1
|
Long X, Liu R, Zhang M, Wu Y, Zhang S, Tang K, Wang H. Integrated proteomics and phosphoproteomics study reveals the potential tumour suppressive function of PCK2 in hepatocellular carcinoma. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2025. [PMID: 40235441 DOI: 10.1039/d5ay00090d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Hepatocellular carcinoma (HCC) is one of the most fatal malignancies worldwide. Patients are often diagnosed at advanced stages and have short overall survivals. Thus, there is an urgent need to understand the underlying mechanism of HCC development and discover new drug targets. In the present study, an ultrahigh performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS)-based approach was used to investigate the proteome and phosphoproteome alterations between tumour and non-tumour tissues of HCC patients. In total, 678 proteins and 350 phosphoproteins were significantly changed between the two groups. Kyoto encyclopedia of genes and genomes (KEGG) pathway analysis revealed that glycolysis/glucogenesis was one of the most significantly enriched pathways of down-regulated phosphoproteins and PCK2 was one hub protein in this pathway. Furthermore, the phosphosite of the Ser304 residue in PCK2 is highly evolutionarily conserved among different species and is located near to the GDP, Mn2+ binding sites and kinase activity site, which indicate that the phosphorylation of Ser304 may regulate PCK2 activity. HepG2 cell proliferation, invasion and migration were significantly increased when treated with 3-mercaptopicolinic acid, an inhibitor of PCK2, which demonstrated the tumour suppressive activity of PCK2 in HCC. In addition, the low mRNA expression of PCK2 was correlated with poor overall survival of live cancer patients. Overall, our proteomic and phosphoproteomic study revealed the tumour suppressive function of PCK2 in HCC and indicated that activation of its expression or activity may be a potential therapeutic strategy for HCC treatment.
Collapse
Affiliation(s)
- Xiao Long
- Institute of Mass Spectrometry, Zhejiang Engineering Research Center of Advanced Mass spectrometry and Clinical Application, School of Material Science and Chemical Engineering, Ningbo University, Ningbo 315211, China.
- Zhenhai Institute of Mass Spectrometry, Ningbo 315211, China
| | - Rong Liu
- Institute of Mass Spectrometry, Zhejiang Engineering Research Center of Advanced Mass spectrometry and Clinical Application, School of Material Science and Chemical Engineering, Ningbo University, Ningbo 315211, China.
- Zhenhai Institute of Mass Spectrometry, Ningbo 315211, China
| | - Meng Zhang
- Institute of Mass Spectrometry, Zhejiang Engineering Research Center of Advanced Mass spectrometry and Clinical Application, School of Material Science and Chemical Engineering, Ningbo University, Ningbo 315211, China.
- Zhenhai Institute of Mass Spectrometry, Ningbo 315211, China
| | - Yannan Wu
- Institute of Mass Spectrometry, Zhejiang Engineering Research Center of Advanced Mass spectrometry and Clinical Application, School of Material Science and Chemical Engineering, Ningbo University, Ningbo 315211, China.
- Zhenhai Institute of Mass Spectrometry, Ningbo 315211, China
| | - Shun Zhang
- Guoke Ningbo Life Science and Health Industry Research Institute, No. 159 Beijiao Road, Ningbo City, Zhejiang Province, China.
| | - Keqi Tang
- Institute of Mass Spectrometry, Zhejiang Engineering Research Center of Advanced Mass spectrometry and Clinical Application, School of Material Science and Chemical Engineering, Ningbo University, Ningbo 315211, China.
- Zhenhai Institute of Mass Spectrometry, Ningbo 315211, China
| | - Hongxia Wang
- Institute of Mass Spectrometry, Zhejiang Engineering Research Center of Advanced Mass spectrometry and Clinical Application, School of Material Science and Chemical Engineering, Ningbo University, Ningbo 315211, China.
- Zhenhai Institute of Mass Spectrometry, Ningbo 315211, China
| |
Collapse
|
2
|
Hwang SH, Baek SH, Lee MJ, Kook Y, Bae SJ, Ahn SG, Jeong J. Clinical Relevance of TP53 Mutation and Its Characteristics in Breast Cancer with Long-Term Follow-Up Date. Cancers (Basel) 2024; 16:3899. [PMID: 39682089 DOI: 10.3390/cancers16233899] [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: 09/25/2024] [Revised: 10/30/2024] [Accepted: 10/30/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND The TP53 mutation is one of the most frequently identified mutations in human cancers and is typically associated with a poor prognosis. However, there are conflicting findings regarding its impact. We aimed to clarify the clinical relevance of TP53 mutations across all breast cancer subtypes and treatments utilizing long-term follow-up data. METHODS We retrospectively identified the data of breast cancer patients who underwent TP53 mutation testing. Stratified log-rank tests and Cox regression analysis were performed to compare oncologic outcomes based on TP53 mutation status and the characteristics of these mutations, including types and locations. Mutations in exons 5-9 were identified using polymerase chain reaction-denaturing high-performance liquid chromatography (PCR-DHPLC) and direct sequencing. RESULTS Between January 2007 and December 2015, 650 breast cancer patients underwent TP53 mutation testing in Gangnam Severance Hospital. The TP53 mutations were identified in 172 patients (26.5%), with 34 (19.8%) exhibiting missense hotspot mutations. Patients with TP53 mutations (TP53-mutated group) had worse prognosis, demonstrated by a 10-year recurrence-free survival (RFS) rate of 83.5% compared to 86.6% in patients without mutations (HR, 1.67; p = 0.026) and a 10-year overall survival (OS) rate of 88.1% versus 91.0% (HR, 3.02; p = 0.003). However, subgroup analyses within the TP53-mutated group did not reveal significant differences in oncologic outcomes based on mutation types and locations. CONCLUSIONS Our findings establish that TP53 mutations are linked to poorer oncologic outcomes in breast cancer across all subtypes. Yet, within the TP53-mutated group, the specific characteristics of TP53 mutations do not influence oncologic outcomes.
Collapse
Affiliation(s)
- Seung Hyun Hwang
- Institute for Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
- Department of Breast and Thyroid Surgery, Sam Hospital, Anyang 14030, Republic of Korea
| | - Seung Ho Baek
- Institute for Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
| | - Min Ji Lee
- Institute for Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
| | - Yoonwon Kook
- Institute for Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
| | - Soong June Bae
- Institute for Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
| | - Sung Gwe Ahn
- Institute for Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
| | - Joon Jeong
- Institute for Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
| |
Collapse
|
3
|
Wang X, Yang J, Yang W, Sheng H, Jia B, Cheng P, Xu S, Hong X, Jiang C, Yang Y, Wu Z, Wang J. Multiple roles of p53 in cancer development: Regulation of tumor microenvironment, m 6A modification and diverse cell death mechanisms. J Adv Res 2024:S2090-1232(24)00481-8. [PMID: 39490612 DOI: 10.1016/j.jare.2024.10.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/02/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024] Open
Abstract
BACKGROUND The protein p53, encoded by the most frequently mutated gene TP53 in human cancers, has diverse functions in tumor suppression. As a best known transcription factor, p53 can regulate various fundamental cellular responses, ranging from the cell-cycle arrest, DNA repair, senescence to the programmed cell death (PCD), which includes autophagy, apoptosis, ferroptosis, cuproptosis, pyroptosis and disulfidoptosis. Accumulating evidence has indicated that the tumor microenvironment (TME), N6-methyladenosine (m6A) modification and diverse PCD are important for the progression, proliferation and metastases of cancers. AIM OF REVIEW This paper aims to systematically and comprehensively summarize the multiple roles of p53 in the development of cancers from the regulation of TME, m6A Modification and diverse PCD. KEY SCIENTIFIC CONCEPTS OF REVIEW TME, a crucial local homeostasis environment, influences every step of tumorigenesis and metastasis. m6A, the most prevalent and abundant endogenous modification in eukaryotic RNAs, plays an essential role in various biological processes, containing the progression of cancers. Additionally, PCD is an evolutionarily conserved mechanism of cell suicide and a common process in living organisms. Some forms of PCD contribute to the occurrence and development of cancer. However, the complex roles of p53 within the TME, m6A modification and diverse PCD mechanisms are still not completely understood. Presently, the function roles of p53 including the wild-type and mutant p53 in different context are summarized. Additionally, the interaction between the cancer immunity, cancer cell death and RNA m6A methylation and the p53 regulation during the development and progress of cancers were discussed. Moreover, the key molecular mechanisms by which p53 participates in the regulation of TME, m6A and diverse PCD are also explored. All the findings will facilitate the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Xiangyu Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Jianhua Yang
- School of Medical Informatics Engineering, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Wanting Yang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Haiyang Sheng
- Global Biometrics and Data Sciences, Bristol Myers Squibb, New York City, USA
| | - Buyun Jia
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Peng Cheng
- The First Affiliated Hospital, Anhui University of Traditional Chinese Medicine, Hefei, Anhui, China
| | - Shanshan Xu
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Xinhui Hong
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Chuanwei Jiang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Yinfeng Yang
- School of Medical Informatics Engineering, Anhui University of Chinese Medicine, Hefei, Anhui, China.
| | - Ziyin Wu
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangsu Kanion Pharmaceutical Co. Ltd, Lianyungang, Jiangsu, China.
| | - Jinghui Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China.
| |
Collapse
|
4
|
Jing N, Tao Z, Du X, Wen Z, Gao WQ, Dong B, Fang YX. Targeting SOX4/PCK2 signaling suppresses neuroendocrine trans-differentiation of castration-resistant prostate cancer. Biol Direct 2024; 19:56. [PMID: 39014441 PMCID: PMC11251300 DOI: 10.1186/s13062-024-00500-2] [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: 04/22/2024] [Accepted: 07/11/2024] [Indexed: 07/18/2024] Open
Abstract
BACKGROUND Neuroendocrine prostate cancer (NEPC), a lethal subset of prostate cancer (PCa), is characterized by loss of AR signaling and resistance to AR-targeted therapy. While it is well reported that second-generation AR blockers induce neuroendocrine (NE) trans-differentiation of castration-resistant prostate cancer (CRPC) to promote the occurrence of NEPC, and pluripotent transcription factors might be potential regulators, the underlying molecular mechanisms remain unclear. METHODS We analyzed the data from public databsets to screen candidate genes and then focused on SOX4, a regulator of NE trans-differentiation. The expression changes of SOX4 and its relationship with tumor progression were validated in clinical tumor tissues. We evaluated malignant characteristics related to NEPC in prostate cancer cell lines with stable overexpression or knockdown of SOX4 in vitro. Tumor xenografts were analyzed after inoculating the relevant cell lines into nude mice. RNA-seq, ATAC-seq, non-targeted metabolomics analysis, as well as molecular and biochemical assays were carried out to determine the mechanism. RESULTS We screened public datasets and identified that expression of SOX4 was significantly elevated in NEPC. Overexpressing SOX4 in C4-2B cells increased cell proliferation and migration, upregulated the expression of NE marker genes, and inhibited AR expression. Consistently, inhibition of SOX4 expression in DU-145 and PC-3 cells reduced the above malignant phenotypes and repressed the expression of NE marker genes. For the in vivo assay, we found that knockdown of SOX4 inhibited tumor growth of subcutaneous xenografts in castrated nude mice which were concomitantly treated with enzalutamide (ENZ). Mechanically, we identified that one of the key enzymes in gluconeogenesis, PCK2, was a novel target of SOX4. The activation of carbohydrate metabolism reprogramming by SOX4 could promote NE trans-differentiation via the SOX4/PCK2 pathway. CONCLUSIONS Our findings reveal that SOX4 promotes NE trans-differentiation both in vitro and in vivo via directly enhancing PCK2 activity to activate carbohydrate metabolism reprogramming. The SOX4/PCK2 pathway and its downstream changes might be novel targets for blocking NE trans-differentiation.
Collapse
Affiliation(s)
- Nan Jing
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Zhenkeke Tao
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Xinxing Du
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Zhenzhen Wen
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Wei-Qiang Gao
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, China.
| | - Baijun Dong
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Yu-Xiang Fang
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| |
Collapse
|
5
|
Li C, Zhang ED, Ye Y, Xiao Z, Huang H, Zeng Z. Association of mitochondrial phosphoenolpyruvate carboxykinase with prognosis and immune regulation in hepatocellular carcinoma. Sci Rep 2024; 14:14051. [PMID: 38890507 PMCID: PMC11189538 DOI: 10.1038/s41598-024-64907-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 06/14/2024] [Indexed: 06/20/2024] Open
Abstract
Mitochondrial phosphoenolpyruvate carboxykinase (PCK2), a mitochondrial isoenzyme, supports the growth of cancer cells under glucose deficiency conditions in vitro. This study investigated the role and potential mechanism of PCK2 in the occurrence and development of Hepatocellular carcinoma (HCC). The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO), and other databases distinguish the expression of PCK2 and verified by qRT-PCR and Western blotting. Kaplan-Meier was conducted to assess PCK2 survival in HCC. The potential biological function of PCK2 was verified by enrichment analysis and gene set enrichment analysis (GSEA). The correlation between PCK2 expression and immune invasion and checkpoint was found by utilizing Tumor Immune Estimation Resource (TIMER). Lastly, the effects of PCK2 on the proliferation and metastasis of hepatocellular carcinoma cells were evaluated by cell tests, and the expressions of Epithelial mesenchymal transformation (EMT) and apoptosis related proteins were detected. PCK2 is down-regulated in HCC, indicating a poor prognosis. PCK2 gene mutation accounted for 1.3% of HCC. Functional enrichment analysis indicated the potential of PCK2 as a metabolism-related therapeutic target. Subsequently, we identified several signaling pathways related to the biological function of PCK2. The involvement of PCK2 in immune regulation was verified and key immune checkpoints were predicted. Ultimately, after PCK2 knockdown, cell proliferation and migration were significantly increased, and N-cadherin and vimentin expression were increased. PCK2 has been implicated in immune regulation, proliferation, and metastasis of hepatocellular carcinoma, and is emerging as a novel predictive biomarker and metabolic-related clinical target.
Collapse
Affiliation(s)
| | | | - Youzhi Ye
- Kunming Medical University, Kunming, China
| | | | - Hanfei Huang
- The First Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, China.
| | - Zhong Zeng
- The First Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, China.
| |
Collapse
|
6
|
Rai R, Lightfoot S, Benbrook DM. Manipulation of metabolic responses enhances SHetA2 efficacy without toxicity in cervical cancer cell lines and xenografts. Gynecol Oncol 2024; 180:44-54. [PMID: 38052108 PMCID: PMC10922646 DOI: 10.1016/j.ygyno.2023.11.013] [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: 10/12/2023] [Revised: 11/10/2023] [Accepted: 11/12/2023] [Indexed: 12/07/2023]
Abstract
OBJECTIVE The high frequency of cervical cancer recurrence after primary therapy necessitates alternative treatments. High-risk human papillomavirus (HR-HPV) causes cervical cancer and it's continued presence supports elevated metabolism, proliferation and survival of cancer cells. The low-to-no toxicity new investigational drug, SHetA2, counteracts high-risk human papillomavirus (HR-HPV) effects on cell proliferation and survival in cervical cancer cells and xenograft tumors by disrupting heat shock protein 70 chaperone protection of oncogenic proteins. Our objective was to study the involvement of metabolism in SHetA2 effects on cervical cancer cells and tumors. METHODS SHetA2-mediated proteomic and metabolic effects were measured in HR-HPV-positive CaSKi and SiHa and HR-HPV-negative C-33 A cervical cancer cell lines. Combined treatment with 2-deoxyglucose (2-DG) was evaluated in cell culture and SiHa xenografts. RESULTS SHetA2 inhibited oxidative phosphorylation (OxPhos) and altered levels of proteins involved in metabolism, protein synthesis, and DNA replication and repair. Cervical cancer cells responded by elevating glycolysis. Inhibition of the glycolytic responses using galactose media or 2-DG increased SHetA2 sensitivity of two HR-HPV-positive, but not an HR-HPV-negative cervical cancer cell line. Interaction of 2-DG and SHetA2 was synergistic in HR-HPV positive cell lines in association with augmentation of SHetA2 ATP reduction, but not SHetA2 DNA damage induction. These results were verified in a SiHa xenograft tumor model without evidence of toxicity. CONCLUSIONS Compensatory glycolysis counteracts OxPhos inhibition in SHetA2-treated HR-HPV-positive cervical cancer cell lines. Prevention of compensatory glycolysis with 2-DG or another glycolysis inhibitor has the potential to improve SHetA2 therapy without toxicity.
Collapse
Affiliation(s)
- Rajani Rai
- Section of Gynecologic Oncology, Department of Obstetrics and Gynecology, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, USA
| | - Stanley Lightfoot
- Department of Pathology, University of Oklahoma Health Sciences Center, USA
| | - Doris Mangiaracina Benbrook
- Section of Gynecologic Oncology, Department of Obstetrics and Gynecology, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, USA.
| |
Collapse
|
7
|
Qiu X, Li Y, Zhang Z. Crosstalk between oxidative phosphorylation and immune escape in cancer: a new concept of therapeutic targets selection. Cell Oncol (Dordr) 2023; 46:847-865. [PMID: 37040057 DOI: 10.1007/s13402-023-00801-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2023] [Indexed: 04/12/2023] Open
Abstract
BACKGROUND Cancer is increasingly recognized as a metabolic disease, with evidence suggesting that oxidative phosphorylation (OXPHOS) plays a significant role in the progression of numerous cancer cells. OXPHOS not only provides sufficient energy for tumor tissue survival but also regulates conditions for tumor proliferation, invasion, and metastasis. Alterations in OXPHOS can also impair the immune function of immune cells in the tumor microenvironment, leading to immune evasion. Therefore, investigating the relationship between OXPHOS and immune escape is crucial in cancer-related research. This review aims to summarize the effects of transcriptional, mitochondrial genetic, metabolic regulation, and mitochondrial dynamics on OXPHOS in different cancers. Additionally, it highlights the role of OXPHOS in immune escape by affecting various immune cells. Finally, it concludes with an overview of recent advances in antitumor strategies targeting both immune and metabolic processes and proposes promising therapeutic targets by analyzing the limitations of current targeted drugs. CONCLUSIONS The metabolic shift towards OXPHOS contributes significantly to tumor proliferation, progression, metastasis, immune escape, and poor prognosis. A thorough investigation of concrete mechanisms of OXPHOS regulation in different types of tumors and the combination usage of OXPHOS-targeted drugs with existing immunotherapies could potentially uncover new therapeutic targets for future antitumor therapies.
Collapse
Affiliation(s)
- Xutong Qiu
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Head and Neck Cancer Surgery, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Yi Li
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Head and Neck Cancer Surgery, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Zhuoyuan Zhang
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China.
- Department of Head and Neck Cancer Surgery, West China School of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
8
|
Yu Y, Li J, Ren K. Phosphoenolpyruvate carboxykinases as emerging targets in cancer therapy. Front Cell Dev Biol 2023; 11:1196226. [PMID: 37250903 PMCID: PMC10217351 DOI: 10.3389/fcell.2023.1196226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 04/19/2023] [Indexed: 05/31/2023] Open
Abstract
Metabolic reprogramming is commonly accompanied by alterations in the expression of metabolic enzymes. These metabolic enzymes not only catalyze the intracellular metabolic reaction, but also participate in a series of molecular events to regulate tumor initiation and development. Thus, these enzymes may act as promising therapeutic targets for tumor management. Phosphoenolpyruvate carboxykinases (PCKs) are the key enzymes involved in gluconeogenesis, which mediates the conversion of oxaloacetate into phosphoenolpyruvate. Two isoforms of PCK, namely cytosolic PCK1 and mitochondrial PCK2, has been found. PCK not only participates in the metabolic adaptation, but also regulates immune response and signaling pathways for tumor progression. In this review, we discussed the regulatory mechanisms of PCKs expression including transcription and post-translational modification. We also summarized the function of PCKs in tumor progression in different cellular contexts and explores its role in developing promising therapeutic opportunities.
Collapse
Affiliation(s)
- Yong Yu
- Department of Ophthalmology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jingying Li
- Department of Health Management, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Kaiming Ren
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
9
|
Safi A, Saberiyan M, Sanaei MJ, Adelian S, Davarani Asl F, Zeinaly M, Shamsi M, Ahmadi R. The role of noncoding RNAs in metabolic reprogramming of cancer cells. Cell Mol Biol Lett 2023; 28:37. [PMID: 37161350 PMCID: PMC10169341 DOI: 10.1186/s11658-023-00447-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 04/11/2023] [Indexed: 05/11/2023] Open
Abstract
Metabolic reprogramming is a well-known feature of cancer that allows malignant cells to alter metabolic reactions and nutrient uptake, thereby promoting tumor growth and spread. It has been discovered that noncoding RNAs (ncRNAs), including microRNA (miRNA), long noncoding RNA (lncRNA), and circular RNA (circRNA), have a role in a variety of biological functions, control physiologic and developmental processes, and even influence disease. They have been recognized in numerous cancer types as tumor suppressors and oncogenic agents. The role of ncRNAs in the metabolic reprogramming of cancer cells has recently been noticed. We examine this subject, with an emphasis on the metabolism of glucose, lipids, and amino acids, and highlight the therapeutic use of targeting ncRNAs in cancer treatment.
Collapse
Affiliation(s)
- Amir Safi
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mohammadreza Saberiyan
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mohammad-Javad Sanaei
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samaneh Adelian
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Fateme Davarani Asl
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mahsa Zeinaly
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht, Iran
| | - Mahdi Shamsi
- Department of Cell and Molecular Biology, Najafabad Branch, Islamic Azad University, Najafabad, Iran
| | - Reza Ahmadi
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Rahmatiyeh Region, Shahrekord, Iran.
| |
Collapse
|
10
|
Jiao Z, Pan Y, Chen F. The Metabolic Landscape of Breast Cancer and Its Therapeutic Implications. Mol Diagn Ther 2023; 27:349-369. [PMID: 36991275 DOI: 10.1007/s40291-023-00645-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/23/2023] [Indexed: 03/31/2023]
Abstract
Breast cancer is the most common malignant tumor globally as of 2020 and remains the second leading cause of cancer-related death among female individuals worldwide. Metabolic reprogramming is well recognized as a hallmark of malignancy owing to the rewiring of multiple biological processes, notably, glycolysis, oxidative phosphorylation, pentose phosphate pathway, as well as lipid metabolism, which support the demands for the relentless growth of tumor cells and allows distant metastasis of cancer cells. Breast cancer cells are well documented to reprogram their metabolism via mutations or inactivation of intrinsic factors such as c-Myc, TP53, hypoxia-inducible factor, and the PI3K/AKT/mTOR pathway or crosstalk with the surrounding tumor microenvironments, including hypoxia, extracellular acidification and interaction with immune cells, cancer-associated fibroblasts, and adipocytes. Furthermore, altered metabolism contributes to acquired or inherent therapeutic resistance. Therefore, there is an urgent need to understand the metabolic plasticity underlying breast cancer progression as well as to dictate metabolic reprogramming that accounts for the resistance to standard of care. This review aims to illustrate the altered metabolism in breast cancer and its underlying mechanisms, as well as metabolic interventions in breast cancer treatment, with the intention to provide strategies for developing novel therapeutic treatments for breast cancer.
Collapse
Affiliation(s)
- Zhuoya Jiao
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, No. 350, Longzihu Road, Xinzhan District, Hefei, 230012, China
| | - Yunxia Pan
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, No. 350, Longzihu Road, Xinzhan District, Hefei, 230012, China
| | - Fengyuan Chen
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, No. 350, Longzihu Road, Xinzhan District, Hefei, 230012, China.
- Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China.
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China.
| |
Collapse
|
11
|
Han Z, Wang Y, Han L, Yang C. RPN2 in cancer: An overview. Gene 2023; 857:147168. [PMID: 36621657 DOI: 10.1016/j.gene.2023.147168] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 12/07/2022] [Accepted: 01/03/2023] [Indexed: 01/07/2023]
Abstract
Oncogenes together with tumor suppresser genes are confirmed to regulate tumor phenotype in human cancers. RPN2, widely verified as an oncogene, encodes a protein that is part of an N-oligosaccharyl transferase, and is observed to be aberrantly expressed in human malignancies. Accumulating evidence unveils the vital functions of RPN2, contributing to tumorigenicity, metastasis, progression, and multi-drug resistance. Furthermore, previous studies partly indicated that RPN2 was involved in tumor progression via contributing to N-glycosylation and regulating multiple signaling pathways. In addition, RPN2 was also confirmed as a downstream target involved in tumor progression. Moreover, with demonstrated prognosis value and therapeutic target, RPN2 was also determined as a promising biomarker for forecasting patients' prognostic and therapy efficacy. In the present review, we aimed to summarize the present studies of RPN2 in cancer, and enhance the understanding of RPN2's extensive functions and clinical significances.
Collapse
Affiliation(s)
- Zhengxuan Han
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China; Hubei Key Laboratory of Tumour Biological Behaviours, Wuhan, China; Hubei Cancer Clinical Study Center, Wuhan, China; The Clinical Medical Research Center of Peritoneal Cancer of Wuhan, Wuhan, China
| | - You Wang
- Department of Stomatology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lei Han
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China; Hubei Key Laboratory of Tumour Biological Behaviours, Wuhan, China; Hubei Cancer Clinical Study Center, Wuhan, China; The Clinical Medical Research Center of Peritoneal Cancer of Wuhan, Wuhan, China
| | - Chaogang Yang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China; Hubei Key Laboratory of Tumour Biological Behaviours, Wuhan, China; Hubei Cancer Clinical Study Center, Wuhan, China; The Clinical Medical Research Center of Peritoneal Cancer of Wuhan, Wuhan, China.
| |
Collapse
|
12
|
Xiong C, Ling H, Hao Q, Zhou X. Cuproptosis: p53-regulated metabolic cell death? Cell Death Differ 2023; 30:876-884. [PMID: 36755067 PMCID: PMC10070433 DOI: 10.1038/s41418-023-01125-0] [Citation(s) in RCA: 109] [Impact Index Per Article: 54.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 09/22/2022] [Accepted: 09/29/2022] [Indexed: 02/10/2023] Open
Abstract
Cuproptosis is a novel type of copper-induced cell death that primarily occurs in cells that utilize oxidative phosphorylation as the main metabolic pathway to produce energy. Copper directly associates with the lipoylated proteins of the tricarboxylic acid cycle, leading to the disulfide-bond-dependent aggregation of these lipoylated proteins, destabilization of the iron-sulfur cluster proteins, and consequent proteotoxic stress. Cancer cells prefer glycolysis (Warburg effect) to oxidative phosphorylation for producing intermediate metabolites and energy, thereby achieving resistance to cuproptosis. Interestingly, the tumor suppressor p53 is a crucial metabolic regulator that inhibits glycolysis and drives a metabolic switch towards oxidative phosphorylation in cancer cells. Additionally, p53 regulates the biogenesis of iron-sulfur clusters and the copper chelator glutathione, which are two critical components of the cuproptotic pathway, suggesting that this tumor suppressor might play a role in cuproptosis. Furthermore, the possible roles of mutant p53 in regulating cuproptosis are discussed. In this essay, we review the recent progress in the understanding of the mechanism underlying cuproptosis, revisit the roles of p53 in metabolic regulation and iron-sulfur cluster and glutathione biosynthesis, and propose several potential mechanisms for wild-type and mutant p53-mediated cuproptosis regulation.
Collapse
Affiliation(s)
- Chen Xiong
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Hong Ling
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Department of Breast Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China.,Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Qian Hao
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Xiang Zhou
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China. .,Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
13
|
Hu Y, Liu H, Xiao X, Yu Q, Deng R, Hua L, Wang J, Wang X. Bone Marrow Mesenchymal Stem Cell-Derived Exosomes Inhibit Triple-Negative Breast Cancer Cell Stemness and Metastasis via an ALKBH5-Dependent Mechanism. Cancers (Basel) 2022; 14:6059. [PMID: 36551544 PMCID: PMC9776833 DOI: 10.3390/cancers14246059] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 12/05/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Abnormal N6-methyladenosine (m6A) modification caused by m6A regulators is a common characteristic in various tumors. However, little is known about the role of m6A regulator AlkB homolog 5 (ALKBH5) in triple-negative breast cancer (TNBC). In this study, we analyzed the influence of ALKBH5 on the stemness of TNBC and the molecular mechanism using bioinformatics analysis and in vivo animal experiments. METHODS RNA expression data and single-cell RNA sequencing (scRNA-seq) data were downloaded from the TCGA and GEO databases. Following intersection analysis, key genes involved in the TNBC cell stemness were determined, which was followed by functional enrichment analysis, PPI and survival analysis. Exosomes were extracted from bone marrow mesenchymal stem cells (BMSC-Exos) where ALKBH5 inhibition assay was conducted to verify their function in the biological characteristics of TNBC cells. RESULTS Bioinformatics analysis revealed 45 key genes of ALKBH5 regulating TNBC cell stemness. In addition, UBE2C was predicted as a key downstream gene and p53 was predicted as a downstream signaling of ALKBH5. In vivo data confirmed that ALKBH5 upregulated UBE2C expression by regulating the m6A modification of UBE2C and reduced p53 expression, thus promoting the stemness, growth and metastasis of TNBC cells. BMSC-Exos suppressed the tumor stemness, growth and metastasis of TNBC cells and ALKBH5 shRNA-loaded BMSC-Exos showed a more significant suppressive role. CONCLUSION Taken together, our findings indicated that ALKBH5 shRNA-loaded BMSC-Exos reduced TNBC cell stemness, growth and metastasis and define a promising strategy to treat TNBC.
Collapse
Affiliation(s)
- Yun Hu
- Department of General Surgery, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing 210009, China
| | - Hanyuan Liu
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210012, China
| | - Xiudi Xiao
- Department of Breast Surgery, Affiliated People’s Hospital of Jiangsu University & Zhenjiang First People’s Hospital, Zhenjiang 212000, China
| | - Qiao Yu
- Department of General Surgery, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing 210009, China
| | - Rong Deng
- Department of General Surgery, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing 210009, China
| | - Lixin Hua
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jinhua Wang
- Department of Female Tumor, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing 210009, China
| | - Xinwei Wang
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing 210009, China
| |
Collapse
|
14
|
Mutant p53 in cancer: from molecular mechanism to therapeutic modulation. Cell Death Dis 2022; 13:974. [PMID: 36400749 PMCID: PMC9674619 DOI: 10.1038/s41419-022-05408-1] [Citation(s) in RCA: 149] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 11/02/2022] [Accepted: 11/04/2022] [Indexed: 11/19/2022]
Abstract
TP53, a crucial tumor suppressor gene, is the most commonly mutated gene in human cancers. Aside from losing its tumor suppressor function, mutant p53 (mutp53) often acquires inherent, novel oncogenic functions, which is termed "gain-of-function". Emerging evidence suggests that mutp53 is highly associated with advanced malignancies and poor prognosis, which makes it a target for development of novel cancer therapies. Herein, we provide a summary of our knowledge of the mutp53 types and mutp53 spectrum in cancers. The mechanisms of mutp53 accumulation and gain-of-function are also summarized. Furthermore, we discuss the gain-of-function of mutp53 in cancers: genetic instability, ferroptosis, microenvironment, and stemness. Importantly, the role of mutp53 in the clinic is also discussed, particularly with regard to chemotherapy and radiotherapy. Last, emphasis is given to emerging strategies on how to target mutp53 for tumor therapy. Thus, this review will contribute to better understanding of the significance of mutp53 as a target for therapeutic strategies.
Collapse
|
15
|
Wang CY, Wang CH, Mai RT, Chen TW, Li CW, Chao CH. Mutant p53-microRNA-200c-ZEB2-Axis-Induced CPT1C Elevation Contributes to Metabolic Reprogramming and Tumor Progression in Basal-Like Breast Cancers. Front Oncol 2022; 12:940402. [PMID: 35936710 PMCID: PMC9351713 DOI: 10.3389/fonc.2022.940402] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 06/08/2022] [Indexed: 11/23/2022] Open
Abstract
TP53 is mutated in more than 80% of basal-like breast cancers (BLBCs). BLBCs with TP53 mutation are usually high-grade and have worse responses to chemotherapy, leading to poor clinical outcomes. Wild-type p53 (WTp53) is well-accepted to promote fatty acid oxidation (FAO); however, in this study, we demonstrate that mutant p53 (Mutp53) enhances FAO activity through constitutively upregulating CPT1C via dysregulating the miR-200c-ZEB2 axis. Sustained CPT1C expression contributes to the metabolic preference of FAO, epithelial-mesenchymal transition (EMT) phenotypes, migration, invasion, and cancer stemness in BLBC, which is mediated by modulating the redox status. Furthermore, interference of CPT1C expression impairs tumor growth and pulmonary colonization of BLBC cells in vivo, and even postpones the occurrence of spontaneous metastasis, resulting in a prolonged disease-specific survival (DSS). Consistently, clinical validation reveals that high CPT1C is observed in breast cancer patients with metastasis and is correlated with poor overall, disease-free, progression-free, and disease-specific survival in BLBC patients. Together, unlike WTp53 which transiently transactivates CPT1C, Mutp53 provides long-term benefits through sustaining CPT1C expression by disturbing the miR-200c-ZEB2 axis, which potentiates FAO and facilitates tumor progression in BLBC, suggesting that targeting Mutp53-CPT1C-driven metabolic reprogramming is promising to serve as novel therapeutic strategies for BLBC in the future.
Collapse
Affiliation(s)
- Chen-Yun Wang
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
- Center For Intelligent Drug Systems and Smart Bio-devices (IDSB), National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Cing-Hong Wang
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
- Center For Intelligent Drug Systems and Smart Bio-devices (IDSB), National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Ru-Tsun Mai
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
- Center For Intelligent Drug Systems and Smart Bio-devices (IDSB), National Yang Ming Chiao Tung University, Hsinchu, Taiwan
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Ting-Wen Chen
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
- Center For Intelligent Drug Systems and Smart Bio-devices (IDSB), National Yang Ming Chiao Tung University, Hsinchu, Taiwan
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Chia-Wei Li
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chi-Hong Chao
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
- Center For Intelligent Drug Systems and Smart Bio-devices (IDSB), National Yang Ming Chiao Tung University, Hsinchu, Taiwan
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
- *Correspondence: Chi-Hong Chao,
| |
Collapse
|
16
|
Wang CY, Chao CH. p53-Mediated Indirect Regulation on Cellular Metabolism: From the Mechanism of Pathogenesis to the Development of Cancer Therapeutics. Front Oncol 2022; 12:895112. [PMID: 35707366 PMCID: PMC9190692 DOI: 10.3389/fonc.2022.895112] [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: 03/13/2022] [Accepted: 04/28/2022] [Indexed: 11/13/2022] Open
Abstract
The transcription factor p53 is the most well-characterized tumor suppressor involved in multiple cellular processes, which has expanded to the regulation of metabolism in recent decades. Accumulating evidence reinforces the link between the disturbance of p53-relevant metabolic activities and tumor development. However, a full-fledged understanding of the metabolic roles of p53 and the underlying detailed molecular mechanisms in human normal and cancer cells remain elusive, and persistent endeavor is required to foster the entry of drugs targeting p53 into clinical use. This mini-review summarizes the indirect regulation of cellular metabolism by wild-type p53 as well as mutant p53, in which mechanisms are categorized into three major groups: through modulating downstream transcriptional targets, protein-protein interaction with other transcription factors, and affecting signaling pathways. Indirect mechanisms expand the p53 regulatory networks of cellular metabolism, making p53 a master regulator of metabolism and a key metabolic sensor. Moreover, we provide a brief overview of recent achievements and potential developments in the therapeutic strategies targeting mutant p53, emphasizing synthetic lethal methods targeting mutant p53 with metabolism. Then, we delineate synthetic lethality targeting mutant p53 with its indirect regulation on metabolism, which expands the synthetic lethal networks of mutant p53 and broadens the horizon of developing novel therapeutic strategies for p53 mutated cancers, providing more opportunities for cancer patients with mutant p53. Finally, the limitations and current research gaps in studies of metabolic networks controlled by p53 and challenges of research on p53-mediated indirect regulation on metabolism are further discussed.
Collapse
Affiliation(s)
- Chen-Yun Wang
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu, Taiwan.,Center For Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Chi-Hong Chao
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu, Taiwan.,Center For Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu, Taiwan.,Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| |
Collapse
|
17
|
Guo Q, Pei XH, Chu AJ, Guo YB, Fan YY, Wang CH, Zhang SJ, Sun SQ, Liu YF, Wang X. The mechanism of action of Fangji Huangqi Decoction on epithelial-mesenchymal transition in breast cancer using high-throughput next-generation sequencing and network pharmacology. JOURNAL OF ETHNOPHARMACOLOGY 2022; 284:114793. [PMID: 34728317 DOI: 10.1016/j.jep.2021.114793] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 10/27/2021] [Accepted: 10/28/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Fangji Huangqi Decoction (FHD) is widely used in traditional Chinese medicine (TCM). FHD has been hypothesized to inhibit the epithelial-mesenchymal transition (EMT) process, which may positively impact breast cancer prevention and treatment. However, its exact mechanism of action is still unknown. AIM OF THE STUDY This study aimed to screen potential targets of FHD for the treatment of EMT in breast cancer through network pharmacology, and to verify their therapeutic effects in vitro experiments and high-throughput second-generation sequencing. MATERIALS AND METHODS The data sets of effective components and targets of FHD were established through the Traditional Chinese Medicine Systems Pharmacology database. The GeneCards and OMIM databases were used to establish breast cancer-related target datasets, which were then matched with the TCM target data. The interaction between key target proteins was analyzed using the STRING database; the gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) databases were used to identify the associated biological processes and enriched signal pathways, respectively. The active ingredient disease target network was analyzed using Cytoscape. Finally, next generation sequencing was used to verify the related pathways of FHD intervention in EMT in breast cancer. High-content screening was used to identify the genes/pathways affected by FHD. MDA-MB-231 and HCC-1937 breast cancer cell lines were used to evaluate the impact of FHD on migration, invasion, and EMT. RESULTS Eighty possible significant targets were identified for the treatment of breast cancer EMT with FHD; GO and KEGG were used to identify 173 cell biological processes associated with breast cancer (P < 0.05), including the NF-κB and PI3K-Akt signaling pathways. The high-throughput sequencing and network pharmacology results were highly consistent. The migration and invasion ability of MDA-MB-231 cells was reduced and their EMT status could be reversed by DSHR2 knockdown. The results of morphology and scratch assays showed that FHD could improve the EMT status of HCC-1973. CONCLUSIONS This study provides more evidence to support the clinical application of FHD, which has reliable interventional effects on breast cancer EMT. Its therapeutic effects may involve a multi-target, multi-pathway, and multi-mechanism effect.
Collapse
Affiliation(s)
- Qi Guo
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, 100029, China.
| | - Xiao-Hua Pei
- Beijing University of Chinese Medicine Eighth Affiliated Hospital, Xiamen, 361001, China.
| | - Ai-Jing Chu
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, 100029, China.
| | - Yu-Bo Guo
- Beijing Municipal Hospital of Traditional Chinese Medicine, Beijing, 100010, China.
| | - Ying-Yi Fan
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, 100029, China.
| | - Chun-Hui Wang
- Fangshan Hospital Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Shu-Jing Zhang
- Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Shi-Qing Sun
- Henan Provincial Hospital of Traditional Chinese Medicine, Zhengzhou, 450000, China.
| | - Yu-Fei Liu
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, 100029, China.
| | - Xuan Wang
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, 100029, China
| |
Collapse
|
18
|
Crosslink between p53 and metastasis: focus on epithelial-mesenchymal transition, cancer stem cell, angiogenesis, autophagy, and anoikis. Mol Biol Rep 2021; 48:7545-7557. [PMID: 34519942 DOI: 10.1007/s11033-021-06706-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/07/2021] [Indexed: 01/05/2023]
Abstract
INTRODUCTION P53, as a tumor suppressor gene, is believed to be one of the most mutated genes in cancer cells. The mutant forms of this protein often play a tumorigenic role in cancer cells. Recent evidence shows that p53 plays a critical role in the migration, metastasis, and invasion of cancer cells. The present article aims to investigate the molecular mechanism that induces metastasis in cancer cells generated by the mutant P53, and to highlight the compounds targeting mutant-p53 together with their clinical applications. METHODS A detailed literature search was conducted to find information about the role of the mutant-p53 in the processes involved in metastasis in various databases. RESULTS A growing body of evidence suggests that Mutant-p53 enhances tumor metastasis affecting the Epithelial-mesenchymal transition (EMT) process, cancer stem cells, angiogenesis, autophagy, anoikis, and any other mechanisms regarding metastasis. CONCLUSIONS Taken together, targeting mutant-p53 by altering the processes involved in metastasis could be a potential therapeutic strategy in the treatment of metastatic cancer.
Collapse
|