1
|
Upadhyay S, Bhardwaj M, Kumar SP, Khan S, Kumar A, Hassan MI. Impact of Cancer-Associated PKM2 Mutations on Enzyme Activity and Allosteric Regulation: Structural and Functional Insights into Metabolic Reprogramming. Biochemistry 2025; 64:1463-1475. [PMID: 40080100 DOI: 10.1021/acs.biochem.5c00009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
Mammalian pyruvate kinase M2 (PKM2) is a key regulator of glycolysis and is highly expressed in proliferative tissues including tumors. Mutations in PKM2 have been identified in various cancers, but their effects on enzyme activity and regulation are not fully understood. This study investigates the structural and functional effects of cancer-associated PKM2 mutations on enzyme kinetics, allosteric regulation, and oligomerization. Using computational modeling, X-ray crystallography, and biochemical assays, we demonstrated how these mutations impact PKM2 activity, substrate binding, and allosteric activation via fructose-1,6-bisphosphate (FBP), contributing to altered enzyme function. In this study, we characterized four cancer-associated PKM2 mutations (P403A, C474S, R516C, and L144P) using computational, structural, and biochemical approaches. Computational modeling revealed disruptions in allosteric signaling pathways, particularly affecting the communication between regulatory sites and the active site. X-ray crystallography demonstrated local conformational changes in the hinge and FBP-binding regions, leading to a shift from the active tetrameric state to a less active dimeric state, particularly in the C474S and R516C mutants. The mutants exhibited reduced maximal velocity, reduced substrate affinity, and altered activation by the allosteric activator fructose-1,6-bisphosphate (FBP). Under alkaline pH conditions, mimicking the tumor microenvironment, these mutations further destabilized the PKM2 oligomeric state, favoring the formation of lower-order species. Our findings suggest that PKM2 is highly sensitive to mutations, and these alterations may contribute to metabolic reprogramming in cancer cells by impairing its enzymatic regulation.
Collapse
Affiliation(s)
- Saurabh Upadhyay
- Kusuma School of Biological Sciences, Indian Institute of Technology, New Delhi 110016, India
| | - Mohit Bhardwaj
- Kusuma School of Biological Sciences, Indian Institute of Technology, New Delhi 110016, India
| | - Sivakumar Prasanth Kumar
- Department of Botany, Bioinformatics and Climate Change Impacts Management, School of Sciences, Gujarat University, Ahmedabad 380009, India
| | - Shumayila Khan
- Kusuma School of Biological Sciences, Indian Institute of Technology, New Delhi 110016, India
- International Health Division, Indian Council of Medical Research, Ansari Nagar, New Delhi 110029, India
| | - Ashwani Kumar
- Macromolecular Crystallography Section, Beamline Development & Application Section, Bhabha Atomic Research Center, Trombay, Mumbai 400085, India
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| |
Collapse
|
2
|
Zhang Y, Xu Y, Zhang Y, Wang S, Zhao M. The multiple functions and mechanisms of long non-coding RNAs in regulating breast cancer progression. Front Pharmacol 2025; 16:1559408. [PMID: 40223929 PMCID: PMC11985786 DOI: 10.3389/fphar.2025.1559408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 03/14/2025] [Indexed: 04/15/2025] Open
Abstract
Breast cancer (BC) is a malignant tumor that has the highest morbidity and mortality rates in the female population, and its high tendency to metastasize is the main cause of poor clinical prognosis. Long non-coding RNAs (lncRNAs) have been extensively documented to exhibit aberrant expression in various cancers and influence tumor progression via multiple molecular pathways. These lncRNAs not only modulate numerous aspects of gene expression in cancer cells, such as transcription, translation, and post-translational modifications, but also play a crucial role in the reprogramming of energy metabolism by regulating metabolic regulators, which is particularly significant in advanced BC. This review examines the characteristics and mechanisms of lncRNAs in regulating BC cells, both intracellularly (e.g., cell cycle, autophagy) and extracellularly (e.g., tumor microenvironment). Furthermore, we explore the potential of specific lncRNAs and their regulatory factors as molecular markers and therapeutic targets. Lastly, we summarize the application of lncRNAs in the treatment of advanced BC, aiming to offer novel personalized therapeutic options for patients.
Collapse
Affiliation(s)
- Yongsheng Zhang
- Qingdao Medical College, Qingdao University, Qingdao, Shandong, China
- Department of Anesthesia and Perioperative Medicine, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| | - Yanjiao Xu
- Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yanping Zhang
- Department of Anesthesia and Perioperative Medicine, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| | - Shoushi Wang
- Department of Anesthesia and Perioperative Medicine, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| | - Mingqiang Zhao
- Department of Anesthesia and Perioperative Medicine, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| |
Collapse
|
3
|
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
|
4
|
Wu M, Jia G, Liu Y, Lou Y, Li Y, Xia M, Li H, Li W. PKM2 controls cochlear development through lactate-dependent transcriptional regulation. Proc Natl Acad Sci U S A 2025; 122:e2410829122. [PMID: 39773029 PMCID: PMC11745320 DOI: 10.1073/pnas.2410829122] [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: 05/31/2024] [Accepted: 11/25/2024] [Indexed: 01/11/2025] Open
Abstract
Understanding the role of metabolic processes during inner ear development is essential for identifying targets for hair cell (HC) regeneration, as metabolic choices play a crucial role in cell proliferation and differentiation. Among the metabolic processes, growing evidence shows that glucose metabolism is closely related to organ development. However, the role of glucose metabolism in mammalian inner ear development and HC regeneration remains unclear. In this study, we found that glycolytic metabolism is highly active during mouse and human cochlear prosensory epithelium expansion. Using mouse cochlear organoids, we revealed that glycolytic activity in cochlear nonsensory epithelial cells was predominantly dominated by pyruvate kinase M2 (PKM2). Deletion of PKM2 induced a metabolic switch from glycolysis to oxidative phosphorylation, impairing cochlear organoid formation. Furthermore, conditional loss of PKM2 in cochlear progenitors hindered sensory epithelium morphogenesis, as demonstrated in PKM2 knockout mice. Mechanistically, pyruvate is generated by PKM2 catalysis and then converted into lactate, which then lactylates histone H3, regulating the transcription of key genes for cochlear development. Specifically, accumulated lactate causes histone H3 lactylation at lysine 9 (H3K9la), upregulating the expression of Sox family transcription factors through epigenetic modification. Moreover, overexpression of PKM2 in supporting cells (SCs) triggered metabolism reprogramming and enhanced HC generation in cultured mouse and human cochlear explants. Our findings uncover a molecular mechanism of sensory epithelium formation driven by glycolysis-lactate flow and suggest unique approaches for mammalian HC regeneration.
Collapse
Affiliation(s)
- Mingxuan Wu
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai200031, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai200032, China
| | - Gaogan Jia
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai200031, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai200032, China
| | - Yaoqian Liu
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai200031, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai200032, China
| | - Yiyun Lou
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai200031, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai200032, China
| | - Yunjie Li
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai200031, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai200032, China
| | - Mingyu Xia
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai200031, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai200031, China
- The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai200032, China
| | - Huawei Li
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai200031, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai200031, China
- The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai200032, China
- Shanghai Engineering Research Centre of Cochlear Implant, Shanghai200031, China
| | - Wenyan Li
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai200031, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai200031, China
- The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai200032, China
- Shanghai Engineering Research Centre of Cochlear Implant, Shanghai200031, China
| |
Collapse
|
5
|
Liu F, Li C. SIRT1-driven mechanism: sevoflurane's interference with mESC neural differentiation via PRRX1/DRD2 cascade. Hum Mol Genet 2024; 33:1758-1770. [PMID: 39087769 DOI: 10.1093/hmg/ddae099] [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: 12/12/2023] [Revised: 05/13/2024] [Accepted: 06/03/2024] [Indexed: 08/02/2024] Open
Abstract
Investigating the sevoflurane-induced perturbation in the differentiation of mouse embryonic stem cells (mESCs) into neural stem cells (mNSCs), our study delineates a novel SIRT1/PRRX1/DRD2/PKM2/NRF2 axis as a key player in this intricate process. Sevoflurane treatment hindered mESC differentiation, evidenced by altered expression patterns of pluripotency and neural lineage markers. Mechanistically, sevoflurane downregulated Sirt1, setting in motion a signaling cascade. Sevoflurane may inhibit PKM2 dimerization and NRF2 signaling pathway activation by inhibiting the expression of SIRT1 and its downstream genes Prrx1 and DRD2, ultimately inhibiting mESCs differentiation into mNSCs. These findings contribute to our understanding of the molecular basis of sevoflurane-induced neural toxicity, presenting a potential avenue for therapeutic intervention in sevoflurane-induced perturbation in the differentiation of mESCs into mNSCs by modulating the SIRT1/PRRX1/DRD2/PKM2/NRF2 axis.
Collapse
Affiliation(s)
- Feifei Liu
- Department of Anesthesiology, The First Affiliated Hospital of Jinzhou Medical University, No. 2, Section 5, Renmin Street, Jinzhou 121000, P. R. China
| | - Chenguang Li
- Department of Neurosurgery, The First Affiliated Hospital of Jinzhou Medical University, No. 2, Section 5, Renmin Street, Jinzhou 121000, P. R. China
| |
Collapse
|
6
|
Zheng X, Zhang S, Ma H, Dong Y, Zheng J, Zeng L, Liu J, Dai Y, Yin Q. Replenishment of TCA cycle intermediates and long-noncoding RNAs regulation in breast cancer. Mol Cell Endocrinol 2024; 592:112321. [PMID: 38936596 DOI: 10.1016/j.mce.2024.112321] [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: 12/02/2023] [Revised: 03/13/2024] [Accepted: 06/24/2024] [Indexed: 06/29/2024]
Abstract
The tricarboxylic acid (TCA) cycle is an essential interface that coordinates cellular metabolism and is as a primary route determining the fate of a variety of fuel sources, including glucose, fatty acid and glutamate. The crosstalk of nutrients replenished TCA cycle regulates breast cancer (BC) progression by changing substrate levels-induced epigenetic alterations, especially the methylation, acetylation, succinylation and lactylation. Long non-coding RNAs (lncRNA) have dual roles in inhibiting or promoting energy reprogramming, and so altering the metabolic flux of fuel sources to the TCA cycle, which may regulate epigenetic modifications at the cellular level of BC. This narrative review discussed the central role of the TCA cycle in interconnecting numerous fuels and the induced epigenetic modifications, and the underlying regulatory mechanisms of lncRNAs in BC.
Collapse
Affiliation(s)
- Xuewei Zheng
- Precision Medicine Laboratory, School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| | - ShunShun Zhang
- Precision Medicine Laboratory, School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| | - HaoDi Ma
- Precision Medicine Laboratory, School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| | - Yirui Dong
- Precision Medicine Laboratory, School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| | - Jiayu Zheng
- Precision Medicine Laboratory, School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| | - Li Zeng
- Precision Medicine Laboratory, School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| | - Jiangbo Liu
- Department of General Surgery, First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, China
| | - Yanzhenzi Dai
- Animal Science, School of Biosciences, University of Nottingham, UK.
| | - Qinan Yin
- Precision Medicine Laboratory, School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China.
| |
Collapse
|
7
|
Zhang X, Lei Y, Zhou H, Liu H, Xu P. The Role of PKM2 in Multiple Signaling Pathways Related to Neurological Diseases. Mol Neurobiol 2024; 61:5002-5026. [PMID: 38157121 DOI: 10.1007/s12035-023-03901-y] [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: 09/09/2023] [Accepted: 12/18/2023] [Indexed: 01/03/2024]
Abstract
Pyruvate kinase M2 (PKM2) is a key rate-limiting enzyme in glycolysis. It is well known that PKM2 plays a vital role in the proliferation of tumor cells. However, PKM2 can also exert its biological functions by mediating multiple signaling pathways in neurological diseases, such as Alzheimer's disease (AD), cognitive dysfunction, ischemic stroke, post-stroke depression, cerebral small-vessel disease, hypoxic-ischemic encephalopathy, traumatic brain injury, spinal cord injury, Parkinson's disease (PD), epilepsy, neuropathic pain, and autoimmune diseases. In these diseases, PKM2 can exert various biological functions, including regulation of glycolysis, inflammatory responses, apoptosis, proliferation of cells, oxidative stress, mitochondrial dysfunction, or pathological autoimmune responses. Moreover, the complexity of PKM2's biological characteristics determines the diversity of its biological functions. However, the role of PKM2 is not entirely the same in different diseases or cells, which is related to its oligomerization, subcellular localization, and post-translational modifications. This article will focus on the biological characteristics of PKM2, the regulation of PKM2 expression, and the biological role of PKM2 in neurological diseases. With this review, we hope to have a better understanding of the molecular mechanisms of PKM2, which may help researchers develop therapeutic strategies in clinic.
Collapse
Affiliation(s)
- Xiaoping Zhang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yihui Lei
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Hongyan Zhou
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Haijun Liu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Ping Xu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| |
Collapse
|
8
|
Dai E, Wang W, Li Y, Ye D, Li Y. Lactate and lactylation: Behind the development of tumors. Cancer Lett 2024; 591:216896. [PMID: 38641309 DOI: 10.1016/j.canlet.2024.216896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/13/2024] [Accepted: 04/11/2024] [Indexed: 04/21/2024]
Abstract
There is growing evidence that lactate can have a wide range of biological impacts in addition to being a waste product of metabolism. Because of the Warburg effect, tumors generate lots of lactate, which create a tumor microenvironment (TME) with low nutrition, hypoxia, and low pH. As a result, the immunosuppressive network is established to gain immune escape potential and regulate tumor growth. Consequently, the tumor lactate pathway is emerging as a possible therapeutic target for tumor. Importantly, Zhao et al. first discovered histone lysine lactylation (Kla) in 2019, which links gene regulation to cell metabolism through dysmetabolic activity and epigenetic modifications, influencing TME and tumor development. Therefore, the aim of this paper is to explore the effects of lactate and lactylation on the TME and tumors, and provide theoretical basis for further research on potential therapeutic targets and biomarkers, with the view to providing new ideas and methods for tumor treatment and prognosis evaluation.
Collapse
Affiliation(s)
- Enci Dai
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 650 Xinsongjiang Road, Shanghai, 201600, China.
| | - Wei Wang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 650 Xinsongjiang Road, Shanghai, 201600, China.
| | - Yingying Li
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 650 Xinsongjiang Road, Shanghai, 201600, China.
| | - Defeng Ye
- Department of Thoracic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Hongkou District, Shanghai, 200080, China.
| | - Yanli Li
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 650 Xinsongjiang Road, Shanghai, 201600, China.
| |
Collapse
|
9
|
Wu B, Liang Z, Lan H, Teng X, Wang C. The role of PKM2 in cancer progression and its structural and biological basis. J Physiol Biochem 2024; 80:261-275. [PMID: 38329688 DOI: 10.1007/s13105-024-01007-0] [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: 06/20/2023] [Accepted: 01/10/2024] [Indexed: 02/09/2024]
Abstract
Pyruvate kinase M2 (PKM2), a subtype of pyruvate kinase (PK), has been shown to play an important role in the development of cancer. It regulates the last step of glycolytic pathway. PKM2 has both pyruvate kinase and protein kinase activity, and the conversion of these two functions of PKM2 depends on the mutual change of dimer and tetramer. The dimerization of PKM2 can promote the proliferation and growth of tumor cells, so inhibiting the dimerization of PKM2 is essential to curing cancer. The aggregation of PKM2 is regulated by both endogenous and exogenous cofactors as well as post-translational modification (PTM). Although there are many studies on the different aggregation of PKM2 in the process of tumor development, there are few summaries in recent years. In this review, we first introduce the role of PKM2 in various biological processes of tumor growth. Then, we summarize the aggregation regulation mechanism of PKM2 by various endogenous cofactors such as Fructose-1, 6-diphosphate (FBP), various amino acids, and post-translational modification (PTMs). Finally, the related inhibitors and agonists of PKM2 are summarized to provide reference for regulating PKM2 aggregation in the treatment of cancer in the future.
Collapse
Affiliation(s)
- Bingxin Wu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Zuhui Liang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Huan Lan
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Xiaojun Teng
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Caiyan Wang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| |
Collapse
|
10
|
Qian J, Huang C, Wang M, Liu Y, Zhao Y, Li M, Zhang X, Gao X, Zhang Y, Wang Y, Huang J, Li J, Zhou Q, Liu R, Wang X, Cui J, Yang Y. Nuclear translocation of metabolic enzyme PKM2 participates in high glucose-promoted HCC metastasis by strengthening immunosuppressive environment. Redox Biol 2024; 71:103103. [PMID: 38471282 PMCID: PMC10945175 DOI: 10.1016/j.redox.2024.103103] [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: 01/18/2024] [Revised: 02/14/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Although some cohort studies have indicated a close association between diabetes and HCC, the underlying mechanism about the contribution of diabetes to HCC progression remains largely unknown. In the study, we applied a novel HCC model in SD rat with diabetes and a series of high glucose-stimulated cell experiments to explore the effect of a high glucose environment on HCC metastasis and its relevant mechanism. Our results uncovered a novel regulatory mechanism by which nuclear translocation of metabolic enzyme PKM2 mediated high glucose-promoted HCC metastasis. Specifically, high glucose-increased PKM2 nuclear translocation downregulates chemerin expression through the redox protein TRX1, and then strengthens immunosuppressive environment to promote HCC metastasis. To the best of our knowledge, this is the first report to elucidate the great contribution of a high glucose environment to HCC metastasis from a new perspective of enhancing the immunosuppressive microenvironment. Simultaneously, this work also highlights a previously unidentified non-metabolic role of PKM2 and opens a novel avenue for cross research and intervention for individuals with HCC and comorbid diabetes.
Collapse
Affiliation(s)
- Jiali Qian
- Department of endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Chuxin Huang
- Department of endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Mimi Wang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Ying Liu
- Department of endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yingying Zhao
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Miao Li
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Xi Zhang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Xiangyu Gao
- Department of endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yawen Zhang
- Department of endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yi Wang
- Department of endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jinya Huang
- Department of endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiajun Li
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Qiwen Zhou
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Rui Liu
- Department of endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xuanchun Wang
- Department of endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiefeng Cui
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China.
| | - Yehong Yang
- Department of endocrinology, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
11
|
Yin L, Shi J, Zhang J, Lin X, Jiang W, Zhu Y, Song Y, Lu Y, Ma Y. PKM2 is a potential prognostic biomarker and related to immune infiltration in lung cancer. Sci Rep 2023; 13:22243. [PMID: 38097666 PMCID: PMC10721847 DOI: 10.1038/s41598-023-49558-4] [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: 10/09/2023] [Accepted: 12/09/2023] [Indexed: 12/17/2023] Open
Abstract
Pyruvate kinase M2 (PKM2), a subtype of pyruvate kinase, plays a crucial role as a key enzyme in the final step of glycolysis. It is involved in regulating the tumor microenvironment and accelerating tumor progression. However, the relationship between PKM2 expression and the prognosis and immune infiltration remains unclear in lung cancer. In this study, we analyzed PKM2 expression in pan-cancer, and investigated its association with prognosis and immune cell infiltration of lung cancer by using multiple online databases, including Gent2, Tumor Immune Estimation Resource (TIMER), Gene Expression Profiling Interactive Analysis (GEPIA), PrognoScan, Kaplan-Meier plotter, and The Human Protein Atlas (HPA). The results showed that PKM2 expression is elevated in tumor tissues compared with the adjacent normal tissues of most cancers, including lung cancer. Prognostic analysis indicated that high expression of PKM2 was associated with poorer prognosis in overall lung cancer patients, especially in lung adenocarcinoma (LUAD). Notably, PKM2 exhibited a strong correlation with B cells and CD4+ T cells in LUAD; and with B cells, CD8+ T cells, CD4+ cells, and macrophages in lung squamous cell carcinoma (LUSC). Furthermore, PKM2 expression displayed a significant negative correlation with the expression of immune cell markers in both LUAD and LUSC. These findings suggested that PKM2 could serve as a promising prognostic biomarker for lung cancer and provided insights into its essential role in modulating the immune cell infiltration.
Collapse
Affiliation(s)
- Lan Yin
- Department of Medical Genetics, West China Hospital, Sichuan University, Chengdu, China
| | - Jiaying Shi
- Department of Medical Genetics, West China Hospital, Sichuan University, Chengdu, China
| | - Jingfei Zhang
- Department of Medical Genetics, West China Hospital, Sichuan University, Chengdu, China
| | - Xinyu Lin
- Department of Medical Genetics, West China Hospital, Sichuan University, Chengdu, China
| | - Wenhao Jiang
- Department of Medical Genetics, West China Hospital, Sichuan University, Chengdu, China
| | - Yingchuan Zhu
- Department of Medical Genetics, West China Hospital, Sichuan University, Chengdu, China
| | - Yue Song
- Department of Medical Genetics, West China Hospital, Sichuan University, Chengdu, China
| | - Yilu Lu
- Department of Medical Genetics, West China Hospital, Sichuan University, Chengdu, China
| | - Yongxin Ma
- Department of Medical Genetics, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
12
|
Wei Y, Miao Q, Zhang Q, Mao S, Li M, Xu X, Xia X, Wei K, Fan Y, Zheng X, Fang Y, Mei M, Zhang Q, Ding J, Fan Y, Lu M, Hu G. Aerobic glycolysis is the predominant means of glucose metabolism in neuronal somata, which protects against oxidative damage. Nat Neurosci 2023; 26:2081-2089. [PMID: 37996529 DOI: 10.1038/s41593-023-01476-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 09/29/2023] [Indexed: 11/25/2023]
Abstract
It is generally thought that under basal conditions, neurons produce ATP mainly through mitochondrial oxidative phosphorylation (OXPHOS), and glycolytic activity only predominates when neurons are activated and need to meet higher energy demands. However, it remains unknown whether there are differences in glucose metabolism between neuronal somata and axon terminals. Here, we demonstrated that neuronal somata perform higher levels of aerobic glycolysis and lower levels of OXPHOS than terminals, both during basal and activated states. We found that the glycolytic enzyme pyruvate kinase 2 (PKM2) is localized predominantly in the somata rather than in the terminals. Deletion of Pkm2 in mice results in a switch from aerobic glycolysis to OXPHOS in neuronal somata, leading to oxidative damage and progressive loss of dopaminergic neurons. Our findings update the conventional view that neurons uniformly use OXPHOS under basal conditions and highlight the important role of somatic aerobic glycolysis in maintaining antioxidant capacity.
Collapse
Affiliation(s)
- Yao Wei
- Department of Pharmacology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - QianQian Miao
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Qian Zhang
- Department of Pharmacology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shiyu Mao
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Mengke Li
- Department of Pharmacology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xing Xu
- Department of Pharmacology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xian Xia
- Department of Pharmacology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ke Wei
- Department of Pharmacology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yu Fan
- Department of Pharmacology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xinlei Zheng
- Department of Pharmacology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yinquan Fang
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Meng Mei
- Department of Pharmacology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qingyu Zhang
- Department of Pharmacology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jianhua Ding
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Yi Fan
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Ming Lu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Gang Hu
- Department of Pharmacology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
13
|
Babu VS, Mallipatna A, Dudeja G, Shetty R, Nair AP, Tun SBB, Ho CEH, Chaurasia SS, Bhattacharya SS, Verma NK, Lakshminarayanan R, Guha N, Heymans S, Barathi VA, Ghosh A. Depleted hexokinase1 and lack of AMPKα activation favor OXPHOS-dependent energetics in retinoblastoma tumors. Transl Res 2023; 261:41-56. [PMID: 37419277 DOI: 10.1016/j.trsl.2023.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 06/03/2023] [Accepted: 07/02/2023] [Indexed: 07/09/2023]
Abstract
Lack of retinoblastoma (Rb) protein causes aggressive intraocular retinal tumors in children. Recently, Rb tumors have been shown to have a distinctly altered metabolic phenotype, such as reduced expression of glycolytic pathway proteins alongside altered pyruvate and fatty acid levels. In this study, we demonstrate that loss of hexokinase 1(HK1) in tumor cells rewires their metabolism allowing enhanced oxidative phosphorylation-dependent energy production. We show that rescuing HK1 or retinoblastoma protein 1 (RB1) in these Rb cells reduced cancer hallmarks such as proliferation, invasion, and spheroid formation and increased their sensitivity to chemotherapy drugs. Induction of HK1 was accompanied by a metabolic shift of the cells to glycolysis and a reduction in mitochondrial mass. Cytoplasmic HK1 bound Liver Kinase B1 and phosphorylated AMP-activated kinase-α (AMPKα Thr172), thereby reducing mitochondria-dependent energy production. We validated these findings in tumor samples from Rb patients compared to age-matched healthy retinae. HK1 or RB1 expression in Rb-/- cells led to a reduction in their respiratory capacity and glycolytic proton flux. HK1 overexpression reduced tumor burden in an intraocular tumor xenograft model. AMPKα activation by AICAR also enhanced the tumoricidal effects of the chemotherapeutic drug topotecan in vivo. Therefore, enhancing HK1 or AMPKα activity can reprogram cancer metabolism and sensitize Rb tumors to lower doses of existing treatments, a potential therapeutic modality for Rb.
Collapse
Affiliation(s)
- Vishnu Suresh Babu
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, India; Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Ashwin Mallipatna
- Retinoblastoma Service, Narayana Nethralaya, Bangalore, Karnataka, India
| | - Gagan Dudeja
- Retinoblastoma Service, Narayana Nethralaya, Bangalore, Karnataka, India
| | - Rohit Shetty
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, India
| | | | | | | | - Shyam S Chaurasia
- Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Shomi S Bhattacharya
- University College London, London, UK; GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, India
| | - Navin Kumar Verma
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore; Singapore Eye Research Institute, Singapore
| | | | - Nilanjan Guha
- Agilent Technologies India Pvt Ltd, New Delhi, Delhi, India
| | - Stephane Heymans
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands; Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology, Leuven, Belgium
| | - Veluchamy Amutha Barathi
- Singapore Eye Research Institute, Singapore; The Ophthalmology and Visual Sciences ACP, Duke-NUS Medical School, Singapore; Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Arkasubhra Ghosh
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, India.
| |
Collapse
|
14
|
Zheng Y, Shi D, Chen L, Yang Y, Yao M. UCHL1-PKM2 axis dysregulation is associated with promoted proliferation and invasiveness of urothelial bladder cancer cells. Aging (Albany NY) 2023; 15:10593-10606. [PMID: 37815895 PMCID: PMC10599732 DOI: 10.18632/aging.205097] [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: 06/21/2023] [Accepted: 09/09/2023] [Indexed: 10/12/2023]
Abstract
BACKGROUND Bladder cancer is one of the most common type of cancers globally, and the majority of cases belong to urothelial bladder carcinoma (UBC) type. Current researches have demonstrated that multiple genomic abnormalities are related to the sensitivity of cisplatin-based chemotherapy in bladder cancer patients. Previous findings have indicated a controversial role of Ubiquitin Carboxy-Terminal Hydrolase L1 (UCHL1) in malignancy, so we aimed to further explore the role of UCHL1 in UBC. METHODS UBC cell lines and The Cancer Genome Atlas (TCGA) in-silico datasets were utilized to investigate UCHL1 expression pattern and functional as well as prognostic impacts in UBC cancer cell line models and patients. UCHL1 overexpression and silencing vectors and subsequent immunoprecipitation/ubiquitination experiments in combination of cellular functional assays were conducted to explore UCHL1-PKM2 interaction axis and its significance in UBC malignancy. RESULTS UCHL1 was significantly up-regulated in UBC cancer cells and UCHL1 high-expression was associated with higher pathology/clinical grade and significantly inferior overall prognosis of UBC patients. UCHL1 interacted with PKM2 and enhanced PKM2 protein level through inhibition of PKM2 protein degradation via ubiquitination process. UCHL1-PKM2 interaction significantly promoted UBC cellular proliferation, metastasis and invasion activities. CONCLUSION UCHL1-PKM2 interaction played an interesting role in UBC tumor cell proliferation, migration and metastasis. Our study suggests PKM2-targeted treatment might have a potential value in metastatic malignancy therapy development in the future.
Collapse
Affiliation(s)
- Yuhui Zheng
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian, China
| | - Dongliang Shi
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian, China
| | - Linlin Chen
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian, China
| | - Yinghong Yang
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian, China
| | - Meihong Yao
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian, China
| |
Collapse
|
15
|
Dimitrijevs P, Makrecka-Kuka M, Bogucka A, Hyvönen M, Pantelejevs T, Arsenyan P. Development of isoselenazolium chlorides as selective pyruvate kinase isoform M2 inhibitors. Eur J Med Chem 2023; 257:115504. [PMID: 37216812 DOI: 10.1016/j.ejmech.2023.115504] [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: 04/16/2023] [Revised: 05/12/2023] [Accepted: 05/16/2023] [Indexed: 05/24/2023]
Abstract
Alterations in cancer metabolic pathways open up an opportunity for targeted and effective elimination of tumor cells. Pyruvate kinase M2 (PKM2) is predominantly expressed in proliferating cells and plays an essential role in directing glucose metabolism in cancer. Here, we report the design of novel class of selective PKM2 inhibitors as anti-cancer agents and their mechanism of action. Compound 5c being the most active with IC50 = 0.35 ± 0.07 μM, also downregulates PKM2 mRNA expression, modulates mitochondrial functionality, induces oxidative burst and is cytotoxic for various cancer types. Isoselenazolium chlorides have an unusual mechanism of PKM2 inhibition, inducing a functionally deficient tetrameric assembly, while exhibiting a competitive inhibitor character. The discovery of robust PKM2 inhibitors not only offers candidates for anticancer therapy but is also crucial for studying the role of PKM2 in cancer.
Collapse
Affiliation(s)
- Pavels Dimitrijevs
- Latvian Institute of Organic Synthesis, Aizkraukles 21, LV1006, Riga, Latvia
| | | | - Agnieszka Bogucka
- Department of Biochemistry, University of Cambridge, Sanger Building, 80 Tennis Ct Rd, Cambridge, CB2 1GA, UK
| | - Marko Hyvönen
- Department of Biochemistry, University of Cambridge, Sanger Building, 80 Tennis Ct Rd, Cambridge, CB2 1GA, UK
| | - Teodors Pantelejevs
- Latvian Institute of Organic Synthesis, Aizkraukles 21, LV1006, Riga, Latvia
| | - Pavel Arsenyan
- Latvian Institute of Organic Synthesis, Aizkraukles 21, LV1006, Riga, Latvia.
| |
Collapse
|
16
|
Wubben TJ, Chaudhury S, Watch BT, Stuckey JA, Weh E, Fernando R, Goswami M, Pawar M, Rech JC, Besirli CG. Development of Novel Small-Molecule Activators of Pyruvate Kinase Muscle Isozyme 2, PKM2, to Reduce Photoreceptor Apoptosis. Pharmaceuticals (Basel) 2023; 16:ph16050705. [PMID: 37242488 DOI: 10.3390/ph16050705] [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: 04/13/2023] [Revised: 05/01/2023] [Accepted: 05/04/2023] [Indexed: 05/28/2023] Open
Abstract
Treatment options are lacking to prevent photoreceptor death and subsequent vision loss. Previously, we demonstrated that reprogramming metabolism via the pharmacologic activation of PKM2 is a novel photoreceptor neuroprotective strategy. However, the features of the tool compound used in those studies, ML-265, preclude its advancement as an intraocular, clinical candidate. This study sought to develop the next generation of small-molecule PKM2 activators, aimed specifically for delivery into the eye. Compounds were developed that replaced the thienopyrrolopyridazinone core of ML-265 and modified the aniline and methyl sulfoxide functional groups. Compound 2 demonstrated that structural changes to the ML-265 scaffold are tolerated from a potency and efficacy standpoint, allow for a similar binding mode to the target, and circumvent apoptosis in models of outer retinal stress. To overcome the low solubility and problematic functional groups of ML-265, compound 2's efficacious and versatile core structure for the incorporation of diverse functional groups was then utilized to develop novel PKM2 activators with improved solubility, lack of structural alerts, and retained potency. No other molecules are in the pharmaceutical pipeline for the metabolic reprogramming of photoreceptors. Thus, this study is the first to cultivate the next generation of novel, structurally diverse, small-molecule PKM2 activators for delivery into the eye.
Collapse
Affiliation(s)
- Thomas J Wubben
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI 48105, USA
| | - Sraboni Chaudhury
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI 48105, USA
| | - Brennan T Watch
- Department of Internal Medicine, Hematology and Oncology, Michigan Center for Therapeutic Innovation, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jeanne A Stuckey
- Departments of Biological Chemistry and Biophysics, Center for Structural Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Eric Weh
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI 48105, USA
| | - Roshini Fernando
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI 48105, USA
| | - Moloy Goswami
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI 48105, USA
| | - Mercy Pawar
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI 48105, USA
| | - Jason C Rech
- Department of Internal Medicine, Hematology and Oncology, Michigan Center for Therapeutic Innovation, University of Michigan, Ann Arbor, MI 48109, USA
| | - Cagri G Besirli
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI 48105, USA
| |
Collapse
|
17
|
Niu N, Ye J, Hu Z, Zhang J, Wang Y. Regulative Roles of Metabolic Plasticity Caused by Mitochondrial Oxidative Phosphorylation and Glycolysis on the Initiation and Progression of Tumorigenesis. Int J Mol Sci 2023; 24:ijms24087076. [PMID: 37108242 PMCID: PMC10139088 DOI: 10.3390/ijms24087076] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/23/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
One important feature of tumour development is the regulatory role of metabolic plasticity in maintaining the balance of mitochondrial oxidative phosphorylation and glycolysis in cancer cells. In recent years, the transition and/or function of metabolic phenotypes between mitochondrial oxidative phosphorylation and glycolysis in tumour cells have been extensively studied. In this review, we aimed to elucidate the characteristics of metabolic plasticity (emphasizing their effects, such as immune escape, angiogenesis migration, invasiveness, heterogeneity, adhesion, and phenotypic properties of cancers, among others) on tumour progression, including the initiation and progression phases. Thus, this article provides an overall understanding of the influence of abnormal metabolic remodeling on malignant proliferation and pathophysiological changes in carcinoma.
Collapse
Affiliation(s)
- Nan Niu
- Shenzhen Engineering Labortaory for Marine Algal Biotechnology, Longhua Innovation Institute for Biotechnology, College of Life Sciences and Oceanography, Lihu Campus of Shenzhen University, Shenzhen 518055, China
- College of Physics and Optoelectronic Engineering, Canghai Campus of Shenzhen University, Shenzhen 518060, China
| | - Jinfeng Ye
- Shenzhen Engineering Labortaory for Marine Algal Biotechnology, Longhua Innovation Institute for Biotechnology, College of Life Sciences and Oceanography, Lihu Campus of Shenzhen University, Shenzhen 518055, China
| | - Zhangli Hu
- Shenzhen Engineering Labortaory for Marine Algal Biotechnology, Longhua Innovation Institute for Biotechnology, College of Life Sciences and Oceanography, Lihu Campus of Shenzhen University, Shenzhen 518055, China
| | - Junbin Zhang
- Shenzhen Engineering Labortaory for Marine Algal Biotechnology, Longhua Innovation Institute for Biotechnology, College of Life Sciences and Oceanography, Lihu Campus of Shenzhen University, Shenzhen 518055, China
| | - Yun Wang
- Shenzhen Engineering Labortaory for Marine Algal Biotechnology, Longhua Innovation Institute for Biotechnology, College of Life Sciences and Oceanography, Lihu Campus of Shenzhen University, Shenzhen 518055, China
| |
Collapse
|
18
|
Yan M, Yao J, Lin Y, Yan J, Xie Y, Fu Z, Zhou Y, Wei J, Li X. Tumor cell density dependent IL-8 secretion induces the fluctuation of tregs/CD8 + T cells infiltration in hepatocellular carcinoma: one prompt for the existence of density checkpoint. J Transl Med 2023; 21:202. [PMID: 36932390 PMCID: PMC10022186 DOI: 10.1186/s12967-023-04060-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 03/12/2023] [Indexed: 03/19/2023] Open
Abstract
BACKGROUND Tumor cell density is a basic pathological feature of solid tumors. Chemotherapy, radiotherapy, and targeted therapy reduce tumor cell density, whereas unrestricted tumor cell proliferation promotes this feature. The impact of tumor cells on the microenvironment following changes in tumor cell density is still unclear. In this study, we focused on the response of key immune cell subsets to tumor cell density in hepatocellular carcinoma (HCC). METHODS We determined the density of tumor and immune cells in the same area by section staining. We then identified potential mediators using polymerase chain reaction (PCR), enzyme-linked immunofluorescence assay (ELISA), 3D and co-culture, flow cytometry, and lentivirus intervention. The mechanism of lactate promotion was verified using lactate tests, bioinformatics, western blotting, and the above methods. The IL-8/DAPK1/lactate/regulatory T cell (Treg) axis was verified using a mouse liver cancer model. Tumor mutation burden was calculated using maftools in R. RESULTS We found that the Treg/CD8 + T cell ratio is not consistent with tumor cell density in HCC, and a decreased Treg/CD8 + T cell ratio in the range of 5000-6000 cells/mm2 may elicit the possibility for immunotherapy in an immunosuppressive microenvironment. We showed that IL-8 mediates this immune fluctuation and promotes the infiltration of Tregs through the DAPK1/pyruvate kinase activity/lactate axis in HCC. Based on tumor ploidy and mutation burden data, we discussed the potential significance of immune fluctuation in the homeostasis of HCC mutation burden and proposed a "density checkpoint" and "entropy model" to describe this phenomenon. CONCLUSIONS In summary, we report the mode of infiltration of Tregs/CD8 + T cells in response to tumor cell density and provide a new theoretical basis for IL-8 as a therapeutic target and the selection of an immunotherapy window in HCC.
Collapse
Affiliation(s)
- Mengchao Yan
- The First School of Clinical Medical, Lanzhou University, No.1 Donggang West Road, Chengguan District, Lanzhou, 730000, Gansu, People's Republic of China
| | - Jia Yao
- The First School of Clinical Medical, Lanzhou University, No.1 Donggang West Road, Chengguan District, Lanzhou, 730000, Gansu, People's Republic of China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou, 730000, Gansu, People's Republic of China
| | - Yan Lin
- The First School of Clinical Medical, Lanzhou University, No.1 Donggang West Road, Chengguan District, Lanzhou, 730000, Gansu, People's Republic of China
| | - Jun Yan
- The First School of Clinical Medical, Lanzhou University, No.1 Donggang West Road, Chengguan District, Lanzhou, 730000, Gansu, People's Republic of China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou, 730000, Gansu, People's Republic of China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
| | - Ye Xie
- The First School of Clinical Medical, Lanzhou University, No.1 Donggang West Road, Chengguan District, Lanzhou, 730000, Gansu, People's Republic of China
| | - Zongli Fu
- SUN YAT-SEN University, Guangzhou, 510000, Guangdong, People's Republic of China
| | - Yongqiang Zhou
- The First School of Clinical Medical, Lanzhou University, No.1 Donggang West Road, Chengguan District, Lanzhou, 730000, Gansu, People's Republic of China
| | - Jiayun Wei
- The First School of Clinical Medical, Lanzhou University, No.1 Donggang West Road, Chengguan District, Lanzhou, 730000, Gansu, People's Republic of China
| | - Xun Li
- The First School of Clinical Medical, Lanzhou University, No.1 Donggang West Road, Chengguan District, Lanzhou, 730000, Gansu, People's Republic of China.
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou, 730000, Gansu, People's Republic of China.
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China.
| |
Collapse
|
19
|
Metabolic remodeling of pyrimidine synthesis pathway and serine synthesis pathway in human glioblastoma. Sci Rep 2022; 12:16277. [PMID: 36175487 PMCID: PMC9522918 DOI: 10.1038/s41598-022-20613-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 09/15/2022] [Indexed: 12/02/2022] Open
Abstract
Glioblastoma is the most common brain tumor with dismal outcomes in adults. Metabolic remodeling is now widely acknowledged as a hallmark of cancer cells, but glioblastoma-specific metabolic pathways remain unclear. Here we show, using a large-scale targeted proteomics platform and integrated molecular pathway-level analysis tool, that the de novo pyrimidine synthesis pathway and serine synthesis pathway (SSP) are the major enriched pathways in vivo for patients with glioblastoma. Among the enzymes associated with nucleotide synthesis, RRM1 and NME1 are significantly upregulated in glioblastoma. In the SSP, SHMT2 and PSPH are upregulated but the upstream enzyme PSAT1 is downregulated in glioblastoma. Kaplan–Meier curves of overall survival for the GSE16011 and The Cancer Genome Atlas datasets revealed that high SSP activity correlated with poor outcome. Enzymes relating to the pyrimidine synthesis pathway and SSP might offer therapeutic targets for new glioblastoma treatments.
Collapse
|
20
|
Nold SP, Sych K, Imre G, Fuhrmann DC, Pfeilschifter J, Vutukuri R, Schnutgen F, Wittig I, Meisterknecht J, Frank S, Goren I. Reciprocal abrogation of
PKM
isoforms: contradictory outcomes and differing impact of splicing signal on
CRISPR
/Cas9 mediates gene editing in keratinocytes. FEBS J 2022; 290:2338-2365. [PMID: 36083715 DOI: 10.1111/febs.16625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 06/28/2022] [Accepted: 09/08/2022] [Indexed: 11/30/2022]
Abstract
The healing of wounded skin is a highly organized process involving a massive cell in- and outflux, proliferation and tissue remodelling. It is well accepted that metabolic constraints such as diabetes mellitus, overweight or anorexia impairs wound healing. Indeed, wound inflammation involves a boost of overall metabolic changes. As wound healing converges inflammatory processes that are also common to transformation, we investigate the functional role of the pro-neoplastic factor pyruvate kinase (PK) M2 and its metabolic active splice variant PKM1 in keratinocytes. Particularly, we challenge the impact of reciprocal ablation of PKM1 or two expression. Here, CRISPR/Cas9 genome editing of the PKM gene in HaCaT reveals an unexpected mutational bias at the 3'SS of exon 9, whereas no preference for any particular kind of mutation at exon 10 3' splice, despite the close vicinity (400 nucleotides apart) and sequence similarity between the two sites. Furthermore, as opposed to transient silencing of PKM2, exclusion splicing of PKM2 via genome editing mutually increases PKM1 mRNA and protein expression and compensates for the absence of PKM2, whereas the reciprocal elimination of PKM1 splicing reduces PKM2 expression and impedes cell proliferation, thus unveiling an essential role for PKM1 in growth and metabolic balance of HaCaT keratinocytes.
Collapse
Affiliation(s)
- Simon P. Nold
- Pharmazentrum Frankfurt/ZAFES, General Pharmacology and Toxicology, Faculty of Medicine Goethe University Frankfurt Germany
| | - Khrystyna Sych
- Pharmazentrum Frankfurt/ZAFES, General Pharmacology and Toxicology, Faculty of Medicine Goethe University Frankfurt Germany
| | - Gergely Imre
- Pharmazentrum Frankfurt/ZAFES, General Pharmacology and Toxicology, Faculty of Medicine Goethe University Frankfurt Germany
| | - Dominik C. Fuhrmann
- Faculty of Medicine, Institute of Biochemistry I Goethe University Frankfurt Germany
| | - Josef Pfeilschifter
- Pharmazentrum Frankfurt/ZAFES, General Pharmacology and Toxicology, Faculty of Medicine Goethe University Frankfurt Germany
| | - Rajkumar Vutukuri
- Pharmazentrum Frankfurt/ZAFES, General Pharmacology and Toxicology, Faculty of Medicine Goethe University Frankfurt Germany
| | - Frank Schnutgen
- Hematology/Oncology, Faculty of Medicine 2, University Hospital Goethe University Frankfurt Germany
- German Cancer Research Center and German Cancer Consortium Heidelberg Germany
- Frankfurt Cancer Institute (FCI) Germany
| | - Ilka Wittig
- Functional Proteomics, Institute of Cardiovascular Physiology Goethe University Frankfurt Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhein‐Main Frankfurt Germany
| | - Jana Meisterknecht
- Functional Proteomics, Institute of Cardiovascular Physiology Goethe University Frankfurt Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhein‐Main Frankfurt Germany
| | - Stefan Frank
- Pharmazentrum Frankfurt/ZAFES, General Pharmacology and Toxicology, Faculty of Medicine Goethe University Frankfurt Germany
| | - Itamar Goren
- Pharmazentrum Frankfurt/ZAFES, General Pharmacology and Toxicology, Faculty of Medicine Goethe University Frankfurt Germany
| |
Collapse
|
21
|
El-Far AH, Al Jaouni SK, Li X, Fu J. Cancer metabolism control by natural products: Pyruvate kinase M2 targeting therapeutics. Phytother Res 2022; 36:3181-3201. [PMID: 35794729 DOI: 10.1002/ptr.7534] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 05/19/2022] [Accepted: 06/12/2022] [Indexed: 12/13/2022]
Abstract
Glycolysis is the primary source of energy for cancer growth and metastasis. The shift in metabolism from mitochondrial oxidative phosphorylation to aerobic glycolysis is called the Warburg effect. Cancer progression due to aerobic glycolysis is often associated with the activation of oncogenes or the loss of tumor suppressors. Therefore, inhibition of glycolysis is one of the effective strategies in cancer control. Pyruvate kinase M2 (PKM2) is a key glycolytic enzyme overexpressed in breast, prostate, lung, colorectal, and liver cancers. Here, we discuss published studies regarding PKM2 inhibitors from natural products that are promising drug candidates for cancer therapy. We have highlighted the potential of natural PKM2 inhibitors for various cancer types. Moreover, we encourage researchers to evaluate the combinational effects between natural and synthetic PKM2 inhibitors. Also, further high-quality studies are needed to firmly establish the clinical efficacy of natural products.
Collapse
Affiliation(s)
- Ali H El-Far
- Department of Biochemistry, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| | - Soad K Al Jaouni
- Department of Hematology/Pediatric Oncology, Yousef Abdulatif Jameel Scientific Chair of Prophetic Medicine Application, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Xiaotao Li
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, China.,School of Arts and Sciences, New York University-Shanghai, Shanghai, China.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Junjiang Fu
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, China
| |
Collapse
|
22
|
Davidson SM, Schmidt DR, Heyman JE, O'Brien JP, Liu AC, Israelsen WJ, Dayton TL, Sehgal R, Bronson RT, Freinkman E, Mak HH, Fanelli GN, Malstrom S, Bellinger G, Carracedo A, Pandolfi PP, Courtney KD, Jha A, DePinho RA, Horner JW, Thomas CJ, Cantley LC, Loda M, Vander Heiden MG. Pyruvate Kinase M1 Suppresses Development and Progression of Prostate Adenocarcinoma. Cancer Res 2022; 82:2403-2416. [PMID: 35584006 PMCID: PMC9256808 DOI: 10.1158/0008-5472.can-21-2352] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 04/19/2022] [Accepted: 05/11/2022] [Indexed: 01/07/2023]
Abstract
SIGNIFICANCE Differential expression of PKM1 and PKM2 impacts prostate tumorigenesis and suggests a potential therapeutic vulnerability in prostate cancer.
Collapse
Affiliation(s)
- Shawn M. Davidson
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Broad Institute of MIT and Harvard University, Cambridge, Massachusetts.,Corresponding Authors: Matthew G. Vander Heiden, Koch Institute/Biology, Massachusetts Institute of Technology, Cambridge, MA 02139. E-mail: ; and Shawn M. Davidson,
| | - Daniel R. Schmidt
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Broad Institute of MIT and Harvard University, Cambridge, Massachusetts.,Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Julia E. Heyman
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - James P. O'Brien
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Amy C. Liu
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - William J. Israelsen
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Talya L. Dayton
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | | | - Roderick T. Bronson
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | | | - Howard H. Mak
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Giuseppe Nicolò Fanelli
- Weill Cornell Medical College, New York, New York.,Division of Pathology, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Scott Malstrom
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Gary Bellinger
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | | | | | | | | | | | | | - Craig J. Thomas
- National Center for Advancing Translational Sciences, NIH, Bethesda, Maryland
| | - Lewis C. Cantley
- Beth Israel Deaconess Medical Center, Boston, Massachusetts.,Weill Cornell Medical College, New York, New York
| | - Massimo Loda
- Weill Cornell Medical College, New York, New York.,Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Matthew G. Vander Heiden
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Broad Institute of MIT and Harvard University, Cambridge, Massachusetts.,Dana-Farber Cancer Institute, Boston, Massachusetts.,Corresponding Authors: Matthew G. Vander Heiden, Koch Institute/Biology, Massachusetts Institute of Technology, Cambridge, MA 02139. E-mail: ; and Shawn M. Davidson,
| |
Collapse
|
23
|
Huang Z, Zhang J, Gu Y, Cai Z, Wei D, Feng X, Yang C. Analysis of the molecular mechanism of inosine monophosphate deposition in Jingyuan chicken muscles using a proteomic approach. Poult Sci 2022; 101:101741. [PMID: 35259688 PMCID: PMC8904228 DOI: 10.1016/j.psj.2022.101741] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 12/24/2021] [Accepted: 01/12/2022] [Indexed: 12/03/2022] Open
Abstract
Inosine monophosphate (IMP) is an indicator of meat taste, and the molecular mechanism underlying IMP deposition in muscle tissues is important to developing superior poultry breeds. The aim of this study was to identify the key proteins regulating IMP deposition in different muscle groups of 180-day-old Jingyuan chickens (Hen) using a proteomics-based approach. We identified 1,300 proteins in the muscle tissues of Jingyuan chickens, of which 322 were differentially expressed between the breast and leg muscles (129 proteins were highly expressed in breast muscles and 193 proteins were highly expressed in leg muscles). PGM1, PKM2, AK1, AMPD1, and PurH/ATIC were among the differentially expressed proteins (DEPs) involved in the purine metabolism pathway, of which purH was highly expressed in leg muscles, while the others were highly expressed in breast muscles. The proteomics screening results were verified by PRM, qPCR, and western blotting, showing consistency with the proteomics results. Our findings are not only significant in terms of protecting the Jingyuan chicken germplasm resources, but also provide the molecular basis for generating high-quality broiler chicken breeds.
Collapse
Affiliation(s)
- Zengwen Huang
- Agriculture College, Ningxia University, Yinchuan, China; Xichang University, Sichuan 615012, China
| | - Juan Zhang
- Agriculture College, Ningxia University, Yinchuan, China
| | - Yaling Gu
- Agriculture College, Ningxia University, Yinchuan, China.
| | - Zhengyun Cai
- Agriculture College, Ningxia University, Yinchuan, China
| | - Dawei Wei
- Agriculture College, Ningxia University, Yinchuan, China
| | - Xiaofang Feng
- Agriculture College, Ningxia University, Yinchuan, China
| | - Chaoyun Yang
- Agriculture College, Ningxia University, Yinchuan, China
| |
Collapse
|
24
|
Li C, You X, Xu X, Wu B, Liu Y, Tong T, Chen J, Li Y, Dai C, Ye Z, Tian X, Wei Y, Hao Z, Jiang L, Wu J, Zhao M. A Metabolic Reprogramming Amino Acid Polymer as an Immunosurveillance Activator and Leukemia Targeting Drug Carrier for T-Cell Acute Lymphoblastic Leukemia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104134. [PMID: 35080145 PMCID: PMC8948613 DOI: 10.1002/advs.202104134] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/14/2021] [Indexed: 05/10/2023]
Abstract
Compromised immunosurveillance leads to chemotherapy resistance and disease relapse of hematological malignancies. Amino acid metabolism regulates immune responses and cancer; however, a druggable amino acid metabolite to enhance antitumor immunosurveillance and improve leukemia targeting-therapy efficacy remains unexplored. Here, an L-phenylalanine polymer, Metabolic Reprogramming Immunosurveillance Activation Nanomedicine (MRIAN), is invented to effectively target bone marrow (BM) and activate the immune surveillance in T-cell acute lymphoblastic leukemia (T-ALL) by inhibiting myeloid-derived suppressor cells (MDSCs) in T-ALL murine model. Stable-isotope tracer and in vivo drug distribution experiments show that T-ALL cells and MDSCs have enhanced cellular uptake of L-phenylalanine and MRIANs than normal hematopoietic cells and progenitors. Therefore, MRIAN assembled Doxorubicin (MRIAN-Dox) specifically targets T-ALL cells and MDSCs but spare normal hematopoietic cells and hematopoietic stem and progenitor cells with enhanced leukemic elimination efficiency. Consequently, MRIAN-Dox has reduced cardiotoxicity and myeloablation side effects in treating T-ALL mice. Mechanistically, MRIAN degrades into L-phenylalanine, which inhibits PKM2 activity and reduces ROS levels in MDSCs to disturb their immunosuppressive function and increase their differentiation toward normal myeloid cells. Overall, a novel amino acid metabolite nanomedicine is invented to treat T-ALL through the combination of leukemic cell targeting and immunosurveillance stimulation.
Collapse
Affiliation(s)
- Changzheng Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat‐Sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education)Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Xinru You
- School of Biomedical EngineeringSun Yat‐sen UniversityShenzhenGuangdongChina
| | - Xi Xu
- Department of HematologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Binghuo Wu
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education)Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Yuye Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat‐Sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Tong Tong
- School of Biomedical EngineeringSun Yat‐sen UniversityShenzhenGuangdongChina
| | - Jie Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat‐Sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Yishan Li
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education)Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Chunlei Dai
- School of Biomedical EngineeringSun Yat‐sen UniversityShenzhenGuangdongChina
| | - Zhitao Ye
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education)Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Xiaobin Tian
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education)Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Yan Wei
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education)Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Zechen Hao
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education)Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Linjia Jiang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat‐Sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Jun Wu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat‐Sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
- School of Biomedical EngineeringSun Yat‐sen UniversityShenzhenGuangdongChina
| | - Meng Zhao
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education)Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouGuangdongChina
| |
Collapse
|
25
|
Ding L, Liang X. Ras related GTP binding D promotes aerobic glycolysis of hepatocellular carcinoma. Ann Hepatol 2022; 23:100307. [PMID: 33434687 DOI: 10.1016/j.aohep.2021.100307] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 12/03/2020] [Accepted: 12/04/2020] [Indexed: 02/04/2023]
Abstract
INTRODUCTION AND OBJECTIVES Warburg effect is attracting increasing attention as it is important for cancer progression. However, how cancer cells regulate glucose metabolism through glycolysis is still unknown. Here, we demonstrated the regulatory role of Ras related GTP binding D (RRAGD) in human hepatocellular carcinoma (HCC) cells. PATIENTS OR MATERIALS AND METHODS Kaplan-Meier's analysis was used to analyze the correlation between RRAGD expression levels and prognosis of HCC patients from the Cancer Genome Atlas database. Two stable RRAGD knockdown HCC cell lines were created using shRNAs to investigate cancer progression and aerobic glycolysis. Western blot and quantitative reverse transcription polymerase chain reaction were performed to detect the expression levels of RRAGD and MYC. RESULTS RRAGD expression was elevated in HCC patients with poor prognosis. RRAGD knockdown could inhibit the proliferation, invasion and migration of Huh-7 and HepG2 cells. Interestingly, silence RRAGD was able to reduce the glucose uptake, lactate production and extracellular acidification rate of HCC. RRAGD expression level was up-regulated by oncogene MYC in HCC cells. CONCLUSION This study highlights RRAGD as an important cancer-promoting factor for cancer progression and aerobic glycolysis, and thereby it is a potential therapeutic target for HCC intervention.
Collapse
Affiliation(s)
- Lishuang Ding
- The First Department of Pu Wai, Xinhua West Road Central Hospital, Cangzhou 061001, Hebei, China
| | - Xuezheng Liang
- Emergency Department, Xinhua West Road Central Hospital, Cangzhou 061001, Hebei, China.
| |
Collapse
|
26
|
Liu X, Wang C, Li S, Qu L, Yin F, Lu D, Luo H, Chen X, Luo Z, Cui N, Wang X, Kong L. Parthenolide Derivatives as PKM2 Activators Showing Potential in Colorectal Cancer. J Med Chem 2021; 64:17304-17325. [PMID: 34847663 DOI: 10.1021/acs.jmedchem.1c01380] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
As a vital kinase in the glycolysis system, PKM2 is extensively expressed in colorectal cancer (CRC) to support the energy and biosynthetic needs. In this study, we designed a series of parthenolide (PTL) derivatives through a stepwise structure optimization, and an excellent derivate 29e showed good activity on PKM2 (AC50 = 86.29 nM) and displayed significant antiproliferative activity against HT29 (IC50 = 0.66 μM) and SW480 (IC50 = 0.22 μM) cells. 29e decreased the expression of total PKM2, prevented nucleus translocation of PKM2 dimer, and inhibited PKM2/STAT3 signaling pathway. 29e remarkably increased OCR and decreased the extracellular acidification rate (ECAR). The antiproliferative effect of 29e depended on PKM2, and the Cys424 of PKM2 was the key binding site. Furthermore, 29e significantly suppressed tumor growth in the HT29 xenograft model without obvious toxicity. These outcomes demonstrate that 29e is a promising drug candidate for the treatment of CRC.
Collapse
Affiliation(s)
- Xingchen Liu
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Cheng Wang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Shang Li
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Lailiang Qu
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Fucheng Yin
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Dehua Lu
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Heng Luo
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Xinye Chen
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Zhongwen Luo
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Ningjie Cui
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Xiaobing Wang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Lingyi Kong
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| |
Collapse
|
27
|
Li M, Lu H, Wang X, Duan C, Zhu X, Zhang Y, Ge X, Ji F, Wang X, Su J, Zhang D. Pyruvate kinase M2 (PKM2) interacts with activating transcription factor 2 (ATF2) to bridge glycolysis and pyroptosis in microglia. Mol Immunol 2021; 140:250-266. [PMID: 34798593 DOI: 10.1016/j.molimm.2021.10.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 09/25/2021] [Accepted: 10/25/2021] [Indexed: 12/14/2022]
Abstract
Pyruvate kinase M2 (PKM2), a glycolytic rate-limiting enzyme, reportedly plays an important role in tumorigenesis and the inflammatory response by regulating the metabolic reprogramming. However, its contribution to microglial activation during neuroinflammation is still unknown. In this study, we observed an enhanced glycolysis level in the lipopolysaccharide (LPS)-activated microglia. Utilizing the glycolysis inhibitor 2-DG, we proved that LPS requires glycolysis to induce microglial pyroptosis. Moreover, the protein expression, dimer/monomer formation, phosphorylation and nuclear translocation of PKM2 were all increased by LPS. Silencing PKM2 or preventing its nuclear translocation by TEPP-46 significantly alleviated the LPS-induced inflammatory response and pyroptosis in microglia. Employing biological mass spectrometry combined with immunoprecipitation technology, we identified for the first time that PKM2 interacts with activating transcription factor 2 (ATF2) in microglia. Inhibition of glycolysis or preventing PKM2 nuclear aggregation significantly reduced the phosphorylation and activation of ATF2. Furthermore, knocking down ATF2 reduced the LPS-induced pyroptosis of microglia. In vivo, we showed the LPS-induced pyroptosis in the cerebral cortex tissues of mice, and first found that an increased PKM2 expression was co-localized with ATF2 in the inflamed mice brain. Collectively, our data suggested for the first time that PKM2, a key rate-limiting enzyme of the Warburg effect, directly interacts with the pro-inflammatory transcription factor ATF2 to bridge glycolysis and pyroptosis in microglia, which might be a pivotal crosstalk between metabolic reprogramming and neuroinflammation in the CNS.
Collapse
Affiliation(s)
- Mengmeng Li
- Medical Research Center, Affiliated Hospital 2 of Nantong University, Nantong 226001, People's Republic of China
| | - Hongjian Lu
- Medical Research Center, Affiliated Hospital 2 of Nantong University, Nantong 226001, People's Republic of China; Department of Rehabilitation Medicine, Affiliated Hospital 2 of Nantong University, Nantong 226001, People's Republic of China
| | - Xueyan Wang
- Medical Research Center, Affiliated Hospital 2 of Nantong University, Nantong 226001, People's Republic of China; Department of Pathogen Biology, Medical College, Nantong University, Nantong 226001, People's Republic of China
| | - Chengwei Duan
- Medical Research Center, Affiliated Hospital 2 of Nantong University, Nantong 226001, People's Republic of China
| | - Xiangyang Zhu
- Neurology Department, Affiliated Hospital 2 of Nantong University, Nantong 226001, People's Republic of China
| | - Yi Zhang
- Neurosurgery Department, Affiliated Hospital 2 of Nantong University, Nantong 226001, People's Republic of China
| | - Xin Ge
- Medical Research Center, Affiliated Hospital 2 of Nantong University, Nantong 226001, People's Republic of China
| | - Feng Ji
- Medical Research Center, Affiliated Hospital 2 of Nantong University, Nantong 226001, People's Republic of China
| | - Xueqin Wang
- Endocrinology Department, Affiliated Hospital 2 of Nantong University, Nantong 226001, People's Republic of China
| | - Jianbin Su
- Endocrinology Department, Affiliated Hospital 2 of Nantong University, Nantong 226001, People's Republic of China
| | - Dongmei Zhang
- Medical Research Center, Affiliated Hospital 2 of Nantong University, Nantong 226001, People's Republic of China; Department of Pathogen Biology, Medical College, Nantong University, Nantong 226001, People's Republic of China.
| |
Collapse
|
28
|
Yang GJ, Wu J, Leung CH, Ma DL, Chen J. A review on the emerging roles of pyruvate kinase M2 in anti-leukemia therapy. Int J Biol Macromol 2021; 193:1499-1506. [PMID: 34740687 DOI: 10.1016/j.ijbiomac.2021.10.213] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/28/2021] [Accepted: 10/28/2021] [Indexed: 12/12/2022]
Abstract
Glycolysis is an important step in respiration and provides energy for cellular processes. Pyruvate kinase M2 (PKM2), a key rate-limiting enzyme of glycolysis, plays an important role in tumor cell metabolism and proliferation. It is also specifically overexpressed in leukemia cells and contributes to leukemic proliferation, differentiation, and drug resistance through both aerobic glycolysis and non-metabolic pathways. In this review, the functions and regulatory roles of PKM2 are firstly introduced. Then, the molecular mechanisms of PKM2 in leukemogenesis are summarized. Next, reported PKM2 modulators and their anti-leukemia mechanisms are described. Finally, the current challenges and the potential opportunities of PKM2 inhibitors or agonists in leukemia therapy are discussed.
Collapse
Affiliation(s)
- Guan-Jun Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo 315211, Zhejiang, China; Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao SAR, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China
| | - Jia Wu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao SAR, China
| | - Chung-Hang Leung
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao SAR, China; Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, 999078, Macao SAR, China.
| | - Dik-Lung Ma
- Department of Chemistry, Hong Kong Baptist University, Kowloon 999077, Hong Kong, China.
| | - Jiong Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo 315211, Zhejiang, China; Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao SAR, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, China.
| |
Collapse
|
29
|
Chen J, LaGue E, Li J, Yang C, Hackett EP, Mendoza M, Alger JR, DeBerardinis RJ, Corbin IR, Billingsley KL, Park JM. Profiling Carbohydrate Metabolism in Liver and Hepatocellular Carcinoma with [ 13C]-Glycerate Probes. ANALYSIS & SENSING 2021; 1:196-202. [PMID: 35693130 PMCID: PMC9187054 DOI: 10.1002/anse.202100034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
The interplay between glycolysis and gluconeogenesis is central to carbohydrate metabolism. Here, we describe novel methods to assess carbohydrate metabolism using [13C]-probes derived from glycerate, a molecule whose metabolic fate in mammals remains underexplored. Isotope-based studies were conducted via NMR and mass spectrometry analyses of freeze-clamped liver tissue extracts after [2,3-13C2]glycerate infusion. The ex vivo investigations were correlated with in vivo measurements using hyperpolarized [1-13C]glycerate. Application of [13C]glycerate to N-nitrosodiethylamine (DEN)-treated rats provided further assessments of intermediary carbohydrate metabolism in hepatocellular carcinoma. This method afforded direct analyses of control versus DEN tissues, and altered ratios of 13C metabolic products as well as unique glycolysis intermediates were observed in the DEN liver/tumor. Isotopomer studies showed increased glycerate uptake and altered carbohydrate metabolism in the DEN rats.
Collapse
Affiliation(s)
- Jun Chen
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-8568 (USA)
| | - Evan LaGue
- Department of Chemistry and Biochemistry, California State University, Fullerton, 800 State College Blvd. Fullerton, CA 92834-6866 (USA)
| | - Junjie Li
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-8568 (USA)
| | - Chendong Yang
- Howard Hughes Medical Institute and Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-8502 (USA)
| | - Edward P Hackett
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-8568 (USA)
| | - Manuel Mendoza
- Department of Chemistry and Biochemistry, California State University, Fullerton, 800 State College Blvd. Fullerton, CA 92834-6866 (USA)
| | - Jeffry R Alger
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-8568 (USA)
| | - Ralph J DeBerardinis
- Howard Hughes Medical Institute and Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-8502 (USA)
| | - Ian R Corbin
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-8568 (USA)
| | - Kelvin L Billingsley
- Department of Chemistry and Biochemistry, California State University, Fullerton, 800 State College Blvd. Fullerton, CA 92834-6866 (USA)
| | - Jae Mo Park
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-8568 (USA)
| |
Collapse
|
30
|
The Intersection of Purine and Mitochondrial Metabolism in Cancer. Cells 2021; 10:cells10102603. [PMID: 34685583 PMCID: PMC8534091 DOI: 10.3390/cells10102603] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/20/2021] [Accepted: 09/23/2021] [Indexed: 12/23/2022] Open
Abstract
Nucleotides are essential to cell growth and survival, providing cells with building blocks for DNA and RNA, energy carriers, and cofactors. Mitochondria have a critical role in the production of intracellular ATP and participate in the generation of intermediates necessary for biosynthesis of macromolecules such as purines and pyrimidines. In this review, we highlight the role of purine and mitochondrial metabolism in cancer and how their intersection influences cancer progression, especially in ovarian cancer. Additionally, we address the importance of metabolic rewiring in cancer and how the evolving landscape of purine synthesis and mitochondria inhibitors can be potentially exploited for cancer treatment.
Collapse
|
31
|
Rathod B, Chak S, Patel S, Shard A. Tumor pyruvate kinase M2 modulators: a comprehensive account of activators and inhibitors as anticancer agents. RSC Med Chem 2021; 12:1121-1141. [PMID: 34355179 PMCID: PMC8292966 DOI: 10.1039/d1md00045d] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 03/25/2021] [Indexed: 12/16/2022] Open
Abstract
Pyruvate kinase M2 (PKM2) catalyzes the conversion of phosphoenolpyruvate (PEP) to pyruvate. It plays a central role in the metabolic reprogramming of cancer cells and is expressed in most human tumors. It is essential in indiscriminate proliferation, survival, and tackling apoptosis in cancer cells. This positions PKM2 as a hot target in cancer therapy. Despite its well-known structure and several reported modulators targeting PKM2 as activators or inhibitors, a comprehensive review focusing on such modulators is lacking. Herein we summarize modulators of PKM2, the assays used to detect their potential, the preferable tense (T) and relaxed (R) states in which the enzyme resides, lacunae in existing modulators, and several strategies that may lead to effective anticancer drug development targeting PKM2.
Collapse
Affiliation(s)
- Bhagyashri Rathod
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad Opposite Air Force Station Gandhinagar Gujarat 382355 India
| | - Shivam Chak
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad Opposite Air Force Station Gandhinagar Gujarat 382355 India
| | - Sagarkumar Patel
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad Opposite Air Force Station Gandhinagar Gujarat 382355 India
| | - Amit Shard
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad Opposite Air Force Station Gandhinagar Gujarat 382355 India
| |
Collapse
|
32
|
Xie M, Pei DS. Serine hydroxymethyltransferase 2: a novel target for human cancer therapy. Invest New Drugs 2021; 39:1671-1681. [PMID: 34215932 DOI: 10.1007/s10637-021-01144-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 06/22/2021] [Indexed: 12/21/2022]
Abstract
Serine and glycine are the primary sources of one-carbon units that are vital for cell proliferation. Their abnormal metabolism is known to be associated with cancer progression. As the key enzyme of serine metabolism, Serine Hydroxymethyltransferase 2 (SHMT2) has been a research hotspot in recent years. SHMT2 is a PLP-dependent tetrameric enzyme that catalyzes the reversible transition from serine to glycine, thus promoting the production of one-carbon units that are indispensable for cell growth and regulation of the redox and epigenetic states of cells. Under a hypoxic environment, SHMT2 can be upregulated and could promote the generation of nicotinamide adenine dinucleotide phosphate (NADPH) and glutathione for maintaining the redox balance. Accumulating evidence confirmed that SHMT2 facilitates cell proliferation and tumor growth and is tightly associated with poor prognosis. In this review, we present insights into the function and research development of SHMT2 and summarize the possible molecular mechanisms of SHMT2 in promoting tumor growth, in the hope that it could provide clues to more effective clinical treatment of cancer.
Collapse
Affiliation(s)
- Min Xie
- Department of Pathology, Xuzhou Medical University, 209 Tong-shan Road, Xuzhou, 221004, Jiangsu, China
| | - Dong-Sheng Pei
- Department of Pathology, Xuzhou Medical University, 209 Tong-shan Road, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
33
|
Zhou L, Ding L, Gong Y, Zhao J, Zhang J, Mao Z, Wang Z, Zhang W, Zhou R. NEK2 Promotes Cell Proliferation and Glycolysis by Regulating PKM2 Abundance via Phosphorylation in Diffuse Large B-Cell Lymphoma. Front Oncol 2021; 11:677763. [PMID: 34168996 PMCID: PMC8217770 DOI: 10.3389/fonc.2021.677763] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/13/2021] [Indexed: 12/17/2022] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most frequent and commonly diagnosed subtype of NHL, which is characterized by high heterogeneity and malignancy, and most DLBCL patients are at advanced stages. The serine/threonine kinase NEK2 (NIMA-related kinase 2), a member of NIMA-related kinase (NEK) family that regulates cell cycle, is upregulated in a variety of malignancies, including diffuse large B-cell lymphoma. However, the role and underlying mechanisms of NEK2 in DLBCL have seldom been discussed. In this study, we identified that NEK2 is upregulated in DLBCL compared to normal lymphoid tissues, and overexpression of NEK2 predicted a worse prognosis of DLBCL patients. Gene set enrichment analysis indicates that NEK2 might participate in regulating glycolysis. Knockdown of NEK2 inhibited growth and glycolysis of DLBCL cells. The interaction between NEK2 and PKM2 was discovered by tandem affinity purification and then was confirmed by immunofluorescence staining, coimmunoprecipitation, and immunoprecipitation. NEK2 bounds to PKM2 and regulates PKM2 abundance via phosphorylation, which increases PKM2 stability. The xenograft tumor model checks the influence of NEK2 on tumor growth in vivo. Thus, NEK2 could be the novel biomarker and target of DLBCL, which remarkably ameliorates the diagnosis and treatment of DLBCL.
Collapse
Affiliation(s)
- Lingna Zhou
- Department of Pathology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Cancer Institute, Department of Pathology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Clinical Science, Department of Pathology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liya Ding
- Department of Pathology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Clinical Science, Department of Pathology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuqi Gong
- Department of Pathology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jing Zhao
- Department of Pathology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jing Zhang
- Department of Pathology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhengrong Mao
- Department of Pathology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhe Wang
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Wei Zhang
- Cancer Institute, Department of Pathology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ren Zhou
- Department of Pathology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
34
|
Zhu S, Guo Y, Zhang X, Liu H, Yin M, Chen X, Peng C. Pyruvate kinase M2 (PKM2) in cancer and cancer therapeutics. Cancer Lett 2021; 503:240-248. [PMID: 33246091 DOI: 10.1016/j.canlet.2020.11.018] [Citation(s) in RCA: 139] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 10/12/2020] [Accepted: 11/15/2020] [Indexed: 02/07/2023]
Abstract
Pyruvate kinase M2 (PKM2), a key rate-limiting enzyme of glycolysis, is a critical regulator in tumor metabolism. PKM2 has been demonstrated to overexpressed in various cancers and promoted proliferation and metastasis of tumor cells. The errant expression of PKM2 has inspired people to investigate the function of PKM2 and the therapeutic potential in cancer. In addition, some studies have shown that the upregulation of PKM2 in tumor tissues is associated with the altered expression of lncRNAs and the poor survival. Therefore, researchers have begun to unravel the specific molecular mechanisms of lncRNA-mediated PKM2 expression in cancer metabolism. As the tumor microenvironment (TME) is essential in tumor development, it is necessary to identify the role of PKM2 in TME. In this review, we will introduce the role of PKM2 in different cancers as well as TME, and summarize the molecular mechanism of PKM2-related lncRNAs in cancer metabolism. We expect that this work will lead to a better understanding of the molecular mechanisms of PKM2 that may help in developing therapeutic strategies in clinic for researchers.
Collapse
Affiliation(s)
- Susi Zhu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, China; Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, China; Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, Hunan, China; Research Center of Molecular Metabolomics, Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yeye Guo
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, China; Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, China; Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, Hunan, China; Research Center of Molecular Metabolomics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xu Zhang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, China; Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, China; Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, Hunan, China; Research Center of Molecular Metabolomics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hong Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, China; Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, China; Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, Hunan, China; Research Center of Molecular Metabolomics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Mingzhu Yin
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, China; Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, China; Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, Hunan, China; Research Center of Molecular Metabolomics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, China; Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, China; Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, Hunan, China; Research Center of Molecular Metabolomics, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Cong Peng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, China; Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, China; Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, Hunan, China; Research Center of Molecular Metabolomics, Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
35
|
Olson-Manning CF. Elaboration of the Corticosteroid Synthesis Pathway in Primates through a Multistep Enzyme. Mol Biol Evol 2021; 37:2257-2267. [PMID: 32196091 DOI: 10.1093/molbev/msaa080] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Metabolic networks are complex cellular systems dependent on the interactions among, and regulation of, the enzymes in the network. Although there is great diversity of types of enzymes that make up metabolic networks, the models meant to understand the possible evolutionary outcomes following duplication neglect specifics about the enzyme, pathway context, and cellular constraints. To illuminate the mechanisms that shape the evolution of biochemical pathways, I functionally characterize the consequences of gene duplication of an enzyme family that performs multiple subsequent enzymatic reactions (a multistep enzyme) in the corticosteroid pathway in primates. The products of the corticosteroid pathway (aldosterone and cortisol) are steroid hormones that regulate metabolism and stress response in tetrapods. These steroid hormones are synthesized by a multistep enzyme Cytochrome P450 11B (CYP11B) that performs subsequent steps on different carbon atoms of the steroid derivatives. Through ancestral state reconstruction and in vitro characterization, I find that the primate ancestor of the CYP11B1 and CYP11B2 paralogs had moderate ability to synthesize both cortisol and aldosterone. Following duplication in Old World primates, the CYP11B1 homolog specialized on the production of cortisol, whereas its paralog, CYP11B2, maintained its ability to perform multiple subsequent steps as in the ancestral pathway. Unlike CYP11B1, CYP11B2 could not specialize on the production of aldosterone because it is constrained to perform earlier steps in the corticosteroid synthesis pathway to achieve the final product aldosterone. These results suggest that enzyme function, pathway context, along with tissue-specific regulation, both play a role in shaping potential outcomes of metabolic network elaboration.
Collapse
Affiliation(s)
- Carrie F Olson-Manning
- Department of Biology, Augustana University, Sioux Falls, SD.,Department of Ecology and Evolution, University of Chicago, Chicago, IL
| |
Collapse
|
36
|
Weglarz-Tomczak E, Mondeel TDGA, Piebes DGE, Westerhoff HV. Simultaneous Integration of Gene Expression and Nutrient Availability for Studying the Metabolism of Hepatocellular Carcinoma Cell Lines. Biomolecules 2021; 11:biom11040490. [PMID: 33805227 PMCID: PMC8064315 DOI: 10.3390/biom11040490] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/07/2021] [Accepted: 03/19/2021] [Indexed: 01/08/2023] Open
Abstract
How cancer cells utilize nutrients to support their growth and proliferation in complex nutritional systems is still an open question. However, it is certainly determined by both genetics and an environmental-specific context. The interactions between them lead to profound metabolic specialization, such as consuming glucose and glutamine and producing lactate at prodigious rates. To investigate whether and how glucose and glutamine availability impact metabolic specialization, we integrated computational modeling on the genome-scale metabolic reconstruction with an experimental study on cell lines. We used the most comprehensive human metabolic network model to date, Recon3D, to build cell line-specific models. RNA-Seq data was used to specify the activity of genes in each cell line and the uptake rates were quantitatively constrained according to nutrient availability. To integrated both constraints we applied a novel method, named Gene Expression and Nutrients Simultaneous Integration (GENSI), that translates the relative importance of gene expression and nutrient availability data into the metabolic fluxes based on an observed experimental feature(s). We applied GENSI to study hepatocellular carcinoma addiction to glucose/glutamine. We were able to identify that proliferation, and lactate production is associated with the presence of glucose but does not necessarily increase with its concentration when the latter exceeds the physiological concentration. There was no such association with glutamine. We show that the integration of gene expression and nutrient availability data into genome-wide models improves the prediction of metabolic phenotypes.
Collapse
Affiliation(s)
- Ewelina Weglarz-Tomczak
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, 1098 XH Amsterdam, The Netherlands; (T.D.G.A.M.); (D.G.E.P.); (H.V.W.)
- Correspondence:
| | - Thierry D. G. A. Mondeel
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, 1098 XH Amsterdam, The Netherlands; (T.D.G.A.M.); (D.G.E.P.); (H.V.W.)
| | - Diewertje G. E. Piebes
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, 1098 XH Amsterdam, The Netherlands; (T.D.G.A.M.); (D.G.E.P.); (H.V.W.)
| | - Hans V. Westerhoff
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, 1098 XH Amsterdam, The Netherlands; (T.D.G.A.M.); (D.G.E.P.); (H.V.W.)
- Molecular Cell Physiology, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands
- Manchester Centre for Integrative Systems Biology, School for Chemical Engineering and Analytical Sciences, University of Manchester, Manchester M1 7DN, UK
| |
Collapse
|
37
|
Gu J, Li X, Zhao L, Yang Y, Xue C, Gao Y, Li J, Han Q, Sun Z, Bai C, Zhao RC. The role of PKM2 nuclear translocation in the constant activation of the NF-κB signaling pathway in cancer-associated fibroblasts. Cell Death Dis 2021; 12:291. [PMID: 33731686 PMCID: PMC7969736 DOI: 10.1038/s41419-021-03579-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 02/23/2021] [Accepted: 02/25/2021] [Indexed: 12/24/2022]
Abstract
Cancer-associated fibroblasts (CAFs) play critical roles in cancer progression by regulating tumor cell proliferation, angiogenesis, and metastasis. Recent studies demonstrated that CAFs induce inhibitory immune cell infiltration and chemotherapy resistance in gastric cancer by activating the NF-κB signaling pathway to secrete IL6, IL8, and other inflammatory factors. Inhibition of the NF-κB signaling pathway in CAFs might be a potential therapeutic strategy in gastric cancer. However, how the NF-κB pathway is activated in CAFs remains unclear. We showed that mesenchymal stem cells (MSCs) differentiated into CAFs, induced by the exosomes derived from gastric cancer cells. During the process of differentiation from MSCs into CAFs, we showed that nuclear PKM2 expression was continuously upregulated and associated with NF-κB P65 acetylation, contributing to P65 nuclear retention in CAFs and constant transcription of IL-6, IL-8, and other inflammatory factors, thus promoting gastric cancer cell proliferation. We showed that NF-κB P65 acetylation was induced by P300. We showed that nuclear PKM2 was derived from exosomes of gastric cancer cell lines and the positive feedback loop induced by PKM2-P65 combination. It is also proved that P300 inhibitors can inhibit tumor proliferation in an AGS subcutaneous xenograft tumor model. Our study showed that gastric cancer cells influence the continuous activation of the NF-κB signaling pathway in CAFs by secreting gastric cancer exosomes containing PKM2, thus inducing abnormal metabolism and inflammation activation. This study provides a new therapeutic target for CAF normalization or deactivation strategies.
Collapse
Affiliation(s)
- Junjie Gu
- Department of Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100005, Beijing, China
| | - Xuechun Li
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory (No. BZO381), 100005, Beijing, China
| | - Lin Zhao
- Department of Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100005, Beijing, China
| | - Ying Yang
- Department of Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100005, Beijing, China
| | - Chunling Xue
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory (No. BZO381), 100005, Beijing, China
| | - Yang Gao
- Department of Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100005, Beijing, China
| | - Jing Li
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory (No. BZO381), 100005, Beijing, China
| | - Qin Han
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory (No. BZO381), 100005, Beijing, China.
| | - Zhao Sun
- Department of Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100005, Beijing, China.
| | - Chunmei Bai
- Department of Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100005, Beijing, China.
| | - Robert Chunhua Zhao
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory (No. BZO381), 100005, Beijing, China.
- School of Life Sciences, Shanghai University, 99 Shangda Road, 200444, Shanghai, China.
| |
Collapse
|
38
|
Damasceno LEA, Prado DS, Veras FP, Fonseca MM, Toller-Kawahisa JE, Rosa MH, Públio GA, Martins TV, Ramalho FS, Waisman A, Cunha FQ, Cunha TM, Alves-Filho JC. PKM2 promotes Th17 cell differentiation and autoimmune inflammation by fine-tuning STAT3 activation. J Exp Med 2021; 217:151965. [PMID: 32697823 PMCID: PMC7537396 DOI: 10.1084/jem.20190613] [Citation(s) in RCA: 165] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 05/28/2019] [Accepted: 05/28/2020] [Indexed: 01/15/2023] Open
Abstract
Th17 cell differentiation and pathogenicity depend on metabolic reprogramming inducing shifts toward glycolysis. Here, we show that the pyruvate kinase M2 (PKM2), a glycolytic enzyme required for cancer cell proliferation and tumor progression, is a key factor mediating Th17 cell differentiation and autoimmune inflammation. We found that PKM2 is highly expressed throughout the differentiation of Th17 cells in vitro and during experimental autoimmune encephalomyelitis (EAE) development. Strikingly, PKM2 is not required for the metabolic reprogramming and proliferative capacity of Th17 cells. However, T cell-specific PKM2 deletion impairs Th17 cell differentiation and ameliorates symptoms of EAE by decreasing Th17 cell-mediated inflammation and demyelination. Mechanistically, PKM2 translocates into the nucleus and interacts with STAT3, enhancing its activation and thereby increasing Th17 cell differentiation. Thus, PKM2 acts as a critical nonmetabolic regulator that fine-tunes Th17 cell differentiation and function in autoimmune-mediated inflammation.
Collapse
Affiliation(s)
- Luis Eduardo Alves Damasceno
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.,Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Douglas Silva Prado
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.,Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Flavio Protasio Veras
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.,Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Miriam M Fonseca
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.,Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Juliana E Toller-Kawahisa
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.,Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Marcos Henrique Rosa
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.,Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Gabriel Azevedo Públio
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.,Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Timna Varela Martins
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.,Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Fernando S Ramalho
- Department of Pathology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Fernando Queiroz Cunha
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.,Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Thiago Mattar Cunha
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.,Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - José Carlos Alves-Filho
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.,Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| |
Collapse
|
39
|
Li R, Ning X, He J, Lin Z, Su Y, Li R, Yin Y. Synthesis of novel sulfonamide derivatives containing pyridin-3-ylmethyl 4-(benzoyl)piperazine-1-carbodithioate moiety as potent PKM2 activators. Bioorg Chem 2021; 108:104653. [PMID: 33517002 DOI: 10.1016/j.bioorg.2021.104653] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 01/08/2021] [Accepted: 01/09/2021] [Indexed: 01/13/2023]
Abstract
Pyruvate kinase M2 isoform (PKM2) plays a key role in cancer progression through both metabolic and non-metabolic functions, thus it is recognized as a potential target for cancer diagnosis and treatment. In this study, we discovered a sulfonamide-dithiocarbamate compound 8a as a novel PKM2 activator from a random screening of an in-house compound library. Then, a series of lead compound 8a analogs were designed and synthesized for screening as potent PKM2 activators. Among them, compound 8b (AC50 = 0.136 µM) and 8k (AC50 = 0.056 µM) showed higher PKM2 activation activities than positive control NZT (AC50 = 0.228 µM), and they (IC50 < 1 µM) exhibited more significant anti-proliferative activities against human tumor cell lines than NZT (IC50 > 10 µM). Especially, compound 8k inhibited the proliferation of multiple cancer cells, but showed little toxicity on normal cells. In addition, we found that compound 8k inhibit the colony formation of MCF7 cells. Western blot analysis demonstrated that 8k could reduce PKM2 nuclear localization and block the downstream signaling pathway of PKM2, resulting in suppression of tumor cell proliferation. Overall, compound 8k may be a promising candidate for further mechanistic investigation of PKM2 and cancer therapy.
Collapse
Affiliation(s)
- Ridong Li
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, PR China.
| | - Xianling Ning
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, PR China
| | - Jianan He
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville Victoria 3010, Australia
| | - Zhiqiang Lin
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, PR China
| | - Yue Su
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, PR China
| | - Runtao Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, PR China
| | - Yuxin Yin
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, PR China; Peking-Tsinghua Center for Life Sciences, Peking University Health Science Center, Beijing 100191, PR China; Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, PR China.
| |
Collapse
|
40
|
Pan WW, Wubben TJ, Besirli CG. Photoreceptor metabolic reprogramming: current understanding and therapeutic implications. Commun Biol 2021; 4:245. [PMID: 33627778 PMCID: PMC7904922 DOI: 10.1038/s42003-021-01765-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 01/28/2021] [Indexed: 02/06/2023] Open
Abstract
Acquired and inherited retinal disorders are responsible for vision loss in an increasing proportion of individuals worldwide. Photoreceptor (PR) death is central to the vision loss individuals experience in these various retinal diseases. Unfortunately, there is a lack of treatment options to prevent PR loss, so an urgent unmet need exists for therapies that improve PR survival and ultimately, vision. The retina is one of the most energy demanding tissues in the body, and this is driven in large part by the metabolic needs of PRs. Recent studies suggest that disruption of nutrient availability and regulation of cell metabolism may be a unifying mechanism in PR death. Understanding retinal cell metabolism and how it is altered in disease has been identified as a priority area of research. The focus of this review is on the recent advances in the understanding of PR metabolism and how it is critical to reduction-oxidation (redox) balance, the outer retinal metabolic ecosystem, and retinal disease. The importance of these metabolic processes is just beginning to be realized and unraveling the metabolic and redox pathways integral to PR health may identify novel targets for neuroprotective strategies that prevent blindness in the heterogenous group of retinal disorders.
Collapse
Affiliation(s)
- Warren W Pan
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, USA
| | - Thomas J Wubben
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, USA.
| | - Cagri G Besirli
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
41
|
Autophagy activation and photoreceptor survival in retinal detachment. Exp Eye Res 2021; 205:108492. [PMID: 33609513 DOI: 10.1016/j.exer.2021.108492] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 12/14/2022]
Abstract
We assess the effect of autophagy inhibition on photoreceptor (PR) survival during experimental retinal detachment (RD) and examine the and examine the relationship between autophagy and the expression of glycolytic enzymes HK2 and PKM2 in the retina. We find that inhibiting autophagy by genetic knock out of the autophagy activator Atg5 in rod PRs resulted in increased apoptotic and necroptotic cell death during RD, demonstrated by elevated terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL)-positive cells, caspase 8 activity, transcript levels of Fas receptor and RIPK3 as compared to controls. The absence of autophagy in rods resulted in downregulation of hexokinase 2 and pyruvate kinase muscle isozyme 2 levels. More than 460 proteins were identified by mass spectroscopy in autophagosomes isolated from detached retinas compared with less than 150 proteins identified in autophagosomes from attached retinas. Among various cellular compartments, proteins from cytoskeleton, cytoplasm and intracellular organelles constituted a large portion of increased autophagosome contents. These proteins represent numerous biological processes, including phototransduction, cell-cell signaling, metabolism and inflammation. Our findings suggest that competent autophagy machinery is necessary for PR homeostasis and improving PR survival during periods of nutrient deprivation.
Collapse
|
42
|
Evaluation of Glycolytic Response to Multiple Classes of Anti-glioblastoma Drugs by Noninvasive Measurement of Pyruvate Kinase M2 Using [ 18F]DASA-23. Mol Imaging Biol 2021; 22:124-133. [PMID: 30989436 DOI: 10.1007/s11307-019-01353-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE Pyruvate kinase M2 (PKM2) catalyzes the final step in glycolysis, the key process of tumor metabolism. PKM2 is found in high levels in glioblastoma (GBM) cells with marginal expression within healthy brain tissue, rendering it a key biomarker of GBM metabolic re-programming. Our group has reported the development of a novel radiotracer, 1-((2-fluoro- 6-[18F]fluorophenyl)sulfonyl)-4-((4-methoxyphenyl)sulfonyl)piperazine ([18F]DASA- 23), to non-invasively detect PKM2 levels with positron emission tomography (PET). PROCEDURE U87 human GBM cells were treated with the IC50 concentration of various agents used in the treatment of GBM, including alkylating agents (temozolomide, carmustine, lomustine, procarbazine), inhibitor of topoisomerase I (irinotecan), vascular endothelial and epidermal growth factor receptor inhibitors (cediranib and erlotinib, respectively) anti-metabolite (5-fluorouracil), microtubule inhibitor (vincristine), and metabolic agents (dichloroacetate and IDH1 inhibitor ivosidenib). Following drug exposure for three or 6 days (n = 6 replicates per condition), the radiotracer uptake of [18F]DASA-23 and 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) was assessed. Changes in PKM2 protein levels were determined via Western blot and correlated to radiotracer uptake. RESULTS Significant interactions were found between the treatment agent (n = 12 conditions total comprised 11 drugs and vehicle) and the duration of treatment (3- or 6-day exposure to each drug) on the cellular uptake of [18F]DASA-23 (p = 0.0001). The greatest change in the cellular uptake of [18F]DASA-23 was found after exposure to alkylating agents (p < 0. 0001) followed by irinotecan (p = 0. 0012), erlotinib (p = 0. 02), and 5-fluorouracil (p = 0. 005). Correlation of PKM2 protein levels and [18F]DASA-23 cellular uptake revealed a moderate correlation (r = 0.44, p = 0.15). CONCLUSIONS These proof of principle studies emphasize the superiority of [18F]DASA-23 to [18F]FDG in detecting the glycolytic response of GBM to multiple classes of anti-neoplastic drugs in cell culture. A clinical trial evaluating the diagnostic utility of [18F]DASA-23 PET in GBM patients (NCT03539731) is ongoing.
Collapse
|
43
|
Patel CB, Beinat C, Xie Y, Chang E, Gambhir SS. Tumor treating fields (TTFields) impairs aberrant glycolysis in glioblastoma as evaluated by [ 18F]DASA-23, a non-invasive probe of pyruvate kinase M2 (PKM2) expression. Neoplasia 2021; 23:58-67. [PMID: 33221711 PMCID: PMC7689378 DOI: 10.1016/j.neo.2020.11.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 12/27/2022]
Abstract
Despite the anti-proliferative and survival benefits from tumor treating fields (TTFields) in human glioblastoma (hGBM), little is known about the effects of this form of alternating electric fields therapy on the aberrant glycolysis of hGBM. [18F]FDG is the most common radiotracer in cancer metabolic imaging, but its utility in hGBM is impaired due to high glucose uptake in normal brain tissue. With TTFields, radiochemistry, Western blot, and immunofluorescence microscopy, we identified pyruvate kinase M2 (PKM2) as a biomarker of hGBM response to therapeutic TTFields. We used [18F]DASA-23, a novel radiotracer that measures PKM2 expression and which has been shown to be safe in humans, to detect a shift away from hGBM aberrant glycolysis in response to TTFields. Compared to unexposed hGBM, [18F]DASA-23 uptake was reduced in hGBM exposed to TTFields (53%, P< 0.05) or temozolomide chemotherapy (33%, P > 0.05) for 3 d. A 6-d TTFields exposure resulted in a 31% reduction (P = 0.043) in 60-min uptake of [18F]DASA-23. [18F]DASA-23 was retained after a 10 but not 30-min wash-out period. Compared to [18F]FDG, [18F]DASA-23 demonstrated a 4- to 9-fold greater uptake, implying an improved tumor-to-background ratio. Furthermore, compared to no-TTFields exposure, a 6-d TTFields exposure caused a 35% reduction in [18F]DASA-23 30-min uptake compared to only an 8% reduction in [18F]FDG 30-min uptake. Quantitative Western blot analysis and qualitative immunofluorescence for PKM2 confirmed the TTFields-induced reduction in PKM2 expression. This is the first study to demonstrate that TTFields impairs hGBM aberrant glycolytic metabolism through reduced PKM2 expression, which can be non-invasively detected by the [18F]DASA-23 radiotracer.
Collapse
Affiliation(s)
- Chirag B Patel
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA; Division of Adult Neuro-Oncology, Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.
| | - Corinne Beinat
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yuanyang Xie
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Edwin Chang
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Sanjiv S Gambhir
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA; Departments of Bioengineering and Materials Science & Engineering, Stanford University, Stanford, CA, USA
| |
Collapse
|
44
|
Maugard M, Vigneron PA, Bolaños JP, Bonvento G. l-Serine links metabolism with neurotransmission. Prog Neurobiol 2020; 197:101896. [PMID: 32798642 DOI: 10.1016/j.pneurobio.2020.101896] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 07/20/2020] [Accepted: 08/09/2020] [Indexed: 12/12/2022]
Abstract
Brain energy metabolism is often considered as a succession of biochemical steps that metabolize the fuel (glucose and oxygen) for the unique purpose of providing sufficient ATP to maintain the huge information processing power of the brain. However, a significant fraction (10-15 %) of glucose is shunted away from the ATP-producing pathway (oxidative phosphorylation) and may be used to support other functions. Recent studies have pointed to the marked compartmentation of energy metabolic pathways between neurons and glial cells. Here, we focused our attention on the biosynthesis of l-serine, a non-essential amino acid that is formed exclusively in glial cells (mostly astrocytes) by re-routing the metabolic fate of the glycolytic intermediate, 3-phosphoglycerate (3PG). This metabolic pathway is called the phosphorylated pathway and transforms 3PG into l-serine via three enzymatic reactions. We first compiled the available data on the mechanisms that regulate the flux through this metabolic pathway. We then reviewed the current evidence that is beginning to unravel the roles of l-serine both in the healthy and diseased brain, leading to the notion that this specific metabolic pathway connects glial metabolism with synaptic activity and plasticity. We finally suggest that restoring astrocyte-mediated l-serine homeostasis may provide new therapeutic strategies for brain disorders.
Collapse
Affiliation(s)
- Marianne Maugard
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | - Pierre-Antoine Vigneron
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | - Juan P Bolaños
- Institute of Functional Biology and Genomics (IBFG), Universidad de Salamanca, CSIC, Salamanca, Spain; Centro de Investigación Biomédica en Red sobre Fragilidad y Envejecimiento Saludable (CIBERFES), Institute of Biomedical Research of Salamanca, 37007, Salamanca, Spain
| | - Gilles Bonvento
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France.
| |
Collapse
|
45
|
Zahra K, Dey T, Ashish, Mishra SP, Pandey U. Pyruvate Kinase M2 and Cancer: The Role of PKM2 in Promoting Tumorigenesis. Front Oncol 2020; 10:159. [PMID: 32195169 PMCID: PMC7061896 DOI: 10.3389/fonc.2020.00159] [Citation(s) in RCA: 318] [Impact Index Per Article: 63.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 01/29/2020] [Indexed: 12/17/2022] Open
Abstract
Pyruvate kinase plays a pivotal role in regulating cell metabolism. The final and rate-limiting step of glycolysis is the conversion of Phosphoenolpyruvate (PEP) to Pyruvate, which is catalyzed by Pyruvate Kinase. There are four isomeric, tissue-specific forms of Pyruvate Kinase found in mammals: PKL, PKR, PKM1, and PKM2. PKM1 and PKM2 are formed bya single mRNA transcript of the PKM gene by alternative splicing. The oligomers of PKM2 exist in high activity tetramer and low activity dimer forms. The dimer PKM2 regulates the rate-limiting step of glycolysis that shifts the glucose metabolism from the normal respiratory chain to lactate production in tumor cells. Besides its role as a metabolic regulator, it also acts as protein kinase, which contributes to tumorigenesis. This review is focused on the metabolic role of pyruvate kinase M2 in normal cells vs. cancerous cells and its regulation at the transcriptional level. The review also highlights the role of PKM2 as a potential diagnostic marker and as a therapeutic target in cancer treatment.
Collapse
Affiliation(s)
- Kulsoom Zahra
- Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Tulika Dey
- Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Ashish
- Department of Anatomy, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Surendra Pratap Mishra
- Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Uma Pandey
- Department of Obstetrics and Gynecology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
46
|
Wubben TJ, Pawar M, Weh E, Smith A, Sajjakulnukit P, Zhang L, Dai L, Hager H, Pai MP, Lyssiotis CA, Besirli CG. Small molecule activation of metabolic enzyme pyruvate kinase muscle isozyme 2, PKM2, circumvents photoreceptor apoptosis. Sci Rep 2020; 10:2990. [PMID: 32076076 PMCID: PMC7031539 DOI: 10.1038/s41598-020-59999-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 02/06/2020] [Indexed: 01/22/2023] Open
Abstract
Photoreceptor cell death is the ultimate cause of vision loss in many retinal disorders, and there is an unmet need for neuroprotective modalities to improve photoreceptor survival. Similar to cancer cells, photoreceptors maintain pyruvate kinase muscle isoform 2 (PKM2) expression, which is a critical regulator in aerobic glycolysis. Unlike PKM1, which has constitutively high catalytic activity, PKM2 is under complex regulation. Recently, we demonstrated that genetically reprogramming photoreceptor metabolism via PKM2-to-PKM1 substitution is a promising neuroprotective strategy. Here, we explored the neuroprotective effects of pharmacologically activating PKM2 via ML-265, a small molecule activator of PKM2, during acute outer retinal stress. We found that ML-265 increased PKM2 activity in 661 W cells and in vivo in rat eyes without affecting the expression of genes involved in glucose metabolism. ML-265 treatment did, however, alter metabolic intermediates of glucose metabolism and those necessary for biosynthesis in cultured cells. Long-term exposure to ML-265 did not result in decreased photoreceptor function or survival under baseline conditions. Notably, though, ML-265-treatment did reduce entrance into the apoptotic cascade in in vitro and in vivo models of outer retinal stress. These data suggest that reprogramming metabolism via activation of PKM2 is a novel, and promising, therapeutic strategy for photoreceptor neuroprotection.
Collapse
Affiliation(s)
- Thomas J Wubben
- University of Michigan, Kellogg Eye Center, Department of Ophthalmology and Visual Sciences, Ann Arbor, USA
| | - Mercy Pawar
- University of Michigan, Kellogg Eye Center, Department of Ophthalmology and Visual Sciences, Ann Arbor, USA
| | - Eric Weh
- University of Michigan, Kellogg Eye Center, Department of Ophthalmology and Visual Sciences, Ann Arbor, USA
| | - Andrew Smith
- University of Michigan, Kellogg Eye Center, Department of Ophthalmology and Visual Sciences, Ann Arbor, USA
| | - Peter Sajjakulnukit
- University of Michigan, Department of Molecular and Integrative Physiology, Ann Arbor, USA
| | - Li Zhang
- University of Michigan Biomedical Research Core Facilities, Metabolomics Core, Ann Arbor, USA
| | - Lipeng Dai
- University of Michigan, College of Pharmacy, Ann Arbor, USA
| | - Heather Hager
- University of Michigan, Kellogg Eye Center, Department of Ophthalmology and Visual Sciences, Ann Arbor, USA
| | | | - Costas A Lyssiotis
- University of Michigan, Department of Molecular and Integrative Physiology, Ann Arbor, USA
| | - Cagri G Besirli
- University of Michigan, Kellogg Eye Center, Department of Ophthalmology and Visual Sciences, Ann Arbor, USA.
| |
Collapse
|
47
|
Pyridoxine induces glutathione synthesis via PKM2-mediated Nrf2 transactivation and confers neuroprotection. Nat Commun 2020; 11:941. [PMID: 32071304 PMCID: PMC7029000 DOI: 10.1038/s41467-020-14788-x] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 02/04/2020] [Indexed: 12/20/2022] Open
Abstract
Oxidative stress is a major pathogenic mechanism in Parkinson's disease (PD). As an important cellular antioxidant, glutathione (GSH) balances the production and incorporation of free radicals to protect neurons from oxidative damage. GSH level is decreased in the brains of PD patients. Hence, clarifying the molecular mechanism of GSH deficiency may help deepen our knowledge of PD pathogenesis. Here we report that the astrocytic dopamine D2 receptor (DRD2) regulates GSH synthesis via PKM2-mediated Nrf2 transactivation. In addition we find that pyridoxine can dimerize PKM2 to promote GSH biosynthesis. Further experiments show that pyridoxine supplementation increases the resistance of nigral dopaminergic neurons to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced neurotoxicity in wild-type mice as well as in astrocytic Drd2 conditional knockout mice. We conclude that dimerizing PKM2 may be a potential target for PD treatment.
Collapse
|
48
|
Liu VM, Howell AJ, Hosios AM, Li Z, Israelsen WJ, Heiden MGV. Cancer-associated mutations in human pyruvate kinase M2 impair enzyme activity. FEBS Lett 2020; 594:646-664. [PMID: 31642061 PMCID: PMC7042059 DOI: 10.1002/1873-3468.13648] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 09/28/2019] [Accepted: 10/15/2019] [Indexed: 12/18/2022]
Abstract
Mammalian pyruvate kinase catalyzes the final step of glycolysis, and its M2 isoform (PKM2) is widely expressed in proliferative tissues. Mutations in PKM2 are found in some human cancers; however, the effects of these mutations on enzyme activity and regulation are unknown. Here, we characterized five cancer-associated PKM2 mutations, occurring at various locations on the enzyme, with respect to substrate kinetics and activation by the allosteric activator fructose-1,6-bisphosphate (FBP). The mutants exhibit reduced maximal velocity, reduced substrate affinity, and/or altered activation by FBP. The kinetic parameters of five additional PKM2 mutants that have been used to study enzyme function or regulation also demonstrate the deleterious effects of mutations on PKM2 function. Our findings indicate that PKM2 is sensitive to many amino acid changes and support the hypothesis that decreased PKM2 activity is selected for in rapidly proliferating cells.
Collapse
Affiliation(s)
- Vivian M. Liu
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States
- Harvard-MIT Health Sciences and Technology Division, Harvard Medical School, Boston, MA 02115, United States
| | - Andrea J. Howell
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States
| | - Aaron M. Hosios
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States
| | - Zhaoqi Li
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States
| | - William J. Israelsen
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Matthew G. Vander Heiden
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States
- Dana-Farber Cancer Institute, Boston, MA 02115, United States
| |
Collapse
|
49
|
Serpa J. Metabolic Remodeling as a Way of Adapting to Tumor Microenvironment (TME), a Job of Several Holders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1219:1-34. [PMID: 32130691 DOI: 10.1007/978-3-030-34025-4_1] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The microenvironment depends and generates dependence on all the cells and structures that share the same niche, the biotope. The contemporaneous view of the tumor microenvironment (TME) agrees with this idea. The cells that make up the tumor, whether malignant or not, behave similarly to classes of elements within a living community. These elements inhabit, modify and benefit from all the facilities the microenvironment has to offer and that will contribute to the survival and growth of the tumor and the progression of the disease.The metabolic adaptation to microenvironment is a crucial process conducting to an established tumor able to grow locally, invade and metastasized. The metastatic cancer cells are reasonable more plastic than non-metastatic cancer cells, because the previous ones must survive in the microenvironment where the primary tumor develops and in addition, they must prosper in the microenvironment in the metastasized organ.The metabolic remodeling requires not only the adjustment of metabolic pathways per se but also the readjustment of signaling pathways that will receive and obey to the extracellular instructions, commanding the metabolic adaptation. Many diverse players are pivotal in cancer metabolic fitness from the initial signaling stimuli, going through the activation or repression of genes, until the phenotype display. The new phenotype will permit the import and consumption of organic compounds, useful for energy and biomass production, and the export of metabolic products that are useless or must be secreted for a further recycling or controlled uptake. In the metabolic network, three subsets of players are pivotal: (1) the organic compounds; (2) the transmembrane transporters, and (3) the enzymes.This chapter will present the "Pharaonic" intent of diagraming the interplay between these three elements in an attempt of simplifying and, at the same time, of showing the complex sight of cancer metabolism, addressing the orchestrating role of microenvironment and highlighting the influence of non-cancerous cells.
Collapse
Affiliation(s)
- Jacinta Serpa
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School | Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal.
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Lisbon, Portugal.
| |
Collapse
|
50
|
Kumar S, Patel AK. Purification and Characterization of Prolyl Hydroxylase 3/Pyruvate Kinase Isoform 2 Protein Complex. Mol Biotechnol 2019; 62:111-118. [PMID: 31760602 DOI: 10.1007/s12033-019-00228-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The prolyl hydroxylase 3 (PHD3) protein is less abundant in normal oxygen conditions (normoxia) but increases under deficient oxygen condition (hypoxia). Since cancerous cells often thrive in hypoxic conditions and predominantly express the Pyruvate kinase isoforms 2 (PKM2), the PHD3/PKM2 interaction might be particularly important in cancer development. In the present study, the PHD3/PKM2 complex was co-expressed and purified by size-exclusion chromatography. The interaction of PHD3 with PKM2 was confirmed in Native gel as well as western blot analysis. The PHD3/PKM2 complex formed discreet crystals under suitable conditions, and diffraction data revealed that crystal belonged to the P1 space group with 3.0 Å resolution. This is the first crystal report of PHD3/PKM2 complex as well as this study demonstrates a direct physical binding through protein-protein interaction. The structural analysis of complex will provide the information regarding the amino acid residues critical for the catalytic mechanism. Based on the structural information thus obtained, pharmacological interference with the PHD3/PKM2 interaction could be used as a novel strategy to reduce the cancer progression.
Collapse
Affiliation(s)
- Sunil Kumar
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Ashok Kumar Patel
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India.
| |
Collapse
|