1
|
Ma J, Zhao J, Zhang C, Tan J, Cheng A, Niu Z, Lin Z, Pan G, Chen C, Ding Y, Zhong M, Zhuang Y, Xiong Y, Zhou H, Zhou S, Xu M, Ye W, Li F, Song Y, Wang Z, Hong X. Cleavage of CAD by caspase-3 determines the cancer cell fate during chemotherapy. Nat Commun 2025; 16:5006. [PMID: 40442064 PMCID: PMC12123037 DOI: 10.1038/s41467-025-60144-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 05/16/2025] [Indexed: 06/02/2025] Open
Abstract
Metabolic heterogeneity resulting from the intra-tumoral heterogeneity mediates massive adverse outcomes of tumor therapy, including chemotherapeutic resistance, but the mechanisms inside remain largely unknown. Here, we find that the de novo pyrimidine synthesis pathway determines the chemosensitivity. Chemotherapeutic drugs promote the degradation of cytosolic Carbamoyl-phosphate synthetase II, Aspartate transcarbamylase, and Dihydroorotase (CAD), an enzyme that is rate-limiting for pyrimidine synthesis, leading to apoptosis. We also find that CAD needs to be cleaved by caspase-3 on its Asp1371 residue, before its degradation. Overexpressing CAD or mutating Asp1371 to block caspase-3 cleavage confers chemoresistance in xenograft and Cldn18-ATK gastric cancer models. Importantly, mutations related to Asp1371 of CAD are found in tumor samples that failed neoadjuvant chemotherapy and pharmacological targeting of CAD-Asp1371 mutations using RMY-186 ameliorates chemotherapy efficacy. Our work reveals the vulnerability of de novo pyrimidine synthesis during chemotherapy, highlighting CAD as a promising therapeutic target and biomarker.
Collapse
Affiliation(s)
- Jingsong Ma
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Municipal Key Laboratory of Gastrointestinal Oncology, Xiamen, China
| | - Jiabao Zhao
- State Key Laboratory for Cellular Stress Biology, Innovation Centre for Cell Signalling Network, School of Life Sciences, Xiamen University, Xiamen, China
| | - Chensong Zhang
- State Key Laboratory for Cellular Stress Biology, Innovation Centre for Cell Signalling Network, School of Life Sciences, Xiamen University, Xiamen, China
| | - Jinshui Tan
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Municipal Key Laboratory of Gastrointestinal Oncology, Xiamen, China
| | - Ao Cheng
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Zhuo Niu
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Zeyang Lin
- Department of Pathology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Guangchao Pan
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Municipal Key Laboratory of Gastrointestinal Oncology, Xiamen, China
| | - Chao Chen
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Yang Ding
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Mengya Zhong
- Department of Radiology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yifan Zhuang
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yubo Xiong
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Municipal Key Laboratory of Gastrointestinal Oncology, Xiamen, China
| | - Huiwen Zhou
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Municipal Key Laboratory of Gastrointestinal Oncology, Xiamen, China
| | - Shengyi Zhou
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Municipal Key Laboratory of Gastrointestinal Oncology, Xiamen, China
| | - Meijuan Xu
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Municipal Key Laboratory of Gastrointestinal Oncology, Xiamen, China
| | - Wenjie Ye
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Municipal Key Laboratory of Gastrointestinal Oncology, Xiamen, China
| | - Funan Li
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China.
| | - Yongxi Song
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, China.
| | - Zhenning Wang
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, China.
| | - Xuehui Hong
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
- Xiamen Municipal Key Laboratory of Gastrointestinal Oncology, Xiamen, China.
| |
Collapse
|
2
|
Zhang Y, Zhang D, Xie Z, Xia T, Zou L, Wang T, Zhong L, Zeng Z, Wang L, Chen G, Liang X. Integrated transcriptomic and metabolomic analysis reveals the effects of EMMPRIN on nucleotide metabolism and 1C metabolism in AS mouse BMDMs. Front Mol Biosci 2025; 11:1460186. [PMID: 40125455 PMCID: PMC11927532 DOI: 10.3389/fmolb.2024.1460186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 12/27/2024] [Indexed: 03/25/2025] Open
Abstract
Background Extracellular matrix metalloproteinase inducer (EMMPRIN) has been considered as a key promoting factor in atherosclerosis (AS). Some studies have shown that regulating EMMPRIN expression in bone marrow-derived macrophages (BMDMs) of ApoE-/- mice can affect plaque stability, but the mechanism was not clear. Methods AS model mice were built from high-fat-feeding ApoE -/- mice, and were divided into siE group and CON group. The BMDMs and aortas from AS mice were harvested following in vivo treatment with either EMMPRIN short interfering (si)RNA (siEMMPRIN) or negative control siRNA. Transcriptomic and metabolomic profiles were analyzed using RNA-sequencing and Liquid chromatography-tandem mass spectrometry (LC-MS/MS), respectively. The efficacy of siEMMPRIN was assessed through real-time quantitative polymerase chain reaction (RT-qPCR) and Western blotting (WB). Immunofluorescence staining was employed to measure EMMPRIN expression within aortic atherosclerotic plaques. Cell proliferation was monitored using the Cell Counting Kit-8 (CCK8), while flow cytometry was utilized to analyze the cell cycle. Additionally, seahorse analysis and oil red O staining were conducted to verify glucose and lipid metabolism, respectively. Results A total of 3,282 differentially expressed metabolites (DEMs) and 16,138 differentially expressed genes (DEGs) were identified between the CON group and siE group. The nucleotide metabolism and one-carbon (1C) metabolism were identified as major altered pathways at both the transcriptional and metabolic levels. Metabolomic results identified increased levels of glycine, serine, betaine and S-adenosyl-L-methionine (SAM) to S-adenosyl-L-homocysteine (SAH) ratio and decreased levels of dimethylglycine (DMG) and SAH in 1C metabolism, accompanied by the accumulation of nucleotides, nucleosides, and bases in nucleotide metabolism. Transcriptomics results shown that Dnmt, Mthfd2 and Dhfr were downregulated, while Mthfr were upregulated in 1C metabolism. And numerous genes involved in de novo nucleotide synthesis, pentose phosphate pathway (PPP) and dNTP production were significantly inhibited, which may be associated with decreased BMDMs proliferation and cell cycle arrest in the G0/G1 phase in siE group. Multi-omics results also showed changes in glucose and lipid metabolism. Seahorse assay confirmed reduced glycolysis and oxidative phosphorylation (OXPHOS) levels and the Oil Red O staining confirmed the decrease of lipid droplets in siE group. Conclusion The integrated metabolomic and transcriptomic analysis suggested that nucleotide metabolism and 1C metabolism may be major metabolic pathways affected by siEMMPRIN in AS mouse BMDMs. Our study contributes to a better understanding of the role of EMMPRIN in AS development.
Collapse
Affiliation(s)
- Yun Zhang
- First Clinical College, Chongqing Medical University, Chongqing, China
| | - Diyuan Zhang
- Second Clinical College, Chongqing Medical University, Chongqing, China
| | - Zulong Xie
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tianli Xia
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lili Zou
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tao Wang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Zhong
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhuo Zeng
- First Clinical College, Chongqing Medical University, Chongqing, China
| | - Lingying Wang
- First Clinical College, Chongqing Medical University, Chongqing, China
| | - Guozhu Chen
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xing Liang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
3
|
Zhou M, Li Y, Qian J, Dong X, Guo Y, Yin L, Liu C, Hao K, Hu Q. P2Y 14R activation facilitates liver regeneration via CREB/DNMT3b/Dact-2/ β-Catenin signals in acute liver failure. Acta Pharm Sin B 2025; 15:919-933. [PMID: 40177539 PMCID: PMC11959937 DOI: 10.1016/j.apsb.2025.01.004] [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: 06/25/2024] [Revised: 09/29/2024] [Accepted: 11/06/2024] [Indexed: 04/05/2025] Open
Abstract
Acute liver failure (ALF) is lack of broadly approved therapeutic strategy except liver transplantation. As a glycogen metabolic intermediate, UDP-glucose (UDP-G) has been considered to accelerate liver repairment. Nevertheless, the role of UDP-G and its receptor P2Y purinoceptor 14 (P2Y14R) in ALF remains unknown. The present study aims to investigate the role and underlying mechanisms of UDP-G/P2Y14R axis in ALF. In this study, hepatic P2Y14R is significantly increased in TAA-induced and partial hepatectomy-induced ALF, while knockout of whole-body P2Y14R aggravates liver failure, manifested by inhibiting β-Catenin-mediated liver regeneration. Consistently, P2Y14R deficiency exhibits impaired liver regeneration in mice suffer partial hepatectomy. Importantly, only hepatocellular specific deletion of P2Y14R (P2Y14R flox/flox Alb cre/+ ) mice shows a similar phenomenon, rather than stellate cell specific deletion of P2Y14R (P2Y14R flox/flox Lrat cre/+ ) mice. Mechanistically, P2Y14R induction regulates methylation of Dact-2 through CREB/DNMT3b signals in hepatocytes, subsequently inhibiting the expression of Dact-2 which is a stabilizer of β-Catenin degradation complex, leading to the activation of β-Catenin -mediated liver regeneration. Interestingly, the administration of exogenous UDP-G can accelerate liver regeneration and liver function recovery after partial hepatectomy in hepatocellular carcinoma mice. Together, the findings propose an unrecognized role of P2Y14R in ALF and provide an effective adjuvant strategy for treatment of ALF.
Collapse
Affiliation(s)
- Mengze Zhou
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Yehong Li
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Jialong Qian
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Xinli Dong
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yanshuo Guo
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Li Yin
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Chunxiao Liu
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Kun Hao
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Qinghua Hu
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
4
|
Li Y, Yu H, Xiong L, Zeng K, Wei Y, Li H, Ji X. Diversity and function of viral AMGs associated with DNA biosynthesis in the Napahai plateau wetland. ENVIRONMENTAL TECHNOLOGY 2024; 45:5521-5535. [PMID: 38126212 DOI: 10.1080/09593330.2023.2296531] [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: 07/18/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023]
Abstract
Viruses play an important role in microbial community structure and biodiversity by lysing host cells, and can also affect host metabolic pathways by expressing auxiliary metabolic genes (AMGs). As a unique low-latitude, high-altitude seasonal plateau wetland in China, Napahai has high research value. However, studies on the genetic diversity of AMGs and viruses associated with DNA biosynthesis have not been reported. Based on metagenomics, with the phylogenetic tree, PCoA, and α diversity analysis, we found that three DNA biosynthesis-related viral AMGs (cobS, mazG, and purM) in the Napahai plateau wetland were rich in genetic diversity, uniqueness, and differences compared with other habitats and host sources. Through the KEGG metabolic pathway and metabolic flow analysis of Pseudomonas mandelii (SW-3) and phage (VSW-3), the AMGs (cobS, mazG, and purM) genes of the three related viruses involved in DNA biosynthesis were upregulated and their expression increased significantly. In general, we systematically described the genetic diversity of AMGs associated with DNA biosynthesis in plateau wetland ecosystems and clarified the contribution of viral AMGs in the Napahai plateau wetland to DNA biosynthesis, as well as the changes of metabolites and genes. It further expands the understanding of phage-host interactions, which is of great significance for further revealing the role of viral AMGs in the biological evolution and biogeochemical cycle of wetland ecosystems.
Collapse
Affiliation(s)
- Yanmei Li
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Hang Yu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Lingling Xiong
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Kun Zeng
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Yunlin Wei
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Haiyan Li
- Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Xiuling Ji
- Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
| |
Collapse
|
5
|
Cao Y, Xu M, Chen Q, Wu D, Lu J, Cai G. Potential nutritional and functional matters in yeast culture prepared by soybean meal fermentation. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:8869-8878. [PMID: 38963133 DOI: 10.1002/jsfa.13713] [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: 04/09/2024] [Revised: 05/20/2024] [Accepted: 06/16/2024] [Indexed: 07/05/2024]
Abstract
BACKGROUND Yeast culture (YC) is a product fermented on a specific medium, which is a type of postbiotic of anaerobic solid-state fermentation. Although YC has positive effects on the animal growth and health, it contains a variety of beneficial metabolites as dark matter, which have not been quantified. In the present study, liquid chromatography-tandem mass spectrometry is employed to identify the unknown metabolites. Following their identification, the important chemicals are quantified using HPLC-diode array detection methods. RESULTS Non-targeted metabolomics studies showed that 670 metabolites in total were identified in YC, of which 23 metabolites significantly increased, including organic acids, amino acids, nucleosides and purines, isoflavones, and other substances. The chemical quantitative analysis showed that the contents of succinic acid, aminobutyric acid, glutamine, purine and daidzein increased by 84.42%, 51.07%, 100%, 68.85% and 4.60%, respectively. CONCLUSION Therefore, the use of non-targeted metabolomics combined with chemical quantitative analysis to reveal the nutritional and functional substances of YC could help to elucidate the postbiotic mechanism and provide theoretical support for the regulation of the directional accumulation of beneficial metabolites. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Yazhuo Cao
- School of Biotechnology and Key Laboratory of Industrial Biotechnology of Ministry of Education, Jiangnan University, Wuxi, China
- National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, China
| | - Minwei Xu
- Department of Plant Sciences, North Dakota State University, Fargo, ND, USA
| | - Qiong Chen
- Department of Plant Sciences, North Dakota State University, Fargo, ND, USA
| | - Dianhui Wu
- National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, China
| | - Jian Lu
- National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, China
| | - Guolin Cai
- School of Biotechnology and Key Laboratory of Industrial Biotechnology of Ministry of Education, Jiangnan University, Wuxi, China
- National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, China
- Jiangsu Provincial Research Center for Bioactive Product Processing Technology, Jiangnan University, Wuxi, China
| |
Collapse
|
6
|
Ma J, Zhao J, Wu Z, Tan J, Xu M, Ye W, Zhong M, Xiong Y, Pan G, Zhou H, Zhou S, Hong X. Dehydroabietylamine exerts antitumor effects by affecting nucleotide metabolism in gastric cancer. Carcinogenesis 2024; 45:759-772. [PMID: 38869064 PMCID: PMC11464700 DOI: 10.1093/carcin/bgae037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 04/28/2024] [Accepted: 06/12/2024] [Indexed: 06/14/2024] Open
Abstract
Nucleotide metabolism is the ultimate and most critical link in the self-replication process of tumors, including gastric cancer (GC). However, in clinical treatment, classic antitumor drugs such as 5-fluorouracil (5-FU) are mostly metabolic analogs of purines or pyrimidines, which lack specificity for tumor cells and therefore have significant side effects. It is unclear whether there are other drugs that can target nucleotide metabolism, except for nucleic acid analogs. Here, we found that a natural compound, dehydroabietylamine (DHAA), significantly reduced the viability and proliferation of GC cells and organoids. DHAA disrupts the purine and pyrimidine metabolism of GC cells, causing DNA damage and further inducing apoptosis. DHAA treatment decreased transcription and protein levels of key enzymes involved in the nucleotide metabolism pathway, with significant reductions in the expression of pyrimidine metabolism key enzymes CAD, DHODH, and purine metabolism key enzymes PAICS. We also found that DHAA directly binds to and reduces the expression of Forkhead box K2 (FOXK2), a common transcription factor for these metabolic enzymes. Ultimately, DHAA was shown to delay tumorigenesis in K19-Wnt1/C2mE transgenic mice model and reduce levels of CAD, DHODH, and PAICS in vivo. We demonstrate that DHAA exerts an anticancer effect on GC by targeting transcription factor FOXK2, reducing transcription of key genes for nucleotide metabolism and impairing nucleotide biosynthesis, thus DHAA is a promising candidate for GC therapy.
Collapse
Affiliation(s)
- Jingsong Ma
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361000, China
- Xiamen Municipal Key Laboratory of Gastrointestinal Oncology, Xiamen 361000, China
| | - Jiabao Zhao
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361000, China
- Xiamen Municipal Key Laboratory of Gastrointestinal Oncology, Xiamen 361000, China
| | - Zhengxin Wu
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, China
| | - Jinshui Tan
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361000, China
- Xiamen Municipal Key Laboratory of Gastrointestinal Oncology, Xiamen 361000, China
| | - Meijuan Xu
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361000, China
- Xiamen Municipal Key Laboratory of Gastrointestinal Oncology, Xiamen 361000, China
| | - Wenjie Ye
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361000, China
- Xiamen Municipal Key Laboratory of Gastrointestinal Oncology, Xiamen 361000, China
| | - Mengya Zhong
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen 361003, China
| | - Yubo Xiong
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen 361003, China
| | - Guangchao Pan
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361000, China
- Xiamen Municipal Key Laboratory of Gastrointestinal Oncology, Xiamen 361000, China
| | - Huiwen Zhou
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361000, China
- Xiamen Municipal Key Laboratory of Gastrointestinal Oncology, Xiamen 361000, China
| | - Shengyi Zhou
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361000, China
- Xiamen Municipal Key Laboratory of Gastrointestinal Oncology, Xiamen 361000, China
| | - Xuehui Hong
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361000, China
- Xiamen Municipal Key Laboratory of Gastrointestinal Oncology, Xiamen 361000, China
| |
Collapse
|
7
|
Zhang L, Tao L, Tian D, Li D. Editorial: Immunometabolism and tumor microenvironment in hepatocellular carcinoma. Front Oncol 2024; 14:1483397. [PMID: 39319056 PMCID: PMC11419957 DOI: 10.3389/fonc.2024.1483397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 08/26/2024] [Indexed: 09/26/2024] Open
Affiliation(s)
- Luyun Zhang
- Department of Gastroenterology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China
- Henan Key Medical Laboratory for Molecular Immunology of Digestive Diseases, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Lianyuan Tao
- Department of Hepatobiliary Pancreatic Surgery, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China
- Henan Provincial Key Medical Laboratory for Hepatobiliary and Pancreatic Diseases, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Dean Tian
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dongxiao Li
- Department of Gastroenterology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China
- Henan Key Medical Laboratory for Molecular Immunology of Digestive Diseases, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| |
Collapse
|
8
|
Deng YN, Chen Y, Gao S, Zhang N, Luo Y, Luo S, Li Q, Fu X, Liang S. RREB1-mediated SUMOylation enhancement promotes chemoresistance partially by transcriptionally upregulating UBC9 in colorectal cancer. Front Pharmacol 2024; 15:1381860. [PMID: 39108750 PMCID: PMC11300207 DOI: 10.3389/fphar.2024.1381860] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 07/10/2024] [Indexed: 03/17/2025] Open
Abstract
Chemoresistance is a main cause of chemotherapy failure and tumor recurrence. The effects of global protein SUMOylation on chemoresistance in colorectal cancer (CRC) remains to be investigated. Herein, we have proposed that the elevated SUMO2/3-modified proteins confer 5-fluorouracil (5-FU) chemoresistance acquisition in CRC. The SUMOylation levels of global proteins in CRC cell lines were elevated compared with normal colon cell line NCM460. 5-FU treatment obviously reduced SUMOylation of global proteins in 5-FU-sensitive CRC cells including HT29, HCT116 and HCT-8. However, in 5-FU-resistant HCT-8/5-FU cells, the expression level of SUMO2/3-modified proteins was increased under 5-FU exposure in a concentration-dependent manner. 5-FU treatment combined with SUMOylation inhibitor ML-792 significantly increased the sensitivity of 5-FU-resistant cells to 5-FU and reduced colony formation numbers in HCT-8/5-FU cells. And UBC9-mediated SUMOylation elevation contributes to 5-FU resistance in HCT116 cells. Moreover, we also identified RREB1 as a regulator of SUMOylation profiling of global cellular proteins via directly binding to the promoter of UBC9. Overexpression of RREB1 promoted 5-FU resistance in CRC, which was partially abolished by treatment of inhibitor ML-792. In conclusion, RREB1-enhanced protein SUMOylation contributes to 5-FU resistance acquisition in CRC.
Collapse
Affiliation(s)
- Ya-nan Deng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Ying Chen
- Department of Medical Oncology, Suining First People’s Hospital, Suining, Sichuan, China
| | - Shan Gao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Nan Zhang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yinheng Luo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Shu Luo
- Department of Medical Oncology, Suining First People’s Hospital, Suining, Sichuan, China
| | - Qiu Li
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xianghui Fu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Shufang Liang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
9
|
El-Mergawy R, Chafin L, Ovando-Ricardez JA, Rosas L, Tsai M, Rojas M, Mora AL, Mallampalli RK. FOXK2 targeting by the SCF-E3 ligase subunit FBXO24 for ubiquitin mediated degradation modulates mitochondrial respiration. J Biol Chem 2024; 300:107359. [PMID: 38735474 PMCID: PMC11209018 DOI: 10.1016/j.jbc.2024.107359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/05/2024] [Accepted: 04/30/2024] [Indexed: 05/14/2024] Open
Abstract
FOXK2 is a crucial transcription factor implicated in a wide array of biological activities and yet understanding of its molecular regulation at the level of protein turnover is limited. Here, we identify that FOXK2 undergoes degradation in lung epithelia in the presence of the virulent pathogens Pseudomonas aeruginosa and Klebsiella pneumoniae through ubiquitin-proteasomal processing. FOXK2 through its carboxyl terminus (aa 428-478) binds the Skp-Cullin-F-box ubiquitin E3 ligase subunit FBXO24 that mediates multisite polyubiquitylation of the transcription factor resulting in its nuclear degradation. FOXK2 was detected within the mitochondria and targeted depletion of the transcription factor or cellular expression of FOXK2 mutants devoid of key carboxy terminal domains significantly impaired mitochondrial function. In experimental bacterial pneumonia, Fbxo24 heterozygous mice exhibited preserved mitochondrial function and Foxk2 protein levels compared to WT littermates. The results suggest a new mode of regulatory control of mitochondrial energetics through modulation of FOXK2 cellular abundance.
Collapse
Affiliation(s)
- Rabab El-Mergawy
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Lexie Chafin
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Jose A Ovando-Ricardez
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Lorena Rosas
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - MuChun Tsai
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Mauricio Rojas
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Ana L Mora
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Rama K Mallampalli
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA.
| |
Collapse
|
10
|
Yang Y, Yu F. Abnormal protein SUMOylation in liver disease: novel target for therapy. J Mol Med (Berl) 2024; 102:719-731. [PMID: 38565749 DOI: 10.1007/s00109-024-02440-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/10/2024] [Accepted: 03/15/2024] [Indexed: 04/04/2024]
Abstract
SUMOylation is an important protein post-translational modification (PTM) process, in which the small ubiquitin-like modifier (SUMO) protein covalently binds to the target protein and regulates stability, subcellular localization, and protein-protein interaction of the target protein. Protein SUMOylation exerts crucial regulatory function in the liver, and its abnormalities are associated with various liver-related disease processes. This review focuses on the biological functions of protein SUMOylation in liver-related diseases in recent years, summarizes the molecular mechanisms of SUMOylation in the replication of hepatitis viruses and the occurrence of hepatocellular carcinoma, and discusses the significance of SUMOylation in liver-related disorders, which is essential for understanding liver biological processes and formulating therapeutic strategies.
Collapse
Affiliation(s)
- Yanfang Yang
- Department of Central Laboratory, Guizhou Provincial People's Hospital, Guiyang, 550002, China.
- NHC Key Laboratory of Pulmonary Immunological Diseases, Guizhou Provincial People's Hospital, Guiyang, 550002, China.
| | - Fuxun Yu
- Department of Central Laboratory, Guizhou Provincial People's Hospital, Guiyang, 550002, China.
- NHC Key Laboratory of Pulmonary Immunological Diseases, Guizhou Provincial People's Hospital, Guiyang, 550002, China.
| |
Collapse
|
11
|
Xing X, Que X, Zheng S, Wang S, Song Q, Yao Y, Zhang P. Emerging roles of FOXK2 in cancers and metabolic disorders. Front Oncol 2024; 14:1376496. [PMID: 38741782 PMCID: PMC11089157 DOI: 10.3389/fonc.2024.1376496] [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: 01/25/2024] [Accepted: 04/15/2024] [Indexed: 05/16/2024] Open
Abstract
FOXK2, a member of the Forkhead box K (FOXK) transcription factor family, is widely expressed in various tissues and organs throughout the body. FOXK2 plays crucial roles in cell proliferation, differentiation, autophagy, de novo nucleotide biosynthesis, DNA damage response, and aerobic glycolysis. Although FOXK2 is recognized as an oncogene in colorectal cancer and hepatocellular carcinoma, it acts as a tumor suppressor in breast cancer, cervical cancer, and non-small cell lung cancer (NSCLC). This review provides an overview of the recent progress in understanding the regulatory mechanisms of FOXK2 and its downstream targets, highlights the significant impact of FOXK2 dysregulation on cancer etiology, and discusses the potential of targeting FOXK2 for cancer treatment.
Collapse
Affiliation(s)
| | | | | | | | - Qibin Song
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yi Yao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Pingfeng Zhang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
12
|
Zhang Y, Wei Z, Zhang M, Wang S, Gao T, Huang H, Zhang T, Cai H, Liu X, Fu T, Liang D. Population Structure and Selection Signal Analysis of Nanyang Cattle Based on Whole-Genome Sequencing Data. Genes (Basel) 2024; 15:351. [PMID: 38540410 PMCID: PMC10970060 DOI: 10.3390/genes15030351] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/07/2024] [Accepted: 03/09/2024] [Indexed: 06/14/2024] Open
Abstract
With a rich breeding history, Nanyang cattle (NY cattle) have undergone extensive natural and artificial selection, resulting in distinctive traits such as high fertility, excellent meat quality, and disease resistance. This makes them an ideal model for studying the mechanisms of environmental adaptability. To assess the population structure and genetic diversity of NY cattle, we performed whole-genome resequencing on 30 individuals. These data were then compared with published whole-genome resequencing data from 432 cattle globally. The results indicate that the genetic structure of NY cattle is significantly different from European commercial breeds and is more similar to North-Central Chinese breeds. Furthermore, among all breeds, NY cattle exhibit the highest genetic diversity and the lowest population inbreeding levels. A genome-wide selection signal analysis of NY cattle and European commercial breeds using Fst, θπ-ratio, and θπ methods revealed significant selection signals in genes associated with reproductive performance and immunity. Our functional annotation analysis suggests that these genes may be responsible for reproduction (MAP2K2, PGR, and GSE1), immune response (NCOA2, HSF1, and PAX5), and olfaction (TAS1R3). We provide a comprehensive overview of sequence variations in the NY cattle genome, revealing insights into the population structure and genetic diversity of NY cattle. Additionally, we identify candidate genes associated with important economic traits, offering valuable references for future conservation and breeding efforts of NY cattle.
Collapse
Affiliation(s)
- Yan Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Y.Z.); (Z.W.); (M.Z.); (S.W.); (T.G.); (H.H.); (T.Z.); (H.C.); (T.F.)
| | - Zhitong Wei
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Y.Z.); (Z.W.); (M.Z.); (S.W.); (T.G.); (H.H.); (T.Z.); (H.C.); (T.F.)
| | - Man Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Y.Z.); (Z.W.); (M.Z.); (S.W.); (T.G.); (H.H.); (T.Z.); (H.C.); (T.F.)
| | - Shiwei Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Y.Z.); (Z.W.); (M.Z.); (S.W.); (T.G.); (H.H.); (T.Z.); (H.C.); (T.F.)
| | - Tengyun Gao
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Y.Z.); (Z.W.); (M.Z.); (S.W.); (T.G.); (H.H.); (T.Z.); (H.C.); (T.F.)
| | - Hetian Huang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Y.Z.); (Z.W.); (M.Z.); (S.W.); (T.G.); (H.H.); (T.Z.); (H.C.); (T.F.)
| | - Tianliu Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Y.Z.); (Z.W.); (M.Z.); (S.W.); (T.G.); (H.H.); (T.Z.); (H.C.); (T.F.)
| | - Hanfang Cai
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Y.Z.); (Z.W.); (M.Z.); (S.W.); (T.G.); (H.H.); (T.Z.); (H.C.); (T.F.)
| | - Xian Liu
- Henan Animal Husbandry Station, Zhengzhou 450008, China;
| | - Tong Fu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Y.Z.); (Z.W.); (M.Z.); (S.W.); (T.G.); (H.H.); (T.Z.); (H.C.); (T.F.)
| | - Dong Liang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Y.Z.); (Z.W.); (M.Z.); (S.W.); (T.G.); (H.H.); (T.Z.); (H.C.); (T.F.)
| |
Collapse
|
13
|
Cai Y, Wang Z, Guo S, Lin C, Yao H, Yang Q, Wang Y, Yu X, He X, Sun W, Qiu S, Guo Y, Tang S, Xie Y, Zhang A. Detection, mechanisms, and therapeutic implications of oncometabolites. Trends Endocrinol Metab 2023; 34:849-861. [PMID: 37739878 DOI: 10.1016/j.tem.2023.08.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 08/10/2023] [Accepted: 08/28/2023] [Indexed: 09/24/2023]
Abstract
Metabolic abnormalities are a hallmark of cancer cells and are essential to tumor progression. Oncometabolites have pleiotropic effects on cancer biology and affect a plethora of processes, from oncogenesis and metabolism to therapeutic resistance. Targeting oncometabolites, therefore, could offer promising therapeutic avenues against tumor growth and resistance to treatments. Recent advances in characterizing the metabolic profiles of cancer cells are shedding light on the underlying mechanisms and associated metabolic networks. This review summarizes the diverse detection methods, molecular mechanisms, and therapeutic targets of oncometabolites, which may lead to targeting oncometabolism for cancer therapy.
Collapse
Affiliation(s)
- Ying Cai
- International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan General Hospital, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, International Joint Research Center on Traditional Chinese and Modern Medicine, Hainan Medical University, Haikou 571199, China; Graduate School, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Zhibo Wang
- International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan General Hospital, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, International Joint Research Center on Traditional Chinese and Modern Medicine, Hainan Medical University, Haikou 571199, China; Graduate School, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Sifan Guo
- International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan General Hospital, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, International Joint Research Center on Traditional Chinese and Modern Medicine, Hainan Medical University, Haikou 571199, China; Graduate School, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Chunsheng Lin
- Second Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Hong Yao
- First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Qiang Yang
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Yan Wang
- International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan General Hospital, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, International Joint Research Center on Traditional Chinese and Modern Medicine, Hainan Medical University, Haikou 571199, China
| | - Xiaodan Yu
- International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan General Hospital, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, International Joint Research Center on Traditional Chinese and Modern Medicine, Hainan Medical University, Haikou 571199, China
| | - Xiaowen He
- International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan General Hospital, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, International Joint Research Center on Traditional Chinese and Modern Medicine, Hainan Medical University, Haikou 571199, China
| | - Wanying Sun
- International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan General Hospital, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, International Joint Research Center on Traditional Chinese and Modern Medicine, Hainan Medical University, Haikou 571199, China
| | - Shi Qiu
- International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan General Hospital, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, International Joint Research Center on Traditional Chinese and Modern Medicine, Hainan Medical University, Haikou 571199, China.
| | - Yu Guo
- International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan General Hospital, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, International Joint Research Center on Traditional Chinese and Modern Medicine, Hainan Medical University, Haikou 571199, China.
| | - Songqi Tang
- International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan General Hospital, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, International Joint Research Center on Traditional Chinese and Modern Medicine, Hainan Medical University, Haikou 571199, China.
| | - Yiqiang Xie
- International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan General Hospital, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, International Joint Research Center on Traditional Chinese and Modern Medicine, Hainan Medical University, Haikou 571199, China.
| | - Aihua Zhang
- International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan General Hospital, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, International Joint Research Center on Traditional Chinese and Modern Medicine, Hainan Medical University, Haikou 571199, China; Graduate School, Heilongjiang University of Chinese Medicine, Harbin 150040, China.
| |
Collapse
|
14
|
Markitantova Y, Fokin A, Boguslavsky D, Simirskii V, Kulikov A. Molecular Signatures Integral to Natural Reprogramming in the Pigment Epithelium Cells after Retinal Detachment in Pleurodeles waltl. Int J Mol Sci 2023; 24:16940. [PMID: 38069262 PMCID: PMC10707686 DOI: 10.3390/ijms242316940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 12/18/2023] Open
Abstract
The reprogramming of retinal pigment epithelium (RPE) cells into retinal cells (transdifferentiation) lies in the bases of retinal regeneration in several Urodela. The identification of the key genes involved in this process helps with looking for approaches to the prevention and treatment of RPE-related degenerative diseases of the human retina. The purpose of our study was to examine the transcriptome changes at initial stages of RPE cell reprogramming in adult newt Pleurodeles waltl. RPE was isolated from the eye samples of day 0, 4, and 7 after experimental surgical detachment of the neural retina and was used for a de novo transcriptome assembly through the RNA-Seq method. A total of 1019 transcripts corresponding to the differently expressed genes have been revealed in silico: the 83 increased the expression at an early stage, and 168 increased the expression at a late stage of RPE reprogramming. We have identified up-regulation of classical early response genes, chaperones and co-chaperones, genes involved in the regulation of protein biosynthesis, suppressors of oncogenes, and EMT-related genes. We revealed the growth in the proportion of down-regulated ribosomal and translation-associated genes. Our findings contribute to revealing the molecular mechanism of RPE reprogramming in Urodela.
Collapse
Affiliation(s)
| | | | | | - Vladimir Simirskii
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia; (Y.M.); (A.K.)
| | | |
Collapse
|