1
|
Nair RS, Patel MN, Kannan T, Gour S, Hariharan MM, Prasanna V, Thirumalai A, Chockalingam R, Vasantharekha R, ThyagaRajan S, Priyanka HP. Effects of 17β-estradiol and estrogen receptor subtype-specific agonists on Jurkat E6.1 T-cell leukemia cells. Toxicol In Vitro 2025; 106:106057. [PMID: 40112934 DOI: 10.1016/j.tiv.2025.106057] [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: 12/16/2024] [Revised: 02/23/2025] [Accepted: 03/17/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Estrogen signaling plays a crucial role in immune regulation and cancer metabolism, yet its impact on T-cell leukemia remains unclear. In hematological malignancies, estrogen receptor (ER) activation may influence metabolic shifts that affect cell survival and proliferation. This study investigates the in vitro effects of 17β-estradiol and estrogen receptor subtype-specific agonists on Jurkat E6.1 T-cell leukemia cells. PURPOSE To assess how estrogen signaling influences metabolic reprogramming, inflammatory response, and survival pathways in Jurkat E6.1 cells through receptor-dependent and independent mechanisms. METHODS Jurkat E6.1 cells incubated with different concentrations of 17β-estradiol (10-12 M, 10-10 M, 10-8 M) or ER-α agonist 4,4',4″-(4-Propyl-[1H]-pyrazole-1,3,5-triyl)trisphenol (10-10 M, 10-8 M, 10-6 M) or ER-β agonist diarylproprionitrile (10-10 M, 10-8 M, 10-6 M) with and without non-specific antagonist ICI 182,780 (10-6 M). The metabolic enzyme activities of hexokinase, pyruvate kinase, and citrate synthase were measured in cell pellets, while supernatants were analyzed for IL-6 and nitric oxide (NO) production. Additionally, PI3K/Akt pathway activation was assessed by measuring p-Akt/Total Akt expression. RESULTS A shift from glycolysis to oxidative phosphorylation was observed on treatment with 17β-estradiol with significant decline in hexokinase activity and a concomitant increase in activities of pyruvate kinase and citrate synthase. CONCLUSION 17β-estradiol mediates its effects on Jurkat E6.1 cells in vitro through receptor-subtype dependent and independent mechanisms involving metabolic enzymes (hexokinase, pyruvate kinase, citrate synthase), cytokines (IL-6), nitric oxide, and signaling molecules (p-Akt).
Collapse
Affiliation(s)
- Rahul S Nair
- Inspire Laboratory, Institute of Advanced Research in Health Sciences, Tamil Nadu Government Multi Super Speciality Hospital, Chennai, Tamil Nadu, India; Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Mantavya N Patel
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Thangamani Kannan
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Shaili Gour
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Murali M Hariharan
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Vijayarengamani Prasanna
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Anupriya Thirumalai
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Ramanathan Chockalingam
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Ramasamy Vasantharekha
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Srinivasan ThyagaRajan
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Hannah P Priyanka
- Inspire Laboratory, Institute of Advanced Research in Health Sciences, Tamil Nadu Government Multi Super Speciality Hospital, Chennai, Tamil Nadu, India; Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India.
| |
Collapse
|
2
|
Li J, Li Y, Fu L, Chen H, Du F, Wang Z, Zhang Y, Huang Y, Miao J, Xiao Y. Targeting ncRNAs to overcome metabolic reprogramming‑mediated drug resistance in cancer (Review). Int J Oncol 2025; 66:35. [PMID: 40116120 PMCID: PMC12002672 DOI: 10.3892/ijo.2025.5741] [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: 01/22/2025] [Accepted: 03/07/2025] [Indexed: 03/23/2025] Open
Abstract
The emergence of resistance to antitumor drugs in cancer cells presents a notable obstacle in cancer therapy. Metabolic reprogramming is characterized by enhanced glycolysis, disrupted lipid metabolism, glutamine dependence and mitochondrial dysfunction. In addition to promoting tumor growth and metastasis, metabolic reprogramming mediates drug resistance through diverse molecular mechanisms, offering novel opportunities for therapeutic intervention. Non‑coding RNAs (ncRNAs), a diverse class of RNA molecules that lack protein‑coding function, represent a notable fraction of the human genome. Due to their distinct expression profiles and multifaceted roles in various cancers, ncRNAs have relevance in cancer pathophysiology. ncRNAs orchestrate metabolic abnormalities associated with drug resistance in cancer cells. The present review provides a comprehensive analysis of the mechanisms by which metabolic reprogramming drives drug resistance, with an emphasis on the regulatory roles of ncRNAs in glycolysis, lipid metabolism, mitochondrial dysfunction and glutamine metabolism. Furthermore, the present review aimed to discuss the potential of ncRNAs as biomarkers for predicting chemotherapy responses, as well as emerging strategies to target ncRNAs that modulate metabolism, particularly in the context of combination therapy with anti‑cancer drugs.
Collapse
Affiliation(s)
- Junxin Li
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Yanyu Li
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Lin Fu
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Huiling Chen
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Fei Du
- Department of Pharmacy, The Fourth Affiliated Hospital of Southwest Medical University, Meishan, Sichuan 64200, P.R. China
| | - Zhongshu Wang
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Yan Zhang
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Yu Huang
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Jidong Miao
- Department of Oncology, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Yi Xiao
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| |
Collapse
|
3
|
Zhang J, Lin F, Xu Y, Sun J, Zhang L, Chen W. Lactylation and Ischemic Stroke: Research Progress and Potential Relationship. Mol Neurobiol 2025; 62:5359-5376. [PMID: 39541071 DOI: 10.1007/s12035-024-04624-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024]
Abstract
Ischemic stroke is caused by interrupted cerebral blood flow and is a leading cause of mortality and disability worldwide. Significant advancements have been achieved in comprehending the pathophysiology of stroke and the fundamental mechanisms responsible for ischemic damage. Lactylation, as a newly discovered post-translational modification, has been reported to participate in several physiological and pathological processes. However, research on lactylation and ischemic stroke is scarce. This review summarized the current function of protein lactylation in other diseases or normal physiological processes and explored their potential link with the pathophysiological process and the reparative mechanism of ischemic stroke. We proposed that neuroinflammation, regulation of metabolism, regulation of messenger RNA translation, angiogenesis, and neurogenesis might be the bridge linking lactylation and ischemic stroke. Our study provided a novel perspective for comprehending the role of protein lactylation in the pathophysiological processes underlying ischemic stroke. Lactylation might be a promising target in drug development of ischemic stroke.
Collapse
Affiliation(s)
- Jingyuan Zhang
- Department of Cerebrovascular Disease, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong Province, China
| | - Feng Lin
- Department of Cerebrovascular Disease, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong Province, China
| | - Yue Xu
- Department of Cerebrovascular Disease, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong Province, China
| | - Jiaxin Sun
- Department of Cerebrovascular Disease, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong Province, China
| | - Lei Zhang
- Department of Cerebrovascular Disease, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong Province, China.
| | - Wenli Chen
- Department of Pharmacy, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong Province, China.
| |
Collapse
|
4
|
Li YE, Liu S, Wang L, Du Y, Wu L, Chen H, Zhu T, Lin J, Xiong S, Wang Y, Zheng Q, Zou R, Lin L, Li Z, Wang L, Ge J, Ren J, Zhang Y. March2 Alleviates Aortic Aneurysm/Dissection by Regulating PKM2 Polymerization. Circ Res 2025; 136:e73-e93. [PMID: 40079144 DOI: 10.1161/circresaha.124.325049] [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: 06/09/2024] [Revised: 02/19/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND Aortic aneurysm/dissection (AAD) is a life-threatening disease lacking effective pharmacological treatment. Protein ubiquitination plays a pivotal role in cardiovascular diseases. However, the possible contribution of the E3 ubiquitin ligase March2 (membrane-associated RING [really interesting new gene] finger protein 2) to the cause of AAD remains elusive. METHODS Integrated single-cell RNA sequencing analysis was conducted in human AAD tissues. Based on the screening results, we generated a mouse line of smooth muscle cell-specific March2 knockout. β-Aminopropionitrile monofumarate was used to establish AAD. Cleavage under targets and tagmentation and cleavage under targets and tagmentation-quantitative polymerase chain reaction were performed to identify possible target genes for histone H3K18 lactylation. RESULTS March2 expression was downregulated in aorta from patients with AAD or β-aminopropionitrile monofumarate-induced AAD mice. β-Aminopropionitrile monofumarate-induced AAD was significantly accentuated in March2 global (March2-/-) and vascular smooth muscle cell-specific deletion (March2fl/fl; TaglnCre) mice, whereas the AAD pathology was rescued by rAAV9-SM22α (smooth muscle 22α)-March2 (recombinant adeno-associated virus serotype 9 expressing Flag-tagged March2 under SM22α promoter). March2 interacted with PKM2 (pyruvate kinase M2) to promote K33-linked polyubiquitination. Deficiency of March2 lessened PKM2 dimer-to-tetramer conversion in AAD and overtly exacerbated AAD-induced histone H3K18 lactylation in vascular smooth muscle cells by fostering glucose metabolism reprogramming, thereby promoting p53-driven apoptotic transcriptional response-a hallmark of AAD pathogenesis. TEPP-46 (tetraethyl pyrophosphate), a PKM2-specific activator, pronouncedly alleviated March2 deficiency-deteriorated AAD pathology. CONCLUSIONS Our findings demonstrated that March2 is a novel endogenous defender that prevents AAD by inhibiting vascular smooth muscle cell apoptosis, suggesting that March2 represents a potential therapeutic target for AAD.
Collapse
Affiliation(s)
- Yiran E Li
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| | - Shuolin Liu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- Department of Vascular Surgery (Z.L., Lixin Wang), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| | - Litao Wang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| | - Yuxin Du
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| | - Lin Wu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| | - Haoran Chen
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| | - Tingfang Zhu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| | - Jie Lin
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| | - Shengjun Xiong
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| | - Yayu Wang
- Department of Cardiovascular Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Guangdong, China (Y.W., Q.Z.)
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, Guangdong (Y.W.)
| | - Qijun Zheng
- Department of Cardiovascular Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Guangdong, China (Y.W., Q.Z.)
| | - Rongjun Zou
- Department of Cardiovascular Surgery, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine (R.Z.)
| | - Ling Lin
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| | - Zheyun Li
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- Department of Vascular Surgery (Z.L., Lixin Wang), Zhongshan Hospital, Fudan University, China
- Vascular Surgery Institute of Fudan University, Shanghai, China (Z.L., Lixin Wang)
| | - Lixin Wang
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- Department of Vascular Surgery (Z.L., Lixin Wang), Zhongshan Hospital, Fudan University, China
- Vascular Surgery Institute of Fudan University, Shanghai, China (Z.L., Lixin Wang)
| | - Junbo Ge
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| | - Jun Ren
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| | - Yingmei Zhang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| |
Collapse
|
5
|
Xiong Y, Du Y, Lin F, Fu B, Guo D, Sha Z, Tian R, Yao R, Wang L, Cong Z, Li B, Lin X, Wu H. SENP1-SIRT3 axis mediates glycolytic reprogramming to suppress inflammation during Listeria monocytogenes infection. mBio 2025; 16:e0252424. [PMID: 40071948 PMCID: PMC11980586 DOI: 10.1128/mbio.02524-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 02/12/2025] [Indexed: 04/10/2025] Open
Abstract
Listeria monocytogenes, a foodborne pathogen, has the ability to invade intestinal mucosal cells, undergo intracellular proliferation, activate host immune responses, and induce diseases such as colitis. We have demonstrated that sentrin-specific protease 1 (SENP1) functions as a protective gene in the host, suppressing the inflammatory response triggered by Listeria monocytogenes. The host's SENP1-SIRT3 axis plays a critical role in regulating inflammation during Listeria monocytogenes infection. Our findings reveal that overexpression of SENP1, particularly under Listeria monocytogenes infection conditions (MOI = 20), effectively suppresses inflammation through modulation of glycolysis. Mechanistically, during Listeria monocytogenes infection, SENP1 accumulates in the mitochondria, facilitating the de-SUMOylation and activation of sirtuin 3 (SIRT3). Activated SIRT3 then regulates the deacetylation of pyruvate kinase M2 (PKM2), leading to a decrease in glycolytic intermediates, downregulation of glycolysis-related gene expression, and suppression of inflammation. Taken together, our study provides a deeper understanding of the mechanistic role of the SENP1-SIRT3 axis in the regulation of inflammation, offering novel insights, and strategies for the treatment and prevention of inflammatory diseases. IMPORTANCE Sentrin-specific protease 1 (SENP1)-sirtuin 3 (SIRT3) has never been reported in the regulation of bacteria-induced inflammation. Our study demonstrated that SENP1 acted as a protective factor against Listeria-induced inflammation by promoting SIRT3 activation and subsequent metabolic reprogramming. The SENP1-SIRT3 axis served not only as an essential signaling pathway for regulating mitochondrial metabolic responses to metabolic stress but also responds to bacterial invasion and plays a protective role in the organism. Our findings provide a basis for further research into targeting the SENP1-SIRT3 signaling pathway for the treatment of bacterial infections.
Collapse
Affiliation(s)
- Yan Xiong
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Yongliang Du
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Feng Lin
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Beibei Fu
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Dong Guo
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Zhou Sha
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Rong Tian
- Department of Pathology, Chongqing Hygeia Hospital, Chongqing, China
| | - Rui Yao
- Department of Pathology, Chongqing Hygeia Hospital, Chongqing, China
| | - Lulu Wang
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Zixuan Cong
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Bohao Li
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Xiaoyuan Lin
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, China
| | - Haibo Wu
- School of Life Sciences, Chongqing University, Chongqing, China
| |
Collapse
|
6
|
Delobelle Q, Inizan TJ, Adjoua O, Lagardère L, Célerse F, Maréchal V, Piquemal J. High-Resolution Molecular-Dynamics Simulations of the Pyruvate Kinase Muscle Isoform 1 and 2 (PKM1/2). Chemistry 2025; 31:e202402534. [PMID: 39614705 PMCID: PMC11973853 DOI: 10.1002/chem.202402534] [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: 07/03/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/01/2024]
Abstract
Glucose metabolism plays a pivotal role in physiological processes and cancer growth. The final stage of glycolysis, converting phosphoenolpyruvate (PEP) into pyruvate, is catalyzed by the pyruvate kinase (PK) enzyme. Whereas PKM1 is mainly expressed in cells with high energy requirements, PKM2 is preferentially expressed in proliferating cells, including tumor cells. Structural analysis of PKM1 and PKM2 is essential to design new molecules with antitumoral activity. To understand their structural dynamics, we performed extensive high-resolution molecular dynamics (MD) simulations using adaptive sampling techniques coupled to the polarizable AMOEBA force field. Performing more than 6 μs of simulation, we considered all oligomerization states of PKM2 and propose structural insights for PKM1 to further study the PKM2-specific allostery. We focused on key sites including the active site and the natural substrate Fructose Bi-Phosphate (FBP) fixation pocket. Additionally, we present the first MD simulation of biologically active PKM1 and uncover important similarities with its PKM2 counterpart bound to FBP. We also analysed TEPP-46's fixation, a pharmacological activator binding a different pocket, on PKM2 and highlighted the structural differences and similarities compared to PKM2 bound to FBP. Finally, we determined potential new cryptic pockets specific to PKM2 for drug targeting.
Collapse
Affiliation(s)
- Quentin Delobelle
- Centre de Recherche Saint-Antoine – Team “Biologie et Thérapeutique du Cancer”, UMRS 938 INSERMParisFrance
- Sorbonne Université, CNRS, Laboratoire de Chimie Théorique, UMR 761675005ParisFrance
| | - Théo Jaffrelot Inizan
- Sorbonne Université, CNRS, Laboratoire de Chimie Théorique, UMR 761675005ParisFrance
- University of California BerkeleyBakar Institute of Digital Materials for the PlanetCollege of Computing, Data Science, and SocietyBerkeley94720USA
| | - Olivier Adjoua
- Sorbonne Université, CNRS, Laboratoire de Chimie Théorique, UMR 761675005ParisFrance
| | - Louis Lagardère
- Sorbonne Université, CNRS, Laboratoire de Chimie Théorique, UMR 761675005ParisFrance
| | - Frédéric Célerse
- Sorbonne Université, CNRS, Laboratoire de Chimie Théorique, UMR 761675005ParisFrance
- Sorbonne Université, CNRS, IPCM75005ParisFrance
| | - Vincent Maréchal
- Centre de Recherche Saint-Antoine – Team “Biologie et Thérapeutique du Cancer”, UMRS 938 INSERMParisFrance
| | - Jean‐Philip Piquemal
- Sorbonne Université, CNRS, Laboratoire de Chimie Théorique, UMR 761675005ParisFrance
| |
Collapse
|
7
|
Luo YY, Guan YP, Zhan HF, Sun CY, Cai LY, Tao KG, Lin Y, Zeng X. Circ_0098181 binds PKM2 to attenuate liver fibrosis. Front Pharmacol 2025; 16:1517250. [PMID: 40248098 PMCID: PMC12003362 DOI: 10.3389/fphar.2025.1517250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/14/2025] [Indexed: 04/19/2025] Open
Abstract
Background Liver cirrhosis seriously harms human health and fibrosis is the essential pathological process of cirrhosis. Recently, circular RNAs (circRNAs) were found to play critical roles in liver fibrosis, but the key circRNAs and precise mechanisms remained unclear. This study aimed to investigate the effect of circ_0098181 in fibrogenesis and explore its mechanism. Methods RNA sequencing was conducted to identify circRNA signatures in human liver cirrhotic tissues. Hepatic stellate cells (HSCs) (including primary rat HSCs, LX2, HSC-T6) and carbon tetrachloride (CCl4) induced liver cirrhosis model were used to explore the role of circ_0098181 on HSC activation and liver fibrogenesis in vitro and in vivo. RNA sequencing, RNA pull-down, mass spectrometry, and RNA immunoprecipitation (RIP) experiments were performed to elucidate the mechanism. Results Circ_0098181 was obviously reduced in human fibrotic liver tissues and activated HSCs. Exogenous administration of circ_0098181 blocked the activation, proliferation, and migration of HSCs in vitro and mitigated the progression of CCl4-induced liver fibrosis in vivo. Mechanistically, adenosine deaminase acting on RNA1 (ADAR1) combined with the intronic complementary sequences (ICSs) in the flanking regions, thereby regulating the biogenesis of circ_0098181. RNA sequencing and qRT-PCR revealed the suppression of circ_0098181 on pro-inflammation cytokines expression (TNFα, Fas, Cxcl11, etc.). RNA pull-down, mass spectrometry, and RIP experiments indicated that pyruvate kinase M2 (PKM2) was the direct target of circ_0098181. Circ_0098181 bound to PKM2, restrained its nuclear translocation and phosphorylation. Conclusion In conclusion, circ_0098181 exerts a significant anti-fibrotic effect by binding PKM2 to repress its nuclear translocation and inhibiting hepatic inflammation, suggesting the promising therapeutic merit in liver cirrhosis.
Collapse
Affiliation(s)
- Yuan-Yuan Luo
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ya-Ping Guan
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hong-Fei Zhan
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chun-Yan Sun
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ling-Yan Cai
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ke-Gong Tao
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yong Lin
- Department of Gastroenterology, Shanghai Changzheng Hospital, Navy Military Medical University, Shanghai, China
| | - Xin Zeng
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
8
|
Mortazavi Farsani SS, Soni J, Jin L, Yadav AK, Bansal S, Mi T, Hilakivi-Clarke L, Clarke R, Youngblood B, Cheema A, Verma V. Pyruvate kinase M2 activation reprograms mitochondria in CD8 T cells, enhancing effector functions and efficacy of anti-PD1 therapy. Cell Metab 2025:S1550-4131(25)00106-8. [PMID: 40199327 DOI: 10.1016/j.cmet.2025.03.003] [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: 11/06/2023] [Revised: 11/27/2024] [Accepted: 03/06/2025] [Indexed: 04/10/2025]
Abstract
Mitochondria regulate T cell functions and response to immunotherapy. We show that pyruvate kinase M2 (PKM2) activation enhances mitochondria-dependent effector functions in CD8 and chimeric antigen receptor (CAR)-T cells. Multi-omics and 13C-glucose tracer studies showed that PKM2 agonism alters one-carbon metabolism, decreasing methionine levels, resulting in hypomethylated nuclear and mitochondrial DNA and enhancing mitochondrial biogenesis and functions. PKM2 activation increased the recall responses and anti-tumor functions of CD8 T cells, enhancing adoptive cell therapy. In preclinical models, the PKM2 agonist induced CD8 T cell-dependent anti-tumor responses that synergized with anti-programmed death 1 (PD1) therapy. Immunologically, PKM2 agonists boosted the activation of effector T cells while reducing FoxP3+ T regulatory (Treg) cells in the tumors. The anti-PD1 combination enhanced the frequency of tumor-specific activated CD8 T cells. Together, PKM2 agonism increased mitochondrial functions supporting cell cytotoxicity. Hence, pharmacological targeting of PKM2 can be a clinically viable strategy for enhancement of adoptive cell therapy, in situ anti-tumor immune responses, and immune checkpoint blockade therapy. VIDEO ABSTRACT.
Collapse
Affiliation(s)
| | - Jignesh Soni
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Lu Jin
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Anil Kumar Yadav
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Shivani Bansal
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
| | - Tian Mi
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | | - Robert Clarke
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Benjamin Youngblood
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Amrita Cheema
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
| | - Vivek Verma
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA; Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
9
|
Ni X, Wei Z, Peng Y, Zheng L, Shang J, Liu F, Li Y, Liu J. Triclosan exposure induces liver fibrosis in mice: The heterogeneous nuclear ribonucleoprotein A1/pyruvate kinase M2 axis drives hepatic stellate cell activation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 294:118113. [PMID: 40157328 DOI: 10.1016/j.ecoenv.2025.118113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 03/23/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Triclosan (TCS) is an effective broad-spectrum antibacterial agent. TCS possesses a stable structure, can easily accumulate in the environment, and may have numerous negative impacts on human health. One organ particularly susceptible to TCS damage is the liver; however, the molecular mechanisms underlying TCS-induced liver damage remain unclear. A long-term TCS exposure model was established in C57BL/6 mice through maternal administration from gestation to postnatal 8-week-old. The offspring were randomly assigned to three groups (0, 50, and 100 mg/kg TCS) with six animals per group, ensuring an equal gender distribution (3 males and 3 females). The results showed that TCS-exposed mice exhibited serum aspartate aminotransferase, alanine aminotransferase, and alkaline phosphatase enzyme activities increased by 1.5-2 times when compared with vehicle-treated mice, along with features of liver fibrosis. In the LX-2 cell line, used as an in vitro model, TCS promoted proliferation and migration and induced the activation of hepatic stellate cells (HSCs). The level of pyruvate kinase M2 (PKM2) dimer increased by 200 % in LX-2 cells treated with TCS. PKM2 dimer overexpression stimulated HSC activation, whereas treatment with TEPP-46 (a PKM2 dimer inhibitor) significantly decreased the activation process. The expression of heterogeneous ribonucleoprotein particle A1 (hnRNPA1) was upregulated in the TCS treatment group and promoted the PKM2 expression. Moreover, disruption of the hnRNPA1/PKM2 axis reduced HSC proliferation and migration activated by TCS. Overall, our findings highlighted that TCS could cause liver fibrosis by stimulating the proliferation and migration of HSCs activated via the hnRNPA1/PKM2 axis, providing promising treatment options for TCS-related liver damage.
Collapse
Affiliation(s)
- Xiao Ni
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Ziyun Wei
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Yuxuan Peng
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Linlin Zheng
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Jianing Shang
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Fu Liu
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Yunwei Li
- Department of Anorectal Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, PR China.
| | - Jieyu Liu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, PR China; Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China.
| |
Collapse
|
10
|
Oska N, Awad AM, Eltanani S, Shawky M, Naghdi A, Yumnamcha T, Singh LP, Ibrahim AS. Glyceraldehyde-3-Phosphate Dehydrogenase/1,3-Bisphosphoglycerate-NADH as Key Determinants in Controlling Human Retinal Endothelial Cellular Functions: Insights from Glycolytic Screening. J Biol Chem 2025:108472. [PMID: 40158853 DOI: 10.1016/j.jbc.2025.108472] [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: 10/26/2024] [Revised: 03/11/2025] [Accepted: 03/26/2025] [Indexed: 04/02/2025] Open
Abstract
Maintaining barrier integrity, along with cell adhesion to the extracellular matrix and the subsequent process of cell spreading, are essential functions of endothelial cells, including human retinal endothelial cells (HRECs). Disruptions in these processes can lead to vision-threatening conditions like diabetic retinopathy. However, the bioenergetic mechanisms that regulate HREC barrier function and cell spreading remain incompletely understood. This study investigates the role of lower glycolytic components in modulating these critical functions of HRECs. In vitro, Electric Cell-Substrate Impedance Sensing (ECIS) technology was used to measure real-time changes in HREC barrier integrity (electrical resistance) and cell spreading (capacitance). Pharmacological inhibitors targeting lower glycolytic components were tested: heptelidic acid for glyceraldehyde-3-phosphate dehydrogenase (GAPDH), NG-52 for phosphoglycerate kinase (PGK), shikonin for pyruvate kinase M (PKM), galloflavin for lactate dehydrogenase (LDH), AZD3965 for lactate transporter (MCT-1), and MSDC-0160 for the mitochondrial pyruvate carrier (MPC). GAPDH knockdown was performed using siRNA, and cell viability was assessed via lactate dehydrogenase (LDH) release assays. For in vivo studies, wild-type C57BL/6J mice received intravitreal injections of heptelidic acid, while control mice received vehicle (DMSO). Retinal vascular permeability was assessed by fluorescein angiography (FA) and retinal albumin leakage. The most significant decrease in electrical resistance and increase in capacitance of HRECs were observed following the dose-dependent inhibition of GAPDH and the resulting reduction in 1,3-bisphosphoglycerate (1,3-BPG) and NADH by heptelidic acid. LDH level analysis at 24-48 hours post-treatment with heptelidic acid (1 and 10 μM) showed no significant difference compared to controls, indicating that the observed disruption of HREC functionality was not due to cell death. Supporting these findings, inhibition of downstream glycolytic steps that result in the accumulation of 1,3-BPG and NADH, such as treatment with NG-52 for PGK or shikonin for PKM, led to a significant increase in electrical resistance and a decrease in cell capacitance. Furthermore, GAPDH knockdown via siRNA also led to a significant decrease in cellular resistance in HRECs. In vivo, FA imaging demonstrated that intravitreal injection of heptelidic acid led to significant retinal vascular leakage, further supported by increased albumin extravasation in treated eyes. Conversely, pharmacological inhibition of other lower glycolytic components, including LDH, MCT, and MPC, did not significantly alter HREC barrier function or spreading behavior. This study highlights the distinct roles of lower glycolytic components in regulating HREC functionality. GAPDH and its downstream products (1,3-BPG and NADH) are shown to play a pivotal role in maintaining barrier integrity and promoting HREC adhesion and spreading. These findings guide the development of targeted interventions that modulate HREC bioenergetics to treat endothelial dysfunction in various retinal disorders, while minimizing potential adverse effects on healthy endothelial cells.
Collapse
Affiliation(s)
- Nicole Oska
- Department of Ophthalmology, Visual, and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Ahmed M Awad
- Department of Ophthalmology, Visual, and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, MI, USA; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura National University, Gamasa, Egypt
| | - Shaimaa Eltanani
- Department of Ophthalmology, Visual, and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Mohamed Shawky
- Department of Ophthalmology, Visual, and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, MI, USA; Department of Biochemistry, Faculty of Pharmacy, Horus University, Egypt
| | - Armaan Naghdi
- Department of Ophthalmology, Visual, and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Thangal Yumnamcha
- Department of Ophthalmology, Visual, and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Lalit Pukhrambam Singh
- Department of Ophthalmology, Visual, and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Ahmed S Ibrahim
- Department of Ophthalmology, Visual, and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, MI, USA; Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt; Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI, USA; Molecular Therapeutics Research Program, Karmanos Cancer Institute (KCI), School of Medicine, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
11
|
Cui H, Xie L, Lu H, Cheng C, Xue F, Wu Z, Liu L, Qiao L, Zhang C, Zhang W, Yang J. Macrophage junctional adhesion molecule-like (JAML) protein promotes NLRP3 inflammasome activation in the development of atherosclerosis. Cell Death Differ 2025:10.1038/s41418-025-01489-5. [PMID: 40148467 DOI: 10.1038/s41418-025-01489-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 03/12/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025] Open
Abstract
Inflammation plays a crucial role in the progression of atherosclerosis. Junctional adhesion molecule-like protein (JAML), a type-I transmembrane glycoprotein, activates downstream signaling pathways. However, the precise role of macrophage-derived JAML in inflammation and atherosclerosis remains unclear. This study aimed to generate mice with macrophage-specific deletion or overexpression of JAML, with the focus of assessing its impact on macrophage function and elucidating its regulatory mechanism in atherosclerosis. High-throughput data screening was employed to investigate JAML expression in atherosclerosis, and macrophage-specific JAML-knockout and transgenic mice models were utilized to examine the effects of JAML on atherosclerosis. Furthermore, the role of JAML was assessed using Oil Red O staining, RNA-sequencing analysis, and co-immunoprecipitation techniques. Increased JAML expression was observed in macrophages from both mice and patients with atherosclerosis. Macrophage-specific JAML deletion attenuated atherosclerosis and inflammation, whereas macrophage-specific JAML overexpression exacerbated these conditions. Mechanistically, JAML deletion inhibited inflammation by decreasing nuclear translocation of pyruvate kinase M2 (PKM2) and PKM2/p65 complex formation, which consequently suppressed the nuclear factor kappa B (NF-κB) pathway and NLRP3 inflammasome activation. Taken together, these findings demonstrate that macrophage-expressed JAML facilitates the progression of atherosclerosis by activating the NF-κB pathway and NLRP3 inflammasome through nuclear migration and phosphorylation of PKM2. Notably, our study revealed a novel mechanism for the regulation of NLRP3 inflammasome activation in atherosclerosis. Therefore, targeting JAML may be an effective treatment strategy for atherosclerosis, a condition characterized by chronic inflammation.
Collapse
Affiliation(s)
- Huiliang Cui
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Lin Xie
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Hanlin Lu
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Cheng Cheng
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Fei Xue
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Zhenguo Wu
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Li Liu
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Lei Qiao
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Cheng Zhang
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
- Cardiovascular Disease Research Center of Shandong First Medical University, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Wencheng Zhang
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
| | - Jianmin Yang
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
12
|
Kim H, Takegahara N, Choi Y. The impact of IgSF11-PKM2 pathway on gene expression in osteoclasts. MICROPUBLICATION BIOLOGY 2025; 2025:10.17912/micropub.biology.001469. [PMID: 40196137 PMCID: PMC11973584 DOI: 10.17912/micropub.biology.001469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/09/2025]
Abstract
Osteoclasts are primary bone resorbing cells. Previously, we described metabolic regulation of osteoclasts through IgSF11-mediated phosphorylation of the glycolytic enzyme PKM2. Here, we report the impact of IgSF11-PKM2-mediated regulation on gene expression in osteoclasts, utilizing RNA sequencing on osteoclasts engineered to express a chimeric protein, lacking IgSF11, and pharmacologically modulating PKM2 activity. Our analysis identified osteoclast-related genes whose expression is altered by the absence of IgSF11 and by changes in PKM2 activity. This study reveals gene expression changes associated with the IgSF11-PKM2 pathway, providing new insight into its role in osteoclasts.
Collapse
Affiliation(s)
- Hyunsoo Kim
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Noriko Takegahara
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Yongwon Choi
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
13
|
Cai C, Jiang J, Li S, Gao C, Pu H, Zhao L, Xiao J. PKM2 regulates osteoclastogenesis by affecting osteoclast precursor cell fusion via downregulation of OC-STAMP and DC-STAMP. J Biol Chem 2025; 301:108439. [PMID: 40122175 DOI: 10.1016/j.jbc.2025.108439] [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: 02/22/2022] [Revised: 02/25/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025] Open
Abstract
Osteoporosis is a common bone disease that has become a serious public health problem with the aging of population. Osteoclasts are the only cells in body that can resorb bone, whose dysfunction is closely related to osteoporosis. Pyruvate kinase M2 (PKM2) is one of the essential rate-limiting enzymes in the process of glycolysis. This study aimed to elucidate the role of PKM2 in osteoclastogenesis and bone resorption. Bone marrow-derived macrophages were transfected with adenovirus to knock down the expression of PKM2 gene or treated with the PKM2 activators, DASA-58 and TEPP-46. Osteoclast formation was detected by tartrate-resistant acid phosphatase staining, osteoclast-specific gene and protein expression was detected by RT-quantitative PCR and Western blotting, and the effect of DASA-58 on osteoclast gene expression at the transcriptional level was examined by RNA sequencing. The results showed that knockdown of PKM2 by adenoviral transfection or treatment with PKM2 activators, DASA-58 and TEPP-46, inhibited osteoclast differentiation and suppressed the expression of osteoclast-associated genes in bone marrow-derived macrophages. Furthermore, PKM2 activators, DASA-58 and TEPP-46, could inhibit several signaling pathways in osteoclasts; knockdown of PKM2 or treatment with PKM2 activators, DASA-58 and TEPP-46, both affected osteoclast precursor cell fusion by inhibiting the expression of osteoclast stimulatory transmembrane protein (OC-STAMP) and dendritic cell-specific transmembrane protein (DC-STAMP). Therefore, PKM2 is closely related to osteoclast differentiation and formation, and the development of new therapeutic strategies targeting the PKM2 gene in osteoclasts may be feasible for the prevention and treatment of osteoporosis.
Collapse
Affiliation(s)
- Cong Cai
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiawei Jiang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Song Li
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chenghao Gao
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongxu Pu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Libo Zhao
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jun Xiao
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
14
|
Wu H, Zhang J, Wang Q, Li Z, Li L, Xie Y. Metformin combined with CB-839 specifically inhibits KRAS-mutant ovarian cancer. Sci Rep 2025; 15:6072. [PMID: 39972191 PMCID: PMC11840008 DOI: 10.1038/s41598-025-90963-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 02/17/2025] [Indexed: 02/21/2025] Open
Abstract
KRAS mutations can cause metabolic reprogramming in ovarian cancer, leading to an increased metastatic capacity. This study investigated the metabolic reprogramming changes induced by KRAS mutations in ovarian cancer and the mechanism of action of metformin combined with a glutaminase 1 inhibitor (CB-839). KRAS-mutant ovarian cancer accounted for 14% of ovarian cancers. The expression of glucose metabolism-related (PFKFB3, HK2, GLUT1, and PDK2) and glutamine metabolism-related enzymes (GLS1 and ASCT2) was elevated in KRAS-mutant ovarian cancer cells compared with that in wild-type cells. KRAS-mutant cells had a higher aerobic oxidative capacity than did wild-type cells. Metformin inhibited proliferation, the expression of glucose metabolism-related enzymes, and the aerobic oxidative capacity of KRAS-mutant cells compared with those of control cells. Furthermore, it enhanced the expression of glutamine metabolism-related enzymes in KRAS-mutant cells. Metformin combined with CB-839 inhibited the proliferation and aerobic oxidation of KRAS-mutant cells to a greater extent than that observed in wild-type cells. Additionally, the inhibitory effects of metformin and CB-839 in the KRAS-mutant ovarian cancer NOD-SCID mouse model were significantly stronger than those in the drug-alone group. KRAS mutations lead to enhanced glucose and glutamine metabolism in ovarian cancer cells, which was inhibited by metformin combined with CB-839.
Collapse
Affiliation(s)
- Han Wu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Jialin Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Qiujie Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Zijiao Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Linlin Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Ya Xie
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
15
|
Liu H, Wang S, Wang J, Guo X, Song Y, Fu K, Gao Z, Liu D, He W, Yang LL. Energy metabolism in health and diseases. Signal Transduct Target Ther 2025; 10:69. [PMID: 39966374 PMCID: PMC11836267 DOI: 10.1038/s41392-025-02141-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 11/08/2024] [Accepted: 12/25/2024] [Indexed: 02/20/2025] Open
Abstract
Energy metabolism is indispensable for sustaining physiological functions in living organisms and assumes a pivotal role across physiological and pathological conditions. This review provides an extensive overview of advancements in energy metabolism research, elucidating critical pathways such as glycolysis, oxidative phosphorylation, fatty acid metabolism, and amino acid metabolism, along with their intricate regulatory mechanisms. The homeostatic balance of these processes is crucial; however, in pathological states such as neurodegenerative diseases, autoimmune disorders, and cancer, extensive metabolic reprogramming occurs, resulting in impaired glucose metabolism and mitochondrial dysfunction, which accelerate disease progression. Recent investigations into key regulatory pathways, including mechanistic target of rapamycin, sirtuins, and adenosine monophosphate-activated protein kinase, have considerably deepened our understanding of metabolic dysregulation and opened new avenues for therapeutic innovation. Emerging technologies, such as fluorescent probes, nano-biomaterials, and metabolomic analyses, promise substantial improvements in diagnostic precision. This review critically examines recent advancements and ongoing challenges in metabolism research, emphasizing its potential for precision diagnostics and personalized therapeutic interventions. Future studies should prioritize unraveling the regulatory mechanisms of energy metabolism and the dynamics of intercellular energy interactions. Integrating cutting-edge gene-editing technologies and multi-omics approaches, the development of multi-target pharmaceuticals in synergy with existing therapies such as immunotherapy and dietary interventions could enhance therapeutic efficacy. Personalized metabolic analysis is indispensable for crafting tailored treatment protocols, ultimately providing more accurate medical solutions for patients. This review aims to deepen the understanding and improve the application of energy metabolism to drive innovative diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Hui Liu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuo Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianhua Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xin Guo
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yujing Song
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kun Fu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenjie Gao
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Danfeng Liu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Wei He
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Lei-Lei Yang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
16
|
Ren J, Gao J, Yao X, Wang X, Kong X, Lin T, Wang H, Ma W, Glebov OO, Wan Q. Bilateral transcranial direct-current stimulation confers neuroprotection through suppression of PKM2 after mouse cerebral ischemia injury. Brain Res 2025; 1849:149353. [PMID: 39603317 DOI: 10.1016/j.brainres.2024.149353] [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: 06/27/2024] [Revised: 11/08/2024] [Accepted: 11/23/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND In its tetrameric form, pyruvate kinase M2 isoform (PKM2) catalyzes the last step of glycolysis and plays a key role in the metabolic reprogramming via regulating the signaling of pentose phosphate pathway (PPP). But the role of PKM2 in cerebral ischemia-reperfusion (I/R) injury remains unknown. METHODS Mice model of middle cerebral artery occlusion (MCAO) and model of oxygen-glucose deprivation (OGD) injury in cultured neurons were established. PKM2 activator or inhibitor were used to test the effects of PKM2 in wild-type and PKM2 (-/-) mice after I/R injury. Biochemical and molecular approach were used to detect the level of PKM2 tetramers and PPP metabolites. RESULTS We showed for the first time that ischemia-induced increase of PKM2 activity promoted neuronal death via the suppression of PPP-dependent antioxidant capacity. To identify therapeutic approach that suppresses ischemia-induced increase of PKM2 activity, we tested the effect of bilateral transcranial direct-current stimulation (BtDCS), a newly established BtDCS approach by us, on PKM2 activity after mouse I/R. Our data demonstrated that BtDCS inhibited PKM2 activity in the ischemic neurons. BtDCS also reduced the cerebral infarct volume and the neurological deficits in stroke mice. We found that BtDCS-induced neuroprotection was mediated through the suppression of PKM2 activity after I/R. CONCLUSIONS Together, this study provided novel evidence that supported PKM2 as a crucial regulator of neuronal metabolism after cerebral I/R injury, and revealed the molecular mechanism by which BtDCS protects against mouse cerebral I/R injury through regulating PKM2-mediated metabolic reprogramming.
Collapse
Affiliation(s)
- Jinyang Ren
- Institute of Neuroregeneration & Neurorehabilitation, Department of Pathophysiology, Qingdao University, 308 Ningxia Street, Qingdao 266071, China
| | - Jingchen Gao
- Institute of Neuroregeneration & Neurorehabilitation, Department of Pathophysiology, Qingdao University, 308 Ningxia Street, Qingdao 266071, China
| | - Xujin Yao
- Institute of Neuroregeneration & Neurorehabilitation, Department of Pathophysiology, Qingdao University, 308 Ningxia Street, Qingdao 266071, China
| | - Xiyuran Wang
- Institute of Neuroregeneration & Neurorehabilitation, Department of Pathophysiology, Qingdao University, 308 Ningxia Street, Qingdao 266071, China
| | - Xiangyi Kong
- Institute of Neuroregeneration & Neurorehabilitation, Department of Pathophysiology, Qingdao University, 308 Ningxia Street, Qingdao 266071, China
| | - Tao Lin
- Institute of Neuroregeneration & Neurorehabilitation, Department of Pathophysiology, Qingdao University, 308 Ningxia Street, Qingdao 266071, China
| | - Hui Wang
- Institute of Neuroregeneration & Neurorehabilitation, Department of Pathophysiology, Qingdao University, 308 Ningxia Street, Qingdao 266071, China
| | - Wenlong Ma
- Institute of Neuroregeneration & Neurorehabilitation, Department of Pathophysiology, Qingdao University, 308 Ningxia Street, Qingdao 266071, China
| | - Oleg O Glebov
- Department of Old Age Psychiatry, The Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, Denmark Hill, London SE5 8AF, United Kingdom.
| | - Qi Wan
- Institute of Neuroregeneration & Neurorehabilitation, Department of Pathophysiology, Qingdao University, 308 Ningxia Street, Qingdao 266071, China; Qingdao Gui-Hong Intelligent Medical Technology Co. Ltd, 7 Fenglong Road, Qingdao High-tech Industrial Development District, Qingdao, China.
| |
Collapse
|
17
|
Liao F, Li Y, Zhang Z, Yu Q, Liu H. Pulsed electromagnetic fields modulate energy metabolism during wound healing process: an in vitro model study. BMC Complement Med Ther 2025; 25:50. [PMID: 39939866 PMCID: PMC11823022 DOI: 10.1186/s12906-025-04792-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 01/28/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Pulsed electromagnetic fields (PEMFs) therapy was extensively investigated to treat wound healing, which is a highly metabolically demanding process. However, the effect of PEMFs on energy metabolism in wound healing remains largely unexplored. Therefore, our study aims to demonstrate the role of PEMFs on energy metabolism in wound healing. METHODS Scratch-wound healing assay and cell viability assay were performed for the in vitro study of the effect of PEMFs on cell migration and viability. Seahorse assay was conducted for energy metabolism analysis while holo-tomographic microscopy for fine changes of L929 cells. Mitochondrial membrane potential assay and intracellular reactive oxygen species (ROS) and pH assay were performed for analyzing the changes of mitochondrial function. RESULTS PEMFs with specific parameter (4mT, 80 Hz) promoted cell migration and viability. Glycolysis stress and mitochondria stress test revealed that PEMFs-exposed L929 cells was highly glycolytic for energy generation. Besides, PEMFs enhanced intracellular acidification and maintained low level of intracellular ROS in L929 cells. Compared to control group, much more vesicles were generated and then transported to regions close to the nuclear in L929 cells treated with PEMFs. CONCLUSIONS Our major findings revealed for the first time that PEMFs induce metabolic reprogramming of fibroblast shifting from mitochondrial respiration to glycolysis, accompanied with an increase of vesicular transport, which is closely related to wound healing in vitro.
Collapse
Affiliation(s)
- Feng Liao
- Department of Orthopedics, Sichuan Provincial People's Hospital,School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, PR China
| | - Yamei Li
- Department of Rehabilitation Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, PR China
| | - Zhou Zhang
- Department of Rehabilitation Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, PR China
| | - Qian Yu
- Department of Rehabilitation Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, PR China.
| | - Huifang Liu
- Department of Rehabilitation Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, PR China.
| |
Collapse
|
18
|
Rojas-Pirela M, Andrade-Alviárez D, Rojas V, Marcos M, Salete-Granado D, Chacón-Arnaude M, Pérez-Nieto MÁ, Kemmerling U, Concepción JL, Michels PAM, Quiñones W. Exploring glycolytic enzymes in disease: potential biomarkers and therapeutic targets in neurodegeneration, cancer and parasitic infections. Open Biol 2025; 15:240239. [PMID: 39904372 PMCID: PMC11793985 DOI: 10.1098/rsob.240239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 02/06/2025] Open
Abstract
Glycolysis, present in most organisms, is evolutionarily one of the oldest metabolic pathways. It has great relevance at a physiological level because it is responsible for generating ATP in the cell through the conversion of glucose into pyruvate and reducing nicotinamide adenine dinucleotide (NADH) (that may be fed into the electron chain in the mitochondria to produce additional ATP by oxidative phosphorylation), as well as for producing intermediates that can serve as substrates for other metabolic processes. Glycolysis takes place through 10 consecutive chemical reactions, each of which is catalysed by a specific enzyme. Although energy transduction by glucose metabolism is the main function of this pathway, involvement in virulence, growth, pathogen-host interactions, immunomodulation and adaptation to environmental conditions are other functions attributed to this metabolic pathway. In humans, where glycolysis occurs mainly in the cytosol, the mislocalization of some glycolytic enzymes in various other subcellular locations, as well as alterations in their expression and regulation, has been associated with the development and progression of various diseases. In this review, we describe the role of glycolytic enzymes in the pathogenesis of diseases of clinical interest. In addition, the potential role of these enzymes as targets for drug development and their potential for use as diagnostic and prognostic markers of some pathologies are also discussed.
Collapse
Affiliation(s)
- Maura Rojas-Pirela
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca37007, Spain
- Unidad de Medicina Molecular, Departamento de Medicina, Universidad de Salamanca, Salamanca37007, Spain
- Servicio de Medicina Interna, Hospital Universitario de Salamanca, Salamanca37007, Spain
| | - Diego Andrade-Alviárez
- Laboratorio de Enzimología de Parásitos, Departamento de Biología, Facultad de Ciencias, Universidad de Los Andes, Mérida5101, Venezuela
| | - Verónica Rojas
- Instituto de Biología, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso2373223, Chile
| | - Miguel Marcos
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca37007, Spain
- Unidad de Medicina Molecular, Departamento de Medicina, Universidad de Salamanca, Salamanca37007, Spain
- Servicio de Medicina Interna, Hospital Universitario de Salamanca, Salamanca37007, Spain
| | - Daniel Salete-Granado
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca37007, Spain
- Unidad de Medicina Molecular, Departamento de Medicina, Universidad de Salamanca, Salamanca37007, Spain
| | - Marirene Chacón-Arnaude
- Laboratorio de Enzimología de Parásitos, Departamento de Biología, Facultad de Ciencias, Universidad de Los Andes, Mérida5101, Venezuela
| | - María Á. Pérez-Nieto
- Unidad de Medicina Molecular, Departamento de Medicina, Universidad de Salamanca, Salamanca37007, Spain
- Fundación Instituto de Estudios de Ciencias de la Salud de Castilla y León, Soria42002, Spain
| | - Ulrike Kemmerling
- Instituto de Ciencias Biomédicas, Universidad de Chile, Facultad de Medicina, Santiago de Chile8380453, Chile
| | - Juan Luis Concepción
- Laboratorio de Enzimología de Parásitos, Departamento de Biología, Facultad de Ciencias, Universidad de Los Andes, Mérida5101, Venezuela
| | - Paul A. M. Michels
- School of Biological Sciences, University of Edinburgh, The King’s Buildings, EdinburghEH9 3FL, UK
| | - Wilfredo Quiñones
- Laboratorio de Enzimología de Parásitos, Departamento de Biología, Facultad de Ciencias, Universidad de Los Andes, Mérida5101, Venezuela
| |
Collapse
|
19
|
Ye F, Zhang Z, Shi L, Lu S, Li X, Mu W, Jiang Q, Yan B. Targeting glycolytic reprogramming by tsRNA-0032 for treating pathological lymphangiogenesis. Cell Death Dis 2025; 16:51. [PMID: 39870617 PMCID: PMC11772812 DOI: 10.1038/s41419-025-07366-w] [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: 08/11/2024] [Revised: 12/27/2024] [Accepted: 01/16/2025] [Indexed: 01/29/2025]
Abstract
Lymphangiogenesis is vital for tissue fluid homeostasis, immune function, and lipid absorption. Abnormal lymphangiogenesis has been implicated in several diseases such as cancers, inflammatory, and autoimmune diseases. In this study, we elucidate the role of tsRNA-0032 in lymphangiogenesis and its molecular mechanism. tsRNA-0032 expression is significantly decreased in corneal suture model and human lymphatic endothelial cell (HLEC) model under inflammatory condition. Overexpression of tsRNA-0032 exerts anti-lymphangiogenic effects by inhibiting HLEC proliferation, migration, and tube formation. Moreover, overexpression of tsRNA-0032 inhibits suture-induced corneal lymphangiogenesis. tsRNA-0032 is mainly located in the cytoplasm and interacts with Ago2 protein. Overexpression of tsRNA-0032 reduces ATP production and decreases pyruvate and lactate levels by targeting PKM2, a key enzyme in glycolysis. This regulation of glycolysis alters cellular energy and metabolic balance in HLECs, contributing to anti-lymphangiogenic effects. Clinical data reveals that tsRNA-0032 levels are significantly reduced in corneal tissues of transplant recipients compared to donors, while PKM2 expression is elevated, highlighting the clinical relevance of tsRNA-0032/PKM2 axis in corneal lymphangiogenesis. This study offers new insights into the regulation of lymphangiogenesis and presents potential therapeutic targets for lymphangiogenesis-related diseases.
Collapse
Affiliation(s)
- Fan Ye
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ziran Zhang
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Lianjun Shi
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Shuting Lu
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Xiumiao Li
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Wan Mu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Qin Jiang
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China.
| | - Biao Yan
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
20
|
Liu Y, Hu P, Cheng H, Xu F, Ye Y. The impact of glycolysis on ischemic stroke: from molecular mechanisms to clinical applications. Front Neurol 2025; 16:1514394. [PMID: 39926015 PMCID: PMC11802445 DOI: 10.3389/fneur.2025.1514394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 01/10/2025] [Indexed: 02/11/2025] Open
Abstract
Ischemic stroke (IS), a leading cause of disability and mortality worldwide, remains a significant challenge due to its complex pathogenesis. Glycolysis, a central metabolic pathway, plays a critical role in bridging the gap between metabolic dysfunction and neurological impairment. During ischemic conditions, glycolysis replaces oxidative phosphorylation as the primary energy source for brain tissue. However, in the ischemia-reperfusion state, neuronal cells show a particular reliance on aerobic glycolysis. Immune cells, such as monocytes, also contribute to atheromatous plaque formation and thrombi through increased aerobic glycolysis. Given glycolysis's involvement in various pathological stages of IS, it offers the potential for improved diagnosis, treatment, and prevention. This review comprehensively explores the role of glycolysis in different phases of IS, addresses existing controversies, and discusses its diagnostic and therapeutic applications. By elucidating the intricate relationship between glycolysis and IS, this review aims to provide novel insights for future research and clinical advancements.
Collapse
Affiliation(s)
- Yingquan Liu
- The First Clinical College of Anhui University of Chinese Medicine, Hefei, China
| | - Peijia Hu
- The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Hongliang Cheng
- The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Fangyuan Xu
- The First Clinical College of Anhui University of Chinese Medicine, Hefei, China
| | - Yu Ye
- Graduate School, Anhui University of Chinese Medicine, Hefei, Anhui, China
| |
Collapse
|
21
|
Cao L, Dong X, Chen F, Li G, Fang J, Han Z, Wang J. Increased Plasma Pyruvate Kinase M2 (PK-M2) in Heart Failure: A Novel Biomarker Related to Cardiac Function and its Clinical Implications. J Am Heart Assoc 2025; 14:e036170. [PMID: 39817549 DOI: 10.1161/jaha.124.036170] [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: 04/21/2024] [Accepted: 11/15/2024] [Indexed: 01/18/2025]
Abstract
BACKGROUND The purpose of this study was to investigate whether circulating pyruvate kinase M2 (PK-M2) levels are elevated in the peripheral blood and to assess their association with diagnosis and prognosis in patients with heart failure (HF). METHODS AND RESULTS We conducted a prospective investigation involving 222 patients with HF and 103 control subjects, measuring PK-M2 concentrations using ELISA. The primary outcome, assessed over a median follow-up of 2 years (interquartile range: 776 to 926 days), was the time to the first occurrence of either rehospitalization for worsening HF or cardiovascular death. Patients with HF had higher PK-M2 levels than controls (17.4±4.1 versus 7.8±2.3 U/mL, P <0.001), and these levels correlated with HF severity (New York Heart Association cardiac function class). Patients with reduced left ventricular ejection fraction had higher PK-M2 concentrations than those with preserved ejection fraction (18.3±4.5 versus 16.7±3.6 U/mL, P <0.01). In a subset of patients with HF (n=52), PK-M2 levels significantly decreased following standardized HF treatment (mean difference, -4.3±0.5 U/mL, P <0.001). A high PK-M2 level had a 1.913-fold higher risk of the primary outcome (P=0.033) after adjusting for multiple cardiovascular risk factors, but not with cardiovascular death. Additionally, PK-M2 added incremental prognostic value beyond clinical predictors and N-terminal pro-brain natriuretic peptide (P <0.05). CONCLUSIONS Elevated PK-M2 levels are associated with primary outcomes and rehospitalization for worsening heart failure in patients with HF. These findings suggest that PK-M2 is a potential biomarker for HF diagnosis and prognosis, warranting consideration for serial patient assessment.
Collapse
Affiliation(s)
- Lu Cao
- Department of Cardiology The First Affiliated Hospital of Nanjing Medical University Nanjing China
- Department of Cardiology Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine Shanghai China
| | - Xiaoyu Dong
- Department of Cardiology The First Affiliated Hospital of Nanjing Medical University Nanjing China
| | - Fuzhong Chen
- Department of Cardiology The First Affiliated Hospital of Nanjing Medical University Nanjing China
| | - Guangjuan Li
- Department of Cardiology The First Affiliated Hospital of Nanjing Medical University Nanjing China
- Department of Cardiology The Friendship Hospital of Ili Kazak Autonomous Prefecture Yining China
| | - Jiale Fang
- Department of Cardiology The First Affiliated Hospital of Nanjing Medical University Nanjing China
| | - Zhijun Han
- Department of Clincal Research Center Jiangnan University Medical Center Wuxi Jiangsu Province China
| | - Junhong Wang
- Department of Cardiology The First Affiliated Hospital of Nanjing Medical University Nanjing China
- Department of Cardiology Liyang People's Hospital Liyang China
| |
Collapse
|
22
|
Wang H, Zhou F, Qin W, Yang Y, Li X, Liu R. Metabolic regulation of myeloid-derived suppressor cells in tumor immune microenvironment: targets and therapeutic strategies. Theranostics 2025; 15:2159-2184. [PMID: 39990210 PMCID: PMC11840731 DOI: 10.7150/thno.105276] [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: 08/21/2024] [Accepted: 12/11/2024] [Indexed: 02/25/2025] Open
Abstract
Cancer remains a major challenge to global public health, with rising incidence and high mortality rates. The tumor microenvironment (TME) is a complex system of immune cells, fibroblasts, extracellular matrix (ECM), and blood vessels that form a space conducive to cancer cell proliferation. Myeloid-derived suppressor cells (MDSCs) are abundant in tumors, and they drive immunosuppression through metabolic reprogramming in the TME. This review describes how metabolic pathways such as glucose metabolism, lipid metabolism, amino acid metabolism, and adenosine metabolism have a significant impact on the function of MDSCs by regulating their immunosuppressive activity and promoting their survival and expansion in tumors. The review also explores key metabolic targets in MDSCs and strategies to modulate MDSC metabolism to improve the tumor immune microenvironment and enhance anti-tumor immune responses. Understanding these pathways can provide insight into potential therapeutic targets for modulating MDSC activity and improving outcomes of cancer immunotherapies.
Collapse
Affiliation(s)
- Hong Wang
- School of Life Sciences, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing, 100029, China
| | - Fei Zhou
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Wenqing Qin
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing, 100029, China
| | - Yun Yang
- School of Life Sciences, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing, 100029, China
| | - Xiaojiaoyang Li
- School of Life Sciences, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing, 100029, China
| | - Runping Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing, 100029, China
| |
Collapse
|
23
|
Liao Q, Deng J, Tong J, Gan Y, Hong W, Dong H, Cao M, Xiong C, Chen Y, Xie B, Yang FY, Alifu A, Zhou GB, Huang S, Xiong J, Hao Q, Zhou X. p53 induces circFRMD4A to suppress cancer development through glycolytic reprogramming and cuproptosis. Mol Cell 2025; 85:132-149.e7. [PMID: 39637854 DOI: 10.1016/j.molcel.2024.11.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 09/15/2024] [Accepted: 11/08/2024] [Indexed: 12/07/2024]
Abstract
Cuproptosis is a type of copper-induced cell death that mainly impacts cells relying on mitochondrial metabolism. Although p53 regulates glycolytic metabolism, its role in cuproptosis remains unclear. Here, we report that the circular RNA, circFRMD4A, is crucial for p53-mediated metabolic reprogramming and cuproptosis. CircFRMD4A originates from the transcript of FRMD4A, which is transcriptionally activated by p53, and the formation of circFRMD4A is facilitated by the RNA-binding protein EWSR1. CircFRMD4A functions as a tumor suppressor and enhances the sensitivity of cancer cells to elesclomol-induced cuproptosis. Mechanistic analysis reveals that circFRMD4A interacts with and inactivates the pyruvate kinase PKM2, leading to a decrease in lactate production and a redirection of glycolytic flux toward the tricarboxylic acid cycle. Finally, p53 agonists and elesclomol coordinately suppress the growth of cancer in a xenograft mouse model. Altogether, our study uncovers that p53 promotes glycolytic reprogramming and cuproptosis via circFRMD4A and suggests a potential combination strategy against cancers with wild-type p53.
Collapse
Affiliation(s)
- Quan Liao
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China; Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang 330006, Jiangxi, China
| | - Jun Deng
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China; Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang 330006, Jiangxi, China
| | - Jing Tong
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yu Gan
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Weiwei Hong
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Hanzhi Dong
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China; Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang 330006, Jiangxi, China
| | - Mingming Cao
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Chen Xiong
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yajie Chen
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Bangxiang Xie
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Fu-Ying Yang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Aikede Alifu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guang-Biao Zhou
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shenglin Huang
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, China; Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Jianping Xiong
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China; Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang 330006, Jiangxi, China.
| | - Qian Hao
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Xiang Zhou
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, China; Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China.
| |
Collapse
|
24
|
de Andrade Borges A, Ouverney G, Arruda ATS, Ribeiro AV, Ribeiro RCB, de Souza AS, da Fonseca ACC, de Queiroz LN, de Almeida ECP, Pontes B, Rabelo VWH, Ferreira V, Abreu PA, de Carvalho da Silva F, da Silva Magalhaes Forezi L, Robbs BK. Determination of Inhibitory Effect of PKM2 Enzyme and Antitumoral Activity of Novel Coumarin-naphthoquinone Hybrids. Curr Med Chem 2025; 32:359-379. [PMID: 38877863 DOI: 10.2174/0109298673298471240605072658] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/08/2024] [Accepted: 05/06/2024] [Indexed: 02/19/2025]
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) represents the primary form of oral cancer, posing a significant global health threat. The existing chemotherapy options are accompanied by notable side effects impacting patient treatment adherence. Consequently, the exploration and development of novel substances with enhanced anticancer effects and fewer side effects have become pivotal in the realms of biological and chemical science. OBJECTIVE This work presents the pioneering examples of naphthoquinone-coumarin hybrids as a new category of highly effective cytotoxic substances targeting oral squamous cell carcinoma (OSCC). METHODS Given the significance of both naphthoquinones and coumarins as essential pharmacophores/ privileged structures in the quest for anticancer compounds, this study focused on the synthesis and evaluation of novel naphthoquinones/coumarin hybrids against oral squamous cell carcinoma. RESULTS By several in vitro, in silico, and in vivo approaches, we demonstrated that compound 6e was highly cytotoxic against OSCC cells and several other cancer cell types and was more selective than current chemotherapeutic drugs (carboplatin) and the naphthoquinone lapachol. Furthermore, compound 6e was non-hemolytic and tolerated in vivo at 50 mg/kg with an LD50 of 62.5 mg/kg. Furthermore, compound 6e did not induce apoptosis and cell cycle arrest but led to intracellular vesicle formation with LC3 aggregation in autophagosomes, suggesting an autophagic cell death. Additionally, 6e had a high-affinity potential for PKM2 protein, higher than the known ligands, such as lapachol or shikonin, and was able to inhibit this enzyme activity in vitro. CONCLUSION We assert that compound 6e shows promise as a potential lead for a novel chemotherapeutic drug targeting OSCC, with potential applicability to other cancer types.
Collapse
Grants
- 1A 301873/2019-4, 301873/2019-4 CNPq, Conselho Nacional de Desenvolvimento Científico e Tecnológico
- E-26/010.101106/2018, E-26/202, 787/2019, E-26/10.002250/2019, E-26/210.085/2022, E-26/010.001318/2019, E-26/211.343/2021, E-26/210.068/2021, E-26/203.191/2017-JCNE, E-26 /202.800/2017-CNE, E-26/010.101106/2018, E-26/200 .870/2021-CNE, E-26/201.369/2021-JCNE, E-26/010/ 001687/2015, E-26/202.787/2019, E-26/210.514/2019, E-26/10.002250/2019, E-26/211.343/2021, E-26/210. 085/2022, E-26/210.068/2021 FAPERJ, Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro
- 001 Coordination for the Improvement of Higher Education Personnel - Brazil (CAPES)
Collapse
Affiliation(s)
- Amanda de Andrade Borges
- Departamento de Química Orgânica, Instituto de Química, Universidade Federal Fluminense, CEP, Niterói, 24020-141, RJ, Brazil
| | - Gabriel Ouverney
- Programa de Pós-graduação em Ciências Aplicadas a Produtos para Saúde, Faculdade de Farmácia, Universidade Federal Fluminense, CEP, Niterói, 24020-141, RJ, Brazil
| | - Afonso Thales Sousa Arruda
- Departamento de Ciência Básica, Universidade Federal Fluminense, Campus Universitário de Nova Friburgo, CEP, Nova Friburgo, 28625-650, RJ, Brazil
| | - Amanda Vieira Ribeiro
- Departamento de Ciência Básica, Universidade Federal Fluminense, Campus Universitário de Nova Friburgo, CEP, Nova Friburgo, 28625-650, RJ, Brazil
| | - Ruan Carlos Busquet Ribeiro
- Departamento de Química Orgânica, Instituto de Química, Universidade Federal Fluminense, CEP, Niterói, 24020-141, RJ, Brazil
| | - Acacio Silva de Souza
- Departamento de Tecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal Fluminense, CEP, Niterói, 24020-141, RJ, Brazil
| | - Anna Carolina Carvalho da Fonseca
- Programa de Pós-graduação em Odontologia, Instituto de Saúde de Nova Friburgo, Universidade Federal Fluminense, CEP, Nova Friburgo, 28625-650, RJ, Brazil
| | - Lucas Nicolau de Queiroz
- Programa de Pós-graduação em Ciências Aplicadas a Produtos para Saúde, Faculdade de Farmácia, Universidade Federal Fluminense, CEP, Niterói, 24020-141, RJ, Brazil
| | - Elan Cardozo Paes de Almeida
- Departamento de Ciência Básica, Universidade Federal Fluminense, Campus Universitário de Nova Friburgo, CEP, Nova Friburgo, 28625-650, RJ, Brazil
| | - Bruno Pontes
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, CEP, Rio de Janeiro, 21941-902, RJ, Brazil
| | - Vitor Won-Held Rabelo
- Instituto de Biodiversidade e Sustentabilidade, Universidade Federal do Rio de Janeiro, CEP , Macaé, 27965-045, RJ, Brazil
| | - Vitor Ferreira
- Departamento de Tecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal Fluminense, CEP, Niterói, 24020-141, RJ, Brazil
| | - Paula Alvarez Abreu
- Instituto de Biodiversidade e Sustentabilidade, Universidade Federal do Rio de Janeiro, CEP , Macaé, 27965-045, RJ, Brazil
| | - Fernando de Carvalho da Silva
- Departamento de Química Orgânica, Instituto de Química, Universidade Federal Fluminense, CEP, Niterói, 24020-141, RJ, Brazil
| | - Luana da Silva Magalhaes Forezi
- Departamento de Química Orgânica, Instituto de Química, Universidade Federal Fluminense, CEP, Niterói, 24020-141, RJ, Brazil
| | - Bruno Kaufmann Robbs
- Departamento de Ciência Básica, Universidade Federal Fluminense, Campus Universitário de Nova Friburgo, CEP, Nova Friburgo, 28625-650, RJ, Brazil
| |
Collapse
|
25
|
Chowdhury MG, Kapoor S, Muthukumar V, Chatterjee DR, Shard A. Development of novel tetrazole-based pyruvate kinase M2 inhibitors targeting U87MG glioblastoma cells. Bioorg Chem 2025; 154:108029. [PMID: 39693922 DOI: 10.1016/j.bioorg.2024.108029] [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: 10/03/2024] [Revised: 11/19/2024] [Accepted: 12/02/2024] [Indexed: 12/20/2024]
Abstract
Glioblastoma (GB), the most aggressive and life-threatening primary brain tumor in adults, poses significant therapeutic challenges. Tumor pyruvate kinase M2 (PKM2) has been implicated in the proliferation and survival of glioma cells. In this study, we designed and synthesized a series of 23 novel tetrazole-based derivatives. The compounds were thoroughly characterized using 1H, 13C, 19F NMR, along with HRMS analysis. Among them, 1-(imidazo[1,2-a]pyrimidin-3-yl)-2-(5-(naphthalen-2-yl)-2H-tetrazol-2-yl)ethan-1-one (9b) exhibited potent and selective antiproliferative activity against U87MG glioma cells, with minimal effects on bEnd (brain endothelial cell line) non-glioma cells. It emerged as a potent PKM2 inhibitor, with an IC50 of 0.307 µM. Apoptosis assays and cell cycle analysis revealed that compound 9b induced early apoptosis and caused G1 phase arrest. A significant decrease in pyruvate concentration further suggested PKM2 inhibition. In silico studies confirmed the binding affinity to the PKM2 inhibitory site, and RT-PCR data demonstrated its inhibitory activity against PKM2. Additionally, it reduced lactate levels, indicating its potential impact on cellular metabolism. Collectively, these findings suggest that the most potent compound holds promise as a therapeutic candidate against GB.
Collapse
Affiliation(s)
- Moumita Ghosh Chowdhury
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad (NIPER-A), Gandhinagar, Gujarat, India
| | - Saumya Kapoor
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad (NIPER-A), Gandhinagar, Gujarat, India
| | - Venkatesh Muthukumar
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad (NIPER-A), Gandhinagar, Gujarat, India
| | - Deep Rohan Chatterjee
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad (NIPER-A), Gandhinagar, Gujarat, India
| | - Amit Shard
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad (NIPER-A), Gandhinagar, Gujarat, India.
| |
Collapse
|
26
|
Tian M, Yang L, Zhao Z, Li J, Wang L, Yin Q, Hu W, Lou Y, Du J, Zhao P. TIPE drives a cancer stem-like phenotype by promoting glycolysis via PKM2/HIF-1α axis in melanoma. eLife 2024; 13:RP92741. [PMID: 39728923 DOI: 10.7554/elife.92741] [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] [Indexed: 12/28/2024] Open
Abstract
TIPE (TNFAIP8) has been identified as an oncogene and participates in tumor biology. However, how its role in the metabolism of tumor cells during melanoma development remains unclear. Here, we demonstrated that TIPE promoted glycolysis by interacting with pyruvate kinase M2 (PKM2) in melanoma. We found that TIPE-induced PKM2 dimerization, thereby facilitating its translocation from the cytoplasm to the nucleus. TIPE-mediated PKM2 dimerization consequently promoted HIF-1α activation and glycolysis, which contributed to melanoma progression and increased its stemness features. Notably, TIPE specifically phosphorylated PKM2 at Ser 37 in an extracellular signal-regulated kinase (ERK)-dependent manner. Consistently, the expression of TIPE was positively correlated with the levels of PKM2 Ser37 phosphorylation and cancer stem cell (CSC) markers in melanoma tissues from clinical samples and tumor bearing mice. In summary, our findings indicate that the TIPE/PKM2/HIF-1α signaling pathway plays a pivotal role in promoting CSC properties by facilitating the glycolysis, which would provide a promising therapeutic target for melanoma intervention.
Collapse
Affiliation(s)
- Maojin Tian
- Center of Translational Medicine, Zibo Central Hospital Affiliated to Binzhou Medical University, Zibo, China
| | - Le Yang
- Shandong First Medical University, Jinan, China
| | - Ziqian Zhao
- The Second Medical College, Xinjiang Medical University, Urumqi, China
| | - Jigang Li
- Center of Translational Medicine, Zibo Central Hospital Affiliated to Binzhou Medical University, Zibo, China
| | - Lianqing Wang
- Center of Translational Medicine, Zibo Central Hospital Affiliated to Binzhou Medical University, Zibo, China
| | | | - Wei Hu
- Center of Translational Medicine, Zibo Central Hospital Affiliated to Binzhou Medical University, Zibo, China
| | - Yunwei Lou
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Jianxin Du
- Center of Translational Medicine, Zibo Central Hospital Affiliated to Binzhou Medical University, Zibo, China
| | - Peiqing Zhao
- Center of Translational Medicine, Zibo Central Hospital Affiliated to Binzhou Medical University, Zibo, China
| |
Collapse
|
27
|
Ravera S, Puddu A, Bertola N, Verzola D, Russo E, Maggi D, Panfoli I. IGF-1 Signaling Modulates Oxidative Metabolism and Stress Resistance in ARPE-19 Cells Through PKM2 Function. Int J Mol Sci 2024; 25:13640. [PMID: 39769402 PMCID: PMC11727907 DOI: 10.3390/ijms252413640] [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: 11/26/2024] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 01/16/2025] Open
Abstract
The retinal pigment epithelium (RPE) contributes to retinal homeostasis, and its metabolic dysfunction is implied in the development of retinal degenerative disease. The isoform M2 of pyruvate kinase (PKM2) is a key factor in cell metabolism, and its function may be affected by insulin-like growth factor 1 (IGF-1). This study aims to investigate the effect of IGF-1 on PKM2 modulation of RPE cells and whether co-treatment with klotho may preserve it. ARPE-19 cells, an ex vivo model of human pigmented epithelium, were exposed to IGF-1. Then, we evaluated PKM2 expression, dimerization and subcellular localization, energy metabolism, and redox balance, and whether pre-treatment with Klotho may antagonize the effects of IGF-1. The results show that IGF-1 favors PKM2 dimerization, thus reducing the activity of PKM2 and leading to an altered cellular energy status coupled with reduced oxidative stress. In conclusion, PKM2 plays a pivotal role in the modulation of RPE metabolism and redox balance and could explain the mechanisms through which IGF-1 participates in the pathogenesis of some retinal diseases. Klotho may exert protective effects by mitigating the IGF-1 signal and its effect on mitochondrial function.
Collapse
Affiliation(s)
- Silvia Ravera
- Department of Experimental Medicine, University of Genoa, Via De Toni 14, 16132 Genova, Italy;
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy;
| | - Alessandra Puddu
- Department of Internal Medicine and Medical Specialties, University of Genova, Viale Benedetto XV 6, 16132 Genova, Italy; (D.V.); (E.R.); (D.M.)
| | - Nadia Bertola
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy;
| | - Daniela Verzola
- Department of Internal Medicine and Medical Specialties, University of Genova, Viale Benedetto XV 6, 16132 Genova, Italy; (D.V.); (E.R.); (D.M.)
| | - Elisa Russo
- Department of Internal Medicine and Medical Specialties, University of Genova, Viale Benedetto XV 6, 16132 Genova, Italy; (D.V.); (E.R.); (D.M.)
- Unit of Nephrology, Dialysis and Transplantation, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Davide Maggi
- Department of Internal Medicine and Medical Specialties, University of Genova, Viale Benedetto XV 6, 16132 Genova, Italy; (D.V.); (E.R.); (D.M.)
| | - Isabella Panfoli
- Department of Pharmacy—(DIFAR), University of Genoa, Viale Benedetto XV 3, 16132 Genova, Italy;
| |
Collapse
|
28
|
Ahluwalia P, Mondal AK, Vashisht A, Singh H, Alptekin A, Ballur K, Omar N, Ahluwalia M, Jones K, Barrett A, Kota V, Kolhe R. Identification of a distinctive immunogenomic gene signature in stage-matched colorectal cancer. J Cancer Res Clin Oncol 2024; 151:9. [PMID: 39673574 PMCID: PMC11646222 DOI: 10.1007/s00432-024-06034-4] [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: 05/21/2024] [Accepted: 11/11/2024] [Indexed: 12/16/2024]
Abstract
BACKGROUND Colorectal cancer (CRC) remains one of the leading causes of cancer-related mortality worldwide. Despite advances in diagnosis and treatment, including surgery, chemotherapy, and immunotherapy, accurate clinical markers are still lacking. The development of prognostic and predictive indicators, particularly in the context of personalized medicine, could significantly improve CRC patient management. METHOD In this retrospective study, we used FFPE blocks of tissue samples from CRC patients at Augusta University (AU) to quantify a custom 15-gene panel. To differentiate the tumor and adjacent normal regions (NAT), H&E staining was utilized. For the quantification of transcripts, we used the NanoString nCounter platform. Kaplan-Meier and Log-rank tests were used to perform survival analyses. Several independent datasets were explored to validate the gene signature. Orthogonal analyses included single-cell profiling, differential gene expression, immune cell deconvolution, neoantigen prediction, and biological pathway assessment. RESULTS A 3-gene signature (GTF3A, PKM, and VEGFA) was found to be associated with overall survival in the AU cohort (HR = 2.26, 95% CI 1.05-4.84, p = 0.02, 93 patients), TCGA cohort (HR = 1.57, 95% CI 1.05-2.35, p < 0.02, 435 patients) and four other GEO datasets. Independent single-cell analysis identified relatively higher expression of the 3-gene signature in the tumor region. Differential analysis revealed dysregulated tissue inflammation, immune dysfunction, and neoantigen load of cell cycle processes among high-risk patients compared to low-risk patients. CONCLUSION We developed a 3-gene signature with the potential for prognostic and predictive clinical assessment of CRC patients. This gene-based stratification offers a cost-effective approach to personalized cancer management. Further research using similar methods could identify therapy-specific gene signatures to strengthen the development of personalized medicine for CRC patients.
Collapse
Affiliation(s)
- Pankaj Ahluwalia
- Department of Pathology, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, BF-207, USA
| | - Ashis K Mondal
- Department of Pathology, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, BF-207, USA
| | - Ashutosh Vashisht
- Department of Pathology, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, BF-207, USA
| | - Harmanpreet Singh
- Department of Pathology, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, BF-207, USA
| | - Ahmet Alptekin
- Department of Pathology, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, BF-207, USA
| | - Kalyani Ballur
- Department of Pathology, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, BF-207, USA
| | - Nivin Omar
- Department of Pathology, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, BF-207, USA
| | | | - Kimya Jones
- Department of Pathology, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, BF-207, USA
| | - Amanda Barrett
- Department of Pathology, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, BF-207, USA
| | - Vamsi Kota
- Georgia Cancer Center at Augusta University, Augusta, GA, 30912, USA
- Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Ravindra Kolhe
- Department of Pathology, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, BF-207, USA.
| |
Collapse
|
29
|
Huang Y, Li B, Gui Z, Gao E, Yuan Y, Yang J, Hekmatyar K, Mishra F, Chan P, Liu Z. Extracellular PKM2 Preserves Cardiomyocytes and Reduces Cardiac Fibrosis During Myocardial Infarction. Int J Mol Sci 2024; 25:13246. [PMID: 39769010 PMCID: PMC11675365 DOI: 10.3390/ijms252413246] [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: 11/05/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 01/11/2025] Open
Abstract
Substantial loss of cardiomyocytes during heart attacks and onset of other cardiovascular diseases is a major cause of mortality. Preservation of cardiomyocytes during cardiac injury would be the most effective strategy to manage these diseases in clinic. However, there is no effective treatment strategy that is able to prevent cardiomyocyte loss. We demonstrate here that the systemic administration of a recombinant PKM2 mutant (G415R) preserves cardiomyocytes and reduces cardiac fibrosis during myocardial infarction. G415R preserves cardiomyocytes by protecting the cardiomyocytes from dying and by promoting cardiomyocyte proliferation. Preservation of cardiomyocytes by extracellular PKM2 (EcPKM2) reduces cardiac fibrosis because of the decreased activation of cardiac fibroblasts. Our experiments show that EcPKM2 (G415R) exerts its action by interacting with integrin avb3 on cardiomyocytes. EcPKM2(G415R) activates the integrin-FAK-PI3K signaling axis, which subsequently suppresses PTEN expression and consequently regulates cardiomyocyte apoptosis resistance and proliferation under hypoxia and oxidative stress conditions. Our studies uncover an important cardiomyocyte protection mechanism. More importantly, the activity/action of EcPKM2 (G415R) in preserving cardiomyocyte suggesting a possible therapeutic strategy and target for the treatment of heart attacks and other cardiovascular diseases.
Collapse
Affiliation(s)
- Yang Huang
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (Y.H.); (B.L.); (Y.Y.); (F.M.); (P.C.)
| | - Bin Li
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (Y.H.); (B.L.); (Y.Y.); (F.M.); (P.C.)
| | - Zongxiang Gui
- Department of Chemistry, Georgia State University, Atlanta, GA 30303, USA; (Z.G.); (J.Y.); (K.H.)
| | - Erhe Gao
- Center for Translational Medicine, Temple University, Philadelphia, PA 19140, USA;
| | - Yi Yuan
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (Y.H.); (B.L.); (Y.Y.); (F.M.); (P.C.)
| | - Jenny Yang
- Department of Chemistry, Georgia State University, Atlanta, GA 30303, USA; (Z.G.); (J.Y.); (K.H.)
| | - Khan Hekmatyar
- Department of Chemistry, Georgia State University, Atlanta, GA 30303, USA; (Z.G.); (J.Y.); (K.H.)
| | - Falguni Mishra
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (Y.H.); (B.L.); (Y.Y.); (F.M.); (P.C.)
| | - Payton Chan
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (Y.H.); (B.L.); (Y.Y.); (F.M.); (P.C.)
| | - Zhiren Liu
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (Y.H.); (B.L.); (Y.Y.); (F.M.); (P.C.)
| |
Collapse
|
30
|
Cheng S, Li Y, Sun X, Liu Z, Guo L, Wu J, Yang X, Wei S, Wu G, Xu S, Yang F, Wu J. The impact of glucose metabolism on inflammatory processes in sepsis-induced acute lung injury. Front Immunol 2024; 15:1508985. [PMID: 39712019 PMCID: PMC11659153 DOI: 10.3389/fimmu.2024.1508985] [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: 10/10/2024] [Accepted: 11/18/2024] [Indexed: 12/24/2024] Open
Abstract
Acute lung injury (ALI) is a prevalent and critical complication of sepsis, marked by high incidence and mortality rates, with its pathogenesis still not being fully elucidated. Recent research has revealed a significant correlation between the metabolic reprogramming of glucose and sepsis-associated ALI (S-ALI). Throughout the course of S-ALI, immune cells, including macrophages and dendritic cells, undergo metabolic shifts to accommodate the intricate demands of immune function that emerge as sepsis advances. Indeed, glucose metabolic reprogramming in S-ALI serves as a double-edged sword, fueling inflammatory immune responses in the initial stages and subsequently initiating anti-inflammatory responses as the disease evolves. In this review, we delineate the current research progress concerning the pathogenic mechanisms linked to glucose metabolic reprogramming in S-ALI, with a focus on the pertinent immune cells implicated. We encapsulate the impact of glucose metabolic reprogramming on the onset, progression, and prognosis of S-ALI. Ultimately, by examining key regulatory factors within metabolic intermediates and enzymes, We have identified potential therapeutic targets linked to metabolic reprogramming, striving to tackle the inherent challenges in diagnosing and treating Severe Acute Lung Injury (S-ALI) with greater efficacy.
Collapse
Affiliation(s)
- Shilei Cheng
- School of Anesthesiology, Shandong Second Medical University, Weifang, China
| | - Yufei Li
- Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Institute of Anesthesia and Respiratory Critical Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Anesthesiology, Jinan, China
- School of Pharmacy, Shandong University of Traditional Chinese Medicine (TCM), Jinan, China
| | - Xiaoliang Sun
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhirui Liu
- Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Liang Guo
- Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Institute of Anesthesia and Respiratory Critical Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Anesthesiology, Jinan, China
| | - Jueheng Wu
- Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Xiaohan Yang
- Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Sisi Wei
- Department of Anesthesiology, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, China
| | - Guanghan Wu
- Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Institute of Anesthesia and Respiratory Critical Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Anesthesiology, Jinan, China
| | - Shilong Xu
- School of Anesthesiology, Shandong Second Medical University, Weifang, China
| | - Fan Yang
- Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Institute of Anesthesia and Respiratory Critical Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Anesthesiology, Jinan, China
| | - Jianbo Wu
- Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Institute of Anesthesia and Respiratory Critical Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Anesthesiology, Jinan, China
| |
Collapse
|
31
|
Yang Y, Xie T, Gao P, Han W, Liu Y, Wang Y. Hsa_Circ_002144 Promotes Glycolysis and Immune Escape of Breast Cancer Through miR-326/PKM Axis. Cancer Biother Radiopharm 2024; 39:755-769. [PMID: 38963787 DOI: 10.1089/cbr.2024.0009] [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] [Indexed: 07/06/2024] Open
Abstract
Background: Breast cancer is a leading cause of cancer-related deaths in women worldwide, posing a significant threat to female health. Therefore, it is crucial to search for new therapeutic targets and prognostic biomarkers for breast cancer patients. Method: Bioinformatics analysis, quantitative real-time PCR (qRT-PCR), and fluorescence in situ hybridization (FISH) were employed to investigate the expression of hsa_circ_002144 in breast cancer. Transwell assay, Western blotting, and cell viability assay were utilized to assess the impact of hsa_circ_002144 on the proliferation, migration, and invasion of breast cancer cells. Additionally, a mouse model was established to validate its functionality. Flow cytometry, WB analysis, enzyme-linked immunosorbent assay (ELISA), qRT-PCR, exosomes isolation, and co-culture system were employed to elucidate the molecular mechanism underlying macrophage polarization. Result: we have discovered for the first time that hsa_circ_002144 is highly expressed in breast cancer. It affected tumor growth and metastasis and could influence macrophage polarization through the glycolytic pathway. Conclusion: This finding provides a new direction for breast cancer treatment and prognosis assessment.
Collapse
Affiliation(s)
- Yong Yang
- College of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang City, China
| | - Tianhao Xie
- General Surgery, The Affiliated Hospital of Hebei University, Baoding City, China
| | - Peng Gao
- Anesthesiology department, Affiliated hospital of Qingdao university, Qingdao City, China
| | - Weijun Han
- Third Surgery, Baoji traditional Chinese Medicine Hospital in Shaanxi Province, Baoji City, China
| | - Yuhong Liu
- Rheumatology and Immunology Department, The Affiliated Hospital of Yan 'an University, Yan 'an City, China
| | - Yanmei Wang
- School of Nursing and Health, Medical College of Yan 'an University, Yan 'an City, China
| |
Collapse
|
32
|
Sada T, Kimura W. Transition from fetal to postnatal state in the heart: Crosstalk between metabolism and regeneration. Dev Growth Differ 2024; 66:438-451. [PMID: 39463005 DOI: 10.1111/dgd.12947] [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: 06/16/2024] [Revised: 09/16/2024] [Accepted: 10/04/2024] [Indexed: 10/29/2024]
Abstract
Cardiovascular disease is the leading cause of mortality worldwide. Myocardial injury resulting from ischemia can be fatal because of the limited regenerative capacity of adult myocardium. Mammalian cardiomyocytes rapidly lose their proliferative capacities, with only a small fraction of adult myocardium remaining proliferative, which is insufficient to support post-injury recovery. Recent investigations have revealed that this decline in myocardial proliferative capacity is closely linked to perinatal metabolic shifts. Predominantly glycolytic fetal myocardial metabolism transitions towards mitochondrial fatty acid oxidation postnatally, which not only enables efficient production of ATP but also causes a dramatic reduction in cardiomyocyte proliferative capacity. Extensive research has elucidated the mechanisms behind this metabolic shift, as well as methods to modulate these metabolic pathways. Some of these methods have been successfully applied to enhance metabolic reprogramming and myocardial regeneration. This review discusses recently acquired insights into the interplay between metabolism and myocardial proliferation, emphasizing postnatal metabolic transitions.
Collapse
Affiliation(s)
- Tai Sada
- Laboratory for Heart Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Wataru Kimura
- Laboratory for Heart Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| |
Collapse
|
33
|
Jin M, Shi L, Wang L, Zhang D, Li Y. Dihydroartemisinin enhances the anti-tumour effect of photodynamic therapy by targeting PKM2-mediated glycolysis in oesophageal cancer cell. J Enzyme Inhib Med Chem 2024; 39:2296695. [PMID: 38111311 PMCID: PMC11722009 DOI: 10.1080/14756366.2023.2296695] [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: 06/09/2023] [Revised: 10/17/2023] [Accepted: 12/13/2023] [Indexed: 12/20/2023] Open
Abstract
Photodynamic therapy (PDT) has been demonstrated to provide immediate relief of oesophageal cancer patients' re-obstruction and extend their lifespan. However, tumour regrowth may occur after PDT due to enhanced aerobic glycolysis. Previous research has confirmed the inhibitory effect of Dihydroartemisinin (DHA) on aerobic glycolysis. Therefore, the current study intends to investigate the function and molecular mechanism of DHA targeting tumour cell aerobic glycolysis in synergia PDT. The combined treatment significantly suppressed glycolysis in vitro and in vivo compared to either monotherapy. Exploration of the mechanism through corresponding experiments revealed that pyruvate kinase M2 (PKM2) was downregulated in treated cells, whereas overexpression of PKM2 nullified the inhibitory effects of DHA and PDT. This study proposes a novel therapeutic strategy for oesophageal cancer through DHA-synergized PDT treatment, targeting inhibit PKM2 to reduce tumour cell proliferation and metastasis.
Collapse
Affiliation(s)
- Mengru Jin
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, P. R. China
| | - Luyao Shi
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, P. R. China
| | - Li Wang
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, P. R. China
| | - Dingyuan Zhang
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, P. R. China
| | - Yanjing Li
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, P. R. China
| |
Collapse
|
34
|
Lu J, Wang X, Shi X, Jiang J, Liu L, Liu L, Ren C, Lu C, Yu Z. PAK5-mediated PKM2 phosphorylation is critical for anaerobic glycolysis in endometriosis. Front Med 2024; 18:1054-1067. [PMID: 39331255 DOI: 10.1007/s11684-024-1069-3] [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: 10/15/2023] [Accepted: 02/08/2024] [Indexed: 09/28/2024]
Abstract
P21-activated kinase 5 (PAK5) belongs to the PAK-II subfamily, which is an important regulator of cell survival, adhesion, and motility. However, the functions of PAK5 in endometriosis remain unclear. Here, PAK5 is strikingly upregulated in endometriosis. Furthermore, the knockdown of PAK5 or its inhibitor GNE 2861 blocks the development of endometriosis, which is equally demonstrated in PAK5-knockout mice. In addition, PAK5 promotes glycolysis by enhancing the protein stability of pyruvate kinase 2 (PKM2) in endometriotic cells, which is a key enzyme for glucose metabolism. Moreover, the phosphorylation of PKM2 at Ser519 by PAK5 mediates endometriosis cell proliferation and metastasis. Collectively, PAK5 plays an indispensable role in endometriosis. Our findings demonstrate that PAK5 is an important target for the treatment of endometriosis.
Collapse
Affiliation(s)
- Jiayi Lu
- Department of Reproductive Medicine, Affiliated Hospital of Shandong Second Medical University, Weifang, 261042, China
- School of Clinical Medicine, Shandong Second Medical University, Weifang, 261053, China
| | - Xiaoyun Wang
- Department of Reproductive Medicine, Affiliated Hospital of Shandong Second Medical University, Weifang, 261042, China
- School of Clinical Medicine, Shandong Second Medical University, Weifang, 261053, China
| | - Xiaodan Shi
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
| | - Junyi Jiang
- Department of Reproductive Medicine, Affiliated Hospital of Shandong Second Medical University, Weifang, 261042, China
- School of Clinical Medicine, Shandong Second Medical University, Weifang, 261053, China
| | - Lan Liu
- Department of Reproductive Medicine, Affiliated Hospital of Shandong Second Medical University, Weifang, 261042, China
- School of Clinical Medicine, Shandong Second Medical University, Weifang, 261053, China
| | - Lu Liu
- Department of Reproductive Medicine, Affiliated Hospital of Shandong Second Medical University, Weifang, 261042, China
- School of Clinical Medicine, Shandong Second Medical University, Weifang, 261053, China
| | - Chune Ren
- Department of Reproductive Medicine, Affiliated Hospital of Shandong Second Medical University, Weifang, 261042, China.
| | - Chao Lu
- Department of Reproductive Medicine, Affiliated Hospital of Shandong Second Medical University, Weifang, 261042, China.
| | - Zhenhai Yu
- Department of Reproductive Medicine, Affiliated Hospital of Shandong Second Medical University, Weifang, 261042, China.
| |
Collapse
|
35
|
Jin Y, Liu Q, Sun B, Li X, Wu J, Lin Z, Ma Y, Jia H. Pralatrexate represses the resistance of HCC cells to molecular targeted agents via the miRNA-34a/Notch pathway. Discov Oncol 2024; 15:709. [PMID: 39585461 PMCID: PMC11589030 DOI: 10.1007/s12672-024-01572-2] [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: 07/26/2024] [Accepted: 11/11/2024] [Indexed: 11/26/2024] Open
Abstract
Metabolism-related pathways are important targets for intervention in the treatment of hepatocellular carcinoma (HCC), but few studies have reported on the combination of inhibitors of folate metabolism-related enzymes and molecularly targeted drugs for HCC. The results of the present work are the first to reveal the effects of an inhibitor of dihydrofolate reductase (DHFR), pralatrexate, on the sensitivity of HCC cells to molecularly targeted agents examined using multiple assays. In HCC cells, knockdown of DHFR or treatment with pralatrexate enhanced the sensitivity of HCC cells to molecularly targeted agents, such as sorafenib, regorafenib, lenvatinib, cabozantinib, or anlotinib. Mechanically, pralatrexate decreased the methylation rates of miRNA-34a's promoter region to enhance the expression of miRNA-34a. Treatment with pralatrexate inhibited the expression of Notch and its downstream factors by enhancing the expression of miRNA-34a in HCC cells. In clinical specimens, the expression of miRNA-34a was negatively correlated with DHFR expression, while DHFR expression was positively correlated with the Notch intracellular domain (NICD) and downstream factors of the Notch pathway. The expression of miRNA-34a was negatively correlated with DHFR expression, while the methylation rates of miRNA-34a's promoter were positively related to DHFR. The effect of pralatrexate on the metabolic profile of HCC cells is very different from that of small molecule inhibitors related to glycolipid metabolism. Therefore, pralatrexate upregulates the sensitivity of HCC cells to molecularly targeted drugs. These results expand our understanding of folate metabolism and HCC and can help provide more options for HCC treatment.
Collapse
Affiliation(s)
- Yang Jin
- The 920th Hospital of the PLA Joint Logistic Support Force, Kunming, 650032, Yunnan, People's Republic of China
| | - Qiming Liu
- Air Force Medical Center, Chinese People's Liberation Army, Beijing, 100142, People's Republic of China
| | - Baisheng Sun
- Department of Critical Care Medicine, The First Medical Centre, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Xiaokang Li
- The 63650 Military Hospital, Chinese People's Liberation Army, Urumqi, 841700, China
| | - Jiahao Wu
- The 63650 Military Hospital, Chinese People's Liberation Army, Urumqi, 841700, China
| | - Zhiyuan Lin
- The 63650 Military Hospital, Chinese People's Liberation Army, Urumqi, 841700, China
| | - Yan Ma
- Department of Gastroenterology and Hepatology, The First Medical Centre, Chinese People's Liberation Army General Hospital, Beijing, 100853, People's Republic of China.
| | - Haijiang Jia
- Department of Quality Management, the 967th Hospital of Joint Logistic Support Force of Chinese People's Liberation Army, No. 80 Shengli Road, Xigang District, Dalian, 116021, Liaoning Province, People's Republic of China.
| |
Collapse
|
36
|
Bögel G, Sváb G, Murányi J, Szokol B, Kukor Z, Kardon T, Őrfi L, Tretter L, Hrabák A. The role of PI3K-Akt-mTOR axis in Warburg effect and its modification by specific protein kinase inhibitors in human and rat inflammatory macrophages. Int Immunopharmacol 2024; 141:112957. [PMID: 39197292 DOI: 10.1016/j.intimp.2024.112957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 07/25/2024] [Accepted: 08/14/2024] [Indexed: 09/01/2024]
Abstract
The Warburg effect occurs both in cancer cells and in inflammatory macrophages. The aim of our work was to demonstrate the role of PI3K-Akt-mTOR axis in the Warburg effect in HL-60 derived, rat peritoneal and human blood macrophages and to investigate the potential of selected inhibitors of this pathway to antagonize it. M1 polarization in HL-60-derived and human blood monocyte-derived macrophages was supported by the increased expression of NOS2 and inflammatory cytokines. All M1 polarized and inflammatory macrophages investigated expressed higher levels of HIF-1α and NOS2, which were reduced by selected kinase inhibitors, supporting the role of PI3K-Akt-mTOR axis. Using Seahorse XF plates, we found that in HL-60-derived and human blood-derived macrophages, glucose loading reduced oxygen consumption (OCR) and increased glycolysis (ECAR) in M1 polarization, which was antagonized by selected kinase inhibitors and by dichloroacetate. In rat peritoneal macrophages, the changes in oxidative and glycolytic metabolism were less marked and the NOS2 inhibitor decreased OCR and increased ECAR. Non-mitochondrial oxygen consumption and ROS production were likely due to NADPH oxidase, expressed in each macrophage type, independently of PI3K-Akt-mTOR axis. Our results suggest that inflammation changed the metabolism in each macrophage model, but a clear relationship between polarization and Warburg effect was confirmed only after glucose loading in HL-60 and human blood derived macrophages. The effect of kinase inhibitors on Warburg effect was variable in different cell types, whereas dichloroacetate caused a shift toward oxidative metabolism. Our findings suggest that these originally anti-cancer inhibitors may also be candidates for anti-inflammatory therapy.
Collapse
Affiliation(s)
- Gábor Bögel
- Department of Molecular Biology, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, H-1094 Tűzoltó u. 37-47, Hungary
| | - Gergely Sváb
- Department of Biochemistry, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, H-1094 Tűzoltó u. 37-47, Hungary
| | - József Murányi
- Department of Molecular Biology, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, H-1094 Tűzoltó u. 37-47, Hungary
| | - Bálint Szokol
- Vichem Chemie Research Ltd., Veszprém, H-8200, Viola u. 2., Hungary
| | - Zoltán Kukor
- Department of Molecular Biology, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, H-1094 Tűzoltó u. 37-47, Hungary
| | - Tamás Kardon
- Department of Molecular Biology, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, H-1094 Tűzoltó u. 37-47, Hungary
| | - László Őrfi
- Vichem Chemie Research Ltd., Veszprém, H-8200, Viola u. 2., Hungary; Department of Pharmaceutical Chemistry, Semmelweis University, Budapest, H-1092 Hőgyes E. u. 9., Hungary
| | - László Tretter
- Department of Biochemistry, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, H-1094 Tűzoltó u. 37-47, Hungary
| | - András Hrabák
- Department of Molecular Biology, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, H-1094 Tűzoltó u. 37-47, Hungary.
| |
Collapse
|
37
|
Wang X, Ding J, Chen K, Hu H, Huang B, Shi G, Li S. Probing Selenium-Deficient Chicken Spleen Th1/Th17 Differentiation Based on Selenoprotein W Targeting of PKM2/HIF1α. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:24808-24822. [PMID: 39441563 DOI: 10.1021/acs.jafc.4c04795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Selenium regulates the differentiation and function of immune cells mainly through selenoproteins. Selenoprotein W (SelW) has been shown to mitigate inflammatory bowel disease in mice by modulating the differentiation of helper T (CD4+ T) cell. Previous studies by our team have underscored SelW's critical role in safeguarding chicken spleens and splenic lymphocytes against inflammatory injury. However, research examining SelW's involvement in regulating CD4+ T cell differentiation in avian spleens remains scarce. Therefore, the selenium-deficient chicken model was constructed in this study. It was found that the spleen of selenium-deficient chickens showed significant inflammatory damage, accompanied by decreased SelW expression, diminished antioxidant capacity, heightened glycolysis, and an elevated count of Th1/Th17 cells. To elucidate the specific mechanism of SelW regulating Th1/Th17 cell differentiation, this study used molecular docking technology, fluorescence colocalization, and co-immunoprecipitation and initially confirmed the targeting relationship between SelW and pyruvate kinase M2 (PKM2). Subsequently, an in vitro model of SelW overexpression, knockdown, and TEPP-46 (PKM2 tetramer activator) cotreatment of chicken primary splenic lymphocytes was replicated. Our findings revealed that selenium deficiency triggers oxidative stress and promotes PKM2 nuclear translocation via SelW downregulation, which stabilizes HIF1α transcription in the nucleus, enhancing glycolysis and skewing chicken splenic CD4+ T cells toward the Th1/Th17 phenotype. Our study, for the first time, demonstrates the existence of an interaction between SelW and PKM2 in poultry, emphasizing SelW's paramount significance in CD4+ T cell differentiation, providing fresh perspectives on the contributions of selenoproteins to T cell biology and immune processes.
Collapse
Affiliation(s)
- Xixi Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Jiayi Ding
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Kai Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Haodong Hu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Bo Huang
- National Selenium-rich Product Quality Supervision and Inspection Center, Enshi 445000, China
| | - Guangliang Shi
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Shu Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
38
|
Hou N, Zhou H, Li J, Xiong X, Deng H, Xiong S. Macrophage polarization and metabolic reprogramming in abdominal aortic aneurysm. Immun Inflamm Dis 2024; 12:e1268. [PMID: 39530309 PMCID: PMC11555488 DOI: 10.1002/iid3.1268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 04/03/2024] [Accepted: 04/22/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) is a macrovascular disease with high morbidity and mortality in the elderly. The limitation of the current management is that most patients can only be followed up until the AAA diameter increases to a threshold, and surgical intervention is recommended. The development of preventive and curative drugs for AAA is urgently needed. Macrophage-mediated immune inflammation is one of the key pathological links in the occurrence and development of AAA. AIMS This review article aims to evaluate the impact of immunometabolism on macrophage biology and its role in AAA. METHODS We analyze publications focusing on the polarization and metabolic reprogramming in macrophages as well as their potential impact on AAA, and summarize the potential interventions that are currently available to regulate these processes. RESULTS The phenotypic and functional changes in macrophages are accompanied by significant alterations in metabolic pathways. The interaction between macrophage polarization and metabolic pathways significantly influences the progression of AAA. CONCLUSION Macrophage polarization is a manifestation of the gross dichotomy of macrophage function into pro-inflammatory killing and tissue repair, that is, classically activated M1 macrophages and alternatively activated M2 macrophages. Macrophage functions are closely linked to metabolic changes, and the emerging field of immunometabolism is providing unique insights into the role of macrophages in AAA. It is essential to further investigate the precise metabolic changes and their functional consequences in AAA-associated macrophages.
Collapse
Affiliation(s)
- Ningxin Hou
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Hongmin Zhou
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Jun Li
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiaoxing Xiong
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanChina
| | - Hongping Deng
- Department of Vascular SurgeryRenmin Hospital of Wuhan UniversityWuhanChina
| | - Sizheng Xiong
- Department of Vascular SurgeryRenmin Hospital of Wuhan UniversityWuhanChina
| |
Collapse
|
39
|
Hu P, Xu Y, Su Y, Wang Y, Xiong Y, Ding Y. Nuclear-localized pyruvate kinases control phosphorylation of histone H3 on threonine 11. NATURE PLANTS 2024; 10:1682-1697. [PMID: 39367257 DOI: 10.1038/s41477-024-01821-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 09/13/2024] [Indexed: 10/06/2024]
Abstract
Phosphorylation of histone H3 at threonine 11 (H3T11ph) affects transcription and chromosome stability. However, the enzymes responsible for depositing H3T11ph and the functions of H3T11ph in plants remain unknown. Here we report that in Arabidopsis thaliana, PYRUVATE KINASE 6 (PK6), PK7 and PK8 enter the nucleus under conditions of sufficient glucose and light exposure, where they interact with SWI2/SNF2-RELATED 1 COMPLEX 4 (SWC4) and phosphorylate H3 at threonine 11. Mutations in these kinases or knockdown of SWC4 resulted in FLC-dependent early flowering, short hypocotyls and short pedicels. Genome-wide, H3T11ph is highly enriched at transcription start sites and transcription termination sites, and positively correlated with gene transcript levels. PK6 and SWC4 targeted FLC, MYB73, PRE1, TCP4 and TCP10, depositing H3T11ph at these loci and promoting their transcription, and PK6 occupancy at these loci requires SWC4. Together, our results reveal that nuclear-localized PK6, PK7 and PK8 modulate H3T11ph and plant growth.
Collapse
Affiliation(s)
- Pengcheng Hu
- Ministry of Education Key Laboratory for Membraneless Organelles and Cellular Dynamics; School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yanmei Xu
- Ministry of Education Key Laboratory for Membraneless Organelles and Cellular Dynamics; School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yanhua Su
- Ministry of Education Key Laboratory for Membraneless Organelles and Cellular Dynamics; School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yuxin Wang
- Ministry of Education Key Laboratory for Membraneless Organelles and Cellular Dynamics; School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yan Xiong
- Haixia Institute of Science and Technology, Synthetic Biology Center, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Yong Ding
- Ministry of Education Key Laboratory for Membraneless Organelles and Cellular Dynamics; School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China.
| |
Collapse
|
40
|
Sun L, Xiao M, Chen L, Ni L, Chen X, Zhang L, Yao J, Shao Z, Zhao B, Chen X, Liu Y. Enhanced Tissue Regeneration Through Immunomodulatory Extracellular Vesicles Encapsulated in Diversified Silk-Based Injectable Hydrogels. Adv Healthc Mater 2024; 13:e2401460. [PMID: 39011805 DOI: 10.1002/adhm.202401460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 06/26/2024] [Indexed: 07/17/2024]
Abstract
Mesenchymal stem cells (MSCs) immunologically trained using lipopolysaccharide (LPS) display enhanced immunomodulatory capabilities. Extracellular vesicles (EVs) derived from MSCs are widely used in regenerative medicine owing to their bioactive properties without the drawbacks of cell therapy. However, it remains unclear whether EVs derived from LPS-stimulated (trained) MSCs (L-EVs) inherit the enhanced reparative potential from their parent cells. Thus, this study first aims to explore the effect of immunological training on the bioactivity of L-EVs. LPS-trained bone marrow-derived MSCs (BMSCs) secrete more EVs, and these EVs significantly promote M2 macrophage polarization. Subsequently, hydrogel systems based on thixotropic injectable silk fibroin are prepared for in vivo EV delivery. These hydrogels have controllable gelation time and exhibit outstanding reparative effects on rat skin wounds and alveolar bone defects. Finally, it is revealed that L-EVs promote M2 macrophage polarization by inhibiting the nuclear translocation of PKM2. Overall, this study shows that the immunological training of BMSCs effectively improves the therapeutic effects of their EVs and provides a convenient and diversified EV delivery strategy using an injectable silk fibroin hydrogel. This strategy has broad clinical application prospects for tissue regeneration.
Collapse
Affiliation(s)
- Liangyan Sun
- Department of Orthodontics, Department of Macromolecular Science, Multidisciplinary Consultant Center, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200001, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, 200001, China
| | - Menglin Xiao
- Department of Orthodontics, Department of Macromolecular Science, Multidisciplinary Consultant Center, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200001, China
- State Key Laboratory of Molecular Engineering of Polymers, Laboratory of Advanced Materials, Fudan University, Shanghai, 200433, China
| | - Ling Chen
- Department of Orthodontics, Department of Macromolecular Science, Multidisciplinary Consultant Center, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200001, China
- State Key Laboratory of Molecular Engineering of Polymers, Laboratory of Advanced Materials, Fudan University, Shanghai, 200433, China
| | - Lingyue Ni
- Department of Orthodontics, Department of Macromolecular Science, Multidisciplinary Consultant Center, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200001, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, 200001, China
| | - Xiaoxuan Chen
- Department of Orthodontics, Department of Macromolecular Science, Multidisciplinary Consultant Center, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200001, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, 200001, China
| | - Lina Zhang
- Department of Biostatistics, Clinical Research Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jinrong Yao
- Department of Orthodontics, Department of Macromolecular Science, Multidisciplinary Consultant Center, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200001, China
- State Key Laboratory of Molecular Engineering of Polymers, Laboratory of Advanced Materials, Fudan University, Shanghai, 200433, China
| | - Zhengzhong Shao
- Department of Orthodontics, Department of Macromolecular Science, Multidisciplinary Consultant Center, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200001, China
- State Key Laboratory of Molecular Engineering of Polymers, Laboratory of Advanced Materials, Fudan University, Shanghai, 200433, China
| | - Bingjiao Zhao
- Department of Orthodontics, Department of Macromolecular Science, Multidisciplinary Consultant Center, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200001, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, 200001, China
| | - Xin Chen
- Department of Orthodontics, Department of Macromolecular Science, Multidisciplinary Consultant Center, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200001, China
- State Key Laboratory of Molecular Engineering of Polymers, Laboratory of Advanced Materials, Fudan University, Shanghai, 200433, China
| | - Yuehua Liu
- Department of Orthodontics, Department of Macromolecular Science, Multidisciplinary Consultant Center, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200001, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, 200001, China
| |
Collapse
|
41
|
Du R, Gao Y, Yan C, Ren X, Qi S, Liu G, Guo X, Song X, Wang H, Rao J, Zang Y, Zheng M, Li J, Huang H. Sirtuin 1/sirtuin 3 are robust lysine delactylases and sirtuin 1-mediated delactylation regulates glycolysis. iScience 2024; 27:110911. [PMID: 39351192 PMCID: PMC11440250 DOI: 10.1016/j.isci.2024.110911] [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: 04/01/2024] [Revised: 07/16/2024] [Accepted: 09/06/2024] [Indexed: 10/04/2024] Open
Abstract
Lysine lactylation (Kla), an epigenetic mark triggered by lactate during glycolysis, including the Warburg effect, bridges metabolism and gene regulation. Enzymes such as p300 and HDAC1/3 have been pivotal in deciphering the regulatory dynamics of Kla, though questions about additional regulatory enzymes, their specific Kla substrates, and the underlying functional mechanisms persist. Here, we identify SIRT1 and SIRT3 as key "erasers" of Kla, shedding light on their selective regulation of both histone and non-histone proteins. Proteomic analysis in SIRT1/SIRT3 knockout HepG2 cells reveals distinct substrate specificities toward Kla, highlighting their unique roles in cellular signaling. Notably, we highlight the role of specific Kla modifications, such as those on the M2 splice isoform of pyruvate kinase (PKM2), in modulating metabolic pathways and cell proliferation, thereby expanding Kla's recognized functions beyond epigenetics. Therefore, this study deepens our understanding of Kla's functional mechanisms and broadens its biological significance.
Collapse
Affiliation(s)
- Runhua Du
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yanmei Gao
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Cong Yan
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xuelian Ren
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Shankang Qi
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Guobin Liu
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xinlong Guo
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiaohan Song
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hanmin Wang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jingxin Rao
- State Key Laboratory of Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yi Zang
- Lingang Laboratory, Shanghai 201203, China
| | - Mingyue Zheng
- State Key Laboratory of Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jia Li
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - He Huang
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
42
|
Zheng C, Jiang L, Gong X, Zhang W, Pu R, Zhang Y, Zhao M, Jiang C, Wang H, Zhang P, Li Y. Cabozantinib-encapsulated and maytansine-conjugated high-density lipoprotein for immunotherapy in colorectal cancer. J Control Release 2024; 376:138-148. [PMID: 39362608 DOI: 10.1016/j.jconrel.2024.09.047] [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: 06/28/2024] [Revised: 09/24/2024] [Accepted: 09/27/2024] [Indexed: 10/05/2024]
Abstract
Advanced colorectal cancer (CRC) responds poorly to current adjuvant therapies, partially due to its immunosuppressive intestinal microenvironment. We found that myeloid-derived suppressor cells (MDSCs) were enriched in orthotopic tumors due to treatment-induced succinate release, which activated tuft cells and upregulated interleukin 25 (IL-25) and interleukin 13 (IL-13). We engineered a cabozantinib (Cabo)-encapsulated and maytansine (DM1)-conjugated synthetic high-density lipoprotein (ECCD-sHDL) to modulate the tumor microenvironment. DM1 induced immunogenic cell death and promoted the maturation of dendritic cells. Meanwhile, Cabo alleviated DM1-induced succinate release, preventing tuft cell activation, downregulating IL-25 and IL-13 secretion, and reducing intratumoral MDSC infiltration. ECCD-sHDL increased the densities of active cytotoxic T lymphocytes (CTLs) and M1 macrophages in the tumors, effectively inhibiting tumor growth and metastasis, thereby prolonging survival in murine CRC models. Our study sheds light on the mechanism of treatment-induced immunosuppression in orthotopic CRC and demonstrates that this combinatorial therapy could be an effective treatment for CRC.
Collapse
Affiliation(s)
- Chao Zheng
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai 201203, China.; State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, Shanghai 201203, China
| | - Linyang Jiang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiang Gong
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, Shanghai 201203, China
| | - Wen Zhang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, Shanghai 201203, China
| | - Rong Pu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yuan Zhang
- Department of Pulmonary and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Mengmeng Zhao
- Department of Pulmonary and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Chen Jiang
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai 201203, China..
| | - Hao Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai 201203, China.; National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, Shanghai 201203, China.
| | - Pengcheng Zhang
- School of Biomedical Engineering & State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China; Shanghai Clinical Research and Trial Center, Shanghai 201203, China.
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China; Nanjing University of Chinese Medicine, Nanjing 210023, China; Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China.
| |
Collapse
|
43
|
Xu Y, Han X, You S, Zhu W, Zhang M, Lu C, He J, Yao Q. SLC45A4 is involved in malignant progression of ovarian cancer through glycolytic metabolic reprogramming. Sci Rep 2024; 14:23031. [PMID: 39363015 PMCID: PMC11450204 DOI: 10.1038/s41598-024-74249-z] [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: 06/06/2024] [Accepted: 09/24/2024] [Indexed: 10/05/2024] Open
Abstract
Tumor cells promote malignant behaviors such as proliferation, invasion, and metastasis of cancer cells through glucose metabolic reprogramming, but the role of the H-dependent sugar cotransporter SLC45A4 in regulating metabolic reprogramming in ovarian cancer (OC) remains largely unknown. This study aimed to investigate the effects of SLC45A4 silencing on the transcriptome spectrum of ovarian cancer cells (OCC), glucose uptake, lactic acid production, intracellular ATP levels, and the expression and activity of HIF-α glycolysis signaling pathway. The results showed that SLC45A4 is overexpressed in OC and its elevated expression correlates with adverse clinical outcomes in OC patients. Silencing of SLC45A4 significantly inhibited the proliferation, invasion, and metastasis of OCC by suppressing glucose uptake and glycolysis, and it also reduced the expression of HIF-α glycolysis signaling pathway in OC tissues. In vivo experiments using shRNA to knock down SLC45A4 in xenograft models in nude mice demonstrated a significant inhibition of tumor growth. These findings suggest that SLC45A4 silencing can restrain the malignant progression of OC by inhibiting glucose uptake in OCC and affecting the reprogramming of glycolytic energy metabolism, indicating that SLC45A4 may serve as a potential therapeutic target for OC intervention.
Collapse
Affiliation(s)
- Yuance Xu
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Xiahui Han
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Shijing You
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Wei Zhu
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Mingyun Zhang
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Changyu Lu
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Junqi He
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Qin Yao
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
| |
Collapse
|
44
|
Jiang R, Li P, Meng E, Cheng X, Wu X, Wu H. Hsa_Circ_0008035 drives immune evasion of gastric cancer via promoting EXT1-mediated nuclear translocation of PKM2. Transl Oncol 2024; 48:102004. [PMID: 39053344 PMCID: PMC11325002 DOI: 10.1016/j.tranon.2024.102004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/15/2024] [Accepted: 05/17/2024] [Indexed: 07/27/2024] Open
Abstract
Circular RNAs (circRNAs) have been reported to be associated with the malignant phenotypes of cancer. However, the role and underlying mechanism of hsa_Circ_0008035 in colorectal cancer (CRC) remains unclear. In this study, we elucidated the pivotal role of hsa_circ_0008035 in gastric cancer progression and immune evasion. Elevated hsa_circ_0008035 levels in gastric cancer patient serum correlated positively with disease advancement, including tumor stages and lymph node metastasis. Functional analyses revealed a negative association between hsa_circ_0008035 and CD8+ T cell number and function. Mechanistically, hsa_circ_0008035 encoded the novel protein EXT1-219aa, suppressing EXT1 phosphorylation and expression. Additionally, hsa_circ_0008035 regulated pyruvate metabolism by influencing the nucleus localization of PKM2. The identified EXT1/PKM2 axis further underscored the intricate regulatory mechanisms orchestrated by hsa_circ_0008035 in gastric cancer, offering potential diagnostic and therapeutic implications in the ongoing pursuit of targeted therapies for gastric cancer patients.
Collapse
Affiliation(s)
- Rongqi Jiang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu Province, PR China
| | - Ping Li
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu Province, PR China
| | - Enqing Meng
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu Province, PR China
| | - Xu Cheng
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu Province, PR China
| | - Xinyi Wu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu Province, PR China
| | - Hao Wu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu Province, PR China.
| |
Collapse
|
45
|
Lemos FO, de Ridder I, Wagner L, Bootman MD, Bultynck G, Yule DI, Parys JB. Tetrameric, active PKM2 inhibits IP 3 receptors, potentially requiring GRP75 as an additional interaction partner. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119796. [PMID: 39038610 DOI: 10.1016/j.bbamcr.2024.119796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/05/2024] [Accepted: 07/08/2024] [Indexed: 07/24/2024]
Abstract
Pyruvate kinase M2 (PKM2) is a key glycolytic enzyme interacting with the inositol 1,4,5-trisphosphate receptor (IP3R). This interaction suppresses IP3R-mediated cytosolic [Ca2+] rises. As PKM2 exists in monomeric, dimeric and tetrameric forms displaying different properties including catalytic activity, we investigated the molecular determinants of PKM2 enabling its interaction with IP3Rs. Treatment of HeLa cells with TEPP-46, a compound stabilizing the tetrameric form of PKM2, increased both its catalytic activity and the suppression of IP3R-mediated Ca2+ signals. Consistently, in PKM2 knock-out HeLa cells, PKM2C424L, a tetrameric, highly active PKM2 mutant, but not inactive PKM2K270M or the less active PKM2K305Q, suppressed IP3R-mediated Ca2+ release. Surprisingly, however, in vitro assays did not reveal a direct interaction between purified PKM2 and either the purified Fragment 5 of IP3R1 (a.a. 1932-2216) or the therein located D5SD peptide (a.a. 2078-2098 of IP3R1), the presumed interaction sites of PKM2 on the IP3R. Moreover, on-nucleus patch clamp of heterologously expressed IP3R1 in DT40 cells devoid of endogenous IP3Rs did not reveal any functional effect of purified wild-type PKM2, mutant PKM2 or PKM1 proteins. These results indicate that an additional factor mediates the regulation of the IP3R by PKM2 in cellulo. Immunoprecipitation of GRP75 using HeLa cell lysates co-precipitated IP3R1, IP3R3 and PKM2. Moreover, the D5SD peptide not only disrupted PKM2:IP3R, but also PKM2:GRP75 and GRP75:IP3R interactions. Our data therefore support a model in which catalytically active, tetrameric PKM2 suppresses Ca2+ signaling via the IP3R through a multiprotein complex involving GRP75.
Collapse
Affiliation(s)
- Fernanda O Lemos
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, KU Leuven, Herestraat 49, Campus Gasthuisberg O&N1 - B802, 3000 Leuven, Belgium.
| | - Ian de Ridder
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, KU Leuven, Herestraat 49, Campus Gasthuisberg O&N1 - B802, 3000 Leuven, Belgium
| | - Larry Wagner
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642, USA
| | - Martin D Bootman
- School of Life, Health and Chemical Sciences, The Open University, Walton Hall, Milton Keynes MK7 6AA, UK
| | - Geert Bultynck
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, KU Leuven, Herestraat 49, Campus Gasthuisberg O&N1 - B802, 3000 Leuven, Belgium
| | - David I Yule
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642, USA
| | - Jan B Parys
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, KU Leuven, Herestraat 49, Campus Gasthuisberg O&N1 - B802, 3000 Leuven, Belgium.
| |
Collapse
|
46
|
Sha S, Jin N, Zhou R, Ruan Y, Ouyang Y. The Activation of PKM2 Induces Pyroptosis in Hippocampal Neurons via the NLRP3/Caspase-1/GSDMD Pathway in Neonatal Rats With Hypoxic-Ischemic Brain Injury. Brain Behav 2024; 14:e70108. [PMID: 39444090 PMCID: PMC11499207 DOI: 10.1002/brb3.70108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 08/27/2024] [Accepted: 10/05/2024] [Indexed: 10/25/2024] Open
Abstract
INTRODUCTION The presence of hypoxic-ischemic brain damage (HIBD) in neonates triggers a strong neuroinflammatory reaction. Pyroptosis, a programmed cell death mechanism associated with inflammation, plays a crucial role in HIBD. Pyruvate kinase M2 (PKM2) plays a significant role in connecting metabolic processes and inflammatory responses, but whether it affects hippocampus pyroptosis in HIBD is unclear. The aim of this study is to elucidate the role of PKM2 in HIBD and to propose a novel therapeutic approach for neonatal ischemic-hypoxic encephalopathy. METHODS In this study, we employed neonatal 7-day-old Sprague Dawley rats to establish a model of HIBD using the Rice-Vannucci surgical technique and a hypoxia device. To inhibit the elevation of PKM2, we utilized the PKM2 inhibitor shikonin. The rats were categorized into four groups: Sham, Shikonin, HIBD, and Shikonin + HIBD. Behavioral tests, hematoxylin eosin staining, immunofluorescence staining, ELISA (IL-1β, IL-18), and LDH were conducted in each group to evaluate neurological function, hippocampal damage, the occurrence of neuronal pyroptosis, and the neuroinflammation. Western blot was used to assess the expression levels of PKM2, NLRP3, Caspase-1, Cleaved Caspase-1, GSDMD, GSDMDN, and IL-1β. RESULTS The expression of PKM2 elevated in hippocampal tissues of the HIBD model and the localization of PKM2 in the hippocampus was activated in neurons instead of microglia during the HIBD. Meanwhile, the inhibition of PKM2 improved the behavioral test scores and the body weight of rats, the neuronal damage in the CA1 region of hippocampal tissue was also attenuated. In addition, inhibiting PKM2 alleviated neuronal pyroptosis by decreasing the expression of PKM2, NLRP3, Caspase-1, Cleaved Caspase-1, GSDMD, GSDMDN. Furthermore, serum levels of LDH and inflammatory factors IL-1β and IL-18 decrease with PKM2 inhibition. CONCLUSIONS Based on these findings, we can conclude that PKM2 plays a crucial role in regulating hippocampal neuronal pyroptosis of HIBD rats via NLRP3/Caspase-1/GSDMD pathway. Therefore, inhibiting PKM2 could be a promising therapeutic strategy for the treatment of neonatal ischemic-hypoxic encephalopathy.
Collapse
Affiliation(s)
- Sha Sha
- Sun Yat‐sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouChina
| | - Ni Jin
- Sun Yat‐sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouChina
| | - Ruiyu Zhou
- The Fifth Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
| | - Yanghao Ruan
- The Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdongChina
| | - Ying Ouyang
- Sun Yat‐sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouChina
| |
Collapse
|
47
|
Jemal M, Getinet M, Amare GA, Tegegne BA, Baylie T, Mengistu EF, Osman EE, Chura Waritu N, Adugna A. Non-metabolic enzyme function of pyruvate kinase M2 in breast cancer. Front Oncol 2024; 14:1450325. [PMID: 39411137 PMCID: PMC11473492 DOI: 10.3389/fonc.2024.1450325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 09/12/2024] [Indexed: 10/19/2024] Open
Abstract
Breast cancer (BC) is a prevalent malignant tumor in women, and its incidence has been steadily increasing in recent years. Compared with other types of cancer, it has the highest mortality and morbidity rates in women. So, it is crucial to investigate the underlying mechanisms of BC development and identify specific therapeutic targets. Pyruvate kinase M2 (PKM2), an important metabolic enzyme in glycolysis, has been found to be highly expressed in BC. It can also move to the nucleus and interact with various transcription factors and proteins, including hypoxia-inducible factor-1α (HIF-1α), signal transducer and activator of transcription 3 (STAT3), β-catenin, cellular-myelocytomatosis oncogene (c-Myc), nuclear factor kappa-light-chain enhancer of activated B cells (NF-κB), and mammalian sterile 20-like kinase 1 (MST1). This interaction leads to non-metabolic functions that control the cell cycle, proliferation, apoptosis, migration, invasion, angiogenesis, and tumor microenvironment in BC. This review provides an overview of the latest advancements in understanding the interactions between PKM2 and different transcription factors and proteins that influence the initiation and progression of BC. It also examined how natural drugs and noncoding RNAs affect various biological processes in BC cells through the regulation of the non-metabolic enzyme functions of PKM2. The findings provide valuable insights for improving the prognosis and developing targeted therapies for BC in the coming years.
Collapse
Affiliation(s)
- Mohammed Jemal
- Department of Biomedical Science, School of Medicine, Debre Markos University, Debre Markos, Ethiopia
| | - Mamaru Getinet
- Department of Biomedical Science, School of Medicine, Debre Markos University, Debre Markos, Ethiopia
| | - Gashaw Azanaw Amare
- Department of Medical Laboratory Sciences, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Bantayehu Addis Tegegne
- Department of Pharmacy, College of Medicine and Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Temesgen Baylie
- Department of Biomedical Science, School of Medicine, Debre Markos University, Debre Markos, Ethiopia
| | - Enyew Fenta Mengistu
- Department of Biomedical Science, School of Medicine, Debre Markos University, Debre Markos, Ethiopia
| | - Enatnesh Essa Osman
- Department of Biomedical Science, School of Medicine, Debre Markos University, Debre Markos, Ethiopia
| | - Nuredin Chura Waritu
- Department of Biomedical Sciences, School of Medicine, Wolaita Sodo University, Wolaita Sodo, Ethiopia
| | - Adane Adugna
- Department of Medical Laboratory Sciences, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| |
Collapse
|
48
|
Lu H, Zhu Z, Fields L, Zhang H, Li L. Mass Spectrometry Structural Proteomics Enabled by Limited Proteolysis and Cross-Linking. MASS SPECTROMETRY REVIEWS 2024. [PMID: 39300771 DOI: 10.1002/mas.21908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 08/31/2024] [Accepted: 09/02/2024] [Indexed: 09/22/2024]
Abstract
The exploration of protein structure and function stands at the forefront of life science and represents an ever-expanding focus in the development of proteomics. As mass spectrometry (MS) offers readout of protein conformational changes at both the protein and peptide levels, MS-based structural proteomics is making significant strides in the realms of structural and molecular biology, complementing traditional structural biology techniques. This review focuses on two powerful MS-based techniques for peptide-level readout, namely limited proteolysis-mass spectrometry (LiP-MS) and cross-linking mass spectrometry (XL-MS). First, we discuss the principles, features, and different workflows of these two methods. Subsequently, we delve into the bioinformatics strategies and software tools used for interpreting data associated with these protein conformation readouts and how the data can be integrated with other computational tools. Furthermore, we provide a comprehensive summary of the noteworthy applications of LiP-MS and XL-MS in diverse areas including neurodegenerative diseases, interactome studies, membrane proteins, and artificial intelligence-based structural analysis. Finally, we discuss the factors that modulate protein conformational changes. We also highlight the remaining challenges in understanding the intricacies of protein conformational changes by LiP-MS and XL-MS technologies.
Collapse
Affiliation(s)
- Haiyan Lu
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Zexin Zhu
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Lauren Fields
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Hua Zhang
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Lingjun Li
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Lachman Institute for Pharmaceutical Development, School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
49
|
Zhang L, Li M, Li X, Xiao T, Zhou H, Zhang W, Wang P. Deciphering the role of PLCD3 in lung cancer: A gateway to glycolytic reprogramming via PKC-Rap1 activation. Heliyon 2024; 10:e37063. [PMID: 39296221 PMCID: PMC11408031 DOI: 10.1016/j.heliyon.2024.e37063] [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: 05/13/2024] [Revised: 08/21/2024] [Accepted: 08/27/2024] [Indexed: 09/21/2024] Open
Abstract
PLCD3 belongs to the phospholipase C delta group and is involved in numerous biological functions, including cell growth, programmed cell death, and specialization. However, the role of PLCD3 in lung cancer still needs further investigation. This research aimed to investigate if PLCD3 influences glycolytic reprogramming and lung cancer development through the PKC-dependent Rap1 signaling pathway. This study found that PLCD3 was increased in lung cancer tissues. PLCD3 promotes the proliferation and invasion of lung cancer cells by activating the PKC-dependent Rap1 pathway. The detailed process involves PLCD3 triggering PKC, which subsequently stimulates the Rap1 pathway, leading to glycolytic reprogramming that supplies adequate energy and metabolic substrates necessary for the growth and spread of lung cancer cells. Moreover, PLCD3 can also promote the metastasis and invasion of lung cancer cells by activating the Rap1 pathway. This study reveals the mechanism of PLCD3 in lung cancer and provides new ideas for the treatment of lung cancer. Inhibiting PLCD3, PKC, and the Rap1 pathway may be an effective strategy for treating lung cancer.
Collapse
Affiliation(s)
- Liang Zhang
- Tianjin Medical University Cancer Institute & Hospital, Tianjin, PR China
- Department of Thoracic Surgery, Tianjin First Central Hospital, Tianjin, 300192, PR China
| | - Mingjiang Li
- Department of Thoracic Surgery, Tianjin First Central Hospital, Tianjin, 300192, PR China
| | - Xiaoping Li
- Department of Thoracic Surgery, Tianjin First Central Hospital, Tianjin, 300192, PR China
| | - Ting Xiao
- College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, PR China
| | - Honggang Zhou
- College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, PR China
| | - Weidong Zhang
- Department of Thoracic Surgery, Tianjin First Central Hospital, Tianjin, 300192, PR China
| | - Ping Wang
- Tianjin Medical University Cancer Institute & Hospital, Tianjin, PR China
| |
Collapse
|
50
|
Rong J, Wang Q, Li T, Qian J, Cheng J. Glucose metabolism in glioma: an emerging sight with ncRNAs. Cancer Cell Int 2024; 24:316. [PMID: 39272133 PMCID: PMC11395608 DOI: 10.1186/s12935-024-03499-8] [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: 03/05/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024] Open
Abstract
Glioma is a primary brain tumor that grows quickly, has an unfavorable prognosis, and can spread intracerebrally. Glioma cells rely on glucose as the major energy source, and glycolysis plays a critical role in tumorigenesis and progression. Substrate utilization shifts throughout glioma progression to facilitate energy generation and biomass accumulation. This metabolic reprogramming promotes glioma cell proliferation and metastasis and ultimately decreases the efficacy of conventional treatments. Non-coding RNAs (ncRNAs) are involved in several glucose metabolism pathways during tumor initiation and progression. These RNAs influence cell viability and glucose metabolism by modulating the expression of key genes of the glycolytic pathway. They can directly or indirectly affect glycolysis in glioma cells by influencing the transcription and post-transcriptional regulation of oncogenes and suppressor genes. In this review, we discussed the role of ncRNAs in the metabolic reprogramming of glioma cells and tumor microenvironments and their abnormal expression in the glucometabolic pathway in glioma. In addition, we consolidated the existing theoretical knowledge to facilitate the use of this emerging class of biomarkers as biological indicators and potential therapeutic targets for glioma.
Collapse
Affiliation(s)
- Jun Rong
- Department of Neurosurgery, Xuancheng People's Hospital, The Affiliated Xuancheng Hospital of Wannan Medical College, Xuancheng, People's Republic of China
| | - Qifu Wang
- Department of Neurosurgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), WuHu, People's Republic of China
| | - Tingzheng Li
- Department of Neurosurgery, Xuancheng Central Hospital, Xuancheng, People's Republic of China
| | - Jin Qian
- Department of Neurosurgery, Xuancheng People's Hospital, The Affiliated Xuancheng Hospital of Wannan Medical College, Xuancheng, People's Republic of China.
| | - Jinchao Cheng
- Department of Neurosurgery, Xuancheng Central Hospital, Xuancheng, People's Republic of China.
| |
Collapse
|