1
|
Tai L, Zhu D, Tang P, Li J, Li J, Li P, Tao Z, Lei H, Miao K, Wang HX, Lin S, Zhang L, Dou M, Han Y, Shen HM, Deng C, Wang L, Di LJ. Reciprocal stabilization of CtBP and TRIM28 represses autophagy to promote metastasis. Nat Struct Mol Biol 2025:10.1038/s41594-025-01554-0. [PMID: 40374929 DOI: 10.1038/s41594-025-01554-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 04/05/2025] [Indexed: 05/18/2025]
Abstract
Deciphering the processes through which cancer cells overcome stress, escape a repressive microenvironment and metastasize remains a challenge. Autophagy has been demonstrated to regulate cancer metastasis and C-terminal binding protein (CtBP) has been previously implicated in promoting metastasis in breast cancer. Here we identify the formation of a complex between CtBP and tripartite-motif-containing protein 28 (TRIM28) in the nucleus. Interestingly, this complex regulates the stability of both proteins, as the removal of either partner leads to degradation of the other. Furthermore, the stability of this complex in the nucleus inhibits autophagy through two independent mechanisms. Firstly, the formation of the complex sequesters TRIM28 in the nucleus, preventing its involvement in and its degradation through autophagy. Secondly, this complex participates in the suppression of PTEN expression and leads to inhibition of Unc-51-like kinase 1-mediated autophagy through activation of the protein kinase B-mammalian target of rapamycin pathway. Using mammary gland-specific CtBP-knockout mice, we demonstrate that repression of autophagy by the CtBP-TRIM28 complex modulates luminal duct formation. In breast cancer models, CtBP-TRIM28-dependent inhibition of cellular autophagy also promotes malignant metastasis. Therefore, our study reveals similarities between the mechanisms driving tumor progression and those involved in normal mammary gland development, potentially helping to pave the way toward targeted intervention in breast cancer metastasis.
Collapse
Affiliation(s)
- Lixin Tai
- Department of Biological Sciences, Faculty of Health Sciences, University of Macau, Macau, China
- Ministry of Education Frontiers Science Center for Precision Oncology (FSCPO), University of Macau, Macau, China
| | - Dongliang Zhu
- Department of Biological Sciences, Faculty of Health Sciences, University of Macau, Macau, China
- Ministry of Education Frontiers Science Center for Precision Oncology (FSCPO), University of Macau, Macau, China
| | - Ping Tang
- Department of Biological Sciences, Faculty of Health Sciences, University of Macau, Macau, China
- Ministry of Education Frontiers Science Center for Precision Oncology (FSCPO), University of Macau, Macau, China
| | - Jiajia Li
- Department of Biological Sciences, Faculty of Health Sciences, University of Macau, Macau, China
- Ministry of Education Frontiers Science Center for Precision Oncology (FSCPO), University of Macau, Macau, China
| | - Junyi Li
- Department of Biological Sciences, Faculty of Health Sciences, University of Macau, Macau, China
- Ministry of Education Frontiers Science Center for Precision Oncology (FSCPO), University of Macau, Macau, China
| | - Peipei Li
- Department of Biological Sciences, Faculty of Health Sciences, University of Macau, Macau, China
- Ministry of Education Frontiers Science Center for Precision Oncology (FSCPO), University of Macau, Macau, China
| | - Zhonghua Tao
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Haipeng Lei
- Department of Biological Sciences, Faculty of Health Sciences, University of Macau, Macau, China
- Ministry of Education Frontiers Science Center for Precision Oncology (FSCPO), University of Macau, Macau, China
| | - Kai Miao
- Department of Biological Sciences, Faculty of Health Sciences, University of Macau, Macau, China
- Ministry of Education Frontiers Science Center for Precision Oncology (FSCPO), University of Macau, Macau, China
| | - Hong-Xia Wang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shuhai Lin
- State Key Laboratory of Cellular Stress Biology, School of Life Science, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Lei Zhang
- Department of Pathology, Basic Medical Science College, Harbin Medical University, Harbin, China
| | - Man Dou
- Department of Biological Sciences, Faculty of Health Sciences, University of Macau, Macau, China
- Metabolomics core, Faculty of Health Sciences, University of Macau, Macau, China
| | - Yu Han
- Department of Biological Sciences, Faculty of Health Sciences, University of Macau, Macau, China
- Genomics, Bioinformatics and Single Cell Analysis Core, Faculty of Health Sciences, University of Macau, Macau, China
| | - Han-Ming Shen
- Department of Biological Sciences, Faculty of Health Sciences, University of Macau, Macau, China
- Ministry of Education Frontiers Science Center for Precision Oncology (FSCPO), University of Macau, Macau, China
| | - Chuxia Deng
- Department of Biological Sciences, Faculty of Health Sciences, University of Macau, Macau, China
- Ministry of Education Frontiers Science Center for Precision Oncology (FSCPO), University of Macau, Macau, China
| | - Li Wang
- Department of Biological Sciences, Faculty of Health Sciences, University of Macau, Macau, China
- Ministry of Education Frontiers Science Center for Precision Oncology (FSCPO), University of Macau, Macau, China
- Metabolomics core, Faculty of Health Sciences, University of Macau, Macau, China
| | - Li-Jun Di
- Department of Biological Sciences, Faculty of Health Sciences, University of Macau, Macau, China.
- Ministry of Education Frontiers Science Center for Precision Oncology (FSCPO), University of Macau, Macau, China.
- Department of Pathology, Basic Medical Science College, Harbin Medical University, Harbin, China.
| |
Collapse
|
2
|
Song Q, Sui J, Yang Y, Zhang H, Ya L, Yang L. Fructose-1,6-bisphosphatase 1 in cancer: Dual roles, mechanistic insights, and therapeutic potential - A comprehensive review. Int J Biol Macromol 2025; 293:139273. [PMID: 39753180 DOI: 10.1016/j.ijbiomac.2024.139273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 12/22/2024] [Accepted: 12/26/2024] [Indexed: 01/07/2025]
Abstract
Fructose-1,6-bisphosphatase 1 (FBP1) is a key gluconeogenic enzyme that plays complex and context-dependent roles in cancer biology. This review comprehensively examines FBP1's dual functions as both a tumor suppressor and an oncogene across various cancer types. In many cancers, such as hepatocellular carcinoma, clear cell renal cell carcinoma, and lung cancer, downregulation of FBP1 contributes to tumor progression through metabolic reprogramming, promoting glycolysis, and altering the tumor microenvironment. Conversely, in certain contexts like breast and prostate cancers, FBP1 overexpression is associated with tumor promotion, indicating its oncogenic potential. The review explores FBP1's interactions with immune cells within the tumor microenvironment, influencing immune surveillance and tumor immune escape mechanisms. Additionally, FBP1 emerges as a promising diagnostic and prognostic biomarker, with expression levels correlating with patient outcomes in multiple cancers. Future therapeutic strategies targeting FBP1 are discussed, including inhibitors, activators, epigenetic modulation, and combination therapies, while addressing the challenges posed by its dual nature. Understanding the multifaceted roles of FBP1 offers valuable insights into cancer metabolism and opens avenues for personalized therapeutic interventions.
Collapse
Affiliation(s)
- Qinghang Song
- School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Jiazhen Sui
- School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Yuxuan Yang
- School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Huhu Zhang
- School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Li Ya
- School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Lina Yang
- School of Basic Medicine, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
3
|
Chen W, Huang D, Wu R, Wen Y, Zhong Y, Guo J, Liu A, Lin L. A multi-functional integrated nanoplatform based on a tumor microenvironment-responsive PtAu/MnO 2 cascade nanoreactor with multi-enzymatic activities for multimodal synergistic tumor therapy. J Colloid Interface Sci 2025; 679:957-974. [PMID: 39486234 DOI: 10.1016/j.jcis.2024.10.160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/24/2024] [Accepted: 10/25/2024] [Indexed: 11/04/2024]
Abstract
The utilization or improvement of tumor microenvironment (TME) has become a breakthrough in emerging oncology therapies. To address the limited therapeutic efficacy of single modality, a multi-functional integrated nanoplatform based on a TME-responsive PtAu/MnO2 cascade nanoreactor with multi-enzymatic activities was developed for multimodal synergistic tumor therapy. Benefiting from the slightly acidic environment and high-level glutathione (GSH) in TME, PtAu/MnO2 cascade nanoreactor consumed GSH, followed by the reductive generation of manganese ion (Mn2+) and the release of PtAu nanoparticles (NPs). Then, the multimodal synergistic tumor therapy was activated as follows. First, GSH depletion inhibited the activity of glutathione peroxidase 4 and led to the accumulation of lipid peroxidation, thereby inducing tumor cell ferroptosis. Second, PtAu NPs exhibited catalase-like, glucose oxidase-like and nicotinamide adenine dinucleotide (NADH) oxidase-like activities, which generated oxygen for the cascade reaction to alleviate hypoxia and further depleted glucose, NADH and adenosine triphosphate, leading to the inhibition of tumor cell proliferation via starvation therapy. Third, the production of reactive oxygen species by the oxidase- and peroxidase-like activities of PtAu NPs and the Fenton-like reaction of Mn2+ simultaneously induced tumor cell apoptosis via chemodynamic therapy. Briefly, the in vitro and in vivo results confirmed that the multi-functional integrated nanoplatform based on a PtAu/MnO2 cascade nanoreactor with five nanozyme activities demonstrated outstanding biocompatibility and greater inhibition of tumor growth via synergistic ferroptosis/starvation therapy/apoptosis.
Collapse
Affiliation(s)
- Wenxin Chen
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Dandan Huang
- Department of Pharmacy, Fujian Children's Hospital, Fuzhou, Fujian 350000, China
| | - Ruimei Wu
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Yujuan Wen
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Yu Zhong
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Jianpeng Guo
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Ailin Liu
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Liqing Lin
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian 350122, China.
| |
Collapse
|
4
|
Hu Y, Zeng C, Li J, Ren S, Shao M, Lei W, Yi J, Han W, Cao J, Zou J, Fei Q, Cheng Z, Liu W. TRIM27 revealing by tumor educated platelet RNA-sequencing, as a potential biomarker for malignant ground-glass opacities diagnosis mediates glycolysis of non-small cell lung cancer cells partially through HOXM1. Transl Lung Cancer Res 2024; 13:2307-2325. [PMID: 39430321 PMCID: PMC11484725 DOI: 10.21037/tlcr-24-157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 07/17/2024] [Indexed: 10/22/2024]
Abstract
Background Efficient ground-glass opacities (GGOs) diagnosis is challenging. A diagnostic method distinguishing malignant from benign GGOs is warranted. In this study, we sought to construct a noninvasive method based on tumor educated platelet (TEP) RNA profiles for malignant GGOs diagnosis and explore the molecular mechanism of the potential biomarker for the first time. Methods Based on TEP RNA-sequencing (TEP RNA-seq) in benign and malignant GGOs, a classification model was constructed using differentially expressed genes (DEGs) and was used to evaluate diagnostic performance. High-throughput quantitative polymerase chain reaction (HT-qPCR) verified 23 genes selected from the top 60 DEGs between benign and malignant GGOs. The correlation between 17 verified DEGs and 22 key glycolytic genes was analyzed. Tripartite motif-containing 27 (TRIM27) overexpressing and knockdown (KD) cell models were constructed using A549 and PC-9 cells, respectively in which cell growth, apoptosis, migration and invasion were evaluated. The protein levels of HK-1/2, PKM1/2, LDHA and GLUT1 were evaluated by western blot. Glycolysis was evaluated through adenosine triphosphate (ATP), reactive oxygen species (ROS), lactate acid (LD) production, glucose uptake, and lactate dehydrogenase (LDH) activity assays. RNA-seq was performed in loss-of TRIM27-KD PC-9 cells to clarify the downstream factors of TRIM27 which was verified using western blot and immunofluorescence double staining. Results In 81 samples, the 1,647-DEG-based classification model exhibited area under the curve (AUC), sensitivity, and specificity values of 0.99 [95% confidence interval (CI): 0.972-1.000], 100%, and 91%, respectively, while the top 60-DEG-based classification model exhibited AUC, sensitivity, and specificity values of 0.986 (95% CI: 0.962-1.000), 98%, and 91%, respectively. TRIM27 achieved AUC of 0.87 in the diagnosis of malignant GGOs, with 83.93% sensitivity, 78.79% specificity, 81.15% accuracy, 77.05% positive predictive value (PPV) and 85.25% negative predictive value (NPV). TRIM27 was highly expressed in non-small cell lung cancer (NSCLC) cells, and accelerated cell migration and invasion. In addition, TRIM27 was found to promote glycolysis in NSCLC cells partially through HMOX1 which was negatively correlated with TRIM27. Conclusions We constructed a novel TEP RNA-seq based classifier for malignant GGOs diagnosis. TRIM27, an important target discovered, could accelerate migration, invasion and regulate glycolysis partially through HMOX1 in NSCLC cells, thus providing scientific support for TRIM27 as a diagnostic biomarker for malignant GGO diagnosis.
Collapse
Affiliation(s)
- Yan Hu
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chao Zeng
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jina Li
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Siying Ren
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Research Unit of Respiratory Disease, Central South University, Hunan Diagnosis and Treatment Center of Respiratory Disease, Changsha, China
| | - Mengqi Shao
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Weixuan Lei
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Junqi Yi
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wei Han
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jieming Cao
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jian Zou
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Quanming Fei
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zeyu Cheng
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wenliang Liu
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
5
|
Hu D, Kobayashi N, Ohki R. FUCA1: An Underexplored p53 Target Gene Linking Glycosylation and Cancer Progression. Cancers (Basel) 2024; 16:2753. [PMID: 39123480 PMCID: PMC11311387 DOI: 10.3390/cancers16152753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/26/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024] Open
Abstract
Cancer is a difficult-to-cure disease with high worldwide incidence and mortality, in large part due to drug resistance and disease relapse. Glycosylation, which is a common modification of cellular biomolecules, was discovered decades ago and has been of interest in cancer research due to its ability to influence cellular function and to promote carcinogenesis. A variety of glycosylation types and structures regulate the function of biomolecules and are potential targets for investigating and treating cancer. The link between glycosylation and carcinogenesis has been more recently revealed by the role of p53 in energy metabolism, including the p53 target gene alpha-L-fucosidase 1 (FUCA1), which plays an essential role in fucosylation. In this review, we summarize roles of glycan structures and glycosylation-related enzymes to cancer development. The interplay between glycosylation and tumor microenvironmental factors is also discussed, together with involvement of glycosylation in well-characterized cancer-promoting mechanisms, such as the epidermal growth factor receptor (EGFR), phosphatidylinositol-3-kinase/protein kinase B (PI3K/Akt) and p53-mediated pathways. Glycan structures also modulate cell-matrix interactions, cell-cell adhesion as well as cell migration and settlement, dysfunction of which can contribute to cancer. Thus, further investigation of the mechanistic relationships among glycosylation, related enzymes and cancer progression may provide insights into potential novel cancer treatments.
Collapse
Affiliation(s)
- Die Hu
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada;
| | - Naoya Kobayashi
- Laboratory of Fundamental Oncology, National Cancer Center Research Institute, Tsukiji 5-1-1, Chuo-ku, Tokyo 104-0045, Japan;
- Department of NCC Cancer Science, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Rieko Ohki
- Laboratory of Fundamental Oncology, National Cancer Center Research Institute, Tsukiji 5-1-1, Chuo-ku, Tokyo 104-0045, Japan;
| |
Collapse
|
6
|
Zhang G, Wei W, Li S, Yang J. Transcription Factor yin-Yang 1 augments nucleoporin 93 oncogene activity and modulates bladder Cancer malignancy. Toxicol In Vitro 2024; 99:105875. [PMID: 38857852 DOI: 10.1016/j.tiv.2024.105875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/12/2024]
Abstract
OBJECTIVE This study aims to investigate the functional interplay between transcription factor YY1 and nucleoporin 93 (NUP93) in regulating the malignancy of bladder cancer cells. METHODS NUP93 expressions in bladder cancer tissues and normal counterparts were analyzed using a public dataset and clinical samples. NUP93 and Yin Yang 1 (YY1) mRNA expression and protein levels in T24 and RT4 cells were determined by Quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting. The effect of NUP93 knockdown on the proliferation, migration, and invasion capabilities of cells was evaluated. Concurrently, transcriptional regulation of NUP93 by YY1 was confirmed using a dual luciferase assay. The effect of NUP93 knockdown on tumorigenesis was evaluate in a subcutaneous xenograft mouse model. RESULTS Elevated levels of NUP93 in bladder cancer tissues and cell lines were observed. Silencing NUP93 significantly suppressed glycolysis, impeded the growth, migration, invasion and tumor formation of bladder cancer cells. The transcription factor YY1 acted as a positive regulator to upregulate NUP93 expression. YY1 overexpression partially rescued the effects of NUP93 silencing on bladder cancer cells. CONCLUSION Our results uncovered transcription factor YY1 as a positive regulator of NUP93 expression, and NUP93 serves as an oncogenic factor to sustain the malignancy of bladder cancer cells. These findings suggest that targeting the YY1-NUP93 axis could offer novel therapeutic strategies for bladder cancer treatment.
Collapse
Affiliation(s)
- Gang Zhang
- Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Wei Wei
- Department of Urology section, Dalian Friendship Hospital, Dalian, Liaoning 116001, China
| | - Shao Li
- Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning,116044, China
| | - Jinyi Yang
- Department of Urology section, Dalian Friendship Hospital, Dalian, Liaoning 116001, China.
| |
Collapse
|
7
|
Fu Y, Tao J, Liu T, Liu Y, Qiu J, Su D, Wang R, Luo W, Cao Z, Weng G, Zhang T, Zhao Y. Unbiasedly decoding the tumor microenvironment with single-cell multiomics analysis in pancreatic cancer. Mol Cancer 2024; 23:140. [PMID: 38982491 PMCID: PMC11232163 DOI: 10.1186/s12943-024-02050-7] [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: 05/07/2024] [Accepted: 06/21/2024] [Indexed: 07/11/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy with a poor prognosis and limited therapeutic options. Research on the tumor microenvironment (TME) of PDAC has propelled the development of immunotherapeutic and targeted therapeutic strategies with a promising future. The emergence of single-cell sequencing and mass spectrometry technologies, coupled with spatial omics, has collectively revealed the heterogeneity of the TME from a multiomics perspective, outlined the development trajectories of cell lineages, and revealed important functions of previously underrated myeloid cells and tumor stroma cells. Concurrently, these findings necessitated more refined annotations of biological functions at the cell cluster or single-cell level. Precise identification of all cell clusters is urgently needed to determine whether they have been investigated adequately and to identify target cell clusters with antitumor potential, design compatible treatment strategies, and determine treatment resistance. Here, we summarize recent research on the PDAC TME at the single-cell multiomics level, with an unbiased focus on the functions and potential classification bases of every cellular component within the TME, and look forward to the prospects of integrating single-cell multiomics data and retrospectively reusing bulk sequencing data, hoping to provide new insights into the PDAC TME.
Collapse
Affiliation(s)
- Yifan Fu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Jinxin Tao
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Tao Liu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yueze Liu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Jiangdong Qiu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Dan Su
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Ruobing Wang
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Wenhao Luo
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Zhe Cao
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Guihu Weng
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Taiping Zhang
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
- Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Yupei Zhao
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
8
|
Jaiswal A, Singh R. A negative feedback loop underlies the Warburg effect. NPJ Syst Biol Appl 2024; 10:55. [PMID: 38789545 PMCID: PMC11126737 DOI: 10.1038/s41540-024-00377-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 04/29/2024] [Indexed: 05/26/2024] Open
Abstract
Aerobic glycolysis, or the Warburg effect, is used by cancer cells for proliferation while producing lactate. Although lactate production has wide implications for cancer progression, it is not known how this effect increases cell proliferation and relates to oxidative phosphorylation. Here, we elucidate that a negative feedback loop (NFL) is responsible for the Warburg effect. Further, we show that aerobic glycolysis works as an amplifier of oxidative phosphorylation. On the other hand, quiescence is an important property of cancer stem cells. Based on the NFL, we show that both aerobic glycolysis and oxidative phosphorylation, playing a synergistic role, are required to achieve cell quiescence. Further, our results suggest that the cells in their hypoxic niche are highly proliferative yet close to attaining quiescence by increasing their NADH/NAD+ ratio through the severity of hypoxia. The findings of this study can help in a better understanding of the link among metabolism, cell cycle, carcinogenesis, and stemness.
Collapse
Affiliation(s)
- Alok Jaiswal
- Department of Chemical Engineering, Indian Institute of Technology Kanpur, Kanpur, 208016, India
| | - Raghvendra Singh
- Department of Chemical Engineering, Indian Institute of Technology Kanpur, Kanpur, 208016, India.
| |
Collapse
|
9
|
Wang M, Shi L, Fan S, Hu C. CTBP2 contributes to cisplatin resistance in lung adenocarcinoma by inhibiting generation of reactive oxygen species. Transl Cancer Res 2024; 13:1695-1706. [PMID: 38737699 PMCID: PMC11082667 DOI: 10.21037/tcr-23-2135] [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: 11/19/2023] [Accepted: 02/29/2024] [Indexed: 05/14/2024]
Abstract
Background Cisplatin (CP) is commonly used for the initial treatment of lung adenocarcinoma (LUAD). Resistance to CP has long been recognized as a significant obstacle to achieving improved therapeutic outcomes. Nevertheless, the intricate molecular mechanisms underlying the phenomenon remain incompletely understood. Methods The present study utilized the University of ALabama at Birmingham CANcer data analysis Portal (UALCAN) and Gene Expression Profiling Interactive Analysis (GEPIA) databases to conduct an analysis of the expression of C-terminal binding protein 2 (CTBP2) in LUAD. The correlation between CTBP2 expression and survival data was investigated by the Kaplan-Meier (K-M) plotter. Subsequently, the roles of CTBP2 in CP resistance were explored by analyzing cell viability, cell apoptosis, reactive oxygen species (ROS), and mitochondrial membrane potential (MMP) in CP-resistant cells (A549/DDP). Results Our data indicated that the CTBP2 expression in LUAD exhibited a significant increase compared to the non-malignant tissues. CTBP2 overexpression showed a correlation to poor survival. CTBP2 knockdown significantly enhanced cell sensitivity to CP in A549/DDP cells. The underlying mechanism is related to promoting ROS production and decreasing MMP after CP treatment. Conclusions CTBP2 expression has been identified as a novel biomarker for resistance to CP, and its downregulation has been found to enhance sensitivity to CP. Therefore, CTBP2 can serve as a predictor related to CP resistance and a viable therapeutic target for CP resistance in LUAD.
Collapse
Affiliation(s)
- Mingqun Wang
- Department of Pharmacy, Changning County Hospital of Traditional Chinese Medicine, Yibin, China
| | - Linshan Shi
- College of Pharmacy (International Academy of Targeted Therapeutics and Innovation), Chongqing University of Arts and Sciences, Chongqing, China
| | - Shiming Fan
- Department of Respiratory and Critical Care Medicine, Changning County Hospital of Traditional Chinese Medicine, Yibin, China
| | - Chunsheng Hu
- College of Pharmacy (International Academy of Targeted Therapeutics and Innovation), Chongqing University of Arts and Sciences, Chongqing, China
- Chongqing Key Laboratory of Chinese Medicine & Health Science, Chongqing Academy of Chinese Materia Medica, Chongqing, China
| |
Collapse
|
10
|
Cheng C, Wang Z, Ding C, Liu P, Xu X, Li Y, Yan Y, Yin X, Chen B, Gu B. Bronchoalveolar Lavage Fluid Microbiota is Associated with the Diagnosis and Prognosis Evaluation of Lung Cancer. PHENOMICS (CHAM, SWITZERLAND) 2024; 4:125-137. [PMID: 38884058 PMCID: PMC11169441 DOI: 10.1007/s43657-023-00135-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 09/25/2023] [Accepted: 10/12/2023] [Indexed: 06/18/2024]
Abstract
The gut microbiota and cancer have been demonstrated to be closely related. However, few studies have explored the bronchoalveolar lavage fluid (BALF) microbiota in patients with lung cancer (LC), specifically the microbiota related to progression-free survival (PFS) in LC. A total of 216 BALF samples were collected including 166 LC and 50 benign pulmonary disease (N-LC) samples, and further sequenced using 16S rRNA amplicon sequencing. Enrolled LC patients were followed up, the therapeutic efficacy was assessed, and PFS was calculated. The associated clinical and microbiota sequencing data were deeply analysed. Distinct differences in the microbial profiles were evident in the lower airways of patients with LC and N-LC, which was also found between non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC). A combined random forest model was built to distinguish NSCLC from SCLC and reached area under curves (AUCs) of 0.919 (95% CI 86.69-97.1%) and 0.893 (95% CI 79.39-99.29%) in the training and test groups, respectively. The lower alpha diversity of the BALF microbiota in NSCLC patients was significantly associated with reduced PFS, although this link was not observed in SCLC. Specifically, NSCLC with a higher abundance of f_Lachnospiraceae, s_Prevotella nigrescens and f_[Mogibacteriaceae] achieved longer PFS. The enrichment of o_Streptophyta and g_Prevotella was observed in SCLC with worse PFS. This study provided a detailed description of the characteristics of BALF microbiota in patients with NSCLC and SCLC simultaneously and provided insights into the role of the diagnosis and prognosis evaluation. Supplementary Information The online version contains supplementary material available at 10.1007/s43657-023-00135-9.
Collapse
Affiliation(s)
- Chen Cheng
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 Jiangsu China
- Branch of National Clinical Research Center for Laboratory Medicine, Nanjing, 210029 Jiangsu China
| | - Zhifeng Wang
- Department of Bioinformatics, 01Life Institute, Shenzhen, 518000 Guangdong China
| | - Chao Ding
- Department of General Surgery, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008 Jiangsu China
| | - Pingli Liu
- Department of Respiratory Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006 Jiangsu China
| | - Xiaoqiang Xu
- Department of Bioinformatics, 01Life Institute, Shenzhen, 518000 Guangdong China
| | - Yan Li
- Department of Respiratory Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006 Jiangsu China
| | - Yi Yan
- Department of Respiratory Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006 Jiangsu China
| | - Xiaocong Yin
- Medical Technology School of Xuzhou Medical University, Xuzhou, 221004 Jiangsu China
| | - Bi Chen
- Department of Respiratory Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006 Jiangsu China
| | - Bing Gu
- Laboratory Medicine, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan 2Nd Rd, Yuexiu District, Guangzhou, 510000 Guangdong China
| |
Collapse
|
11
|
Yin F, Zhang X, Zhang Z, Zhang M, Yin Y, Yang Y, Gao Y. ERK/PKM2 Is Mediated in the Warburg Effect and Cell Proliferation in Arsenic-Induced Human L-02 Hepatocytes. Biol Trace Elem Res 2024; 202:493-503. [PMID: 37237135 DOI: 10.1007/s12011-023-03706-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 05/13/2023] [Indexed: 05/28/2023]
Abstract
This study aimed to investigate the potential role of pyruvate kinase M2 (PKM2) and extracellular regulated protein kinase (ERK) in arsenic-induced cell proliferation. L-02 cells were treated with 0.2 and 0.4 μmol/L As3+, glycolysis inhibitor (2-deoxy-D-glucose,2-DG), ERK inhibitor [1,4-diamino-2,3-dicyano-1,4-bis(2-aminophenylthio)-butadiene, U0126] or transfected with PKM2 plasmid. Cell viability, proliferation, lactate acid production, and glucose intake capacity were determined by CCK-8 assay, EdU assay, lactic acid kit and 2-deoxy-2-[(7-nitro-2,1,3-benzoxadiazol-4-yl) amino]-D-glucose (2-NBDG) uptake kit, respectively. Also, levels of PKM2, phospho-PKM2S37, glucose transporter protein 1 (GLUT1), lactate dehydrogenase A (LDHA), ERK, and phospho-ERK were detected using Western blot and the subcellular localization of PKM2 in L-02 cells was detected by immunocytochemistry (ICC). Treatment with 0.2 and 0.4 μmol/L As3+ for 48 h increased the viability and proliferation of L-02 cells, the proportion of 2-NBDG+ cell and lactic acid in the culture medium, and GLUT1, LDHA, PKM2, phospho-PKM2S37, and phospho-ERK levels and PKM2 in nucleus. Compared with the 0.2 μmol/L As3+ treatment group, the lactic acid in the culture medium, cell proliferation and cell viability, and the expression of GLUT1 and LDHA were reduced in the group co-treated with siRNA-PKM2 and arsenic or in the group co-treated with U0126. Moreover, the arsenic-increased phospho-PKM2S37/PKM2 was decreased by U0126. Therefore, ERK/PKM2 plays a key role in the Warburg effect and proliferation of L-02 cells induced by arsenic, and also might be involved in arsenic-induced upregulation of GLUT1 and LDHA. This study provides a theoretical basis for further elucidating the carcinogenic mechanism of arsenic.
Collapse
Affiliation(s)
- Fanshuo Yin
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, No.157 Baojian Road, Nangang District, Harbin, 150081, Heilongjiang Province, China
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health of P. R. China, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
| | - Xin Zhang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, No.157 Baojian Road, Nangang District, Harbin, 150081, Heilongjiang Province, China
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health of P. R. China, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
| | - Zaihong Zhang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, No.157 Baojian Road, Nangang District, Harbin, 150081, Heilongjiang Province, China
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health of P. R. China, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
| | - Meichen Zhang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, No.157 Baojian Road, Nangang District, Harbin, 150081, Heilongjiang Province, China
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health of P. R. China, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
| | - Yunyi Yin
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, No.157 Baojian Road, Nangang District, Harbin, 150081, Heilongjiang Province, China
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health of P. R. China, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
| | - Yanmei Yang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, No.157 Baojian Road, Nangang District, Harbin, 150081, Heilongjiang Province, China.
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health of P. R. China, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China.
| | - Yanhui Gao
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, No.157 Baojian Road, Nangang District, Harbin, 150081, Heilongjiang Province, China.
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health of P. R. China, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China.
| |
Collapse
|
12
|
Luo W, Lu J, Zheng X, Wang J, Qian S, Bai Z, Wu M. A novel prognostic N 7-methylguanosine-related long non-coding RNA signature in clear cell renal cell carcinoma. Sci Rep 2023; 13:18454. [PMID: 37891201 PMCID: PMC10611723 DOI: 10.1038/s41598-023-45287-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: 02/03/2023] [Accepted: 10/18/2023] [Indexed: 10/29/2023] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is regulated by methylation modifications and long noncoding RNAs (lncRNAs). However, knowledge of N7-methylguanosine (m7G)-related lncRNAs that predict ccRCC prognosis remains insufficient. A prognostic multi-lncRNA signature was created using LASSO regression to examine the differential expression of m7G-related lncRNAs in ccRCC. Furthermore, we performed Kaplan-Meier analysis and area under the curve (AUC) analysis for diagnosis. In all, a model based on five lncRNAs was developed. Principal component analysis (PCA) indicated that the risk model precisely separated the patients into different groups. The IC50 value for drug sensitivity divided patients into two risk groups. High-risk group of patients was more susceptible to A.443654, A.770041, ABT.888, AMG.706, and AZ628. Moreover, a lower tumor mutation burden combined with low-risk scores was associated with a better prognosis of ccRCC. Quantitative real-time polymerase chain reaction (qRT-PCR) exhibited that the expression levels of LINC01507, AC093278.2 were very high in all five ccRCC cell lines, AC084876.1 was upregulated in all ccRCC cell lines except 786-O, and the levels of AL118508.1 and DUXAP8 were upregulated in the Caki-1 cell line. This risk model may be promising for the clinical prediction of prognosis and immunotherapeutic responses in patients with ccRCC.
Collapse
Affiliation(s)
- Wang Luo
- School of Stomatology, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Jing Lu
- Department of Clinical, Zunyi Medical and Pharmaceutical College, Zunyi, 563000, Guizhou, China
| | - Xiang Zheng
- Department of Medical Genetics, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - JinJing Wang
- Department of Pathology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - ShengYan Qian
- School of Stomatology, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - ZhiXun Bai
- Department of Nephrology, the Second Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China.
| | - MingSong Wu
- School of Stomatology, Zunyi Medical University, Zunyi, 563000, Guizhou, China.
| |
Collapse
|
13
|
Li X, Duan Z, Chen X, Pan D, Luo Q, Gu L, Xu G, Li Y, Zhang H, Gong Q, Chen R, Gu Z, Luo K. Impairing Tumor Metabolic Plasticity via a Stable Metal-Phenolic-Based Polymeric Nanomedicine to Suppress Colorectal Cancer. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2300548. [PMID: 36917817 DOI: 10.1002/adma.202300548] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/08/2023] [Indexed: 06/09/2023]
Abstract
Targeting metabolic vulnerability of tumor cells is a promising anticancer strategy. However, the therapeutic efficacy of existing metabolism-regulating agents is often compromised due to tolerance resulting from tumor metabolic plasticity, as well as their poor bioavailability and tumor-targetability. Inspired by the inhibitive effect of N-ethylmaleimide on the mitochondrial function, a dendronized-polymer-functionalized metal-phenolic nanomedicine (pOEG-b-D-SH@NP) encapsulating maleimide-modified doxorubicin (Mal-DOX) is developed to enable improvement in the overall delivery efficiency and inhibition of the tumor metabolism via multiple pathways. It is observed that Mal-DOX and its derived nanomedicine induces energy depletion of CT26 colorectal cancer cells more efficiently than doxorubicin, and shifts the balance of programmed cell death from apoptosis toward necroptosis. Notably, pOEG-b-D-SH@NP simultaneously inhibits cellular oxidative phosphorylation and glycolysis, thus potently suppressing cancer growth and peritoneal intestinal metastasis in mouse models. Overall, the study provides a promising dendronized-polymer-derived nanoplatform for the treatment of cancers through impairing metabolic plasticity.
Collapse
Affiliation(s)
- Xiaoling Li
- Huaxi MR Research Center (HMRRC), Animal Experimental Center, Department of Radiology, National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhenyu Duan
- Huaxi MR Research Center (HMRRC), Animal Experimental Center, Department of Radiology, National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaoting Chen
- Huaxi MR Research Center (HMRRC), Animal Experimental Center, Department of Radiology, National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Dayi Pan
- Huaxi MR Research Center (HMRRC), Animal Experimental Center, Department of Radiology, National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qiang Luo
- Huaxi MR Research Center (HMRRC), Animal Experimental Center, Department of Radiology, National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lei Gu
- Huaxi MR Research Center (HMRRC), Animal Experimental Center, Department of Radiology, National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Gang Xu
- Huaxi MR Research Center (HMRRC), Animal Experimental Center, Department of Radiology, National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Functional and molecular imaging Key Laboratory of Sichuan Province, Key Laboratory of Transplant Engineering and Immunology, NHC, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| | - Yinggang Li
- Huaxi MR Research Center (HMRRC), Animal Experimental Center, Department of Radiology, National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hu Zhang
- Amgen Bioprocessing Centre, Keck Graduate Institute, Claremont, CA, 91711, USA
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC), Animal Experimental Center, Department of Radiology, National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Functional and molecular imaging Key Laboratory of Sichuan Province, Key Laboratory of Transplant Engineering and Immunology, NHC, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
- Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen, Fujian, 361000, China
| | - Rongjun Chen
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
| | - Zhongwei Gu
- Huaxi MR Research Center (HMRRC), Animal Experimental Center, Department of Radiology, National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Animal Experimental Center, Department of Radiology, National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Functional and molecular imaging Key Laboratory of Sichuan Province, Key Laboratory of Transplant Engineering and Immunology, NHC, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| |
Collapse
|
14
|
Fumonisin B 1 disrupts mitochondrial function in oxidatively poised HepG2 liver cells by disrupting oxidative phosphorylation complexes and potential participation of lincRNA-p21. Toxicon 2023; 225:107057. [PMID: 36796496 DOI: 10.1016/j.toxicon.2023.107057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 02/06/2023] [Accepted: 02/13/2023] [Indexed: 02/16/2023]
Abstract
Fumonisin B1 (FB1) is etiologically linked to cancer, yet the underlying mechanisms remain largely unclear. It is also not known if mitochondrial dysfunction is involved as a contributor to FB1-induced metabolic toxicity. This study investigated the effects of FB1 on mitochondrial toxicity and its implications in cultured human liver (HepG2) cells. HepG2 cells poised to undergo oxidative and glycolytic metabolism were exposed to FB1 for 6 h. We determined mitochondrial toxicity, reducing equivalent levels and mitochondrial sirtuin activity using luminometric, fluorometric and spectrophotometric methods. Molecular pathways involved were determined using western blots and PCR. Our data confirm that FB1 is a mitochondrial toxin capable of disrupting the stability of complexes I and V of the mitochondrial electron transport and decreasing the NAD:NADH ratio in galactose supplemented HepG2 cells. We further showed that in cells treated with FB1, p53 acts as a metabolic stress-responsive transcription factor that induces the expression of lincRNA-p21, which plays a crucial role in stabilising HIF-1α. The findings provide novel insights into the impact of this mycotoxin in the dysregulation of energy metabolism and may contribute to the growing body of evidence of its tumor promoting effects.
Collapse
|
15
|
Liu C, Wu K, Li J, Mu X, Gao H, Xu X. Nanoparticle-mediated therapeutic management in cholangiocarcinoma drug targeting: Current progress and future prospects. Biomed Pharmacother 2023; 158:114135. [PMID: 36535198 DOI: 10.1016/j.biopha.2022.114135] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Patients with cholangiocarcinoma (CCA) often have an unfavorable prognosis because of its insidious nature, low resectability rate, and poor response to anticancer drugs and radiotherapy, which makes early detection and treatment difficult. At present, CCA has a five-year overall survival rate (OS) of only 5%, despite advances in therapies. New an increasing number of evidence suggests that nanoplatforms may play a crucial role in enhancing the pharmacological effects and in reducing both short- and long-term side effects of cancer treatment. This document reviews the advantages and shortcomings of nanoparticles such as liposomes, polymeric nanoparticle,inorganic nanoparticle, nano-metals and nano-alloys, carbon dots, nano-micelles, dendrimer, nano-capsule, bio-Nanomaterials in the diagnosis and treatment of CCA and discuss the current challenges in of nanoplatforms for CCA.
Collapse
Affiliation(s)
- Chunkang Liu
- Department of Gastrointestinal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Kunzhe Wu
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jianyang Li
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xupeng Mu
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Huan Gao
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xiaohua Xu
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, China.
| |
Collapse
|
16
|
Understanding the Contribution of Lactate Metabolism in Cancer Progress: A Perspective from Isomers. Cancers (Basel) 2022; 15:cancers15010087. [PMID: 36612084 PMCID: PMC9817756 DOI: 10.3390/cancers15010087] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/13/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Lactate mediates multiple cell-intrinsic effects in cancer metabolism in terms of development, maintenance, and metastasis and is often correlated with poor prognosis. Its functions are undertaken as an energy source for neighboring carcinoma cells and serve as a lactormone for oncogenic signaling pathways. Indeed, two isomers of lactate are produced in the Warburg effect: L-lactate and D-lactate. L-lactate is the main end-production of glycolytic fermentation which catalyzes glucose, and tiny D-lactate is fabricated through the glyoxalase system. Their production inevitably affects cancer development and therapy. Here, we systematically review the mechanisms of lactate isomers production, and highlight emerging evidence of the carcinogenic biological effects of lactate and its isomers in cancer. Accordingly, therapy that targets lactate and its metabolism is a promising approach for anticancer treatment.
Collapse
|
17
|
Song J, Zheng J, Liu X, Dong W, Yang C, Wang D, Ruan X, Zhao Y, Liu L, Wang P, Zhang M, Liu Y. A novel protein encoded by ZCRB1-induced circHEATR5B suppresses aerobic glycolysis of GBM through phosphorylation of JMJD5. J Exp Clin Cancer Res 2022; 41:171. [PMID: 35538499 PMCID: PMC9086421 DOI: 10.1186/s13046-022-02374-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 04/26/2022] [Indexed: 12/28/2022] Open
Abstract
Abstract
Background
RNA-binding proteins (RBPs) and circular RNAs (circRNAs) play important roles in glioblastoma multiforme (GBM). Aerobic glycolysis is a metabolic characteristic of GBM. However, the roles of RBPs and circRNAs in aerobic glycolysis in GBM remain unclear. The aim of this study is to explore the mechanisms by which RBPs and circRNAs regulate aerobic glycolysis in GBM cells.
Methods
RNA sequencing and circRNA microarray analysis were performed to identify RBPs and circRNAs for further study. Mass spectrometry validated the encoded protein and its interacting proteins. Quantitative reverse transcription PCR and western blot assays were used to determine the mRNA and protein expression, respectively. Furthermore, immunofluorescence and fluorescence in situ hybridization assays were used to determine the protein and RNA localization, respectively. Glucose and lactate measurement assays, Seahorse XF glycolysis stress assays and cell viability assays were conducted to investigate the effects on glycolysis and proliferation in GBM cells.
Results
We selected zinc finger CCHC-type and RNA-binding motif 1 (ZCRB1) and circRNA HEAT repeat containing 5B (circHEATR5B) as candidates for this study. These genes were expressed at low levels in GBM tissues and cells. Both ZCRB1 and circHEATR5B overexpression suppressed aerobic glycolysis and proliferation in GBM cells. ZCRB1 overexpression promoted the Alu element-mediated formation of circHEATR5B. In addition, circHEATR5B encoded a novel protein HEATR5B-881aa which interacted directly with Jumonji C-domain-containing 5 (JMJD5) and reduced its stability by phosphorylating S361. JMJD5 knockdown increased pyruvate kinase M2 (PKM2) enzymatic activity and suppressed glycolysis and proliferation in GBM cells. Finally, ZCRB1, circHEATR5B and HEATR5B-881aa overexpression inhibited GBM xenograft growth and prolonged the survival time of nude mice.
Conclusions
This study reveals a novel mechanism of regulating aerobic glycolysis and proliferation in GBM cells through the ZCRB1/circHEATR5B/HEATR5B-881aa/JMJD5/PKM2 pathway, which can provide novel strategies and potential targets for GBM therapy.
Collapse
|
18
|
Warburg effect in colorectal cancer: the emerging roles in tumor microenvironment and therapeutic implications. J Hematol Oncol 2022; 15:160. [PMID: 36319992 PMCID: PMC9628128 DOI: 10.1186/s13045-022-01358-5] [Citation(s) in RCA: 135] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 09/26/2022] [Indexed: 11/07/2022] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer and the second leading cause of cancer-related death worldwide. Countless CRC patients undergo disease progression. As a hallmark of cancer, Warburg effect promotes cancer metastasis and remodels the tumor microenvironment, including promoting angiogenesis, immune suppression, cancer-associated fibroblasts formation and drug resistance. Targeting Warburg metabolism would be a promising method for the treatment of CRC. In this review, we summarize information about the roles of Warburg effect in tumor microenvironment to elucidate the mechanisms governing Warburg effect in CRC and to identify novel targets for therapy.
Collapse
|
19
|
Xu Y, Tran L, Tang J, Nguyen V, Sewell E, Xiao J, Hino C, Wasnik S, Francis-Boyle OL, Zhang KK, Xie L, Zhong JF, Baylink DJ, Chen CS, Reeves ME, Cao H. FBP1-Altered Carbohydrate Metabolism Reduces Leukemic Viability through Activating P53 and Modulating the Mitochondrial Quality Control System In Vitro. Int J Mol Sci 2022; 23:ijms231911387. [PMID: 36232688 PMCID: PMC9570078 DOI: 10.3390/ijms231911387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/17/2022] [Accepted: 09/23/2022] [Indexed: 11/16/2022] Open
Abstract
Acute myeloid leukemia (AML)—the most frequent form of adult blood cancer—is characterized by heterogeneous mechanisms and disease progression. Developing an effective therapeutic strategy that targets metabolic homeostasis and energy production in immature leukemic cells (blasts) is essential for overcoming relapse and improving the prognosis of AML patients with different subtypes. With respect to metabolic regulation, fructose-1,6-bisphosphatase 1 (FBP1) is a gluconeogenic enzyme that is vital to carbohydrate metabolism, since gluconeogenesis is the central pathway for the production of important metabolites and energy necessary to maintain normal cellular activities. Beyond its catalytic activity, FBP1 inhibits aerobic glycolysis—known as the “Warburg effect”—in cancer cells. Importantly, while downregulation of FBP1 is associated with carcinogenesis in major human organs, restoration of FBP1 in cancer cells promotes apoptosis and prevents disease progression in solid tumors. Recently, our large-scale sequencing analyses revealed FBP1 as a novel inducible therapeutic target among 17,757 vitamin-D-responsive genes in MV4-11 or MOLM-14 blasts in vitro, both of which were derived from AML patients with FLT3 mutations. To investigate FBP1′s anti-leukemic function in this study, we generated a new AML cell line through lentiviral overexpression of an FBP1 transgene in vitro (named FBP1-MV4-11). Results showed that FBP1-MV4-11 blasts are more prone to apoptosis than MV4-11 blasts. Mechanistically, FBP1-MV4-11 blasts have significantly increased gene and protein expression of P53, as confirmed by the P53 promoter assay in vitro. However, enhanced cell death and reduced proliferation of FBP1-MV4-11 blasts could be reversed by supplementation with post-glycolytic metabolites in vitro. Additionally, FBP1-MV4-11 blasts were found to have impaired mitochondrial homeostasis through reduced cytochrome c oxidase subunit 2 (COX2 or MT-CO2) and upregulated PTEN-induced kinase (PINK1) expressions. In summary, this is the first in vitro evidence that FBP1-altered carbohydrate metabolism and FBP1-activated P53 can initiate leukemic death by activating mitochondrial reprogramming in AML blasts, supporting the clinical potential of FBP1-based therapies for AML-like cancers.
Collapse
Affiliation(s)
- Yi Xu
- Division of Hematology and Oncology, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Cancer Center, Loma Linda University, Loma Linda, CA 92354, USA
- Correspondence: ; Tel.: +1-909-651-5887
| | - Lily Tran
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Janet Tang
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Vinh Nguyen
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Elisabeth Sewell
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Jeffrey Xiao
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Christopher Hino
- Division of Hematology and Oncology, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Samiksha Wasnik
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Olivia L. Francis-Boyle
- Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Loma Linda University, Loma Linda, CA 92354, USA
- Department of Pathology & Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Ke K. Zhang
- Department of Nutrition, Texas A&M University, College Station, TX 77030, USA
- Center for Epigenetics & Disease Prevention, Institute of Biosciences & Technology, College of Medicine, Texas A&M University, Houston, TX 77030, USA
| | - Linglin Xie
- Department of Nutrition, Texas A&M University, College Station, TX 77030, USA
| | - Jiang F. Zhong
- Cancer Center, Loma Linda University, Loma Linda, CA 92354, USA
- Department of Basic Science, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - David J. Baylink
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Chien-Shing Chen
- Division of Hematology and Oncology, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Cancer Center, Loma Linda University, Loma Linda, CA 92354, USA
| | - Mark E. Reeves
- Division of Hematology and Oncology, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Cancer Center, Loma Linda University, Loma Linda, CA 92354, USA
| | - Huynh Cao
- Division of Hematology and Oncology, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Cancer Center, Loma Linda University, Loma Linda, CA 92354, USA
| |
Collapse
|
20
|
Gu L, Ma G, Li C, Lin J, Zhao G. New insights into the prognosis of intraocular malignancy: Interventions for association mechanisms between cancer and diabetes. Front Oncol 2022; 12:958170. [PMID: 36003786 PMCID: PMC9393514 DOI: 10.3389/fonc.2022.958170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/12/2022] [Indexed: 11/19/2022] Open
Abstract
The intraocular malignancies, which mostly originate from the retina and uvea, exhibit a high incidence of blindness and even death. Uveal melanoma (UM) and retinoblastoma (RB) are the most common intraocular malignancies in adults and children, respectively. The high risks of distant metastases lead to an extremely poor prognosis. Nowadays, various epidemiological studies have demonstrated that diabetes is associated with the high incidence and mortality of cancers, such as liver cancer, pancreatic cancer, and bladder cancer. However, the mechanisms and interventions associated with diabetes and intraocular malignancies have not been reviewed. In this review, we have summarized the associated mechanisms between diabetes and intraocular malignancy. Diabetes mellitus is a chronic metabolic disease characterized by prolonged periods of hyperglycemia. Recent studies have reported that the abnormal glucose metabolism, insulin resistance, and the activation of the IGF/insulin-like growth factor-1 receptor (IGF-1R) signaling axis in diabetes contribute to the genesis, growth, proliferation, and metastases of intraocular malignancy. In addition, diabetic patients are more prone to suffer severe complications and poor prognosis after radiotherapy for intraocular malignancy. Based on the common pathogenesis shared by diabetes and intraocular malignancy, they may be related to interventions and treatments. Therefore, interventions targeting the abnormal glucose metabolism, insulin resistance, and IGF-1/IGF-1R signaling axis show therapeutic potentials to treat intraocular malignancy.
Collapse
Affiliation(s)
- Lingwen Gu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Guofeng Ma
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Cui Li
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jing Lin
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Guiqiu Zhao
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
- *Correspondence: Guiqiu Zhao,
| |
Collapse
|
21
|
Li S, Qian J, Xu M, Yang J, He Z, Zhao T, Zhao J, Fang R. A new adenine nucleotide transporter located in the ER is essential for maintaining the growth of Toxoplasma gondii. PLoS Pathog 2022; 18:e1010665. [PMID: 35788770 PMCID: PMC9286291 DOI: 10.1371/journal.ppat.1010665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 07/15/2022] [Accepted: 06/12/2022] [Indexed: 11/30/2022] Open
Abstract
The lumen of the endoplasmic reticulum (ER) is the subcellular site where secretory protein folding, glycosylation and sulfation of membrane-bound proteins, proteoglycans, and lipids occur. The protein folding and degradation in the lumen of the ER require high levels of energy in the form of ATP. Biochemical and genetic approaches show that ATP must first be translocated across ER membrane by particular transporters before serving as substrates and energy sources in the lumenal reactions. Here we describe an ATP/ADP transporter residing in the ER membranes of T.gondii. Immunofluorescence (IFA) assay in transgenic TgANT1-HA tag revealed that TgANT1 is a protein specifically expressed in the ER. In vitro assays, functional integration of TgANT in the cytoplasmic membrane of intact E. coli cells reveals high specificity for an ATP/ADP antiport. The depletion of TgANT leads to fatal growth defects in T.gondii, including a significant slowdown in replication, no visible plaque formation, and reduced ability to invade. We also found that the amino acid mutations in two domains of TgANT lead to the complete loss of its function. Since these two domains are conserved in multiple species, they may share the same transport mechanism. Our results indicate that TgANT is the only ATP/ADP transporter in the ER of T. gondii, and the lack of ATP in the ER is the cause of the death of T. gondii. The secretory protein of T. gondii is essential for its invasion and normal growth in host cells, all the secretory proteins are synthesized in the ER before being destined for these distinct organelles, such as apicoplast, microneme, dense granule and rhoptry. ER ATP is demanded to support secretory protein folding and trafficking, and the level of ER ATP determines which proteins are able to be directed to the distinct organelles. In theory, the supply of ATP in the ER is necessary for T. gondii. However, the transport mechanism and importance of the ER ATP in T. gondii are still unclear. In our study, we identified an ATP/ADP transporter (TgANT) located in the ER and verified its function through various methods. Unlike the ER ATP/ADP transporter in mammals, we proved that TgANT is functionally specific; the deletion of TgANT caused the interruption of the supply of ATP in the ER, which leads to fatal phenotypic defects of T. gondii. Our research further expands the understanding of the growth regulation in T. gondii.
Collapse
Affiliation(s)
- Senyang Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei Province, China
| | - Jiahui Qian
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei Province, China
| | - Ming Xu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei Province, China
| | - Jing Yang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei Province, China
| | - Zhengming He
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei Province, China
| | - Tongjie Zhao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei Province, China
| | - Junlong Zhao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei Province, China
| | - Rui Fang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei Province, China
- * E-mail:
| |
Collapse
|
22
|
Jerves T, Blau N, Ferreira CR. Clinical and biochemical footprints of inherited metabolic diseases. VIII. Neoplasias. Mol Genet Metab 2022; 136:118-124. [PMID: 35422340 PMCID: PMC9189061 DOI: 10.1016/j.ymgme.2022.03.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 12/21/2022]
Abstract
Cancer, caused by multiple cumulative pathogenic variants in tumor suppressor genes and proto-oncogenes, is a leading cause of mortality worldwide. The uncontrolled and rapid cell growth of the tumors requires a reprogramming of the complex cellular metabolic network to favor anabolism. Adequate management and treatment of certain inherited metabolic diseases might prevent the development of certain neoplasias, such as hepatocellular carcinoma in tyrosinemia type 1 or hepatocellular adenomas in glycogen storage disorder type 1a. We reviewed and updated the list of known metabolic etiologies associated with various types of benign and malignant neoplasias, finding 64 relevant inborn errors of metabolism. This is the eighth article of the series attempting to create a comprehensive list of clinical and metabolic differential diagnosis by system involvement.
Collapse
Affiliation(s)
- Teodoro Jerves
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nenad Blau
- Division of Metabolism, University Children's Hospital, Zürich, Switzerland.
| | - Carlos R Ferreira
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
23
|
Hyperglycemia induces gastric carcinoma proliferation and migration via the Pin1/BRD4 pathway. Cell Death Dis 2022; 8:224. [PMID: 35461311 PMCID: PMC9035156 DOI: 10.1038/s41420-022-01030-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 11/18/2022]
Abstract
Diabetes is a potential risk factor for gastric cancer (GC). Pin1, a peptidyl–prolyl cis/trans isomerase, promotes GC cell proliferation and migration. The role and underlying mechanism of the Pin1/BRD4 axis in hyperglycemia-induced proliferation and migration of GC cells were analyzed in vivo and in vitro. Proliferation and migration of GC cells were measured; Pin1 and BRD4 expression of the cell cycle were determined. Pin1 and BRD4 were downregulated by transfecting Pin1 shRNA lentivirus into GC cells and JQ1-intervention GC cells. Tumor formation and lung metastasis were assessed in vivo. Inhibition of Pin1 and BRD4 significantly suppressed high-glucose (HG)-induced GC cell proliferation and migration. HG enhanced G1/S cell-cycle transition, associated with increased Pin1 and BRD4 expression. Silencing Pin1 significantly downregulated the expression of BRD4 and NAP1L1 and upregulated that of P21 in GC cells. In vivo studies indicated that hyperglycemia promotes tumor growth and lung metastasis by inducing Pin1 and BRD4 expression. Thus, Pin1/BRD4 plays an important role in hyperglycemia-promoted tumor growth. The significance of these findings toward improved prognosis of diabetic patients with GC cannot be underestimated.
Collapse
|
24
|
San Martín A, Arce-Molina R, Aburto C, Baeza-Lehnert F, Barros LF, Contreras-Baeza Y, Pinilla A, Ruminot I, Rauseo D, Sandoval PY. Visualizing physiological parameters in cells and tissues using genetically encoded indicators for metabolites. Free Radic Biol Med 2022; 182:34-58. [PMID: 35183660 DOI: 10.1016/j.freeradbiomed.2022.02.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 02/07/2023]
Abstract
The study of metabolism is undergoing a renaissance. Since the year 2002, over 50 genetically-encoded fluorescent indicators (GEFIs) have been introduced, capable of monitoring metabolites with high spatial/temporal resolution using fluorescence microscopy. Indicators are fusion proteins that change their fluorescence upon binding a specific metabolite. There are indicators for sugars, monocarboxylates, Krebs cycle intermediates, amino acids, cofactors, and energy nucleotides. They permit monitoring relative levels, concentrations, and fluxes in living systems. At a minimum they report relative levels and, in some cases, absolute concentrations may be obtained by performing ad hoc calibration protocols. Proper data collection, processing, and interpretation are critical to take full advantage of these new tools. This review offers a survey of the metabolic indicators that have been validated in mammalian systems. Minimally invasive, these indicators have been instrumental for the purposes of confirmation, rebuttal and discovery. We envision that this powerful technology will foster metabolic physiology.
Collapse
Affiliation(s)
- A San Martín
- Centro de Estudios Científicos (CECs), Valdivia, Chile.
| | - R Arce-Molina
- Centro de Estudios Científicos (CECs), Valdivia, Chile
| | - C Aburto
- Centro de Estudios Científicos (CECs), Valdivia, Chile; Universidad Austral de Chile, Valdivia, Chile
| | | | - L F Barros
- Centro de Estudios Científicos (CECs), Valdivia, Chile
| | - Y Contreras-Baeza
- Centro de Estudios Científicos (CECs), Valdivia, Chile; Universidad Austral de Chile, Valdivia, Chile
| | - A Pinilla
- Centro de Estudios Científicos (CECs), Valdivia, Chile; Universidad Austral de Chile, Valdivia, Chile
| | - I Ruminot
- Centro de Estudios Científicos (CECs), Valdivia, Chile
| | - D Rauseo
- Centro de Estudios Científicos (CECs), Valdivia, Chile; Universidad Austral de Chile, Valdivia, Chile
| | - P Y Sandoval
- Centro de Estudios Científicos (CECs), Valdivia, Chile
| |
Collapse
|
25
|
Ruan Y, Fang X, Guo T, Liu Y, Hu Y, Wang X, Hu Y, Gao L, Li Y, Pi J, Xu Y. Metabolic reprogramming in the arsenic carcinogenesis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 229:113098. [PMID: 34952379 DOI: 10.1016/j.ecoenv.2021.113098] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 12/06/2021] [Accepted: 12/14/2021] [Indexed: 06/14/2023]
Abstract
Chronic exposure to arsenic has been associated with a variety of cancers with the mechanisms undefined. Arsenic exposure causes alterations in metabolites in bio-samples. Recent research progress on cancer biology suggests that metabolic reprogramming contributes to tumorigenesis. Therefore, metabolic reprogramming provides a new clue for the mechanisms of arsenic carcinogenesis. In the present manuscript, we review the latest findings in reprogramming of glucose, lipids, and amino acids in response to arsenic exposure. Most studies focused on glucose reprogramming and found that arsenic exposure enhanced glycolysis. However, in vivo studies observed "reverse Warburg effect" in some cases due to the complexity of the disease evolution and microenvironment. Arsenic exposure has been reported to disturb lipid deposition by inhibiting lipolysis, and induce serine-glycine one-carbon pathway. As a dominant mechanism for arsenic toxicity, oxidative stress is considered to link with metabolism reprogramming. Few studies analyzed the causal relationship between metabolic reprogramming and arsenic-induced cancers. Metabolic alterations may vary with exposure doses and periods. Identifying metabolic alterations common among humans and experiment models with human-relevant exposure characteristics may guide future investigations.
Collapse
Affiliation(s)
- Yihui Ruan
- Group of Chronic Disease and Environmental Genomics, School of Public Health, China Medical University, P.R. China
| | - Xin Fang
- Group of Chronic Disease and Environmental Genomics, School of Public Health, China Medical University, P.R. China
| | - Tingyue Guo
- Group of Chronic Disease and Environmental Genomics, School of Public Health, China Medical University, P.R. China
| | - Yiting Liu
- Group of Chronic Disease and Environmental Genomics, School of Public Health, China Medical University, P.R. China
| | - Yu Hu
- Group of Chronic Disease and Environmental Genomics, School of Public Health, China Medical University, P.R. China
| | - Xuening Wang
- Group of Chronic Disease and Environmental Genomics, School of Public Health, China Medical University, P.R. China
| | - Yuxin Hu
- Experimental Teaching Center, School of Public Health, China Medical University, P.R. China
| | - Lanyue Gao
- Experimental Teaching Center, School of Public Health, China Medical University, P.R. China
| | - Yongfang Li
- The Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, China Medical University, P.R. China
| | - Jingbo Pi
- The Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, China Medical University, P.R. China; Program of Environmental Toxicology, School of Public Health, China Medical University, P.R. China
| | - Yuanyuan Xu
- Group of Chronic Disease and Environmental Genomics, School of Public Health, China Medical University, P.R. China; The Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, China Medical University, P.R. China.
| |
Collapse
|
26
|
Feng S, Liu H, Dong X, Du P, Guo H, Pang Q. Identification and validation of an autophagy-related signature for predicting survival in lower-grade glioma. Bioengineered 2021; 12:9692-9708. [PMID: 34696669 PMCID: PMC8810042 DOI: 10.1080/21655979.2021.1985818] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Abnormal levels of autophagy have been implicated in the pathogenesis of multiple diseases, including cancer. However, little is known about the role of autophagy-related genes (ARGs) in low-grade gliomas (LGG). Accordingly, the aims of this study were to assess the prognostic values of ARGs and to establish a genetic signature for LGG prognosis. Expression profile data from patients with and without primary LGG were obtained from The Cancer Genome Atlas (TCGA) and Genome Tissue Expression databases, respectively, and consensus clustering was used to identify clusters of patients with distinct prognoses. Nineteen differentially expressed ARGs were selected with threshold values of FDR < 0.05 and |log2 fold change (FC)| ≥ 2, and functional analysis revealed that these genes were associated with autophagy processes as expected. An autophagy-related signature was established using a Cox regression model of six ARGs that separated patients from TCGA training cohort into high- and low-risk groups. Univariate and multivariate Cox regression analysis indicated that the signature-based risk score was an independent prognostic factor. The signature was successfully validated using the TCGA testing, TCGA entire, and Chinese Glioma Genome Atlas cohorts. Stratified analyses demonstrated that the signature was associated with clinical features and prognosis, and gene set enrichment analysis revealed that autophagy- and cancer-related pathways were more enriched in high-risk patients than in low-risk patients. The prognostic value and expression of the six signature-related genes were also investigated. Thus, the present study constructed and validated an autophagy-related prognostic signature that could optimize individualized survival prediction in LGG patients.
Collapse
Affiliation(s)
- Shaobin Feng
- Department of Neurosurgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Huiling Liu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Xushuai Dong
- Department of Neurosurgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Peng Du
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Hua Guo
- Department of Neurosurgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.,Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Qi Pang
- Department of Neurosurgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
27
|
Insulin Resistance and Cancer: In Search for a Causal Link. Int J Mol Sci 2021; 22:ijms222011137. [PMID: 34681797 PMCID: PMC8540232 DOI: 10.3390/ijms222011137] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 12/12/2022] Open
Abstract
Insulin resistance (IR) is a condition which refers to individuals whose cells and tissues become insensitive to the peptide hormone, insulin. Over the recent years, a wealth of data has made it clear that a synergistic relationship exists between IR, type 2 diabetes mellitus, and cancer. Although the underlying mechanism(s) for this association remain unclear, it is well established that hyperinsulinemia, a hallmark of IR, may play a role in tumorigenesis. On the other hand, IR is strongly associated with visceral adiposity dysfunction and systemic inflammation, two conditions which favor the establishment of a pro-tumorigenic environment. Similarly, epigenetic modifications, such as DNA methylation, histone modifications, and non-coding RNA, in IR states, have been often associated with tumorigenesis in numerous types of human cancer. In addition to these observations, it is also broadly accepted that gut microbiota may play an intriguing role in the development of IR-related diseases, including type 2 diabetes and cancer, whereas potential chemopreventive properties have been attributed to some of the most commonly used antidiabetic medications. Herein we provide a concise overview of the most recent literature in this field and discuss how different but interrelated molecular pathways may impact on tumor development.
Collapse
|
28
|
Igelmann S, Lessard F, Uchenunu O, Bouchard J, Fernandez-Ruiz A, Rowell MC, Lopes-Paciencia S, Papadopoli D, Fouillen A, Ponce KJ, Huot G, Mignacca L, Benfdil M, Kalegari P, Wahba HM, Pencik J, Vuong N, Quenneville J, Guillon J, Bourdeau V, Hulea L, Gagnon E, Kenner L, Moriggl R, Nanci A, Pollak MN, Omichinski JG, Topisirovic I, Ferbeyre G. A hydride transfer complex reprograms NAD metabolism and bypasses senescence. Mol Cell 2021; 81:3848-3865.e19. [PMID: 34547241 DOI: 10.1016/j.molcel.2021.08.028] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 06/25/2021] [Accepted: 08/20/2021] [Indexed: 01/23/2023]
Abstract
Metabolic rewiring and redox balance play pivotal roles in cancer. Cellular senescence is a barrier for tumorigenesis circumvented in cancer cells by poorly understood mechanisms. We report a multi-enzymatic complex that reprograms NAD metabolism by transferring reducing equivalents from NADH to NADP+. This hydride transfer complex (HTC) is assembled by malate dehydrogenase 1, malic enzyme 1, and cytosolic pyruvate carboxylase. HTC is found in phase-separated bodies in the cytosol of cancer or hypoxic cells and can be assembled in vitro with recombinant proteins. HTC is repressed in senescent cells but induced by p53 inactivation. HTC enzymes are highly expressed in mouse and human prostate cancer models, and their inactivation triggers senescence. Exogenous expression of HTC is sufficient to bypass senescence, rescue cells from complex I inhibitors, and cooperate with oncogenic RAS to transform primary cells. Altogether, we provide evidence for a new multi-enzymatic complex that reprograms metabolism and overcomes cellular senescence.
Collapse
Affiliation(s)
- Sebastian Igelmann
- CRCHUM, 900 Saint-Denis St, Montréal, QC H2X 0A9, Canada; Département de Biochimie et Médecine Moléculaire, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Frédéric Lessard
- Département de Biochimie et Médecine Moléculaire, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Oro Uchenunu
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, QC H3T1E2, Canada; Department of Experimental Medicine, McGill University, Montreal, QC H4A3T2, Canada
| | - Jacob Bouchard
- Département de Biochimie et Médecine Moléculaire, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | | | | | | | - David Papadopoli
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, QC H3T1E2, Canada; Gerald Bronfman Department of Oncology, McGill University, Montreal, QC H4A3T2, Canada
| | - Aurélien Fouillen
- Faculté de médecine dentaire, Université de Montréal, Montréal, QC H3C 3J7, Canada; Département de Biochimie et Médecine Moléculaire, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Katia Julissa Ponce
- Faculté de médecine dentaire, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Geneviève Huot
- Département de Biochimie et Médecine Moléculaire, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Lian Mignacca
- Département de Biochimie et Médecine Moléculaire, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Mehdi Benfdil
- Département de Biochimie et Médecine Moléculaire, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Paloma Kalegari
- CRCHUM, 900 Saint-Denis St, Montréal, QC H2X 0A9, Canada; Département de Biochimie et Médecine Moléculaire, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Haytham M Wahba
- Department of Biochemistry, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62521, Egypt; Département de Biochimie et Médecine Moléculaire, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Jan Pencik
- Department of Pathology, Medical University of Vienna, Vienna, Austria; Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA; Center for Biomarker Research in Medicine, 8010 Graz, Austria
| | - Nhung Vuong
- CRCHUM, 900 Saint-Denis St, Montréal, QC H2X 0A9, Canada
| | - Jordan Quenneville
- Institut de recherche en immunologie et en cancérologie (IRIC), Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Jordan Guillon
- CRCHUM, 900 Saint-Denis St, Montréal, QC H2X 0A9, Canada
| | - Véronique Bourdeau
- Département de Biochimie et Médecine Moléculaire, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Laura Hulea
- Maisonneuve-Rosemont Hospital Research Centre, Montréal, QC H1T 2M4, Canada, Département de Médecine, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Etienne Gagnon
- Institut de recherche en immunologie et en cancérologie (IRIC), Université de Montréal, Montréal, QC H3C 3J7, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Lukas Kenner
- Department of Pathology, Medical University of Vienna, Vienna, Austria; Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, Vienna, Austria; Christian Doppler Laboratory for Applied Metabolomics, Vienna, Austria; CBmed GmbH - Center for Biomarker Research in Medicine, Graz, Styria, Austria
| | - Richard Moriggl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Antonio Nanci
- Faculté de médecine dentaire, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Michael N Pollak
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, QC H3T1E2, Canada
| | - James G Omichinski
- Département de Biochimie et Médecine Moléculaire, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Ivan Topisirovic
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, QC H3T1E2, Canada; Department of Experimental Medicine, McGill University, Montreal, QC H4A3T2, Canada; Department of Biochemistry, McGill University, Montreal, QC H4A 3T2, Canada; Gerald Bronfman Department of Oncology, McGill University, Montreal, QC H4A3T2, Canada.
| | - Gerardo Ferbeyre
- CRCHUM, 900 Saint-Denis St, Montréal, QC H2X 0A9, Canada; Département de Biochimie et Médecine Moléculaire, Université de Montréal, Montréal, QC H3C 3J7, Canada.
| |
Collapse
|
29
|
Huang Z, Wang S, Zhang HJ, Zhou YL, Tang X, Shi JH. Characteristics of hypoxic tumor microenvironment in non-small cell lung cancer, involving molecular patterns and prognostic signature. Transl Lung Cancer Res 2021; 10:2132-2147. [PMID: 34164265 PMCID: PMC8182713 DOI: 10.21037/tlcr-20-1314] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Background The mechanisms of hypoxia or immune microenvironment in cancer have been studied respectively, but the role of hypoxia immune microenvironment in non-small cell lung cancer (NSCLC) still needs further exploration. Methods By applying the K-means algorithm, 1,121 patients with NSCLC were divided into three categories. We evaluated the constructed signature in order to link it with the prognosis, which was constructed by univariate and least absolute shrinkage operator (LASSO) Cox regression analysis. Results A total of three clusters were obtained by clustering five Gene Expression Omnibus (GEO) data sets. Gene Set Variation Analysis (GSVA) and immune infiltration analysis were performed to explore the biological behavior. Cluster one presented an activated state of oncogenic pathways, and compared with the other two clusters, the median risk score was the highest, which was the reason for its poor survival. Cluster three showed that the immune pathway was active and the median risk score was the lowest, so the survival was the best. However, cluster two presented a state in which both immune and matrix pathways were activate. This was manifested as mutual antagonism, and its risk score was in the middle. Its survival was in the middle. Conclusions This work revealed the role of hypoxia related genes (HRGs) modification in tumor microenvironment, which was conducive to our comprehensive analysis of the prognosis of NSCLC, and provided direction and guidance for clinical immunotherapy.
Collapse
Affiliation(s)
- Zhanghao Huang
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, and Research Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, Nantong, China.,Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China.,Medical College of Nantong University, Nantong, China
| | - Shuo Wang
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, and Research Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, Nantong, China.,Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China.,Medical College of Nantong University, Nantong, China
| | - Hai-Jian Zhang
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - You Lang Zhou
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Xin Tang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Jia-Hai Shi
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, and Research Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, Nantong, China.,Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
30
|
Cao H, Tong H, Zhu J, Xie C, Qin Z, Li T, Liu X, He W. A Glycolysis-Based Long Non-coding RNA Signature Accurately Predicts Prognosis in Renal Carcinoma Patients. Front Genet 2021; 12:638980. [PMID: 33868376 PMCID: PMC8047215 DOI: 10.3389/fgene.2021.638980] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 03/16/2021] [Indexed: 12/18/2022] Open
Abstract
Background The prognosis of renal cell carcinoma (RCC) varies greatly among different risk groups, and the traditional indicators have limited effect in the identification of risk grade in patients with RCC. The purpose of our study is to explore a glycolysis-based long non-coding RNAs (lncRNAs) signature and verify its potential clinical significance in prognostic prediction of RCC patients. Methods In this study, RNA data and clinical information were downloaded from The Cancer Genome Atlas (TCGA) database. Univariate and multivariate cox regression displayed six significantly related lncRNAs (AC124854.1, AC078778.1, EMX2OS, DLGAP1-AS2, AC084876.1, and AC026401.3) which were utilized in construction of risk score by a formula. The accuracy of risk score was verified by a series of statistical methods such as receiver operating characteristic (ROC) curves, nomogram and Kaplan-Meier curves. Its potential clinical significance was excavated by gene enrichment analysis. Results Kaplan-Meier curves and ROC curves showed reliability of the risk score to predict the prognosis of RCC patients. Stratification analysis indicated that the risk score was independent predictor compare to other traditional clinical parameters. The clinical nomogram showed highly rigorous with index of 0.73 and precisely predicted 1-, 3-, and 5-year survival time of RCC patients. Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene set enrichment analysis (GSEA) depicted the top ten correlated pathways in both high-risk group and low-risk group. There are 6 lncRNAs and 25 related mRNAs including 36 lncRNA-mRNA links in lncRNA-mRNA co-expression network. Conclusion This research demonstrated that glycolysis-based lncRNAs possessed an important value in survival prediction of RCC patients, which would be a potential target for future treatment.
Collapse
Affiliation(s)
- Honghao Cao
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Department of Urology, Rongchang Traditional Chinese Medicine Hospital, Chongqing, China
| | - Hang Tong
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Central Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Junlong Zhu
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Central Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chenchen Xie
- Central Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zijia Qin
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Central Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tinghao Li
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Central Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xudong Liu
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Weiyang He
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
31
|
Chen Y, Cui J, Gong Y, Wei S, Wei Y, Yi L. MicroRNA: a novel implication for damage and protection against ionizing radiation. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:15584-15596. [PMID: 33533004 PMCID: PMC7854028 DOI: 10.1007/s11356-021-12509-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 01/12/2021] [Indexed: 04/16/2023]
Abstract
Ionizing radiation (IR) is a form of high energy. It poses a serious threat to organisms, but radiotherapy is a key therapeutic strategy for various cancers. It is significant to reduce radiation injury but maximize the effect of radiotherapy. MicroRNAs (miRNAs) are posttranscriptionally regulatory factors involved in cellular radioresponse. In this review, we show how miRNAs regulate important genes on cellular response to IR-induced damage and how miRNAs participate in IR-induced carcinogenesis. Additionally, we summarize the experimental and clinical evidence for miRNA involvement in radiotherapy and discuss their potential for improvement of radiotherapy. Finally, we highlight the role that miRNAs play in accident exposure to IR or radiotherapy as predictive biomarker. miRNA therapeutics have shown great perspective in radiobiology; miRNA may become a novel strategy for damage and protection against IR.
Collapse
Affiliation(s)
- Yonglin Chen
- Hengyang Medical College, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, University of South China, Hengyang, 421001, Hunan Province, People's Republic of China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, 421001, Hunan Province, People's Republic of China
| | - Jian Cui
- Hengyang Medical College, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, University of South China, Hengyang, 421001, Hunan Province, People's Republic of China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, 421001, Hunan Province, People's Republic of China
| | - Yaqi Gong
- Hengyang Medical College, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, University of South China, Hengyang, 421001, Hunan Province, People's Republic of China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, 421001, Hunan Province, People's Republic of China
| | - Shuang Wei
- Hengyang Medical College, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, University of South China, Hengyang, 421001, Hunan Province, People's Republic of China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, 421001, Hunan Province, People's Republic of China
| | - Yuanyun Wei
- Hengyang Medical College, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, University of South China, Hengyang, 421001, Hunan Province, People's Republic of China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, 421001, Hunan Province, People's Republic of China
| | - Lan Yi
- Hengyang Medical College, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, University of South China, Hengyang, 421001, Hunan Province, People's Republic of China.
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, 421001, Hunan Province, People's Republic of China.
| |
Collapse
|
32
|
Asadullah, Kumar S, Saxena N, Sarkar M, Barai A, Sen S. Combined heterogeneity in cell size and deformability promotes cancer invasiveness. J Cell Sci 2021; 134:jcs.250225. [PMID: 33602741 DOI: 10.1242/jcs.250225] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 01/29/2021] [Indexed: 01/27/2023] Open
Abstract
Phenotypic heterogeneity is increasingly acknowledged to confer several advantages to cancer progression and drug resistance. Here, we probe the collective importance of heterogeneity in cell size and deformability in breast cancer invasion. A computational model of invasion of a heterogeneous cell aggregate predicts that combined heterogeneity in cell size and deformability enhances invasiveness of the whole population, with maximum invasiveness at intermediate cell-cell adhesion. We then show that small cells of varying deformability, a subpopulation predicted to be enriched at the invasive front, exhibit considerable overlap with the biophysical properties of cancer stem cells (CSCs). In MDA-MB-231 cells, these include CD44 hi CD24- mesenchymal CSCs, which are small and soft, and CD44 hi CD24+ hybrid CSCs, which exhibit a wide range of size and deformability. We validate our predictions by tracking the pattern of cell invasion from spheroids implanted in three-dimensional collagen gels, wherein we show temporal enrichment of CD44 hi cells at the invasive front. Collectively, our results illustrate the advantages imparted by biophysical heterogeneity in enhancing cancer invasiveness.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Asadullah
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Sandeep Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Neha Saxena
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Madhurima Sarkar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Amlan Barai
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Shamik Sen
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| |
Collapse
|
33
|
Kreuzer M, Banerjee A, Birts CN, Darley M, Tavassoli A, Ivan M, Blaydes JP. Glycolysis, via NADH-dependent dimerisation of CtBPs, regulates hypoxia-induced expression of CAIX and stem-like breast cancer cell survival. FEBS Lett 2020; 594:2988-3001. [PMID: 32618367 DOI: 10.1002/1873-3468.13874] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 06/12/2020] [Accepted: 06/17/2020] [Indexed: 02/06/2023]
Abstract
Adaptive responses to hypoxia are mediated by the hypoxia-inducible factor (HIF) family of transcription factors. These responses include the upregulation of glycolysis to maintain ATP production. This also generates acidic metabolites, which require HIF-induced carbonic anhydrase IX (CAIX) for their neutralisation. C-terminal binding proteins (CtBPs) are coregulators of gene transcription and couple glycolysis with gene transcription due to their regulation by the glycolytic coenzyme NADH. Here, we find that experimental manipulation of glycolysis and CtBP function in breast cancer cells through multiple complementary approaches supports a hypothesis whereby the expression of known HIF-inducible genes, and CAIX in particular, adapts to available glucose in the microenvironment through a mechanism involving CtBPs. This novel pathway promotes the survival of stem cell-like cancer (SCLC) cells in hypoxia.
Collapse
Affiliation(s)
- Mira Kreuzer
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, Hants, UK.,Institute for Life Sciences, University of Southampton, Southampton, Hants, UK
| | - Arindam Banerjee
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, Hants, UK
| | - Charles N Birts
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, Hants, UK.,Institute for Life Sciences, University of Southampton, Southampton, Hants, UK
| | - Matthew Darley
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, Hants, UK
| | - Ali Tavassoli
- Institute for Life Sciences, University of Southampton, Southampton, Hants, UK.,School of Chemistry, University of Southampton, Southampton, Hants, UK
| | - Mircea Ivan
- Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Jeremy P Blaydes
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, Hants, UK.,Institute for Life Sciences, University of Southampton, Southampton, Hants, UK
| |
Collapse
|