncology School of Fujian Medical University, Fuzhou, 350004, Fujian, Fujian, P.R. China
- Department of Colorectal Surgery, Clinical Oncology School of Fujian Medical University, 420# Fuma Road, 350011, Fuzhou, Fujian, China
| | - Chunkang Yang
- Department of Colorectal Surgery, Clinical Oncology School of Fujian Medical University, Fuzhou, 350004, Fujian, Fujian, P.R. China.
- Fujian Key Laboratory of Translational Cancer Medicine, 350014, Fuzhou, P.R. China.
- Department of Colorectal Surgery, Clinical Oncology School of Fujian Medical University, 420# Fuma Road, 350011, Fuzhou, Fujian, China.
| |
Collapse
|
13
|
Liu Q, Song Y, Su J, Yang S, Lian Q, Wang T, Wei H, Fang J. PUF60 Promotes Chemoresistance Through Drug Efflux and Reducing Apoptosis in Gastric Cancer. Int J Med Sci 2025; 22:269-282. [PMID: 39781520 PMCID: PMC11704696 DOI: 10.7150/ijms.102976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 11/15/2024] [Indexed: 01/12/2025] Open
Abstract
Background: Chemotherapy resistance is a great challenge in the treatment of gastric cancer (GC), so it is urgent to explore the prognostic markers of chemoresistance. PUF60 (Poly (U)-binding splicing factor 60) is a nucleic acid-binding protein that has been shown to regulate transcription and link to tumorigenesis in various cancers. However, its biological role and function in chemotherapy resistance of GC is unclear. Methods: The expression and prognostic value of PUF60 in GC chemotherapy-resistant patients were analyzed by databases and K-M Plotter. The functional effect of PUF60 on chemoresistance in GC was studied by by RNA interference, CCK8 test, colony formation test and apoptosis detection. Moreover, further validation and mechanism exploration were conducted in clinical samples. Results: PUF60 was highly expressed in both GC and chemoresistant tissues, and was positively correlated with poor prognosis in GC patients treated with 5-fluorouracil (5-FU). In addition, the knockdown of PUF60 significantly reduced the proliferation of human gastric cancer cells and increased sensitivity to chemotherapy drugs, such as 5-FU and cisplatin (CDDP). Mechanistically, PUF60 enhances chemotherapy resistance in gastric cancer (GC) cells by actively excluding chemotherapy drugs via the recombinant ATP Binding Cassette Transporter A1 (ABCA1) and ATP Binding Cassette Subfamily C Member 1 (ABCC1). This process further affects the cell cycle, reduces cell apoptosis, and ultimately promotes resistance to chemotherapy in GC. Conclusion: PUF60 promotes chemoresistance in GC, resulting in poor prognosis of GC patients treated with 5-FU, and providing a new idea for overcoming the chemoresistance in GC.
Collapse
Affiliation(s)
- Qianhui Liu
- Department of Gastrointestinal Surgery, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yingqiu Song
- Department of Gastrointestinal Surgery, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jing Su
- Department of Nursing, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shangbin Yang
- Department of Gastrointestinal Surgery, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qinghai Lian
- Department of Cell-Gene Therapy Translational Medicine Research Center, the Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Tiantian Wang
- Department of Medical Oncology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hongbo Wei
- Department of Gastrointestinal Surgery, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jiafeng Fang
- Department of Gastrointestinal Surgery, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
14
|
Chen Y, Yang X, Li Q. PDLIM1 Inhibits Chemoresistance by Blocking DNA Damage Repair in Gastric Cancer. Recent Pat Anticancer Drug Discov 2025; 20:260-273. [PMID: 38779728 DOI: 10.2174/0115748928307544240502064448] [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] [Revised: 04/06/2024] [Accepted: 04/18/2024] [Indexed: 05/25/2024]
Abstract
OBJECTIVE Current cisplatin (CDDP) resistance remains a major challenge in the treatment of advanced gastric cancer. To address the issue of drug resistance, we explored the regulatory functions of PDZ and LIM structural domain protein 1 (PDLIM1) in CDDP chemotherapy for gastric cancer. METHODS In this study, we analyzed PDLIM1 expression and prognosis using bioinformatics on publicly available data. PDLIM1 expression in a gastric mucosal epithelial cell line (GSE-1), CDDP- sensitive (SGC7901, BGC823) and CDDP-resistant gastric cancer cells was detected by RTqPCR and Western blotting. Cell proliferative capacity was assessed by knockdown of PDLIM1 and overexpression of PDLIM1 in cells administered in combination with cisplatin, and apoptotic levels were measured by CCK-8 and colony formation assay and by flow cytometry. Expression of breast cancer susceptibility gene 1 (BRCA1) and γH2AX was determined by Western blotting or immunofluorescence staining. RESULTS Downregulation of PDLIM1 was found in tumor tissues and cells, which was associated with poor clinical outcomes. Knockdown of PDLIM1 enhanced proliferation and attenuated apoptosis in gastric cancer cells. In addition, the therapeutic effects of CDDP on proliferation, apoptosis, and DNA damage repair were attenuated by PDLIM1 deletion.PDLIM1 expression was downregulated in CDDP-resistant tumor cells. Overexpression of PDLIM1 overcomes CDDP resistance in tumor cells as BRCA1 expression decreases and γH2AX expression increases. CONCLUSION Our findings demonstrate that PDLIM1 enables to alleviate gastric cancer progression and resistance to cisplatin via impeding DNA damage repair.
Collapse
Affiliation(s)
- Yuli Chen
- Health Management Center, The Third People's Hospital of Chengdu, Sichuan, Chengdu, 610014, China
| | - Xin Yang
- School of Clinical Medicine, Chengdu Medical College, Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
| | - Qiang Li
- Department of Emergency, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| |
Collapse
|
15
|
Yang C, Hu Y, Gao L, Li Z, Zhang Y, Zhuo R, Du Y, Liu H, Ji Q, Liu M, Pan J, Lu J, Xiao P, Tian Y, He S, Ling J, Hu S. Anagrelide and idarubicin combination induces GSDME-mediated pyroptosis as a potential therapy for high-PDE3A acute myeloid leukemia. Leukemia 2025; 39:98-111. [PMID: 39406931 DOI: 10.1038/s41375-024-02437-x] [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: 01/05/2024] [Revised: 09/09/2024] [Accepted: 10/07/2024] [Indexed: 10/18/2024]
Abstract
Acute myeloid leukemia (AML) is an invasive hematopoietic malignancy requiring novel treatment strategies. In this study, we identified phosphodiesterase 3 A (PDE3A) as a potential new target for drug repositioning in AML. PDE3A was preferentially overexpressed in AML cells than in normal cells, and high expression of PDE3A was correlated with lower event-free survival (EFS) in de novo AML patients. The PDE3A inhibitor anagrelide (ANA) profoundly suppresses the proliferation of high PDE3A-expressing AML cells while exhibiting minimal impact on those with low PDE3A expression. Moreover, synergistic effect of ANA with other chemotherapeutic drugs in high PDE3A expression AML cells was observed. The ANA-idarubicin (IDA) combination showed the most remarkable synergistic effect among all ANA-chemotherapeutic drugs commonly used in AML cell line models. Mechanistically, the synergy between ANA and IDA inhibited the survival of PDE3Ahigh AML cell lines through pyroptosis. This mechanism was initiated by GSDME cleavage triggered by caspase-3 activation. In vivo combination treatment of leukemic animals with high PDE3A expression significantly reduced leukemia burden and prolonged survival time compared with single-drug and vehicle control treatments. Our findings suggest that combined ANA and IDA treatment is an innovative and promising therapeutic strategy for AML patients with high PDE3A expression.
Collapse
MESH Headings
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/metabolism
- Humans
- Animals
- Mice
- Pyroptosis/drug effects
- Idarubicin/pharmacology
- Idarubicin/administration & dosage
- Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism
- Cyclic Nucleotide Phosphodiesterases, Type 3/genetics
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Xenograft Model Antitumor Assays
- Cell Proliferation/drug effects
- Drug Synergism
- Female
- Cell Line, Tumor
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Chenwei Yang
- Department of Pediatric Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, 215000, China
| | - Yixin Hu
- Department of Pediatric Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, 215000, China
| | - Li Gao
- Department of Pediatric Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, 215000, China
| | - Zhiheng Li
- Institution of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215000, China
| | - Yongping Zhang
- Department of Pediatric Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, 215000, China
| | - Ran Zhuo
- Department of Pediatric Surgery, Children's Hospital of Soochow University, Suzhou, 215000, China
| | - Yayun Du
- State Key Laboratory of Common Mechanism Research for Major Diseases and Key Laboratory of Synthetic Biology Regulatory Elements, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China
| | - Hu Liu
- Department of Pediatric Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, 215000, China
| | - Qi Ji
- Department of Pediatric Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, 215000, China
| | - Minyuan Liu
- Department of Pediatric Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, 215000, China
| | - Jian Pan
- Institution of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215000, China
| | - Jun Lu
- Department of Pediatric Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, 215000, China
| | - Peifang Xiao
- Department of Pediatric Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, 215000, China
| | - Yuanyuan Tian
- Institution of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215000, China.
| | - Sudan He
- State Key Laboratory of Common Mechanism Research for Major Diseases and Key Laboratory of Synthetic Biology Regulatory Elements, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China.
| | - Jing Ling
- Department of Transfusion Medicine, Children's Hospital of Soochow University, Suzhou, 215000, China.
| | - Shaoyan Hu
- Department of Pediatric Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, 215000, China.
- Jiangsu Pediatric Hematology & Oncology Center, Jiangsu, China.
| |
Collapse
|
16
|
Shen Y, Zhang T, Jia X, Xi F, Jing W, Wang Y, Huang M, Na R, Xu L, Ji W, Qiao Y, Zhang X, Sun W, Li S, Wu J. MEF2A, a gene associated with mitochondrial biogenesis, promotes drug resistance in gastric cancer. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167497. [PMID: 39237047 DOI: 10.1016/j.bbadis.2024.167497] [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: 08/24/2024] [Accepted: 08/30/2024] [Indexed: 09/07/2024]
Abstract
Chemotherapeutic resistance is a major obstacle to the effectiveness of cisplatin-based chemotherapy for gastric cancer (GC), leading to treatment failure and poor survival rates. However, the underlying mechanisms are not fully understood. Our study demonstrated that the transcription factor myocyte enhancer factor 2A (MEF2A) plays a role in chemotherapeutic drug resistance by regulating the transcription of PGC1α and KEAP1, promoting mitochondrial biogenesis. It was found that increased MEF2A expression is linked with poor prognosis, cisplatin insensitivity, and mitochondrial function in GC. MEF2A overexpression significantly decreases GC cell sensitivity in vitro and in vivo, while MEF2A knockdown enhances the sensitivity to cisplatin. Mechanistically, MEF2A activates the transcription of PGC1α, leading to increased mitochondrial biogenesis. In addition, MEF2A inhibits KEAP1 transcription, reduces NRF2 ubiquitination degradation, and activates the KEAP1/NRF2 signaling pathway, which modulates the reactive oxygen species level. The present study identifies MEF2A as a new critical oncogene involved in GC chemoresistance, suggesting a novel therapeutic target for GC.
Collapse
Affiliation(s)
- Yao Shen
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China; Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China
| | - Tong Zhang
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China; Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China
| | - Xueyuan Jia
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China; Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China
| | - Fei Xi
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China; Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China
| | - Wanting Jing
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China; Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China
| | - Yusi Wang
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China; Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China
| | - Min Huang
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China; Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China
| | - Ruisi Na
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Lidan Xu
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China; Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China
| | - Wei Ji
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China; Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China
| | - Yuandong Qiao
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China; Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China
| | - Xuelong Zhang
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China; Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China
| | - Wenjing Sun
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China; Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China
| | - Shuijie Li
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, Harbin, China.
| | - Jie Wu
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China; Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China; Future Medical Laboratory, the 2nd Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
17
|
Lin Q, Liu HM, Wu LZ, Yu DD, Hua CY, Zou Y, Jiao WE, Li XP, Chen SM. Effect and underlying mechanism of a photochemotherapy dual-function nanodrug delivery system for head and neck squamous cell carcinoma. J Transl Med 2024; 22:1043. [PMID: 39563418 DOI: 10.1186/s12967-024-05855-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 11/04/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND The novel nanomaterials PNA-TN (PN) and PNA-TN-Dox (PND) have been shown to have strong inhibitory effects on breast cancer; however, it is unclear whether PN and PND have anti-head and neck squamous cell carcinoma (HNSCC) activity, and their potential mechanisms of activity are unknown. So, our study aims to explore the therapeutic effects of PN and PND on HNSCC and their possible mechanisms. METHODS We used a series of phenotypic research to evaluate the effects of PN + Laser (L) and PND + L on the biological function of HNSCC cells in vitro and in vivo. We subsequently used mechanism research to examine changes in mRNA and protein expression related to apoptosis, epithelial‒mesenchymal transition (EMT), and the JNK signalling pathway. RESULTS Our study revealed that PN and PND have strong inhibitory effects on HNSCC cells both in vitro and in vivo. In vitro, PN and PND significantly inhibited the proliferation, migration, invasion and EMT ability of HNSCC cells and promoted apoptosis; the inhibitory effect in the PND + L group was significantly greater than that in the PN + L group. In vivo, both treatments led to significant reductions in tumour volume and weight. Notably, the tumour volume and weight in the PND + L group were significantly lower than those in the PN + L group. Mechanism research confirmed that PN + L activated the expression of apoptosis-related proteins and inhibited the expression of EMT-related proteins via the JNK pathway. Furthermore, the anti-HNSCC effect of PN + L was blocked after the use of a JNK pathway inhibitor. CONCLUSION Treatment with PN + L or PND + L significantly inhibited the malignant progress of HNSCC cells, and the therapeutic effect of PND + L was significantly stronger than that of PN + L. The JNK signalling pathway is a key mechanism by which PN exerts its anti-HNSCC activity.
Collapse
Affiliation(s)
- Qian Lin
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, P. R. China
| | - Hui-Min Liu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, No. 238, Jie-Fang Road, Wuhan, Hubei, 430060, P. R. China
| | - Li-Zhi Wu
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, P. R. China
| | - Dong-Dong Yu
- Department of Oncology, Renmin Hospital of Wuhan University, No. 238, Jie-Fang Road, Wuhan, Hubei, 430060, P. R. China
| | - Cheng-Yu Hua
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, P. R. China
| | - You Zou
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, P. R. China
| | - Wo-Er Jiao
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, P. R. China
| | - Xiang-Pan Li
- Department of Oncology, Renmin Hospital of Wuhan University, No. 238, Jie-Fang Road, Wuhan, Hubei, 430060, P. R. China.
| | - Shi-Ming Chen
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, P. R. China.
| |
Collapse
|
18
|
Yao Z, Lu Y, Wang P, Chen Z, Zhou L, Sang X, Yang Q, Wang K, Hao M, Cao G. The role of JNK signaling pathway in organ fibrosis. J Adv Res 2024:S2090-1232(24)00431-4. [PMID: 39366483 DOI: 10.1016/j.jare.2024.09.029] [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: 07/25/2024] [Revised: 09/28/2024] [Accepted: 09/29/2024] [Indexed: 10/06/2024] Open
Abstract
BACKGROUND Fibrosis is a tissue damage repair response caused by multiple pathogenic factors which could occur in almost every apparatus and leading to the tissue structure damage, physiological abnormality, and even organ failure until death. Up to now, there is still no specific drugs or strategies can effectively block or changeover tissue fibrosis. JNKs, a subset of mitogen-activated protein kinases (MAPK), have been reported that participates in various biological processes, such as genetic expression, DNA damage, and cell activation/proliferation/death pathways. Increasing studies indicated that abnormal regulation of JNK signal pathway has strongly associated with tissue fibrosis. AIM OF REVIEW This review designed to sum up the molecular mechanism progresses in the role of JNK signal pathway in organ fibrosis, hoping to provide a novel therapy strategy to tackle tissue fibrosis. KEY SCIENTIFIC CONCEPTS OF REVIEW Recent evidence shows that JNK signaling pathway could modulates inflammation, immunoreaction, oxidative stress and Multiple cell biological functions in organ fibrosis. Therefore, targeting the JNK pathway may be a useful strategy in cure fibrosis.
Collapse
Affiliation(s)
- Zhouhui Yao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yandan Lu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Pingping Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Ziyan Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Licheng Zhou
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xianan Sang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Qiao Yang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Kuilong Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Min Hao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Songyang Research Institute of Zhejiang Chinese Medical University, Songyang, 323400, China.
| | - Gang Cao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
19
|
Zhang J, Chen Y, Gong X, Yang Y, Gu Y, Huang L, Fu J, Zhao M, Huang Y, Li L, Liu W, Wan Y, He X, Ma Z, Zhao W, Zhang M, Tang T, Wang Y, Thiery JP, Zheng X, Chen L. GATA factor TRPS1, a new DNA repair protein, cooperates with reversible PARylation to promote chemoresistance in patients with breast cancer. J Biol Chem 2024; 300:107780. [PMID: 39276941 PMCID: PMC11490888 DOI: 10.1016/j.jbc.2024.107780] [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: 11/19/2023] [Revised: 08/26/2024] [Accepted: 09/02/2024] [Indexed: 09/17/2024] Open
Abstract
Resistance to DNA-damaging agents is a major unsolved challenge for breast cancer patients undergoing chemotherapy. Here, we show that elevated expression of transcriptional repressor GATA binding 1 (TRPS1) is associated with lower drug sensitivity, reduced response rate, and poor prognosis in chemotherapy-treated breast cancer patients. Mechanistically, elevated TRPS1 expression promotes hyperactivity of DNA damage repair (DDR) in breast cancer cells. We provide evidence that TRPS1 dynamically localizes to DNA breaks in a Ku70-and Ku80-dependent manner and that TRPS1 is a new member of the DDR protein family. We also discover that the dynamics of TRPS1 assembly at DNA breaks is regulated by its reversible PARylation in the DDR, and that mutations of the PARylation sites on TRPS1 lead to increased sensitivity to chemotherapeutic drugs. Taken together, our findings provide new mechanistic insights into the DDR and chemoresistance in breast cancer patients and identify TRPS1 as a critical DDR protein. TRPS1 may also be considered as a target to improve chemo-sensitization strategies and, consequently, clinical outcomes for breast cancer patients.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Yatao Chen
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Xue Gong
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China; Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China
| | - Yongfeng Yang
- State Key Lab of Protein and Plant Gene Research, Department of Biochemistry and Molecular Biology, School of Life Sciences, Peking University, Beijing, China
| | - Yun Gu
- Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China
| | - Ling Huang
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Jianfeng Fu
- State Key Lab of Protein and Plant Gene Research, Department of Biochemistry and Molecular Biology, School of Life Sciences, Peking University, Beijing, China
| | - Menglu Zhao
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Yehong Huang
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Lulu Li
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Wenzhuo Liu
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Yajie Wan
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Xilin He
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Zhifang Ma
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China; Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China
| | - Weiyong Zhao
- Department of Radiation Oncology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Meng Zhang
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Tao Tang
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Yuzhi Wang
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | | | - Xiaofeng Zheng
- State Key Lab of Protein and Plant Gene Research, Department of Biochemistry and Molecular Biology, School of Life Sciences, Peking University, Beijing, China.
| | - Liming Chen
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China; Jiangsu Institute of Cancer Research, Jiangsu Cancer Hospital, the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
20
|
Hou YM, Xu BH, Zhang QT, Cheng J, Zhang X, Yang HR, Wang ZY, Wang P, Zhang MX. Deficiency of smooth muscle cell ILF3 alleviates intimal hyperplasia via HMGB1 mRNA degradation-mediated regulation of the STAT3/DUSP16 axis. J Mol Cell Cardiol 2024; 190:62-75. [PMID: 38583797 DOI: 10.1016/j.yjmcc.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/03/2024] [Accepted: 04/04/2024] [Indexed: 04/09/2024]
Abstract
Intimal hyperplasia is a complicated pathophysiological phenomenon attributable to in-stent restenosis, and the underlying mechanism remains unclear. Interleukin enhancer-binding factor 3 (ILF3), a double-stranded RNA-binding protein involved in regulating mRNA stability, has been recently demonstrated to assume a crucial role in cardiovascular disease; nevertheless, its impact on intimal hyperplasia remains unknown. In current study, we used samples of human restenotic arteries and rodent models of intimal hyperplasia, we found that vascular smooth muscle cell (VSMC) ILF3 expression was markedly elevated in human restenotic arteries and murine ligated carotid arteries. SMC-specific ILF3 knockout mice significantly suppressed injury induced neointimal formation. In vitro, platelet-derived growth factor type BB (PDGF-BB) treatment elevated the level of VSMC ILF3 in a dose- and time-dependent manner. ILF3 silencing markedly inhibited PDGF-BB-induced phenotype switching, proliferation, and migration in VSMCs. Transcriptome sequencing and RNA immunoprecipitation sequencing depicted that ILF3 maintained its stability upon binding to the mRNA of the high-mobility group box 1 protein (HMGB1), thereby exerting an inhibitory effect on the transcription of dual specificity phosphatase 16 (DUSP16) through enhanced phosphorylation of signal transducer and activator of transcription 3 (STAT3). Therefore, the results both in vitro and in vivo indicated that the loss of ILF3 in VSMC ameliorated neointimal hyperplasia by regulating the STAT3/DUSP16 axis through the degradation of HMGB1 mRNA. Our findings revealed that vascular injury activates VSMC ILF3, which in turn promotes intima formation. Consequently, targeting specific VSMC ILF3 may present a potential therapeutic strategy for ameliorating cardiovascular restenosis.
Collapse
Affiliation(s)
- Ya-Min Hou
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Bo-Han Xu
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Qiu-Ting Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Jie Cheng
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Xu Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Hong-Rui Yang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Ze-Ying Wang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Peng Wang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Ming-Xiang Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
21
|
Enomoto A, Fukasawa T, Terunuma H, Nakagawa K, Yoshizaki A, Sato S, Hosoya N, Miyagawa K. Deregulated JNK signaling enhances apoptosis during hyperthermia. Int J Hyperthermia 2024; 41:2335199. [PMID: 38565204 DOI: 10.1080/02656736.2024.2335199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 03/22/2024] [Indexed: 04/04/2024] Open
Abstract
PURPOSE c-Jun N-terminal kinases (JNKs) comprise a subfamily of mitogen-activated protein kinases (MAPKs). The JNK group is known to be activated by a variety of stimuli. However, the molecular mechanism underlying heat-induced JNK activation is largely unknown. The aim of this study was to clarify how JNK activity is stimulated by heat. METHODS AND MATERIALS The expression levels of various MAPK members in HeLa cells, with or without hyperthermia treatment, were evaluated via western blotting. The kinase activity of MAPK members was assessed through in vitro kinase assays. Cell death was assessed in the absence or presence of siRNAs targeting MAPK-related members. RESULTS Hyperthermia decreased the levels of MAP3Ks, such as ASK1 and MLK3 which are JNK kinase kinase members, but not those of the downstream MAP2K/SEK1 and MAPK/JNK. Despite the reduced or transient phosphorylation of ASK1, MLK3, or SEK1, downstream JNK was phosphorylated in a temperature-dependent manner. In vitro kinase assays demonstrated that heat did not directly stimulate SEK1 or JNK. However, the expression levels of DUSP16, a JNK phosphatase, were decreased upon hyperthermia treatment. DUSP16 knockdown enhanced the heat-induced activation of ASK1-SEK1-JNK pathway and apoptosis. CONCLUSION JNK was activated in a temperature-dependent manner despite reduced or transient phosphorylation of the upstream MAP3K and MAP2K. Hyperthermia-induced degradation of DUSP16 may induce activation of the ASK1-SEK1-JNK pathway and subsequent apoptosis.
Collapse
Affiliation(s)
- Atsushi Enomoto
- Laboratory of Molecular Radiology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takemichi Fukasawa
- Department of Dermatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Clinical Cannabinoid Research, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | | | - Keiichi Nakagawa
- Comprehensive Radiation Oncology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ayumi Yoshizaki
- Department of Dermatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Clinical Cannabinoid Research, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shinichi Sato
- Department of Dermatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Noriko Hosoya
- Laboratory of Molecular Radiology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kiyoshi Miyagawa
- Laboratory of Molecular Radiology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
22
|
Kou X, Yang X, Zhao Z, Li L. HSPA8-mediated stability of the CLPP protein regulates mitochondrial autophagy in cisplatin-resistant ovarian cancer cells. Acta Biochim Biophys Sin (Shanghai) 2024; 56:356-365. [PMID: 38419499 PMCID: PMC10984867 DOI: 10.3724/abbs.2023246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 08/07/2023] [Indexed: 03/02/2024] Open
Abstract
Currently, platinum agents remain the mainstay of chemotherapy for ovarian cancer (OC). However, cisplatin (DDP) resistance is a major reason for chemotherapy failure. Thus, it is extremely important to elucidate the mechanism of resistance to DDP. Here, we establish two DDP-resistant ovarian cancer cell lines and find that caseinolytic protease P (CLPP) level is significantly downregulated in DDP-resistant cell lines compared to wild-type ovarian cancer cell lines (SK-OV-3 and OVcar3). Next, we investigate the functions of CLPP in DDP-resistant and wild-type ovarian cancer cells using various assays, including cell counting kit-8 assay, western blot analysis, immunofluorescence staining, and detection of reactive oxygen species (ROS) and apoptosis. Our results show that CLPP knockdown significantly increases the half maximal inhibitory concentration (IC 50) and mitophagy of wild-type SK-OV-3 and OVcar3 cells, while CLPP overexpression reduces the IC 50 values and mitophagy of DDP-resistant SK-OV-3 and OVcar3 cells. Next, we perform database predictions and confirmation experiments, which show that heat shock protein family A member 8 (HSPA8) regulates CLPP protein stability. The dynamic effects of the HSPA8/CLPP axis in ovarian cancer cells are also examined. HSPA8 increases mitophagy and the IC 50 values of SK-OV-3 and OVcar3 cells but inhibits their ROS production and apoptosis. In addition, CLPP partly reverses the effects induced by HSPA8 in SK-OV-3 and OVcar3 cells. In conclusion, CLPP increases DDP resistance in ovarian cancer by inhibiting mitophagy and promoting cellular stress. Meanwhile, HSPA8 promotes the degradation of CLPP protein by regulating its stability.
Collapse
Affiliation(s)
- Xinxin Kou
- />Department of GynecologyCancer Hospital Affiliated to Zhengzhou UniversityZhengzhou450008China
| | - Xiaoxia Yang
- />Department of GynecologyCancer Hospital Affiliated to Zhengzhou UniversityZhengzhou450008China
| | - Zheng Zhao
- />Department of GynecologyCancer Hospital Affiliated to Zhengzhou UniversityZhengzhou450008China
| | - Lei Li
- />Department of GynecologyCancer Hospital Affiliated to Zhengzhou UniversityZhengzhou450008China
| |
Collapse
|
23
|
Zou JY, Chen QL, Luo XC, Damdinjav D, Abdelmohsen UR, Li HY, Battulga T, Chen HB, Wang YQ, Zhang JY. Natural products reverse cancer multidrug resistance. Front Pharmacol 2024; 15:1348076. [PMID: 38572428 PMCID: PMC10988293 DOI: 10.3389/fphar.2024.1348076] [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: 12/01/2023] [Accepted: 02/19/2024] [Indexed: 04/05/2024] Open
Abstract
Cancer stands as a prominent global cause of death. One of the key reasons why clinical tumor chemotherapy fails is multidrug resistance (MDR). In recent decades, accumulated studies have shown how Natural Product-Derived Compounds can reverse tumor MDR. Discovering novel potential modulators to reduce tumor MDR by Natural Product-Derived Compounds has become a popular research area across the globe. Numerous studies mainly focus on natural products including flavonoids, alkaloids, terpenoids, polyphenols and coumarins for their MDR modulatory activity. Natural products reverse MDR by regulating signaling pathways or the relevant expressed protein or gene. Here we perform a deep review of the previous achievements, recent advances in the development of natural products as a treatment for MDR. This review aims to provide some insights for the study of multidrug resistance of natural products.
Collapse
Affiliation(s)
- Jia-Yu Zou
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qi-Lei Chen
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
| | - Xiao-Ci Luo
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Davaadagva Damdinjav
- School of Pharmacy, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Usama Ramadan Abdelmohsen
- Deraya Center for Scientific Research, Deraya University, New Minia, Egypt
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Hong-Yan Li
- Ministry of Education Engineering Research Center of Tibetan Medicine Detection Technology, Xizang Minzu University, Xianyang, China
| | - Tungalag Battulga
- School of Pharmacy, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Hu-Biao Chen
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
| | - Yu-Qing Wang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- The Affiliated TCM Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jian-Ye Zhang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- The Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, China
| |
Collapse
|
24
|
Tao J, Xue C, Cao M, Ye J, Sun Y, Chen H, Guan Y, Zhang W, Zhang W, Yao Y. Protein disulfide isomerase family member 4 promotes triple-negative breast cancer tumorigenesis and radiotherapy resistance through JNK pathway. Breast Cancer Res 2024; 26:1. [PMID: 38167446 PMCID: PMC10759449 DOI: 10.1186/s13058-023-01758-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 12/20/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Despite radiotherapy ability to significantly improve treatment outcomes and survival in triple-negative breast cancer (TNBC) patients, acquired resistance to radiotherapy poses a serious clinical challenge. Protein disulfide isomerase exists in endoplasmic reticulum and plays an important role in promoting protein folding and post-translational modification. However, little is known about the role of protein disulfide isomerase family member 4 (PDIA4) in TNBC, especially in the context of radiotherapy resistance. METHODS We detected the presence of PDIA4 in TNBC tissues and paracancerous tissues, then examined the proliferation and apoptosis of TNBC cells with/without radiotherapy. As part of the validation process, xenograft tumor mouse model was used. Mass spectrometry and western blot analysis were used to identify PDIA4-mediated molecular signaling pathway. RESULTS Based on paired clinical specimens of TNBC patients, we found that PDIA4 expression was significantly higher in tumor tissues compared to adjacent normal tissues. In vitro, PDIA4 knockdown not only increased apoptosis of tumor cells with/without radiotherapy, but also decreased the ability of proliferation. In contrast, overexpression of PDIA4 induced the opposite effects on apoptosis and proliferation. According to Co-IP/MS results, PDIA4 prevented Tax1 binding protein 1 (TAX1BP1) degradation by binding to TAX1BP1, which inhibited c-Jun N-terminal kinase (JNK) activation. Moreover, PDIA4 knockdown suppressed tumor growth xenograft model in vivo, which was accompanied by an increase in apoptosis and promoted tumor growth inhibition after radiotherapy. CONCLUSIONS The results of this study indicate that PDIA4 is an oncoprotein that promotes TNBC progression, and targeted therapy may represent a new and effective anti-tumor strategy, especially for patients with radiotherapy resistance.
Collapse
Affiliation(s)
- Jinqiu Tao
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Cailin Xue
- Division of Hepatobilliary Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Meng Cao
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Jiahui Ye
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Yulu Sun
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Hao Chen
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Yinan Guan
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Wenjie Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
| | - Weijie Zhang
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
| | - Yongzhong Yao
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
| |
Collapse
|
25
|
Liang L, Zhang X, Su X, Zeng T, Suo D, Yun J, Wang X, Guan XY, Li Y. Fibroblasts in metastatic lymph nodes confer cisplatin resistance to ESCC tumor cells via PI16. Oncogenesis 2023; 12:50. [PMID: 37914722 PMCID: PMC10620422 DOI: 10.1038/s41389-023-00495-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 10/11/2023] [Accepted: 10/20/2023] [Indexed: 11/03/2023] Open
Abstract
Although many studies have compared tumor fibroblasts (T-Fbs) and nontumor fibroblasts (N-Fbs), less is understood about the stromal contribution of metastatic lymph node fibroblasts (LN-Fbs) to the evolving microenvironment. Here, we explored the characteristics of LN-Fbs in esophageal squamous cell carcinoma (ESCC) and the interactions between fibroblasts and ESCC tumor cells in metastatic lymph nodes. Fibroblasts were isolated from tumor, nontumor and metastatic lymph node tissues from different patients with ESCC. Transcriptome sequencing was performed on the fibroblasts. Tumor growth and drug-resistance assays were carried out, and characteristics of T-Fbs, N-Fbs and LN-Fbs were determined. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) was used to assay the culture medium of fibroblasts. The results demonstrated that fibroblasts derived from different tissues had different characteristics. Coculture with LN-Fbs conditioned medium inhibited ESCC tumor cell growth and induced chemoresistance in ESCC cells. LN-Fbs induced chemoresistance to cisplatin in ESCC cells by secreting PI16. Coculture with LN-Fbs conditioned medium decreased cisplatin-induced apoptosis in ESCC cells by regulating the p38 and JNK cell signaling pathways. Survival analyses showed that patients with high PI16 expression in Fbs of lymph nodes exhibited worse overall survival. We also examined PI16 expression in interstitial tissues in ESCC tumor samples of patients receiving platinum-based therapy postsurgery and found that high PI16 expression in tumor interstitial tissues was an independent prognostic factor for ESCC patients. In addition, an in vivo assay demonstrated that PI16 knockdown increased the sensitivity of ESCC cells to cisplatin. Our results suggest that fibroblasts in metastatic lymph nodes decrease apoptosis of ESCC cells via PI16, thereby providing a cisplatin-resistance niche and supporting ESCC tumor cells to survive in metastatic lymph nodes. PI16 is also a potential target for effectively blocking the chemoresistance niche signaling circuit in response to cisplatin.
Collapse
Affiliation(s)
- Lily Liang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Guangdong Esophageal Cancer Institute, Department of Thoracic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xu Zhang
- Guangdong Esophageal Cancer Institute, Department of Thoracic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiaodong Su
- Guangdong Esophageal Cancer Institute, Department of Thoracic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Tingting Zeng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Daqin Suo
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jingping Yun
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xin Wang
- Guangdong Esophageal Cancer Institute, Department of Thoracic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xin-Yuan Guan
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China
- Guangdong-Hongkong Joint Laboratory for RNA medicine, Sun Yat-sen University, Guangzhou, China
| | - Yan Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China.
- Guangdong Esophageal Cancer Institute, Department of Thoracic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.
- Guangdong-Hongkong Joint Laboratory for RNA medicine, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
26
|
Tomic U, Nikolic N, Carkic J, Mihailovic D, Jelovac D, Milasin J, Pucar A. Streptococcus mitis and Prevotella melaninogenica Influence Gene Expression Changes in Oral Mucosal Lesions in Periodontitis Patients. Pathogens 2023; 12:1194. [PMID: 37887710 PMCID: PMC10610332 DOI: 10.3390/pathogens12101194] [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: 08/20/2023] [Revised: 09/12/2023] [Accepted: 09/20/2023] [Indexed: 10/28/2023] Open
Abstract
Oral microbiome disruptions in periodontitis are related to the chronic inflammatory reactions that could in turn lead to the development of multiple oral diseases. The objective of the study was to assess the frequencies of Streptococcus mitis, Prevotella melaninogenica, and Prevotella intermedia in oral benign lesions, oral potentially malignant disorders (OPMDs), and oral squamous cell carcinomas (OSCCs) and investigate the impact of these bacteria on the expression patterns of the selected (potential) target genes (PI3CA/AKT2/mTOR, DUSP16/MAPK14, and COX2). After sample collection (25 benign lesions, 30 OPMDs, and 35 OSCCs) and DNA/RNA extraction, quantitative real-time polymerase chain reaction (qPCR) was performed to detect bacterial presence and assess relative gene expression levels in different lesion groups. Prevotella melaninogenica was the most prevalent of the three analyzed bacteria, with the frequency being 60% in benign lesions, 87% in OPMDs (p = 0.024), and 77% in OSCC. The OPMD tissues in which Prevotella melaninogenica was present exhibited a higher expression level of AKT2 (p = 0.042). Significantly lower expression of DUSP16 was observed in OSCC tissues containing Streptococcus mitis (p = 0.011). The obtained results indicate a substantial contribution of P. melaninogenica and Str. mitis in the pathogenesis of oral mucosal lesions, possibly via AKT2 upregulation and DUSP16 downregulation.
Collapse
Affiliation(s)
- Uros Tomic
- Clinic for Periodontology and Oral Medicine, School of Dental Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Nadja Nikolic
- Department of Human Genetics, School of Dental Medicine, University of Belgrade, 11000 Belgrade, Serbia; (J.C.); (J.M.)
| | - Jelena Carkic
- Department of Human Genetics, School of Dental Medicine, University of Belgrade, 11000 Belgrade, Serbia; (J.C.); (J.M.)
| | - Djordje Mihailovic
- Department of Dentistry, Faculty of Medical Sciences Pristina, University of Pristina, 38220 Kosovska Mitrovica, Serbia;
| | - Drago Jelovac
- Clinic for Maxillofacial Surgery, School of Dental Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Jelena Milasin
- Department of Human Genetics, School of Dental Medicine, University of Belgrade, 11000 Belgrade, Serbia; (J.C.); (J.M.)
| | - Ana Pucar
- Clinic for Periodontology and Oral Medicine, School of Dental Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| |
Collapse
|
27
|
Wei X, Png CW, Weerasooriya M, Li H, Zhu C, Chen G, Xu C, Zhang Y, Xu X. Tumor Promoting Function of DUSP10 in Non-Small Cell Lung Cancer Is Associated With Tumor-Promoting Cytokines. Immune Netw 2023; 23:e34. [PMID: 37670811 PMCID: PMC10475826 DOI: 10.4110/in.2023.23.e34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 07/09/2023] [Accepted: 07/31/2023] [Indexed: 09/07/2023] Open
Abstract
Lung cancer, particularly non-small cell lung cancer (NSCLC) which contributes more than 80% to totally lung cancer cases, remains the leading cause of cancer death and the 5-year survival is less than 20%. Continuous understanding on the mechanisms underlying the pathogenesis of this disease and identification of biomarkers for therapeutic application and response to treatment will help to improve patient survival. Here we found that a molecule known as DUSP10 (also known as MAPK phosphatase 5) is oncogenic in NSCLC. Overexpression of DUSP10 in NSCLC cells resulted in reduced activation of ERK and JNK, but increased activation of p38, which was associated with increased cellular growth and migration. When inoculated in immunodeficient mice, the DUSP10-overexpression NSCLC cells formed larger tumors compared to control cells. The increased growth of DUSP10-overexpression NSCLC cells was associated with increased expression of tumor-promoting cytokines including IL-6 and TGFβ. Importantly, higher DUSP10 expression was associated with poorer prognosis of NSCLC patients. Therefore, DUSP10 could severe as a biomarker for NSCLC prognosis and could be a target for development of therapeutic method for lung cancer treatment.
Collapse
Affiliation(s)
- Xing Wei
- Breast Surgery Department, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310006, China
| | - Chin Wen Png
- Department of Microbiology and Immunology, and NUSMED Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore
- Immunology Programme, Institute of Life Sciences, National University of Singapore, Singapore 117545, Singapore
| | - Madhushanee Weerasooriya
- Department of Microbiology and Immunology, and NUSMED Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore
- Immunology Programme, Institute of Life Sciences, National University of Singapore, Singapore 117545, Singapore
| | - Heng Li
- Department of Microbiology and Immunology, and NUSMED Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore
- Immunology Programme, Institute of Life Sciences, National University of Singapore, Singapore 117545, Singapore
| | - Chenchen Zhu
- Breast Surgery Department, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310006, China
| | - Guiping Chen
- Breast Surgery Department, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310006, China
| | - Chuan Xu
- Department of Oncology & Cancer Institute, Sichuan Academy of Medical Sciences, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu 610056, China
| | - Yongliang Zhang
- Department of Microbiology and Immunology, and NUSMED Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore
- Immunology Programme, Institute of Life Sciences, National University of Singapore, Singapore 117545, Singapore
| | - Xiaohong Xu
- Breast Surgery Department, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310006, China
| |
Collapse
|
28
|
Zheng XX, Chen JJ, Sun YB, Chen TQ, Wang J, Yu SC. Mitochondria in cancer stem cells: Achilles heel or hard armor. Trends Cell Biol 2023; 33:708-727. [PMID: 37137792 DOI: 10.1016/j.tcb.2023.03.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 03/14/2023] [Accepted: 03/15/2023] [Indexed: 05/05/2023]
Abstract
Previous studies have shown that mitochondria play core roles in not only cancer stem cell (CSC) metabolism but also the regulation of CSC stemness maintenance and differentiation, which are key regulators of cancer progression and therapeutic resistance. Therefore, an in-depth study of the regulatory mechanism of mitochondria in CSCs is expected to provide a new target for cancer therapy. This article mainly introduces the roles played by mitochondria and related mechanisms in CSC stemness maintenance, metabolic transformation, and chemoresistance. The discussion mainly focuses on the following aspects: mitochondrial morphological structure, subcellular localization, mitochondrial DNA, mitochondrial metabolism, and mitophagy. The manuscript also describes the recent clinical research progress on mitochondria-targeted drugs and discusses the basic principles of their targeted strategies. Indeed, an understanding of the application of mitochondria in the regulation of CSCs will promote the development of novel CSC-targeted strategies, thereby significantly improving the long-term survival rate of patients with cancer.
Collapse
Affiliation(s)
- Xiao-Xia Zheng
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing 400038, China; Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing 400038, China
| | - Jun-Jie Chen
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing 400038, China; Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing 400038, China
| | - Yi-Bo Sun
- College of Basic Medical Sciences, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Tian-Qing Chen
- School of Pharmacy, Shanxi Medical University, Taiyuan, 030002, Shanxi, China
| | - Jun Wang
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing 400038, China; Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing 400038, China
| | - Shi-Cang Yu
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing 400038, China; Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing 400038, China; College of Basic Medical Sciences, Third Military Medical University (Army Medical University), Chongqing 400038, China; Jin-feng Laboratory, Chongqing 401329, China.
| |
Collapse
|
29
|
Xu T, Yang Y, Chen Z, Wang J, Wang X, Zheng Y, Wang C, Wang Y, Zhu Z, Ding X, Zhou J, Li G, Zhang H, Zhang W, Wu Y, Song X. TNFAIP2 confers cisplatin resistance in head and neck squamous cell carcinoma via KEAP1/NRF2 signaling. J Exp Clin Cancer Res 2023; 42:190. [PMID: 37525222 PMCID: PMC10391982 DOI: 10.1186/s13046-023-02775-1] [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: 03/08/2023] [Accepted: 07/22/2023] [Indexed: 08/02/2023] Open
Abstract
BACKGROUND Drug resistance limits the treatment effect of cisplatin-based chemotherapy in head and neck squamous cell carcinoma (HNSCC), and the underlying mechanism is not fully understood. The aim of this study was to explore the cause of cisplatin resistance in HNSCC. METHODS We performed survival and gene set variation analyses based on HNSCC cohorts and identified the critical role of tumor necrosis factor alpha-induced protein 2 (TNFAIP2) in cisplatin-based chemotherapy resistance. Half-maximal inhibitory concentration (IC50) examination, colony formation assays and flow cytometry assays were conducted to examine the role of TNFAIP2 in vitro, while xenograft models in nude mice and 4-nitroquinoline N-oxide (4NQO)-induced HNSCC models in C57BL/6 mice were adopted to verify the effect of TNFAIP2 in vivo. Gene set enrichment analysis (GSEA) and coimmunoprecipitation coupled with mass spectrometry (Co-IP/MS) were performed to determine the mechanism by which TNFAIP2 promotes cisplatin resistance. RESULTS High expression of TNFAIP2 is associated with a poor prognosis, cisplatin resistance, and low reactive oxygen species (ROS) levels in HNSCC. Specifically, it protects cancer cells from cisplatin-induced apoptosis by inhibiting ROS-mediated c-JUN N-terminal kinase (JNK) phosphorylation. Mechanistically, the DLG motif contained in TNFAIP2 competes with nuclear factor-erythroid 2-related factor 2 (NRF2) by directly binding to the Kelch domain of Kelch-like ECH-associated protein 1 (KEAP1), which prevents NRF2 from undergoing ubiquitin proteasome-mediated degradation. This results in the accumulation of NRF2 and confers cisplatin resistance. Positive correlations between TNFAIP2 protein levels and NRF2 as well as its downstream target genes were validated in HNSCC specimens. Moreover, the small interfering RNA (siRNA) targeting TNFAIP2 significantly enhanced the cisplatin treatment effect in a 4NQO-induced HNSCC mouse model. CONCLUSIONS Our results reveal the antioxidant and cisplatin resistance-regulating roles of the TNFAIP2/KEAP1/NRF2/JNK axis in HNSCC, suggesting that TNFAIP2 might be a potential target in improving the cisplatin treatment effect, particularly for patients with cisplatin resistance.
Collapse
Affiliation(s)
- Teng Xu
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Yuemei Yang
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Zhihong Chen
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Jinsong Wang
- Department of Pathology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xiaolei Wang
- Department of Pathology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yang Zheng
- Department of Oral Maxillofacial & Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center of Stomatology, Shanghai, China
| | - Chao Wang
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Yachen Wang
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Zaiou Zhu
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Xu Ding
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Junbo Zhou
- Department of Stomatology, Nanjing Integrated Traditional Chinese and Western Medicine Hospital, Nanjing, China
| | - Gang Li
- Department of Stomatology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Hongchuang Zhang
- Department of Stomatology, Xuzhou No. 1 Peoples Hospital, Xuzhou, China
| | - Wei Zhang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China.
| | - Yunong Wu
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China.
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China.
| | - Xiaomeng Song
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China.
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
30
|
Nguyen TT, Wei S, Nguyen TH, Jo Y, Zhang Y, Park W, Gariani K, Oh CM, Kim HH, Ha KT, Park KS, Park R, Lee IK, Shong M, Houtkooper RH, Ryu D. Mitochondria-associated programmed cell death as a therapeutic target for age-related disease. Exp Mol Med 2023; 55:1595-1619. [PMID: 37612409 PMCID: PMC10474116 DOI: 10.1038/s12276-023-01046-5] [Citation(s) in RCA: 104] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/24/2023] [Accepted: 04/27/2023] [Indexed: 08/25/2023] Open
Abstract
Mitochondria, ubiquitous double-membrane-bound organelles, regulate energy production, support cellular activities, harbor metabolic pathways, and, paradoxically, mediate cell fate. Evidence has shown mitochondria as points of convergence for diverse cell death-inducing pathways that trigger the various mechanisms underlying apoptotic and nonapoptotic programmed cell death. Thus, dysfunctional cellular pathways eventually lead or contribute to various age-related diseases, such as neurodegenerative, cardiovascular and metabolic diseases. Thus, mitochondrion-associated programmed cell death-based treatments show great therapeutic potential, providing novel insights in clinical trials. This review discusses mitochondrial quality control networks with activity triggered by stimuli and that maintain cellular homeostasis via mitohormesis, the mitochondrial unfolded protein response, and mitophagy. The review also presents details on various forms of mitochondria-associated programmed cell death, including apoptosis, necroptosis, ferroptosis, pyroptosis, parthanatos, and paraptosis, and highlights their involvement in age-related disease pathogenesis, collectively suggesting therapeutic directions for further research.
Collapse
Affiliation(s)
- Thanh T Nguyen
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Shibo Wei
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea
| | - Thu Ha Nguyen
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju, 26426, Republic of Korea
| | - Yunju Jo
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Yan Zhang
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea
| | - Wonyoung Park
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Karim Gariani
- Service of Endocrinology, Diabetes, Nutrition and Patient Therapeutic Education, Geneva University Hospitals, Geneva, 1205, Switzerland
| | - Chang-Myung Oh
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Hyeon Ho Kim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06351, Republic of Korea
| | - Ki-Tae Ha
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Kyu Sang Park
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju, 26426, Republic of Korea
| | - Raekil Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - In-Kyu Lee
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, Republic of Korea
| | - Minho Shong
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - Riekelt H Houtkooper
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
- Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
- Amsterdam Cardiovascular Sciences, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| | - Dongryeol Ryu
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea.
| |
Collapse
|
31
|
Jing W, Chen C, Wang G, Han M, Chen S, Jiang X, Shi C, Sun P, Yang Z, Shi B, Jiang X. Metabolic Modulation of Intracellular Ammonia via Intravesical Instillation of Nanoporter-Encased Hydrogel Eradicates Bladder Carcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206893. [PMID: 36775865 PMCID: PMC10131795 DOI: 10.1002/advs.202206893] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/25/2023] [Indexed: 06/18/2023]
Abstract
Tumor protein 53 (TP53) mutation in bladder carcinoma (BC), upregulates the transcription of carbamoyl phosphate synthetase 1 (CPS1), to reduce intracellular ammonia toxicity. To leverage ammonia combating BC, here, an intravesically perfusable nanoporter-encased hydrogel system is reported. A biomimetic fusogenic liposomalized nanoporter (FLNP) that is decorated with urea transporter-B (UT-B) is first synthesized with protonated chitosan oligosaccharide for bladder tumor-targeted co-delivery of urease and small interfering RNA targeting CPS1 (siCPS1). Mussel-inspired hydrogel featured with dual functions of bio-adhesion and injectability is then fabricated as the reservoir for intravesical immobilization of FLNP. It is found that FLNP-mediated UT-B immobilization dramatically induces urea transportation into tumor cells, and co-delivery of urease and siCPS1 significantly boosts ammonia accumulation in tumor inducing cell apoptosis. Treatment with hybrid system exhibits superior anti-tumor effect in orthotopic bladder tumor mouse model and patient-derived xenograft model, respectively. Combined with high-protein diet, the production of urinary urea increases, leading to an augmented intracellular deposition of ammonia in BC cells, and ultimately an enhanced tumor inhibition. Together, the work establishes that cascade modulation of ammonia in tumor cells could induce tumor apoptosis and may be a practical strategy for eradication of TP53-mutated bladder cancer.
Collapse
Affiliation(s)
- Weiqiang Jing
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Cultural West Road, Jinan, Shandong Province, 250012, China
| | - Chen Chen
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Cultural West Road, Jinan, Shandong Province, 250012, China
| | - Ganyu Wang
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Cultural West Road, Jinan, Shandong Province, 250012, China
| | - Maosen Han
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Cultural West Road, Jinan, Shandong Province, 250012, China
| | - Shouzhen Chen
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Cultural West Road, Jinan, Shandong Province, 250012, China
| | - Xin Jiang
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Cultural West Road, Jinan, Shandong Province, 250012, China
| | - Chongdeng Shi
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Cultural West Road, Jinan, Shandong Province, 250012, China
| | - Peng Sun
- Shandong University of Traditional Chinese Medicine, University Road, Jinan, Shandong Province, 250355, China
| | - Zhenmei Yang
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Cultural West Road, Jinan, Shandong Province, 250012, China
| | - Benkang Shi
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Cultural West Road, Jinan, Shandong Province, 250012, China
| | - Xinyi Jiang
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Cultural West Road, Jinan, Shandong Province, 250012, China
| |
Collapse
|
32
|
Kajihara N, Kobayashi T, Otsuka R, Nio-Kobayashi J, Oshino T, Takahashi M, Imanishi S, Hashimoto A, Wada H, Seino KI. Tumor-derived interleukin-34 creates an immunosuppressive and chemoresistant tumor microenvironment by modulating myeloid-derived suppressor cells in triple-negative breast cancer. Cancer Immunol Immunother 2023; 72:851-864. [PMID: 36104597 DOI: 10.1007/s00262-022-03293-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 09/06/2022] [Indexed: 10/14/2022]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype characterized by a lack of therapeutic targets. The paucity of effective treatment options motivated a number of studies to tackle this problem. Immunosuppressive cells infiltrated into the tumor microenvironment (TME) of TNBC are currently considered as candidates for new therapeutic targets. Myeloid-derived suppressor cells (MDSCs) have been reported to populate in the TME of TNBC, but their roles in the clinical and biological features of TNBC have not been clarified. This study identified that interleukin-34 (IL-34) released by TNBC cells is a crucial immunomodulator to regulate MDSCs accumulation in the TME. We provide evidence that IL-34 induces a differentiation of myeloid stem cells into monocytic MDSCs (M-MDSCs) that recruits regulatory T (Treg) cells, while suppressing a differentiation into polymorphonuclear MDSCs (PMN-MDSCs). As a result, the increase in M-MDSCs contributes to the creation of an immunosuppressive TME, and the decrease in PMN-MDSCs suppresses angiogenesis, leading to an acquisition of resistance to chemotherapy. Accordingly, blockade of M-MDSC differentiation with an estrogen receptor inhibitor or anti-IL-34 monoclonal antibody suppressed M-MDSCs accumulation causing retardation of tumor growth and restores chemosensitivity of the tumor by promoting PMN-MDSCs accumulation. This study demonstrates previously poorly understood mechanisms of MDSCs-mediated chemoresistance in the TME of TNBC, which is originated from the existence of IL-34, suggesting a new rationale for TNBC treatment.
Collapse
Affiliation(s)
- Nabeel Kajihara
- Division of Immunobiology, Graduate School of Medicine, Institute for Genetic Medicine, Hokkaido University, Kita-15 Nishi-7, Kita-ku, Sapporo city, Hokkaido, 060-0815, Japan
| | - Takuto Kobayashi
- Division of Immunobiology, Graduate School of Medicine, Institute for Genetic Medicine, Hokkaido University, Kita-15 Nishi-7, Kita-ku, Sapporo city, Hokkaido, 060-0815, Japan
| | - Ryo Otsuka
- Division of Immunobiology, Graduate School of Medicine, Institute for Genetic Medicine, Hokkaido University, Kita-15 Nishi-7, Kita-ku, Sapporo city, Hokkaido, 060-0815, Japan
| | - Junko Nio-Kobayashi
- Laboratory of Histology and Cytology, Graduate School of Medicine, Hokkaido University, Kita-15 Nishi-7, Kita-ku, Sapporo city, Hokkaido, 060-8638, Japan
| | - Tomohiro Oshino
- Department of Breast Surgery, Hokkaido University Hospital, Kita-14 Nishi-5, Kita-ku, Sapporo city, Hokkaido, 060-8648, Japan
| | - Masato Takahashi
- Department of Breast Surgery, Hokkaido University Hospital, Kita-14 Nishi-5, Kita-ku, Sapporo city, Hokkaido, 060-8648, Japan
| | - Seiichi Imanishi
- Department of Breast Surgery, Osaka Rosai Hospital, Nagasone-cho 1179-3, Kita-ku, Sakai city, Osaka, 591-8025, Japan
| | - Ari Hashimoto
- Department of Molecular Biology, Faculty of Medicine, Hokkaido University, Kita-15 Nishi-7, Kita-ku, Sapporo city, Hokkaido, 060-8638, Japan
| | - Haruka Wada
- Division of Immunobiology, Graduate School of Medicine, Institute for Genetic Medicine, Hokkaido University, Kita-15 Nishi-7, Kita-ku, Sapporo city, Hokkaido, 060-0815, Japan
| | - Ken-Ichiro Seino
- Division of Immunobiology, Graduate School of Medicine, Institute for Genetic Medicine, Hokkaido University, Kita-15 Nishi-7, Kita-ku, Sapporo city, Hokkaido, 060-0815, Japan.
| |
Collapse
|
33
|
Abstract
Phosphatases and kinases maintain an equilibrium of dephosphorylated and phosphorylated proteins, respectively, that are required for critical cellular functions. Imbalance in this equilibrium or irregularity in their function causes unfavorable cellular effects that have been implicated in the development of numerous diseases. Protein tyrosine phosphatases (PTPs) catalyze the dephosphorylation of protein substrates on tyrosine residues, and their involvement in cell signaling and diseases such as cancer and inflammatory and metabolic diseases has made them attractive therapeutic targets. However, PTPs have proved challenging in therapeutics development, garnering them the unfavorable reputation of being undruggable. Nonetheless, great strides have been made toward the inhibition of PTPs over the past decade. Here, we discuss the advancement in small-molecule inhibition for the PTP subfamily known as the mitogen-activated protein kinase (MAPK) phosphatases (MKPs). We review strategies and inhibitor discovery tools that have proven successful for small-molecule inhibition of the MKPs and discuss what the future of MKP inhibition potentially might yield.
Collapse
Affiliation(s)
- Shanelle R Shillingford
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA;
- Department of Chemistry, Yale University, New Haven, Connecticut, USA
| | - Anton M Bennett
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA;
- Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
34
|
Chen H, Zhou H, Yang J, Wan H, He Y. Guhong injection mitigates myocardial ischemia/reperfusion injury by activating GST P to inhibit ASK1-JNK/p38 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 109:154603. [PMID: 36610111 DOI: 10.1016/j.phymed.2022.154603] [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: 10/11/2022] [Revised: 11/30/2022] [Accepted: 12/12/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Guhong injection (GHI), a novel compound preparation that is composed of a chemical drug, namely aceglutamide, and the aqueous extract of safflower (Carthamus tinctorius L.), exhibits extreme antioxidative, antiapoptotic, anti-inflammatory, and neuroprotective effects. Since oxidative stress, apoptosis, and inflammatory response are all the dominant mechanisms of myocardial ischemia/reperfusion (MI/R) injury, we probe into the protective mechanism of GHI on MI/R injury for the first time. METHODS In this research, we first employed molecular docking to determine whether three active ingredients in GHI, acetylglutamine (NAG), hydroxysafflor yellow A (HSYA), and syringin, possessed the potential activity to modulate the protein, glutathione S-transferase P (GST P). We further identified the protective effect of GHI on myocardial tissue with TTC staining, HE staining, TUNEL staining, and ELISA, and on H9c2 with flow cytometry and ELISA. We next explored whether the cardioprotective effect of GHI on left anterior descending ligation-reperfusion in rats and hypoxia/reoxygenation (H/R) in H9c2 cells was related to activate GST P to inhibit ASK1-JNK/p38 pathway via approaches of qRT-PCR and Western blot. RESULTS Results of molecular docking indicated that all three compounds spontaneously docked to GST P, among them the binding affinities of both HSYA and syringin to GST P were higher than NAG. In vivo, GHI reduced myocardial infarction size and mitigated myocardial pathological injury. In vitro, GHI enhanced cell viability and extenuated depolarization of mitochondrial membrane potential. In addition, the results of in vivo and in vitro studies demonstrated that the cardioprotection of GHI was associated with improving the mRNA and protein expression levels of GST P to modulate oxidative stress, and inhibiting the levels of mRNA expression and protein phosphorylation of ASK1, JNK, and p38. However, the suppressed effect of GHI on ASK1-JNK/p38 pathway was reversed by ethacrynic acid (EA, a GST inhibitor), indicating that the regulation of GHI on ASK1-JNK/p38 was related to the activity of GST P. Besides, the in vitro results of qRT-PCR and western-blot also certified that the inhibited JNK and p38 further reduced Bax expression and elevated Bcl-2 expression to reduce the expression of caspase-3 to exert anti-apoptosis effects. CONCLUSION Taken together, the cardioprotection of GHI mainly incarnated in activating GST P to relieve oxidation properties, thereby inhibiting ASK1-JNK/p38 pathway to suppress apoptosis.
Collapse
Affiliation(s)
- Haiyang Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Huifen Zhou
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Jiehong Yang
- School of Basic Medicine Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Haitong Wan
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| | - Yu He
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| |
Collapse
|
35
|
Effendy WNFWE, S. M. N. Mydin RB, Gazzali AM, Sreekantan S. Localised Delivery of Cisplatin from Chitosan-Coated Titania Nanotube Array Nanosystems Targeting Nasopharyngeal Carcinoma. Adv Pharm Bull 2023; 13:104-112. [PMID: 36721810 PMCID: PMC9871279 DOI: 10.34172/apb.2023.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 10/01/2021] [Accepted: 12/31/2021] [Indexed: 02/03/2023] Open
Abstract
Pupose: Cisplatin (CDDP), while amongst the recognised chemotherapeutic drugs currently available, is known to have limitations; the lack of a single treatment approach and non-specific targeted therapies. Therefore, the development of an innovative strategy that could achieve localised CDDP treatment is an urgent undertaking. Recent advances in titania nanotube arrays (TNAs) technology have demonstrated promising applications for localised chemotherapeutic drug treatment. The present work investigated the efficiency of a TNA nanosystem for the localised CDDP treatment of nasopharyngeal carcinoma (NPC). Methods: Two models of the TNA nanosystem were prepared: CDDP loaded onto the TNA nanosystem surface (CDDP-TNA) and the other consisted of chitosan-coated CDDP-TNA. CDDP release from these two nanosystems was comprehensively tested on the NPC cells NPC/HK-1 and C666-1. The NPC cytotoxicity profile of the two CDDP-TNA nanosystems was evaluated after incubation for 24, 48 and 72 hours. Intracellular damage profiles were studied using fluorescence microscopy analysis with Hoechst 33342, acridine orange and propidium iodide. Results: The half-maximal inhibitory concentrations (IC50) of CDDP at 24 hours were 0.50 mM for NPC/HK-1 and 0.05 mM for C666-1. CDDP in the CDDP-TNA and chitosan-coated CDDPTNA models presented a significant degree of NPC inhibition (P<0.05) after 24, 48 and 72 hours of exposure. The outcome revealed cellular damage and shrinkage of the cell membranes after 48 hours of exposure to CDDP-TNA. Conclusion: This in vitro work demonstrated the effectiveness of TNA nanosystems for the localised CDDP treatment of NPC cells. Further in vivo studies are needed to support the findings.
Collapse
Affiliation(s)
| | - Rabiatul Basria S. M. N. Mydin
- Department of Biomedical Science, Advanced Medical and Dental Institute, Universiti Sains Malaysia, 13200 Bertam, Kepala Batas, Pulau Pinang, Malaysia.,Corresponding Author: Rabiatul Basria S. M. N. Mydin,
| | - Amirah Mohd Gazzali
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800, Minden, Pulau Pinang, Malaysia
| | - Srimala Sreekantan
- School of Materials and Mineral Resources Engineering, Universiti Sains Malaysia, Engineering Campus, 14300, Nibong Tebal, Pulau Pinang, Malaysia
| |
Collapse
|
36
|
A novel site on dual-specificity phosphatase MKP7/DUSP16 is required for catalysis and MAPK binding. J Biol Chem 2022; 298:102617. [PMID: 36272649 PMCID: PMC9676401 DOI: 10.1016/j.jbc.2022.102617] [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: 02/25/2022] [Revised: 10/06/2022] [Accepted: 10/11/2022] [Indexed: 11/18/2022] Open
Abstract
The dual-specificity phosphatases responsible for the inactivation of the mitogen-activated protein kinases (MAPKs) are designated as the MAPK phosphatases (MKPs). We demonstrated previously that MKP5 is regulated through a novel allosteric site suggesting additional regulatory mechanisms of catalysis exist amongst the MKPs. Here, we sought to determine whether the equivalent site within the phosphatase domain of a highly similar MKP family member, MKP7, is also important for phosphatase function. We found that mutation of tyrosine 271 (Y271) in MKP7, which represents the comparable Y435 within the MKP5 allosteric pocket, inhibited MKP7 catalytic activity. Consistent with this, when MKP7 Y271 mutants were overexpressed in cells, the substrates of MKP7, p38 MAPK or JNK, failed to undergo dephosphorylation. The binding efficiency of MKP7 to p38 MAPK and JNK1/2 was also reduced when MKP7 Y271 is mutated. Consistent with reduced MAPK binding, we observed a greater accumulation of nuclear p38 MAPK and JNK when the MKP7 Y271 mutants are expressed in cells as compared with WT MKP7, which sequesters p38 MAPK/JNK in the cytoplasm. Therefore, we propose that Y271 is critical for effective MAPK dephosphorylation through a mechanism whereby binding to this residue precedes engagement of the catalytic site and upon overexpression, MKP7 allosteric site mutants potentiate MAPK signaling. These results provide insight into the regulatory mechanisms of MKP7 catalysis and interactions with the MAPKs. Furthermore, these data support the generality of the MKP allosteric site and provide a basis for small molecule targeting of MKP7.
Collapse
|
37
|
Tang X, Liang Y, Sun G, He Q, Hou Z, Jiang X, Gao P, Qu H. Upregulation of CRABP2 by TET1-mediated DNA hydroxymethylation attenuates mitochondrial apoptosis and promotes oxaliplatin resistance in gastric cancer. Cell Death Dis 2022; 13:848. [PMID: 36195596 PMCID: PMC9532395 DOI: 10.1038/s41419-022-05299-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 09/22/2022] [Accepted: 09/26/2022] [Indexed: 11/07/2022]
Abstract
Oxaliplatin is the main chemotherapy drug for gastric cancer (GC), but quite a few patients are resistant to oxaliplatin, which contributes to the poor prognosis of GC patients. There is therefore an urgent need to identify potential targets for reversing chemotherapy resistance in GC patients. In this study, we analyzed the tumor samples of GC patients who received neoadjuvant chemotherapy based on oxaliplatin through quantitative proteomics and identified the potential chemoresistance-related protein cellular retinoic acid binding protein 2 (CRABP2). CRABP2 was significantly upregulated in the tumor tissues of chemoresistant GC patients and was closely related to prognosis. The results of cell function experiments showed that CRABP2 can promote the oxaliplatin resistance of GC cells in vitro. Coimmunoprecipitation and GST pulldown assays showed that CRAPB2 expedited the binding of BAX and PARKIN in GC cells and facilitated the ubiquitination-mediated degradation of BAX. Furthermore, both the in vitro assay and cell-derived xenograft (CDX) in vivo model verified that CRABP2 promoted oxaliplatin resistance by inhibiting BAX-dependent cell apoptosis. Further experiments proved that the abnormally high expression of CRABP2 in oxaliplatin-resistant GC cells was affected by TET1-mediated DNA hydroxymethylation. The patient-derived xenograft (PDX) model suggested that interference with CRABP2 reversed oxaliplatin resistance in GC in vivo. In conclusion, the results of our study show that CRABP2 was a key molecule in oxaliplatin resistance regulation and could be a new target for reversing the chemoresistance of GC.
Collapse
Affiliation(s)
- Xiaolong Tang
- grid.452402.50000 0004 1808 3430Department of General Surgery, Qilu Hospital of Shandong University, Jinan, 250012 China
| | - Yahang Liang
- grid.452402.50000 0004 1808 3430Department of General Surgery, Qilu Hospital of Shandong University, Jinan, 250012 China
| | - Guorui Sun
- grid.452402.50000 0004 1808 3430Department of General Surgery, Qilu Hospital of Shandong University, Jinan, 250012 China
| | - Qingsi He
- grid.452402.50000 0004 1808 3430Department of General Surgery, Qilu Hospital of Shandong University, Jinan, 250012 China
| | - Zhenyu Hou
- grid.452402.50000 0004 1808 3430Department of General Surgery, Qilu Hospital of Shandong University, Jinan, 250012 China
| | - Xingzhi Jiang
- grid.452402.50000 0004 1808 3430Department of General Surgery, Qilu Hospital of Shandong University, Jinan, 250012 China
| | - Peng Gao
- grid.452402.50000 0004 1808 3430Department of Pathology, Qilu Hospital of Shandong University, Jinan, 250012 China
| | - Hui Qu
- grid.452402.50000 0004 1808 3430Department of General Surgery, Qilu Hospital of Shandong University, Jinan, 250012 China
| |
Collapse
|
38
|
CAF-derived exosomal WEE2-AS1 facilitates colorectal cancer progression via promoting degradation of MOB1A to inhibit the Hippo pathway. Cell Death Dis 2022; 13:796. [PMID: 36123327 PMCID: PMC9485119 DOI: 10.1038/s41419-022-05240-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 01/22/2023]
Abstract
Cancer-associated fibroblasts (CAFs) are the most abundant stromal components in the tumor microenvironment (TME) and closely involved in tumor progression. However, the precise biological functions and molecular mechanisms of CAFs in the TME have yet to be understood. Here, we demonstrate that WEE2-AS1 is highly expressed in the CAF-derived small extracellular vesicles (sEVs). Moreover, WEE2-AS1 is markedly higher in plasma sEVs of CRC patients than in healthy subjects and its high level predicts advanced pathological staging and poor survival. Then, we conducted a series of in vitro and in vivo experiments. Elevated expression of WEE2-AS1 in sEVs increases CRC cell proliferation in vitro. Importantly, aberrant CAF-sEVsWEE2-AS1 leads to tumor formation and progression in BALB/c nude mice and promotes AOM/DSS-induced tumorigenesis. Mechanistically, WEE2-AS1 functions as a modular scaffold for the MOB1A and E3 ubiquitin-protein ligase praja2 complexes, leading to MOB1A degradation via the ubiquitin-proteasome pathway. The Hippo pathway is then inhibited and more YAP are transported into the nucleus, where they activate downstream gene transcription. Together, our data reveal that CAF-sEVsWEE2-AS1 interacts with MOB1A, promotes degradation of MOB1A, inhibits the Hippo pathway, and facilitates the growth of CRC cells. Hence, exosomal WEE2-AS1 may be a promising therapeutic target and circulating biomarker for CRC diagnosis and prognosis.
Collapse
|
39
|
Yang EY, Howard GR, Brock A, Yankeelov TE, Lorenzo G. Mathematical characterization of population dynamics in breast cancer cells treated with doxorubicin. Front Mol Biosci 2022; 9:972146. [PMID: 36172049 PMCID: PMC9510895 DOI: 10.3389/fmolb.2022.972146] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/17/2022] [Indexed: 11/20/2022] Open
Abstract
The development of chemoresistance remains a significant cause of treatment failure in breast cancer. We posit that a mathematical understanding of chemoresistance could assist in developing successful treatment strategies. Towards that end, we have developed a model that describes the cytotoxic effects of the standard chemotherapeutic drug doxorubicin on the MCF-7 breast cancer cell line. We assume that treatment with doxorubicin induces a compartmentalization of the breast cancer cell population into surviving cells, which continue proliferating after treatment, and irreversibly damaged cells, which gradually transition from proliferating to treatment-induced death. The model is fit to experimental data including variations in drug concentration, inter-treatment interval, and number of doses. Our model recapitulates tumor cell dynamics in all these scenarios (as quantified by the concordance correlation coefficient, CCC > 0.95). In particular, superior tumor control is observed with higher doxorubicin concentrations, shorter inter-treatment intervals, and a higher number of doses (p < 0.05). Longer inter-treatment intervals require adapting the model parameterization after each doxorubicin dose, suggesting the promotion of chemoresistance. Additionally, we propose promising empirical formulas to describe the variation of model parameters as functions of doxorubicin concentration (CCC > 0.78). Thus, we conclude that our mathematical model could deepen our understanding of the cytotoxic effects of doxorubicin and could be used to explore practical drug regimens achieving optimal tumor control.
Collapse
Affiliation(s)
- Emily Y. Yang
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX, United States
| | - Grant R. Howard
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States
| | - Amy Brock
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States
- Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
- Interdisciplinary Life Sciences Program, The University of Texas at Austin, Austin, TX, United States
| | - Thomas E. Yankeelov
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX, United States
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States
- Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
- Department of Diagnostic Medicine, The University of Texas at Austin, Austin, TX, United States
- Department of Oncology, The University of Texas at Austin, Austin, TX, United States
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Guillermo Lorenzo
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX, United States
- Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy
| |
Collapse
|
40
|
Wu D, Zhao L, Sui B, Tan L, Lu L, Mao X, Liao G, Shi S, Cao Y, Yang X, Kou X. An Appearance Data-Driven Model Visualizes Cell State and Predicts Mesenchymal Stem Cell Regenerative Capacity. SMALL METHODS 2022; 6:e2200087. [PMID: 35674483 DOI: 10.1002/smtd.202200087] [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: 01/20/2022] [Revised: 04/14/2022] [Indexed: 06/15/2023]
Abstract
Mesenchymal stem cells (MSCs) are widely used in treating various diseases. However, lack of a reliable evaluation approach to characterize the potency of MSCs has dampened their clinical applications. Here, a function-oriented mathematical model is established to evaluate and predict the regenerative capacity (RC) of MSCs. Processed by exhaustive testing, the model excavates four optimal fitted indices, including nucleus roundness, nucleus/cytoplasm ratio, side-scatter height, and ERK1/2 from the given index combinations. Notably, three of them except ERK1/2 are cell appearance-associated features. The predictive power of the model is validated via screening experiments of these indices by predicting the RC of newly enrolled and chemical inhibitor-treated MSCs. Further RNA-sequencing analysis reveals that cell appearance-based indices may serve as major indicators to visualize the results of integration-weighted signals in and out of cells and reflect MSC stemness. In general, this study proposes an appearance data-driven predictive model for the RC and stemness of MSCs.
Collapse
Affiliation(s)
- Di Wu
- Hospital of Stomatology, Guanghua School of Stomatology, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
- Hospital of Stomatology, Guanghua School of Stomatology, Department of Oral and Maxillofacial Surgery, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
- Hospital of Stomatology, Guanghua School of Stomatology, Department of Orthodontics, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
| | - Lu Zhao
- Hospital of Stomatology, Guanghua School of Stomatology, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
| | - Bingdong Sui
- Hospital of Stomatology, Guanghua School of Stomatology, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Lingping Tan
- Hospital of Stomatology, Guanghua School of Stomatology, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
| | - Lu Lu
- Hospital of Stomatology, Guanghua School of Stomatology, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
| | - Xueli Mao
- Hospital of Stomatology, Guanghua School of Stomatology, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
| | - Guiqing Liao
- Hospital of Stomatology, Guanghua School of Stomatology, Department of Oral and Maxillofacial Surgery, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
| | - Songtao Shi
- Hospital of Stomatology, Guanghua School of Stomatology, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
- Key Laboratory of Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yang Cao
- Hospital of Stomatology, Guanghua School of Stomatology, Department of Orthodontics, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
| | - Xiaobao Yang
- School of Physics and Optoelectronics, South China University of Technology, Guangzhou, 510640, China
| | - Xiaoxing Kou
- Hospital of Stomatology, Guanghua School of Stomatology, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
- Key Laboratory of Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China
| |
Collapse
|
41
|
Liu JY, Zou T, Yin JY, Wang Z, Liu C, Huang HX, Ding FX, Lei MR, Wang Y, Liu M, Liu ZQ, Tan LM, Chen J. Genetic Variants in Double-Strand Break Repair Pathway Genes to Predict Platinum-Based Chemotherapy Prognosis in Patients With Lung Cancer. Front Pharmacol 2022; 13:915822. [PMID: 35899106 PMCID: PMC9309806 DOI: 10.3389/fphar.2022.915822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/30/2022] [Indexed: 11/21/2022] Open
Abstract
Objective: The purpose of this study was to investigate the associations of genetic variants in double-strand break (DSB) repair pathway genes with prognosis in patients with lung cancer treated with platinum-based chemotherapy. Methods: Three hundred ninety-nine patients with lung cancer who received platinum-based chemotherapy for at least two cycles were included in this study. A total of 35 single nucleotide polymorphisms (SNPs) in DSB repair, base excision repair (BER), and nucleotide excision repair (NER) repair pathway genes were genotyped, and were used to evaluate the overall survival (OS) and the progression-free survival (PFS) of patients who received platinum-based chemotherapy using Cox proportional hazard models. Results: The PFS of patients who carried the MAD2L2 rs746218 GG genotype was shorter than that in patients with the AG or AA genotypes (recessive model: p = 0.039, OR = 5.31, 95% CI = 1.09–25.93). Patients with the TT or GT genotypes of TNFRSF1A rs4149570 had shorter OS times than those with the GG genotype (dominant model: p = 0.030, OR = 0.57, 95% CI = 0.34–0.95). We also investigated the influence of age, gender, histology, smoking, stage, and metastasis in association between SNPs and OS or PFS in patients with lung cancer. DNA repair gene SNPs were significantly associated with PFS and OS in the subgroup analyses. Conclusion: Our study showed that variants in MAD2L2 rs746218 and TNFRSF1A rs4149570 were associated with shorter PFS or OS in patients with lung cancer who received platinum-based chemotherapy. These variants may be novel biomarkers for the prediction of prognosis of patients with lung cancer who receive platinum-based chemotherapy.
Collapse
Affiliation(s)
- Jun-Yan Liu
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Ting Zou
- National Institution of Drug Clinical Trial, Xiangya Hospital, Central South University, Changsha, China
| | - Ji-Ye Yin
- Departments of Clinical Pharmacology, Xinagya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacology and Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China
| | - Zhan Wang
- Lung Cancer and Gastrointestinal Unit, Department of Medical Oncology, Hunan Cancer Hospital, Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha, China
| | - Chong Liu
- Institute of Clinical Pharmacology and Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China
| | - Han-Xue Huang
- Institute of Clinical Pharmacology and Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China
| | - Fei-Xiang Ding
- Institute of Clinical Pharmacology and Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China
| | - Meng-Rong Lei
- Institute of Clinical Pharmacology and Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China
| | - Ying Wang
- Hunan Clinical Research Center in Gynecologic Cancer, Hunan Cancer Hospital, Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha, China
| | - Min Liu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Zhao-Qian Liu
- National Institution of Drug Clinical Trial, Xiangya Hospital, Central South University, Changsha, China
- Departments of Clinical Pharmacology, Xinagya Hospital, Central South University, Changsha, China
| | - Li-Ming Tan
- Department of Pharmacy, The Second People's Hospital of Huaihua City, Huaihua, China
| | - Juan Chen
- Department of Pharmacy, Xinagya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
42
|
Ramalingam V, Narendra Kumar N, Harshavardhan M, Sampath Kumar HM, Tiwari AK, Suresh Babu K, Mudiam MKR. Chemical profiling of marine seaweed Halimeda gracilis using UPLC-ESI-Q-TOF-MSE and evaluation of anticancer activity targeting PI3K/AKT and intrinsic apoptosis signaling pathway. Food Res Int 2022; 157:111394. [DOI: 10.1016/j.foodres.2022.111394] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 12/24/2022]
|
43
|
Mei Y, Feng X, Feng T, Yan M, Zhu Z, Li T, Zhu Z. Adjuvant Chemotherapy in pT2N0M0 Gastric Cancer: Findings From a Retrospective Study. Front Pharmacol 2022; 13:845261. [PMID: 35250596 PMCID: PMC8891981 DOI: 10.3389/fphar.2022.845261] [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: 12/29/2021] [Accepted: 02/04/2022] [Indexed: 11/13/2022] Open
Abstract
Background: There is no global consensus on adjuvant chemotherapy (ACT) for pT2N0M0 gastric cancer. We conducted a retrospective study to reveal the role of ACT in such patients. Methods: Patients with pT2N0M0 gastric cancer who underwent radical resection with D2 lymphadenectomy for primary gastric cancer between January 2012 and May 2016 were included. Kaplan-Meier and Cox regression were used to evaluate overall survival (OS), disease-specific survival (DSS) and predictors of prognosis. Stratified analysis based on high-risk factors was conducted. Results: Of enrolled 307 patients, 111 patients underwent surgery alone and 196 patients received ACT. Surgery alone (HR = 2.913, 95% CI: 1.494-5.682, p = 0.002) and total gastrectomy (HR = 2.445, 95% CI: 1.279-4.675, p = 0.007) were independently associated with decreased OS. With the median follow-up of 73.1 months, the 5-year OS rate was 87.9% and 5-year DSS rate was 91.8%. Patients receiving ACT showed a better 5-year OS rate (92.9 vs. 79.3%, p < 0.001) and DSS rate (96.8 vs. 83.0%, p < 0.001) than patients underwent surgery alone. Patients receiving monotherapy (n = 130) had a relatively poor prognosis compared to patients receiving dual-drug (n = 66) without a significant difference (92.3 vs. 93.9%, p = 0.637). In patients without high-risk factors based on the Chinese Society of Clinical Oncology (CSCO) Guidelines, ACT also provided survival benefit (96.0 vs 82.9%, p = 0.038). Conclusions: ACT was accompanied with higher 5-year OS and DSS rates of patients with pT2N0M0 gastric cancer. Patients with pT2N0M0 gastric cancer, regardless of high-risk factors based on the CSCO guidelines, might be considered candidates for ACT. In regard to the therapy regimen, monotherapy might be the optimal choice, considering the adverse events.
Collapse
Affiliation(s)
- Yu Mei
- Department of General Surgery, Gastrointestinal Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xijia Feng
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tienan Feng
- Clinical Research Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Yan
- Department of General Surgery, Gastrointestinal Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenggang Zhu
- Department of General Surgery, Gastrointestinal Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Zhenglun Zhu
- Department of General Surgery, Gastrointestinal Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
44
|
Labadie JD, Savas S, Harrison TA, Banbury B, Huang Y, Buchanan DD, Campbell PT, Gallinger SJ, Giles GG, Gunter MJ, Hoffmeister M, Hsu L, Jenkins MA, Lin Y, Ogino S, Phipps AI, Slattery ML, Steinfelder RS, Sun W, Van Guelpen B, Hua X, Figuieredo JC, Pai RK, Nassir R, Qi L, Chan AT, Peters U, Newcomb PA. Genome-wide association study identifies tumor anatomical site-specific risk variants for colorectal cancer survival. Sci Rep 2022; 12:127. [PMID: 34996992 PMCID: PMC8741984 DOI: 10.1038/s41598-021-03945-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 12/06/2021] [Indexed: 12/16/2022] Open
Abstract
Identification of new genetic markers may improve the prediction of colorectal cancer prognosis. Our objective was to examine genome-wide associations of germline genetic variants with disease-specific survival in an analysis of 16,964 cases of colorectal cancer. We analyzed genotype and colorectal cancer-specific survival data from a consortium of 15 studies. Approximately 7.5 million SNPs were examined under the log-additive model using Cox proportional hazards models, adjusting for clinical factors and principal components. Additionally, we ran secondary analyses stratifying by tumor site and disease stage. We used a genome-wide p-value threshold of 5 × 10-8 to assess statistical significance. No variants were statistically significantly associated with disease-specific survival in the full case analysis or in the stage-stratified analyses. Three SNPs were statistically significantly associated with disease-specific survival for cases with tumors located in the distal colon (rs698022, HR = 1.48, CI 1.30-1.69, p = 8.47 × 10-9) and the proximal colon (rs189655236, HR = 2.14, 95% CI 1.65-2.77, p = 9.19 × 10-9 and rs144717887, HR = 2.01, 95% CI 1.57-2.58, p = 3.14 × 10-8), whereas no associations were detected for rectal tumors. Findings from this large genome-wide association study highlight the potential for anatomical-site-stratified genome-wide studies to identify germline genetic risk variants associated with colorectal cancer-specific survival. Larger sample sizes and further replication efforts are needed to more fully interpret these findings.
Collapse
Affiliation(s)
- Julia D Labadie
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Sevtap Savas
- Discipline of Genetics, Faculty of Medicine, Memorial University, St. John's, NL, Canada
- Discipline of Oncology, Faculty of Medicine, Memorial University, St. John's, NL, Canada
| | - Tabitha A Harrison
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Barb Banbury
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Yuhan Huang
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Daniel D Buchanan
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia
- Colorectal Oncogenomics Group, Genetic Epidemiology Laboratory, Department of Pathology, The University of Melbourne, Parkville, VIC, Australia
- Genetic Medicine and Family Cancer Clinic, The Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Peter T Campbell
- Department of Population Science, American Cancer Society, Atlanta, GA, USA
| | - Steven J Gallinger
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, ON, Canada
| | - Graham G Giles
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, VIC, Australia
- Medicine, School of Clinical Sciences at Monash Health, Monash University, VIC, Australia
| | - Marc J Gunter
- Nutrition and Metabolism Section, International Agency for Research On Cancer, World Health Organization, Lyon, France
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Li Hsu
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Mark A Jenkins
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Yi Lin
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Shuji Ogino
- Program in Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Cancer Immunology Program, Dana-Farber Harvard Cancer Center, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Amanda I Phipps
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Martha L Slattery
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Robert S Steinfelder
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Wei Sun
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Bethany Van Guelpen
- Department of Radiation Sciences, Oncology Unit, Umeå University, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| | - Xinwei Hua
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Jane C Figuieredo
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Rish K Pai
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Rami Nassir
- Department of Pathology, School of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Lihong Qi
- Department of Public Health Sciences, University of California Davis, Davis, CA, USA
| | - Andrew T Chan
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Ulrike Peters
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Polly A Newcomb
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
- Department of Epidemiology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
45
|
Zhu M, Zhang P, Yu S, Tang C, Wang Y, Shen Z, Chen W, Liu T, Cui Y. Targeting ZFP64/GAL-1 axis promotes therapeutic effect of nab-paclitaxel and reverses immunosuppressive microenvironment in gastric cancer. J Exp Clin Cancer Res 2022; 41:14. [PMID: 34996504 PMCID: PMC8740411 DOI: 10.1186/s13046-021-02224-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 12/13/2021] [Indexed: 01/03/2023] Open
Abstract
Background Chemoresistance is a main obstacle in gastric cancer (GC) treatment, but its molecular mechanism still needs to be elucidated. Here, we aim to reveal the underlying mechanisms of nanoparticle albumin-bound paclitaxel (nab-paclitaxel) resistance in GC. Methods We performed RNA sequencing (RNA-seq) on samples from patients who were resistant or sensitive to nab-paclitaxel, and identified Zinc Finger Protein 64 (ZFP64) as critical for nab-paclitaxel resistance in GC. CCK8, flow cytometry, TUNEL staining, sphere formation assays were performed to investigate the effects of ZFP64 in vitro, while subcutaneous tumor formation models were established in nude mice or humanized mice to evaluate the biological roles of ZFP64 in vivo. Chromatin immunoprecipitation sequencing (CHIP-seq) and double-luciferase reporter gene assay were conducted to reveal the underlying mechanism of ZFP64. Results ZFP64 overexpression was linked with aggressive phenotypes, nab-paclitaxel resistance and served as an independent prognostic factor in GC. As a transcription factor, ZFP64 directly binds to Galectin-1 (GAL-1) promoter and promoted GAL-1 transcription, thus inducing stem-cell like phenotypes and immunosuppressive microenvironment in GC. Importantly, compared to treatment with nab-paclitaxel alone, nab-paclitaxel plus GAL-1 blockade significantly enhanced the anti-tumor effect in mouse models, particularly in humanized mice. Conclusions Our data support a pivotal role for ZFP64 in GC progression by simultaneously promoting cellular chemotherapy resistance and tumor immunosuppression. Treatment with the combination of nab-paclitaxel and a GAL-1 inhibitor might benefit a subgroup of GC patients. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-02224-x.
Collapse
Affiliation(s)
- Mengxuan Zhu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.,Center of Evidence-based Medicine, Fudan University, Shanghai, China
| | - Pengfei Zhang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.,Center of Evidence-based Medicine, Fudan University, Shanghai, China
| | - Shan Yu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Cheng Tang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yan Wang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhenbin Shen
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Weidong Chen
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tianshu Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China. .,Center of Evidence-based Medicine, Fudan University, Shanghai, China.
| | - Yuehong Cui
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China. .,Center of Evidence-based Medicine, Fudan University, Shanghai, China.
| |
Collapse
|
46
|
Posch F, Prinz F, Balihodzic A, Mayr C, Kiesslich T, Klec C, Jonas K, Barth DA, Riedl JM, Gerger A, Pichler M. MiR-200c-3p Modulates Cisplatin Resistance in Biliary Tract Cancer by ZEB1-Independent Mechanisms. Cancers (Basel) 2021; 13:cancers13163996. [PMID: 34439151 PMCID: PMC8392278 DOI: 10.3390/cancers13163996] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 08/03/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Biliary tract cancer is a rare malignancy with poor overall survival. The majority of patients are faced with advanced disease stage. Cisplatin-based treatment schedules represent the mainstay of first-line therapeutic strategy, yet only a small portion of patients develop a treatment response. One of the main reasons is acquired drug resistance. Previous studies correlated certain microRNAs (miRNAs), including miR-200c-3p, with drug resistance in various cancer types. However, limited knowledge exists about miR-200c-3p expression and cisplatin resistance in biliary tract cancer. Thus, the main aim of this study was to investigate the influence of miR-200c-3p on the cisplatin resistance in this cancer entity. We demonstrated that miR-200c-3p contributes to cisplatin resistance independently of its known influence on ZEB1 expression. Abstract Biliary tract cancer is a major global health issue in cancer-related mortality. Therapeutic options are limited, and cisplatin-based treatment schedules represent the mainstay of first-line therapeutic strategies. Although the gain of survival by the addition of cisplatin to gemcitabine is moderate, acquired cisplatin resistance frequently leads to treatment failures with mechanisms that are still poorly understood. Epithelial–mesenchymal transition (EMT) is a dynamic process that changes the shape, function, and gene expression pattern of biliary tract cancer cells. In this study, we explored the influence of the EMT-regulating miR-200c-3p on cisplatin sensitivity in biliary tract cancer cells. Using gain of function experiments, we demonstrated that miR-200c-3p regulates epithelial cell markers through the downregulation of the transcription factor ZEB1. MiR-200c-3p upregulation led to a decreased sensitivity against cisplatin, as observed in transient overexpression models as well as in cell lines stably overexpressing miR-200c-3p. The underlying mechanism seems to be independent of miR-200c-3p’s influence on ZEB1 expression, as ZEB1 knockdown resulted in the opposite effect on cisplatin resistance, which was abolished when ZEB1 knockdown and miR-200c-3p overexpression occurred in parallel. Using a gene panel of 40 genes that were previously associated with cisplatin resistance, two (Dual Specificity Phosphatase 16 (DUSP16) and Stratifin (SFN)) were identified as significantly (>2 fold, p-value < 0.05) up-regulated in miR-200c-3p overexpressing cells. In conclusion, miR-200c-3p might be an important contributor to cisplatin resistance in biliary tract cancer, independently of its interaction with ZEB1.
Collapse
Affiliation(s)
- Florian Posch
- Department of Internal Medicine, Division of Oncology, Medical University of Graz, 8036 Graz, Austria; (F.P.); (F.P.); (A.B.); (C.K.); (K.J.); (D.A.B.); (J.M.R.); (A.G.)
| | - Felix Prinz
- Department of Internal Medicine, Division of Oncology, Medical University of Graz, 8036 Graz, Austria; (F.P.); (F.P.); (A.B.); (C.K.); (K.J.); (D.A.B.); (J.M.R.); (A.G.)
- Research Unit “Non-Coding RNAs and Genome Editing in Cancer”, Division of Oncology, Medical University of Graz, 8036 Graz, Austria
| | - Amar Balihodzic
- Department of Internal Medicine, Division of Oncology, Medical University of Graz, 8036 Graz, Austria; (F.P.); (F.P.); (A.B.); (C.K.); (K.J.); (D.A.B.); (J.M.R.); (A.G.)
- Research Unit “Non-Coding RNAs and Genome Editing in Cancer”, Division of Oncology, Medical University of Graz, 8036 Graz, Austria
| | - Christian Mayr
- Institute of Physiology and Pathophysiology, Paracelsus Medical University, 5020 Salzburg, Austria; (C.M.); (T.K.)
- Department of Internal Medicine I, University Clinics Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Tobias Kiesslich
- Institute of Physiology and Pathophysiology, Paracelsus Medical University, 5020 Salzburg, Austria; (C.M.); (T.K.)
- Department of Internal Medicine I, University Clinics Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Christiane Klec
- Department of Internal Medicine, Division of Oncology, Medical University of Graz, 8036 Graz, Austria; (F.P.); (F.P.); (A.B.); (C.K.); (K.J.); (D.A.B.); (J.M.R.); (A.G.)
- Research Unit “Non-Coding RNAs and Genome Editing in Cancer”, Division of Oncology, Medical University of Graz, 8036 Graz, Austria
| | - Katharina Jonas
- Department of Internal Medicine, Division of Oncology, Medical University of Graz, 8036 Graz, Austria; (F.P.); (F.P.); (A.B.); (C.K.); (K.J.); (D.A.B.); (J.M.R.); (A.G.)
- Research Unit “Non-Coding RNAs and Genome Editing in Cancer”, Division of Oncology, Medical University of Graz, 8036 Graz, Austria
| | - Dominik A. Barth
- Department of Internal Medicine, Division of Oncology, Medical University of Graz, 8036 Graz, Austria; (F.P.); (F.P.); (A.B.); (C.K.); (K.J.); (D.A.B.); (J.M.R.); (A.G.)
- Research Unit “Non-Coding RNAs and Genome Editing in Cancer”, Division of Oncology, Medical University of Graz, 8036 Graz, Austria
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jakob M. Riedl
- Department of Internal Medicine, Division of Oncology, Medical University of Graz, 8036 Graz, Austria; (F.P.); (F.P.); (A.B.); (C.K.); (K.J.); (D.A.B.); (J.M.R.); (A.G.)
| | - Armin Gerger
- Department of Internal Medicine, Division of Oncology, Medical University of Graz, 8036 Graz, Austria; (F.P.); (F.P.); (A.B.); (C.K.); (K.J.); (D.A.B.); (J.M.R.); (A.G.)
| | - Martin Pichler
- Department of Internal Medicine, Division of Oncology, Medical University of Graz, 8036 Graz, Austria; (F.P.); (F.P.); (A.B.); (C.K.); (K.J.); (D.A.B.); (J.M.R.); (A.G.)
- Research Unit “Non-Coding RNAs and Genome Editing in Cancer”, Division of Oncology, Medical University of Graz, 8036 Graz, Austria
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Correspondence:
| |
Collapse
|