1
|
Mandal M, Banerjee I, Mandal M. Effective approaches in conquering chemoresistance of glioblastoma: potential for nanoformulations. Drug Deliv Transl Res 2025:10.1007/s13346-025-01859-z. [PMID: 40259195 DOI: 10.1007/s13346-025-01859-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2025] [Indexed: 04/23/2025]
Abstract
Glioblastoma Multiforme is an aggressive and complex cancer affecting mostly elderly patients above the age of 60 years. Originally classified as the fourth stage of glioma, it has an abysmal prognosis along with limited therapeutic options. Surgical removal of tumors, radiotherapy, and chemotherapy are prevalent treatment strategies with numerous therapeutic obstacles, including undefined boundary of tumor mass leaving traces even after excision, chances of secondary cancer formation, and presence of blood-brain barrier. These blood-brain and blood-brain tumor barriers actively restrict the permeability of many molecules from blood circulation to enter the central nervous system. Therefore, many conventional antineoplastic drugs fail to reach the tumor periphery except temozolomide. Meanwhile, active stem cells in the tumor microenvironment, genetic mutation inducing tumor growth, and epigenetic pattern alteration make this cancer chemoresistant. Our review delineates the recent approaches to resensitize the existing clinical drugs through specifically designed nanoformulations. Nanoparticles with modified physiological characteristics and modified through technological parameters can reduce the tumor's stemness, which increases tumor cells' apoptosis rate. Moreover, these nanoparticles can efficiently traverse the blood-brain barrier and escape from endosomal degradation with minimum toxicological impact. Overall, this review discusses the cancer chemoresistance phenomena and related pathways and highlights the potential of nanoformulation in reversing chemoresistance. Also, the existing limitations of this unique approach and suggestions are discussed at the end of the article, which may facilitate the identification of new directions for advancement of the nanoparticle-mediated reversal of chemoresistance.
Collapse
Affiliation(s)
- Madhurima Mandal
- School of Medical Science & Technology, Indian Institute of Technology, Kharagpur, West Bengal, 721302, India
| | - Indranil Banerjee
- Department of Pharmaceutical Technology, JIS University, 81, Nilgunj Road, Agarpara, Kolkata, West Bengal, 700109, India.
| | - Mahitosh Mandal
- School of Medical Science & Technology, Indian Institute of Technology, Kharagpur, West Bengal, 721302, India.
| |
Collapse
|
2
|
Guo D, Meng Y, Zhao G, Wu Q, Lu Z. Moonlighting functions of glucose metabolic enzymes and metabolites in cancer. Nat Rev Cancer 2025:10.1038/s41568-025-00800-3. [PMID: 40175621 DOI: 10.1038/s41568-025-00800-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/13/2025] [Indexed: 04/04/2025]
Abstract
Glucose metabolic enzymes and their metabolites not only provide energy and building blocks for synthesizing macromolecules but also possess non-canonical or moonlighting functions in response to extracellular and intracellular signalling. These moonlighting functions modulate various cellular activities, including gene expression, cell cycle progression, DNA repair, autophagy, senescence and apoptosis, cell proliferation, remodelling of the tumour microenvironment and immune responses. These functions integrate glucose metabolism with other essential cellular activities, driving cancer progression. Targeting these moonlighting functions could open new therapeutic avenues and lead to cancer-specific treatments.
Collapse
Affiliation(s)
- Dong Guo
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Ying Meng
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Gaoxiang Zhao
- Department of Oncology, Cancer Institute of The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, China
| | - Qingang Wu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Zhimin Lu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China.
| |
Collapse
|
3
|
Jaiswal A, Kaushik N, Patel P, Acharya TR, Mukherjee S, Choi EH, Kaushik NK. Nonthermal plasma boosted dichloroacetate induces metabolic shifts to combat glioblastoma CSCs via oxidative stress. Free Radic Biol Med 2025; 229:264-275. [PMID: 39724987 DOI: 10.1016/j.freeradbiomed.2024.12.045] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/11/2024] [Accepted: 12/23/2024] [Indexed: 12/28/2024]
Abstract
Glioblastoma (GBM) remains a formidable clinical challenge, with cancer stem cells (CSCs) contributing to treatment resistance and tumor recurrence. Conventional treatments often fail to eradicate these CSCs characterized by enhanced resistance to standard therapies through metabolic plasticity making them key targets for novel treatment approaches. Addressing this challenge, this study introduces a novel combination therapy of dichloroacetate (DCA), a metabolic modulator and nonthermal plasma to induce oxidative stress in glioblastomas. Our results demonstrate that DCA and nonthermal plasma (NTP) synergistically increase ROS production, resulting in endoplasmic reticulum (ER) stress and mitochondrial reprogramming, key factors in the initiation of programmed cell death. Furthermore, the combination downregulated key stemness markers indicating effective CSCs suppression. Upregulation of pro-apoptotic proteins and downregulation of anti-apoptotic factors highlight the induction of apoptosis in glioma stem cells. This study provides compelling evidence that the combination of DCA and NTP offers a novel and effective strategy for targeting glioma CSCs by inducing oxidative and metabolic stress, underscoring potential therapeutic advancements in glioblastoma treatment.
Collapse
Affiliation(s)
- Apurva Jaiswal
- Plasma Bioscience Research Center/Department of Electrical and Biological Physics, Kwangwoon University, Seoul, 01897, South Korea
| | - Neha Kaushik
- Department of Biotechnology, College of Engineering, The University of Suwon, Hwaseong, 18323, South Korea
| | - Paritosh Patel
- Plasma Bioscience Research Center/Department of Electrical and Biological Physics, Kwangwoon University, Seoul, 01897, South Korea
| | - Tirtha Raj Acharya
- Plasma Bioscience Research Center/Department of Electrical and Biological Physics, Kwangwoon University, Seoul, 01897, South Korea
| | - Subhadip Mukherjee
- Plasma Bioscience Research Center/Department of Electrical and Biological Physics, Kwangwoon University, Seoul, 01897, South Korea
| | - Eun Ha Choi
- Plasma Bioscience Research Center/Department of Electrical and Biological Physics, Kwangwoon University, Seoul, 01897, South Korea.
| | - Nagendra Kumar Kaushik
- Plasma Bioscience Research Center/Department of Electrical and Biological Physics, Kwangwoon University, Seoul, 01897, South Korea.
| |
Collapse
|
4
|
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
|
5
|
Koltai T, Fliegel L. Dichloroacetate for Cancer Treatment: Some Facts and Many Doubts. Pharmaceuticals (Basel) 2024; 17:744. [PMID: 38931411 PMCID: PMC11206832 DOI: 10.3390/ph17060744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/23/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Rarely has a chemical elicited as much controversy as dichloroacetate (DCA). DCA was initially considered a dangerous toxic industrial waste product, then a potential treatment for lactic acidosis. However, the main controversies started in 2008 when DCA was found to have anti-cancer effects on experimental animals. These publications showed contradictory results in vivo and in vitro such that a thorough consideration of this compound's in cancer is merited. Despite 50 years of experimentation, DCA's future in therapeutics is uncertain. Without adequate clinical trials and health authorities' approval, DCA has been introduced in off-label cancer treatments in alternative medicine clinics in Canada, Germany, and other European countries. The lack of well-planned clinical trials and its use by people without medical training has discouraged consideration by the scientific community. There are few thorough clinical studies of DCA, and many publications are individual case reports. Case reports of DCA's benefits against cancer have been increasing recently. Furthermore, it has been shown that DCA synergizes with conventional treatments and other repurposable drugs. Beyond the classic DCA target, pyruvate dehydrogenase kinase, new target molecules have also been recently discovered. These findings have renewed interest in DCA. This paper explores whether existing evidence justifies further research on DCA for cancer treatment and it explores the role DCA may play in it.
Collapse
Affiliation(s)
- Tomas Koltai
- Hospital del Centro Gallego de Buenos Aires, Buenos Aires 2199, Argentina
| | - Larry Fliegel
- Department of Biochemistry, University Alberta, Edmonton, AB T6G 2H7, Canada;
| |
Collapse
|
6
|
Upadhyay S, Khan S, Hassan MI. Exploring the diverse role of pyruvate kinase M2 in cancer: Navigating beyond glycolysis and the Warburg effect. Biochim Biophys Acta Rev Cancer 2024; 1879:189089. [PMID: 38458358 DOI: 10.1016/j.bbcan.2024.189089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/25/2024] [Accepted: 03/03/2024] [Indexed: 03/10/2024]
Abstract
Pyruvate Kinase M2, a key enzyme in glycolysis, has garnered significant attention in cancer research due to its pivotal role in the metabolic reprogramming of cancer cells. Originally identified for its association with the Warburg effect, PKM2 has emerged as a multifaceted player in cancer biology. The functioning of PKM2 is intricately regulated at multiple levels, including controlling the gene expression via various transcription factors and non-coding RNAs, as well as adding post-translational modifications that confer distinct functions to the protein. Here, we explore the diverse functions of PKM2, encompassing newly emerging roles in non-glycolytic metabolic regulation, immunomodulation, inflammation, DNA repair and mRNA processing, beyond its canonical role in glycolysis. The ever-expanding list of its functions has recently grown to include roles in subcellular compartments such as the mitochondria and extracellular milieu as well, all of which make PKM2 an attractive drug target in the pursuit of therapeutics for cancer.
Collapse
Affiliation(s)
- Saurabh Upadhyay
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Shumayila Khan
- International Health Division, Indian Council of Medical Research, Ansari Nagar, New Delhi 110029, India
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India.
| |
Collapse
|
7
|
Rihan M, Sharma SS. Cardioprotective potential of compound 3K, a selective PKM2 inhibitor in isoproterenol-induced acute myocardial infarction: A mechanistic study. Toxicol Appl Pharmacol 2024; 485:116905. [PMID: 38521371 DOI: 10.1016/j.taap.2024.116905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/20/2024] [Accepted: 03/15/2024] [Indexed: 03/25/2024]
Abstract
Myocardial infarction (MI) or heart attack arises from acute or chronic prolonged ischemic conditions in the myocardium. Although several risk factors are associated with MI pathophysiology, one of the risk factors is an imbalance in the oxygen supply. The current available MI therapies are still inadequate due to the complexity of MI pathophysiology. Pyruvate kinase M2 (PKM2) has been implicated in numerous CVDs pathologies. However, the effect of specific pharmacological intervention targeting PKM2 has not been studied in MI. Therefore, in this study, we explored the effect of compound 3K, a PKM2-specific inhibitor, in isoproterenol-induced acute MI model. In this study, in order to induce MI in rats, isoproterenol (ISO) was administered at a dose of 100 mg/kg over two days at an interval of 24 h. Specific PKM2 inhibitor, compound 3K (2 and 4 mg/kg), was administered in MI rats to investigate its cardioprotective potential. After the last administration of compound 3K, ECG and hemodynamic parameters were recorded using a PV-loop system. Cardiac histology, western blotting, and plasmatic cardiac damage markers were evaluated to elucidate the underlying mechanisms. Treatment of compound 3K significantly reduced ISO-induced alterations in ECG, ventricular functions, cardiac damage, infarct size, and cardiac fibrosis. Compound 3K treatment produced significant increase in PKM1 expression and decrease in PKM2 expression. In addition, HIF-1α, caspase-3, c-Myc, and PTBP1 expression were also reduced after compound 3K treatment. This study demonstrates the cardioprotective potential of compound 3K in MI, and its mechanisms of cardioprotective action.
Collapse
Affiliation(s)
- Mohd Rihan
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S Nagar, Mohali 160062, Punjab, India
| | - Shyam Sunder Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S Nagar, Mohali 160062, Punjab, India.
| |
Collapse
|
8
|
Xu S, Wang L, Zhao Y, Mo T, Wang B, Lin J, Yang H. Metabolism-regulating non-coding RNAs in breast cancer: roles, mechanisms and clinical applications. J Biomed Sci 2024; 31:25. [PMID: 38408962 PMCID: PMC10895768 DOI: 10.1186/s12929-024-01013-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 01/17/2024] [Indexed: 02/28/2024] Open
Abstract
Breast cancer is one of the most common malignancies that pose a serious threat to women's health. Reprogramming of energy metabolism is a major feature of the malignant transformation of breast cancer. Compared to normal cells, tumor cells reprogram metabolic processes more efficiently, converting nutrient supplies into glucose, amino acid and lipid required for malignant proliferation and progression. Non-coding RNAs(ncRNAs) are a class of functional RNA molecules that are not translated into proteins but regulate the expression of target genes. NcRNAs have been demonstrated to be involved in various aspects of energy metabolism, including glycolysis, glutaminolysis, and fatty acid synthesis. This review focuses on the metabolic regulatory mechanisms and clinical applications of metabolism-regulating ncRNAs involved in breast cancer. We summarize the vital roles played by metabolism-regulating ncRNAs for endocrine therapy, targeted therapy, chemotherapy, immunotherapy, and radiotherapy resistance in breast cancer, as well as their potential as therapeutic targets and biomarkers. Difficulties and perspectives of current targeted metabolism and non-coding RNA therapeutic strategies are discussed.
Collapse
Affiliation(s)
- Shiliang Xu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, People's Republic of China
| | - Lingxia Wang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, People's Republic of China
| | - Yuexin Zhao
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, People's Republic of China
| | - Tong Mo
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, People's Republic of China
| | - Bo Wang
- Department of Oncology, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu, 215004, People's Republic of China
| | - Jun Lin
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, People's Republic of China.
| | - Huan Yang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, People's Republic of China.
| |
Collapse
|
9
|
Huang PC, Chang CW, Lin YC, Chen CY, Chen TY, Chuang LT, Liu CJ, Huang CL, Li WC. Pyruvate Kinase Differentially Alters Metabolic Signatures during Head and Neck Carcinogenesis. Int J Mol Sci 2023; 24:16639. [PMID: 38068962 PMCID: PMC10706023 DOI: 10.3390/ijms242316639] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/13/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
During glycolysis, the muscle isoform of pyruvate kinase PKM2 produces ATP in exchange for dephosphorylation of phosphoenolpyruvate (PEP) into pyruvate. PKM2 has been considered as a tumor-promoting factor in most cancers, whereas the regulatory role of PKM2 during head and neck carcinogenesis remained to be delineated. PKM2 mRNA and protein expression was examined in head and neck tumorous specimens. The role of PKM2 in controlling cellular malignancy was determined in shRNA-mediated PKM2-deficient head and neck squamous cell carcinoma (HNSC) cells. In agreement with the results in other cancers, PKM2 expression is enriched in both mouse and human HNSC tissues. Nevertheless, PKM2 mRNA expression reversely correlated with tumor stage, and greater recurrence-free survival rates are evident in the PKM2high HNSC population, arguing that PKM2 may be tumor-suppressive. Multifaceted analyses showed a greater in vivo xenografic tumor growth and an enhanced cisplatin resistance in response to PKM2 loss, whereas PKM2 silencing led to reduced cell motility. At the molecular level, metabolic shifts towards mitochondrial metabolism and activation of oncogenic Protein kinase B (PKB/Akt) and extracellular signal-regulated kinase (ERK) signals were detected in PKM2-silencing HNSC cells. In sum, our findings demonstrated that PKM2 differentially modulated head and neck tumorigenicity via metabolic reprogramming.
Collapse
Affiliation(s)
- Pei-Chun Huang
- Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; (P.-C.H.); (C.-Y.C.); (T.-Y.C.)
| | - Ching-Wen Chang
- Graduate Institute of Metabolism and Obesity Sciences (GIMOS), College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan;
- Taipei Cancer Center, Taipei Medical University, Taipei 11031, Taiwan
| | - Yu-Cheng Lin
- Department of Dentistry, College of Dentistry, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; (Y.-C.L.); (C.-J.L.)
- Oral Medicine Innovation Center (OMIC), National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Chang-Yi Chen
- Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; (P.-C.H.); (C.-Y.C.); (T.-Y.C.)
| | - Tsai-Ying Chen
- Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; (P.-C.H.); (C.-Y.C.); (T.-Y.C.)
| | - Lu-Te Chuang
- Department of Biotechnology and Pharmaceutical Technology, Yuanpei University of Medical Technology, Hsinchu 30015, Taiwan;
| | - Chung-Ji Liu
- Department of Dentistry, College of Dentistry, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; (Y.-C.L.); (C.-J.L.)
- Department of Oral and Maxillofacial Surgery, MacKay Memorial Hospital, Taipei 10449, Taiwan
- Department of Medical Research, MacKay Memorial Hospital, Taipei 10449, Taiwan
| | - Chien-Ling Huang
- Department of Health Technology and Informatics (HTI), The Hong Kong Polytechnic University (PolyU), Hung Hom, Kowloon, Hong Kong SAR, China;
| | - Wan-Chun Li
- Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; (P.-C.H.); (C.-Y.C.); (T.-Y.C.)
- Department of Dentistry, College of Dentistry, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; (Y.-C.L.); (C.-J.L.)
- Oral Medicine Innovation Center (OMIC), National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| |
Collapse
|
10
|
Doualle C, Gouju J, Nouari Y, Wery M, Guittonneau C, Codron P, Rousseau A, Saulnier P, Eyer J, Letournel F. Dedifferentiated cells obtained from glioblastoma cell lines are an easy and robust model for mesenchymal glioblastoma stem cells studies. Am J Cancer Res 2023; 13:1425-1442. [PMID: 37168329 PMCID: PMC10164819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 02/12/2023] [Indexed: 05/13/2023] Open
Abstract
Glioblastoma is an aggressive brain tumor with a poor prognosis. Glioblastoma Stem Cells (GSC) are involved in glioblastoma resistance and relapse. Effective glioblastoma treatment must include GSC targeting strategy. Robust and well defined in vitroGSC models are required for new therapies evaluation. In this study, we extensively characterized 4 GSC models obtained by dedifferentiation of commercially available glioblastoma cell lines and compared them to 2 established patient derived GSC lines (Brain Tumor Initiating Cells). Dedifferentiated cells formed gliospheres, typical for GSC, with self-renewal ability. Gene expression and protein analysis revealed an increased expression of several stemness associated markers such as A2B5, integrin α6, Nestin, SOX2 and NANOG. Cells were oriented toward a mesenchymal GSC phenotype as shown by elevated levels of mesenchymal and EMT related markers (CD44, FN1, integrin α5). Dedifferentiated GSC were similar to BTIC in terms of size and heterogeneity. The characterization study also revealed that CXCR4 pathway was activated by dedifferentiation, emphasizing its role as a potential therapeutic target. The expression of resistance-associated markers and the phenotypic diversity of the 4 GSC models obtained by dedifferentiation make them relevant to challenge future GSC targeting therapies.
Collapse
Affiliation(s)
- Cécile Doualle
- Univ Angers, CHU Angers, Inserm, CNRS, MINT, SFR ICATF-49000 Angers, France
| | - Julien Gouju
- Univ Angers, CHU Angers, Inserm, CNRS, MINT, SFR ICATF-49000 Angers, France
- Département de Pathologie, CHU AngersF-49000 Angers, France
| | - Yousra Nouari
- Univ Angers, CHU Angers, Inserm, CNRS, MINT, SFR ICATF-49000 Angers, France
| | | | - Clélia Guittonneau
- Univ Angers, CHU Angers, Inserm, CNRS, MINT, SFR ICATF-49000 Angers, France
| | - Philippe Codron
- Département de Pathologie, CHU AngersF-49000 Angers, France
- Univ Angers, CHU Angers, Inserm, CNRS, MITOVASC, SFR ICATF-49000 Angers, France
| | - Audrey Rousseau
- Département de Pathologie, CHU AngersF-49000 Angers, France
- Univ Angers, Nantes Université, CHU Angers, Inserm, CNRS, CRCI2NA, SFR ICATF-49000 Angers, France
| | - Patrick Saulnier
- Univ Angers, CHU Angers, Inserm, CNRS, MINT, SFR ICATF-49000 Angers, France
| | - Joël Eyer
- Univ Angers, CHU Angers, Inserm, CNRS, MINT, SFR ICATF-49000 Angers, France
| | - Franck Letournel
- Univ Angers, CHU Angers, Inserm, CNRS, MINT, SFR ICATF-49000 Angers, France
- Département de Pathologie, CHU AngersF-49000 Angers, France
| |
Collapse
|
11
|
Bikfalvi A, da Costa CA, Avril T, Barnier JV, Bauchet L, Brisson L, Cartron PF, Castel H, Chevet E, Chneiweiss H, Clavreul A, Constantin B, Coronas V, Daubon T, Dontenwill M, Ducray F, Enz-Werle N, Figarella-Branger D, Fournier I, Frenel JS, Gabut M, Galli T, Gavard J, Huberfeld G, Hugnot JP, Idbaih A, Junier MP, Mathivet T, Menei P, Meyronet D, Mirjolet C, Morin F, Mosser J, Moyal ECJ, Rousseau V, Salzet M, Sanson M, Seano G, Tabouret E, Tchoghandjian A, Turchi L, Vallette FM, Vats S, Verreault M, Virolle T. Challenges in glioblastoma research: focus on the tumor microenvironment. Trends Cancer 2023; 9:9-27. [PMID: 36400694 DOI: 10.1016/j.trecan.2022.09.005] [Citation(s) in RCA: 129] [Impact Index Per Article: 64.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 09/20/2022] [Accepted: 09/30/2022] [Indexed: 11/17/2022]
Abstract
Glioblastoma (GBM) is the most deadly type of malignant brain tumor, despite extensive molecular analyses of GBM cells. In recent years, the tumor microenvironment (TME) has been recognized as an important player and therapeutic target in GBM. However, there is a need for a full and integrated understanding of the different cellular and molecular components involved in the GBM TME and their interactions for the development of more efficient therapies. In this review, we provide a comprehensive report of the GBM TME, which assembles the contributions of physicians and translational researchers working on brain tumor pathology and therapy in France. We propose a holistic view of the subject by delineating the specific features of the GBM TME at the cellular, molecular, and therapeutic levels.
Collapse
Affiliation(s)
- Andreas Bikfalvi
- Bordeaux University, INSERM, U1312 BRIC, Tumor and Vascular Biology Laboratory, F-33600, Pessac, France.
| | - Cristine Alves da Costa
- Côte d'Azur University, INSERM, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Team "Laboratory of Excellence (LABEX) Distalz", F-06560 Nice, France
| | - Tony Avril
- Rennes University, Inserm U1242, Centre de Lutte contre le Cancer Eugène Marquis, F- 35000 Rennes, France
| | - Jean-Vianney Barnier
- Institute of Neuroscience Paris-Saclay, UMR9197, CNRS, Univ. Paris-Saclay, F-91191 Gif-sur-Yvette, France
| | - Luc Bauchet
- Montpellier University Medical Center, Department of Neurosurgery, INSERM U1191, F-34090 Montpellier, France
| | - Lucie Brisson
- Bordeaux University, INSERM, U1312 BRIC, Tumor and Vascular Biology Laboratory, F-33600, Pessac, France
| | | | - Hélène Castel
- Normandie University, INSERM U1239, DC2N, Institute for Research and Innovation in Biomedicine (IRIB), F-76000 Rouen, France
| | - Eric Chevet
- Rennes University, Inserm U1242, Centre de Lutte contre le Cancer Eugène Marquis, F- 35000 Rennes, France
| | - Hervé Chneiweiss
- Sorbonne University, CNRS UMR8246, Inserm U1130, IBPS-Neuroscience Paris Seine, F- 75005 Paris, France
| | - Anne Clavreul
- Angers University, CHU d'Angers, CRCINA, F-49000 Angers, France
| | - Bruno Constantin
- Poitiers University, CNRS UMR 6041, Laboratory Channels & Connexins in Cancers and Cell Stemness, F-86000 Poitiers, France
| | - Valérie Coronas
- Poitiers University, CNRS UMR 6041, Laboratory Channels & Connexins in Cancers and Cell Stemness, F-86000 Poitiers, France
| | - Thomas Daubon
- Bordeaux University, CNRS, IBGC, UMR 5095, F-33 077 Bordeaux, France
| | - Monique Dontenwill
- Strasbourg University, Laboratoire de Bioimagerie et Pathologie, UMR7021 CNRS, F-67401 Illkirch-Graffenstaden, France
| | - Francois Ducray
- Lyon I University, Cancer Research Centre of Lyon (CRCL) INSERM 1052&CNRS UMR5286, Centre Léon Bérard, Lyon 69008, France., F-69622 Villeurbanne, France
| | - Natacha Enz-Werle
- Strasbourg University, Laboratoire de Bioimagerie et Pathologie, UMR7021 CNRS, F-67401 Illkirch-Graffenstaden, France
| | - Dominique Figarella-Branger
- Aix-Marseille University, Service d'Anatomie Pathologique et de Neuropathologie, Hôpital de la Timone, F-13385 Marseille, France
| | - Isabelle Fournier
- Lille University, Inserm, CHU Lille, U1192, Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), F-59000 Lille, France
| | - Jean-Sébastien Frenel
- Normandie University, INSERM U1239, DC2N, Institute for Research and Innovation in Biomedicine (IRIB), F-76000 Rouen, France
| | - Mathieu Gabut
- Lyon I University, Cancer Research Centre of Lyon (CRCL) INSERM 1052&CNRS UMR5286, Centre Léon Bérard, Lyon 69008, France., F-69622 Villeurbanne, France
| | - Thierry Galli
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Membrane Traffic in Healthy & Diseased Brain, GHU PARIS Psychiatrie & Neurosciences, F-75014 Paris, France
| | - Julie Gavard
- CRCI2NA, INSERM U1307, CNRS UMR6075, Nantes Universite, 44007 Nantes, France
| | - Gilles Huberfeld
- College de France, Center for Interdisciplinary Research in Biology (CIRB), CNRS, INSERM, Université PSL, Paris 75005, France
| | - Jean-Philippe Hugnot
- Montpellier University, Institut de Génomique Fonctionnelle, CNRS, INSERM, F-34094 Montpellier, France
| | - Ahmed Idbaih
- Sorbonne University, AP-HP, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, F-75013, Paris, France
| | - Marie-Pierre Junier
- Sorbonne University, CNRS UMR8246, Inserm U1130, IBPS-Neuroscience Paris Seine, F- 75005 Paris, France
| | - Thomas Mathivet
- Bordeaux University, INSERM, U1312 BRIC, Tumor and Vascular Biology Laboratory, F-33600, Pessac, France
| | - Philippe Menei
- Angers University, CHU d'Angers, CRCINA, F-49000 Angers, France
| | - David Meyronet
- Institute of Neuropathology, Hospices Civils de Lyon, F-69008, Lyon, France
| | - Céline Mirjolet
- Centre Georges-François Leclerc, UNICANCER, Dijon, France. Inserm U1231, Equipe Cadir, F-21000 Dijon, France
| | - Fabrice Morin
- Normandie University, INSERM U1239, DC2N, Institute for Research and Innovation in Biomedicine (IRIB), F-76000 Rouen, France
| | - Jean Mosser
- Rennes University, Inserm U1242, Centre de Lutte contre le Cancer Eugène Marquis, F- 35000 Rennes, France
| | - Elisabeth Cohen-Jonathan Moyal
- Institut Claudius Regaud, NSERM 1037, CRCT Team RADOPT, Département de Radiothérapie, IUCT-Oncopole, F-31100 Toulouse, France
| | - Véronique Rousseau
- Institute of Neuroscience Paris-Saclay, UMR9197, CNRS, Univ. Paris-Saclay, F-91191 Gif-sur-Yvette, France
| | - Michel Salzet
- Lille University, Inserm, CHU Lille, U1192, Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), F-59000 Lille, France
| | - Marc Sanson
- Sorbonne University, AP-HP, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, F-75013, Paris, France
| | - Giorgio Seano
- Curie Institute Research Center, Tumor Microenvironment Laboratory, PSL Research University, Inserm U1021, CNRS UMR3347, F-91898 Orsay, France
| | - Emeline Tabouret
- Aix-Marseille University, CNRS, INP, Inst Neurophysiopathol, F-13005 Marseille, France
| | - Aurélie Tchoghandjian
- Aix-Marseille University, CNRS, INP, Inst Neurophysiopathol, F-13005 Marseille, France
| | - Laurent Turchi
- Côte D'Azur University, CNRS, INSERM, Institut de Biologie Valrose, Team INSERM "Cancer Stem Cell Plasticity and Functional Intra-tumor Heterogeneity", F-06108 Nice, France
| | - Francois M Vallette
- CRCI2NA, INSERM U1307, CNRS UMR6075, Nantes Universite, 44007 Nantes, France
| | - Somya Vats
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Membrane Traffic in Healthy & Diseased Brain, GHU PARIS Psychiatrie & Neurosciences, F-75014 Paris, France
| | - Maité Verreault
- Sorbonne University, AP-HP, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, F-75013, Paris, France
| | - Thierry Virolle
- Côte D'Azur University, CNRS, INSERM, Institut de Biologie Valrose, Team INSERM "Cancer Stem Cell Plasticity and Functional Intra-tumor Heterogeneity", F-06108 Nice, France
| |
Collapse
|
12
|
Circular RNA KIF4A Promotes Liver Metastasis of Breast Cancer by Reprogramming Glucose Metabolism. JOURNAL OF ONCOLOGY 2022; 2022:8035083. [PMID: 36052282 PMCID: PMC9427241 DOI: 10.1155/2022/8035083] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 07/08/2022] [Indexed: 11/18/2022]
Abstract
Background Circular RNAs (circRNAs) regulate complex functional processes and play crucial roles in cancer development and progression. It was reported that circKIF4 regulates the progression of triple-negative breast cancer (TNBC). This study evaluates the role of circKIF4 in breast cancer distant metastasis and metabolic reprogramming. Methods RT-qPCR was performed to verify the expression of circKIF4A in breast cancer, liver metastatic tissues, and cell lines. The function of circKIF4A in metastasis was evaluated both in vitro and in vivo through a series of experiments, including cell migration and glucose intake experiments. Additionally, we conducted molecular experiments to clarify the regulatory role of circKIF4A. We then conducted a Luciferase reporter assay and an RNA immunoprecipitation assay to identify the molecular interactions between circKIF4A and miRNA. Results circKIF4A was overexpressed in breast cancer cell lines and tissues, inhibiting its expression and suppressing breast cancer growth and metastasis. Interestingly, we observed that circKIF4A reprogrammed the glucose metabolism of breast cancer, and silencing circKIF4A greatly affected glucose uptake and lactate production in breast cancer cells. miR-335 can be sponged by circKIF4A, which affected the expression of ALDOA/OCT4 protein and regulated HK2/PKM2 expression. Conclusions This study demonstrated that the circKIF4A-miR-335-OCT4/ALDOA-HK2/PKM2 axis is critical to breast cancer metabolic reprogramming, indicating that this axis could be a novel therapeutic target for the treatment of liver metastasis of breast cancer.
Collapse
|
13
|
El-Far AH, Al Jaouni SK, Li X, Fu J. Cancer metabolism control by natural products: Pyruvate kinase M2 targeting therapeutics. Phytother Res 2022; 36:3181-3201. [PMID: 35794729 DOI: 10.1002/ptr.7534] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 05/19/2022] [Accepted: 06/12/2022] [Indexed: 12/13/2022]
Abstract
Glycolysis is the primary source of energy for cancer growth and metastasis. The shift in metabolism from mitochondrial oxidative phosphorylation to aerobic glycolysis is called the Warburg effect. Cancer progression due to aerobic glycolysis is often associated with the activation of oncogenes or the loss of tumor suppressors. Therefore, inhibition of glycolysis is one of the effective strategies in cancer control. Pyruvate kinase M2 (PKM2) is a key glycolytic enzyme overexpressed in breast, prostate, lung, colorectal, and liver cancers. Here, we discuss published studies regarding PKM2 inhibitors from natural products that are promising drug candidates for cancer therapy. We have highlighted the potential of natural PKM2 inhibitors for various cancer types. Moreover, we encourage researchers to evaluate the combinational effects between natural and synthetic PKM2 inhibitors. Also, further high-quality studies are needed to firmly establish the clinical efficacy of natural products.
Collapse
Affiliation(s)
- Ali H El-Far
- Department of Biochemistry, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| | - Soad K Al Jaouni
- Department of Hematology/Pediatric Oncology, Yousef Abdulatif Jameel Scientific Chair of Prophetic Medicine Application, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Xiaotao Li
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, China.,School of Arts and Sciences, New York University-Shanghai, Shanghai, China.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Junjiang Fu
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, China
| |
Collapse
|
14
|
Hossain AJ, Islam R, Kim JG, Dogsom O, Cap KC, Park JB. Pyruvate Dehydrogenase A1 Phosphorylated by Insulin Associates with Pyruvate Kinase M2 and Induces LINC00273 through Histone Acetylation. Biomedicines 2022; 10:biomedicines10061256. [PMID: 35740278 PMCID: PMC9220252 DOI: 10.3390/biomedicines10061256] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/14/2022] [Accepted: 05/25/2022] [Indexed: 02/07/2023] Open
Abstract
Insulin potently promotes cell proliferation and anabolic metabolism along with a reduction in blood glucose levels. Pyruvate dehydrogenase (PDH) plays a pivotal role in glucose metabolism. Insulin increase PDH activity by attenuating phosphorylated Ser293 PDH E1α (p-PDHA1) in normal liver tissue. In contrast to normal hepatocytes, insulin enhanced p-PDHA1 level and induced proliferation of hepatocellular carcinoma HepG2 cells. Here, we attempted to find a novel function of p-PDHA1 in tumorigenesis upon insulin stimulation. We found that p-Ser293 E1α, but not the E2 or E3 subunit of pyruvate dehydrogenase complex (PDC), co-immunoprecipitated with pyruvate kinase M2 (PKM2) upon insulin. Of note, the p-PDHA1 along with PKM2 translocated to the nucleus. The p-PDHA1/PKM2 complex was associated with the promoter of long intergenic non-protein coding (LINC) 00273 gene (LINC00273) and recruited p300 histone acetyl transferase (HAT) and ATP citrate lyase (ACL), leading to histone acetylation. Consequently, the level of transcription factor ZEB1, an epithelial–mesenchymal transition (EMT) marker, was promoted through increased levels of LINC00273, resulting in cell migration upon insulin. p-PDHA1, along with PKM2, may be crucial for transcriptional regulation of specific genes through epigenetic regulation upon insulin in hepatocarcinoma cells.
Collapse
Affiliation(s)
- Abu Jubayer Hossain
- Department of Biochemistry, Hallym University College of Medicine, Chuncheon 24252, Kangwon-do, Korea; (A.J.H.); (R.I.); (J.-G.K.); (O.D.); (K.C.C.)
- Institute of Cell Differentiation and Aging, Hallym University College of Medicine, Chuncheon 24252, Kangwon-do, Korea
| | - Rokibul Islam
- Department of Biochemistry, Hallym University College of Medicine, Chuncheon 24252, Kangwon-do, Korea; (A.J.H.); (R.I.); (J.-G.K.); (O.D.); (K.C.C.)
- Institute of Cell Differentiation and Aging, Hallym University College of Medicine, Chuncheon 24252, Kangwon-do, Korea
- Department of Biotechnology and Genetic Engineering, Faculty of Biological Science, Islamic University, Kushtia 7003, Bangladesh
| | - Jae-Gyu Kim
- Department of Biochemistry, Hallym University College of Medicine, Chuncheon 24252, Kangwon-do, Korea; (A.J.H.); (R.I.); (J.-G.K.); (O.D.); (K.C.C.)
- Institute of Cell Differentiation and Aging, Hallym University College of Medicine, Chuncheon 24252, Kangwon-do, Korea
| | - Oyungerel Dogsom
- Department of Biochemistry, Hallym University College of Medicine, Chuncheon 24252, Kangwon-do, Korea; (A.J.H.); (R.I.); (J.-G.K.); (O.D.); (K.C.C.)
- Department of Biology, School of Bio-Medicine, Mongolian National University of Medical Sciences, Ulaanbaatar 14210, Mongolia
| | - Kim Cuong Cap
- Department of Biochemistry, Hallym University College of Medicine, Chuncheon 24252, Kangwon-do, Korea; (A.J.H.); (R.I.); (J.-G.K.); (O.D.); (K.C.C.)
- Institute of Cell Differentiation and Aging, Hallym University College of Medicine, Chuncheon 24252, Kangwon-do, Korea
- Institute of Research and Development, Duy Tan University, Danang 550000, Vietnam
| | - Jae-Bong Park
- Department of Biochemistry, Hallym University College of Medicine, Chuncheon 24252, Kangwon-do, Korea; (A.J.H.); (R.I.); (J.-G.K.); (O.D.); (K.C.C.)
- Institute of Cell Differentiation and Aging, Hallym University College of Medicine, Chuncheon 24252, Kangwon-do, Korea
- Correspondence: ; Tel.: +82-33-248-2542; Fax: +82-33-244-8425
| |
Collapse
|
15
|
Park JH, Lee JS, Oh Y, Lee JS, Park HE, Lee H, Park YS, Kyung SY, Kim HS, Yoon S. PKM2 Is Overexpressed in Glioma Tissues, and Its Inhibition Highly Increases Late Apoptosis in U87MG Cells With Low-density Specificity. In Vivo 2022; 36:694-703. [PMID: 35241524 PMCID: PMC8931915 DOI: 10.21873/invivo.12755] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 12/08/2021] [Accepted: 12/10/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND/AIM Pyruvate kinase M2 (PKM2) functions as an important rate-limiting enzyme in aerobic glycolysis and is involved in tumor initiation and progression. However, there are few studies on the correlation between PKM2 expression and its role in glioma. MATERIALS AND METHODS PKM2 expression was immunohistochemically examined in human brain tumor samples. Furthermore, we studied the effects of two PKM2 inhibitors (shikonin and compound 3K) on the U87MG glioma cell line. RESULTS PKM2 was overexpressed in most glioma tissues when compared to controls. Interestingly, glioma-adjacent tissues from showed slight PKM2 overexpression. This suggests that PKM2 overexpression maybe an important trigger factor for glioma tumorigenesis. We found that the PKM2 inhibitor shikonin was effective against U87MG cells at a relatively low dose and was largely dependent on low cellular density compared to the effects of the anticancer drug vincristine. Shikonin highly increased late-apoptosis of U87MG cells. We also demonstrated that autophagy was involved in the increase in late-apoptosis levels caused by shikonin. Although vincristine treatment led to a high level of G2-phase arrest in U87MG cells, shikonin did not increase G2 arrest. Co-treatment with two PKM2 inhibitors, shikonin and compound 3K, increased the inhibitory effects. CONCLUSION Combination therapy with PKM2 inhibitors together might be more effective than combination therapy with anticancer drugs. Our findings encourage the application of PKM2-targeting in gliomas, and lay the foundation for the development of PKM2 inhibitors as promising antitumor agents for glioma.
Collapse
Affiliation(s)
- Jae Hyeon Park
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Jin-Sol Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Yunmoon Oh
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Ji Sun Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Hae Eun Park
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Haeun Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Yeon Su Park
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - So Young Kyung
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Hyung Sik Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Sungpil Yoon
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| |
Collapse
|
16
|
Li Y, Gao X. LINC00883 Promotes Drug Resistance of Glioma Through a microRNA-136/NEK1-Dependent Mechanism. Front Oncol 2022; 11:692265. [PMID: 35083134 PMCID: PMC8785904 DOI: 10.3389/fonc.2021.692265] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 11/08/2021] [Indexed: 12/22/2022] Open
Abstract
Objective Accumulating evidence has highlighted the roles of long noncoding RNAs (lncRNAs) as competing endogenous RNAs (ceRNAs) of microRNAs (miRNAs) through their binding sites in the progression of glioma. Hereby, we aim to explore the role of LINC00883 as a regulator of miR-136 and its target, NIMA-related kinase 1 (NEK1), thus, its involvement in the drug resistance of glioma cells. Methods and Results Mechanistic investigations by dual-luciferase reporter, RNA pull-down, and RNA-binding protein immunoprecipitation (RIP) assays indicated that LINC00883 bound to miR-136, thereby blocking miR-136-induced downregulation of NEK1. Through gain-of-function experiments in U251 cells that presented a high drug resistance, we found that ectopic expression of LINC00883 resulted in increased MRP (encoding multidrug resistance-associated protein), limited cell apoptosis, and increased proliferation. Expectedly, depleting LINC00883 yielded tumor-suppressive and anti-chemoresistance effects on U251 cells by increasing miR-136 and inhibiting NEK1. Next, drug-resistant glioma cell line SOWZ1, drug-sensitive glioma cell line SOWZ2, and drug-resistant glioma cell line SOWZ2-BCNU (SOWZ2 cultured in BCNU) were applied to validate the roles of LINC00883 in the regulation of multidrug resistance. LINC00883 knockdown suppressed the viability of SWOZ1, SWOZ2, and SWOZ2-BCNU cells. Conclusion In conclusion, LINC00883 knockdown reduces drug resistance in glioma. Hence, our study provides a future strategy to prevent drug resistance-induced therapeutic failure in glioma.
Collapse
Affiliation(s)
- Yongzhe Li
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xin Gao
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
17
|
Ma WK, Voss DM, Scharner J, Costa ASH, Lin KT, Jeon HY, Wilkinson JE, Jackson M, Rigo F, Bennett CF, Krainer AR. ASO-based PKM splice-switching therapy inhibits hepatocellular carcinoma growth. Cancer Res 2021; 82:900-915. [PMID: 34921016 DOI: 10.1158/0008-5472.can-20-0948] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 10/22/2021] [Accepted: 12/14/2021] [Indexed: 11/16/2022]
Abstract
The M2 pyruvate kinase (PKM2) isoform is upregulated in most cancers and plays a crucial role in regulation of the Warburg effect, which is characterized by the preference for aerobic glycolysis over oxidative phosphorylation for energy metabolism. PKM2 is an alternative-splice isoform of the PKM gene and is a potential therapeutic target. Antisense oligonucleotides (ASO) that switch PKM splicing from the cancer-associated PKM2 to the PKM1 isoform have been shown to induce apoptosis in cultured glioblastoma cells when delivered by lipofection. Here, we explore the potential of ASO-based PKM splice switching as a targeted therapy for liver cancer. A more potent lead cEt/DNA ASO induced PKM splice-switching and inhibited the growth of cultured hepatocellular carcinoma (HCC) cells. This PKM isoform switch increased pyruvate-kinase activity and altered glucose metabolism. In an orthotopic HCC xenograft mouse model, the lead ASO and a second ASO targeting a non-overlapping site inhibited tumor growth. Finally, in a genetic HCC mouse model, a surrogate mouse-specific ASO induced Pkm splice switching and inhibited tumorigenesis, without observable toxicity. These results lay the groundwork for a potential ASO-based splicing therapy for HCC.
Collapse
Affiliation(s)
| | - Dillon M Voss
- Medical Scientist Training Program (MSTP), Stony Brook University School of Medicine
| | | | - Ana S H Costa
- Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai
| | | | | | - John E Wilkinson
- Unit for Laboratory Animal Medicine, University of Michigan–Ann Arbor
| | | | | | | | | |
Collapse
|
18
|
Liu J, Zhi Q, Liu Y, Wang Y, Chen L, Ke Y, Zeng L, Wu X, Yang X, Guleng B, Liu H, Ren J. Insulin promotes hepatocarcinoma tumorigenesis by up-regulating PKM2 expression. Exp Cell Res 2021; 408:112872. [PMID: 34648844 DOI: 10.1016/j.yexcr.2021.112872] [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: 09/04/2020] [Revised: 10/02/2021] [Accepted: 10/05/2021] [Indexed: 11/22/2022]
Abstract
Insulin, as a growth factor, can increase the risk of certain types of cancer. The present study showed that insulin promoted the proliferation of hepatocellular carcinoma cells in vitro and in vivo through pyruvate kinase M2 (PKM2), which is a rate-limiting enzyme in the process of glycolysis. Moreover, the expression of PKM2 was up-regulated by insulin at the posttranslational level in a nuclear orphan receptor TR3-dependent manner. In addition, insulin could enhance the interaction between PKM2 and TR3 and protect PKM2 from degradation. Our results identified a specific mechanism of insulin affecting cancer metabolism and thus promoting cancer progression, and they contribute to a better understanding of the observation that insulin is linked to an increased cancer risk under hyperinsulinemic conditions.
Collapse
Affiliation(s)
- Jingjing Liu
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, 201 Hubin South Road, Xiamen, 361004, Fujian Province, PR China; Xiamen Key Laboratory of Intestinal Microbiome and Human Health, Zhongshan Hospital Affiliated to Xiamen University, 201 Hubin South Road, Xiamen, 361004, Fujian Province, PR China.
| | - Qiang Zhi
- Faculty of Clinical Medicine, School of Medicine, Xiamen University, 168 University Road, Xiamen, 361005, Fujian Province, PR China
| | - Yunpeng Liu
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, 201 Hubin South Road, Xiamen, 361004, Fujian Province, PR China; Xiamen Key Laboratory of Intestinal Microbiome and Human Health, Zhongshan Hospital Affiliated to Xiamen University, 201 Hubin South Road, Xiamen, 361004, Fujian Province, PR China
| | - Ying Wang
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, 201 Hubin South Road, Xiamen, 361004, Fujian Province, PR China
| | - Linlin Chen
- Faculty of Clinical Medicine, School of Medicine, Xiamen University, 168 University Road, Xiamen, 361005, Fujian Province, PR China
| | - Yuhao Ke
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, 201 Hubin South Road, Xiamen, 361004, Fujian Province, PR China
| | - Lingsu Zeng
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, 201 Hubin South Road, Xiamen, 361004, Fujian Province, PR China
| | - Xiaoling Wu
- Faculty of Clinical Medicine, School of Medicine, Xiamen University, 168 University Road, Xiamen, 361005, Fujian Province, PR China
| | - Xiaoning Yang
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, 201 Hubin South Road, Xiamen, 361004, Fujian Province, PR China; Xiamen Key Laboratory of Intestinal Microbiome and Human Health, Zhongshan Hospital Affiliated to Xiamen University, 201 Hubin South Road, Xiamen, 361004, Fujian Province, PR China
| | - Bayasi Guleng
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, 201 Hubin South Road, Xiamen, 361004, Fujian Province, PR China; Xiamen Key Laboratory of Intestinal Microbiome and Human Health, Zhongshan Hospital Affiliated to Xiamen University, 201 Hubin South Road, Xiamen, 361004, Fujian Province, PR China; Faculty of Clinical Medicine, School of Medicine, Xiamen University, 168 University Road, Xiamen, 361005, Fujian Province, PR China; Faculty of Clinical Medicine & Institute of Mirobial Ecology, Medical College of Xiamen University, 168 University Road, Xiamen, 361005, Fujian Province, PR China; Department of Digestive Disease, School of Medicine, Xiamen University, 168 University Road, Xiamen, 361005, Fujian Province, PR China
| | - Hao Liu
- General Hospital of Ningxia Medical University, 804 Shengli Street, Yinchuan, 750004, Ningxia Hui Autonomous Region, PR China
| | - Jianlin Ren
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, 201 Hubin South Road, Xiamen, 361004, Fujian Province, PR China; Xiamen Key Laboratory of Intestinal Microbiome and Human Health, Zhongshan Hospital Affiliated to Xiamen University, 201 Hubin South Road, Xiamen, 361004, Fujian Province, PR China; Faculty of Clinical Medicine, School of Medicine, Xiamen University, 168 University Road, Xiamen, 361005, Fujian Province, PR China; Faculty of Clinical Medicine & Institute of Mirobial Ecology, Medical College of Xiamen University, 168 University Road, Xiamen, 361005, Fujian Province, PR China; Department of Digestive Disease, School of Medicine, Xiamen University, 168 University Road, Xiamen, 361005, Fujian Province, PR China.
| |
Collapse
|
19
|
Li L, Song D, Qi L, Jiang M, Wu Y, Gan J, Cao K, Li Y, Bai Y, Zheng T. Photodynamic therapy induces human esophageal carcinoma cell pyroptosis by targeting the PKM2/caspase-8/caspase-3/GSDME axis. Cancer Lett 2021; 520:143-159. [PMID: 34256094 DOI: 10.1016/j.canlet.2021.07.014] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 06/24/2021] [Accepted: 07/08/2021] [Indexed: 12/17/2022]
Abstract
Photodynamic therapy (PDT) uses a photosensitizer (PS) and visible light to induce cancer cell death. Pyroptosis is a new type of programmed cell death that is associated with the gasdermin protein family. However, the precise mechanism of pyroptosis in PDT-induced suppression of esophageal cancer remains unknown. We demonstrate that PDT can induce gasdermin E (GSDME)-mediated pyroptosis, which is characterized by the formation of pyroptotic blebs in esophageal squamous cell carcinoma (ESCC), which burst and release intracellular contents and pro-inflammatory mediators. Mechanistically, PDT may inhibit pyruvate kinase M2 (PKM2) and consequently, activate caspase-8 and caspase-3, which ultimately releases N-GSDME and triggers pyroptosis in ESCC. Moreover, PDT decreased the efficiency of pyroptosis in the presence of a glycolytic inhibitor. Overall, our results show that PDT induces pyroptosis in ESCC by targeting the PKM2/caspase-8/caspase-3/GSDME axis. This is the first in-depth study of the specific mechanism underlying PKM2-mediated pyroptosis under PDT in ESCC, and potentially has great implications for the clinical application of PDT in ESCC.
Collapse
Affiliation(s)
- Lisha Li
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, 150 Haping St, Nangang District, Harbin, Heilongjiang, 150081, PR China
| | - Dongfeng Song
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, 150 Haping St, Nangang District, Harbin, Heilongjiang, 150081, PR China
| | - Ling Qi
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, 150 Haping St, Nangang District, Harbin, Heilongjiang, 150081, PR China
| | - Mingxia Jiang
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, 150 Haping St, Nangang District, Harbin, Heilongjiang, 150081, PR China
| | - Yiming Wu
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, 150 Haping St, Nangang District, Harbin, Heilongjiang, 150081, PR China
| | - Junqing Gan
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, 150 Haping St, Nangang District, Harbin, Heilongjiang, 150081, PR China
| | - Kui Cao
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, 150 Haping St, Nangang District, Harbin, Heilongjiang, 150081, PR China
| | - Yanjing Li
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, 150 Haping St, Nangang District, Harbin, Heilongjiang, 150081, PR China.
| | - Yuxian Bai
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, 150 Haping St, Nangang District, Harbin, Heilongjiang, 150081, PR China.
| | - Tongsen Zheng
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, 150 Haping St, Nangang District, Harbin, Heilongjiang, 150081, PR China.
| |
Collapse
|
20
|
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: 25] [Impact Index Per Article: 6.3] [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
|
21
|
Dierolf JG, Watson AJ, Betts DH. Differential localization patterns of pyruvate kinase isoforms in murine naïve, formative, and primed pluripotent states. Exp Cell Res 2021; 405:112714. [PMID: 34181938 DOI: 10.1016/j.yexcr.2021.112714] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 05/17/2021] [Accepted: 06/22/2021] [Indexed: 12/28/2022]
Abstract
Mouse embryonic stem cells (mESCs) and mouse epiblast stem cells (mEpiSCs) represent opposite ends of the pluripotency continuum, referred to as naïve and primed pluripotent states, respectively. These divergent pluripotent states differ in several ways, including growth factor requirements, transcription factor expression, DNA methylation patterns, and metabolic profiles. Naïve cells employ both glycolysis and oxidative phosphorylation (OXPHOS), whereas primed cells preferentially utilize aerobic glycolysis, a trait shared with cancer cells referred to as the Warburg Effect. Until recently, metabolism has been regarded as a by-product of cell fate, however, evidence now supports metabolism as being a driver of stem cell state and fate decisions. Pyruvate kinase muscle isoforms (PKM1 and PKM2) are important for generating and maintaining pluripotent stem cells (PSCs) and mediating the Warburg Effect. Both isoforms catalyze the final, rate limiting step of glycolysis, generating adenosine triphosphate and pyruvate, however, the precise role(s) of PKM1/2 in naïve and primed pluripotency is not well understood. The primary objective of this study was to characterize the cellular expression and localization patterns of PKM1 and PKM2 in mESCs, chemically transitioned epiblast-like cells (mEpiLCs) representing formative pluripotency, and mEpiSCs using immunoblotting and confocal microscopy. The results indicate that PKM1 and PKM2 are not only localized to the cytoplasm, but also accumulate in differential subnuclear regions of mESC, mEpiLCs, and mEpiSCs as determined by a quantitative confocal microscopy employing orthogonal projections and airyscan processing. Importantly, we discovered that the subnuclear localization of PKM1/2 changes during the transition from mESCs, mEpiLCs, and mEpiSCs. Finally, we have comprehensively validated the appropriateness and power of the Pearson's correlation coefficient and Manders's overlap coefficient for assessing nuclear and cytoplasmic protein colocalization in PSCs by immunofluorescence confocal microscopy. We propose that nuclear PKM1/2 may assist with distinct pluripotency state maintenance and lineage priming by non-canonical mechanisms. These results advance our understanding of the overall mechanisms controlling naïve, formative, and primed pluripotency.
Collapse
Affiliation(s)
- Joshua G Dierolf
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Canada
| | - Andrew J Watson
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Canada; Department of Obstetrics and Gynecology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Canada; The Children's Health Research Institute (CHRI), Lawson Health Research Institute, London, Canada
| | - Dean H Betts
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Canada; Department of Obstetrics and Gynecology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Canada; The Children's Health Research Institute (CHRI), Lawson Health Research Institute, London, Canada.
| |
Collapse
|
22
|
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: 15] [Impact Index Per Article: 3.8] [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
|
23
|
Abstract
Clear cell renal cell carcinoma (ccRCC) is a major cancer yet has long evaded extensive efforts to target it chemotherapeutically. Recent efforts to characterize its proteome and metabolome in a grade-defined manner has resulted in a global proteometabolomic reprogramming model yielding a number of potential drug targets, many of which are under the control of transcription factor and MYC proto-oncogene, bHLH transcription factor. Furthermore, through the use of conventional technologies such as immunohistochemistry, protein moonlighting, a phenomenon wherein a single protein performs more than one distinct biochemical or biophysical functions, is emerging as a second mode of operation for ccRCC metabolo-proteomic reprogramming. This renders the subcellular localization of the grade-defining biomarkers an additional layer of grade-defining ccRCC molecular signature, although its functional significance in ccRCC etiology is only beginning to emerge.
Collapse
Affiliation(s)
- Tatsuto Ishimaru
- Division of Nephrology, Department of Internal Medicine, University of California, Davis, CA.
| |
Collapse
|
24
|
Gu J, Li X, Zhao L, Yang Y, Xue C, Gao Y, Li J, Han Q, Sun Z, Bai C, Zhao RC. The role of PKM2 nuclear translocation in the constant activation of the NF-κB signaling pathway in cancer-associated fibroblasts. Cell Death Dis 2021; 12:291. [PMID: 33731686 PMCID: PMC7969736 DOI: 10.1038/s41419-021-03579-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 02/23/2021] [Accepted: 02/25/2021] [Indexed: 12/24/2022]
Abstract
Cancer-associated fibroblasts (CAFs) play critical roles in cancer progression by regulating tumor cell proliferation, angiogenesis, and metastasis. Recent studies demonstrated that CAFs induce inhibitory immune cell infiltration and chemotherapy resistance in gastric cancer by activating the NF-κB signaling pathway to secrete IL6, IL8, and other inflammatory factors. Inhibition of the NF-κB signaling pathway in CAFs might be a potential therapeutic strategy in gastric cancer. However, how the NF-κB pathway is activated in CAFs remains unclear. We showed that mesenchymal stem cells (MSCs) differentiated into CAFs, induced by the exosomes derived from gastric cancer cells. During the process of differentiation from MSCs into CAFs, we showed that nuclear PKM2 expression was continuously upregulated and associated with NF-κB P65 acetylation, contributing to P65 nuclear retention in CAFs and constant transcription of IL-6, IL-8, and other inflammatory factors, thus promoting gastric cancer cell proliferation. We showed that NF-κB P65 acetylation was induced by P300. We showed that nuclear PKM2 was derived from exosomes of gastric cancer cell lines and the positive feedback loop induced by PKM2-P65 combination. It is also proved that P300 inhibitors can inhibit tumor proliferation in an AGS subcutaneous xenograft tumor model. Our study showed that gastric cancer cells influence the continuous activation of the NF-κB signaling pathway in CAFs by secreting gastric cancer exosomes containing PKM2, thus inducing abnormal metabolism and inflammation activation. This study provides a new therapeutic target for CAF normalization or deactivation strategies.
Collapse
Affiliation(s)
- Junjie Gu
- Department of Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100005, Beijing, China
| | - Xuechun Li
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory (No. BZO381), 100005, Beijing, China
| | - Lin Zhao
- Department of Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100005, Beijing, China
| | - Ying Yang
- Department of Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100005, Beijing, China
| | - Chunling Xue
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory (No. BZO381), 100005, Beijing, China
| | - Yang Gao
- Department of Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100005, Beijing, China
| | - Jing Li
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory (No. BZO381), 100005, Beijing, China
| | - Qin Han
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory (No. BZO381), 100005, Beijing, China.
| | - Zhao Sun
- Department of Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100005, Beijing, China.
| | - Chunmei Bai
- Department of Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100005, Beijing, China.
| | - Robert Chunhua Zhao
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory (No. BZO381), 100005, Beijing, China.
- School of Life Sciences, Shanghai University, 99 Shangda Road, 200444, Shanghai, China.
| |
Collapse
|
25
|
Integrated Metabolomics and Transcriptomics Analysis of Monolayer and Neurospheres from Established Glioblastoma Cell Lines. Cancers (Basel) 2021; 13:cancers13061327. [PMID: 33809510 PMCID: PMC8001840 DOI: 10.3390/cancers13061327] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 03/10/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Glioblastomas are very aggressive tumours without efficient treatment, where cancer stem-like cells are thought to be responsible for relapse. This pilot study investigated the metabolic discrepancies between monolayer and neurosphere cultures of two glioblastoma cell lines using transcriptomics and metabolomics. We show that the two culture systems display substantial differences regarding their metabolome and transcriptome. Specifically, we found that metabolic reactions connected to arginine biosynthesis are crucial to support the different metabolic needs of neurospheres from the two cell lines. By identifying metabolic vulnerabilities in different glioblastoma subpopulations, new therapeutic strategies may be emerging that can be explored to treat this disease. Moreover, this data set may be of great value as a resource for the scientific community. Abstract Altered metabolic processes contribute to carcinogenesis by modulating proliferation, survival and differentiation. Tumours are composed of different cell populations, with cancer stem-like cells being one of the most prominent examples. This specific pool of cells is thought to be responsible for cancer growth and recurrence and plays a particularly relevant role in glioblastoma (GBM), the most lethal form of primary brain tumours. Here, we have analysed the transcriptome and metabolome of an established GBM cell line (U87) and a patient-derived GBM stem-like cell line (NCH644) exposed to neurosphere or monolayer culture conditions. By integrating transcriptome and metabolome data, we identified key metabolic pathways and gene signatures that are associated with stem-like and differentiated states in GBM cells, and demonstrated that neurospheres and monolayer cells differ substantially in their metabolism and gene regulation. Furthermore, arginine biosynthesis was identified as the most significantly regulated pathway in neurospheres, although individual nodes of this pathway were distinctly regulated in the two cellular systems. Neurosphere conditions, as opposed to monolayer conditions, cause a transcriptomic and metabolic rewiring that may be crucial for the regulation of stem-like features, where arginine biosynthesis may be a key metabolic pathway. Additionally, TCGA data from GBM patients showed significant regulation of specific components of the arginine biosynthesis pathway, providing further evidence for the importance of this metabolic pathway in GBM.
Collapse
|
26
|
Lee SA, Ho C, Troxler M, Lin CY, Chung SH. Non-Metabolic Functions of PKM2 Contribute to Cervical Cancer Cell Proliferation Induced by the HPV16 E7 Oncoprotein. Viruses 2021; 13:433. [PMID: 33800513 PMCID: PMC8001101 DOI: 10.3390/v13030433] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/02/2021] [Accepted: 03/04/2021] [Indexed: 12/12/2022] Open
Abstract
Pyruvate kinase M2 (PKM2) mainly catalyzes glycolysis, but it also exerts non-glycolytic functions in several cancers. While it has been shown to interact with the human papillomavirus 16 (HPV16) E7 oncoprotein, the functional significance of PKM2 in HPV-associated cervical cancer has been elusive. Here, we show that HPV16 E7 increased the expression of PKM2 in cervical cancer cells. TCGA data analyses revealed a higher level of PKM2 in HPV+ than HPV- cervical cancers and a worse prognosis for patients with high PKM2 expression. Functionally, we demonstrate that shRNA-mediated PKM2 knockdown decreased the proliferation of HPV+ SiHa cervical cancer cells. PKM2 knockdown also inhibited the E7-induced proliferation of cervical cancer cells. ML265 activating the pyruvate kinase function of PKM2 inhibited cell cycle progression and colony formation. ML265 treatments decreased phosphorylation of PKM2 at the Y105 position that has been associated with non-glycolytic functions. On the contrary, HPV16 E7 increased the PKM2 phosphorylation. Our results indicate that E7 increases PKM2 expression and activates a non-glycolytic function of PKM2 to promote cervical cancer cell proliferation.
Collapse
Affiliation(s)
| | | | | | | | - Sang-Hyuk Chung
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA; (S.-A.L.); (C.H.); (M.T.); (C.-Y.L.)
| |
Collapse
|
27
|
Egan G, Khan DH, Lee JB, Mirali S, Zhang L, Schimmer AD. Mitochondrial and Metabolic Pathways Regulate Nuclear Gene Expression to Control Differentiation, Stem Cell Function, and Immune Response in Leukemia. Cancer Discov 2021; 11:1052-1066. [PMID: 33504581 DOI: 10.1158/2159-8290.cd-20-1227] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/16/2020] [Accepted: 11/24/2020] [Indexed: 11/16/2022]
Abstract
Mitochondria are involved in many biological processes including cellular homeostasis, energy generation, and apoptosis. Moreover, mitochondrial and metabolic pathways are interconnected with gene expression to regulate cellular functions such as cell growth, survival, differentiation, and immune recognition. Metabolites and mitochondrial enzymes regulate chromatin-modifying enzymes, chromatin remodeling, and transcription regulators. Deregulation of mitochondrial pathways and metabolism leads to alterations in gene expression that promote cancer development, progression, and evasion of the immune system. This review highlights how mitochondrial and metabolic pathways function as a central mediator to control gene expression, specifically on stem cell functions, differentiation, and immune response in leukemia. SIGNIFICANCE: Emerging evidence demonstrates that mitochondrial and metabolic pathways influence gene expression to promote tumor development, progression, and immune evasion. These data highlight new areas of cancer biology and potential new therapeutic strategies.
Collapse
Affiliation(s)
- Grace Egan
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Dilshad H Khan
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Jong Bok Lee
- Toronto General Hospital Research Institute, Toronto, Ontario, Canada
| | - Sara Mirali
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Li Zhang
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.,Toronto General Hospital Research Institute, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathology, University of Toronto, Toronto, Ontario, Canada.,Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Aaron D Schimmer
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada. .,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
28
|
Chen X, Chen S, Yu D. Protein kinase function of pyruvate kinase M2 and cancer. Cancer Cell Int 2020; 20:523. [PMID: 33292198 PMCID: PMC7597019 DOI: 10.1186/s12935-020-01612-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 10/20/2020] [Indexed: 02/07/2023] Open
Abstract
Pyruvate kinase is a terminal enzyme in the glycolytic pathway, where it catalyzes the conversion of phosphoenolpyruvate to pyruvate and production of ATP via substrate level phosphorylation. PKM2 is one of four isoforms of pyruvate kinase and is widely expressed in many types of tumors and associated with tumorigenesis. In addition to pyruvate kinase activity involving the metabolic pathway, increasing evidence demonstrates that PKM2 exerts a non-metabolic function in cancers. PKM2 has been shown to be translocated into nucleus, where it serves as a protein kinase to phosphorylate various protein targets and contribute to multiple physiopathological processes. We discuss the nuclear localization of PKM2, its protein kinase function and association with cancers, and regulation of PKM2 activity.
Collapse
Affiliation(s)
- Xun Chen
- Department of Oral and Maxillofacial Surgery, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, 56 Lingyuan West Road, Guangzhou, 510055, People's Republic of China
| | - Shangwu Chen
- Department of Biochemistry, Guangdong Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, People's Republic of China.
| | - Dongsheng Yu
- Department of Oral and Maxillofacial Surgery, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, 56 Lingyuan West Road, Guangzhou, 510055, People's Republic of China.
| |
Collapse
|
29
|
Kim DK, Song B, Han S, Jang H, Bae SH, Kim HY, Lee SH, Lee S, Kim JK, Kim HS, Hong KM, Lee BI, Youn HD, Kim SY, Kang SW, Jang H. Phosphorylation of OCT4 Serine 236 Inhibits Germ Cell Tumor Growth by Inducing Differentiation. Cancers (Basel) 2020; 12:cancers12092601. [PMID: 32932964 PMCID: PMC7565739 DOI: 10.3390/cancers12092601] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/09/2020] [Accepted: 09/09/2020] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Octamer-binding transcription factor 4 (OCT4) plays an important role in early embryonic development, but is rarely expressed in adults. However, in many cancer cells, this gene is re-expressed, making the cancer malignant. This present study revealed that inhibiting OCT4 transcriptional activity induces cancer cell differentiation and growth retardation. Specifically, when the phosphorylation of OCT4 serine 236 increases by interfering with the binding of protein phosphatase 1 (PP1) to OCT4, OCT4 loses its transcriptional activity and cancer cells differentiate. Therefore, this study presents the basis for the development of protein-protein interaction inhibitors that inhibit the binding of OCT4 and PP1 for cancer treatment. Abstract Octamer-binding transcription factor 4 (Oct4) plays an important role in maintaining pluripotency in embryonic stem cells and is closely related to the malignancies of various cancers. Although posttranslational modifications of Oct4 have been widely studied, most of these have not yet been fully characterized, especially in cancer. In this study, we investigated the role of phosphorylation of serine 236 of OCT4 [OCT4 (S236)] in human germ cell tumors (GCTs). OCT4 was phosphorylated at S236 in a cell cycle-dependent manner in a patient sample and GCT cell lines. The substitution of endogenous OCT4 by a mimic of phosphorylated OCT4 with a serine-to-aspartate mutation at S236 (S236D) resulted in tumor cell differentiation, growth retardation, and inhibition of tumor sphere formation. GCT cells expressing OCT4 S236D instead of endogenous OCT4 were similar to cells with OCT4 depletion at the mRNA transcript level as well as in the phenotype. OCT4 S236D also induced tumor cell differentiation and growth retardation in mouse xenograft experiments. Inhibition of protein phosphatase 1 by chemicals or short hairpin RNAs increased phosphorylation at OCT4 (S236) and resulted in the differentiation of GCTs. These results reveal the role of OCT4 (S236) phosphorylation in GCTs and suggest a new strategy for suppressing OCT4 in cancer.
Collapse
Affiliation(s)
- Dong Keon Kim
- Research Institute, National Cancer Center, Goyang 10408, Korea; (D.K.K.); (B.S.); (S.H.); (H.J.); (S.-H.B.); (H.Y.K.); (S.-H.L.); (S.L.); (J.K.K.); (K.-M.H.); (B.I.L.); (S.-Y.K.)
| | - Bomin Song
- Research Institute, National Cancer Center, Goyang 10408, Korea; (D.K.K.); (B.S.); (S.H.); (H.J.); (S.-H.B.); (H.Y.K.); (S.-H.L.); (S.L.); (J.K.K.); (K.-M.H.); (B.I.L.); (S.-Y.K.)
- Department of Life Science, Ewha Womans University, Seoul 03760, Korea;
| | - Suji Han
- Research Institute, National Cancer Center, Goyang 10408, Korea; (D.K.K.); (B.S.); (S.H.); (H.J.); (S.-H.B.); (H.Y.K.); (S.-H.L.); (S.L.); (J.K.K.); (K.-M.H.); (B.I.L.); (S.-Y.K.)
| | - Hansol Jang
- Research Institute, National Cancer Center, Goyang 10408, Korea; (D.K.K.); (B.S.); (S.H.); (H.J.); (S.-H.B.); (H.Y.K.); (S.-H.L.); (S.L.); (J.K.K.); (K.-M.H.); (B.I.L.); (S.-Y.K.)
- Department of Cancer Biomedical Science, National Cancer Center Graduate School of Cancer Science and Policy, Goyang 10408, Korea
| | - Seung-Hyun Bae
- Research Institute, National Cancer Center, Goyang 10408, Korea; (D.K.K.); (B.S.); (S.H.); (H.J.); (S.-H.B.); (H.Y.K.); (S.-H.L.); (S.L.); (J.K.K.); (K.-M.H.); (B.I.L.); (S.-Y.K.)
- Department of Cancer Biomedical Science, National Cancer Center Graduate School of Cancer Science and Policy, Goyang 10408, Korea
| | - Hee Yeon Kim
- Research Institute, National Cancer Center, Goyang 10408, Korea; (D.K.K.); (B.S.); (S.H.); (H.J.); (S.-H.B.); (H.Y.K.); (S.-H.L.); (S.L.); (J.K.K.); (K.-M.H.); (B.I.L.); (S.-Y.K.)
- Department of Life Science, Ewha Womans University, Seoul 03760, Korea;
| | - Seon-Hyeong Lee
- Research Institute, National Cancer Center, Goyang 10408, Korea; (D.K.K.); (B.S.); (S.H.); (H.J.); (S.-H.B.); (H.Y.K.); (S.-H.L.); (S.L.); (J.K.K.); (K.-M.H.); (B.I.L.); (S.-Y.K.)
| | - Seungjin Lee
- Research Institute, National Cancer Center, Goyang 10408, Korea; (D.K.K.); (B.S.); (S.H.); (H.J.); (S.-H.B.); (H.Y.K.); (S.-H.L.); (S.L.); (J.K.K.); (K.-M.H.); (B.I.L.); (S.-Y.K.)
- Department of Cancer Biomedical Science, National Cancer Center Graduate School of Cancer Science and Policy, Goyang 10408, Korea
| | - Jong Kwang Kim
- Research Institute, National Cancer Center, Goyang 10408, Korea; (D.K.K.); (B.S.); (S.H.); (H.J.); (S.-H.B.); (H.Y.K.); (S.-H.L.); (S.L.); (J.K.K.); (K.-M.H.); (B.I.L.); (S.-Y.K.)
| | - Han-Seong Kim
- Department of Pathology, Inje University Ilsan Paik Hospital, Goyang 10308, Korea;
| | - Kyeong-Man Hong
- Research Institute, National Cancer Center, Goyang 10408, Korea; (D.K.K.); (B.S.); (S.H.); (H.J.); (S.-H.B.); (H.Y.K.); (S.-H.L.); (S.L.); (J.K.K.); (K.-M.H.); (B.I.L.); (S.-Y.K.)
| | - Byung Il Lee
- Research Institute, National Cancer Center, Goyang 10408, Korea; (D.K.K.); (B.S.); (S.H.); (H.J.); (S.-H.B.); (H.Y.K.); (S.-H.L.); (S.L.); (J.K.K.); (K.-M.H.); (B.I.L.); (S.-Y.K.)
- Department of Cancer Biomedical Science, National Cancer Center Graduate School of Cancer Science and Policy, Goyang 10408, Korea
| | - Hong-Duk Youn
- National Creative Research Center for Epigenome Reprogramming Network, Department of Biomedical Sciences, Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 03080; Korea;
| | - Soo-Youl Kim
- Research Institute, National Cancer Center, Goyang 10408, Korea; (D.K.K.); (B.S.); (S.H.); (H.J.); (S.-H.B.); (H.Y.K.); (S.-H.L.); (S.L.); (J.K.K.); (K.-M.H.); (B.I.L.); (S.-Y.K.)
| | - Sang Won Kang
- Department of Life Science, Ewha Womans University, Seoul 03760, Korea;
| | - Hyonchol Jang
- Research Institute, National Cancer Center, Goyang 10408, Korea; (D.K.K.); (B.S.); (S.H.); (H.J.); (S.-H.B.); (H.Y.K.); (S.-H.L.); (S.L.); (J.K.K.); (K.-M.H.); (B.I.L.); (S.-Y.K.)
- Department of Cancer Biomedical Science, National Cancer Center Graduate School of Cancer Science and Policy, Goyang 10408, Korea
- Correspondence: ; Tel.: +82-31-920-2239
| |
Collapse
|
30
|
Oizel K, Yang C, Renoult O, Gautier F, Do QN, Joalland N, Gao X, Ko B, Vallette F, Ge WP, Paris F, DeBerardinis RJ, Pecqueur C. Glutamine uptake and utilization of human mesenchymal glioblastoma in orthotopic mouse model. Cancer Metab 2020; 8:9. [PMID: 32789014 PMCID: PMC7416393 DOI: 10.1186/s40170-020-00215-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 03/24/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Glioblastoma (GBM) are highly heterogeneous on the cellular and molecular basis. It has been proposed that glutamine metabolism of primary cells established from human tumors discriminates aggressive mesenchymal GBM subtype to other subtypes. METHODS To study glutamine metabolism in vivo, we used a human orthotopic mouse model for GBM. Tumors evolving from the implanted primary GBM cells expressing different molecular signatures were analyzed using mass spectrometry for their metabolite pools and enrichment in carbon 13 (13C) after 13C-glutamine infusion. RESULTS Our results showed that mesenchymal GBM tumors displayed increased glutamine uptake and utilization compared to both control brain tissue and other GBM subtypes. Furthermore, both glutamine synthetase and transglutaminase-2 were expressed accordingly to GBM metabolic phenotypes. CONCLUSION Thus, our results outline the specific enhanced glutamine flux in vivo of the aggressive mesenchymal GBM subtype.
Collapse
Affiliation(s)
- Kristell Oizel
- Children’s Research Institute, UT Southwestern Medical Center, Dallas, TX 75390 USA
| | - Chendong Yang
- Children’s Research Institute, UT Southwestern Medical Center, Dallas, TX 75390 USA
| | | | - Fabien Gautier
- Université de Nantes, CNRS, INSERM, CRCINA, Nantes, France
- Institut de Cancérologie de l’Ouest, Saint-Herblain, France
| | - Quyen N. Do
- Department of Radiology, UT Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390-9061 USA
| | - Noemie Joalland
- Université de Nantes, CNRS, INSERM, CRCINA, Nantes, France
- LabEx IGO “Immunotherapy, Graft, Oncology”, Nantes, France
| | - Xiaofei Gao
- Children’s Research Institute, UT Southwestern Medical Center, Dallas, TX 75390 USA
| | - Bookyung Ko
- Children’s Research Institute, UT Southwestern Medical Center, Dallas, TX 75390 USA
| | - François Vallette
- Université de Nantes, CNRS, INSERM, CRCINA, Nantes, France
- Institut de Cancérologie de l’Ouest, Saint-Herblain, France
| | - Woo-Ping Ge
- Children’s Research Institute, UT Southwestern Medical Center, Dallas, TX 75390 USA
- Department of Pediatrics, Neuroscience, Neurology & Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - François Paris
- Université de Nantes, CNRS, INSERM, CRCINA, Nantes, France
- Institut de Cancérologie de l’Ouest, Saint-Herblain, France
| | - Ralph J. DeBerardinis
- Children’s Research Institute, UT Southwestern Medical Center, Dallas, TX 75390 USA
- Howard Hughes Medical Institute, Chevy Chase, USA
| | - Claire Pecqueur
- Université de Nantes, CNRS, INSERM, CRCINA, Nantes, France
- LabEx IGO “Immunotherapy, Graft, Oncology”, Nantes, France
| |
Collapse
|
31
|
Shen YA, Pan SC, Chu I, Lai RY, Wei YH. Targeting cancer stem cells from a metabolic perspective. Exp Biol Med (Maywood) 2020; 245:465-476. [PMID: 32102562 PMCID: PMC7082881 DOI: 10.1177/1535370220909309] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The process of cancer development and progression is driven by distinct subsets of cancer stem cells (CSCs) that contribute the self-renewal capacity as the major impetus to the metastatic dissemination and main impediments in cancer treatment. Given that CSCs are so scarce in the tumor mass, there are debatable points on the metabolic signatures of CSCs. As opposed to differentiated tumor progenies, CSCs display exquisite patterns of metabolism that, depending on the type of cancer, predominately rely on glycolysis, oxidative metabolism of glutamine, fatty acids, or amino acids for ATP production. Metabolic heterogeneity of CSCs, which attributes to differences in type and microenvironment of tumors, confers CSCs to have the plasticity to cope with the endogenous mitochondrial stress and exogenous microenvironment. In essence, CSCs and normal stem cells are like mirror images of each other in terms of metabolism. To achieve reprogramming, CSCs not only need to upregulate their metabolic engine for self-renewal and defense mechanism, but also expedite the antioxidant defense to sustain the redox homeostasis. In the context of these pathways, this review portrays the connection between the metabolic features of CSCs and cancer stemness. Identification of the metabolic features in conferring resistance to anticancer treatment dictated by CSCs can enhance the opportunity to open up a new therapeutic dimension, which might not only improve the effectiveness of cancer therapies but also annihilate the whole tumor without recurrence. Henceforth, we highlight current findings of potential therapeutic targets for the design of alternative strategies to compromise the growth, drug resistance, and metastasis of CSCs by altering their metabolic phenotypes. Perturbing the versatile skills of CSCs by barricading metabolic signaling might bring about plentiful approaches to discover novel therapeutic targets for clinical application in cancer treatments.Impact statementThis minireview highlights the current evidence on the mechanisms of pivotal metabolic pathways that attribute to cancer stem cells (CSCs) with a special focus on developing metabolic strategies of anticancer treatment that can be exploited in preclinical and clinical settings. Specific metabolic inhibitors that can overwhelm the properties of CSCs may impede tumor recurrence and metastasis, and potentially achieve a permanent cure of cancer patients.
Collapse
Affiliation(s)
- Yao-An Shen
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Siao-Cian Pan
- Center for Mitochondrial Medicine and Free Radical Research, Changhua Christian Hospital, Changhua City 500, Taiwan
| | - I Chu
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Ruo-Yun Lai
- Department of Pathology, Taipei Medical University Hospital, Taipei Medical University, Taipei 110, Taiwan
| | - Yau-Huei Wei
- Center for Mitochondrial Medicine and Free Radical Research, Changhua Christian Hospital, Changhua City 500, Taiwan
| |
Collapse
|
32
|
Dichloroacetate (DCA) and Cancer: An Overview towards Clinical Applications. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8201079. [PMID: 31827705 PMCID: PMC6885244 DOI: 10.1155/2019/8201079] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 09/12/2019] [Accepted: 10/11/2019] [Indexed: 12/19/2022]
Abstract
An extensive body of literature describes anticancer property of dichloroacetate (DCA), but its effective clinical administration in cancer therapy is still limited to clinical trials. The occurrence of side effects such as neurotoxicity as well as the suspicion of DCA carcinogenicity still restricts the clinical use of DCA. However, in the last years, the number of reports supporting DCA employment against cancer increased also because of the great interest in targeting metabolism of tumour cells. Dissecting DCA mechanism of action helped to understand the bases of its selective efficacy against cancer cells. A successful coadministration of DCA with conventional chemotherapy, radiotherapy, other drugs, or natural compounds has been tested in several cancer models. New drug delivery systems and multiaction compounds containing DCA and other drugs seem to ameliorate bioavailability and appear more efficient thanks to a synergistic action of multiple agents. The spread of reports supporting the efficiency of DCA in cancer therapy has prompted additional studies that let to find other potential molecular targets of DCA. Interestingly, DCA could significantly affect cancer stem cell fraction and contribute to cancer eradication. Collectively, these findings provide a strong rationale towards novel clinical translational studies of DCA in cancer therapy.
Collapse
|
33
|
Sun X, Yao L, Liang H, Wang D, He Y, Wei Y, Ye L, Wang K, Li L, Chen J, Zhang CY, Xu G, Wang F, Zen K. Intestinal epithelial PKM2 serves as a safeguard against experimental colitis via activating β-catenin signaling. Mucosal Immunol 2019; 12:1280-1290. [PMID: 31462699 DOI: 10.1038/s41385-019-0197-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 07/22/2019] [Accepted: 08/08/2019] [Indexed: 02/04/2023]
Abstract
The pyruvate kinase M2 (PKM2)-mediated aerobic glycolysis has been shown to play a critical role in promoting cell survival and proliferation. However, little is known about the function of intestinal epithelial PKM2 in intestine homeostasis. Here we investigate whether and how intestinal epithelial PKM2 modulates the morphology and function of the adult intestine in experimental colitis. Analyzing colonoscopic biopsies from Crohn's disease and ulcerative colitis patients, we found significantly decreased level of intestinal epithelial PKM2 in patients compared to that in non-inflamed tissues. Similar reduction of intestinal epithelial PKM2 was observed in mice with dextran sulfate sodium-induced colitis. Moreover, intestinal epithelial-specific PKM2-knockout (Pkm2-/-) mice displayed more severe intestinal inflammation, as evidenced by a shortened colon, disruption of epithelial tight junctions, an increase in inflammatory cytokine levels, and immune cell infiltration, when compared to wild-type mice. Gene profiling, western blot, and function analyses indicated that cell survival signals, particularly the Wnt/β-catenin pathways, were associated with PKM2 activity. Increasing mouse intestinal epithelial PKM2 expression via delivery of a PKM2-expressing plasmid attenuated experimental colitis. In conclusion, our studies demonstrate that intestinal epithelial PKM2 increases cell survival and wound healing under the colitic condition via activating the Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Xinlei Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210093, China
| | - Li Yao
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210093, China
| | - Hongwei Liang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210093, China
| | - Dong Wang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, 210009, China
| | - Yueqin He
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210093, China
| | - Yao Wei
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210093, China
- School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Lei Ye
- Department of Gastroenterology and Hepatology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, 210093, China
| | - Kai Wang
- Research Institute of General Surgery, Jinling Hospital, Nanjing University School of Medicine, No. 305 East Zhongshan Road, Nanjing, Jiangsu, 210002, China
| | - Limin Li
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210093, China
| | - Jiangning Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210093, China
| | - Chen-Yu Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210093, China
| | - Guifang Xu
- Department of Gastroenterology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210008, China.
| | - Fangyu Wang
- Department of Gastroenterology and Hepatology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, 210093, China.
| | - Ke Zen
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210093, China.
| |
Collapse
|
34
|
Regulation of chromatin and gene expression by metabolic enzymes and metabolites. Nat Rev Mol Cell Biol 2019; 19:563-578. [PMID: 29930302 DOI: 10.1038/s41580-018-0029-7] [Citation(s) in RCA: 304] [Impact Index Per Article: 50.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Metabolism and gene expression, which are two fundamental biological processes that are essential to all living organisms, reciprocally regulate each other to maintain homeostasis and regulate cell growth, survival and differentiation. Metabolism feeds into the regulation of gene expression via metabolic enzymes and metabolites, which can modulate chromatin directly or indirectly - through regulation of the activity of chromatin trans-acting proteins, including histone-modifying enzymes, chromatin-remodelling complexes and transcription regulators. Deregulation of these metabolic activities has been implicated in human diseases, prominently including cancer.
Collapse
|
35
|
Zhang Z, Deng X, Liu Y, Liu Y, Sun L, Chen F. PKM2, function and expression and regulation. Cell Biosci 2019; 9:52. [PMID: 31391918 PMCID: PMC6595688 DOI: 10.1186/s13578-019-0317-8] [Citation(s) in RCA: 274] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 06/20/2019] [Indexed: 12/14/2022] Open
Abstract
Pyruvate kinase (PK), as one of the key enzymes for glycolysis, can encode four different subtypes from two groups of genes, although the M2 subtype PKM2 is expressed mainly during embryonic development in normal humans, and is closely related to tissue repair and regeneration, with the deepening of research, the role of PKM2 in tumor tissue has received increasing attention. PKM2 can be aggregated into tetrameric and dimeric forms, PKM2 in the dimer state can enter the nuclear to regulate gene expression, the transformation between them can play an important role in tumor cell energy supply, epithelial-mesenchymal transition (EMT), invasion and metastasis and cell proliferation. We will use the switching effect of PKM2 in glucose metabolism as the entry point to expand and enrich the Warburg effect. In addition, PKM2 can also regulate each other with various proteins by phosphorylation, acetylation and other modifications, mediate the different intracellular localization of PKM2 and then exert specific biological functions. In this paper, we will illustrate each of these points.
Collapse
Affiliation(s)
- Ze Zhang
- Department of General Surgery, The First Hospital of Jilin University, Changchun, 130021 China
| | - Xinyue Deng
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, 130021 China
| | - Yuanda Liu
- Department of Gastrointestinal Surgery, The Second Hospital of Jilin University, Changchun, 130041 China
| | - Yahui Liu
- Department of General Surgery, The First Hospital of Jilin University, Changchun, 130021 China
| | - Liankun Sun
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, 130021 China
| | - Fangfang Chen
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130021 China
| |
Collapse
|
36
|
Amin S, Yang P, Li Z. Pyruvate kinase M2: A multifarious enzyme in non-canonical localization to promote cancer progression. Biochim Biophys Acta Rev Cancer 2019; 1871:331-341. [PMID: 30826427 DOI: 10.1016/j.bbcan.2019.02.003] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 01/21/2019] [Accepted: 02/13/2019] [Indexed: 12/13/2022]
Abstract
Rewiring glucose metabolism, termed as Warburg effect or aerobic glycolysis, is a common signature of cancer cells to meet their high energetic and biosynthetic demands of rapid growth and proliferation. Pyruvate kinase M2 isoform (PKM2) is a key player in such metabolic reshuffle, which functions as a rate-limiting glycolytic enzyme in the cytosol of highly-proliferative cancer cells. During the recent decades, PKM2 has been extensively studied in non-canonical localizations such as nucleus, mitochondria, and extracellular secretion, and pertained to novel biological functions in tumor progression. Such functions of PKM2 open a new avenue for cancer researchers. This review summarizes up-to-date functions of PKM2 at various subcellular localizations of cancer cells and draws attention to the translocation of PKM2 from cytosol into the nucleus induced by posttranslational modifications. Moreover, PKM2 in tumor cells could have an important role in resistance acquisition processes against various chemotherapeutic drugs, which have raised a concern on PKM2 as a potential therapeutic target. Finally, we summarize the current status and future perspectives to improve the potential of PKM2 as a therapeutic target for the development of anticancer therapeutic strategies.
Collapse
Affiliation(s)
- Sajid Amin
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, China; Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China
| | - Peng Yang
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, China; Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China
| | - Zhuoyu Li
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, China; School of Life Science, Shanxi University, Taiyuan 030006, China.
| |
Collapse
|
37
|
Garnier D, Renoult O, Alves-Guerra MC, Paris F, Pecqueur C. Glioblastoma Stem- Like Cells, Metabolic Strategy to Kill a Challenging Target. Front Oncol 2019; 9:118. [PMID: 30895167 PMCID: PMC6415584 DOI: 10.3389/fonc.2019.00118] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 02/11/2019] [Indexed: 01/25/2023] Open
Abstract
Over the years, substantial evidence has definitively confirmed the existence of cancer stem-like cells within tumors such as Glioblastoma (GBM). The importance of Glioblastoma stem-like cells (GSCs) in tumor progression and relapse clearly highlights that cancer eradication requires killing of GSCs that are intrinsically resistant to conventional therapies as well as eradication of the non-GSCs cells since GSCs emergence relies on a dynamic process. The past decade of research highlights that metabolism is a significant player in tumor progression and actually might orchestrate it. The growing interest in cancer metabolism reprogrammation can lead to innovative approaches exploiting metabolic vulnerabilities of cancer cells. These approaches are challenging since they require overcoming the compensatory and adaptive responses of GSCs. In this review, we will summarize the current knowledge on GSCs with a particular focus on their metabolic complexity. We will also discuss potential approaches targeting GSCs metabolism to potentially improve clinical care.
Collapse
Affiliation(s)
| | | | | | - François Paris
- CRCINA, INSERM CNRS, Université de Nantes, Nantes, France.,Institut de Cancérologie de l'Ouest - René Gauducheau, St Herblain, France
| | - Claire Pecqueur
- CRCINA, INSERM CNRS, Université de Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| |
Collapse
|
38
|
Archer SL. Pyruvate Kinase and Warburg Metabolism in Pulmonary Arterial Hypertension: Uncoupled Glycolysis and the Cancer-Like Phenotype of Pulmonary Arterial Hypertension. Circulation 2019; 136:2486-2490. [PMID: 29255124 DOI: 10.1161/circulationaha.117.031655] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Stephen L Archer
- Department of Medicine, Queen's University, Kingston, Ontario, Canada.
| |
Collapse
|
39
|
Abramek J, Bogucki J, Ziaja-Sołtys M, Stępniewski A, Bogucka-Kocka A. Effect of sodium dichloroacetate on apoptotic gene expression in human leukemia cell lines. Pharmacol Rep 2018; 71:248-256. [PMID: 30822618 DOI: 10.1016/j.pharep.2018.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 11/20/2018] [Accepted: 12/11/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Sodium dichloroacetate (DCA) is an agent with anticancer properties against solid tumors. DCA also seems to have antileukemic activity. In order to affirm it we investigate the effect of DCA on cell viability and apoptotic gene expression profiles in leukemia cell lines: CEM/C1, CCRF/CEM, HL-60, HL-60/MX2. METHODS Cell viability was assessed by trypan blue staining. The expression of 93 genes involved in the process of apoptosis was determined by real-time PCR method using Taqman Low Density Array (TLDA). RESULTS CEM/C1, CCRF/CEM, HL-60, HL-60/MX2 cells were exposed to DCA for 24 h. The sensitivity of each cell line to DCA is different and depends on the concentration. CEM/C1 was the most sensitive with an half-maximal inhibitory concentration (IC50) value of 30 mM, while HL-60/MX2 was the most resistant with an IC50 value of 75 mM. Exposure of leukemia cells to DCA causes differences in gene expression profiles which cannot indicate that any particular pathway of apoptosis is initiated. However, the presence of 388 statistically significant correlations between expression pattern of gens was determined. CONCLUSION We showed that DCA causes a decrease in viability of leukemia cells. The decline depends on DCA concentration. The induction of any particular apoptosis pathway is not shown in cells after DCA treatment. For that reason, studies on the molecular mechanism of cell death after exposure to DCA should be continued.
Collapse
Affiliation(s)
- Jagoda Abramek
- Chair and Department of Biology and Genetics, Medical University of Lublin, Lublin, Poland.
| | - Jacek Bogucki
- Department of Clinical Genetics, Medical University of Lublin, Lublin, Poland.
| | - Marta Ziaja-Sołtys
- Chair and Department of Biology and Genetics, Medical University of Lublin, Lublin, Poland.
| | | | - Anna Bogucka-Kocka
- Chair and Department of Biology and Genetics, Medical University of Lublin, Lublin, Poland.
| |
Collapse
|
40
|
Transcriptomic but not genomic variability confers phenotype of breast cancer stem cells. Cancer Commun (Lond) 2018; 38:56. [PMID: 30231942 PMCID: PMC6146522 DOI: 10.1186/s40880-018-0326-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 09/08/2018] [Indexed: 12/19/2022] Open
Abstract
Background Breast cancer stem cells (BCSCs) are considered responsible for cancer relapse and drug resistance. Understanding the identity of BCSCs may open new avenues in breast cancer therapy. Although several discoveries have been made on BCSC characterization, the factors critical to the origination of BCSCs are largely unclear. This study aimed to determine whether genomic mutations contribute to the acquisition of cancer stem-like phenotype and to investigate the genetic and transcriptional features of BCSCs. Methods We detected potential BCSC phenotype-associated mutation hotspot regions by using whole-genome sequencing on parental cancer cells and derived serial-generation spheres in increasing order of BCSC frequency, and then performed target deep DNA sequencing at bulk-cell and single-cell levels. To identify the transcriptional program associated with BCSCs, bulk-cell and single-cell RNA sequencing was performed. Results By using whole-genome sequencing of bulk cells, potential BCSC phenotype-associated mutation hotspot regions were detected. Validation by target deep DNA sequencing, at both bulk-cell and single-cell levels, revealed no genetic changes specifically associated with BCSC phenotype. Moreover, single-cell RNA sequencing showed profound transcriptomic variability in cancer cells at the single-cell level that predicted BCSC features. Notably, this transcriptomic variability was enriched during the transcription of 74 genes, revealed as BCSC markers. Breast cancer patients with a high risk of relapse exhibited higher expression levels of these BCSC markers than those with a low risk of relapse, thereby highlighting the clinical significance of predicting breast cancer prognosis with these BCSC markers. Conclusions Transcriptomic variability, not genetic mutations, distinguishes BCSCs from non-BCSCs. The identified 74 BCSC markers have the potential of becoming novel targets for breast cancer therapy. Electronic supplementary material The online version of this article (10.1186/s40880-018-0326-8) contains supplementary material, which is available to authorized users.
Collapse
|
41
|
Li YH, Li XF, Liu JT, Wang H, Fan LL, Li J, Sun GP. PKM2, a potential target for regulating cancer. Gene 2018; 668:48-53. [PMID: 29775756 DOI: 10.1016/j.gene.2018.05.038] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 03/07/2018] [Accepted: 05/13/2018] [Indexed: 01/09/2023]
Abstract
Aberrated glucose metabolism is a key future of cancer cells. Unlike normal cells, tumor cells favor glycolysis even in the presence of sufficient oxygen. Pyruvate kinase (PK), a key glucose metabolic enzyme, converts phosphoenolpyruvate (PEP) to pyruvate by transferring the high-energy phosphate group to adenosine diphosphate (ADP) to produce adenosine triphosphate (ATP). Pyruvate kinase M2 (PKM2), one of the four isozyme of PK, which universally expressed in rapidly proliferating cells such as embryonic cells and cancer cells. Recent years, more and more research suggested PKM2 plays a crucial role in cancer progression through both metabolic and non-metabolic pathways. On the one hand, the middle product of glycolysis, such as amino acids, nucleotides, lipids is necessary to rapid growth of cancer cells. On the other hand, PKM2 supports tumor growth through regulating the expression of gene that involved in cell proliferation, migration and apoptosis. In this article, we review the recent advances to further understand the regulation and function of PKM2 in tumorigenesis. Given its multiple effects on cancer, PKM2 may be a potential target for cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Yu-Huan Li
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, China
| | - Xiao-Feng Li
- School of Pharmacy, Anhui Medical University, Mei Shan Road, Hefei 230032, Anhui, China
| | - Jia-Tao Liu
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, China
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, China
| | - Lu-Lu Fan
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, China
| | - Jun Li
- School of Pharmacy, Anhui Medical University, Mei Shan Road, Hefei 230032, Anhui, China.
| | - Guo-Ping Sun
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, China
| |
Collapse
|
42
|
Abstract
Cancer stem cells are a subpopulation of cells within a tumour believed to confer resistance to standard cancer therapies. Although many studies have addressed the specific mechanisms of tumour recurrence driven by cancer stem cells, cellular metabolism is an often-neglected attribute. The metabolic features of cancer stem cells are still poorly understood, and they thus constitute a promising field in cancer research. The findings published so far point to a distinct metabolic phenotype in cancer stem cells, which might depend on the cancer type, the model system used or even the experimental design, and several controversies still need to be tackled. This Review describes the metabolic phenotype of cancer stem cells by addressing the main metabolic traits in different tumours, including glycolysis and oxidative, glutamine, fatty acid and amino acid metabolism. In the context of these pathways, we also mention the specific alterations in metabolic enzymes and metabolite levels that have a role in the regulation of cancer stemness. Determining the role of metabolism in supporting resistance to therapy driven by cancer stem cells can raise the opportunity for novel therapeutic targets, which might not only eliminate this resistant population, but, more importantly, eradicate the whole tumour in a relapse-free scenario. Summary: The intrinsic mechanisms that define cancer stem cells, specifically their metabolic properties, are summarized in this Review, in an attempt to point out the benefit of targeting metabolism as a novel therapeutic approach.
Collapse
Affiliation(s)
- Joana Peixoto
- Cancer Signalling and Metabolism Group, Instituto de Investigação e Inovação em Saúde (I3S), Universidade do Porto, 4200-135 Porto, Portugal.,Cancer Signalling and Metabolism Group, Institute of Molecular Pathology and Immunology of the University of Porto (Ipatimup), 4200-465 Porto, Portugal.,Medical Faculty of the University of Porto, 4200-319 Porto, Portugal.,Department of Biochemistry and Molecular Biology, Theodor-Boveri-Institute, Biocenter, 97074 Würzburg, Germany
| | - Jorge Lima
- Cancer Signalling and Metabolism Group, Instituto de Investigação e Inovação em Saúde (I3S), Universidade do Porto, 4200-135 Porto, Portugal .,Cancer Signalling and Metabolism Group, Institute of Molecular Pathology and Immunology of the University of Porto (Ipatimup), 4200-465 Porto, Portugal.,Medical Faculty of the University of Porto, 4200-319 Porto, Portugal
| |
Collapse
|
43
|
Yang YC, Chien MH, Liu HY, Chang YC, Chen CK, Lee WJ, Kuo TC, Hsiao M, Hua KT, Cheng TY. Nuclear translocation of PKM2/AMPK complex sustains cancer stem cell populations under glucose restriction stress. Cancer Lett 2018; 421:28-40. [PMID: 29408265 DOI: 10.1016/j.canlet.2018.01.075] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 01/26/2018] [Accepted: 01/30/2018] [Indexed: 02/07/2023]
Abstract
Cancer cells encounter metabolic stresses such as hypoxia and nutrient limitations because they grow and divide more quickly than their normal counterparts. In response to glucose restriction, we found that nuclear translocation of the glycolic enzyme, pyruvate kinase M2 (PKM2), helped cancer cells survive under the metabolic stress. Restriction of glucose stimulated AMPK activation and resulted in co-translocation of AMPK and PKM2 through Ran-mediated nuclear transport. Nuclear PKM2 subsequently bound to Oct4 and promoted the expression of cancer stemness-related genes, which might enrich the cancer stem cell population under the metabolic stress. Nuclear PKM2 was also capable of promoting cancer metastasis in an orthotopic xenograft model. In summary, we found that cytosolic AMPK helped PKM2 carry out its nonmetabolic functions in the nucleus under glucose restriction and that nuclear PKM2 promoted cancer stemness and metastasis. These findings suggested a potential new targeting pathway for cancer therapy in the future.
Collapse
Affiliation(s)
- Yi-Chieh Yang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC; Genomics Research Center, Academia Sinica, Taipei, Taiwan, ROC
| | - Ming-Hsien Chien
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
| | - Hsin-Yi Liu
- Graduate Institute of Biochemical Sciences, College of Life Science Sciences, National Taiwan University, Taipei, Taiwan, ROC
| | - Yu-Chan Chang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan, ROC
| | - Chi-Kuan Chen
- Genomics Research Center, Academia Sinica, Taipei, Taiwan, ROC; Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Wei-Jiunn Lee
- Department of Urology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan, ROC
| | - Tsang-Chih Kuo
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei, Taiwan, ROC; Department of Biochemistry, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
| | - Kuo-Tai Hua
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Tsu-Yao Cheng
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan, ROC.
| |
Collapse
|
44
|
Snaebjornsson MT, Schulze A. Non-canonical functions of enzymes facilitate cross-talk between cell metabolic and regulatory pathways. Exp Mol Med 2018; 50:1-16. [PMID: 29657328 PMCID: PMC5938058 DOI: 10.1038/s12276-018-0065-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 12/22/2017] [Indexed: 12/25/2022] Open
Abstract
The metabolic rewiring that occurs during cell transformation is a hallmark of cancer. It is diverse in different cancers as it reflects different combinations of oncogenic drivers, tumor suppressors, and the microenvironment. Metabolic rewiring is essential to cancer as it enables uncontrolled proliferation and adaptation to the fluctuating availability of nutrients and oxygen caused by poor access to the vasculature due to tumor growth and a foreign microenvironment encountered during metastasis. Increasing evidence now indicates that the metabolic state in cancer cells also plays a causal role in tumor growth and metastasis, for example through the action of oncometabolites, which modulate cell signaling and epigenetic pathways to promote malignancy. In addition to altering the metabolic state in cancer cells, some multifunctional enzymes possess non-metabolic functions that also contribute to cell transformation. Some multifunctional enzymes that are highly expressed in cancer, such as pyruvate kinase M2 (PKM2), have non-canonical functions that are co-opted by oncogenic signaling to drive proliferation and inhibit apoptosis. Other multifunctional enzymes that are frequently downregulated in cancer, such as fructose-bisphosphatase 1 (FBP1), are tumor suppressors, directly opposing mitogenic signaling via their non-canonical functions. In some cases, the enzymatic and non-canonical roles of these enzymes are functionally linked, making the modulation of non-metabolic cellular processes dependent on the metabolic state of the cell.
Collapse
Affiliation(s)
- Marteinn T Snaebjornsson
- Department of Biochemistry and Molecular Biology, Theodor-Boveri-Institute, Biocenter, Am Hubland, 97074, Würzburg, Germany.,Comprehensive Cancer Center Mainfranken, Josef-Schneider Strasse 6, 97080, Würzburg, Germany
| | - Almut Schulze
- Department of Biochemistry and Molecular Biology, Theodor-Boveri-Institute, Biocenter, Am Hubland, 97074, Würzburg, Germany. .,Comprehensive Cancer Center Mainfranken, Josef-Schneider Strasse 6, 97080, Würzburg, Germany.
| |
Collapse
|
45
|
Lin Z, Lu Y, Meng Q, Wang C, Li X, Yang Y, Xin X, Zheng Q, Xu J, Gui X, Li T, Pu H, Xiong W, Li J, Jia S, Lu D. miR372 Promotes Progression of Liver Cancer Cells by Upregulating erbB-2 through Enhancement of YB-1. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 11:494-507. [PMID: 29858084 PMCID: PMC5992473 DOI: 10.1016/j.omtn.2018.04.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 03/30/2018] [Accepted: 04/04/2018] [Indexed: 12/16/2022]
Abstract
MicroRNAs are known to be involved in carcinogenesis. Recently, microRNA-372 (miR372) has been proven to play a substantial role in several human cancers, but its functions in liver cancer remain unclear. Herein, our results demonstrate that miR372 accelerates growth of liver cancer cells in vitro and in vivo. Mechanistically, miR372 enhances expression of Y-box-binding protein 1 (YB-1) by targeting for phosphatase and tensin homolog (PTEN) directly and consequently promotes phosphorylation of YB-1 via HULC looping dependent on ERK1/2 and PTEN. In particular, HULC knockdown or PTEN overexpression abrogated this miR372 action. Moreover, miR372 inhibits the degradation of β-catenin dependent on phosphorylation of YB-1 and then enhances the expression and activity of pyruvate kinase M2 isoform (PKM2) by β-catenin-LEF/TCF4 pathway. Furthermore, the loading of LEF/TCF4 on PKM2 promoter region was significantly increased in miR372 overexpressing Hep3B, and thus, glycolytic proton efflux rate (glycoPER) was significantly increased in rLV-miR372 group compared to the rLV group. Moreover, β-catenin knockdown abrogates this function of miR372. Ultimately, miR372 promotes the expression of erbB-2 through PKM2-pH3T11-acetylation on histone H3 lysine 9 (H3K9Ac) pathway. Of significance, both YB-1 knockdown and erbB-2 knockdown abrogate oncogenic action of miR372. Our observations suggest that miR372 promotes liver cancer cell cycle progress by activating cyclin-dependent kinase 2 (CDK2)-cyclin E-P21/Cip1 complex through miR372-YB-1-β-catenin-LEF/TCF4-PKM2-erbB-2 axis. This study elucidates a novel mechanism for miR372 in liver cancer cells and suggests that miR372 can be used as a novel therapeutic target of liver cancer.
Collapse
Affiliation(s)
- Zhuojia Lin
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Yanan Lu
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Qiuyu Meng
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Chen Wang
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Xiaonan Li
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Yuxin Yang
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Xiaoru Xin
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Qidi Zheng
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Jie Xu
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Xin Gui
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Tianming Li
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Hu Pu
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Wujun Xiong
- Department of Hepatology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Jiao Li
- School of Medicine, Tongji University, Shanghai 200092, China
| | - Song Jia
- School of Medicine, Tongji University, Shanghai 200092, China
| | - Dongdong Lu
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, Shanghai 200092, China.
| |
Collapse
|
46
|
Hsu MC, Hung WC. Pyruvate kinase M2 fuels multiple aspects of cancer cells: from cellular metabolism, transcriptional regulation to extracellular signaling. Mol Cancer 2018; 17:35. [PMID: 29455645 PMCID: PMC5817853 DOI: 10.1186/s12943-018-0791-3] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 02/01/2018] [Indexed: 12/11/2022] Open
Abstract
Originally identified as a metabolic enzyme that catalyzes the transfer of a phosphate group from phosphoenolpyruvate (PEP) to ADP in the glycolytic pathway, pyruvate kinase M2-type (PKM2) has been shown to exhibit novel biological activities in the nucleus and outside the cells. Although cell-based studies reveal new non-canonical functions of PKM2 in gene transcription, epigenetic modulation and cell cycle progression, the importance of these non-canonical functions in PKM2-mediated tumorigenesis is still under debate because studies in genetically modified mice do not consistently echo the findings observed in cultured cancer cells. In addition to regulation of gene expression, the existence of PKM2 in exosomes opens a new venue to study the potential role of this glycolytic enzyme in cell-cell communication and extracellular signal initiation. In this review, we briefly summarize current understanding of PKM2 in metabolic switch and gene regulation. We will then emphasize recent progress of PKM2 in extracellular signaling and tumor microenvironment reprogramming. Finally, the discrepancy of some PKM2’s functions in vitro and in vivo, and the application of PKM2 in cancer detection and treatment will be discussed.
Collapse
Affiliation(s)
- Ming-Chuan Hsu
- National Institute of Cancer Research, National Health Research Institutes, No. 367, Shengli Road, Tainan, 704, Taiwan
| | - Wen-Chun Hung
- National Institute of Cancer Research, National Health Research Institutes, No. 367, Shengli Road, Tainan, 704, Taiwan. .,Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 802, Taiwan.
| |
Collapse
|
47
|
Zheng Q, Lin Z, Xu J, Lu Y, Meng Q, Wang C, Yang Y, Xin X, Li X, Pu H, Gui X, Li T, Xiong W, Lu D. Long noncoding RNA MEG3 suppresses liver cancer cells growth through inhibiting β-catenin by activating PKM2 and inactivating PTEN. Cell Death Dis 2018; 9:253. [PMID: 29449541 PMCID: PMC5833746 DOI: 10.1038/s41419-018-0305-7] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 01/09/2018] [Accepted: 01/11/2018] [Indexed: 12/13/2022]
Abstract
Maternally expressed gene 3 (MEG3) encodes an lncRNA which is suggested to function as a tumor suppressor and has been showed to involve in a variety of cancers. Herein, our findings demonstrate that MEG3 inhibits the malignant progression of liver cancer cells in vitro and in vivo. Mechanistically, MEG3 promotes the expression and maturition of miR122 which targets PKM2. Therefore, MEG3 decreases the expression and nuclear location of PKM2 dependent on miR122. Furthermore, MEG3 also inhibits CyclinD1 and C-Myc via PKM2 in liver cancer cells. On the other hand, MEG3 promotes β-catenin degradation through ubiquitin-proteasome system dependent on PTEN. Strikingly, MEG3 inhibits β-catenin activity through PKM2 reduction and PTEN increase. Significantly, we also found that excessive β-catenin abrogated the effect of MEG3 in liver cancer. In conclusion, our study for the first time demonstrates that MEG3 acts as a tumor suppressor by negatively regulating the activity of the PKM2 and β-catenin signaling pathway in hepatocarcinogenesis and could provide potential therapeutic targets for the treatment of liver cancer.
Collapse
Affiliation(s)
- Qidi Zheng
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China
| | - Zhuojia Lin
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China
| | - Jie Xu
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China
| | - Yanan Lu
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China
| | - Qiuyu Meng
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China
| | - Chen Wang
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China
| | - Yuxin Yang
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China
| | - Xiaoru Xin
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China
| | - Xiaonan Li
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China
| | - Hu Pu
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China
| | - Xin Gui
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China
| | - Tianming Li
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China
| | - Wujun Xiong
- Department of Hepatology, Shanghai East Hospital, Tongji University School of Medicine, 200120, Shanghai, China
| | - Dongdong Lu
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China.
| |
Collapse
|
48
|
Phage display library selection of a hypoxia-binding scFv antibody for liver cancer metabolic marker discovery. Oncotarget 2018; 7:38105-38121. [PMID: 27203546 PMCID: PMC5122375 DOI: 10.18632/oncotarget.9460] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 05/04/2016] [Indexed: 01/29/2023] Open
Abstract
Hypoxia, which is frequently observed in liver cancer and metastasis, influences tumor progression and resistance to therapy. Although hypoxia-associated biomarkers are of use in other cancers, none is recognized as a surrogate for hypoxia in liver cancer. In this study, we generated seven unique human single-chain Fv (scFv) antibodies (Abs) specific to hypoxic liver cancer cells, using normoxia-depleted vs hypoxia-selected phage library panning technology. By developing the scFv immunoprecipitation-based mass spectrometry method, the antigen that bound with one of the Abs (H103) was identified as the M2 splice isoform of pyruvate kinase (PKM2), an enzyme that is a key regulator of aerobic glycolysis in cancer cells. Increased expression of PKM2 was induced by hypoxia in liver cancer cell lines. Immunohistochemical (IHC) staining showed that PKM2 was highly expressed in moderately and well differentiated hepatocellular carcinoma (HCC) tissues with a hypovascular staining pattern. High expression of PKM2 was also localized in the perinecrotic area of intrahepatic cholangiocarcinoma (ICC) tissues. The percentage of the HCC or ICC tumor expressing PKM2 was significantly higher with more tumor necrosis, low microvessel density, and advanced stage. Moreover, the H103 scFv Ab was efficiently internalized into hypoxic liver cancer cells and could have potential for targeted drug delivery. Conclusion: our study, for the first time, developed hypoxia-specific scFv Ab H103 to liver cancer cells, and revealed that PKM2 is a promising biomarker for hypoxia in HCC and ICC tissues. These allow further exploration of this valuable Ab and PKM2 antigen for hypoxia targeting in liver cancer.
Collapse
|
49
|
Horiuchi Y, Nakatsu D, Kano F, Murata M. Pyruvate kinase M1 interacts with A-Raf and inhibits endoplasmic reticulum stress-induced apoptosis by activating MEK1/ERK pathway in mouse insulinoma cells. Cell Signal 2017; 38:212-222. [PMID: 28743549 DOI: 10.1016/j.cellsig.2017.07.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 07/20/2017] [Accepted: 07/20/2017] [Indexed: 12/19/2022]
Abstract
Apoptotic death of pancreatic β cells is a major cause of type 2 diabetes mellitus (T2D) progression. Two isoforms of pyruvate kinase, PKM1 and PKM2, have been reported to participate in cell death in several cell types; however, little is known about their causal pathways in pancreatic β-cell death. We examined whether the suppression of PKM1 or PKM2 affects endoplasmic reticulum (ER) stress-induced apoptosis in a pancreatic β-cell line, MIN6, and Beta-TC-6 and found that knockdown of PKM1, but not of PKM2, leads to the induction of ER stress-induced apoptosis in these cells. We also investigated the mechanism by which PKM1 inhibits ER stress-induced apoptosis. We confirmed that PKM1 interacts with A-Raf, an upstream regulator of the MEK/ERK pathway, and that this interaction contributes to MEK1 phosphorylation by A-Raf. PKM1 knockdown suppresses the phosphorylation of MEK, ERK, and caspase-9 (Thr125), which is phosphorylated by the MEK/ERK pathway, thereby inhibiting the cleavage and activation of caspase-9. Thus, PKM1 knockdown activates the caspase-9/caspase-3 pathway under ER stress conditions and leads to apoptosis.
Collapse
Affiliation(s)
- Yuta Horiuchi
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 153-8902, Japan
| | - Daiki Nakatsu
- Cell Biology Unit, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan
| | - Fumi Kano
- Cell Biology Unit, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan
| | - Masayuki Murata
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 153-8902, Japan; Cell Biology Unit, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan.
| |
Collapse
|
50
|
Metabolic Reprogramming, Autophagy, and Reactive Oxygen Species Are Necessary for Primordial Germ Cell Reprogramming into Pluripotency. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:4745252. [PMID: 28757909 PMCID: PMC5516724 DOI: 10.1155/2017/4745252] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 04/07/2017] [Accepted: 05/25/2017] [Indexed: 12/23/2022]
Abstract
Cellular reprogramming is accompanied by a metabolic shift from oxidative phosphorylation (OXPHOS) toward glycolysis. Previous results from our laboratory showed that hypoxia alone is able to reprogram primordial germ cells (PGCs) into pluripotency and that this action is mediated by hypoxia-inducible factor 1 (HIF1). As HIF1 exerts a myriad of actions by upregulating several hundred genes, to ascertain whether the metabolic switch toward glycolysis is solely responsible for reprogramming, PGCs were cultured in the presence of a pyruvate kinase M2 isoform (PKM2) activator, or glycolysis was promoted by manipulating PPARγ. Conversely, OXPHOS was stimulated by inhibiting PDK1 activity in normoxic or in hypoxic conditions. Inhibition or promotion of autophagy and reactive oxygen species (ROS) production was performed to ascertain their role in cell reprogramming. Our results show that a metabolic shift toward glycolysis, autophagy, and mitochondrial inactivation and an early rise in ROS levels are necessary for PGC reprogramming. All of these processes are governed by HIF1/HIF2 balance and strict intermediate Oct4 levels. Histone acetylation plays a role in reprogramming and is observed under all reprogramming conditions. The pluripotent cells thus generated were unable to self-renew, probably due to insufficient Blimp1 downregulation and a lack of Klf4 and cMyc expression.
Collapse
|