1
|
Pan Z, Liu Y, Li H, Qiu H, Zhang P, Li Z, Wang X, Tian Y, Feng Z, Zhu S, Wang X. The role and mechanism of aerobic glycolysis in nasopharyngeal carcinoma. PeerJ 2025; 13:e19213. [PMID: 40191756 PMCID: PMC11971989 DOI: 10.7717/peerj.19213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 03/05/2025] [Indexed: 04/09/2025] Open
Abstract
This review delves into the pivotal role and intricate mechanisms of aerobic glycolysis in nasopharyngeal carcinoma (NPC). NPC, a malignancy originating from the nasopharyngeal epithelium, displays distinct geographical and clinical features. The article emphasizes the significance of aerobic glycolysis, a pivotal metabolic alteration in cancer cells, in NPC progression. Key enzymes such as hexokinase 2, lactate dehydrogenase A, phosphofructokinase 1, and pyruvate kinase M2 are discussed for their regulatory functions in NPC glycolysis through signaling pathways like PI3K/Akt and mTOR. Further, the article explores how oncogenic signaling pathways and transcription factors like c-Myc and HIF-1α modulate aerobic glycolysis, thereby affecting NPC's proliferation, invasion, metastasis, angiogenesis, and immune evasion. By elucidating these mechanisms, the review aims to advance research and clinical practice in NPC, informing the development of targeted therapeutic strategies that enhance treatment precision and reduce side effects. Overall, this review offers a broad understanding of the multifaceted role of aerobic glycolysis in NPC and its potential impact on therapeutic outcomes.
Collapse
Affiliation(s)
- Zhiyong Pan
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Yuyi Liu
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Hui Li
- Department of Ophthalmology, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Huisi Qiu
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Pingmei Zhang
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Zhiying Li
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Xinyu Wang
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Yuxiao Tian
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Zhengfu Feng
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Song Zhu
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Xin Wang
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| |
Collapse
|
2
|
Stepanov Y, Kolesnik D, Yakshibaeva Y, Solyanik G. EFFECT OF ADHESIVE LLC CELL PRETREATMENT BY OXAMATE ON THE SURVIVAL INDEXES AFTER TRANSITION TO DE-ADHESIVE GROWTH. Exp Oncol 2024; 46:237-243. [PMID: 39704457 DOI: 10.15407/exp-oncology.2024.03.237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND The ability to metabolic reprogramming is a distinctive feature of metastatically active tumor cells. A classic example of metabolic reprogramming, characteristic of almost all malignant cells, is aerobic glycolysis. Therefore, inhibition of glycolysis in tumor cells is considered a promising strategy for antitumor therapy. AIM To generate Lewis lung carcinoma (LLC) cell subpopulation after pretreatment by a lactate dehydrogenase (LDH) inhibitor - oxamate in adhesive growth conditions, and then to study the metabolism of this subpopulation in the anchorage-independent growth conditions. MATERIALS AND METHODS LLC cells were cultured without oxamate or with 17 mM oxamate in the adhesive growth conditions with the following transition to the anchorage-independent growth conditions without oxamate. A distribution of LLC cells by cell cycle phases, apoptosis rate, levels of reactive oxygen species (ROS), E-cadherin, and vimentin were determined by flow cytometry. Glucose consumption and lactate production were determined by spectrophotometry. RESULTS 48-h oxamate treatment in adhesive growth conditions resulted in a 30% decrease of the total number of LLC cells compared to the control. In 72 h after the transfer of both oxamate-treated and control cells into the anchorage-independent growth condition without oxamate, the number of viable cells pretreated with oxamate was reduced by 17% (p < 0.05) compared to the control cells. However, the distribution of cells by cell cycle phases did not differ. In cells pre-treated with oxamate, the rate of glucose consumption decreased by 20% (p < 0.05), ROS generation was reduced by 17%, vimentin expression decreased by 10% while the rate of lactate production was the same in oxamate-pretreated and control cells. CONCLUSION The cytostatic effect of oxamate demonstrated in adhesive growth conditions persisted for 72 h in the anchorage-independent growth conditions. The absence of differences in the cell cycle phase distribution and a decrease in the ROS generation may indicate the initial stage of overcoming the cytostatic effect of oxamate after 72 h of culturing LLC cells in anchorage- independent growth conditions.
Collapse
Affiliation(s)
- Yu Stepanov
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - D Kolesnik
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Yu Yakshibaeva
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - G Solyanik
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| |
Collapse
|
3
|
Wang H, Hu J, Zhou W, Qian A. Metabolic reprogramming in the pathogenesis and progression of nasopharyngeal carcinoma: molecular mechanisms and therapeutic implications. Am J Cancer Res 2024; 14:4049-4064. [PMID: 39267663 PMCID: PMC11387871 DOI: 10.62347/vyat9271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a unique head and neck cancer with a complex etiology involving genetic predispositions, environmental factors, and Epstein-Barr virus (EBV) infection. Despite progress in radiotherapy and chemotherapy, the prognosis for advanced NPC is still unfavorable, prompting the need for innovative therapeutic approaches. Metabolic reprogramming plays a crucial role in the development and progression of NPC, marked by substantial changes in glycolysis, lipid, and amino acid metabolism. These alterations aid tumor cell proliferation, survival under stress, and immune evasion, with features such as enhanced aerobic glycolysis (Warburg effect) and shifts in lipid and amino acid pathways. Oncogenic drivers like MYC, RAS, EGFR, and the loss of tumor suppressors such as TP53 and PTEN, along with key signaling pathways including mTOR, AMPK, and HIF-1α, orchestrate these metabolic changes. This review discusses the molecular mechanisms of metabolic reprogramming in NPC and outlines potential therapeutic targets within these pathways. Advances in metabolic imaging and biomarker discovery are also enhancing the precision of diagnostics and treatment monitoring, fostering personalized medicine in NPC treatment. This manuscript aims to provide a detailed overview of the current research and its implications for improving NPC management and patient outcomes through targeted metabolic therapies.
Collapse
Affiliation(s)
- Hongli Wang
- Department of Otolaryngology, The Affiliated People's Hospital of Ningbo University Ningbo, Zhejiang, China
| | - Jiandao Hu
- Department of Otolaryngology, The Affiliated People's Hospital of Ningbo University Ningbo, Zhejiang, China
| | - Weibang Zhou
- Department of Otolaryngology, The Affiliated People's Hospital of Ningbo University Ningbo, Zhejiang, China
| | - Aijuan Qian
- Department of Otolaryngology, The Affiliated People's Hospital of Ningbo University Ningbo, Zhejiang, China
| |
Collapse
|
4
|
Zhou Z, Xu F, Zhang T. Circular RNA COL1A1 promotes Warburg effect and tumor growth in nasopharyngeal carcinoma. Discov Oncol 2024; 15:120. [PMID: 38619648 PMCID: PMC11018599 DOI: 10.1007/s12672-024-00941-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 03/18/2024] [Indexed: 04/16/2024] Open
Abstract
OBJECTIVE Circular RNAs (circRNAs), pivotal in the pathogenesis and progression of nasopharyngeal carcinoma (NPC), remain a significant point of investigation for potential therapeutic interventions. Our research was driven by the objective to decipher the roles and underlying mechanisms of hsa_circ_0044569 (circCOL1A1) in governing the malignant phenotypes and the Warburg effect in NPC. METHODS We systematically collected samples from NPC tissues and normal nasopharyngeal epithelial counterparts. The expression levels of circCOL1A1, microRNA-370-5p (miR-370-5p), and prothymosin alpha (PTMA) were quantitatively determined using quantitative polymerase chain reaction (qPCR) and Western blotting. Transfections in NPC cell lines were conducted using small interfering RNAs (siRNAs) or vectors carrying the pcDNA 3.1 construct for overexpression studies. We interrogated the circCOL1A1/miR-370-5p/PTMA axis's role in cellular functions through a series of assays: 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide for cell viability, colony formation for growth, Transwell assays for migration and invasion, and Western blotting for protein expression profiling. To elucidate the molecular interactions, we employed luciferase reporter assays and RNA immunoprecipitation techniques. RESULTS Our investigations revealed that circCOL1A1 was a stable circRNA, highly expressed in both NPC tissues and derived cell lines. A correlation analysis with clinical pathological features demonstrated a significant association between circCOL1A1 expression, lymph node metastasis, and the tumor node metastasis staging system of NPC. Functionally, silencing circCOL1A1 led to substantial suppression of cell proliferation, migration, invasion, and metabolic alterations characteristic of the Warburg effect in NPC cells. At the molecular level, circCOL1A1 appeared to modulate PTMA expression by acting as a competitive endogenous RNA or 'sponge' for miR-370-5p, which in turn promoted the malignant characteristics of NPC cells. CONCLUSION To conclude, our findings delineate that circCOL1A1 exerts its oncogenic influence in NPC through the modulation of the miR-370-5p/PTMA signaling axis.
Collapse
Affiliation(s)
- ZeJun Zhou
- Department of Otolaryngology, The First Affiliated Hospital of Jinan University, No. 613 West Huangpu Avenue, Tianhe District, Guangzhou, 510630, Guangdong, China
| | - Fang Xu
- Health Management Center, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, Guangdong, China
| | - Tao Zhang
- Department of Otolaryngology, The First Affiliated Hospital of Jinan University, No. 613 West Huangpu Avenue, Tianhe District, Guangzhou, 510630, Guangdong, China.
| |
Collapse
|
5
|
Jia C, Wu Y, Gao F, Liu W, Li N, Chen Y, Sun L, Wang S, Yu C, Bao Y, Song Z. The opposite role of lactate dehydrogenase a (LDHA) in cervical cancer under energy stress conditions. Free Radic Biol Med 2024; 214:2-18. [PMID: 38307156 DOI: 10.1016/j.freeradbiomed.2024.01.043] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/16/2024] [Accepted: 01/24/2024] [Indexed: 02/04/2024]
Abstract
Due to insufficient and defective vascularization, the tumor microenvironment is often nutrient-depleted. LDHA has been demonstrated to play a tumor-promoting role by facilitating the glycolytic process. However, whether and how LDHA regulates cell survival in the nutrient-deficient tumor microenvironment is still unclear. Here, we sought to investigate the role and mechanism of LDHA in regulating cell survival and proliferation under energy stress conditions. Our results showed that the aerobic glycolysis levels, cell survival and proliferation of cervical cancer cells decreased significantly after inhibition of LDHA under normal culture condition while LDHA deficiency greatly inhibited glucose starvation-induced ferroptosis and promoted cell proliferation and tumor formation under energy stress conditions. Mechanistic studies suggested that glucose metabolism shifted from aerobic glycolysis to mitochondrial OXPHOS under energy stress conditions and LDHA knockdown increased accumulation of pyruvate in the cytosol, which entered the mitochondria and upregulated the level of oxaloacetate by phosphoenolpyruvate carboxylase (PC). Importantly, the increase in oxaloacetate production after absence of LDHA remarkably activated AMP-activated protein kinase (AMPK), which increased mitochondrial biogenesis and mitophagy, promoted mitochondrial homeostasis, thereby decreasing ROS level. Moreover, repression of lipogenesis by activation of AMPK led to elevated levels of reduced nicotinamide adenine dinucleotide phosphate (NADPH), which effectively resisted ROS-induced cell ferroptosis and enhanced cell survival under energy stress conditions. These results suggested that LDHA played an opposing role in survival and proliferation of cervical cancer cells under energy stress conditions, and inhibition of LDHA may not be a suitable treatment strategy for cervical cancer.
Collapse
Affiliation(s)
- Chaoran Jia
- NMPA Key Laboratory for Quality Control of Cell and Gene Therapy Medicine Products, Northeast Normal University, Changchun, 130024, China; National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, 130117, China
| | - Yulun Wu
- NMPA Key Laboratory for Quality Control of Cell and Gene Therapy Medicine Products, Northeast Normal University, Changchun, 130024, China
| | - Feng Gao
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, 130117, China
| | - Wei Liu
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, 130117, China
| | - Na Li
- NMPA Key Laboratory for Quality Control of Cell and Gene Therapy Medicine Products, Northeast Normal University, Changchun, 130024, China
| | - Yao Chen
- NMPA Key Laboratory for Quality Control of Cell and Gene Therapy Medicine Products, Northeast Normal University, Changchun, 130024, China; National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, 130117, China
| | - Luguo Sun
- NMPA Key Laboratory for Quality Control of Cell and Gene Therapy Medicine Products, Northeast Normal University, Changchun, 130024, China
| | - Shuyue Wang
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, 130117, China
| | - Chunlei Yu
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, 130117, China
| | - Yongli Bao
- NMPA Key Laboratory for Quality Control of Cell and Gene Therapy Medicine Products, Northeast Normal University, Changchun, 130024, China.
| | - Zhenbo Song
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, 130117, China.
| |
Collapse
|
6
|
Li Y, Chen L, Zheng Q, Liu G, Wang M, Wei S, Chen T. Lactate dehydrogenase A promotes nasopharyngeal carcinoma progression through the TAK1/NF-κB Axis. Mol Biol Rep 2024; 51:152. [PMID: 38236332 DOI: 10.1007/s11033-023-09130-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 12/07/2023] [Indexed: 01/19/2024]
Abstract
BACKGROUND Nasopharyngeal carcinoma (NPC) is a malignant tumor that originates in the nasopharyngeal mucosa and is common in China and Southeast Asian countries. Cancer cells reprogram glycolytic metabolism to promote their growth, survival and metastasis. Glycolysis plays an important role in NPC development, but the underlying mechanisms remain incompletely elucidated. Lactate dehydrogenase A (LDHA) is a crucial glycolytic enzyme, catalyzing the last step of glycolysis. This study aims to investigate the exact role of LDHA, which catalyzes the conversion of pyruvate into lactate, in NPC development. METHODS AND RESULTS The western blot and immunohistochemical (IHC) results indicated that LDHA was significantly upregulated in NPC cells and clinical samples. LDHA knockdown by shRNA significantly inhibited NPC cell proliferation and invasion. Further knockdown of LDHA dramatically weakened the tumorigenicity of NPC cells in vivo. Mechanistic studies showed that LDHA activated TGF-β-activated kinase 1 (TAK1) and subsequent nuclear factor κB (NF-κB) signaling to promote NPC cell proliferation and invasion. Exogenous lactate supplementation restored NPC cell proliferation and invasion inhibited by LDHA knockdown, and this restorative effect was reversed by NF-κB inhibitor (BAY 11-7082) or TAK1 inhibitor (5Z-7-oxozeaenol) treatment. Moreover, clinical sample analyses showed that LDHA expression was positively correlated with TAK1 Thr187 phosphorylation and poor prognosis. CONCLUSIONS Our results suggest that LDHA and its major metabolite lactate drive NPC progression by regulating TAK1 and its downstream NF-κB signaling, which could become a therapeutic target in NPC.
Collapse
Affiliation(s)
- Yingzi Li
- State Key Laboratory of Respiratory Disease at People's Hospital of Yangjiang, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
- Yangjiang Key Laboratory of Respiratory Disease, Department of Respiratory Medicine, People's Hospital of Yangjiang, Yangjiang, 529500, Guangdong, China
| | - Lanfang Chen
- State Key Laboratory of Respiratory Disease at People's Hospital of Yangjiang, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Qiaochong Zheng
- Yangjiang Key Laboratory of Respiratory Disease, Department of Respiratory Medicine, People's Hospital of Yangjiang, Yangjiang, 529500, Guangdong, China
| | - Guanxin Liu
- State Key Laboratory of Respiratory Disease at People's Hospital of Yangjiang, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Mengjiao Wang
- Clinical Research Lab Center, Guizhou Provincial People's Hospital, Guizhou University Medical College, Guiyang, 550025, Guizhou, China
| | - Shupei Wei
- State Key Laboratory of Respiratory Disease, Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong Higher Education Institutes, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, 510095, Guangdong, China.
| | - Tao Chen
- State Key Laboratory of Respiratory Disease at People's Hospital of Yangjiang, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China.
- Yangjiang Key Laboratory of Respiratory Disease, Department of Respiratory Medicine, People's Hospital of Yangjiang, Yangjiang, 529500, Guangdong, China.
| |
Collapse
|
7
|
Chen MK, Xiao ZY, Huang ZP, Xue KY, Xia H, Zhou JW, Liao DY, Liang ZJ, Xie X, Wei QZ, Zhong L, Yang JK, Liu CD, Liu Y, Zhao SC. Glycine Decarboxylase (GLDC) Plays a Crucial Role in Regulating Energy Metabolism, Invasion, Metastasis and Immune Escape for Prostate Cancer. Int J Biol Sci 2023; 19:4726-4743. [PMID: 37781511 PMCID: PMC10539704 DOI: 10.7150/ijbs.85893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 08/29/2023] [Indexed: 10/03/2023] Open
Abstract
Glycine decarboxylase (GLDC) is one of the core enzymes for glycine metabolism, and its biological roles in prostate cancer (PCa) are unclear. First, we found that GLDC plays a central role in glycolysis in 540 TCGA PCa patients. Subsequently, a metabolomic microarray showed that GLDC enhanced aerobic glycolysis in PCa cells, and GLDC and its enzyme activity enhanced glucose uptake, lactate production and lactate dehydrogenase (LDH) activity in PCa cells. Next, we found that GLDC was highly expressed in PCa, was directly regulated by hypoxia-inducible factor (HIF1-α) and regulated downstream LDHA expression. In addition, GLDC and its enzyme activity showed a strong ability to promote the migration and invasion of PCa both in vivo and in vitro. Furthermore, we found that the GLDC-high group had a higher TP53 mutation frequency, lower CD8+ T-cell infiltration, higher immune checkpoint expression, and higher immune exclusion scores than the GLDC-low group. Finally, the GLDC-based prognostic risk model by applying LASSO Cox regression also showed good predictive power for the clinical characteristics and survival in PCa patients. This evidence indicates that GLDC plays crucial roles in glycolytic metabolism, invasion and metastasis, and immune escape in PCa, and it is a potential therapeutic target for prostate cancer.
Collapse
Affiliation(s)
- Ming-kun Chen
- Department of Urology, NanFang Hospital, Southern Medical University, Guangzhou, 510515, China
- Department of Urology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, China
- The Third Clinical College of Southern Medical University, Guangzhou, 510630, China
| | - Zhuo-Yu Xiao
- Department of Urology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, China
- The Third Clinical College of Southern Medical University, Guangzhou, 510630, China
- Medical College of Shaoguan University, Shaoguan, 512026, China
| | - Zhi-Peng Huang
- Department of Urology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, China
- The Third Clinical College of Southern Medical University, Guangzhou, 510630, China
| | - Kang-Yi Xue
- Department of Urology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, China
- The Third Clinical College of Southern Medical University, Guangzhou, 510630, China
| | - Hui Xia
- Department of Urology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, China
- The Third Clinical College of Southern Medical University, Guangzhou, 510630, China
| | - Jia-Wei Zhou
- Department of Urology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, China
- The Third Clinical College of Southern Medical University, Guangzhou, 510630, China
| | - De-Ying Liao
- Department of Urology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, China
- The Third Clinical College of Southern Medical University, Guangzhou, 510630, China
| | - Zhi-Jian Liang
- Department of Urology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, China
- The Third Clinical College of Southern Medical University, Guangzhou, 510630, China
| | - Xiao Xie
- Department of Urology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, China
- The Third Clinical College of Southern Medical University, Guangzhou, 510630, China
| | - Qing-Zhu Wei
- Department of Pathology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, China
| | - Lin Zhong
- Department of Pathology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, China
| | - Jian-Kun Yang
- Department of Urology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, China
- The Third Clinical College of Southern Medical University, Guangzhou, 510630, China
| | - Cun-Dong Liu
- Department of Urology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, China
- The Third Clinical College of Southern Medical University, Guangzhou, 510630, China
| | - Yang Liu
- Department of Urology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, China
- The Third Clinical College of Southern Medical University, Guangzhou, 510630, China
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150007, China
| | - Shan-Chao Zhao
- Department of Urology, NanFang Hospital, Southern Medical University, Guangzhou, 510515, China
- Department of Urology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, China
- The Third Clinical College of Southern Medical University, Guangzhou, 510630, China
| |
Collapse
|
8
|
Tang Y, Gu S, Zhu L, Wu Y, Zhang W, Zhao C. LDHA: The Obstacle to T cell responses against tumor. Front Oncol 2022; 12:1036477. [PMID: 36518315 PMCID: PMC9742379 DOI: 10.3389/fonc.2022.1036477] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 11/03/2022] [Indexed: 11/16/2023] Open
Abstract
Immunotherapy has become a successful therapeutic strategy in certain solid tumors and hematological malignancies. However, this efficacy of immunotherapy is impeded by limited success rates. Cellular metabolic reprogramming determines the functionality and viability in both cancer cells and immune cells. Extensive research has unraveled that the limited success of immunotherapy is related to immune evasive metabolic reprogramming in tumor cells and immune cells. As an enzyme that catalyzes the final step of glycolysis, lactate dehydrogenase A (LDHA) has become a major focus of research. Here, we have addressed the structure, localization, and biological features of LDHA. Furthermore, we have discussed the various aspects of epigenetic regulation of LDHA expression, such as histone modification, DNA methylation, N6-methyladenosine (m6A) RNA methylation, and transcriptional control by noncoding RNA. With a focus on the extrinsic (tumor cells) and intrinsic (T cells) functions of LDHA in T-cell responses against tumors, in this article, we have reviewed the current status of LDHA inhibitors and their combination with T cell-mediated immunotherapies and postulated different strategies for future therapeutic regimens.
Collapse
Affiliation(s)
- Yu Tang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Shuangshuang Gu
- Shanghai Institute of Rheumatology, Shanghai Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Liqun Zhu
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yujiao Wu
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Wei Zhang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Chuanxiang Zhao
- Institute of Medical Genetics and Reproductive Immunity, School of Medical Science and Laboratory Medicine, Jiangsu College of Nursing, Huai’an, Jiangsu, China
| |
Collapse
|
9
|
Herheliuk TS, Perepelytsina OM, Chmelnytska YM, Kuznetsova GM, Dzjubenko NV, Raksha NG, Gorbach OI, Sydorenko MV. Study of Cancer Stem Cell Subpopulations in Breast Cancer Models. CYTOL GENET+ 2022. [DOI: 10.3103/s0095452722040041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
10
|
The Emerging Significance of Histone Lysine Demethylases as Prognostic Markers and Therapeutic Targets in Head and Neck Cancers. Cells 2022; 11:cells11061023. [PMID: 35326475 PMCID: PMC8946939 DOI: 10.3390/cells11061023] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 02/04/2023] Open
Abstract
Epigenetic aberrations, associated with altered DNA methylation profiles and global changes in the level of histone modifications, are commonly detected in head and neck squamous cell carcinomas (HNSCC). Recently, histone lysine demethylases have been implicated in the pathogenesis of HNSCC and emerged as potential molecular targets. Histone lysine demethylases (KDMs) catalyze the removal of methyl groups from lysine residues in histones. By affecting the methylation of H3K4, H3K9, H3K27, or H3K36, these enzymes take part in transcriptional regulation, which may result in changes in the level of expression of tumor suppressor genes and protooncogenes. KDMs are involved in many biological processes, including cell cycle control, senescence, DNA damage response, and heterochromatin formation. They are also important regulators of pluripotency. The overexpression of most KDMs has been observed in HNSCC, and their inhibition affects cell proliferation, apoptosis, cell motility, invasiveness, and stemness. Of all KDMs, KDM1, KDM4, KDM5, and KDM6 proteins are currently regarded as the most promising prognostic and therapeutic targets in head and neck cancers. The aim of this review is to present up-to-date knowledge on the significance of histone lysine demethylases in head and neck carcinogenesis and to discuss the possibility of using them as prognostic markers and pharmacological targets in patients’ treatment.
Collapse
|
11
|
Wang X, Chen P. Aberrant miR-362-3p is Associated with EBV-Infection and Prognosis in Nasopharyngeal Carcinoma and Involved in Tumor Progression by Targeting JMJD2A. Onco Targets Ther 2022; 15:121-131. [PMID: 35115787 PMCID: PMC8806052 DOI: 10.2147/ott.s325100] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 09/02/2021] [Indexed: 12/14/2022] Open
Abstract
Background Many microRNAs (miRNAs) are involved in the progression of nasopharyngeal carcinoma (NPC). This study aimed to examine the expression and clinical significance of microRNA (miR)-362-3p in NPC, especially in Epstein–Barr virus (EBV)-positive patients, and explore its potential mechanism in NPC progression. Methods miR-362-3p levels and Jumonji C domain 2A (JMJD2A) mRNA levels were detected by quantitative real-time PCR. The diagnostic value of miR-362-3p to distinguish NPC patients and EBV-positive cases was evaluated using receiver operating characteristic analysis. The association of miR-362-3p with NPC survival was assessed by Kaplan–Meier curves and Cox regression analysis. NPC cell proliferation, migration and invasion were determined using Cell Counting Kit-8 and Transwell assays, respectively. A luciferase reporter assay was used to confirm the interaction between miR-362-3p and JMJD2A. Results miR-362-3p expression was decreased in the serum and tissues of NPC patients and had diagnostic value for screening NPC. According to the survival follow-up, NPC survivors had significantly higher miR-362-3p, and miR-326-3p was demonstrated as an independent prognostic indicator of NPC. Interestingly, it is found that EBV-positive NPC patients and cells had significantly lower miR-362-3p compared with EBV-negative NPC patients and cells and had certain ability to distinguish EBV-positive patients. Moreover, miR-362-3p inhibited the proliferation, migration and invasion of both EBV-positive and -negative NPC cells, and these effects might be mediated by targeting JMJD2A. Conclusion Abnormal miR-362-3p expression is related to EBV-infection and prognosis in NPC patients and may be involved in NPC progression by targeting JMJD2A.
Collapse
Affiliation(s)
- Xiangyun Wang
- Department of Otorhinolaryngology, Dongying People’s Hospital, Dongying, Shandong, 257091, People’s Republic of China
- Correspondence: Xiangyun Wang, Department of Otorhinolaryngology, Dongying People’s Hospital, No. 317 Nanyi Road, Dongying, Shandong, 257091, People’s Republic of China, Tel/Fax + 86-0546-8901191, Email
| | - Ping Chen
- Department of Otorhinolaryngology, Dongying People’s Hospital, Dongying, Shandong, 257091, People’s Republic of China
| |
Collapse
|
12
|
Zoccarato A, Nabeebaccus AA, Oexner RR, Santos CXC, Shah AM. The nexus between redox state and intermediary metabolism. FEBS J 2021; 289:5440-5462. [PMID: 34496138 DOI: 10.1111/febs.16191] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/20/2021] [Accepted: 09/07/2021] [Indexed: 12/12/2022]
Abstract
Reactive oxygen species (ROS) are not just a by-product of cellular metabolic processes but act as signalling molecules that regulate both physiological and pathophysiological processes. A close connection exists in cells between redox homeostasis and cellular metabolism. In this review, we describe how intracellular redox state and glycolytic intermediary metabolism are closely coupled. On the one hand, ROS signalling can control glycolytic intermediary metabolism by direct regulation of the activity of key metabolic enzymes and indirect regulation via redox-sensitive transcription factors. On the other hand, metabolic adaptation and reprogramming in response to physiological or pathological stimuli regulate intracellular redox balance, through mechanisms such as the generation of reducing equivalents. We also discuss the impact of these intermediary metabolism-redox circuits in physiological and disease settings across different tissues. A better understanding of the mechanisms regulating these intermediary metabolism-redox circuits will be crucial to the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Anna Zoccarato
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Adam A Nabeebaccus
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Rafael R Oexner
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Celio X C Santos
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Ajay M Shah
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| |
Collapse
|
13
|
Xu QL, Luo Z, Zhang B, Qin GJ, Zhang RY, Kong XY, Tang HY, Jiang W. Methylation-associated silencing of miR-9-1 promotes nasopharyngeal carcinoma progression and glycolysis via HK2. Cancer Sci 2021; 112:4127-4138. [PMID: 34382305 PMCID: PMC8486208 DOI: 10.1111/cas.15103] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 08/06/2021] [Accepted: 08/08/2021] [Indexed: 12/23/2022] Open
Abstract
Characteristically, cancer cells metabolize glucose through aerobic glycolysis, known as the Warburg effect. Accumulating evidence suggest that during cancer formation, microRNAs (miRNAs) could regulate such metabolic reprogramming. In the present study, miR‐9‐1 was identified as significantly hypermethylated in nasopharyngeal carcinoma (NPC) cell lines and clinical tissues. Ectopic expression of miR‐9‐1 inhibited NPC cell growth and glycolytic metabolism, including reduced glycolysis, by reducing lactate production, glucose uptake, cellular glucose‐6‐phosphate levels, and ATP generation in vitro and tumor proliferation in vivo. HK2 (encoding hexokinase 2) was identified as a direct target of miR‐9‐1 using luciferase reporter assays and Western blotting. In NPC cells, hypermethylation regulates miR‐9‐1 expression and inhibits HK2 translation by directly targeting its 3' untranslated region. MiR‐9‐1 overexpression markedly reduced HK2 protein levels. Restoration of HK2 expression attenuated the inhibitory effect of miR‐9‐1 on NPC cell proliferation and glycolysis. Fluorescence in situ hybridization results indicated that miR‐9‐1 expression was an independent prognostic factor in NPC. Our findings revealed the role of the miR‐9‐1/HK2 axis in the metabolic reprogramming of NPC, providing a potential therapeutic strategy for NPC.
Collapse
Affiliation(s)
- Qian-Lan Xu
- Department of Radiation Oncology, Affiliated Hospital of Guilin Medical University, Guilin, China.,Department of Laboratory Animal Center, Southern Medical University, Guangzhou, China
| | - Zan Luo
- Department of Radiation Oncology, Affiliated Hospital of Guilin Medical University, Guilin, China.,Guangxi Key Laboratory of Tumor Immunology and Receptor Targeted Therapy, Guilin Medical University, Guilin, China.,Department of Oncology, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, China
| | - Bin Zhang
- Department of Radiation Oncology, Wuzhou Red Cross Hospital, Wuzhou, China
| | - Guan-Jie Qin
- Department of Radiation Oncology, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Ru-Yun Zhang
- Department of Radiation Oncology, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Xiang-Yun Kong
- Department of Radiation Oncology, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Hua-Ying Tang
- Department of Radiation Oncology, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Wei Jiang
- Department of Radiation Oncology, Affiliated Hospital of Guilin Medical University, Guilin, China.,Department of Laboratory Animal Center, Southern Medical University, Guangzhou, China
| |
Collapse
|
14
|
Zhao J, Li B, Ren Y, Liang T, Wang J, Zhai S, Zhang X, Zhou P, Zhang X, Pan Y, Gao F, Zhang S, Li L, Yang Y, Deng X, Li X, Chen L, Yang D, Zheng Y. Histone demethylase KDM4A plays an oncogenic role in nasopharyngeal carcinoma by promoting cell migration and invasion. Exp Mol Med 2021; 53:1207-1217. [PMID: 34385569 PMCID: PMC8417295 DOI: 10.1038/s12276-021-00657-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 06/07/2021] [Accepted: 06/15/2021] [Indexed: 02/07/2023] Open
Abstract
Compelling evidence has indicated the vital role of lysine-specific demethylase 4 A (KDM4A), hypoxia-inducible factor-1α (HIF1α) and the mechanistic target of rapamycin (mTOR) signaling pathway in nasopharyngeal carcinoma (NPC). Therefore, we aimed to investigate whether KDM4A affects NPC progression by regulating the HIF1α/DDIT4/mTOR signaling pathway. First, NPC and adjacent tissue samples were collected, and KDM4A protein expression was examined by immunohistochemistry. Then, the interactions among KDM4A, HIF1α and DDIT4 were assessed. Gain- and loss-of-function approaches were used to alter KDM4A, HIF1α and DDIT4 expression in NPC cells. The mechanism of KDM4A in NPC was evaluated both in vivo and in vitro via RT-qPCR, Western blot analysis, MTT assay, Transwell assay, flow cytometry and tumor formation experiments. KDM4A, HIF1α, and DDIT4 were highly expressed in NPC tissues and cells. Mechanistically, KDM4A inhibited the enrichment of histone H3 lysine 9 trimethylation (H3K9me3) in the HIF1α promoter region and thus inhibited the methylation of HIF1α to promote HIF1α expression, thus upregulating DDIT4 and activating the mTOR signaling pathway. Overexpression of KDM4A, HIF1α, or DDIT4 or activation of the mTOR signaling pathway promoted SUNE1 cell proliferation, migration, and invasion but inhibited apoptosis. KDM4A silencing blocked the mTOR signaling pathway by inhibiting the HIF1α/DDIT4 axis to inhibit the growth of SUNE1 cells in vivo. Collectively, KDM4A silencing could inhibit NPC progression by blocking the activation of the HIF1α/DDIT4/mTOR signaling pathway by increasing H3K9me3, highlighting a promising therapeutic target for NPC.
Collapse
Affiliation(s)
- Jingyi Zhao
- Radiotherapy Department, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Bingyan Li
- Radiotherapy Department, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Yongxia Ren
- Radiotherapy Department, Huaihe Hospital of Henan University, Kaifeng, PR China
| | - Tiansong Liang
- Radiotherapy Department, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Juan Wang
- Radiotherapy Department, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Suna Zhai
- Radiotherapy Department, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Xiqian Zhang
- Radiotherapy Department, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Pengcheng Zhou
- Radiotherapy Department, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Xiangxian Zhang
- Radiotherapy Department, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Yuanyuan Pan
- Radiotherapy Department, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Fangfang Gao
- Radiotherapy Department, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Sulan Zhang
- Institute of Radiation Therapy and Tumor Critical Care of Zhengzhou University, Zhengzhou, PR China
| | - Liming Li
- Henan Key Laboratory of Molecular Radiotherapy, Zhengzhou, PR China
| | - Yongqiang Yang
- Radiotherapy Department, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Xiaoyu Deng
- Radiotherapy Department, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Xiaole Li
- Radiotherapy Department, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Linhui Chen
- Radiotherapy Department, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Daoke Yang
- Radiotherapy Department, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China.
| | - Yingjuan Zheng
- Radiotherapy Department, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China.
| |
Collapse
|
15
|
Rewiring of lactate-IL-1β auto-regulatory loop with Clock-Bmal1: A feed-forward circuit in glioma. Mol Cell Biol 2021; 41:e0044920. [PMID: 34124933 DOI: 10.1128/mcb.00449-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
De-synchronized circadian rhythm in tumors is coincident with aberrant inflammation and dysregulated metabolism. As their inter-relationship in cancer etiology is largely unknown, we investigated the link between the three in glioma. Tumor metabolite lactate- mediated increase in pro-inflammatory cytokine IL-1β was concomitant with elevated levels of core circadian regulators Clock and Bmal1. siRNA mediated knockdown of Bmal1 and Clock decreased (i) LDHA and IL-1β levels and (ii) release of lactate and pro-inflammatory cytokines. Lactate mediated deacetylation of Bmal1 and its interaction with Clock, regulate IL-1β levels and vice versa. Site-directed mutagenesis and luciferase reporter assay indicated the functionality of E-box sites on LDHA and IL-1β promoters. ChIP-re-ChIP revealed that lactate-IL-1β crosstalk positively affects co-recruitment of Clock-Bmal1 to these E-box sites. Clock-Bmal1 enrichment was accompanied by decreased H3K9me3, and increased H3K9ac and RNA pol II occupancy. Lactate-IL-1β-Clock (LIC) loop positively regulated expression of genes associated with cell cycle, DNA damage and cytoskeletal organization involved in glioma progression. TCGA data analysis suggested the presence of lactate- IL-1β-crosstalk in other cancers. The responsiveness of stomach and cervical cancer cells to lactate inhibition followed the same trend exhibited by glioma cells. In addition, components of LIC loop were found to be correlated with (i) patient survival, (ii) clinically actionable genes, and (iii) anti-cancer drug sensitivity. Our findings provide evidence for a potential cancer-specific axis wiring of IL-1β and LDHA through Clock -Bmal1, the outcome of which is to fuel an IL-1β-lactate autocrine loop that drives pro-inflammatory and oncogenic signals.
Collapse
|
16
|
Wu J, You K, Chen C, Zhong H, Jiang Y, Mo H, Song J, Qiu X, Liu Y. High Pretreatment LDH Predicts Poor Prognosis in Hypopharyngeal Cancer. Front Oncol 2021; 11:641682. [PMID: 33777804 PMCID: PMC7991725 DOI: 10.3389/fonc.2021.641682] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 02/02/2021] [Indexed: 12/30/2022] Open
Abstract
Background Elevated pretreatment lactate dehydrogenase (LDH) has been associated with poor prognosis in various malignancies; however, its prognostic role in hypopharyngeal cancer remains elusive. In this study, we aimed to assess the association between pretreatment LDH and clinical outcome of hypopharyngeal cancer. Methods We retrospectively collected 198 hypopharyngeal cancer patients treated with surgery in our institution between 2004 and 2018. The prognostic role of pretreatment LDH was explored by using univariate and multivariate analyses. Besides, subgroup analysis was performed based on T stage. Results Three-year and Five-year of disease-free survival (DFS, 67.0 vs. 57.4%, 65.8 vs. 39.8%, p = 0.007) and overall survival (OS, 74.8 vs. 68.9%, 66.8 vs. 50.8%, p = 0.006) exhibited significant differences between low LDH level and high LDH level groups. Univariate analysis showed that pretreatment elevated serum LDH served as an unfavorable determinant with regard to DFS and OS. Further multivariate analysis also confirmed that LDH was an independent predictor for DFS and OS. Additionally, N status and age were also found to be significantly associated with both DFS and OS. Conclusion Pretreatment elevated serum LDH is an inferior prognostic factor for patients with hypopharyngeal cancer. These results should be validated by more multicenter and prospective studies.
Collapse
Affiliation(s)
- Jialing Wu
- Department of Radiation Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Kaiyun You
- Department of Radiation Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Changlong Chen
- Department of Radiation Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Huimin Zhong
- Department of Radiation Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yanhui Jiang
- Department of Radiation Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Huaqian Mo
- Department of Radiation Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Juanjuan Song
- Department of Radiation Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xingsheng Qiu
- Department of Radiation Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yimin Liu
- Department of Radiation Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
17
|
Mechanistic insights into KDM4A driven genomic instability. Biochem Soc Trans 2021; 49:93-105. [PMID: 33492339 PMCID: PMC7925003 DOI: 10.1042/bst20191219] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 12/11/2020] [Accepted: 12/15/2020] [Indexed: 12/19/2022]
Abstract
Alterations in global epigenetic signatures on chromatin are well established to contribute to tumor initiation and progression. Chromatin methylation status modulates several key cellular processes that maintain the integrity of the genome. KDM4A, a demethylase that belongs to the Fe-II dependent dioxygenase family that uses α-ketoglutarate and molecular oxygen as cofactors, is overexpressed in several cancers and is associated with an overall poor prognosis. KDM4A demethylates lysine 9 (H3K9me2/3) and lysine 36 (H3K36me3) methyl marks on histone H3. Given the complexity that exists with these marks on chromatin and their effects on transcription and proliferation, it naturally follows that demethylation serves an equally important role in these cellular processes. In this review, we highlight the role of KDM4A in transcriptional modulation, either dependent or independent of its enzymatic activity, arising from the amplification of this demethylase in cancer. KDM4A modulates re-replication of distinct genomic loci, activates cell cycle inducers, and represses proteins involved in checkpoint control giving rise to proliferative damage, mitotic disturbances and chromosomal breaks, ultimately resulting in genomic instability. In parallel, emerging evidence of non-nuclear substrates of epigenetic modulators emphasize the need to investigate the role of KDM4A in regulating non-nuclear substrates and evaluate their contribution to genomic instability in this context. The existence of promising KDM-specific inhibitors makes these demethylases an attractive target for therapeutic intervention in cancers.
Collapse
|
18
|
Construction and Analysis of lncRNA-Mediated ceRNA Network in Nasopharyngeal Carcinoma Based on Weighted Correlation Network Analysis. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1468980. [PMID: 33102573 PMCID: PMC7569441 DOI: 10.1155/2020/1468980] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/12/2020] [Accepted: 09/25/2020] [Indexed: 12/24/2022]
Abstract
Increasing evidence indicated that aberrant expression of long noncoding RNAs (lncRNAs) are involved in tumorigenesis of nasopharyngeal carcinoma (NPC). The purpose of this study was to construct a lncRNA-mediated ceRNA network based on weighted correlation network analysis (WGCNA). First, modules with highly correlated genes were identified from GSE102349 via WGCNA, and the preservation of the modules was evaluated by GSE68799. Then, the differentially expressed lncRNAs and mRNAs identified from GSE12452 which belonged to the same WGCNA modules and the differentially expressed miRNAs identified from GSE32960 were used to construct a ceRNA network. The prognostic value of the network was evaluated by survival analysis. Furthermore, a risk score model for predicting progression-free survival (PFS) of NPC patients was established via LASSO-penalized Cox regression, and the differences in the expression of the lncRNAs between high- and low-risk groups were investigated. Finally, 14 stable modules were identified, and a ceRNA network composed of 11 lncRNAs, 15 miRNAs, and 40 mRNAs was established. The lncRNAs and mRNAs in the network belonged to the turquoise and salmon modules. Survival analysis indicated that ZNF667-AS1, LDHA, LMNB2, TPI1, UNG, and hsa-miR-142-3p were significantly correlated with the prognosis of NPC. Gene set enrichment analysis indicated that the upregulation of ZNF667-AS1 was associated with some immune-related pathways. Besides, a risk score model consisting of 12 genes was constructed and showed a good performance in predicting PFS for NPC patients. Among the 11 lncRNAs in the ceRNA network, SNHG16, SNHG17, and THAP9-AS1 were upregulated in the high-risk group of NPC, while ZNF667-AS1 was downregulated in the high-risk group of NPC. These results will promote our understanding of the crosstalk among lncRNAs, miRNAs, and mRNAs in the tumorigenesis and progression of NPC.
Collapse
|
19
|
Sellam LS, Zappasodi R, Chettibi F, Djennaoui D, Yahi-Ait Mesbah N, Amir-Tidadini ZC, Touil-Boukoffa C, Ouahioune W, Merghoub T, Bourouba M. Silibinin down-regulates PD-L1 expression in nasopharyngeal carcinoma by interfering with tumor cell glycolytic metabolism. Arch Biochem Biophys 2020; 690:108479. [PMID: 32679194 PMCID: PMC8507490 DOI: 10.1016/j.abb.2020.108479] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/09/2020] [Accepted: 06/21/2020] [Indexed: 02/06/2023]
Abstract
The upregulation of checkpoint inhibitor PD-L1 expression has recently been associated with nasopharyngeal carcinoma (NPC) resistance to therapy. The mechanism of induction of PD-L1 has also been linked to enhanced aerobic glycolysis promoted by HIF1-α dysregulation and LDH-A activity in cancer. Here, we investigated the effect of the anti-tumoral compound Silibinin on HIF-1α/LDH-A mediated cancer cell metabolism and PD-L1 expression in NPC. Our results demonstrate that exposure to Silibinin potently inhibits tumor growth and promotes a shift from aerobic glycolysis toward oxidative phosphorylation. The EBV + NPC cell line C666-1 and glycolytic human tumor explants treated with Silibinin displayed a reduction in LDH-A activity which consistently associated with a reduction in lactate levels. This effect was accompanied by an increase in intracellular citrate levels in C666-1 cells. Accordingly, expression of HIF-1α, a critical regulator of glycolysis, was down-regulated after treatment. This event associated with a down-regulation in PD-L1. Altogether, our results provide evidence that silibinin can alter PD-L1 expression by interfering with HIF-1α/LDH-A mediated cell metabolism in NPC. These results provide a new perspective for Silibinin use to overcome PD-L1 mediated NPC resistance to therapy.
Collapse
Affiliation(s)
- Leïla Sarah Sellam
- Cell and Molecular Biology Laboratory (LBCM), Team Cytokines and Nitric oxide Synthase: Immunity and Pathogeny, Faculty of Biological Sciences, University of Sciences and Technology Houari Boumediene (USTHB), Bab Ezzouar, Algiers, Algeria
| | - Roberta Zappasodi
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Fayçal Chettibi
- Otorhinolaryngology Department, Mustapha Pacha Hospital, Algiers, Algeria
| | - Djamel Djennaoui
- Otorhinolaryngology Department, Mustapha Pacha Hospital, Algiers, Algeria
| | | | | | - Chafia Touil-Boukoffa
- Cell and Molecular Biology Laboratory (LBCM), Team Cytokines and Nitric oxide Synthase: Immunity and Pathogeny, Faculty of Biological Sciences, University of Sciences and Technology Houari Boumediene (USTHB), Bab Ezzouar, Algiers, Algeria
| | - Wahiba Ouahioune
- Central Laboratory for Anatomopathology, Frantz Fanon Hospital, Blida, Algeria
| | - Taha Merghoub
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Medicine, New York, NY 10065, USA
| | - Mehdi Bourouba
- Cell and Molecular Biology Laboratory (LBCM), Team Cytokines and Nitric oxide Synthase: Immunity and Pathogeny, Faculty of Biological Sciences, University of Sciences and Technology Houari Boumediene (USTHB), Bab Ezzouar, Algiers, Algeria.
| |
Collapse
|
20
|
Thomas R, Shaath H, Naik A, Toor SM, Elkord E, Decock J. Identification of two HLA-A*0201 immunogenic epitopes of lactate dehydrogenase C (LDHC): potential novel targets for cancer immunotherapy. Cancer Immunol Immunother 2020; 69:449-463. [PMID: 31932876 PMCID: PMC7044258 DOI: 10.1007/s00262-020-02480-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 01/04/2020] [Indexed: 12/13/2022]
Abstract
Lactate dehydrogenase C (LDHC) is an archetypical cancer testis antigen with limited expression in adult tissues and re-expression in tumors. This restricted expression pattern together with the important role of LDHC in cancer metabolism renders LDHC a potential target for immunotherapy. This study is the first to investigate the immunogenicity of LDHC using T cells from healthy individuals. LDHC-specific T cell responses were induced by in vitro stimulation with synthetic peptides, or by priming with autologous peptide-pulsed dendritic cells. We evaluated T cell activation by IFN-γ ELISpot and determined cytolytic activity of HLA-A*0201-restricted T cells in breast cancer cell co-cultures. In vitro T cell stimulation induced IFN-γ secretion in response to numerous LDHC-derived peptides. Analysis of HLA-A*0201 responses revealed a significant T cell activation after stimulation with peptide pools 2 (PP2) and 8 (PP8). The PP2- and PP8-specific T cells displayed cytolytic activity against breast cancer cells with endogenous LDHC expression within a HLA-A*0201 context. We identified peptides LDHC41−55 and LDHC288−303 from PP2 and PP8 to elicit a functional cellular immune response. More specifically, we found an increase in IFN-γ secretion by CD8 + T cells and cancer-cell-killing of HLA-A*0201/LDHC positive breast cancer cells by LDHC41−55- and LDHC288−303-induced T cells, albeit with a possible antigen recognition threshold. The majority of induced T cells displayed an effector memory phenotype. To conclude, our findings support the rationale to assess LDHC as a targetable cancer testis antigen for immunotherapy, and in particular the HLA-A*0201 restricted LDHC41–55 and LDHC288–303 peptides within LDHC.
Collapse
Affiliation(s)
- Remy Thomas
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Hibah Shaath
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Adviti Naik
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Salman M Toor
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Eyad Elkord
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Julie Decock
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar.
| |
Collapse
|
21
|
Zhao P, Zhou M, Chen R, Su R. Suppressed "Warburg Effect" in Nasopharyngeal Carcinoma Via the Inhibition of Pyruvate Kinase Type M2-Mediated Energy Generation Pathway. Technol Cancer Res Treat 2020; 19:1533033820945804. [PMID: 32815467 PMCID: PMC7444150 DOI: 10.1177/1533033820945804] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/31/2020] [Accepted: 06/19/2020] [Indexed: 12/24/2022] Open
Abstract
Warburg effect describes the abnormal energy metabolism in cancer cells and pyruvate kinase type M2 is involved in the regulation of this effect. In the current study, the role of pyruvate kinase type M2 in the initiation of Warburg effect in nasopharyngeal carcinoma cells was explored. The expression status of pyruvate kinase type M2 was detected in nasopharyngeal carcinoma samples and analyzed by different clinicopathological characteristics. Then the level of pyruvate kinase type M2 was suppressed in 2 nasopharyngeal carcinoma cell lines. The effects of pyruvate kinase type M2 inhibition on cell viability, apoptosis, invasion, glucose uptake, ATP generation, and glycolysis metabolism were determined. The data showed that the high expression of pyruvate kinase type M2 in nasopharyngeal carcinoma tissues was associated with the larger tumor size and advanced metastasis in the patients. The inhibition of pyruvate kinase type M2 resulted in the repressed proliferation and invasion in nasopharyngeal carcinoma cells, along with the increased apoptotic rate. The lack of pyruvate kinase type M2 function inhibited glucose uptake, while increased ATP generation in nasopharyngeal carcinoma cells. Moreover, the production of glycolysis metabolites, including pyruvic acid, lactate, citrate, and malate, was also suppressed by pyruvate kinase type M2 inhibition. At molecular level, the expressions of glucose transporter and hexokinase 2 were downregulated by pyruvate kinase type M2 inhibition, confirming the changes in glucose metabolism. Collectively, the current study demonstrated that the function of pyruvate kinase type M2 was important to maintain the proliferation and invasion of nasopharyngeal carcinoma cells, and the inhibition of the factor would antagonize nasopharyngeal carcinoma by blocking Warburg effect.
Collapse
Affiliation(s)
- Penglong Zhao
- Department of Otolaryngology-Head and Neck Surgery, The First People’s Hospital of Wenling, Wenling, Zhejiang Province, China
| | - Mengyan Zhou
- Department of Otolaryngology-Head and Neck Surgery, The First People’s Hospital of Wenling, Wenling, Zhejiang Province, China
| | - Ruixiang Chen
- Department of Otolaryngology-Head and Neck Surgery, The First People’s Hospital of Wenling, Wenling, Zhejiang Province, China
| | - Renjie Su
- Department of Otolaryngology-Head and Neck Surgery, The First People’s Hospital of Wenling, Wenling, Zhejiang Province, China
| |
Collapse
|
22
|
Cai H, Li J, Zhang Y, Liao Y, Zhu Y, Wang C, Hou J. LDHA Promotes Oral Squamous Cell Carcinoma Progression Through Facilitating Glycolysis and Epithelial-Mesenchymal Transition. Front Oncol 2019; 9:1446. [PMID: 31921691 PMCID: PMC6930919 DOI: 10.3389/fonc.2019.01446] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 12/03/2019] [Indexed: 12/26/2022] Open
Abstract
Aerobic glycolysis is the main pathway for energy metabolism in cancer cells. It provides energy and biosynthetic substances for tumor progression and metastasis by increasing lactate production. Lactate dehydrogenase A (LDHA) promotes glycolysis process by catalyzing the conversion of pyruvate to lactate. Despite LDHA exhibiting carcinogenesis in various cancers, its role in oral squamous cell carcinoma (OSCC) remains unclear. This study demonstrated that LDHA was over-expressed in both OSCC tissues and cell lines, and was significantly associated with lower overall survival rates in patients with OSCC. Using weighted gene correlation network analysis and gene set enrichment analysis for the gene expression data of patients with OSCC (obtained from The Cancer Genome Atlas database), a close association was identified between epithelial–mesenchymal transition (EMT) and LDHA in promoting OSCC progression. The knockdown of LDHA suppressed EMT, cell proliferation, and migration and invasion of OSCC cells in vitro. Moreover, the silencing of LDHA inhibited tumor growth in vivo. Oxamate, as a competitive LDHA inhibitor, was also suppressed diverse malignant biocharacteristics of OSCC cells. Our findings reveal that LDHA acts as an oncogene to promote malignant progression of OSCC by facilitating glycolysis and EMT, and LDHA may be a potential anticancer therapeutic target.
Collapse
Affiliation(s)
- Hongshi Cai
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Jiaxin Li
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Yadong Zhang
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Yan Liao
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Yue Zhu
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Cheng Wang
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Jinsong Hou
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
23
|
Lactate Dehydrogenases as Metabolic Links between Tumor and Stroma in the Tumor Microenvironment. Cancers (Basel) 2019; 11:cancers11060750. [PMID: 31146503 PMCID: PMC6627402 DOI: 10.3390/cancers11060750] [Citation(s) in RCA: 186] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 05/20/2019] [Accepted: 05/23/2019] [Indexed: 02/07/2023] Open
Abstract
Cancer is a metabolic disease in which abnormally proliferating cancer cells rewire metabolic pathways in the tumor microenvironment (TME). Molecular reprogramming in the TME helps cancer cells to fulfill elevated metabolic demands for bioenergetics and cellular biosynthesis. One of the ways through which cancer cell achieve this is by regulating the expression of metabolic enzymes. Lactate dehydrogenase (LDH) is the primary metabolic enzyme that converts pyruvate to lactate and vice versa. LDH also plays a significant role in regulating nutrient exchange between tumor and stroma. Thus, targeting human lactate dehydrogenase for treating advanced carcinomas may be of benefit. LDHA and LDHB, two isoenzymes of LDH, participate in tumor stroma metabolic interaction and exchange of metabolic fuel and thus could serve as potential anticancer drug targets. This article reviews recent research discussing the roles of lactate dehydrogenase in cancer metabolism. As molecular regulation of LDHA and LDHB in different cancer remains obscure, we also review signaling pathways regulating LDHA and LDHB expression. We highlight on the role of small molecule inhibitors in targeting LDH activity and we emphasize the development of safer and more effective LDH inhibitors. We trust that this review will also generate interest in designing combination therapies based on LDH inhibition, with LDHA being targeted in tumors and LDHB in stromal cells for better treatment outcome.
Collapse
|
24
|
Urbańska K, Orzechowski A. Unappreciated Role of LDHA and LDHB to Control Apoptosis and Autophagy in Tumor Cells. Int J Mol Sci 2019; 20:ijms20092085. [PMID: 31035592 PMCID: PMC6539221 DOI: 10.3390/ijms20092085] [Citation(s) in RCA: 178] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/23/2019] [Accepted: 04/24/2019] [Indexed: 12/19/2022] Open
Abstract
Tumor cells possess a high metabolic plasticity, which drives them to switch on the anaerobic glycolysis and lactate production when challenged by hypoxia. Among the enzymes mediating this plasticity through bidirectional conversion of pyruvate and lactate, the lactate dehydrogenase A (LDHA) and lactate dehydrogenase B (LDHB), are indicated. LDHA has a higher affinity for pyruvate, preferentially converting pyruvate to lactate, and NADH to NAD+ in anaerobic conditions, whereas LDHB possess a higher affinity for lactate, preferentially converting lactate to pyruvate, and NAD+ to NADH, when oxygen is abundant. Apart from the undisputed role of LDHA and LDHB in tumor cell metabolism and adaptation to unfavorable environmental or cellular conditions, these enzymes participate in the regulation of cell death. This review presents the latest progress made in this area on the roles of LDHA and LDHB in apoptosis and autophagy of tumor cells. Several examples of how LDHA and LDHB impact on these processes, as well as possible molecular mechanisms, will be discussed in this article. The information included in this review points to the legitimacy of modulating LDHA and/or LDHB to target tumor cells in the context of human and veterinary medicine.
Collapse
Affiliation(s)
- Kaja Urbańska
- Department of Morphological Sciences, Warsaw University of Life Sciences-SGGW, Nowoursynowska 159, 02-776 Warsaw, Poland.
| | - Arkadiusz Orzechowski
- Department of Physiological Sciences, Warsaw University of Life Sciences-SGGW, Nowoursynowska 159, 02-776 Warsaw, Poland.
| |
Collapse
|
25
|
Zhai S, Zhao L, Lin T, Wang W. Downregulation of miR-33b promotes non-small cell lung cancer cell growth through reprogramming glucose metabolism miR-33b regulates non-small cell lung cancer cell growth. J Cell Biochem 2019; 120:6651-6660. [PMID: 30368888 PMCID: PMC6587718 DOI: 10.1002/jcb.27961] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 10/04/2018] [Indexed: 12/16/2022]
Abstract
Glucose metabolism is a common target for cancer regulation and microRNAs (miRNAs) are important regulators of this process. Here we aim to investigate a tumor-suppressing miRNA, miR-33b, in regulating the glucose metabolism of non-small cell lung cancer (NSCLC). In our study, quantitative real-time polymerase chain reaction (qRT-PCR) showed that miR-33b was downregulated in NSCLC tissues and cell lines, which was correlated with increased cell proliferation and colony formation. Overexpression of miR-33b through miR-33b mimics transfection suppressed NSCLC proliferation, colony formation, and induced cell-cycle arrest and apoptosis. Meanwhile, miR-33b overexpression inhibited glucose metabolism in NSCLC cells. Luciferase reporter assay confirmed that miR-33b directly binds to the 3'-untranslated region of lactate dehydrogenase A (LDHA). qRT-PCR and Western blot analysis showed that miR-33b downregulated the expression of LDHA. Moreover, introducing LDHA mRNA into cells over-expressing miR-33b attenuated the inhibitory effect of miR-33b on the growth and glucose metabolism in NSCLC cells. Taken together, these results confirm that miR-33b is an anti-oncogenic miRNA, which inhibits NSCLC cell growth by targeting LDHA through reprogramming glucose metabolism.
Collapse
Affiliation(s)
- Shengping Zhai
- Department of RespiratoryYantai Yuhuangding Hospital Affiliated to Qingdao UniversityYantaiShandongChina
| | - Lingyan Zhao
- Department of RespiratoryYantai Yuhuangding Hospital Affiliated to Qingdao UniversityYantaiShandongChina
| | - Tiantian Lin
- Department of RespiratoryYantai Yuhuangding Hospital Affiliated to Qingdao UniversityYantaiShandongChina
| | - Wei Wang
- Department of Thoracic SurgeryYantai Yuhuangding Hospital Affiliated to Qingdao UniversityYantaiShandongChina
| |
Collapse
|
26
|
Pfeifhofer-Obermair C, Tymoszuk P, Petzer V, Weiss G, Nairz M. Iron in the Tumor Microenvironment-Connecting the Dots. Front Oncol 2018; 8:549. [PMID: 30534534 PMCID: PMC6275298 DOI: 10.3389/fonc.2018.00549] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 11/06/2018] [Indexed: 12/18/2022] Open
Abstract
Iron metabolism and tumor biology are intimately linked. Iron facilitates the production of oxygen radicals, which may either result in iron-induced cell death, ferroptosis, or contribute to mutagenicity and malignant transformation. Once transformed, malignant cells require high amounts of iron for proliferation. In addition, iron has multiple regulatory effects on the immune system, thus affecting tumor surveillance by immune cells. For these reasons, inconsiderate iron supplementation in cancer patients has the potential of worsening disease course and outcome. On the other hand, chronic immune activation in the setting of malignancy alters systemic iron homeostasis and directs iron fluxes into myeloid cells. While this response aims at withdrawing iron from tumor cells, it may impair the effector functions of tumor-associated macrophages and will result in iron-restricted erythropoiesis and the development of anemia, subsequently. This review summarizes our current knowledge of the interconnections of iron homeostasis with cancer biology, discusses current clinical controversies in the treatment of anemia of cancer and focuses on the potential roles of iron in the solid tumor microenvironment, also speculating on yet unknown molecular mechanisms.
Collapse
Affiliation(s)
- Christa Pfeifhofer-Obermair
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Innsbruck, Austria
| | - Piotr Tymoszuk
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Innsbruck, Austria
| | - Verena Petzer
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Innsbruck, Austria
| | - Günter Weiss
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Innsbruck, Austria.,Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Innsbruck, Austria
| | - Manfred Nairz
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|