1
|
Liu L, Xing G, Guo X, Chen H, Li J, Wang J, Li Y, Liang G, Liu M. Inhibition of colorectal cancer cell growth by downregulation of M2-PK and reduction of aerobic glycolysis by clove active ingredients. Front Pharmacol 2025; 16:1552486. [PMID: 40308769 PMCID: PMC12041220 DOI: 10.3389/fphar.2025.1552486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Accepted: 03/24/2025] [Indexed: 05/02/2025] Open
Abstract
Exploring the anti-tumor molecular mechanisms of traditional Chinese medicines has become an important strategy to develop novel anti-tumor drugs in the clinic. Several pharmacological studies have reported the antioxidant, antibacterial, anti-inflammatory, and anti-tumor effects of clove. Previously, we have shown that the active fraction from clove (AFC) can inhibit the growth of tumor cells, particularly colon cancer cells, in vitro. However, the mechanism of action regarding the anti-colon cancer activity of AFC, especially in aerobic glycolysis, has not been adequately investigated. In this study, we found that AFC significantly inhibited the growth of five types of colon cancer cells, downregulated the mRNA and protein levels of M2-type pyruvate kinase (PKM2), and reduced aerobic glycolysis capacity. Transfection of PKM2-siRNA mimicked the inhibitory effects of AFC on aerobic glycolysis in colon cancer cells. Furthermore, the highly expressed, tumor-specific targets c-myc and cyclin D1 in cells were also found to be downregulated following the action of AFC. In the HCT116 cell xenograft nude mice models, the results after AFC administration were consistent with those of the cellular experiments, while AFC caused less liver injury and weight loss than the conventional chemotherapeutic agent 5- fluorouracil (5-FU). In conclusion, AFC inhibits colon cancer growth by downregulating PKM2 to inhibit aerobic glycolysis and reduce the tumor-specific high expression of c-myc and cyclin D1. Future work should explore how it downregulates pyruvate kinase (PK) in the first place, along with the intrinsic mechanism between the downregulation of PKM2 and the downregulation of c-myc.
Collapse
Affiliation(s)
- Lin Liu
- School of Pharmacy, Southwest Medical University, Luzhou, China
- Drug Dispending Department, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, China
| | - Gang Xing
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xiaoyi Guo
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Hui Chen
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jian Li
- Pharmacy Department, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Jian Wang
- Discipline Construction Office, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Yaling Li
- Pharmacy Department, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Gang Liang
- Pharmacy Department, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Minghua Liu
- School of Pharmacy, Southwest Medical University, Luzhou, China
| |
Collapse
|
2
|
Carmona OG, Kleinjung J, Anastasiou D, Oostenbrink C, Fraternali F. AllohubPy: Detecting Allosteric Signals Through An Information-theoretic Approach. J Mol Biol 2025:168969. [PMID: 39900284 DOI: 10.1016/j.jmb.2025.168969] [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: 12/15/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 02/05/2025]
Abstract
Allosteric regulation is crucial for biological processes like signal transduction, transcriptional regulation, and metabolism, yet the mechanisms and macromolecular properties that govern it are still not well understood. Several methods have been developed over the years to study allosterism through different angles. Among the possible ways to study allosterism, information-theoretic approaches, like AlloHubMat or GSAtools, can be particularly effective due to their use of robust statistics and the possibility to be combined with graph analysis. These methods capture local conformational changes associated with global motions from molecular dynamics simulations through the use of a Structural Alphabet, which simplifies the complexity of the Cartesian space by reducing the dimensionality down to a string of encoded fragments, representing sets of internal coordinates that still capture the overall conformation changes. In this work, we present "AllohubPy," an improved and standardized methodology of AlloHubMat and GSAtools coded in Python. We analyse the performance, limitations and sampling requirements of AllohubPy by using extensive molecular dynamics simulations of model allosteric systems and apply convergence analysis techniques to estimate result reliability. Additionally, we expand the methodology to use different dimensionality reduction Structural Alphabets, such as the 3DI alphabet, and integrate Protein Language Models (PLMs) to refine allosteric hub communication detection by monitoring the detected evolutionary constraints. Overall, AllohubPy expands its preceding methods and simplifies the use and reliability of the method to effectively capture dynamic allosteric motions and residue pathways. AllohubPy is freely available on GitHub (https://github.com/Fraternalilab/AlloHubPy) as a package and as a Jupyter Notebook.
Collapse
Affiliation(s)
- Oriol Gracia Carmona
- Department of Structural and Molecular Biology, Division of Biosciences and Institute of Structural and Molecular Biology, University College London, London WC1E 6BT, United Kingdom; Department of Biological Sciences Birkbeck, University of London, London WC1E 7HX, United Kingdom; Randall Centre for Cell & Molecular Biosciences, King's College London, London SE1 1UL, United Kingdom
| | - Jens Kleinjung
- Nxera Pharma, Steinmetz & Cori Buildings, Granta Park, Great Abington, Cambridge CB21 6DG, United Kingdom
| | - Dimitrios Anastasiou
- Cancer Metabolism Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom
| | - Chris Oostenbrink
- Institute for Molecular Modeling and Simulation, Department of Material Sciences and Process Engineering, BOKU University 1190 Vienna, Austria
| | - Franca Fraternali
- Department of Structural and Molecular Biology, Division of Biosciences and Institute of Structural and Molecular Biology, University College London, London WC1E 6BT, United Kingdom; Department of Biological Sciences Birkbeck, University of London, London WC1E 7HX, United Kingdom.
| |
Collapse
|
3
|
Zhu C, Yang J, Liu L, Li B, Sun T, Sheng W, He Q. Bibliometric analysis of glycolysis and prostate cancer research from 2004 to 2024. Discov Oncol 2025; 16:34. [PMID: 39800812 PMCID: PMC11725561 DOI: 10.1007/s12672-025-01790-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 01/08/2025] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Prostate cancer (PCa) ranks as the second most common disease among men and the fourth most prevalent cancer worldwide. Enhanced glycolysis and excessive lactate secretion are recognized as critical factors driving the progression of various cancers. This study systematically investigated the research trends associated with glycolysis in PCa through bibliometric analysis. METHOD In this study, we conducted a systematic search of the Web of Science and PubMed databases for literature pertaining to the glycolysis of PCa that was published between January 1, 2004, and June 30, 2024. To achieve this objective, we employed CiteSpace software to generate visualizations that illustrate countries/regions, institutions, journals, and keywords. Additionally, we extracted pertinent quantitative data. Furthermore, we utilized VOSviewer software to create a collaboration network map among various journals. RESULTS Between 2004 and 2024, a total of 408 research articles on glycolysis in PCa were published, indicating a consistent upward trend in the annual publication rate. In this field, the United States not only leads in the volume of research papers but also has the highest degree of centrality. The journal "Cancer Research" is recognized as the most influential in the field, whereas "Prostate and Cancer" serves as a significant platform for disseminating research related to glycolysis in PCa. Keyword analysis has identified four primary research directions that have dominated this field over the past two decades. The role of glycolysis and its associated enzymes in PCa underpins this research. Glycolysis has also demonstrated significant clinical value in the diagnosis and prognosis of PCa. Moreover, drugs targeting glycolytic inhibitors and natural products have exhibited therapeutic potential against this disease. By modulating glycolytic mechanisms, there is potential to increase resistance in PCa. Currently, leading research in this area encompasses the application of nanotechnology to PCa glycolysis, the roles of long noncoding RNAs (lncRNAs) and microRNAs (miRNAs) in this metabolic pathway, and the interactions between glycolysis and other biological processes in PCa. CONCLUSION This study employs bibliometric analysis to provide a comprehensive overview of research on glycolysis in PCa over the past two decades. It highlights the current state of knowledge in this field, identifies key research hotspots, and explores emerging frontiers, particularly nanotechnology, lncRNA, and miRNA, which are driving innovative research directions.
Collapse
Affiliation(s)
- Congxu Zhu
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, No. 300 Bachelor's Road, Changsha, 410208, China
| | - Jingjing Yang
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, No. 300 Bachelor's Road, Changsha, 410208, China
| | - Lumei Liu
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, No. 300 Bachelor's Road, Changsha, 410208, China
| | - Bonan Li
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, No. 300 Bachelor's Road, Changsha, 410208, China
- Hunan Normal University Affiliated Changsha Hospital, No. 200 North Jinxing Road, Changsha, 410023, China
| | - Tiansong Sun
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, No. 300 Bachelor's Road, Changsha, 410208, China
| | - Wen Sheng
- School of Rehabilitation Medicine and Health Care, Hunan University of Medicine, No. 492 Jinxi South Road, Huaihua, 418000, China.
| | - Qinghu He
- School of Rehabilitation Medicine and Health Care, Hunan University of Medicine, No. 492 Jinxi South Road, Huaihua, 418000, China.
| |
Collapse
|
4
|
Liao Q, Deng J, Tong J, Gan Y, Hong W, Dong H, Cao M, Xiong C, Chen Y, Xie B, Yang FY, Alifu A, Zhou GB, Huang S, Xiong J, Hao Q, Zhou X. p53 induces circFRMD4A to suppress cancer development through glycolytic reprogramming and cuproptosis. Mol Cell 2025; 85:132-149.e7. [PMID: 39637854 DOI: 10.1016/j.molcel.2024.11.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 09/15/2024] [Accepted: 11/08/2024] [Indexed: 12/07/2024]
Abstract
Cuproptosis is a type of copper-induced cell death that mainly impacts cells relying on mitochondrial metabolism. Although p53 regulates glycolytic metabolism, its role in cuproptosis remains unclear. Here, we report that the circular RNA, circFRMD4A, is crucial for p53-mediated metabolic reprogramming and cuproptosis. CircFRMD4A originates from the transcript of FRMD4A, which is transcriptionally activated by p53, and the formation of circFRMD4A is facilitated by the RNA-binding protein EWSR1. CircFRMD4A functions as a tumor suppressor and enhances the sensitivity of cancer cells to elesclomol-induced cuproptosis. Mechanistic analysis reveals that circFRMD4A interacts with and inactivates the pyruvate kinase PKM2, leading to a decrease in lactate production and a redirection of glycolytic flux toward the tricarboxylic acid cycle. Finally, p53 agonists and elesclomol coordinately suppress the growth of cancer in a xenograft mouse model. Altogether, our study uncovers that p53 promotes glycolytic reprogramming and cuproptosis via circFRMD4A and suggests a potential combination strategy against cancers with wild-type p53.
Collapse
Affiliation(s)
- Quan Liao
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China; Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang 330006, Jiangxi, China
| | - Jun Deng
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China; Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang 330006, Jiangxi, China
| | - Jing Tong
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yu Gan
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Weiwei Hong
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Hanzhi Dong
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China; Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang 330006, Jiangxi, China
| | - Mingming Cao
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Chen Xiong
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yajie Chen
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Bangxiang Xie
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Fu-Ying Yang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Aikede Alifu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guang-Biao Zhou
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shenglin Huang
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, China; Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Jianping Xiong
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China; Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang 330006, Jiangxi, China.
| | - Qian Hao
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Xiang Zhou
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, China; Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China.
| |
Collapse
|
5
|
Hu Y, Tang J, Xu Q, Fang Z, Li R, Yang M, Zhao J, Chen X. Role of pyruvate kinase M2 in regulating sepsis (Review). Mol Med Rep 2024; 30:185. [PMID: 39155878 DOI: 10.3892/mmr.2024.13309] [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: 06/11/2024] [Accepted: 07/29/2024] [Indexed: 08/20/2024] Open
Abstract
Glycolysis occurs in all living organisms as a form of energy supply. Pyruvate kinase M2 (PKM2) is one of the rate‑limiting enzymes in the glycolytic process. PKM2 is considered to serve an important role in several terminal diseases, including sepsis. However, to the best of our knowledge, the specific mechanistic role of PKM2 in sepsis remains to be systematically summarised. Therefore, the present review aims to summarise the roles of PKM2 in sepsis progression. In addition, potential treatment strategies for patients with sepsis are discussed. The present review hopes to lay the groundwork for studying the role of PKM2 and developing therapeutic strategies against metabolic disorders that occur during sepsis.
Collapse
Affiliation(s)
- Yifei Hu
- Department of Clinical Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang 321000, P.R. China
| | - Jing Tang
- Department of Clinical Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang 321000, P.R. China
| | - Qiao Xu
- Department of Clinical Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang 321000, P.R. China
| | - Zenghui Fang
- Department of Clinical Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang 321000, P.R. China
| | - Rongqing Li
- Department of Clinical Medicine, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu 225000, P.R. China
| | - Mengxuan Yang
- Department of Clinical Laboratory, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang 310000, P.R. China
| | - Jie Zhao
- Department of Clinical Medicine, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu 225000, P.R. China
| | - Xin Chen
- Department of Clinical Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang 321000, P.R. China
| |
Collapse
|
6
|
Jemal M, Getinet M, Amare GA, Tegegne BA, Baylie T, Mengistu EF, Osman EE, Chura Waritu N, Adugna A. Non-metabolic enzyme function of pyruvate kinase M2 in breast cancer. Front Oncol 2024; 14:1450325. [PMID: 39411137 PMCID: PMC11473492 DOI: 10.3389/fonc.2024.1450325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 09/12/2024] [Indexed: 10/19/2024] Open
Abstract
Breast cancer (BC) is a prevalent malignant tumor in women, and its incidence has been steadily increasing in recent years. Compared with other types of cancer, it has the highest mortality and morbidity rates in women. So, it is crucial to investigate the underlying mechanisms of BC development and identify specific therapeutic targets. Pyruvate kinase M2 (PKM2), an important metabolic enzyme in glycolysis, has been found to be highly expressed in BC. It can also move to the nucleus and interact with various transcription factors and proteins, including hypoxia-inducible factor-1α (HIF-1α), signal transducer and activator of transcription 3 (STAT3), β-catenin, cellular-myelocytomatosis oncogene (c-Myc), nuclear factor kappa-light-chain enhancer of activated B cells (NF-κB), and mammalian sterile 20-like kinase 1 (MST1). This interaction leads to non-metabolic functions that control the cell cycle, proliferation, apoptosis, migration, invasion, angiogenesis, and tumor microenvironment in BC. This review provides an overview of the latest advancements in understanding the interactions between PKM2 and different transcription factors and proteins that influence the initiation and progression of BC. It also examined how natural drugs and noncoding RNAs affect various biological processes in BC cells through the regulation of the non-metabolic enzyme functions of PKM2. The findings provide valuable insights for improving the prognosis and developing targeted therapies for BC in the coming years.
Collapse
Affiliation(s)
- Mohammed Jemal
- Department of Biomedical Science, School of Medicine, Debre Markos University, Debre Markos, Ethiopia
| | - Mamaru Getinet
- Department of Biomedical Science, School of Medicine, Debre Markos University, Debre Markos, Ethiopia
| | - Gashaw Azanaw Amare
- Department of Medical Laboratory Sciences, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Bantayehu Addis Tegegne
- Department of Pharmacy, College of Medicine and Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Temesgen Baylie
- Department of Biomedical Science, School of Medicine, Debre Markos University, Debre Markos, Ethiopia
| | - Enyew Fenta Mengistu
- Department of Biomedical Science, School of Medicine, Debre Markos University, Debre Markos, Ethiopia
| | - Enatnesh Essa Osman
- Department of Biomedical Science, School of Medicine, Debre Markos University, Debre Markos, Ethiopia
| | - Nuredin Chura Waritu
- Department of Biomedical Sciences, School of Medicine, Wolaita Sodo University, Wolaita Sodo, Ethiopia
| | - Adane Adugna
- Department of Medical Laboratory Sciences, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| |
Collapse
|
7
|
Wu J, Ding Z, Zhong M, Xi J, He Y, Zhang B, Fang J. Polyphyllin II Induces Apoptosis in Fibrosarcoma Cells via Activating Pyruvate Kinase M2. Chem Res Toxicol 2024; 37:1394-1403. [PMID: 39066737 DOI: 10.1021/acs.chemrestox.4c00154] [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: 07/30/2024]
Abstract
Aerobic glycolysis is a metabolic reprogramming of tumor cells that is essential for sustaining their phenotype of fast multiplication by continuously supplying energy and mass. Pyruvate kinase M2 (PKM2) has a vital role in this process, which has given it high interest as a target for anticancer drug development. With potent toxicity to many types of cancer cells, polyphyllin II (PP2), a steroidal saponin isolated from the herbaceous plant Rhizoma paridis, brought to our attention that it might interfere with the PKM2 activity. In this study, we discovered that PP2 was a novel agonist of PKM2. PP2 activated recombinant PKM2 and changed the protein's oligomeric state to activate intracellular PKM2. At the same time, PP2 suppressed its protein kinase function by decreasing the content of nuclear PKM2. The mRNA levels of its downstream genes, such as Glut1, LDHA, and MYC, were inhibited. In addition, PP2 induced oxidative stress by downregulating the expression and activity of antioxidant proteins such as NQO1, TrxR, and Trx in HT-1080 cells, which in turn led to mitochondrial dysfunction and ultimately induced apoptosis. Moreover, PP2 reduced the proliferation and migration of HT-1080 cells. Thus, targeting the glycolysis pathway offers an unprecedented mode of action for comprehending PP2's pharmacological impacts and advances PP2's further development in fibrosarcoma therapy.
Collapse
Affiliation(s)
- Jun Wu
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Zhenjiang Ding
- Beijing Key Laboratory of the Innovative Development of Functional Staple and Nutritional Intervention for Chronic Diseases, China National Research Institute of Food and Fermentation Industries, Beijing 100015, China
| | - Miao Zhong
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Junmin Xi
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Ying He
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Baoxin Zhang
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Jianguo Fang
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing 210094 , Jiangsu, China
| |
Collapse
|
8
|
Ma L, Li H, Xu H, Liu D. The potential roles of PKM2 in cerebrovascular diseases. Int Immunopharmacol 2024; 139:112675. [PMID: 39024754 DOI: 10.1016/j.intimp.2024.112675] [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: 05/29/2024] [Revised: 07/06/2024] [Accepted: 07/10/2024] [Indexed: 07/20/2024]
Abstract
Pyruvate kinase M2 (PKM2), a key enzyme involved in glycolysis,plays an important role in regulating cell metabolism and growth under different physiological conditions. PKM2 has been intensively investigated in multiple cancer diseases. Recent years, many studies have found its pivotal role in cerebrovascular diseases (CeVDs), the disturbances in intracranial blood circulation. CeVDs has been confirmed to be closely associated with oxidative stress (OS), mitochondrial dynamics, systemic inflammation, and local neuroinflammation in the brain. It has further been revealed that PKM2 exerts various biological functions in the regulation of energy supply, OS, inflammatory responses, and mitochondrial dysfunction. The roles of PKM2 are closely related to its different isoforms, expression levels in subcellular localization, and post-translational modifications. Therefore, summarizing the roles of PKM2 in CeVDs will help further understanding the molecular mechanisms of CeVDs. In this review, we illustrate the characteristics of PKM2, the regulated PKM2 expression, and the biological roles of PKM2 in CeVDs.
Collapse
Affiliation(s)
- Ling Ma
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong 250033, China
| | - Huatao Li
- Department of Stroke Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China
| | - Hu Xu
- Department of Stroke Center, Shandong Second Medical University, Weifang, Shandong 261000, China
| | - Dianwei Liu
- Department of Stroke Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China; Department of Neurosurgery, XuanWu Hospital Capital Medical University Jinan Branch, Jinan, Shandong 250100, China.
| |
Collapse
|
9
|
Fu Z, Deng M, Zhou Q, Li S, Liu W, Cao S, Zhang L, Deng Y, Xi S. Arsenic activated GLUT1-mTORC1/HIF-1α-PKM2 positive feedback networks promote proliferation and migration of bladder epithelial cells. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 947:174538. [PMID: 38977090 DOI: 10.1016/j.scitotenv.2024.174538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 07/03/2024] [Accepted: 07/04/2024] [Indexed: 07/10/2024]
Abstract
Arsenic (As) is recognized as a potent environmental contaminant associated with bladder carcinogenesis. However, its molecular mechanism remains unclear. Metabolic reprogramming is one of the hallmarks of cancer and is as a central feature of malignancy. Here, we performed the study of cross-talk between the mammalian target of rapamycin complex 1 (mTORC1)/ Hypoxia-inducible factor 1 alpha (HIF-1α) pathway and aerobic glycolysis in promoting the proliferation and migration of bladder epithelial cells treated by arsenic in vivo and in vitro. We demonstrated that arsenite promoted N-methyl-N-nitrosourea (MNU)-induced tumor formation in the bladder of rats and the malignant behavior of human ureteral epithelial (SV-HUC-1) cell. We found that arsenite positively regulated the mTORC1/HIF-1α pathway through glucose transporter protein 1 (GLUT1), which involved in the malignant progression of bladder epithelial cells relying on glycolysis. In addition, pyruvate kinase M2 (PKM2) increased by arsenite reduced the protein expressions of succinate dehydrogenase (SDH) and fumarate hydratase (FH), leading to the accumulation of tumor metabolites of succinate and fumarate. Moreover, heat shock protein (HSP)90, functioning as a chaperone protein, stabilized PKM2 and thereby regulated the proliferation and aerobic glycolysis in arsenite treated SV-HUC-1 cells. Taken together, these results provide new insights into mTORC1/HIF-1α and PKM2 networks as critical molecular targets that contribute to the arsenic-induced malignant progression of bladder epithelial cells.
Collapse
Affiliation(s)
- Zhushan Fu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning 110122, China; The Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, Shenyang, Liaoning 110122, China; Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China
| | - Meiqi Deng
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning 110122, China; Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qing Zhou
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning 110122, China; The Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, Shenyang, Liaoning 110122, China; Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China
| | - Sihao Li
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning 110122, China; The Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, Shenyang, Liaoning 110122, China; Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China
| | - Weijue Liu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning 110122, China; The Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, Shenyang, Liaoning 110122, China; Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China
| | - Siyan Cao
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning 110122, China; The Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, Shenyang, Liaoning 110122, China; Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China
| | - Lei Zhang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning 110122, China; The Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, Shenyang, Liaoning 110122, China; Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China
| | - Yu Deng
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning 110122, China; The Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, Shenyang, Liaoning 110122, China; Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China.
| | - Shuhua Xi
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning 110122, China; The Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, Shenyang, Liaoning 110122, China; Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China.
| |
Collapse
|
10
|
Wang Y, Shu H, Qu Y, Jin X, Liu J, Peng W, Wang L, Hao M, Xia M, Zhao Z, Dong K, Di Y, Tian M, Hao F, Xia C, Zhang W, Ba X, Feng Y, Wei M. PKM2 functions as a histidine kinase to phosphorylate PGAM1 and increase glycolysis shunts in cancer. EMBO J 2024; 43:2368-2396. [PMID: 38750259 PMCID: PMC11183095 DOI: 10.1038/s44318-024-00110-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 03/24/2024] [Accepted: 03/27/2024] [Indexed: 06/19/2024] Open
Abstract
Phosphoglycerate mutase 1 (PGAM1) is a key node enzyme that diverts the metabolic reactions from glycolysis into its shunts to support macromolecule biosynthesis for rapid and sustainable cell proliferation. It is prevalent that PGAM1 activity is upregulated in various tumors; however, the underlying mechanism remains unclear. Here, we unveil that pyruvate kinase M2 (PKM2) moonlights as a histidine kinase in a phosphoenolpyruvate (PEP)-dependent manner to catalyze PGAM1 H11 phosphorylation, that is essential for PGAM1 activity. Moreover, monomeric and dimeric but not tetrameric PKM2 are efficient to phosphorylate and activate PGAM1. In response to epidermal growth factor signaling, Src-catalyzed PGAM1 Y119 phosphorylation is a prerequisite for PKM2 binding and the subsequent PGAM1 H11 phosphorylation, which constitutes a discrepancy between tumor and normal cells. A PGAM1-derived pY119-containing cell-permeable peptide or Y119 mutation disrupts the interaction of PGAM1 with PKM2 and PGAM1 H11 phosphorylation, dampening the glycolysis shunts and tumor growth. Together, these results identify a function of PKM2 as a histidine kinase, and illustrate the importance of enzyme crosstalk as a regulatory mode during metabolic reprogramming and tumorigenesis.
Collapse
Affiliation(s)
- Yang Wang
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, 130024, Changchun, Jilin, China
| | - Hengyao Shu
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, 130024, Changchun, Jilin, China
| | - Yanzhao Qu
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, 130024, Changchun, Jilin, China
| | - Xin Jin
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, 130024, Changchun, Jilin, China
| | - Jia Liu
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, 130024, Changchun, Jilin, China
| | - Wanting Peng
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, 130024, Changchun, Jilin, China
| | - Lihua Wang
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, 130024, Changchun, Jilin, China
| | - Miao Hao
- Science Research Center, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, 130033, Changchun, Jilin, China
| | - Mingjie Xia
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, 130024, Changchun, Jilin, China
| | - Zhexuan Zhao
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, 130024, Changchun, Jilin, China
| | - Kejian Dong
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, 130024, Changchun, Jilin, China
| | - Yao Di
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, 130024, Changchun, Jilin, China
| | - Miaomiao Tian
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, 130024, Changchun, Jilin, China
| | - Fengqi Hao
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, 130024, Changchun, Jilin, China
| | - Chaoyi Xia
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, 130024, Changchun, Jilin, China
| | - Wenxia Zhang
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, 130024, Changchun, Jilin, China
| | - Xueqing Ba
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, 130024, Changchun, Jilin, China.
| | - Yunpeng Feng
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, 130024, Changchun, Jilin, China.
| | - Min Wei
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, 130024, Changchun, Jilin, China.
| |
Collapse
|
11
|
Su G, Liu J, Duan C, Fang P, Fang L, Zhou Y, Xiao S. Enteric coronavirus PDCoV evokes a non-Warburg effect by hijacking pyruvic acid as a metabolic hub. Redox Biol 2024; 71:103112. [PMID: 38461791 PMCID: PMC10938170 DOI: 10.1016/j.redox.2024.103112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 02/21/2024] [Accepted: 03/03/2024] [Indexed: 03/12/2024] Open
Abstract
The Warburg effect, also referred as aerobic glycolysis, is a common metabolic program during viral infection. Through targeted metabolomics combined with biochemical experiments and various cell models, we investigated the central carbon metabolism (CCM) profiles of cells infected with porcine deltacoronavirus (PDCoV), an emerging enteropathogenic coronavirus with zoonotic potential. We found that PDCoV infection required glycolysis but decreased glycolytic flux, exhibiting a non-Warburg effect characterized by pyruvic acid accumulation. Mechanistically, PDCoV enhanced pyruvate kinase activity to promote pyruvic acid anabolism, a process that generates pyruvic acid with concomitant ATP production. PDCoV also hijacked pyruvic acid catabolism to increase biosynthesis of non-essential amino acids (NEAAs), suggesting that pyruvic acid is an essential hub for PDCoV to scavenge host energy and metabolites. Furthermore, PDCoV facilitated glutaminolysis to promote the synthesis of NEAA and pyrimidines for optimal proliferation. Our work supports a novel CCM model after viral infection and provides potential anti-PDCoV drug targets.
Collapse
Affiliation(s)
- Guanning Su
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Jiao Liu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Chenrui Duan
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Puxian Fang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Liurong Fang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Yanrong Zhou
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China.
| | - Shaobo Xiao
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China.
| |
Collapse
|
12
|
Zhou X, Li Y, Pan M, Lu T, Liu C, Wang Z, Tang F, Hu G. PKM2 promotes lymphatic metastasis of hypopharyngeal carcinoma via regulating epithelial-mesenchymal transition: an experimental research. Diagn Pathol 2024; 19:48. [PMID: 38431604 PMCID: PMC10907999 DOI: 10.1186/s13000-024-01474-5] [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: 10/17/2023] [Accepted: 02/25/2024] [Indexed: 03/05/2024] Open
Abstract
BACKGROUND Patients with hypopharyngeal carcinoma (HPC) have a poor prognosis mainly because of lymphatic metastasis. This research aimed to determine the PKM2 role in lymphatic metastasis in HPC and the underlying molecular mechanism contributing to this phenomenon. METHODS PKM2 in HPC was studied for its expression and its likelihood of overall survival using TCGA dataset. Western blotting, qRT-PCR, and IHC were employed to confirm PKM2 expression. Methods including gain- and loss-of-function were used to examine the PKM2 role in HPC metastasis in vitro and in vivo. In vitro and in vivo studies also confirmed lymphatic metastasis's mechanism. RESULTS Prominent PKM2 overexpression was seen in patients with lymphatic metastasis of HPC, and there was an inherent relationship between a high PKM2 level and poor prognosis. In vitro research showed that knocking down PKM2 decreased tumor cell invasion, migration, and proliferation while promoting apoptosis and inhibiting epithelial-mesenchymal transition, but overexpressing PKM2 had the reverse effect. Animal studies suggested that PKM2 may facilitate tumor development and lymphatic metastasis. CONCLUSIONS Our findings suggest that PKM2 may be a tumor's promoter gene of lymphatic metastasis, which may promote lymphatic metastasis of HPC by regulating epithelial-mesenchymal transition. PKM2 may be a biomarker of metastatic potential, ultimately providing a basis for exploring new therapeutic targets.
Collapse
Affiliation(s)
- Xin Zhou
- Department of Otolaryngology, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
- Department of Otolaryngology, Chongqing Traditional Chinese Medicine Hospital, Chongqing, China
| | - Yanshi Li
- Department of Otolaryngology, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Min Pan
- Department of Otolaryngology, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Tao Lu
- Department of Otolaryngology, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Chuan Liu
- Department of Otolaryngology, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Zhihai Wang
- Department of Otolaryngology, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Fengxiang Tang
- Department of Otolaryngology, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Guohua Hu
- Department of Otolaryngology, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Road, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
13
|
Zhang A, Zhang H, Wang R, He H, Song B, Song R. Bactericidal bissulfone B 7 targets bacterial pyruvate kinase to impair bacterial biology and pathogenicity in plants. SCIENCE CHINA. LIFE SCIENCES 2024; 67:391-402. [PMID: 37987940 DOI: 10.1007/s11427-023-2449-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 09/11/2023] [Indexed: 11/22/2023]
Abstract
The prevention and control of rice bacterial leaf blight (BLB) disease has not yet been achieved due to the lack of effective agrochemicals and available targets. Herein, we develop a series of novel bissulfones and a novel target with a unique mechanism to address this challenge. The developed bissulfones can control Xanthomonas oryzae pv. oryzae (Xoo), and 2-(bis(methylsulfonyl)methylene)-N-(4-chlorophenyl) hydrazine-1-carboxamide (B7) is more effective than the commercial drugs thiodiazole copper (TC) and bismerthiazol (BT). Pyruvate kinase (PYK) in Xoo has been identified for the first time as the target protein of our bissulfone B7. PYK modulates bacterial virulence via a CRP-like protein (Clp)/two-component system regulatory protein (regR) axis. The elucidation of this pathway facilitates the use of B7 to reduce PYK expression at the transcriptional level, block PYK activity at the protein level, and impair the interaction within the PYK-Clp-regR complex via competitive inhibition, thereby attenuating bacterial biology and pathogenicity. This study offers insights into the molecular and mechanistic aspects underlying anti-Xoo strategies that target PYK. We believe that these valuable discoveries will be used for bacterial disease control in the future.
Collapse
Affiliation(s)
- Awei Zhang
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang, 550025, China
| | - Haizhen Zhang
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang, 550025, China
| | - Ronghua Wang
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang, 550025, China
| | - Hongfu He
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang, 550025, China
| | - Baoan Song
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang, 550025, China.
| | - Runjiang Song
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang, 550025, China.
| |
Collapse
|
14
|
Hermawan A, Putri H, Fatimah N, Prasetio HH. Transcriptomics analysis reveals distinct mechanism of breast cancer stem cells regulation in mammospheres from MCF-7 and T47D cells. Heliyon 2024; 10:e24356. [PMID: 38304813 PMCID: PMC10831612 DOI: 10.1016/j.heliyon.2024.e24356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 12/04/2023] [Accepted: 01/08/2024] [Indexed: 02/03/2024] Open
Abstract
Luminal A breast cancer, constituting 70 % of breast cancer cases, presents a challenge due to the development of resistance and recurrence caused by breast cancer stem cells (BCSC). Luminal breast tumors are characterized by TP53 expression, a tumor suppressor gene involved in maintaining stem cell attributes in cancer. Although a previous study successfully developed mammospheres (MS) from MCF-7 (with wild-type TP53) and T47D (with mutant TP53) luminal breast cancer cells for BCSC enrichment, their transcriptomic profiles remain unclear. We aimed to elucidate the transcriptomic disparities between MS of MCF-7 and T47D cells using bioinformatics analyses of differentially expressed genes (DEGs), including the KEGG pathway, Gene Ontology (GO), drug-gene association, disease-gene association, Gene Set Enrichment Analysis (GSEA), DNA methylation analysis, correlation analysis of DEGs with immune cell infiltration, and association analysis of genes and small-molecule compounds via the Connectivity Map (CMap). Upregulated DEGs were enriched in metabolism-related KEGG pathways, whereas downregulated DEGs were enriched in the MAPK signaling pathway. Drug-gene association analysis revealed that both upregulated and downregulated DEGs were associated with fostamatinib. The KEGG pathway GSEA results indicated that the DEGs were enriched for oxidative phosphorylation, whereas the downregulated DEGs were negatively enriched for the p53 signaling pathway. Examination of DNA methylation revealed a noticeable disparity in the expression patterns of the PKM2, ERO1L, SLC6A6, EPAS1, APLP2, RPL10L, and NEDD4 genes when comparing cohorts with low- and high-risk breast cancer. Furthermore, a significant positive correlation was identified between SLC6A6 expression and macrophage presence, as well as MSN, and AKR1B1 expression and neutrophil and dentritic cell infiltration. CMap analysis unveiled SA-83851 as a potential candidate to counteract the effects of DEGs, specifically in cells harbouring mutant TP53. Further research, including in vitro and in vivo validations, is warranted to develop drugs targeting BCSCs.
Collapse
Affiliation(s)
- Adam Hermawan
- Laboratory of Macromolecular Engineering, Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Universitas Gadjah Mada Sekip Utara II, 55281, Yogyakarta, Indonesia
- Cancer Chemoprevention Research Center, Faculty of Pharmacy, Universitas Gadjah Mada Sekip Utara II, 55281, Yogyakarta, Indonesia
- Laboratory of Advanced Pharmaceutical Sciences. APSLC Building, Faculty of Pharmacy, Universitas Gadjah Mada Sekip Utara II, 55281, Yogyakarta, Indonesia
| | - Herwandhani Putri
- Cancer Chemoprevention Research Center, Faculty of Pharmacy, Universitas Gadjah Mada Sekip Utara II, 55281, Yogyakarta, Indonesia
| | - Nurul Fatimah
- Laboratory of Advanced Pharmaceutical Sciences. APSLC Building, Faculty of Pharmacy, Universitas Gadjah Mada Sekip Utara II, 55281, Yogyakarta, Indonesia
| | - Heri Himawan Prasetio
- Laboratory of Macromolecular Engineering, Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Universitas Gadjah Mada Sekip Utara II, 55281, Yogyakarta, Indonesia
| |
Collapse
|
15
|
Li X, Hu S, Cai Y, Liu X, Luo J, Wu T. Revving the engine: PKB/AKT as a key regulator of cellular glucose metabolism. Front Physiol 2024; 14:1320964. [PMID: 38264327 PMCID: PMC10804622 DOI: 10.3389/fphys.2023.1320964] [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: 10/13/2023] [Accepted: 12/12/2023] [Indexed: 01/25/2024] Open
Abstract
Glucose metabolism is of critical importance for cell growth and proliferation, the disorders of which have been widely implicated in cancer progression. Glucose uptake is achieved differently by normal cells and cancer cells. Even in an aerobic environment, cancer cells tend to undergo metabolism through glycolysis rather than the oxidative phosphorylation pathway. Disordered metabolic syndrome is characterized by elevated levels of metabolites that can cause changes in the tumor microenvironment, thereby promoting tumor recurrence and metastasis. The activation of glycolysis-related proteins and transcription factors is involved in the regulation of cellular glucose metabolism. Changes in glucose metabolism activity are closely related to activation of protein kinase B (PKB/AKT). This review discusses recent findings on the regulation of glucose metabolism by AKT in tumors. Furthermore, the review summarizes the potential importance of AKT in the regulation of each process throughout glucose metabolism to provide a theoretical basis for AKT as a target for cancers.
Collapse
Affiliation(s)
- Xia Li
- General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Shuying Hu
- General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yaoting Cai
- General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xuelian Liu
- General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jing Luo
- General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Tao Wu
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
16
|
Sharma N, Chaudhary A, Sachdeva M. An insight into the structure-activity relationship studies of anticancer medicinal attributes of 7-azaindole derivatives: a review. Future Med Chem 2023; 15:2309-2323. [PMID: 38112047 DOI: 10.4155/fmc-2023-0216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 11/29/2023] [Indexed: 12/20/2023] Open
Abstract
In the current portfolio, there is a lot of interest in the 7-azaindole building block for drug discovery. The creation of synthetic, sophisticated methods for the modification of 7-azaindoles is a promising area of research. This review covers the structure-activity relationship of 7-azaindole analogs, which have been shown to be effective anticancer agents in the literature of the past two decades. Positions 1, 3 and 5 of the 7-azaindole ring are the most active sites. Disubstitution is used for the synthesis of a new analog of the 7-azaindole moiety. All positions are used to create novel molecules that are effective anticancer agents. The alkyl, aryl carboxamide group and heterocyclic ring are the most successful types of substitution.
Collapse
Affiliation(s)
- Neha Sharma
- Rajkumar Goel Institute of Technology (Pharmacy), NH-58, Ghaziabad, 201001, India
| | - Anurag Chaudhary
- Department of Pharmaceutical Technology, Meerut Institute of Engineering & Technology, Meerut, 250005, India
| | - Monika Sachdeva
- Rajkumar Goel Institute of Technology (Pharmacy), NH-58, Ghaziabad, 201001, India
| |
Collapse
|
17
|
Bailleul J, Ruan Y, Abdulrahman L, Scott AJ, Yazal T, Sung D, Park K, Hoang H, Nathaniel J, Chu FI, Palomera D, Sehgal A, Tsang JE, Nathanson DA, Xu S, Park JO, ten Hoeve J, Bhat K, Qi N, Kornblum HI, Schaue D, McBride WH, Lyssiotis CA, Wahl DR, Vlashi E. M2 isoform of pyruvate kinase rewires glucose metabolism during radiation therapy to promote an antioxidant response and glioblastoma radioresistance. Neuro Oncol 2023; 25:1989-2000. [PMID: 37279645 PMCID: PMC10628945 DOI: 10.1093/neuonc/noad103] [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: 01/14/2023] [Indexed: 06/08/2023] Open
Abstract
BACKGROUND Resistance to existing therapies is a significant challenge in improving outcomes for glioblastoma (GBM) patients. Metabolic plasticity has emerged as an important contributor to therapy resistance, including radiation therapy (RT). Here, we investigated how GBM cells reprogram their glucose metabolism in response to RT to promote radiation resistance. METHODS Effects of radiation on glucose metabolism of human GBM specimens were examined in vitro and in vivo with the use of metabolic and enzymatic assays, targeted metabolomics, and FDG-PET. Radiosensitization potential of interfering with M2 isoform of pyruvate kinase (PKM2) activity was tested via gliomasphere formation assays and in vivo human GBM models. RESULTS Here, we show that RT induces increased glucose utilization by GBM cells, and this is accompanied with translocation of GLUT3 transporters to the cell membrane. Irradiated GBM cells route glucose carbons through the pentose phosphate pathway (PPP) to harness the antioxidant power of the PPP and support survival after radiation. This response is regulated in part by the PKM2. Activators of PKM2 can antagonize the radiation-induced rewiring of glucose metabolism and radiosensitize GBM cells in vitro and in vivo. CONCLUSIONS These findings open the possibility that interventions designed to target cancer-specific regulators of metabolic plasticity, such as PKM2, rather than specific metabolic pathways, have the potential to improve the radiotherapeutic outcomes in GBM patients.
Collapse
Affiliation(s)
- Justine Bailleul
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Yangjingyi Ruan
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Lobna Abdulrahman
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Andrew J Scott
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Taha Yazal
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - David Sung
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Keunseok Park
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, California, USA
| | - Hanna Hoang
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Juan Nathaniel
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Fang-I Chu
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Daisy Palomera
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Anahita Sehgal
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Jonathan E Tsang
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - David A Nathanson
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Shili Xu
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
- Crump Institute for Molecular Imaging, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Junyoung O Park
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, California, USA
| | - Johanna ten Hoeve
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Kruttika Bhat
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Nathan Qi
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Harley I Kornblum
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California, USA
- Neuropsychiatric Institute–Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, California, USA
| | - Dorthe Schaue
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - William H McBride
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Costas A Lyssiotis
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Daniel R Wahl
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Erina Vlashi
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California, USA
| |
Collapse
|
18
|
Xu X, Zhao J, Yang M, Han L, Yuan X, Chi W, Jiang J. The emerging roles of N6-methyladenosine RNA modifications in thyroid cancer. Eur J Med Res 2023; 28:475. [PMID: 37915103 PMCID: PMC10621220 DOI: 10.1186/s40001-023-01382-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 09/19/2023] [Indexed: 11/03/2023] Open
Abstract
Thyroid cancer (TC) is the most predominant malignancy of the endocrine system, with steadily growing occurrence and morbidity worldwide. Although diagnostic and therapeutic methods have been rapidly developed in recent years, the underlying molecular mechanisms in the pathogenesis of TC remain enigmatic. The N6-methyladenosine(m6A) RNA modification is designed to impact RNA metabolism and further gene regulation. This process is intricately regulated by a variety of regulators, such as methylases and demethylases. Aberrant m6A regulators expression is related to the occurrence and development of TC and play an important role in drug resistance. This review comprehensively analyzes the effect of m6A methylation on TC progression and the potential clinical value of m6A regulators as prognostic markers and therapeutic targets in this disease.
Collapse
Affiliation(s)
- Xiaoxin Xu
- Heilongjiang University of Chinese Medicine, Harbin, 150040, Heilongjiang, People's Republic of China
| | - Jiayao Zhao
- Heilongjiang University of Chinese Medicine, Harbin, 150040, Heilongjiang, People's Republic of China
| | - Mingyue Yang
- The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Lutuo Han
- Heilongjiang University of Chinese Medicine, Harbin, 150040, Heilongjiang, People's Republic of China
| | - Xingxing Yuan
- Heilongjiang University of Chinese Medicine, Harbin, 150040, Heilongjiang, People's Republic of China
- Heilongjiang Academy of Traditional Chinese Medicine Science, No. 33 of West Dazhi Street, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Wencheng Chi
- Heilongjiang University of Chinese Medicine, Harbin, 150040, Heilongjiang, People's Republic of China.
- Heilongjiang Academy of Traditional Chinese Medicine Science, No. 33 of West Dazhi Street, Harbin, 150001, Heilongjiang, People's Republic of China.
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China.
| | - Jiakang Jiang
- Heilongjiang University of Chinese Medicine, Harbin, 150040, Heilongjiang, People's Republic of China.
- Heilongjiang Academy of Traditional Chinese Medicine Science, No. 33 of West Dazhi Street, Harbin, 150001, Heilongjiang, People's Republic of China.
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China.
| |
Collapse
|
19
|
Baba SK, Baba SK, Mir R, Elfaki I, Algehainy N, Ullah MF, Barnawi J, Altemani FH, Alanazi M, Mustafa SK, Masoodi T, Akil ASA, Bhat AA, Macha MA. Long non-coding RNAs modulate tumor microenvironment to promote metastasis: novel avenue for therapeutic intervention. Front Cell Dev Biol 2023; 11:1164301. [PMID: 37384249 PMCID: PMC10299194 DOI: 10.3389/fcell.2023.1164301] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 05/22/2023] [Indexed: 06/30/2023] Open
Abstract
Cancer is a devastating disease and the primary cause of morbidity and mortality worldwide, with cancer metastasis responsible for 90% of cancer-related deaths. Cancer metastasis is a multistep process characterized by spreading of cancer cells from the primary tumor and acquiring molecular and phenotypic changes that enable them to expand and colonize in distant organs. Despite recent advancements, the underlying molecular mechanism(s) of cancer metastasis is limited and requires further exploration. In addition to genetic alterations, epigenetic changes have been demonstrated to play an important role in the development of cancer metastasis. Long non-coding RNAs (lncRNAs) are considered one of the most critical epigenetic regulators. By regulating signaling pathways and acting as decoys, guides, and scaffolds, they modulate key molecules in every step of cancer metastasis such as dissemination of carcinoma cells, intravascular transit, and metastatic colonization. Gaining a good knowledge of the detailed molecular basis underlying lncRNAs regulating cancer metastasis may provide previously unknown therapeutic and diagnostic lncRNAs for patients with metastatic disease. In this review, we concentrate on the molecular mechanisms underlying lncRNAs in the regulation of cancer metastasis, the cross-talk with metabolic reprogramming, modulating cancer cell anoikis resistance, influencing metastatic microenvironment, and the interaction with pre-metastatic niche formation. In addition, we also discuss the clinical utility and therapeutic potential of lncRNAs for cancer treatment. Finally, we also represent areas for future research in this rapidly developing field.
Collapse
Affiliation(s)
- Sana Khurshid Baba
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Awantipora, Kashmir, India
| | - Sadaf Khursheed Baba
- Department of Microbiology, Sher-I-Kashmir Institute of Medical Science (SKIMS), Soura, Kashmir, India
| | - Rashid Mir
- Department of Medical Lab Technology, Prince Fahd Bin Sultan Research Chair Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Imadeldin Elfaki
- Department of Biochemistry, Faculty of Science, University of Tabuk, Tabuk, Saudi Arabia
| | - Naseh Algehainy
- Department of Medical Lab Technology, Prince Fahd Bin Sultan Research Chair Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Mohammad Fahad Ullah
- Department of Medical Lab Technology, Prince Fahd Bin Sultan Research Chair Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Jameel Barnawi
- Department of Medical Lab Technology, Prince Fahd Bin Sultan Research Chair Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Faisal H. Altemani
- Department of Medical Lab Technology, Prince Fahd Bin Sultan Research Chair Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Mohammad Alanazi
- Department of Biochemistry, Faculty of Science, University of Tabuk, Tabuk, Saudi Arabia
| | - Syed Khalid Mustafa
- Department of Chemistry, Faculty of Science, University of Tabuk, Tabuk, Saudi Arabia
| | - Tariq Masoodi
- Human Immunology Department, Research Branch, Sidra Medicine, Doha, Qatar
| | - Ammira S. Alshabeeb Akil
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity, and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Ajaz A. Bhat
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity, and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Muzafar A. Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Awantipora, Kashmir, India
| |
Collapse
|
20
|
You M, Xie Z, Zhang N, Zhang Y, Xiao D, Liu S, Zhuang W, Li L, Tao Y. Signaling pathways in cancer metabolism: mechanisms and therapeutic targets. Signal Transduct Target Ther 2023; 8:196. [PMID: 37164974 PMCID: PMC10172373 DOI: 10.1038/s41392-023-01442-3] [Citation(s) in RCA: 95] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 03/20/2023] [Accepted: 04/17/2023] [Indexed: 05/12/2023] Open
Abstract
A wide spectrum of metabolites (mainly, the three major nutrients and their derivatives) can be sensed by specific sensors, then trigger a series of signal transduction pathways and affect the expression levels of genes in epigenetics, which is called metabolite sensing. Life body regulates metabolism, immunity, and inflammation by metabolite sensing, coordinating the pathophysiology of the host to achieve balance with the external environment. Metabolic reprogramming in cancers cause different phenotypic characteristics of cancer cell from normal cell, including cell proliferation, migration, invasion, angiogenesis, etc. Metabolic disorders in cancer cells further create a microenvironment including many kinds of oncometabolites that are conducive to the growth of cancer, thus forming a vicious circle. At the same time, exogenous metabolites can also affect the biological behavior of tumors. Here, we discuss the metabolite sensing mechanisms of the three major nutrients and their derivatives, as well as their abnormalities in the development of various cancers, and discuss the potential therapeutic targets based on metabolite-sensing signaling pathways to prevent the progression of cancer.
Collapse
Affiliation(s)
- Mengshu You
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 410078, Changsha, Hunan, China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 410078, Changsha, Hunan, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 410078, Changsha, Hunan, China
| | - Zhuolin Xie
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 410078, Changsha, Hunan, China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 410078, Changsha, Hunan, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 410078, Changsha, Hunan, China
| | - Nan Zhang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 410078, Changsha, Hunan, China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 410078, Changsha, Hunan, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 410078, Changsha, Hunan, China
| | - Yixuan Zhang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 410078, Changsha, Hunan, China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 410078, Changsha, Hunan, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 410078, Changsha, Hunan, China
| | - Desheng Xiao
- Department of Pathology, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Shuang Liu
- Department of Oncology, Institute of Medical Sciences, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Wei Zhuang
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, People's Republic of China.
| | - Lili Li
- Cancer Epigenetics Laboratory, Department of Clinical Oncology, State Key Laboratory of Translational Oncology, Sir YK Pao Centre for Cancer and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Ma Liu Shui, Hong Kong.
| | - Yongguang Tao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 410078, Changsha, Hunan, China.
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 410078, Changsha, Hunan, China.
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 410078, Changsha, Hunan, China.
- Department of Thoracic Surgery, Hunan Key Laboratory of Early Diagnosis and Precision Therapy in Lung Cancer, Second Xiangya Hospital, Central South University, 410011, Changsha, China.
| |
Collapse
|
21
|
Ji X, Lv C, Huang J, Dong W, Sun W, Zhang H. ALKBH5-induced circular RNA NRIP1 promotes glycolysis in thyroid cancer cells by targeting PKM2. Cancer Sci 2023. [PMID: 36851875 DOI: 10.1111/cas.15772] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 02/14/2023] [Accepted: 02/21/2023] [Indexed: 03/01/2023] Open
Abstract
Although circular RNAs (circRNAs) are involved in cell proliferation, differentiation, apoptosis, and invasion, the underlying regulatory mechanisms of circRNAs in thyroid cancer have not been fully elucidated. This article aimed to study the role of circRNA regulated by N6-methyladenosine modification in papillary thyroid cancer (PTC). Quantitative real-time PCR, western blotting, and immunohistochemistry were used to investigate the expressions of circRNA nuclear receptor-interacting protein 1 (circNRIP1) in PTC tissues and adjacent noncancerous thyroid tissues. In vitro and in vivo assays were carried out to assess the effects of circNRIP1 on PTC glycolysis and growth. The N6-methyladenosine mechanisms of circNRIP1 were evaluated by methylated RNA immunoprecipitation sequencing, luciferase reporter gene, and RNA stability assays. Results showed that circNRIP1 levels were significantly upregulated in PTC tissues. Furthermore, elevated circNRIP1 levels in PTC patients were correlated with high tumor lymph node metastasis stage and larger tumor sizes. Functionally, circNRIP1 significantly promoted glycolysis, PTC cell proliferation in vitro, and tumorigenesis in vivo. Mechanistically, circNRIP1 acted as a sponge for microRNA (miR)-541-5p and miR-3064-5p and jointly upregulated pyruvate kinase M2 (PKM2) expression. Knockdown of m6 A demethylase α-ketoglutarate-dependent dioxygenase alkB homolog 5 (ALKBH5) significantly enhanced circNRIP1 m6 A modification and upregulated its expression. These results show that ALKBH5 knockdown upregulates circNRIP1, thus promoting glycolysis in PTC cells. Therefore, circNRIP1 can be a prognostic biomarker and therapeutic target for PTC by acting as a sponge for oncogenic miR-541-5p and miR-3064-5p to upregulate PKM2 expression.
Collapse
Affiliation(s)
- Xiaoyu Ji
- Department of Thyroid Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Chengzhou Lv
- Department of Thyroid Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Jiapeng Huang
- Department of Thyroid Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Wenwu Dong
- Department of Thyroid Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Wei Sun
- Department of Thyroid Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Hao Zhang
- Department of Thyroid Surgery, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
22
|
Wang X, Guo W, Han J, Li J, Zhao Q, Mao Y, Wang S. Oral spatial-to-point cascade targeting "sugar-coated bullets" for precise and safe chemotherapy by intervention Warburg effect. Colloids Surf B Biointerfaces 2023; 222:113108. [PMID: 36586235 DOI: 10.1016/j.colsurfb.2022.113108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 12/10/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022]
Abstract
Glycolysis plays a vital role in the development and progression of tumors. Inhibiting glycolysis via smart and safe methods serves as a promising target for cancer therapy. Here, an oral "sugar-coated bullet" aiming at intervening Warburg effect is designed by coating colloidal mesoporous silica nanoparticles (CMS) encapsulating glycolysis inhibitor shikonin (SHK) with dextran, namely DCMS/SHK. The solubility and drug-loading capacity of SHK were enhanced by the special structure of CMS. Besides, the tempting bullets possess the spatial-to-point cascade targeting ability in delivering SHK from the colonic lumen to colon cancer cells and finally to PKM2. After DCMS/SHK reaches the colon, the dextran is hydrolyzed by dextranase especially existing in the colon site to glucose and the carriers become glucose-coated nanoparticles. The glucose-cloak nanoparticles would be largely endocytosed by tumor cells and complete the efficient delivery of SHK. The encapsulated SHK can prevent the glycolysis of cancer cells and thus inhibit tumor growth effectively. This work presents an ingenious cascade colon-targeting strategy to treat colon cancer by destroying cell energy metabolism.
Collapse
Affiliation(s)
- Xiudan Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Wen Guo
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Jianan Han
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Jia Li
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Qinfu Zhao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Yuling Mao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China.
| | - Siling Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China.
| |
Collapse
|
23
|
Chhimpa N, Singh N, Puri N, Kayath HP. The Novel Role of Mitochondrial Citrate Synthase and Citrate in the Pathophysiology of Alzheimer's Disease. J Alzheimers Dis 2023; 94:S453-S472. [PMID: 37393492 PMCID: PMC10473122 DOI: 10.3233/jad-220514] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2023] [Indexed: 07/03/2023]
Abstract
Citrate synthase is a key mitochondrial enzyme that utilizes acetyl-CoA and oxaloacetate to form citrate in the mitochondrial membrane, which participates in energy production in the TCA cycle and linked to the electron transport chain. Citrate transports through a citrate malate pump and synthesizes acetyl-CoA and acetylcholine (ACh) in neuronal cytoplasm. In a mature brain, acetyl-CoA is mainly utilized for ACh synthesis and is responsible for memory and cognition. Studies have shown low citrate synthase in different regions of brain in Alzheimer's disease (AD) patients, which reduces mitochondrial citrate, cellular bioenergetics, neurocytoplasmic citrate, acetyl-CoA, and ACh synthesis. Reduced citrate mediated low energy favors amyloid-β (Aβ) aggregation. Citrate inhibits Aβ25-35 and Aβ1-40 aggregation in vitro. Hence, citrate can be a better therapeutic option for AD by improving cellular energy and ACh synthesis, and inhibiting Aβ aggregation, which prevents tau hyperphosphorylation and glycogen synthase kinase-3 beta. Therefore, we need clinical studies if citrate reverses Aβ deposition by balancing mitochondrial energy pathway and neurocytoplasmic ACh production. Furthermore, in AD's silent phase pathophysiology, when neuronal cells are highly active, they shift ATP utilization from oxidative phosphorylation to glycolysis and prevent excessive generation of hydrogen peroxide and reactive oxygen species (oxidative stress) as neuroprotective action, which upregulates glucose transporter-3 (GLUT3) and pyruvate dehydrogenase kinase-3 (PDK3). PDK3 inhibits pyruvate dehydrogenase, which decreases mitochondrial-acetyl-CoA, citrate, and cellular bioenergetics, and decreases neurocytoplasmic citrate, acetyl-CoA, and ACh formation, thus initiating AD pathophysiology. Therefore, GLUT3 and PDK3 can be biomarkers for silent phase of AD.
Collapse
Affiliation(s)
- Neeraj Chhimpa
- Department of Pharmacology, Post Graduate Institute of Medical Education & Research, Chandigarh, India
- Department of Pharmacology, Meharishi Markandeshwar College of Medical Science & Research, Ambala, India
| | - Neha Singh
- Department of Pharmacology, Post Graduate Institute of Medical Education & Research, Chandigarh, India
| | - Nikkita Puri
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, India
| | | |
Collapse
|
24
|
Shang X, Wang Y, Hou Z, Wang X, Zhang H, Yang C, Li J, Huang X, Song S, Yao G. A natural PKM2 targeting agent as a potential drug for breast cancer treatment. Clin Transl Med 2022; 13:e1157. [PMID: 36579385 PMCID: PMC9798039 DOI: 10.1002/ctm2.1157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 12/07/2022] [Accepted: 12/12/2022] [Indexed: 12/30/2022] Open
Affiliation(s)
- Xin‐Yue Shang
- Key Laboratory of Computational Chemistry‐Based Natural Antitumor Drug Research & DevelopmentEngineering Research Center of Natural Medicine Active Molecule Research & DevelopmentShenyangChina,Key Laboratory of Natural Bioactive Compounds Discovery & ModificationShenyangLiaoningChina,Department of Natural Medicine Chemistry, School of Traditional Chinese Materia MedicaShenyang Pharmaceutical UniversityShenyangLiaoningChina,Department of PharmacologyShenyang Medical CollegeShenyangLiaoningChina
| | - Yu‐Jue Wang
- Key Laboratory of Computational Chemistry‐Based Natural Antitumor Drug Research & DevelopmentEngineering Research Center of Natural Medicine Active Molecule Research & DevelopmentShenyangChina,Key Laboratory of Natural Bioactive Compounds Discovery & ModificationShenyangLiaoningChina,Department of Natural Medicine Chemistry, School of Traditional Chinese Materia MedicaShenyang Pharmaceutical UniversityShenyangLiaoningChina
| | - Zi‐Lin Hou
- Key Laboratory of Computational Chemistry‐Based Natural Antitumor Drug Research & DevelopmentEngineering Research Center of Natural Medicine Active Molecule Research & DevelopmentShenyangChina,Key Laboratory of Natural Bioactive Compounds Discovery & ModificationShenyangLiaoningChina,Department of Natural Medicine Chemistry, School of Traditional Chinese Materia MedicaShenyang Pharmaceutical UniversityShenyangLiaoningChina
| | - Xin‐Ye Wang
- Key Laboratory of Computational Chemistry‐Based Natural Antitumor Drug Research & DevelopmentEngineering Research Center of Natural Medicine Active Molecule Research & DevelopmentShenyangChina,Key Laboratory of Natural Bioactive Compounds Discovery & ModificationShenyangLiaoningChina,Department of Natural Medicine Chemistry, School of Traditional Chinese Materia MedicaShenyang Pharmaceutical UniversityShenyangLiaoningChina
| | - Hao Zhang
- Key Laboratory of Computational Chemistry‐Based Natural Antitumor Drug Research & DevelopmentEngineering Research Center of Natural Medicine Active Molecule Research & DevelopmentShenyangChina,Key Laboratory of Natural Bioactive Compounds Discovery & ModificationShenyangLiaoningChina,Department of Natural Medicine Chemistry, School of Traditional Chinese Materia MedicaShenyang Pharmaceutical UniversityShenyangLiaoningChina
| | - Chen‐Yu Yang
- Key Laboratory of Computational Chemistry‐Based Natural Antitumor Drug Research & DevelopmentEngineering Research Center of Natural Medicine Active Molecule Research & DevelopmentShenyangChina,Key Laboratory of Natural Bioactive Compounds Discovery & ModificationShenyangLiaoningChina,Department of Natural Medicine Chemistry, School of Traditional Chinese Materia MedicaShenyang Pharmaceutical UniversityShenyangLiaoningChina
| | - Ji‐Chong Li
- Key Laboratory of Computational Chemistry‐Based Natural Antitumor Drug Research & DevelopmentEngineering Research Center of Natural Medicine Active Molecule Research & DevelopmentShenyangChina,Key Laboratory of Natural Bioactive Compounds Discovery & ModificationShenyangLiaoningChina,Department of Natural Medicine Chemistry, School of Traditional Chinese Materia MedicaShenyang Pharmaceutical UniversityShenyangLiaoningChina
| | - Xiao‐Xiao Huang
- Key Laboratory of Computational Chemistry‐Based Natural Antitumor Drug Research & DevelopmentEngineering Research Center of Natural Medicine Active Molecule Research & DevelopmentShenyangChina,Key Laboratory of Natural Bioactive Compounds Discovery & ModificationShenyangLiaoningChina,Department of Natural Medicine Chemistry, School of Traditional Chinese Materia MedicaShenyang Pharmaceutical UniversityShenyangLiaoningChina
| | - Shao‐Jiang Song
- Key Laboratory of Computational Chemistry‐Based Natural Antitumor Drug Research & DevelopmentEngineering Research Center of Natural Medicine Active Molecule Research & DevelopmentShenyangChina,Key Laboratory of Natural Bioactive Compounds Discovery & ModificationShenyangLiaoningChina,Department of Natural Medicine Chemistry, School of Traditional Chinese Materia MedicaShenyang Pharmaceutical UniversityShenyangLiaoningChina
| | - Guo‐Dong Yao
- Key Laboratory of Computational Chemistry‐Based Natural Antitumor Drug Research & DevelopmentEngineering Research Center of Natural Medicine Active Molecule Research & DevelopmentShenyangChina,Key Laboratory of Natural Bioactive Compounds Discovery & ModificationShenyangLiaoningChina,Department of Natural Medicine Chemistry, School of Traditional Chinese Materia MedicaShenyang Pharmaceutical UniversityShenyangLiaoningChina
| |
Collapse
|
25
|
Orofiamma LA, Vural D, Antonescu CN. Control of cell metabolism by the epidermal growth factor receptor. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119359. [PMID: 36089077 DOI: 10.1016/j.bbamcr.2022.119359] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 08/24/2022] [Accepted: 09/01/2022] [Indexed: 06/15/2023]
Abstract
The epidermal growth factor receptor (EGFR) triggers the activation of many intracellular signals that control cell proliferation, growth, survival, migration, and differentiation. Given its wide expression, EGFR has many functions in development and tissue homeostasis. Some of the cellular outcomes of EGFR signaling involve alterations of specific aspects of cellular metabolism, and alterations of cell metabolism are emerging as driving influences in many physiological and pathophysiological contexts. Here we review the mechanisms by which EGFR regulates cell metabolism, including by modulation of gene expression and protein function leading to control of glucose uptake, glycolysis, biosynthetic pathways branching from glucose metabolism, amino acid metabolism, lipogenesis, and mitochondrial function. We further examine how this regulation of cell metabolism by EGFR may contribute to cell proliferation and differentiation and how EGFR-driven control of metabolism can impact certain diseases and therapy outcomes.
Collapse
Affiliation(s)
- Laura A Orofiamma
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario M5B 2K3, Canada; Graduate Program in Molecular Science, Toronto Metropolitan University, Toronto, Ontario M5B 2K3, Canada
| | - Dafne Vural
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario M5B 2K3, Canada; Graduate Program in Molecular Science, Toronto Metropolitan University, Toronto, Ontario M5B 2K3, Canada
| | - Costin N Antonescu
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario M5B 2K3, Canada; Graduate Program in Molecular Science, Toronto Metropolitan University, Toronto, Ontario M5B 2K3, Canada.
| |
Collapse
|
26
|
Cao F, Yang D, Tang F, Lu C, He X, Chen S, Yang Z, Gong S, Sun L, Enomoto A, Takahashi M, Weng L. Girdin Promotes Tumorigenesis and Chemoresistance in Lung Adenocarcinoma by Interacting with PKM2. Cancers (Basel) 2022; 14:cancers14225688. [PMID: 36428781 PMCID: PMC9688487 DOI: 10.3390/cancers14225688] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 10/15/2022] [Accepted: 11/14/2022] [Indexed: 11/22/2022] Open
Abstract
Girdin, an Akt substrate, has been reported to promote tumorigenesis in various tumors. However, the role of Girdin in a spontaneous tumor model has not yet been explored. Here, we studied the role of Girdin in lung adenocarcinoma (LUAD) using the autochthonous mouse model and found that Girdin led to LUAD progression and chemoresistance by enhancing the Warburg effect. Mechanistically, Girdin interacted with pyruvate kinase M2 (PKM2), which played a vital role in aerobic glycolysis. Furthermore, Girdin impaired Platelet Derived Growth Factor Receptor Beta (PDGFRβ) degradation, which in turn, promoted PKM2 tyrosine residue 105 (Y105) phosphorylation and inhibited PKM2 activity, subsequently promoting aerobic glycolysis in cancer cells. Taken together, our study demonstrates that Girdin is a crucial regulator of tumor growth and may be a potential therapeutic target for overcoming the resistance of LUAD cells to chemotherapy.
Collapse
Affiliation(s)
- Fuyang Cao
- Department of Oncology, Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha 410008, China
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Desong Yang
- Hunan Clinical Medical Research Center of Accurate Diagnosis and Treatment for Esophageal Carcinoma, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, China
- Thoracic Surgery Department 2, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Feiyu Tang
- Department of Oncology, Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha 410008, China
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Can Lu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xiang He
- Department of Oncology, Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha 410008, China
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Songming Chen
- Department of Oncology, Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha 410008, China
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Zhanghuan Yang
- Department of Oncology, Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha 410008, China
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Siyuan Gong
- Department of Oncology, Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha 410008, China
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Lunquan Sun
- Department of Oncology, Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha 410008, China
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan International Science and Technology Collaboration Base of Precision Medicine for Cancer, Changsha 410008, China
- Institute of Gerontological Cancer Research, National Clinical Research Center for Gerontology, Changsha 410008, China
- Center for Molecular Imaging of Central South University, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Atsushi Enomoto
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Masahide Takahashi
- International Center for Cell and Gene Therapy, Fujita Health University, Toyoake 470-1192, Japan
- Correspondence: (M.T.); (L.W.)
| | - Liang Weng
- Department of Oncology, Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha 410008, China
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan International Science and Technology Collaboration Base of Precision Medicine for Cancer, Changsha 410008, China
- Institute of Gerontological Cancer Research, National Clinical Research Center for Gerontology, Changsha 410008, China
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha 410008, China
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha 410008, China
- Correspondence: (M.T.); (L.W.)
| |
Collapse
|
27
|
Lv S, Zhang Y, Song J, Chen J, Huang B, Luo Y, Zhao Y. Cerulenin suppresses ErbB2-overexpressing breast cancer by targeting ErbB2/PKM2 pathway. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 40:5. [PMID: 36308575 DOI: 10.1007/s12032-022-01872-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/30/2022] [Indexed: 01/17/2023]
Abstract
Cerulenin is a fungal metabolite and a specific inhibitor of fatty acid synthase (FASN), which has shown a potential anticancer activity. 20-25% of breast cancer patients with ErbB2-overexpressing develop resistance to treatment. Therefore, it is urgent to find an effective new target for the treatment of ErbB2-overexpressing breast cancer. Our previous study found that cerulenin inhibits the glycolysis and migration of SK-BR-3 cells, but the effect of cerulenin on other malignant phenotypes of breast cancer is still unknown. Furthermore, the mechanism by which cerulenin displays its inhibitory effects is not fully understood. In this study, we systematically investigate the inhibitory effects of cerulenin on proliferation, migration, invasion and glycolysis of ErbB2-overexpressing breast cancer cells and its molecular mechanism. We found that cerulenin obviously suppresses the proliferation, migration, invasion as well as glycolysis. Through bioinformatic analyses, we found that PKM2 might be a target of cerulenin. In addition, ErbB2 and its signaling pathway upregulated PKM2 protein levels. Furthermore, we demonstrated that cerulenin downregulated the protein levels of ErbB2, PKM2 and EMT markers (MMP9, MMP2 and Snail2) in a dose- and time-dependent manner. Finally, the inhibitory of cerulenin on colony formation, migration, invasion and glycolysis, as well as protein levels of EMT markers were rescued by replenishing with PKM2. These findings illustrated that cerulenin inhibits proliferation, migration, invasion and glycolysis by targeting ErbB2/PKM2 pathway in ErbB2-overexpressing breast cancer cells.
Collapse
Affiliation(s)
- Sinan Lv
- Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, No. 17, Section 3, South Renmin Road, Chengdu, 610041, China
| | - Yunwu Zhang
- Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, No. 17, Section 3, South Renmin Road, Chengdu, 610041, China
| | - Jiawei Song
- Department of Physiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Jingruo Chen
- Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, No. 17, Section 3, South Renmin Road, Chengdu, 610041, China
| | - Bohan Huang
- Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, No. 17, Section 3, South Renmin Road, Chengdu, 610041, China
| | - Yuhan Luo
- Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, No. 17, Section 3, South Renmin Road, Chengdu, 610041, China
| | - Yuhua Zhao
- Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, No. 17, Section 3, South Renmin Road, Chengdu, 610041, China.
| |
Collapse
|
28
|
Cui Y, Li C, Sang F, Cao W, Qin Z, Zhang P. Natural products targeting glycolytic signaling pathways-an updated review on anti-cancer therapy. Front Pharmacol 2022; 13:1035882. [PMID: 36339566 PMCID: PMC9631946 DOI: 10.3389/fphar.2022.1035882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 09/30/2022] [Indexed: 11/30/2022] Open
Abstract
Glycolysis is a complex metabolic process that occurs to convert glucose into pyruvate to produce energy for living cells. Normal cells oxidized pyruvate into adenosine triphosphate and carbon dioxide in the presence of oxygen in mitochondria while cancer cells preferentially metabolize pyruvate to lactate even in the presence of oxygen in order to maintain a slightly acidic micro-environment of PH 6.5 and 6.9, which is beneficial for cancer cell growth and metastasis. Therefore targeting glycolytic signaling pathways provided new strategy for anti-cancer therapy. Natural products are important sources for the treatment of diseases with a variety of pharmacologic activities. Accumulated studies suggested that natural products exhibited remarkable anti-cancer properties both in vitro and in vivo. Plenty of studies suggested natural products like flavonoids, terpenoids and quinones played anti-cancer properties via inhibiting glucose metabolism targets in glycolytic pathways. This study provided an updated overview of natural products controlling glycolytic pathways, which also provide insight into druggable mediators discovery targeting cancer glucose metabolism.
Collapse
Affiliation(s)
- Yuting Cui
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, China
| | - Chuang Li
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, China
| | - Feng Sang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, China
| | - Weiling Cao
- Department of Pharmacy, Shenzhen Luohu People’s Hospital, Shenzhen, Guangdong, China
- *Correspondence: Weiling Cao, ; Zhuo Qin, ; Peng Zhang,
| | - Zhuo Qin
- Department of Pharmacy, Shenzhen Luohu People’s Hospital, Shenzhen, Guangdong, China
- *Correspondence: Weiling Cao, ; Zhuo Qin, ; Peng Zhang,
| | - Peng Zhang
- Department of Pharmacy, Shenzhen Luohu People’s Hospital, Shenzhen, Guangdong, China
- *Correspondence: Weiling Cao, ; Zhuo Qin, ; Peng Zhang,
| |
Collapse
|
29
|
He W, He X, Li E. Comprehensive analysis of aerobic glycolysis-related genes for prognosis, immune features and drug treatment strategy in prostate cancer. Front Oncol 2022; 12:905888. [PMID: 36249009 PMCID: PMC9556868 DOI: 10.3389/fonc.2022.905888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 08/29/2022] [Indexed: 12/01/2022] Open
Abstract
Background The dysregulated expression of aerobic glycolysis-related genes is closely related to prostate cancer progression and metastasis. However, reliable prognostic signatures based on aerobic glycolysis have not been well established. Methods We screened aerobic glycolysis-related gene modules by weighted gene co-expression network analysis (WGCNA) and established the aerobic glycolysis-related prognostic risk score (AGRS) by univariate Cox and lasso-Cox. In addition, enriched pathways, genomic mutation, and tumor-infiltrating immune cells were analyzed in AGRS subgroups and compared to each other. We also assessed chemotherapeutic drug sensitivity and immunotherapy response among two subgroups. Results An aerobic glycolysis-related 14-gene prognostic model has been established. This model has good predictive prognostic performance both in the training dataset and in two independent validation datasets. Higher AGRS group patients had better immunotherapy response. Different AGRS patients were also associated with sensitivity of multiple prostate cancer chemotherapeutic drugs. We also predicted eight aerobic glycolysis-related small-molecule drugs by differentially expressed genes. Conclusion In summary, the aerobic glycolysis-derived signatures are promising biomarkers to predict clinical outcomes and therapeutic responses in prostate cancer.
Collapse
|
30
|
Ning X, Qi H, Yuan Y, Li R, Wang Y, Lin Z, Yin Y. Identification of a new small molecule that initiates ferroptosis in cancer cells by inhibiting the system Xc - to deplete GSH. Eur J Pharmacol 2022; 934:175304. [PMID: 36174666 DOI: 10.1016/j.ejphar.2022.175304] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 09/22/2022] [Accepted: 09/22/2022] [Indexed: 11/03/2022]
Abstract
Ferroptosis is a non-apoptotic cell death characterized by iron-mediated ROS accumulation and increasing lipid peroxidation. The activation of ferroptosis results in the destruction of cancer cells and overcoming the drug resistance associated with existing chemotherapeutic agents. It is essential to develop new ferroptosis inducers to provide new opportunities for cancer therapy. In this study, we found a small molecule Compound 8 which we had demonstrated to inhibit tumor growth in vivo initiated ferroptosis. Compound 8 treatment elevated the ferroptosis-related genes PTGS2 and CHAC1 mRNA levels in tumor cells. Ferroptosis inhibitors but not the necroptosis inhibitor or the apoptosis inhibitor can suppress the cell death induced by Compound 8. Compound 8 causes overall greater quantity of lipid peroxidation than the classic ferroptosis inducer Erastin through Flow cytometry analysis. The non-targeted lipidomic analysis also showed Compound 8 treatment resulted in oxidized lipid metabolites, similar to Erastin. The mechanism research showed that Compound 8 initiated ferroptosis by inhibiting the system Xc- to deplete GSH. Based on our previous study that Compound 8 blocked the interaction of PKM2 and VDAC3 (a regulator of ferroptosis) to inhibit tumor growth in vivo, Compound 8 may also trigger ferroptosis by regulating VADC3. Thus, Compound 8 not only will offer a potential tumor therapeutic alternative, but also provide an entrance to explore the new mechanism of ferroptosis.
Collapse
Affiliation(s)
- Xianling Ning
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Department of Pathology, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, 100191, China
| | - Hailong Qi
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Department of Pathology, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, 100191, China; Hebei Immune Cell Application Engineering Research Center, Baoding Newish Technology Co., Ltd./Newish Technology (Beijing) Co., Ltd., Beijing, 100176,China
| | - Yuyao Yuan
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Department of Pathology, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, 100191, China
| | - Ridong Li
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Department of Pathology, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, 100191, China
| | - Yuanyuan Wang
- National Engineering Laboratory for Digital and Material Technology of Stomatology, School and Hospital of Stomatology, Peking University, Beijing, 100081, China
| | - Zhiqiang Lin
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Department of Pathology, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, 100191, China; National Engineering Laboratory for Digital and Material Technology of Stomatology, School and Hospital of Stomatology, Peking University, Beijing, 100081, China.
| | - Yuxin Yin
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Department of Pathology, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, 100191, China.
| |
Collapse
|
31
|
He D, Feng H, Sundberg B, Yang J, Powers J, Christian AH, Wilkinson JE, Monnin C, Avizonis D, Thomas CJ, Friedman RA, Kluger MD, Hollingsworth MA, Grandgenett PM, Klute KA, Toste FD, Chang CJ, Chio IIC. Methionine oxidation activates pyruvate kinase M2 to promote pancreatic cancer metastasis. Mol Cell 2022; 82:3045-3060.e11. [PMID: 35752173 PMCID: PMC9391305 DOI: 10.1016/j.molcel.2022.06.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 04/06/2022] [Accepted: 06/02/2022] [Indexed: 02/07/2023]
Abstract
Cancer mortality is primarily a consequence of its metastatic spread. Here, we report that methionine sulfoxide reductase A (MSRA), which can reduce oxidized methionine residues, acts as a suppressor of pancreatic ductal adenocarcinoma (PDA) metastasis. MSRA expression is decreased in the metastatic tumors of PDA patients, whereas MSRA loss in primary PDA cells promotes migration and invasion. Chemoproteomic profiling of pancreatic organoids revealed that MSRA loss results in the selective oxidation of a methionine residue (M239) in pyruvate kinase M2 (PKM2). Moreover, M239 oxidation sustains PKM2 in an active tetrameric state to promote respiration, migration, and metastasis, whereas pharmacological activation of PKM2 increases cell migration and metastasis in vivo. These results demonstrate that methionine residues can act as reversible redox switches governing distinct signaling outcomes and that the MSRA-PKM2 axis serves as a regulatory nexus between redox biology and cancer metabolism to control tumor metastasis.
Collapse
Affiliation(s)
- Dan He
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Huijin Feng
- Institute for Cancer Genetics, Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Belen Sundberg
- Institute for Cancer Genetics, Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Jiaxing Yang
- Institute for Cancer Genetics, Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Justin Powers
- Institute for Cancer Genetics, Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Alec H Christian
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720, USA
| | | | - Cian Monnin
- Metabolomics Innovation Resource, Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada
| | - Daina Avizonis
- Metabolomics Innovation Resource, Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada
| | - Craig J Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA; Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Richard A Friedman
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Biomedical Informatics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Michael D Kluger
- Division of Gastrointestinal & Endocrine Surgery, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Michael A Hollingsworth
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Paul M Grandgenett
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Kelsey A Klute
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - F Dean Toste
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Christopher J Chang
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA.
| | - Iok In Christine Chio
- Institute for Cancer Genetics, Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
32
|
Zhao K, Wang X, Zhao D, Lin Q, Zhang Y, Hu Y. lncRNA HITT Inhibits Lactate Production by Repressing PKM2 Oligomerization to Reduce Tumor Growth and Macrophage Polarization. RESEARCH (WASHINGTON, D.C.) 2022; 2022:9854904. [PMID: 35909936 PMCID: PMC9285634 DOI: 10.34133/2022/9854904] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 05/20/2022] [Indexed: 12/21/2022]
Abstract
Lactic acid acidifies the tumor microenvironment and promotes multiple critical oncogenic processes, including immune evasion. Pyruvate kinase M2 (PKM2) is a dominant form of pyruvate kinase (PK) expressed in cancers that plays essential roles in metabolic reprograming and lactate production, rendering it as an attractive therapeutic target of cancer. However, the mechanism underlying PKM2 regulation remains unclear. Here, we show that long noncoding RNA (lncRNA) HIF-1α inhibitor at transcription level (HITT) inhibits lactate production in a PKM2-dependent manner. Mechanistically, it physically interacts with PKM2 mapped to a region that has been involved in both dimer (less-active) and tetramer (more-active) formation, inhibiting PKM2 oligomerization and leading to dramatic reduction of PK activity. Under glucose starvation, HITT was reduced as a result of miR-106 induction, which subsequently facilitates PKM2 oligomerization and increases vulnerability to apoptosis under glucose starvation stress. In addition, the interaction also reduces lactate secretion from cancer cells, which subsequently polarizes macrophages toward an M2-like anti-inflammatory phenotype and thus possibly contributes to immune escape in vivo. This study highlights an important role of an lncRNA in regulating PKM2 activity and also reveals a metabolic regulatory effect of PKM2 on macrophage polarization.
Collapse
Affiliation(s)
- Kunming Zhao
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, China 150001.,School of Public Health, Qingdao University, Qingdao, China 266071
| | - Xingwen Wang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, China 150001
| | - Dong Zhao
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, China 150001
| | - Qingyu Lin
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, China 150001
| | - Yi Zhang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, China 150001
| | - Ying Hu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, China 150001
| |
Collapse
|
33
|
Wang Y, Shu H, Liu J, Jin X, Wang L, Qu Y, Xia M, Peng P, Feng Y, Wei M. EGF promotes PKM2 O-GlcNAcylation by stimulating O-GlcNAc transferase phosphorylation at Y976 and their subsequent association. J Biol Chem 2022; 298:102340. [PMID: 35931120 PMCID: PMC9436816 DOI: 10.1016/j.jbc.2022.102340] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 07/22/2022] [Accepted: 07/23/2022] [Indexed: 11/30/2022] Open
Abstract
Epidermal growth factor (EGF) is one of the most well-characterized growth factors and plays a crucial role in cell proliferation and differentiation. Its receptor EGFR has been extensively explored as a therapeutic target against multiple types of cancers, such as lung cancer and glioblastoma. Recent studies have established a connection between deregulated EGF signaling and metabolic reprogramming, especially rewiring in aerobic glycolysis, which is also known as the Warburg effect and recognized as a hallmark in cancer. Pyruvate kinase M2 (PKM2) is a rate-limiting enzyme controlling the final step of glycolysis and serves as a major regulator of the Warburg effect. We previously showed that PKM2 T405/S406 O-GlcNAcylation, a critical mark important for PKM2 detetramerization and activity, was markedly upregulated by EGF. However, the mechanism by which EGF regulates PKM2 O-GlcNAcylation still remains uncharacterized. Here, we demonstrated that EGF promoted O-GlcNAc transferase (OGT) binding to PKM2 by stimulating OGT Y976 phosphorylation. As a consequence, we found PKM2 O-GlcNAcylation and detetramerization were upregulated, leading to a significant decrease in PKM2 activity. Moreover, distinct from PKM2, we observed that the association of additional phosphotyrosine-binding proteins with OGT was also enhanced when Y976 was phosphorylated. These proteins included STAT1, STAT3, STAT5, PKCδ, and p85, which are reported to be O-GlcNAcylated. Together, we show EGF-dependent Y976 phosphorylation is critical for OGT-PKM2 interaction and propose that this posttranslational modification might be important for substrate selection by OGT.
Collapse
Affiliation(s)
- Yang Wang
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, Changchun, Jilin, 130024, China
| | - Hengyao Shu
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, Changchun, Jilin, 130024, China
| | - Jia Liu
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, Changchun, Jilin, 130024, China
| | - Xin Jin
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, Changchun, Jilin, 130024, China
| | - Lihua Wang
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, Changchun, Jilin, 130024, China
| | - Yanzhao Qu
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, Changchun, Jilin, 130024, China
| | - Mingjie Xia
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, Changchun, Jilin, 130024, China
| | - Pinghui Peng
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, Changchun, Jilin, 130024, China
| | - Yunpeng Feng
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, Changchun, Jilin, 130024, China.
| | - Min Wei
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, Changchun, Jilin, 130024, China.
| |
Collapse
|
34
|
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
|
35
|
Read GH, Bailleul J, Vlashi E, Kesarwala AH. Metabolic response to radiation therapy in cancer. Mol Carcinog 2022; 61:200-224. [PMID: 34961986 PMCID: PMC10187995 DOI: 10.1002/mc.23379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 12/01/2021] [Accepted: 12/01/2021] [Indexed: 11/11/2022]
Abstract
Tumor metabolism has emerged as a hallmark of cancer and is involved in carcinogenesis and tumor growth. Reprogramming of tumor metabolism is necessary for cancer cells to sustain high proliferation rates and enhanced demands for nutrients. Recent studies suggest that metabolic plasticity in cancer cells can decrease the efficacy of anticancer therapies by enhancing antioxidant defenses and DNA repair mechanisms. Studying radiation-induced metabolic changes will lead to a better understanding of radiation response mechanisms as well as the identification of new therapeutic targets, but there are few robust studies characterizing the metabolic changes induced by radiation therapy in cancer. In this review, we will highlight studies that provide information on the metabolic changes induced by radiation and oxidative stress in cancer cells and the associated underlying mechanisms.
Collapse
Affiliation(s)
- Graham H. Read
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Justine Bailleul
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Erina Vlashi
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California
| | - Aparna H. Kesarwala
- Department of Radiation Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
36
|
Fan M, Sun W, Gu X, Lu S, Shen Q, Liu X, Zhang X. The critical role of STAT3 in biogenesis of tumor-derived exosomes with potency of inducing cancer cachexia in vitro and in vivo. Oncogene 2022; 41:1050-1062. [PMID: 35034093 DOI: 10.1038/s41388-021-02151-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/26/2021] [Accepted: 12/08/2021] [Indexed: 02/07/2023]
Abstract
Tumor-derived exosomes are emerging mediators of cancer cachexia. Clarifying the regulation of exosome biogenesis and finding possible targets for cancer cachexia therapy are important and necessary. In the present study, systemic analysis of the roles of STAT3 in controlling exosome biogenesis of murine C26 colon tumor cells and its contribution to the development of cancer cachexia is conducted. The genetic manipulation of STAT3 expression, STAT3 knockout (KO) or overexpression (OE), significantly affected the exosome biogenesis and also the potency of C26 conditioned medium (CM) in inducing muscle atrophy and lipolysis in vitro. The genetic manipulation of STAT3 expression caused change in phosphorylation of PKM2 and glycolysis. PKM2/SNAP23 pathway was involved in regulation of exosome biogenesis by STAT3 genetic manipulation as well as by STAT3 inhibitors in C26 cells. Mice inoculated with STAT3 knockout or overexpression C26 cells exhibited ameliorated or aggravated cancer cachexia symptoms, with a positive correlation with the serum exosome and IL-6 levels. The STAT3/PKM2/SNAP23 pathway was affected in C26 tumor tissues with genetic manipulation of STAT3 expression. The capacity of exosome biogenesis of different human cancer cells also exhibited a positive correlation with the activation of STAT3/PKM2/SNAP23 pathway. The research presented here confirms that STAT3 plays a critical role in regulating biogenesis of tumor-derived exosomes which could contribute to cancer cachexia development.
Collapse
Affiliation(s)
- Meng Fan
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Weikuan Sun
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Xiaofan Gu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Shanshan Lu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Qiang Shen
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xuan Liu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Xiongwen Zhang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China.
| |
Collapse
|
37
|
Wang M, Yu F, Zhang Y, Zhang L, Chang W, Wang K. The Emerging Roles of Circular RNAs in the Chemoresistance of Gastrointestinal Cancer. Front Cell Dev Biol 2022; 10:821609. [PMID: 35127685 PMCID: PMC8814461 DOI: 10.3389/fcell.2022.821609] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/06/2022] [Indexed: 12/24/2022] Open
Abstract
Gastrointestinal (GI) cancer represents a major global health problem due to its aggressive characteristics and poor prognosis. Despite the progress achieved in the development of treatment regimens, the clinical outcomes and therapeutic responses of patients with GI cancer remain unsatisfactory. Chemoresistance arising throughout the clinical intervention is undoubtedly a critical barrier for the successful treatment of GI cancer. However, the precise mechanisms associated with chemoresistance in GI cancer remain unclear. In the past decade, accumulating evidence has indicated that circular RNAs (circRNAs) play a key role in regulating cancer progression and chemoresistance. Notably, circRNAs function as molecular sponges that sequester microRNAs (miRNAs) and/or proteins, and thus indirectly control the expression of specific genes, which eventually promote or suppress drug resistance in GI cancer. Therefore, circRNAs may represent potential therapeutic targets for overcoming drug resistance in patients with GI cancer. This review comprehensively summarizes the regulatory roles of circRNAs in the development of chemoresistance in different GI cancers, including colorectal cancer, gastric cancer and esophageal cancer, as well as deciphers the underlying mechanisms and key molecules involved. Increasing knowledge of the important functions of circRNAs underlying drug resistance will provide new opportunities for developing efficacious therapeutic strategies against GI cancer.
Collapse
Affiliation(s)
- Man Wang
- *Correspondence: Man Wang, ; Kun Wang,
| | | | | | | | | | - Kun Wang
- *Correspondence: Man Wang, ; Kun Wang,
| |
Collapse
|
38
|
Zhang C, Tang Q, Xia H, Xu H, Bi F. PKM2 compensates for proteasome dysfunction by mediating the formation of the CHIP-HSP70-BAG3 complex and the aggregation of ubiquitinated proteins. FASEB J 2021; 36:e22121. [PMID: 34951719 DOI: 10.1096/fj.202101342rr] [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: 08/23/2021] [Revised: 12/01/2021] [Accepted: 12/13/2021] [Indexed: 02/05/2023]
Abstract
Protein aggregation and degradation via autophagy (aggrephagy) are major strategies adopted by cells to remove misfolded polypeptides when there is proteasome dysfunction. The functional protein complex consisting of heat shock protein 70 (Hsp70), cochaperone ubiquitin ligase carboxyl-terminal of Hsp70/Hsp90 interacting protein (CHIP), and co-chaperone Bcl-2-associated athanogene 3 (BAG3) has been associated with the activation of protein aggregation. However, data on the mechanisms of action of the complex in the protein degradation remains scant. Here, we report that upon proteasome stress, the M2 isoform of pyruvate kinase (PKM2) promotes the aggregation of ubiquitinated proteins and its knockout or knockdown aggravates the sensitivity of cells to proteasome inhibitors. Besides, following proteasome inhibition, PKM2 promotes the interaction of BAG3 with CHIP and HSP70. Interestingly, re-expression of loss-of-function mutants in PKM2-knockout cells showed that the regulatory function of PKM2 in this progress does not depend on the activity of glycolytic enzymes or protein kinases. Taken together, these findings demonstrate that PKM2 mediates the formation of the CHIP-HSP70-BAG3 protein complex and promotes the aggregation of ubiquitinated misfolded proteins, thus compensating for proteasome stress in cells.
Collapse
Affiliation(s)
- Chenliang Zhang
- Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital of Sichuan University, Chengdu, China
| | - Qiulin Tang
- Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital of Sichuan University, Chengdu, China
| | - Hongwei Xia
- Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital of Sichuan University, Chengdu, China
| | - Huanji Xu
- Department of Medical Oncology, Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Feng Bi
- Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital of Sichuan University, Chengdu, China.,Department of Medical Oncology, Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
39
|
Ripoll C, Roldan M, Ruedas-Rama MJ, Orte A, Martin M. Breast Cancer Cell Subtypes Display Different Metabolic Phenotypes That Correlate with Their Clinical Classification. BIOLOGY 2021; 10:biology10121267. [PMID: 34943182 PMCID: PMC8698801 DOI: 10.3390/biology10121267] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/28/2021] [Accepted: 11/29/2021] [Indexed: 12/11/2022]
Abstract
Simple Summary Recent studies on cancer cell metabolism have achieved notable breakthroughs that have led to a new scientific paradigm. How cancer cell metabolic reprogramming is orchestrated and the decisive role of this reprogramming in the oncogenic process and tumor adaptative evolution has been characterized at the molecular level. Despite this knowledge, it is essential to understand how cancer cells can metabolically respond as a living whole to ensure their survival and adaptation potential. In this work, we investigated whether different cancers and different subtypes display different metabolic phenotypes with a focus on breast cancer cell models representative of each clinical subtype. The potential results might have significant translational implications for diagnostic, prognostic and therapeutic applications. Abstract Metabolic reprogramming of cancer cells represents an orchestrated network of evolving molecular and functional adaptations during oncogenic progression. In particular, how metabolic reprogramming is orchestrated in breast cancer and its decisive role in the oncogenic process and tumor evolving adaptations are well consolidated at the molecular level. Nevertheless, potential correlations between functional metabolic features and breast cancer clinical classification still represent issues that have not been fully studied to date. Accordingly, we aimed to investigate whether breast cancer cell models representative of each clinical subtype might display different metabolic phenotypes that correlate with current clinical classifications. In the present work, functional metabolic profiling was performed for breast cancer cell models representative of each clinical subtype based on the combination of enzyme inhibitors for key metabolic pathways, and isotope-labeled tracing dynamic analysis. The results indicated the main metabolic phenotypes, so-called ‘metabophenotypes’, in terms of their dependency on glycolytic metabolism or their reliance on mitochondrial oxidative metabolism. The results showed that breast cancer cell subtypes display different metabophenotypes. Importantly, these metabophenotypes are clearly correlated with the current clinical classifications.
Collapse
Affiliation(s)
- Consuelo Ripoll
- Nanoscopy-UGR Laboratory, Departamento de Fisicoquimica, Unidad de Excelencia de Química Aplicada a Biomedicina y Medioambiente, Facultad de Farmacia, Universidad de Granada, Campus Cartuja, 18071 Granada, Spain; (C.R.); (M.J.R.-R.)
- GENYO, Pfizer-Universidad de Granada-Junta de Andalucia Centre for Genomics and Oncological Research, Avda Ilustracion 114, PTS, 18016 Granada, Spain;
| | - Mar Roldan
- GENYO, Pfizer-Universidad de Granada-Junta de Andalucia Centre for Genomics and Oncological Research, Avda Ilustracion 114, PTS, 18016 Granada, Spain;
- Departamento de Bioquímica y Biología Molecular I, Facultad de Ciencias, Universidad de Granada, Avda. Fuentenueva, 18071 Granada, Spain
| | - Maria J. Ruedas-Rama
- Nanoscopy-UGR Laboratory, Departamento de Fisicoquimica, Unidad de Excelencia de Química Aplicada a Biomedicina y Medioambiente, Facultad de Farmacia, Universidad de Granada, Campus Cartuja, 18071 Granada, Spain; (C.R.); (M.J.R.-R.)
| | - Angel Orte
- Nanoscopy-UGR Laboratory, Departamento de Fisicoquimica, Unidad de Excelencia de Química Aplicada a Biomedicina y Medioambiente, Facultad de Farmacia, Universidad de Granada, Campus Cartuja, 18071 Granada, Spain; (C.R.); (M.J.R.-R.)
- Correspondence: (A.O.); (M.M.)
| | - Miguel Martin
- GENYO, Pfizer-Universidad de Granada-Junta de Andalucia Centre for Genomics and Oncological Research, Avda Ilustracion 114, PTS, 18016 Granada, Spain;
- Departamento de Bioquímica y Biología Molecular I, Facultad de Ciencias, Universidad de Granada, Avda. Fuentenueva, 18071 Granada, Spain
- Correspondence: (A.O.); (M.M.)
| |
Collapse
|
40
|
Zhou B, Lu D, Wang A, Cui J, Zhang L, Li J, Fan L, Wei W, Liu J, Sun G. Endoplasmic reticulum stress promotes sorafenib resistance via miR-188-5p/hnRNPA2B1-mediated upregulation of PKM2 in hepatocellular carcinoma. MOLECULAR THERAPY-NUCLEIC ACIDS 2021; 26:1051-1065. [PMID: 34786210 PMCID: PMC8569435 DOI: 10.1016/j.omtn.2021.09.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 07/22/2021] [Accepted: 09/24/2021] [Indexed: 01/27/2023]
Abstract
Emerging evidence has shown that endoplasmic reticulum (ER) stress promotes sorafenib resistance in hepatocellular carcinoma (HCC). However, the underlying mechanisms are poorly understood. The purpose of this study was to explore the mechanism by which ER stress promotes sorafenib resistance in HCC. We found that pyruvate kinase isoform M2 (PKM2) was highly expressed in human HCC tissues and co-related with worse clinicopathologic features and overall survival. Activation of ER stress positively correlated with PKM2 expression both in HCC tissue samples and tunicamycin (TM)-induced HCC cell lines. PKM2 knockdown increased sorafenib-induced apoptosis and decreased the ability of colony formation, while upregulation of PKM2 reverses this phenomenon. Furthermore, high-throughput sequencing identified that activation of ER stress significantly downregulated the expression of miR-188-5p in HCC cells. According to bioinformatics analysis and dual-luciferase assays, we further confirmed that hnRNPA2B1 is the target gene of miR-188-5p. Downregulating the expression of hnRNPA2B1 with siRNA could decrease the expression of PKM2 and enhance sorafenib-induced apoptosis in HepG2 cells. Our study demonstrated that ER stress could promote sorafenib resistance through upregulating PKM2 via miR-188-5p/hnRNPA2B1. Therefore, targeting the miR-188-5p/hnRNPA2B1/PKM2 pathway and ER stress may prove instrumental in overcoming sorafenib resistance in HCC treatment.
Collapse
Affiliation(s)
- Bei Zhou
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, China
| | - Donghui Lu
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, China
| | - Anqi Wang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, China
| | - Jie Cui
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, China
| | - Li Zhang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, China
| | - Jian Li
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, China
| | - Lulu Fan
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei 230032, Anhui, China
| | - Jiatao Liu
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, China.,Department of Pharmacy, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, China
| | - Guoping Sun
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, China
| |
Collapse
|
41
|
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
|
42
|
Hu Y, Mu H, Deng Z. The transcription factor TEAD4 enhances lung adenocarcinoma progression through enhancing PKM2 mediated glycolysis. Cell Biol Int 2021; 45:2063-2073. [PMID: 34196069 DOI: 10.1002/cbin.11654] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 04/20/2021] [Accepted: 06/16/2021] [Indexed: 01/24/2023]
Abstract
Lung adenocarcinoma (LUAD) is a deadly disease with a hallmark of aberrant metabolism. TEA domain 4 (TEAD4) is involved in the progression of several forms of cancer including LUAD. However, the role of TEAD4 in LUAD glucose metabolism is rarely reported as well as its potential mechanisms. Pyruvate kinase isozymes M2 (PKM2), the key regulatory enzymes in glycolysis, was predicted to be a target for TEAD4 by bioinformatics analysis. Thus, we aimed to explore whether TEAD4/PKM2 axis was related to LUAD glucose metabolism and malignant phenotype. The expression level of TEAD4 and PKM2 was measured by quantitative real-time PCR and Western blot. Luciferase reporter assay were employed to verify the effect of TEAD4 on PKM2 promoter as well as TEAD4/PKM2 axis on reporter activity of hypoxia inducible factor-1α (HIF-1α). Glycolysis was investigated according to glucose consumption, lactate production and the extracellular acidification rate. The present study indicated that TEAD4 and PKM2 were upregulated in LUAD and closely related to prognosis. Mechanistic investigations identified that TEAD4 played a key role as a transcription factor and promoted PKM2 transcription and expression, which further altered the reporter activity of HIF-1α and upregulated HIF-1α-targeted glycolytic genes glucose transporter-1 and hexokinase II. Functional assays revealed that TEAD4 and PKM2 affected glycolytic and 2-DG blocked the positive function of TEAD4 and PKM2 on glycolytic. Besides, TEAD4/PKM2 axis affects LUAD cell viability, apoptosis, migration, and invasion. Together, these data provided evidence that both TEAD4 and PKM2 were poor prognosticator. Targeting TEAD4/PKM2 axis might be an effective therapeutic strategy for LUAD.
Collapse
Affiliation(s)
- Yan Hu
- Department of Respiratory, The First People's Hospital of Zigong City, Zigong, Sichuan, China
| | - Hanshuo Mu
- Department of Clinical Medicine, Nantong University, Nantong, Jiangsu, China
| | - Zhiping Deng
- Department of Respiratory, The First People's Hospital of Zigong City, Zigong, Sichuan, China
| |
Collapse
|
43
|
Huang P, Zhu S, Liang X, Zhang Q, Liu C, Song L. Revisiting Lung Cancer Metastasis: Insight From the Functions of Long Non-coding RNAs. Technol Cancer Res Treat 2021; 20:15330338211038488. [PMID: 34431723 PMCID: PMC8392855 DOI: 10.1177/15330338211038488] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Globally, lung cancer is the most common cause of cancer-related deaths. After
diagnosis at all stages, <7% of patients survive for 10 years. Thus,
diagnosis at later stages and the lack of effective and personalized drugs
reflect a significant need to better understand the mechanisms underpinning lung
cancer progression. Metastasis should be responsible for the high lethality and
recurrence rates seen in lung cancer. Metastasis depends on multiple crucial
steps, including epithelial–mesenchymal transition, vascular remodeling, and
colonization. Therefore, in-depth investigations of metastatic molecular
mechanisms can provide valuable insights for lung cancer treatment. Recently,
long noncoding RNAs (lncRNAs) have attracted considerable attention owing to
their complex roles in cancer progression. In lung cancer, multiple lncRNAs have
been reported to regulate metastasis. In this review, we highlight the major
molecular mechanisms underlying lncRNA-mediated regulation of lung cancer
metastasis, including (1) lncRNAs acting as competing endogenous RNAs, (2)
lncRNAs regulating the transduction of several signal pathways, and (3) lncRNA
coordination with enhancer of zeste homolog 2. Thus, lncRNAs appear to execute
their functions on lung cancer metastasis by regulating angiogenesis, autophagy,
aerobic glycolysis, and immune escape. However, more comprehensive studies are
required to characterize these lncRNA regulatory networks in lung cancer
metastasis, which can provide promising and innovative novel therapeutic
strategies to combat this disease.
Collapse
Affiliation(s)
- Peng Huang
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, P.R. China
| | - Shaomi Zhu
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, P.R. China
| | - Xin Liang
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, P.R. China
| | - Qinxiu Zhang
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, P.R. China
| | - Chi Liu
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, P.R. China
| | - Linjiang Song
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, P.R. China
| |
Collapse
|
44
|
Lee YB, Min JK, Kim JG, Cap KC, Islam R, Hossain AJ, Dogsom O, Hamza A, Mahmud S, Choi DR, Kim YS, Koh YH, Kim HA, Chung WS, Suh SW, Park JB. Multiple functions of pyruvate kinase M2 in various cell types. J Cell Physiol 2021; 237:128-148. [PMID: 34311499 DOI: 10.1002/jcp.30536] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/28/2021] [Accepted: 07/13/2021] [Indexed: 02/06/2023]
Abstract
Glucose metabolism is a mechanism by which energy is produced in form of adenosine triphosphate (ATP) by mitochondria and precursor metabolites are supplied to enable the ultimate enrichment of mature metabolites in the cell. Recently, glycolytic enzymes have been shown to have unconventional but important functions. Among these enzymes, pyruvate kinase M2 (PKM2) plays several roles including having conventional metabolic enzyme activity, and also being a transcriptional regulator and a protein kinase. Compared with the closely related PKM1, PKM2 is highly expressed in cancer cells and embryos, whereas PKM1 is dominant in mature, differentiated cells. Posttranslational modifications such as phosphorylation and acetylation of PKM2 change its cellular functions. In particular, PKM2 can translocate to the nucleus, where it regulates the transcription of many target genes. It is notable that PKM2 also acts as a protein kinase to phosphorylate several substrate proteins. Besides cancer cells and embryonic cells, astrocytes also highly express PKM2, which is crucial for lactate production via expression of lactate dehydrogenase A (LDHA), while mature neurons predominantly express PKM1. The lactate produced in cancer cells promotes tumor progress and that in astrocytes can be supplied to neurons and may act as a major source for neuronal ATP energy production. Thereby, we propose that PKM2 along with its different posttranslational modifications has specific purposes for a variety of cell types, performing unique functions.
Collapse
Affiliation(s)
- Yoon-Beom Lee
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Jung K Min
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Jae-Gyu Kim
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, Republic of Korea.,Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Kim Cuong Cap
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, Republic of Korea.,eLmed Inc. #3419, Hallym University, Chuncheon, Kangwon-do, Republic of Korea.,Institute of Research and Development, Duy Tan University, Danang, Vietnam
| | - Rokibul Islam
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, Republic of Korea.,Department of Biotechnology and Genetic Engineering, Faculty of Biological Science, Islamic University, Kushtia, Bangladesh
| | - Abu J Hossain
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Oyungerel Dogsom
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, Republic of Korea.,Department of Biology, School of Bio-Medicine, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Amir Hamza
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Shohel Mahmud
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, Republic of Korea.,National Institute of Biotechnology, Ganakbari, Savar, Dhaka, Bangladesh
| | - Dae R Choi
- Department of Internal Medicine, Chuncheon Sacred Heart Hospital, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Yong-Sun Kim
- Ilsong Institute of Life Science, Hallym University, Seoul, Republic of Korea
| | - Young-Ho Koh
- Ilsong Institute of Life Science, Hallym University, Seoul, Republic of Korea
| | - Hyun-A Kim
- Department of Internal Medicine, Hallym Sacred Heart Hospital, College of Medicine, Hallym University, Ahnyang, Republic of Korea
| | - Won-Suk Chung
- Department of Biological Science, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Sang W Suh
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Jae-Bong Park
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, Republic of Korea.,Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon, Republic of Korea.,eLmed Inc. #3419, Hallym University, Chuncheon, Kangwon-do, Republic of Korea
| |
Collapse
|
45
|
Wang Z, Zhang X, Zhang H, Tang Y, Pan H, Wang H, Ji T, Guo Y, Gao Q, Song T, Zhang Z. Discovery of a Fluorogenic Probe for In Situ Pyruvate Kinase M2 Isoform (PKM2) Labeling through Chemoselective S NAr with a Binding Site Lysine Residue. Anal Chem 2021; 93:9669-9676. [PMID: 34219457 DOI: 10.1021/acs.analchem.1c00208] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The key challenge of developing reaction-based turn-on probes is to establish latent electrophilic fluorophores exhibiting high reactivity only upon binding to a specific protein(s). Herein, we identified such a fluorophore, 6-arylthioether-substituted 3-cyano-1-oxo-1H-phenalene-2-carboxylate, which chemoselectively labels binding site Cys or Lys residues. Based on this fluorophore, we developed the first reaction-based turn-on pyruvate kinase M2 isoform (PKM2) fluorescent probe AT-OPC1, which selectively labels PKM2 with the binding site Lys305. The latent electrophilic reactivity of the fluorophore endows the probe with precise detection of the expression of PKM2 in situ by means of both in-gel fluorescence imaging at the proteome level and real-time no-wash cell imaging approaches, which has the potential to be applied in cancer diagnoses.
Collapse
Affiliation(s)
- Ziqian Wang
- State Key Laboratory of Fine Chemicals, Zhang Dayu School of Chemistry, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Xiaodong Zhang
- State Key Laboratory of Fine Chemicals, Zhang Dayu School of Chemistry, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Hong Zhang
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Yao Tang
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Hao Pan
- State Key Laboratory of Fine Chemicals, Zhang Dayu School of Chemistry, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Hang Wang
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Tong Ji
- State Key Laboratory of Fine Chemicals, Zhang Dayu School of Chemistry, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Yafei Guo
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Qishuang Gao
- State Key Laboratory of Fine Chemicals, Zhang Dayu School of Chemistry, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Ting Song
- State Key Laboratory of Fine Chemicals, Zhang Dayu School of Chemistry, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Zhichao Zhang
- State Key Laboratory of Fine Chemicals, Zhang Dayu School of Chemistry, Dalian University of Technology, Dalian, Liaoning 116024, China
| |
Collapse
|
46
|
Wiese EK, Hitosugi S, Loa ST, Sreedhar A, Andres-Beck LG, Kurmi K, Pang YP, Karnitz LM, Gonsalves WI, Hitosugi T. Enzymatic activation of pyruvate kinase increases cytosolic oxaloacetate to inhibit the Warburg effect. Nat Metab 2021; 3:954-968. [PMID: 34226744 PMCID: PMC8316326 DOI: 10.1038/s42255-021-00424-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 06/04/2021] [Indexed: 11/30/2022]
Abstract
Pharmacological activation of the glycolytic enzyme PKM2 or expression of the constitutively active PKM1 isoform in cancer cells results in decreased lactate production, a phenomenon known as the PKM2 paradox in the Warburg effect. Here we show that oxaloacetate (OAA) is a competitive inhibitor of human lactate dehydrogenase A (LDHA) and that elevated PKM2 activity increases de novo synthesis of OAA through glutaminolysis, thereby inhibiting LDHA in cancer cells. We also show that replacement of human LDHA with rabbit LDHA, which is relatively resistant to OAA inhibition, eliminated the paradoxical correlation between the elevated PKM2 activity and the decreased lactate concentration in cancer cells treated with a PKM2 activator. Furthermore, rabbit LDHA-expressing tumours, compared to human LDHA-expressing tumours in mice, displayed resistance to the PKM2 activator. These findings describe a mechanistic explanation for the PKM2 paradox by showing that OAA accumulates and inhibits LDHA following PKM2 activation.
Collapse
Affiliation(s)
- Elizabeth K Wiese
- Molecular Pharmacology and Experimental Therapeutics Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Sadae Hitosugi
- Division of Oncology Research, Mayo Clinic, Rochester, MN, USA
| | - Sharon T Loa
- Division of Oncology Research, Mayo Clinic, Rochester, MN, USA
| | | | - Lindsey G Andres-Beck
- Molecular Pharmacology and Experimental Therapeutics Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Kiran Kurmi
- Division of Oncology Research, Mayo Clinic, Rochester, MN, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Yuan-Ping Pang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Larry M Karnitz
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
- Division of Oncology Research, Mayo Clinic, Rochester, MN, USA
| | | | - Taro Hitosugi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA.
- Division of Oncology Research, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
47
|
Verma H, Cholia RP, Kaur S, Dhiman M, Mantha AK. A short review on cross-link between pyruvate kinase (PKM2) and Glioblastoma Multiforme. Metab Brain Dis 2021; 36:751-765. [PMID: 33651273 DOI: 10.1007/s11011-021-00690-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 02/10/2021] [Indexed: 12/23/2022]
Abstract
Pyruvate kinase (PK) catalyzes the last irreversible reaction of glycolysis pathway, generating pyruvate and ATP, from Phosphoenol Pyruvate (PEP) and ADP precursors. In mammals, four different tissue-specific isoforms (M1, M2, L and R) of PK exist, which are translated from two genes (PKL and PKR). PKM2 is the highly expressed isoform of PK in cancers, which regulates the aerobic glycolysis via reprogramming cancer cell's metabolic pathways to provide an anabolic advantage to the tumor cells. In addition to the established role of PKM2 in aerobic glycolysis of multiple cancer types, various recent findings have highlighted the non-metabolic functions of PKM2 in brain tumor development. Nuclear PKM2 acts as a co-activator and directly regulates gene transcription. PKM2 dependent transactivation of various oncogenic genes is instrumental in the progression and aggressiveness of Glioblastoma Multiforme (GBM). Also, PKM2 acts as a protein kinase in histone modification which regulates gene expression and tumorigenesis. Ongoing research has explored novel regulatory mechanisms of PKM2 and its association in GBM progression. This review enlists and summarizes the metabolic and non-metabolic roles of PKM2 at the cellular level, and its regulatory function highlights the importance of the nuclear functions of PKM2 in GBM progression, and an emerging role of PKM2 as novel cancer therapeutics.
Collapse
Affiliation(s)
- Harkomal Verma
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Village Ghudda, Bathinda, Punjab, Pin Code: 151 401, India
| | - Ravi P Cholia
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Village Ghudda, Bathinda, Punjab, Pin Code: 151 401, India
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Sharanjot Kaur
- Department of Microbiology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India
| | - Monisha Dhiman
- Department of Microbiology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India
| | - Anil K Mantha
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Village Ghudda, Bathinda, Punjab, Pin Code: 151 401, India.
| |
Collapse
|
48
|
Pei L, Le Y, Chen H, Feng J, Liu Z, Zhu J, Wang C, Chen L, Dou X, Lu D. Cynaroside prevents macrophage polarization into pro-inflammatory phenotype and alleviates cecal ligation and puncture-induced liver injury by targeting PKM2/HIF-1α axis. Fitoterapia 2021; 152:104922. [PMID: 33984439 DOI: 10.1016/j.fitote.2021.104922] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 04/19/2021] [Accepted: 04/29/2021] [Indexed: 12/12/2022]
Abstract
The treatment of sepsis is still challenging and the liver is an important target of sepsis-related injury. Macrophages are important innate immune cells in liver, and modulation of macrophages M1/M2 polarization may be a promising strategy for septic liver injury treatment. Macrophage polarization and inflammation of liver tissue has been shown regulated by pyruvate kinase M2 (PKM2)-mediated aerobic glycolysis and immune inflammatory pathways. Therefore, modulating PKM2-mediated immunometabolic reprogramming presents a novel strategy for inflammation-associated diseases. In this study, cynaroside, a flavonoid compound, promoted macrophage phenotypic transition from pro-inflammatory M1 to anti-inflammatory M2, and mitigated sepsis-associated liver inflammatory damage. We established that cynaroside reduced binding of PKM2 to hypoxia-inducible factor-1α (HIF-1α) by abolishing translocation of PKM2 to the nucleus and promoting PKM2 tetramer formation, as well as suppressing phosphorylation of PKM2 at Y105 in vivo and in vitro. Moreover, cynaroside restored pyruvate kinase activity, inhibited glycolysis-related proteins including PFKFB3, HK2 and HIF-1α, and inhibited glycolysis-related hyperacetylation of HMGB1 in septic liver. Therefore, this study reports a novel function of cynaroside in hepatic macrophage polarization, and cecum ligation and puncture-induced liver injury in septic mice. The findings provide crucial information with regard to therapeutic efficacy of cynaroside in the treatment of sepsis.
Collapse
Affiliation(s)
- Liuhua Pei
- College of Life Science, Zhejiang Chinese Medical University, 310053 Hangzhou, China
| | - Yifei Le
- College of Life Science, Zhejiang Chinese Medical University, 310053 Hangzhou, China
| | - Hang Chen
- College of Life Science, Zhejiang Chinese Medical University, 310053 Hangzhou, China
| | - Jiafan Feng
- College of Life Science, Zhejiang Chinese Medical University, 310053 Hangzhou, China
| | - Zhijun Liu
- College of Life Science, Zhejiang Chinese Medical University, 310053 Hangzhou, China
| | - Ji Zhu
- Clinical Laboratory, The Third Affiliated Hospital of Zhejiang Chinese Medical University, 330106 Hangzhou, China
| | - Cui Wang
- College of Life Science, Zhejiang Chinese Medical University, 310053 Hangzhou, China
| | - Lin Chen
- College of Life Science, Zhejiang Chinese Medical University, 310053 Hangzhou, China
| | - Xiaobing Dou
- College of Life Science, Zhejiang Chinese Medical University, 310053 Hangzhou, China.
| | - Dezhao Lu
- College of Life Science, Zhejiang Chinese Medical University, 310053 Hangzhou, China.
| |
Collapse
|
49
|
Ma R, Wu Y, Li S, Yu X. Interplay Between Glucose Metabolism and Chromatin Modifications in Cancer. Front Cell Dev Biol 2021; 9:654337. [PMID: 33987181 PMCID: PMC8110832 DOI: 10.3389/fcell.2021.654337] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/19/2021] [Indexed: 12/13/2022] Open
Abstract
Cancer cells reprogram glucose metabolism to meet their malignant proliferation needs and survival under a variety of stress conditions. The prominent metabolic reprogram is aerobic glycolysis, which can help cells accumulate precursors for biosynthesis of macromolecules. In addition to glycolysis, recent studies show that gluconeogenesis and TCA cycle play important roles in tumorigenesis. Here, we provide a comprehensive review about the role of glycolysis, gluconeogenesis, and TCA cycle in tumorigenesis with an emphasis on revealing the novel functions of the relevant enzymes and metabolites. These functions include regulation of cell metabolism, gene expression, cell apoptosis and autophagy. We also summarize the effect of glucose metabolism on chromatin modifications and how this relationship leads to cancer development. Understanding the link between cancer cell metabolism and chromatin modifications will help develop more effective cancer treatments.
Collapse
Affiliation(s)
- Rui Ma
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Environmental Microbial Technology Center of Hubei, School of Life Sciences, Hubei University, Wuhan, China
| | - Yinsheng Wu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Environmental Microbial Technology Center of Hubei, School of Life Sciences, Hubei University, Wuhan, China
| | - Shanshan Li
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Environmental Microbial Technology Center of Hubei, School of Life Sciences, Hubei University, Wuhan, China.,College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, China
| | - Xilan Yu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Environmental Microbial Technology Center of Hubei, School of Life Sciences, Hubei University, Wuhan, China
| |
Collapse
|
50
|
Hauck L, Dadson K, Chauhan S, Grothe D, Billia F. Inhibiting the Pkm2/b-catenin axis drives in vivo replication of adult cardiomyocytes following experimental MI. Cell Death Differ 2021; 28:1398-1417. [PMID: 33288902 PMCID: PMC8027412 DOI: 10.1038/s41418-020-00669-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 11/01/2020] [Accepted: 11/02/2020] [Indexed: 12/19/2022] Open
Abstract
Adult mammalian cardiomyocytes (CM) are postmitotic, differentiated cells that cannot re-enter the cell cycle after any appreciable injury. Therefore, understanding the factors required to induce CM proliferation for repair is of great clinical importance. While expression of muscle pyruvate kinase 2 (Pkm2), a cytosolic enzyme catalyzing the final step in glycolysis, is high in end-stage heart failure (HF), the loss of Pkm2 promotes proliferation in some cellular systems, in vivo. We hypothesized that in the adult heart CM proliferation may require low Pkm2 activity. Thus, we investigated the potential for Pkm2 to regulate CM proliferation in a mouse model of myocardial infarction (MI) employing inducible, cardiac-specific Pkm2 gene knockout (Pkm2KOi) mice. We found a lack of cardiac hypertrophy or expression of the fetal gene program in Pkm2KOi mice post MI, as compared to vehicle control animals (P < 0.01), correlating with smaller infarct size, improved mitochondrial (mt) function, enhanced angiogenesis, reduced degree of CM apoptosis, and reduced oxidative stress post MI. There was significantly higher numbers of dividing CM in the infarct zone between 3-9 days post MI (P < 0.001). Mechanistically, we determined that Pkm2 interacts with β-catenin (Ctnnb1) in the cytoplasm of CM, inhibiting Ctnnb1 phosphorylation at serine 552 and tyrosine 333, by Akt. In the absence of Pkm2, Ctnnb1 translocates to the nucleus leading to transcriptional activation of proliferation-associated target genes. All these effects are abrogated by genetic co-deletion of Pkm2 and Ctnnb1. Collectively, this work supports a novel antiproliferative function for Pkm2 in CM through the sequestration of Ctnnb1 in the cytoplasm of CM whereas loss of Pkm2 is essential for CM proliferation. Reducing cardiac Pkm2 expression may provide a useful strategy for cardiac repair after MI in patients.
Collapse
Affiliation(s)
- Ludger Hauck
- Toronto General Research Institute, 100 College St., M5G 1L7, Toronto, ON, Canada
| | - Keith Dadson
- Toronto General Research Institute, 100 College St., M5G 1L7, Toronto, ON, Canada
| | - Shelly Chauhan
- Toronto General Research Institute, 100 College St., M5G 1L7, Toronto, ON, Canada
| | - Daniela Grothe
- Toronto General Research Institute, 100 College St., M5G 1L7, Toronto, ON, Canada
| | - Filio Billia
- Toronto General Research Institute, 100 College St., M5G 1L7, Toronto, ON, Canada.
- Division of Cardiology, University Health Network (UHN), 200 Elizabeth St., Toronto, ON, M5G 2C4, Canada.
| |
Collapse
|