101
|
Lee YB, Min JK, Kim JG, Cap KC, Islam R, Hossain AJ, Dogsom O, Hamza A, Mahmud S, Choi DR, Kim YS, Koh YH, Kim HA, Chung WS, Suh SW, Park JB. Multiple functions of pyruvate kinase M2 in various cell types. J Cell Physiol 2021; 237:128-148. [PMID: 34311499 DOI: 10.1002/jcp.30536] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/28/2021] [Accepted: 07/13/2021] [Indexed: 02/06/2023]
Abstract
Glucose metabolism is a mechanism by which energy is produced in form of adenosine triphosphate (ATP) by mitochondria and precursor metabolites are supplied to enable the ultimate enrichment of mature metabolites in the cell. Recently, glycolytic enzymes have been shown to have unconventional but important functions. Among these enzymes, pyruvate kinase M2 (PKM2) plays several roles including having conventional metabolic enzyme activity, and also being a transcriptional regulator and a protein kinase. Compared with the closely related PKM1, PKM2 is highly expressed in cancer cells and embryos, whereas PKM1 is dominant in mature, differentiated cells. Posttranslational modifications such as phosphorylation and acetylation of PKM2 change its cellular functions. In particular, PKM2 can translocate to the nucleus, where it regulates the transcription of many target genes. It is notable that PKM2 also acts as a protein kinase to phosphorylate several substrate proteins. Besides cancer cells and embryonic cells, astrocytes also highly express PKM2, which is crucial for lactate production via expression of lactate dehydrogenase A (LDHA), while mature neurons predominantly express PKM1. The lactate produced in cancer cells promotes tumor progress and that in astrocytes can be supplied to neurons and may act as a major source for neuronal ATP energy production. Thereby, we propose that PKM2 along with its different posttranslational modifications has specific purposes for a variety of cell types, performing unique functions.
Collapse
Affiliation(s)
- Yoon-Beom Lee
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Jung K Min
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Jae-Gyu Kim
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, Republic of Korea.,Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Kim Cuong Cap
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, Republic of Korea.,eLmed Inc. #3419, Hallym University, Chuncheon, Kangwon-do, Republic of Korea.,Institute of Research and Development, Duy Tan University, Danang, Vietnam
| | - Rokibul Islam
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, Republic of Korea.,Department of Biotechnology and Genetic Engineering, Faculty of Biological Science, Islamic University, Kushtia, Bangladesh
| | - Abu J Hossain
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Oyungerel Dogsom
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, Republic of Korea.,Department of Biology, School of Bio-Medicine, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Amir Hamza
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Shohel Mahmud
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, Republic of Korea.,National Institute of Biotechnology, Ganakbari, Savar, Dhaka, Bangladesh
| | - Dae R Choi
- Department of Internal Medicine, Chuncheon Sacred Heart Hospital, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Yong-Sun Kim
- Ilsong Institute of Life Science, Hallym University, Seoul, Republic of Korea
| | - Young-Ho Koh
- Ilsong Institute of Life Science, Hallym University, Seoul, Republic of Korea
| | - Hyun-A Kim
- Department of Internal Medicine, Hallym Sacred Heart Hospital, College of Medicine, Hallym University, Ahnyang, Republic of Korea
| | - Won-Suk Chung
- Department of Biological Science, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Sang W Suh
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Jae-Bong Park
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, Republic of Korea.,Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon, Republic of Korea.,eLmed Inc. #3419, Hallym University, Chuncheon, Kangwon-do, Republic of Korea
| |
Collapse
|
102
|
Rathod B, Chak S, Patel S, Shard A. Tumor pyruvate kinase M2 modulators: a comprehensive account of activators and inhibitors as anticancer agents. RSC Med Chem 2021; 12:1121-1141. [PMID: 34355179 PMCID: PMC8292966 DOI: 10.1039/d1md00045d] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 03/25/2021] [Indexed: 12/16/2022] Open
Abstract
Pyruvate kinase M2 (PKM2) catalyzes the conversion of phosphoenolpyruvate (PEP) to pyruvate. It plays a central role in the metabolic reprogramming of cancer cells and is expressed in most human tumors. It is essential in indiscriminate proliferation, survival, and tackling apoptosis in cancer cells. This positions PKM2 as a hot target in cancer therapy. Despite its well-known structure and several reported modulators targeting PKM2 as activators or inhibitors, a comprehensive review focusing on such modulators is lacking. Herein we summarize modulators of PKM2, the assays used to detect their potential, the preferable tense (T) and relaxed (R) states in which the enzyme resides, lacunae in existing modulators, and several strategies that may lead to effective anticancer drug development targeting PKM2.
Collapse
Affiliation(s)
- Bhagyashri Rathod
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad Opposite Air Force Station Gandhinagar Gujarat 382355 India
| | - Shivam Chak
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad Opposite Air Force Station Gandhinagar Gujarat 382355 India
| | - Sagarkumar Patel
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad Opposite Air Force Station Gandhinagar Gujarat 382355 India
| | - Amit Shard
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad Opposite Air Force Station Gandhinagar Gujarat 382355 India
| |
Collapse
|
103
|
Ye Y, Xu L, Ding H, Wang X, Luo J, Zhang Y, Zen K, Fang Y, Dai C, Wang Y, Zhou Y, Jiang L, Yang J. Pyruvate kinase M2 mediates fibroblast proliferation to promote tubular epithelial cell survival in acute kidney injury. FASEB J 2021; 35:e21706. [PMID: 34160104 DOI: 10.1096/fj.202100040r] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 05/14/2021] [Accepted: 05/17/2021] [Indexed: 12/29/2022]
Abstract
Acute kidney injury (AKI) is a devastating condition with high morbidity and mortality rates. The pathological features of AKI are tubular injury, infiltration of inflammatory cells, and impaired vascular integrity. Pyruvate kinase is the final rate-limiting enzyme in the glycolysis pathway. We previously showed that pyruvate kinase M2 (PKM2) plays an important role in regulating the glycolytic reprogramming of fibroblasts in renal interstitial fibrosis. The present study aimed to determine the role of PKM2 in fibroblast activation during the pathogenesis of AKI. We found increased numbers of S100A4 positive cells expressing PKM2 in renal tissues from mice with AKI induced via folic acid or ischemia/reperfusion (I/R). The loss of PKM2 in fibroblasts impaired fibroblast proliferation and promoted tubular epithelial cell death including apoptosis, necroptosis, and ferroptosis. Mechanistically, fibroblasts produced less hepatocyte growth factor (HGF) in response to a loss of PKM2. Moreover, in two AKI mouse models, fibroblast-specific deletion of PKM2 blocked HGF signal activation and aggravated AKI after it was induced in mice via ischemia or folic acid. Fibroblast proliferation mediated by PKM2 elicits pro-survival signals that repress tubular cell death and may help to prevent AKI progression. Fibroblast activation mediated by PKM2 in AKI suggests that targeting PKM2 expression could be a novel strategy for treating AKI.
Collapse
Affiliation(s)
- Yinyin Ye
- Center for Kidney Disease, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China.,Department of Nephrology, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Lingling Xu
- Center for Kidney Disease, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Hao Ding
- Center for Kidney Disease, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Xiao Wang
- Center for Kidney Disease, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Jing Luo
- Center for Kidney Disease, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Yu Zhang
- Center for Kidney Disease, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Ke Zen
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University Advanced Institute of Life Sciences, Nanjing University, Nanjing, China
| | - Yi Fang
- Center for Kidney Disease, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Chunsun Dai
- Center for Kidney Disease, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Yuwei Wang
- Department of Nephrology, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Yang Zhou
- Center for Kidney Disease, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Lei Jiang
- Center for Kidney Disease, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Junwei Yang
- Center for Kidney Disease, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
104
|
Verma H, Cholia RP, Kaur S, Dhiman M, Mantha AK. A short review on cross-link between pyruvate kinase (PKM2) and Glioblastoma Multiforme. Metab Brain Dis 2021; 36:751-765. [PMID: 33651273 DOI: 10.1007/s11011-021-00690-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 02/10/2021] [Indexed: 12/23/2022]
Abstract
Pyruvate kinase (PK) catalyzes the last irreversible reaction of glycolysis pathway, generating pyruvate and ATP, from Phosphoenol Pyruvate (PEP) and ADP precursors. In mammals, four different tissue-specific isoforms (M1, M2, L and R) of PK exist, which are translated from two genes (PKL and PKR). PKM2 is the highly expressed isoform of PK in cancers, which regulates the aerobic glycolysis via reprogramming cancer cell's metabolic pathways to provide an anabolic advantage to the tumor cells. In addition to the established role of PKM2 in aerobic glycolysis of multiple cancer types, various recent findings have highlighted the non-metabolic functions of PKM2 in brain tumor development. Nuclear PKM2 acts as a co-activator and directly regulates gene transcription. PKM2 dependent transactivation of various oncogenic genes is instrumental in the progression and aggressiveness of Glioblastoma Multiforme (GBM). Also, PKM2 acts as a protein kinase in histone modification which regulates gene expression and tumorigenesis. Ongoing research has explored novel regulatory mechanisms of PKM2 and its association in GBM progression. This review enlists and summarizes the metabolic and non-metabolic roles of PKM2 at the cellular level, and its regulatory function highlights the importance of the nuclear functions of PKM2 in GBM progression, and an emerging role of PKM2 as novel cancer therapeutics.
Collapse
Affiliation(s)
- Harkomal Verma
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Village Ghudda, Bathinda, Punjab, Pin Code: 151 401, India
| | - Ravi P Cholia
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Village Ghudda, Bathinda, Punjab, Pin Code: 151 401, India
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Sharanjot Kaur
- Department of Microbiology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India
| | - Monisha Dhiman
- Department of Microbiology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India
| | - Anil K Mantha
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Village Ghudda, Bathinda, Punjab, Pin Code: 151 401, India.
| |
Collapse
|
105
|
Ushio-Fukai M, Ash D, Nagarkoti S, Belin de Chantemèle EJ, Fulton DJR, Fukai T. Interplay Between Reactive Oxygen/Reactive Nitrogen Species and Metabolism in Vascular Biology and Disease. Antioxid Redox Signal 2021; 34:1319-1354. [PMID: 33899493 PMCID: PMC8418449 DOI: 10.1089/ars.2020.8161] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Reactive oxygen species (ROS; e.g., superoxide [O2•-] and hydrogen peroxide [H2O2]) and reactive nitrogen species (RNS; e.g., nitric oxide [NO•]) at the physiological level function as signaling molecules that mediate many biological responses, including cell proliferation, migration, differentiation, and gene expression. By contrast, excess ROS/RNS, a consequence of dysregulated redox homeostasis, is a hallmark of cardiovascular disease. Accumulating evidence suggests that both ROS and RNS regulate various metabolic pathways and enzymes. Recent studies indicate that cells have mechanisms that fine-tune ROS/RNS levels by tight regulation of metabolic pathways, such as glycolysis and oxidative phosphorylation. The ROS/RNS-mediated inhibition of glycolytic pathways promotes metabolic reprogramming away from glycolytic flux toward the oxidative pentose phosphate pathway to generate nicotinamide adenine dinucleotide phosphate (NADPH) for antioxidant defense. This review summarizes our current knowledge of the mechanisms by which ROS/RNS regulate metabolic enzymes and cellular metabolism and how cellular metabolism influences redox homeostasis and the pathogenesis of disease. A full understanding of these mechanisms will be important for the development of new therapeutic strategies to treat diseases associated with dysregulated redox homeostasis and metabolism. Antioxid. Redox Signal. 34, 1319-1354.
Collapse
Affiliation(s)
- Masuko Ushio-Fukai
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Department of Medicine (Cardiology) and Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Dipankar Ash
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Department of Medicine (Cardiology) and Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Sheela Nagarkoti
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Department of Medicine (Cardiology) and Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Eric J Belin de Chantemèle
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Department of Medicine (Cardiology) and Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - David J R Fulton
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Tohru Fukai
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, USA
| |
Collapse
|
106
|
Sun L, Zhang H, Gao P. Metabolic reprogramming and epigenetic modifications on the path to cancer. Protein Cell 2021; 13:877-919. [PMID: 34050894 PMCID: PMC9243210 DOI: 10.1007/s13238-021-00846-7] [Citation(s) in RCA: 352] [Impact Index Per Article: 88.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 04/02/2021] [Indexed: 02/07/2023] Open
Abstract
Metabolic rewiring and epigenetic remodeling, which are closely linked and reciprocally regulate each other, are among the well-known cancer hallmarks. Recent evidence suggests that many metabolites serve as substrates or cofactors of chromatin-modifying enzymes as a consequence of the translocation or spatial regionalization of enzymes or metabolites. Various metabolic alterations and epigenetic modifications also reportedly drive immune escape or impede immunosurveillance within certain contexts, playing important roles in tumor progression. In this review, we focus on how metabolic reprogramming of tumor cells and immune cells reshapes epigenetic alterations, in particular the acetylation and methylation of histone proteins and DNA. We also discuss other eminent metabolic modifications such as, succinylation, hydroxybutyrylation, and lactylation, and update the current advances in metabolism- and epigenetic modification-based therapeutic prospects in cancer.
Collapse
Affiliation(s)
- Linchong Sun
- Guangzhou First People's Hospital, School of Medicine, Institutes for Life Sciences, South China University of Technology, Guangzhou, 510006, China.
| | - Huafeng Zhang
- The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230027, China. .,CAS Centre for Excellence in Cell and Molecular Biology, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
| | - Ping Gao
- Guangzhou First People's Hospital, School of Medicine, Institutes for Life Sciences, South China University of Technology, Guangzhou, 510006, China. .,School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, China. .,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, 510005, China.
| |
Collapse
|
107
|
Jin X, Zhang W, Wang Y, Liu J, Hao F, Li Y, Tian M, Shu H, Dong J, Feng Y, Wei M. Pyruvate Kinase M2 Promotes the Activation of Dendritic Cells by Enhancing IL-12p35 Expression. Cell Rep 2021; 31:107690. [PMID: 32460017 DOI: 10.1016/j.celrep.2020.107690] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 03/06/2020] [Accepted: 05/01/2020] [Indexed: 12/30/2022] Open
Abstract
Dendritic cells (DCs) play a central role in both innate and adaptive immunity. Emerging evidence has demonstrated metabolic reprogramming during DC activation. However, how DC activation is linked with metabolic reprogramming remains unclear. Here we show that pyruvate kinase M2 (PKM2), the rate-limiting enzyme in the last step of glycolysis, is critical for LPS-induced DC activation. Upon DC activation, JNK signaling stimulated p300 association with PKM2 for the acetylation of lysine 433, a classic posttranslational modification critical for PKM2 destabilization and nuclear re-localization. Subsequently, nuclear PKM2 partnered with c-Rel to enhance Il12p35 expression, which is important for Th1 cell differentiation. Meanwhile, decreased enzymatic activity of PKM2 due to detetramerization facilitated glycolysis and fatty acid synthesis, helping DCs meet their need for biomacromolecules. Together, we provide evidence for metabolic control of DC activation and offer insights into aberrant immune responses due to dysregulated Th1 functions.
Collapse
Affiliation(s)
- Xin Jin
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, Changchun, Jilin, 130024, People's Republic of China
| | - Wenxia Zhang
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, Changchun, Jilin, 130024, People's Republic of China
| | - Yang Wang
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, Changchun, Jilin, 130024, People's Republic of China
| | - Jia Liu
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, Changchun, Jilin, 130024, People's Republic of China
| | - Fengqi Hao
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, Changchun, Jilin, 130024, People's Republic of China
| | - Yunlong Li
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, Changchun, Jilin, 130024, People's Republic of China
| | - Miaomiao Tian
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, Changchun, Jilin, 130024, People's Republic of China
| | - Hengyao Shu
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, Changchun, Jilin, 130024, People's Republic of China
| | - Jiaxin Dong
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, Changchun, Jilin, 130024, People's Republic of China
| | - Yunpeng Feng
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, Changchun, Jilin, 130024, People's Republic of China.
| | - Min Wei
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, Changchun, Jilin, 130024, People's Republic of China.
| |
Collapse
|
108
|
Gao F, Zhang X, Wang S, Zheng L, Sun Y, Wang G, Song Z, Bao Y. TSP50 promotes the Warburg effect and hepatocyte proliferation via regulating PKM2 acetylation. Cell Death Dis 2021; 12:517. [PMID: 34016961 PMCID: PMC8138007 DOI: 10.1038/s41419-021-03782-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 04/30/2021] [Accepted: 04/30/2021] [Indexed: 12/16/2022]
Abstract
Metabolic reprogramming is a hallmark of malignancy. Testes-specific protease 50 (TSP50), a newly identified oncogene, has been shown to play an important role in tumorigenesis. However, its role in tumor cell metabolism remains unclear. To investigate this issue, LC-MS/MS was employed to identify TSP50-binding proteins and pyruvate kinase M2 isoform (PKM2), a known key enzyme of aerobic glycolysis, was identified as a novel binding partner of TSP50. Further studies suggested that TSP50 promoted aerobic glycolysis in HCC cells by maintaining low pyruvate kinase activity of the PKM2. Mechanistically, TSP50 promoted the Warburg effect by increasing PKM2 K433 acetylation level and PKM2 acetylation site (K433R) mutation remarkably abrogated the TSP50-induced aerobic glycolysis, cell proliferation in vitro and tumor formation in vivo. Our findings indicate that TSP50-mediated low PKM2 pyruvate kinase activity is an important determinant for Warburg effect in HCC cells and provide a mechanistic link between TSP50 and tumor metabolism.
Collapse
Affiliation(s)
- Feng Gao
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, China
| | - Xiaojun Zhang
- Research Center of Agriculture and Medicine Gene Engineering of Ministry of Education, Northeast Normal University, Changchun, China
| | - Shuyue Wang
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, China
| | - Lihua Zheng
- Research Center of Agriculture and Medicine Gene Engineering of Ministry of Education, Northeast Normal University, Changchun, China
| | - Ying Sun
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal University, Changchun, Jilin, China
| | - Guannan Wang
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal University, Changchun, Jilin, China
| | - Zhenbo Song
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, China.
| | - Yongli Bao
- Research Center of Agriculture and Medicine Gene Engineering of Ministry of Education, Northeast Normal University, Changchun, China.
| |
Collapse
|
109
|
Oncogenic HPV promotes the expression of the long noncoding RNA lnc-FANCI-2 through E7 and YY1. Proc Natl Acad Sci U S A 2021; 118:2014195118. [PMID: 33436409 DOI: 10.1073/pnas.2014195118] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) play diverse roles in biological processes, but their expression profiles and functions in cervical carcinogenesis remain unknown. By RNA-sequencing (RNA-seq) analyses of 18 clinical specimens and selective validation by RT-qPCR analyses of 72 clinical samples, we provide evidence that, relative to normal cervical tissues, 194 lncRNAs are differentially regulated in high-risk (HR)-HPV infection along with cervical lesion progression. One such lncRNA, lnc-FANCI-2, is extensively characterized because it is expressed from a genomic locus adjacent to the FANCI gene encoding an important DNA repair factor. Both genes are up-regulated in HPV lesions and in in vitro model systems of HR-HPV18 infection. We observe a moderate reciprocal regulation of lnc-FANCI-2 and FANCI in cervical cancer CaSki cells. In these cells, lnc-FANCI-2 is transcribed from two alternative promoters, alternatively spliced, and polyadenylated at one of two alternative poly(A) sites. About 10 copies of lnc-FANCI-2 per cell are detected preferentially in the cytoplasm. Mechanistically, HR-HPVs, but not low-risk (LR)-HPV oncogenes induce lnc-FANCI-2 in primary and immortalized human keratinocytes. The induction is mediated primarily by E7, and to a lesser extent by E6, mostly independent of p53/E6AP and pRb/E2F. We show that YY1 interacts with an E7 CR3 core motif and transactivates the promoter of lnc-FANCI-2 by binding to two critical YY1-binding motifs. Moreover, HPV18 increases YY1 expression by reducing miR-29a, which targets the 3' untranslated region of YY1 mRNA. These data have provided insights into the mechanisms of how HR-HPV infections contribute to cervical carcinogenesis.
Collapse
|
110
|
Role of miRNAs in cervical cancer: A comprehensive novel approach from pathogenesis to therapy. J Gynecol Obstet Hum Reprod 2021; 50:102159. [PMID: 33965650 DOI: 10.1016/j.jogoh.2021.102159] [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: 11/02/2020] [Revised: 04/04/2021] [Accepted: 04/30/2021] [Indexed: 11/20/2022]
Abstract
Human papillomaviruses (HPV) infection is a major causative agent and strongly associated with the development of cervical cancer. Understanding the mechanisms of HPV-induced cervical cancer is extremely useful in therapeutic strategies for primary prevention (HPV vaccines) and secondary prevention (screening and diagnosis of precancerous lesions). However, due to the lack of proper implementation of screening programs in developing countries, cervical cancer is usually diagnosed at advanced stages that result in poor treatment responses. Nearly half of the patients will experience disease recurrence within two years post treatment. Therefore, it is vital to identify new tools for early diagnosis, prognosis, and treatment prediction. MicroRNAs (miRNAs) are small non-coding RNAs, implicated in posttranscriptional regulation of gene expression. Growing evidence has shown that abnormal miRNA expression is associated with cervical cancer progression, metastasis, and influences treatment outcomes. In this review, we provide comprehensive information about miRNA and their potential utility in cervical cancer diagnosis, prognosis, and clinical management to improve patient outcomes.
Collapse
|
111
|
Zhang W, Zhang X, Huang S, Chen J, Ding P, Wang Q, Li L, Lv X, Li L, Zhang P, Zhou D, Wen W, Wang Y, Lei Q, Wu J, Hu W. FOXM1D potentiates PKM2-mediated tumor glycolysis and angiogenesis. Mol Oncol 2021; 15:1466-1485. [PMID: 33314660 PMCID: PMC8096781 DOI: 10.1002/1878-0261.12879] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/16/2020] [Accepted: 12/10/2020] [Indexed: 02/06/2023] Open
Abstract
Tumor growth, especially in the late stage, requires adequate nutrients and rich vasculature, in which PKM2 plays a convergent role. It has been reported that PKM2, together with FOXM1D, is upregulated in late-stage colorectal cancer and associated with metastasis; however, their underlying mechanism for promoting tumor progression remains elusive. Herein, we revealed that FOXM1D potentiates PKM2-mediated glycolysis and angiogenesis through multiple protein-protein interactions. In the presence of FBP, FOXM1D binds to tetrameric PKM2 and assembles a heterooctamer, restraining PKM2 metabolic activity by about a half and thereby promoting aerobic glycolysis. Furthermore, FOXM1D interacts with PKM2 and NF-κB and induces their nuclear translocation with the assistance of the nuclear transporter importin 4. Once in the nucleus, PKM2 and NF-κB complexes subsequently augment VEGFA transcription. The increased VEGFA is secreted extracellularly via exosomes, an event potentiated by the interaction of FOXM1 with VPS11, eventually promoting tumor angiogenesis. Based on these findings, our study provides another insight into the role of PKM2 in the regulation of glycolysis and angiogenesis.
Collapse
Affiliation(s)
- Wei Zhang
- Fudan University Shanghai Cancer Center and Institutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Xin Zhang
- Fudan University Shanghai Cancer Center and Institutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Sheng Huang
- Department of Breast SurgeryBreast Cancer InstituteFudan University Shanghai Cancer CenterShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Jianfeng Chen
- Fudan University Shanghai Cancer Center and Institutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Peipei Ding
- Fudan University Shanghai Cancer Center and Institutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Qi Wang
- Fudan University Shanghai Cancer Center and Institutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Luying Li
- Fudan University Shanghai Cancer Center and Institutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Xinyue Lv
- Fudan University Shanghai Cancer Center and Institutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Ling Li
- Fudan University Shanghai Cancer Center and Institutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Pingzhao Zhang
- Fudan University Shanghai Cancer Center and Institutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Danlei Zhou
- Fudan University Shanghai Cancer Center and Institutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Wenyu Wen
- Fudan University Shanghai Cancer Center and Institutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Yiping Wang
- Fudan University Shanghai Cancer Center and Institutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Qun‐Ying Lei
- Fudan University Shanghai Cancer Center and Institutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Jiong Wu
- Department of Breast SurgeryBreast Cancer InstituteFudan University Shanghai Cancer CenterShanghai Medical CollegeFudan UniversityShanghaiChina
- Key Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterFudan UniversityShanghaiChina
| | - Weiguo Hu
- Fudan University Shanghai Cancer Center and Institutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
- Key Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterFudan UniversityShanghaiChina
| |
Collapse
|
112
|
Ma R, Wu Y, Li S, Yu X. Interplay Between Glucose Metabolism and Chromatin Modifications in Cancer. Front Cell Dev Biol 2021; 9:654337. [PMID: 33987181 PMCID: PMC8110832 DOI: 10.3389/fcell.2021.654337] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/19/2021] [Indexed: 12/13/2022] Open
Abstract
Cancer cells reprogram glucose metabolism to meet their malignant proliferation needs and survival under a variety of stress conditions. The prominent metabolic reprogram is aerobic glycolysis, which can help cells accumulate precursors for biosynthesis of macromolecules. In addition to glycolysis, recent studies show that gluconeogenesis and TCA cycle play important roles in tumorigenesis. Here, we provide a comprehensive review about the role of glycolysis, gluconeogenesis, and TCA cycle in tumorigenesis with an emphasis on revealing the novel functions of the relevant enzymes and metabolites. These functions include regulation of cell metabolism, gene expression, cell apoptosis and autophagy. We also summarize the effect of glucose metabolism on chromatin modifications and how this relationship leads to cancer development. Understanding the link between cancer cell metabolism and chromatin modifications will help develop more effective cancer treatments.
Collapse
Affiliation(s)
- Rui Ma
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Environmental Microbial Technology Center of Hubei, School of Life Sciences, Hubei University, Wuhan, China
| | - Yinsheng Wu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Environmental Microbial Technology Center of Hubei, School of Life Sciences, Hubei University, Wuhan, China
| | - Shanshan Li
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Environmental Microbial Technology Center of Hubei, School of Life Sciences, Hubei University, Wuhan, China.,College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, China
| | - Xilan Yu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Environmental Microbial Technology Center of Hubei, School of Life Sciences, Hubei University, Wuhan, China
| |
Collapse
|
113
|
Pandkar MR, Dhamdhere SG, Shukla S. Oxygen gradient and tumor heterogeneity: The chronicle of a toxic relationship. Biochim Biophys Acta Rev Cancer 2021; 1876:188553. [PMID: 33915221 DOI: 10.1016/j.bbcan.2021.188553] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 04/08/2021] [Accepted: 04/21/2021] [Indexed: 12/21/2022]
Abstract
The commencement of cancer is attributed to one or a few cells that become rogue and attain the property of immortality. The inception of distinct cancer cell clones during the hyperplastic and dysplastic stages of cancer progression is the utimate consequence of the dysregulated cellular pathways and the proliferative potential itself. Furthermore, a critical factor that adds a layer of complexity to this pre-existent intra-tumoral heterogeneity (ITH) is the foundation of an oxygen gradient, that is established due to the improper architecture of the tumor vasculature. Therefore, as a resultant effect, the poorly oxygenated regions thus formed and characterized as hypoxic, promote the emergence of aggressive and treatment-resistant cancer cell clones. The extraordinary property of the hypoxic cancer cells to exist harmoniously with cancerous and non-cancerous cells in the tumor microenvironment (TME) further increases the intricacies of ITH. Here in this review, the pivotal influence of differential oxygen concentrations in shaping the ITH is thoroughly discussed. We also emphasize on the vitality of the interacting networks that govern the overall fate of oxygen gradient-dependent origin of tumor heterogeneity. Additionally, the implications of less-appreciated reverse Warburg effect, a symbiotic metabolic coupling, and the associated epigenetic regulation of rewiring of cancer metabolism in response to oxygen gradients, have been highlighted as critical influencers of ITH.
Collapse
Affiliation(s)
- Madhura R Pandkar
- Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, Madhya Pradesh 462066, India
| | - Shruti G Dhamdhere
- Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, Madhya Pradesh 462066, India
| | - Sanjeev Shukla
- Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, Madhya Pradesh 462066, India.
| |
Collapse
|
114
|
Chen M, Liu H, Li Z, Ming AL, Chen H. Mechanism of PKM2 affecting cancer immunity and metabolism in Tumor Microenvironment. J Cancer 2021; 12:3566-3574. [PMID: 33995634 PMCID: PMC8120184 DOI: 10.7150/jca.54430] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 03/24/2021] [Indexed: 12/24/2022] Open
Abstract
PKM2 is the enzyme that regulates the final rate-limiting step of glycolysis. PKM2 expression can reinforce the utilization of oxygen and synthesis of growth substances in cancer cells by enhancing OXPHOS and the Warburg effect. In cancer immunity, PKM2 can modulate the expression of PD-L1 in M2 macrophage and decrease the amount and activity of CD8+ T cells. This affects cancer cell killing and immune escape sequentially. How PKM2 regulates PD-L1 expression through immunometabolism is summarized. PKM2 builds a bridge between energy metabolism and cancer immunity. The activator and inhibitor of PKM2 both promote the anti-cancer immune response and inhibit cancer growth and metastasis by regulating the metabolism of cancer cells and immune cells in the tumor microenvironment through HIF-1α/PKM2 pathway. This review focuses on the precise role of PKM2 modulating immunometabolism, providing valuable suggestions for further study in this field.
Collapse
Affiliation(s)
- Mengxi Chen
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
| | - Huan Liu
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
| | - Zhang Li
- Department of Pathology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, P. R. China
| | - Alex Lau Ming
- Department of Pathology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, P. R. China
| | - Honglei Chen
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
- Department of Pathology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, P. R. China
| |
Collapse
|
115
|
Electrophilic Natural Products as Drug Discovery Tools. Trends Pharmacol Sci 2021; 42:434-447. [PMID: 33902949 DOI: 10.1016/j.tips.2021.03.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 12/22/2022]
Abstract
Electrophilic natural products (ENPs) are a rich source of bioactive molecules with tremendous therapeutic potential. While their synthetic complexity may hinder their direct use as therapeutics, they represent tools for elucidation of suitable molecular targets and serve as inspiration for the design of simplified synthetic counterparts. Here, we review the recent use of various activity-based protein profiling methods to uncover molecular targets of ENPs. Beyond target identification, these examples also showcase further development of synthetic ligands from natural product starting points. Two examples demonstrate how ENPs can progress the emerging fields of targeted protein degradation and molecular glues. Though challenges still remain in the synthesis of ENP-based probes, and in their synthetic simplification, their potential for discovery of novel mechanisms of action makes it well worth the effort.
Collapse
|
116
|
Nandi S, Razzaghi M, Srivastava D, Dey M. Structural basis for allosteric regulation of pyruvate kinase M2 by phosphorylation and acetylation. J Biol Chem 2021; 295:17425-17440. [PMID: 33453989 DOI: 10.1074/jbc.ra120.015800] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/18/2020] [Indexed: 01/01/2023] Open
Abstract
Pyruvate kinase muscle isoform 2 (PKM2) is a key glycolytic enzyme and transcriptional coactivator and is critical for tumor metabolism. In cancer cells, native tetrameric PKM2 is phosphorylated or acetylated, which initiates a switch to a dimeric/monomeric form that translocates into the nucleus, causing oncogene transcription. However, it is not known how these post-translational modifications (PTMs) disrupt the oligomeric state of PKM2. We explored this question via crystallographic and biophysical analyses of PKM2 mutants containing residues that mimic phosphorylation and acetylation. We find that the PTMs elicit major structural reorganization of the fructose 1,6-bisphosphate (FBP), an allosteric activator, binding site, impacting the interaction with FBP and causing a disruption in oligomerization. To gain insight into how these modifications might cause unique outcomes in cancer cells, we examined the impact of increasing the intracellular pH (pHi) from ∼7.1 (in normal cells) to ∼7.5 (in cancer cells). Biochemical studies of WT PKM2 (wtPKM2) and the two mimetic variants demonstrated that the activity decreases as the pH is increased from 7.0 to 8.0, and wtPKM2 is optimally active and amenable to FBP-mediated allosteric regulation at pHi 7.5. However, the PTM mimetics exist as a mixture of tetramer and dimer, indicating that physiologically dimeric fraction is important and might be necessary for the modified PKM2 to translocate into the nucleus. Thus, our findings provide insight into how PTMs and pH regulate PKM2 and offer a broader understanding of its intricate allosteric regulation mechanism by phosphorylation or acetylation.
Collapse
Affiliation(s)
- Suparno Nandi
- Department of Chemistry, University of Iowa, Iowa City, Iowa, USA
| | | | | | - Mishtu Dey
- Department of Chemistry, University of Iowa, Iowa City, Iowa, USA.
| |
Collapse
|
117
|
Coassolo S, Davidson G, Negroni L, Gambi G, Daujat S, Romier C, Davidson I. Citrullination of pyruvate kinase M2 by PADI1 and PADI3 regulates glycolysis and cancer cell proliferation. Nat Commun 2021; 12:1718. [PMID: 33741961 PMCID: PMC7979715 DOI: 10.1038/s41467-021-21960-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 02/23/2021] [Indexed: 12/11/2022] Open
Abstract
Chromodomain helicase DNA binding protein 4 (CHD4) is an ATPase subunit of the Nucleosome Remodelling and Deacetylation (NuRD) complex that regulates gene expression. CHD4 is essential for growth of multiple patient derived melanoma xenografts and for breast cancer. Here we show that CHD4 regulates expression of PADI1 (Protein Arginine Deiminase 1) and PADI3 in multiple cancer cell types modulating citrullination of arginine residues of the allosterically-regulated glycolytic enzyme pyruvate kinase M2 (PKM2). Citrullination of PKM2 R106 reprogrammes cross-talk between PKM2 ligands lowering its sensitivity to the inhibitors Tryptophan, Alanine and Phenylalanine and promoting activation by Serine. Citrullination thus bypasses normal physiological regulation by low Serine levels to promote excessive glycolysis and reduced cell proliferation. We further show that PADI1 and PADI3 expression is up-regulated by hypoxia where PKM2 citrullination contributes to increased glycolysis. We provide insight as to how conversion of arginines to citrulline impacts key interactions within PKM2 that act in concert to reprogramme its activity as an additional mechanism regulating this important enzyme.
Collapse
Affiliation(s)
- Sébastien Coassolo
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Equipe Labélise Ligue Contre le Cancer, Illkirch, France
- Centre National de la Recherche Scientifique, Paris, France
- Institut National de la Santé et de la Recherche Médicale, Paris, France
- Université de Strasbourg, Strasbourg, France
- Discovery Oncology, Genentech, South San Francisco, CA, USA
| | - Guillaume Davidson
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Equipe Labélise Ligue Contre le Cancer, Illkirch, France
- Centre National de la Recherche Scientifique, Paris, France
- Institut National de la Santé et de la Recherche Médicale, Paris, France
- Université de Strasbourg, Strasbourg, France
| | - Luc Negroni
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Equipe Labélise Ligue Contre le Cancer, Illkirch, France
- Centre National de la Recherche Scientifique, Paris, France
- Institut National de la Santé et de la Recherche Médicale, Paris, France
- Université de Strasbourg, Strasbourg, France
| | - Giovanni Gambi
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Equipe Labélise Ligue Contre le Cancer, Illkirch, France
- Centre National de la Recherche Scientifique, Paris, France
- Institut National de la Santé et de la Recherche Médicale, Paris, France
- Université de Strasbourg, Strasbourg, France
| | - Sylvain Daujat
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Equipe Labélise Ligue Contre le Cancer, Illkirch, France
- Centre National de la Recherche Scientifique, Paris, France
- Institut National de la Santé et de la Recherche Médicale, Paris, France
- Université de Strasbourg, Strasbourg, France
- Biotechnology and Cell Signaling, CNRS UMR7242, 300 Bd Sébastien Brandt, Illkirch, France
| | - Christophe Romier
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Equipe Labélise Ligue Contre le Cancer, Illkirch, France
- Centre National de la Recherche Scientifique, Paris, France
- Institut National de la Santé et de la Recherche Médicale, Paris, France
- Université de Strasbourg, Strasbourg, France
| | - Irwin Davidson
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Equipe Labélise Ligue Contre le Cancer, Illkirch, France.
- Centre National de la Recherche Scientifique, Paris, France.
- Institut National de la Santé et de la Recherche Médicale, Paris, France.
- Université de Strasbourg, Strasbourg, France.
| |
Collapse
|
118
|
Das Gupta K, Shakespear MR, Curson JEB, Murthy AMV, Iyer A, Hodson MP, Ramnath D, Tillu VA, von Pein JB, Reid RC, Tunny K, Hohenhaus DM, Moradi SV, Kelly GM, Kobayashi T, Gunter JH, Stevenson AJ, Xu W, Luo L, Jones A, Johnston WA, Blumenthal A, Alexandrov K, Collins BM, Stow JL, Fairlie DP, Sweet MJ. Class IIa Histone Deacetylases Drive Toll-like Receptor-Inducible Glycolysis and Macrophage Inflammatory Responses via Pyruvate Kinase M2. Cell Rep 2021; 30:2712-2728.e8. [PMID: 32101747 DOI: 10.1016/j.celrep.2020.02.007] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 09/30/2019] [Accepted: 02/03/2020] [Indexed: 12/14/2022] Open
Abstract
Histone deacetylases (HDACs) drive innate immune cell-mediated inflammation. Here we identify class IIa HDACs as key molecular links between Toll-like receptor (TLR)-inducible aerobic glycolysis and macrophage inflammatory responses. A proteomic screen identified the glycolytic enzyme pyruvate kinase M isoform 2 (Pkm2) as a partner of proinflammatory Hdac7 in murine macrophages. Myeloid-specific Hdac7 overexpression in transgenic mice amplifies lipopolysaccharide (LPS)-inducible lactate and promotes a glycolysis-associated inflammatory signature. Conversely, pharmacological or genetic targeting of Hdac7 and other class IIa HDACs attenuates LPS-inducible glycolysis and accompanying inflammatory responses in macrophages. We show that an Hdac7-Pkm2 complex acts as an immunometabolism signaling hub, whereby Pkm2 deacetylation at lysine 433 licenses its proinflammatory functions. Disrupting this complex suppresses inflammatory responses in vitro and in vivo. Class IIa HDACs are thus pivotal intermediates connecting TLR-inducible glycolysis to inflammation via Pkm2.
Collapse
Affiliation(s)
- Kaustav Das Gupta
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, Queensland 4072, Australia; IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Melanie R Shakespear
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, Queensland 4072, Australia; IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - James E B Curson
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, Queensland 4072, Australia; IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Ambika M V Murthy
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, Queensland 4072, Australia; IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Abishek Iyer
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, Queensland 4072, Australia; IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland 4072, Australia; ARC Centre of Excellence in Advanced Molecular Imaging, IMB, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Mark P Hodson
- School of Pharmacy, The University of Queensland, Brisbane, Queensland 4072, Australia; Metabolomics Australia, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia; Victor Chang Cardiac Research Institute, Sydney, New South Wales 2010, Australia
| | - Divya Ramnath
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, Queensland 4072, Australia; IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Vikas A Tillu
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Jessica B von Pein
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, Queensland 4072, Australia; IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Robert C Reid
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, Queensland 4072, Australia; IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland 4072, Australia; ARC Centre of Excellence in Advanced Molecular Imaging, IMB, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Kathryn Tunny
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Daniel M Hohenhaus
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Shayli Varasteh Moradi
- CSIRO-QUT Synthetic Biology Alliance, Centre for Tropical Crops and Biocommodities, Queensland University of Technology (QUT), Gardens Point Campus, Brisbane, Queensland 4000, Australia
| | - Gregory M Kelly
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Takumi Kobayashi
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Jennifer H Gunter
- Australian Prostate Cancer Research Centre-Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Translational Research Institute, Queensland University of Technology (QUT), Brisbane, Queensland 4102, Australia
| | - Alexander J Stevenson
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Weijun Xu
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, Queensland 4072, Australia; IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland 4072, Australia; ARC Centre of Excellence in Advanced Molecular Imaging, IMB, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Lin Luo
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, Queensland 4072, Australia; IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Alun Jones
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Wayne A Johnston
- CSIRO-QUT Synthetic Biology Alliance, Centre for Tropical Crops and Biocommodities, Queensland University of Technology (QUT), Gardens Point Campus, Brisbane, Queensland 4000, Australia
| | - Antje Blumenthal
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Kirill Alexandrov
- CSIRO-QUT Synthetic Biology Alliance, Centre for Tropical Crops and Biocommodities, Queensland University of Technology (QUT), Gardens Point Campus, Brisbane, Queensland 4000, Australia
| | - Brett M Collins
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Jennifer L Stow
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, Queensland 4072, Australia; IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - David P Fairlie
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, Queensland 4072, Australia; IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland 4072, Australia; ARC Centre of Excellence in Advanced Molecular Imaging, IMB, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Matthew J Sweet
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, Queensland 4072, Australia; IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland 4072, Australia.
| |
Collapse
|
119
|
Damasceno LEA, Prado DS, Veras FP, Fonseca MM, Toller-Kawahisa JE, Rosa MH, Públio GA, Martins TV, Ramalho FS, Waisman A, Cunha FQ, Cunha TM, Alves-Filho JC. PKM2 promotes Th17 cell differentiation and autoimmune inflammation by fine-tuning STAT3 activation. J Exp Med 2021; 217:151965. [PMID: 32697823 PMCID: PMC7537396 DOI: 10.1084/jem.20190613] [Citation(s) in RCA: 165] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 05/28/2019] [Accepted: 05/28/2020] [Indexed: 01/15/2023] Open
Abstract
Th17 cell differentiation and pathogenicity depend on metabolic reprogramming inducing shifts toward glycolysis. Here, we show that the pyruvate kinase M2 (PKM2), a glycolytic enzyme required for cancer cell proliferation and tumor progression, is a key factor mediating Th17 cell differentiation and autoimmune inflammation. We found that PKM2 is highly expressed throughout the differentiation of Th17 cells in vitro and during experimental autoimmune encephalomyelitis (EAE) development. Strikingly, PKM2 is not required for the metabolic reprogramming and proliferative capacity of Th17 cells. However, T cell-specific PKM2 deletion impairs Th17 cell differentiation and ameliorates symptoms of EAE by decreasing Th17 cell-mediated inflammation and demyelination. Mechanistically, PKM2 translocates into the nucleus and interacts with STAT3, enhancing its activation and thereby increasing Th17 cell differentiation. Thus, PKM2 acts as a critical nonmetabolic regulator that fine-tunes Th17 cell differentiation and function in autoimmune-mediated inflammation.
Collapse
Affiliation(s)
- Luis Eduardo Alves Damasceno
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.,Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Douglas Silva Prado
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.,Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Flavio Protasio Veras
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.,Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Miriam M Fonseca
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.,Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Juliana E Toller-Kawahisa
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.,Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Marcos Henrique Rosa
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.,Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Gabriel Azevedo Públio
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.,Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Timna Varela Martins
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.,Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Fernando S Ramalho
- Department of Pathology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Fernando Queiroz Cunha
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.,Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Thiago Mattar Cunha
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.,Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - José Carlos Alves-Filho
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.,Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| |
Collapse
|
120
|
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
|
121
|
Abstract
Targeting glycolysis in T helper 17 (Th17) cells presents an attractive
opportunity to treat Th17 cell-mediated autoimmune diseases such as multiple
sclerosis (MS). Pyruvate kinase isoform 2 (PKM2) is a glycolytic enzyme
expressed in T cells infiltrating the central nervous system in a mouse model of
MS, suggesting PKM2 modulation could provide a new avenue for MS therapeutics.
In a recent article in Science Signaling, Seki et al. show that
pharmacological modulation of PKM2 alters but does not ameliorate disease in a
mouse model of MS. These results warrant further consideration of PKM2
modulators to treat Th17 cell-mediated autoimmunity.
Collapse
|
122
|
Hao L, Park J, Jang HY, Bae EJ, Park BH. Inhibiting Protein Kinase Activity of Pyruvate Kinase M2 by SIRT2 Deacetylase Attenuates Psoriasis. J Invest Dermatol 2021; 141:355-363.e6. [DOI: 10.1016/j.jid.2020.06.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 06/03/2020] [Accepted: 06/17/2020] [Indexed: 01/16/2023]
|
123
|
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: 27] [Impact Index Per Article: 6.8] [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
|
124
|
Discovery of Functional Alternatively Spliced PKM Transcripts in Human Cancers. Cancers (Basel) 2021; 13:cancers13020348. [PMID: 33478099 PMCID: PMC7835739 DOI: 10.3390/cancers13020348] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/15/2021] [Accepted: 01/17/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Pyruvate kinase muscle type (PKM) is a key enzyme in glycolysis and is a mediator of the Warburg effect in tumors. The association of PKM with survival of cancer patients is controversial. In this study, we investigated the associations of the alternatively spliced transcripts of PKM with cancer patients’ survival outcomes and explained the conflicts in previous studies. We discovered three poorly studied alternatively spliced PKM transcripts that exhibited opposite prognostic indications in different human cancers based on integrative systems analysis. We also detected their protein products and explored their potential biological functions based on in-vitro experiments. Our analysis demonstrated that alternatively spliced transcripts of not only PKM but also other genes should be considered in cancer studies, since it may enable the discovery and targeting of the right protein product for development of the efficient treatment strategies. Abstract Pyruvate kinase muscle type (PKM) is a key enzyme in glycolysis and plays an important oncological role in cancer. However, the association of PKM expression and the survival outcome of patients with different cancers is controversial. We employed systems biology methods to reveal prognostic value and potential biological functions of PKM transcripts in different human cancers. Protein products of transcripts were shown and detected by western blot and mass spectrometry analysis. We focused on different transcripts of PKM and investigated the associations between their mRNA expression and the clinical survival of the patients in 25 different cancers. We find that the transcripts encoding PKM2 and three previously unstudied transcripts, namely ENST00000389093, ENST00000568883, and ENST00000561609, exhibited opposite prognostic indications in different cancers. Moreover, we validated the prognostic effect of these transcripts in an independent kidney cancer cohort. Finally, we revealed that ENST00000389093 and ENST00000568883 possess pyruvate kinase enzymatic activity and may have functional roles in metabolism, cell invasion, and hypoxia response in cancer cells. Our study provided a potential explanation to the controversial prognostic indication of PKM, and could invoke future studies focusing on revealing the biological and oncological roles of these alternative spliced variants of PKM.
Collapse
|
125
|
Xu D, Shao F, Bian X, Meng Y, Liang T, Lu Z. The Evolving Landscape of Noncanonical Functions of Metabolic Enzymes in Cancer and Other Pathologies. Cell Metab 2021; 33:33-50. [PMID: 33406403 DOI: 10.1016/j.cmet.2020.12.015] [Citation(s) in RCA: 138] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Key pathological, including oncogenic, signaling pathways regulate the canonical functions of metabolic enzymes that serve the cellular metabolic needs. Importantly, these signaling pathways also confer a large number of metabolic enzymes to have noncanonical or nonmetabolic functions that are referred to as "moonlighting" functions. In this review, we highlight how aberrantly regulated metabolic enzymes with such activities play critical roles in the governing of a wide spectrum of instrumental cellular activities, including gene expression, cell-cycle progression, DNA repair, cell proliferation, survival, apoptosis, and tumor microenvironment remodeling, thereby promoting the pathologic progression of disease, including cancer.
Collapse
Affiliation(s)
- Daqian Xu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China
| | - Fei Shao
- The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, Shandong 266003, China
| | - Xueli Bian
- The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, Shandong 266003, China
| | - Ying Meng
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China
| | - Tingbo Liang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China
| | - Zhimin Lu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China; Zhejiang University Cancer Center, Hangzhou 310029, China.
| |
Collapse
|
126
|
Proteins moonlighting in tumor metabolism and epigenetics. Front Med 2021; 15:383-403. [PMID: 33387254 DOI: 10.1007/s11684-020-0818-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 07/27/2020] [Indexed: 02/07/2023]
Abstract
Cancer development is a complicated process controlled by the interplay of multiple signaling pathways and restrained by oxygen and nutrient accessibility in the tumor microenvironment. High plasticity in using diverse nutrients to adapt to metabolic stress is one of the hallmarks of cancer cells. To respond to nutrient stress and to meet the requirements for rapid cell proliferation, cancer cells reprogram metabolic pathways to take up more glucose and coordinate the production of energy and intermediates for biosynthesis. Such actions involve gene expression and activity regulation by the moonlighting function of oncoproteins and metabolic enzymes. The signal - moonlighting protein - metabolism axis facilitates the adaptation of tumor cells under varying environment conditions and can be therapeutically targeted for cancer treatment.
Collapse
|
127
|
Chen S, Luo L, Chen H, He C. The Current State of Research Regarding the Role of Non-Coding RNAs in Cutaneous Squamous Cell Carcinoma. Onco Targets Ther 2020; 13:13151-13158. [PMID: 33380805 PMCID: PMC7767711 DOI: 10.2147/ott.s271346] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 10/26/2020] [Indexed: 12/18/2022] Open
Abstract
Skin cancers, including those of both both melanoma and non-melanoma subtypes, remain among the most common forms of human cancer. Non-melanoma skin cancers are typically further differentiated into the basal cell carcinoma and cutaneous squamous cell carcinoma (cSCC) categories. Current approaches to diagnosing and treating cSCC remain unsatisfactory, and the prognosis for patients with this disease is relatively poor. Recent advances in high-throughput sequencing have led to an increasingly robust understanding of the diversity of non-coding RNAs (ncRNAs) expressed in both physiological and pathological contexts. These ncRNAs include microRNAs, long ncRNAs, and circular RNAs, all of which have been found to play key functional roles and/or to have value as diagnostic biomarkers or therapeutic targets in a range of different disease contexts. The number of ncRNAs associated with cSCC continues to rise, and as such, there is clear value in comprehensively reviewing the functional roles of these molecules in this form of cancer in order to highlight future avenues for research and clinical development.
Collapse
Affiliation(s)
- Shuang Chen
- Department of Dermatology, No.1 Hospital of China Medical University, Key Laboratory of Immunodermatology, Shenyang, Liaoning 110001, People's Republic of China
| | - Limin Luo
- Department of Dermatology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, People's Republic of China
| | - Hongduo Chen
- Department of Dermatology, No.1 Hospital of China Medical University, Key Laboratory of Immunodermatology, Shenyang, Liaoning 110001, People's Republic of China
| | - Chundi He
- Department of Dermatology, No.1 Hospital of China Medical University, Key Laboratory of Immunodermatology, Shenyang, Liaoning 110001, People's Republic of China
| |
Collapse
|
128
|
Zhang R, Shen M, Wu C, Chen Y, Lu J, Li J, Zhao L, Meng H, Zhou X, Huang G, Zhao X, Liu J. HDAC8-dependent deacetylation of PKM2 directs nuclear localization and glycolysis to promote proliferation in hepatocellular carcinoma. Cell Death Dis 2020; 11:1036. [PMID: 33279948 PMCID: PMC7719180 DOI: 10.1038/s41419-020-03212-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 11/04/2020] [Accepted: 11/06/2020] [Indexed: 01/11/2023]
Abstract
Pyruvate kinase M2 (PKM2) is not only a key rate-limiting enzyme that guides glycolysis, but also acts as a non-metabolic protein in regulating gene transcription. In recent years, a series of studies have confirmed that post-translational modification has become an important mechanism for regulating the function of PKM2, which in turn affects tumorigenesis. In this study, we found that K62 residues were deacetylated, which is related to the prognosis of HCC. Further studies indicate that HDAC8 binds and deacetylates the K62 residue of PKM2. Mechanistically, K62 deacetylation facilitate PKM2 transport into the nucleus and bind β-catenin, thereby promoting CCND1 gene transcription and cell cycle progression. In addition, the deacetylation of K62 affects the enzyme activity of PKM2 and the flux of glucose metabolism. Therefore, these results suggest that HDAC8 / PKM2 signaling may become a new target for the treatment of HCC.
Collapse
Affiliation(s)
- Ruixue Zhang
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Mengqin Shen
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Chunhua Wu
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yumei Chen
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Jiani Lu
- Division of Physical Therapy Education, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jiajin Li
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Li Zhao
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Huannan Meng
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Xiang Zhou
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Gang Huang
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xiaoping Zhao
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Jianjun Liu
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
- Division of Physical Therapy Education, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
129
|
Wang X, Liang C, Yao X, Yang RH, Zhang ZS, Liu FY, Li WQ, Pei SH, Ma J, Xie SQ, Fang D. PKM2-Induced the Phosphorylation of Histone H3 Contributes to EGF-Mediated PD-L1 Transcription in HCC. Front Pharmacol 2020; 11:577108. [PMID: 33324209 PMCID: PMC7725877 DOI: 10.3389/fphar.2020.577108] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 10/19/2020] [Indexed: 12/19/2022] Open
Abstract
High expression of programmed death-ligand-1 (PD-L1) in hepatocellular carcinoma (HCC) cells usually inhibits the proliferation and functions of T cells, leading to immune suppression in tumor microenvironment. However, very little has been described regarding the mechanism of PD-L1 overexpression in HCC cells. In the present study, we found epidermal growth factor (EGF) stimulation promoted the expression of PD-L1 mRNA and protein in HCC cells. Inhibition of epidermal growth factor receptor (EGFR) could reverse EGF-induced the expression of PD-L1 mRNA and protein. Subsequently, we also observed that the phosphorylation level of Pyruvate kinase isoform M2 (PKM2) at Ser37 site was also increased in response to EGF stimulation. Expression of a phosphorylation-mimic PKM2 S37D mutant stimulated PD-L1 expression as well as H3-Thr11 phosphorylation in HCC cells, while inhibition of PKM2 significantly blocked EGF-induced PD-L1 expression and H3-Thr11 phosphorylation. Furthermore, mutation of Thr11 of histone H3 into alanine abrogated EGF-induced mRNA and protein expression of PD-L1, Chromatin immunoprecipitation (ChIP) assay also suggested that EGF treatment resulted in enhanced H3-Thr11 phosphorylation at the PD-L1 promoter. In a diethylnitrosamine (DEN)-induced rat model of HCC, we found that the expression of phosphorylated EGFR, PKM2 nuclear expression, H3-Thr11 phosphorylation as well as PD-L1 mRNA and protein was higher in the livers than that in normal rat livers. Taken together, our study suggested that PKM2-dependent histone H3-Thr11 phosphorylation was crucial for EGF-induced PD-L1 expression at transcriptional level in HCC. These findings may provide an alternative target for the treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Xiao Wang
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng, China
| | - Chao Liang
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng, China
| | - Xin Yao
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng, China
| | - Ruo-Han Yang
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng, China
| | - Zhan-Sheng Zhang
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng, China
| | - Fan-Ye Liu
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng, China
| | - Wen-Qi Li
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng, China
| | - Shu-Hua Pei
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng, China
| | - Jing Ma
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng, China
| | - Song-Qiang Xie
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng, China.,Institute of Chemical Biology, School of Pharmacy, Henan University, Kaifeng, China
| | - Dong Fang
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng, China
| |
Collapse
|
130
|
Niu J, Li W, Liang C, Wang X, Yao X, Yang RH, Zhang ZS, Liu HF, Liu FY, Pei SH, Li WQ, Sun H, Fang D, Xie SQ. EGF promotes
DKK1
transcription in hepatocellular carcinoma by enhancing the phosphorylation and acetylation of histone H3. Sci Signal 2020; 13:13/657/eabb5727. [DOI: 10.1126/scisignal.abb5727] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Jie Niu
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
| | - Wei Li
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
| | - Chao Liang
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
| | - Xiao Wang
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
| | - Xin Yao
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
| | - Ruo-Han Yang
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
| | - Zhan-Sheng Zhang
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
| | - Han-Fang Liu
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
| | - Fan-Ye Liu
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
| | - Shu-Hua Pei
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
| | - Wen-Qi Li
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
| | - Hua Sun
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
| | - Dong Fang
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
| | - Song-Qiang Xie
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
| |
Collapse
|
131
|
Wei X, Jin XH, Meng XW, Hua J, Ji FH, Wang LN, Yang JP. Platelet-rich plasma improves chronic inflammatory pain by inhibiting PKM2-mediated aerobic glycolysis in astrocytes. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1456. [PMID: 33313201 PMCID: PMC7723564 DOI: 10.21037/atm-20-6502] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Astrocytes are highly glycolytic cells that play a crucial role in chronic pain. Recently it has been found that inflammation and metabolism are related to the inflammatory stimuli closely that cause cellular metabolic changes. Pyruvate kinase M2 (PKM2) is a critical metabolic kinase in aerobic glycolysis or the Warburg effect. Besides, it also plays a crucial role in cell proliferation and signal transduction, but its role in astrocytes is still unclear. Methods The chronic inflammatory pain model was set up by intraplantar injection of complete Freund’s adjuvant (CFA) in Sprague Dawley (SD) rats as well as the cell model was constructed by lipopolysaccharide-treated primary astrocytes. Von Frey filament stimulation was used to continuously observe the changes of pain behavior in rats after modeling. Then, immunofluorescence staining and Western blot tests were used to observe the expression levels of glial fibrillary acidic protein (GFAP), pyruvate kinase (PKM2), signal transducers and activators of transcription 3 (STAT3) and high mobility group box-1 protein (HMGB1). After that, specific kits measured lactate contents. Finally, we observed the platelet-rich plasma’s (PRP) effect on mechanical hyperalgesia in rats with inflammatory pain induced by CFA and its effect on related signal molecules. Results We found that in the CFA-induced inflammatory pain model, astrocytes were significantly activated, GFAP was increased, PKM2 was significantly up-regulated, and the glycolytic product lactate was increased. Also, intrathecal injection of PRP increased the pain threshold, inhibited the activation of astrocytes, and decreased the expression of PKM2 and aerobic glycolysis; in LPS-activated primary astrocytes as an in vitro model, we found PKM2 translocation activationSTAT3 signaling resulted in sustained activation of astrocyte marker GFAP, and the expression level and localization of p-STAT3 were correlated with PKM2. PRP could inhibit the activation of astrocytes, reduce the expression of PKM2 and the expression levels of glycolysis and GFAP, GLUT1, and p-STAT3 in astrocytes. Conclusions Our findings suggest PKM2 not only plays a glycolytic role in astrocytes, but also plays a crucial role in astrocyte-activated signaling pathways, and PRP attenuates CFA induced inflammatory pain by inhibiting aerobic glycolysis in astrocytes, providing a new therapeutic target for the treatment of inflammatory pain.
Collapse
Affiliation(s)
- Xiang Wei
- Department of Anesthesiology and Pain Management, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiao-Hong Jin
- Department of Anesthesiology and Pain Management, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiao-Wen Meng
- Department of Anesthesiology and Pain Management, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jie Hua
- Department of Anesthesiology and Pain Management, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Fu-Hai Ji
- Department of Anesthesiology and Pain Management, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Li-Na Wang
- Department of Anesthesiology and Pain Management, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jian-Ping Yang
- Department of Anesthesiology and Pain Management, the First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
132
|
Zhang L, Yang J, Luo Y, Liu F, Yuan Y, Zhuang S. A p53/lnc-Ip53 Negative Feedback Loop Regulates Tumor Growth and Chemoresistance. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2001364. [PMID: 33173727 PMCID: PMC7610266 DOI: 10.1002/advs.202001364] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/08/2020] [Indexed: 06/03/2023]
Abstract
Acetylation is a critical mechanism to modulate tumor-suppressive activity of p53, but the causative roles of long non-coding RNAs (lncRNAs) in p53 acetylation and their biological significance remain unexplored. Here, lncRNA LOC100294145 is discovered to be transactivated by p53 and is thus designated as lnc-Ip53 for lncRNA induced by p53. Furthermore, lnc-Ip53 impedes p53 acetylation by interacting with histone deacetylase 1 (HDAC1) and E1A binding protein p300 (p300) to prevent HDAC1 degradation and attenuate p300 activity, resulting in abrogation of p53 activity and subsequent cell proliferation and apoptosis resistance. Mouse xenograft models reveal that lnc-Ip53 promotes tumor growth and chemoresistance in vivo, which is attenuated by an HDAC inhibitor. Silencing lnc-Ip53 inhibits the growth of xenografts with wild-type p53, but not those expressing acetylation-resistant p53. Consistently, lnc-Ip53 is upregulated in multiple cancer types, including hepatocellular carcinoma (HCC). High levels of lnc-Ip53 is associated with low levels of acetylated p53 in human HCC and mouse xenografts, and is also correlated with poor survival of HCC patients. These findings identify a novel p53/lnc-Ip53 negative feedback loop in cells and indicate that abnormal upregulation of lnc-Ip53 represents an important mechanism to inhibit p53 acetylation/activity and thereby promote tumor growth and chemoresistance, which may be exploited for anticancer therapy.
Collapse
Affiliation(s)
- Li‐Zhen Zhang
- MOE Key Laboratory of Gene Function and RegulationSchool of Life SciencesCollaborative Innovation Center for Cancer MedicineSun Yat‐sen UniversityGuangzhou510275China
| | - Jin‐E Yang
- MOE Key Laboratory of Gene Function and RegulationSchool of Life SciencesCollaborative Innovation Center for Cancer MedicineSun Yat‐sen UniversityGuangzhou510275China
| | - Yu‐Wei Luo
- MOE Key Laboratory of Gene Function and RegulationSchool of Life SciencesCollaborative Innovation Center for Cancer MedicineSun Yat‐sen UniversityGuangzhou510275China
| | - Feng‐Ting Liu
- MOE Key Laboratory of Gene Function and RegulationSchool of Life SciencesCollaborative Innovation Center for Cancer MedicineSun Yat‐sen UniversityGuangzhou510275China
| | - Yun‐Fei Yuan
- Department of Hepatobilliary OncologyCancer CenterSun Yat‐sen UniversityGuangzhou510060China
| | - Shi‐Mei Zhuang
- MOE Key Laboratory of Gene Function and RegulationSchool of Life SciencesCollaborative Innovation Center for Cancer MedicineSun Yat‐sen UniversityGuangzhou510275China
- Key Laboratory of Liver Disease of Guangdong ProvinceThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| |
Collapse
|
133
|
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: 56] [Impact Index Per Article: 11.2] [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
|
134
|
Wang C, Xiao Y, Lao M, Wang J, Xu S, Li R, Xu X, Kuang Y, Shi M, Zou Y, Wang Q, Liang L, Zheng SG, Xu H. Increased SUMO-activating enzyme SAE1/UBA2 promotes glycolysis and pathogenic behavior of rheumatoid fibroblast-like synoviocytes. JCI Insight 2020; 5:135935. [PMID: 32938830 PMCID: PMC7526534 DOI: 10.1172/jci.insight.135935] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 08/12/2020] [Indexed: 12/13/2022] Open
Abstract
Fibroblast-like synoviocytes (FLSs) are critical to joint inflammation and destruction in rheumatoid arthritis (RA). Increased glycolysis in RA FLSs contributes to persistent joint damage. SUMOylation, a posttranslational modification of proteins, plays an important role in initiation and development of many diseases. However, the role of small ubiquitin-like modifier–activating (SUMO-activating) enzyme 1 (SAE1)/ubiquitin like modifier activating enzyme 2 (UBA2) in regulating the pathogenic FLS behaviors is unknown. Here, we found an increased expression of SAE1 and UBA2 in FLSs and synovial tissues from patients with RA. SAE1 or UBA2 knockdown by siRNA and treatment with GA, an inhibitor of SAE1/UBA2-mediated SUMOylation, resulted in reduced glycolysis, aggressive phenotype, and inflammation. SAE1/UBA2-mediated SUMOylation of pyruvate kinase M2 (PKM2) promoted its phosphorylation and nuclear translocation and decreased PK activity. Moreover, inhibition of PKM2 phosphorylation increased PK activity and suppressed glycolysis, aggressive phenotype, and inflammation. We further demonstrated that STAT5A mediated SUMOylated PKM2-induced glycolysis and biological behaviors. Interestingly, GA treatment attenuated the severity of arthritis in mice with collagen-induced arthritis and human TNF-α transgenic mice. These findings suggest that an increase in synovial SAE1/UBA2 may contribute to synovial glycolysis and joint inflammation in RA and that targeting SAE1/UBA2 may have therapeutic potential in patients with RA. SUMO-activating enzyme SAE1/UBA2 promotes glycolysis and pathogenic behavior of rheumatoid fibroblast-like synoviocytes through SUMOylation of pyruvate kinase M2.
Collapse
Affiliation(s)
- Cuicui Wang
- Department of Rheumatology and Immunology and
| | - Youjun Xiao
- Department of Rheumatology and Immunology and
| | - Minxi Lao
- Department of Rheumatology and Immunology and
| | | | - Siqi Xu
- Department of Rheumatology and Immunology and
| | - Ruiru Li
- Department of Rheumatology and Immunology and
| | - Xuanxian Xu
- Department of Anesthesia, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yu Kuang
- Department of Rheumatology and Immunology and
| | - Maohua Shi
- Department of Rheumatology and Immunology and
| | - Yaoyao Zou
- Department of Rheumatology and Immunology and
| | - Qingwen Wang
- Department of Rheumatism and Immunology, Peking University People's Hospital, Shenzhen, China
| | | | - Song Guo Zheng
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University College of Medicine and The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Hanshi Xu
- Department of Rheumatology and Immunology and
| |
Collapse
|
135
|
Tan KN, Avery VM, Carrasco-Pozo C. Metabolic Roles of Androgen Receptor and Tip60 in Androgen-Dependent Prostate Cancer. Int J Mol Sci 2020; 21:ijms21186622. [PMID: 32927797 PMCID: PMC7555377 DOI: 10.3390/ijms21186622] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/08/2020] [Accepted: 09/09/2020] [Indexed: 01/10/2023] Open
Abstract
Androgen receptor (AR)-mediated signaling is essential for the growth and differentiation of the normal prostate and is the primary target for androgen deprivation therapy in prostate cancer. Tat interactive protein 60 kDa (Tip60) is a histone acetyltransferase that is critical for AR activation. It is well known that cancer cells rewire their metabolic pathways in order to sustain aberrant proliferation. Growing evidence demonstrates that the AR and Tip60 modulate key metabolic processes to promote the survival of prostate cancer cells, in addition to their classical roles. AR activation enhances glucose metabolism, including glycolysis, tricarboxylic acid cycle and oxidative phosphorylation, as well as lipid metabolism in prostate cancer. The AR also interacts with other metabolic regulators, including calcium/calmodulin-dependent kinase kinase 2 and mammalian target of rapamycin. Several studies have revealed the roles of Tip60 in determining cell fate indirectly by modulating metabolic regulators, such as c-Myc, hypoxia inducible factor 1α (HIF-1α) and p53 in various cancer types. Furthermore, Tip60 has been shown to regulate the activity of key enzymes in gluconeogenesis and glycolysis directly through acetylation. Overall, both the AR and Tip60 are master metabolic regulators that mediate cellular energy metabolism in prostate cancer, providing a framework for the development of novel therapeutic targets in androgen-dependent prostate cancer.
Collapse
Affiliation(s)
- Kah Ni Tan
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111, Australia; (K.N.T.); (V.M.A.)
- CRC for Cancer Therapeutics, Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111, Australia
| | - Vicky M. Avery
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111, Australia; (K.N.T.); (V.M.A.)
- CRC for Cancer Therapeutics, Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111, Australia
| | - Catalina Carrasco-Pozo
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111, Australia; (K.N.T.); (V.M.A.)
- CRC for Cancer Therapeutics, Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111, Australia
- Correspondence: ; Tel.: +617-3735-6034
| |
Collapse
|
136
|
Zou B, Zhao D, He G, Nian Y, Da D, Yan J, Li C. Acetylation and Phosphorylation of Proteins Affect Energy Metabolism and Pork Quality. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:7259-7268. [PMID: 32543862 DOI: 10.1021/acs.jafc.0c01822] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Preslaughter handling has been shown to significantly affect meat quality, but the mechanisms are not fully understood. In this study, we investigated protein phosphorylation and acetylation in pig muscles at early postmortem time and their associations with meat quality attributes. Thirty pigs were randomly assigned to traditional (TH, n = 15) or mild handling (MH, n = 15). Compared with TH, MH reduced the incidence of pale, soft, and exudative (PSE) or dark, firm, and dry (DFD) pork. MH induced 65 and 20 peptides that match with 39 and 12 proteins to be more highly phosphorylated and acetylated, respectively. Creatine kinase, β-enolase, α-1,4-glucan phosphorylase, tropomyosin, and myosin heavy chain isoforms 1, 4, and 7 were found to be simultaneously phosphorylated and acetylated, which may involve glycolysis, tight junctions, and muscle contraction. The phosphorylation and acetylation levels of differential proteins showed significant correlations with meat quality traits. These findings indicate that preslaughter MH can improve meat quality by regulating protein phosphorylation and acetylation involving energy metabolism in muscle.
Collapse
Affiliation(s)
- Bo Zou
- Key Laboratory of Meat Processing and Quality Control, MOE; Key Laboratory of Meat Processing, MARA; Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, 210095 Nanjing, China
| | - Di Zhao
- Key Laboratory of Meat Processing and Quality Control, MOE; Key Laboratory of Meat Processing, MARA; Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, 210095 Nanjing, China
| | - Guangjie He
- Key Laboratory of Meat Processing and Quality Control, MOE; Key Laboratory of Meat Processing, MARA; Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, 210095 Nanjing, China
| | - Yingqun Nian
- Key Laboratory of Meat Processing and Quality Control, MOE; Key Laboratory of Meat Processing, MARA; Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, 210095 Nanjing, China
| | - Dandan Da
- Key Laboratory of Meat Processing and Quality Control, MOE; Key Laboratory of Meat Processing, MARA; Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, 210095 Nanjing, China
| | - Jing Yan
- Key Laboratory of Meat Processing and Quality Control, MOE; Key Laboratory of Meat Processing, MARA; Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, 210095 Nanjing, China
| | - Chunbao Li
- Key Laboratory of Meat Processing and Quality Control, MOE; Key Laboratory of Meat Processing, MARA; Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, 210095 Nanjing, China
| |
Collapse
|
137
|
Feng J, Li J, Wu L, Yu Q, Ji J, Wu J, Dai W, Guo C. Emerging roles and the regulation of aerobic glycolysis in hepatocellular carcinoma. J Exp Clin Cancer Res 2020; 39:126. [PMID: 32631382 PMCID: PMC7336654 DOI: 10.1186/s13046-020-01629-4] [Citation(s) in RCA: 403] [Impact Index Per Article: 80.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 06/25/2020] [Indexed: 12/14/2022] Open
Abstract
Liver cancer has become the sixth most diagnosed cancer and the fourth leading cause of cancer death worldwide. Hepatocellular carcinoma (HCC) is responsible for up to 75-85% of primary liver cancers, and sorafenib is the first targeted drug for advanced HCC treatment. However, sorafenib resistance is common because of the resultant enhancement of aerobic glycolysis and other molecular mechanisms. Aerobic glycolysis was firstly found in HCC, acts as a hallmark of liver cancer and is responsible for the regulation of proliferation, immune evasion, invasion, metastasis, angiogenesis, and drug resistance in HCC. The three rate-limiting enzymes in the glycolytic pathway, including hexokinase 2 (HK2), phosphofructokinase 1 (PFK1), and pyruvate kinases type M2 (PKM2) play an important role in the regulation of aerobic glycolysis in HCC and can be regulated by many mechanisms, such as the AMPK, PI3K/Akt pathway, HIF-1α, c-Myc and noncoding RNAs. Because of the importance of aerobic glycolysis in the progression of HCC, targeting key factors in its pathway such as the inhibition of HK2, PFK or PKM2, represent potential new therapeutic approaches for the treatment of HCC.
Collapse
Affiliation(s)
- Jiao Feng
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, number 1291, Jiangning road, Putuo, Shanghai, 200060, China
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China
| | - Jingjing Li
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, number 1291, Jiangning road, Putuo, Shanghai, 200060, China
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China
| | - Liwei Wu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China
| | - Qiang Yu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China
| | - Jie Ji
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China
| | - Jianye Wu
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, number 1291, Jiangning road, Putuo, Shanghai, 200060, China.
| | - Weiqi Dai
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, number 1291, Jiangning road, Putuo, Shanghai, 200060, China.
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China.
- Department of Gastroenterology, Zhongshan Hospital of Fudan University, Shanghai, 200032, China.
- Shanghai Institute of Liver Diseases, Zhongshan Hospital of Fudan University, Shanghai, 200032, China.
- Shanghai Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200336, China.
| | - Chuanyong Guo
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, number 1291, Jiangning road, Putuo, Shanghai, 200060, China.
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China.
| |
Collapse
|
138
|
Masaki S, Hashimoto K, Kihara D, Tsuzuki C, Kataoka N, Suzuki K. The cysteine residue at 424th of pyruvate kinase M2 is crucial for tetramerization and responsiveness to oxidative stress. Biochem Biophys Res Commun 2020; 526:973-977. [PMID: 32295714 DOI: 10.1016/j.bbrc.2020.03.182] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 03/30/2020] [Indexed: 12/24/2022]
Abstract
Alternative splicing of the pyruvate kinase M (PKM) pre-mRNA generates two isoforms, PKM1 and PKM2. PKM catalyzes the conversion of phosphoenol-pyruvate to pyruvate in glycolytic pathway. PKM1 exist as a stable tetramer that is at an active enzyme state, while PKM2 is in equilibrium among monomer, dimer and tetramer under the regulation of its allosteric activators. Many cancer cells show the feature of higher glucose uptake and lactate production in spite of oxygen availability, which is known as the Warburg effect. PKM2 is upregulated in most cancer types and the inactive PKM2 lead to the cancer metabolism. In addition, dimeric PKM2 induces its nuclear translocation through posttranslational modification and acts as a transcriptional co-activator for the expression of oncogenes. Therefore, it is important to elucidate mechanisms for modulation of an active or inactive state of PKM2, namely the tetramer-to-dimer-transition. The definitive difference between PKM1 and PKM2 is to constitutively form tetramer or not in the cytoplasm, which is ascribed to 22 amino acids derived from exon 9 (PKM1) or exon 10 (PKM2). In this study, we generated 22 different PKM1-mimetic point mutants of PKM2, and demonstrated that replacement of cysteine424 residue of PKM2 with leucine424 conserved in PKM1 (C424L) promote its tetramerization. PKM2(C424L) formed a tetramer without allosteric activator, and escaped the inhibitory effects by oxidative stress, like PKM1. Our findings intensely suggest that C424 or L424 determines the different catalytic and modulatory properties between PKM splicing isoforms.
Collapse
Affiliation(s)
- So Masaki
- Laboratory of Molecular Medicinal Science, Department of Pharmaceutical Sciences, Ritsumeikan University, Shiga, Japan; Laboratory for Malignancy Control Research, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Kozue Hashimoto
- Laboratory of Molecular Medicinal Science, Department of Pharmaceutical Sciences, Ritsumeikan University, Shiga, Japan
| | - Daiki Kihara
- Laboratory of Molecular Medicinal Science, Department of Pharmaceutical Sciences, Ritsumeikan University, Shiga, Japan
| | - Chizuru Tsuzuki
- Laboratory for Malignancy Control Research, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Naoyuki Kataoka
- Laboratory for Malignancy Control Research, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan; Laboratory of Cellular Biochemistry, Department of Animal Resource Sciences, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Kenji Suzuki
- Laboratory of Molecular Medicinal Science, Department of Pharmaceutical Sciences, Ritsumeikan University, Shiga, Japan
| |
Collapse
|
139
|
Li Q, Leng K, Liu Y, Sun H, Gao J, Ren Q, Zhou T, Dong J, Xia J. The impact of hyperglycaemia on PKM2-mediated NLRP3 inflammasome/stress granule signalling in macrophages and its correlation with plaque vulnerability: an in vivo and in vitro study. Metabolism 2020; 107:154231. [PMID: 32298723 DOI: 10.1016/j.metabol.2020.154231] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 04/05/2020] [Accepted: 04/11/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND The mechanism of pyruvate kinase M2 (PKM2)-mediated inflammatory signalling in macrophages when plaques rupture and the impact of hyperglycaemia on the signalling are unclear. The present study aimed to explore the impact of hyperglycaemia on PKM2-mediated NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome/stress granule signalling in macrophages and its correlation with plaque vulnerability in vivo and in vitro. METHODS From July to December 2019, 80 patients with coronary heart disease (CHD) were divided into acute ST-segment elevation myocardial infarction (STEMI) (n = 57) (DM-STEMI, n = 21; non-DM-STEMI, n = 36) and stable CHD (SCHD) groups (n = 23). Circulating mononuclear cells were isolated. The value of peak troponin I (TnI), the Global Registry of Acute Coronary Events (GRACE) risk score, and the expression levels of the related markers were quantified and compared. In vitro studies on the THP-1 cells were also performed. RESULTS The DM-STEMI group had a higher value of peak TnI and a higher GRACE risk score than the non-DM-STEMI group (p < 0.05). The highest expression levels of PKM2, NLRP3, interleukin (IL)-1β, and IL-18 and the lowest expression level of GTPase activating protein (SH3 domain)-binding protein 1 (G3BP1) (a stress granule marker protein) were observed in the DM-STEMI group, and they were followed by the non-DM-STEMI group and the SCHD group (p < 0.05). In vitro studies showed similar results and that TEPP-46 (a PKM2 activator) and 2-deoxy-d-glucose (a toxic glucose analogue) reversed the hyperglycaemia-induced increase in the NLRP3 inflammasome and decrease in G3BP1 expression. CONCLUSION Hyperglycaemia might increase the activation of PKM2-mediated NLRP3 inflammasome/stress granule signalling and increase plaque vulnerability, associating it with worse prognosis. PKM2 may be a novel prognostic indicator and a new target for the treatment of patients with CHD and DM.
Collapse
Affiliation(s)
- Qinxue Li
- Department of Cardiology, Xuanwu Hospital, Capital Medical University, National Clinical Research Centre for Geriatric Diseases, Beijing 100053, China
| | - Kunkun Leng
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang 110122, China
| | - Yayun Liu
- Department of Cardiology, Xuanwu Hospital, Capital Medical University, National Clinical Research Centre for Geriatric Diseases, Beijing 100053, China
| | - Haichen Sun
- Surgical Laboratory, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Jinhuan Gao
- Department of Cardiology, Xuanwu Hospital, Capital Medical University, National Clinical Research Centre for Geriatric Diseases, Beijing 100053, China
| | - Quanxin Ren
- Beijing Fangshan District Liangxiang Hospital, Beijing 102501, China
| | - Tian Zhou
- Department of Cardiology, Xuanwu Hospital, Capital Medical University, National Clinical Research Centre for Geriatric Diseases, Beijing 100053, China
| | - Jing Dong
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang 110122, China.
| | - Jinggang Xia
- Department of Cardiology, Xuanwu Hospital, Capital Medical University, National Clinical Research Centre for Geriatric Diseases, Beijing 100053, China.
| |
Collapse
|
140
|
Zhang C, Gou X, He W, Yang H, Yin H. A glycolysis-based 4-mRNA signature correlates with the prognosis and cell cycle process in patients with bladder cancer. Cancer Cell Int 2020; 20:177. [PMID: 32467671 PMCID: PMC7238531 DOI: 10.1186/s12935-020-01255-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 05/10/2020] [Indexed: 12/17/2022] Open
Abstract
Background Bladder cancer is one of the most prevalent malignancies worldwide. However, traditional indicators have limited predictive effects on the clinical outcomes of bladder cancer. The aim of this study was to develop and validate a glycolysis-related gene signature for predicting the prognosis of patients with bladder cancer that have limited therapeutic options. Methods mRNA expression profiling was obtained from patients with bladder cancer from The Cancer Genome Atlas (TCGA) database. Gene set enrichment analysis (GSEA) was conducted to identify glycolytic gene sets that were significantly different between bladder cancer tissues and paired normal tissues. A prognosis-related gene signature was constructed by univariate and multivariate Cox analysis. Kaplan–Meier curves and time-dependent receiver operating characteristic (ROC) curves were utilized to evaluate the signature. A nomogram combined with the gene signature and clinical parameters was constructed. Correlations between glycolysis-related gene signature and molecular characterization as well as cancer subtypes were analyzed. RT-qPCR was applied to analyze gene expression. Functional experiments were performed to determine the role of PKM2 in the proliferation of bladder cancer cells. Results Using a Cox proportional regression model, we established that a 4-mRNA signature (NUP205, NUPL2, PFKFB1 and PKM) was significantly associated with prognosis in bladder cancer patients. Based on the signature, patients were split into high and low risk groups, with different prognostic outcomes. The gene signature was an independent prognostic indicator for overall survival. The ability of the 4-mRNA signature to make an accurate prognosis was tested in two other validation datasets. GSEA was performed to explore the 4-mRNA related canonical pathways and biological processes, such as the cell cycle, hypoxia, p53 pathway, and PI3K/AKT/mTOR pathway. A heatmap showing the correlation between risk score and cell cycle signature was generated. RT-qPCR revealed the genes that were differentially expressed between normal and cancer tissues. Experiments showed that PKM2 plays essential roles in cell proliferation and the cell cycle. Conclusion The established 4‑mRNA signature may act as a promising model for generating accurate prognoses for patients with bladder cancer, but the specific biological mechanism needs further verification.
Collapse
Affiliation(s)
- Chen Zhang
- 2Department of Gynecology and Obstetrics, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Chongqing, 400016 China.,4Chongqing Key Laboratory of Maternal and Fetal Medicine, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Chongqing, 400016 China
| | - Xin Gou
- 1Department of Urology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Chongqing, 400016 China
| | - Weiyang He
- 1Department of Urology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Chongqing, 400016 China
| | - Huaan Yang
- Department of Urology, Yubei District People's Hospital, No. 69 Jianshe Road, Chongqing, 400016 China
| | - Hubin Yin
- 1Department of Urology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Chongqing, 400016 China.,3Central Laboratory, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Chongqing, 400016 China
| |
Collapse
|
141
|
Hu K, Xu J, Fan K, Zhou D, Li L, Tang L, Peng X, Zhang L, Wang Y. Nuclear accumulation of pyruvate kinase M2 promotes liver regeneration via activation of signal transducer and activator of transcription 3. Life Sci 2020; 250:117561. [PMID: 32198052 DOI: 10.1016/j.lfs.2020.117561] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/07/2020] [Accepted: 03/16/2020] [Indexed: 12/29/2022]
Abstract
AIMS Pyruvate kinase M2 (PKM2), a unique isoform of the pyruvate kinases, not only acts as a crucial metabolic enzyme when it locates in the cytoplasm, but also plays important roles in tumor formation and growth when it accumulates in the nuclei. Our aim was to investigate the potential role of PKM2 in liver regeneration in mice insulted with carbon tetrachloride (CCl4). MATERIAL AND METHODS The liver regeneration model was established by intraperitoneal injection of CCl4 for 48 h in male BALB/c mice. The expression of PKM2, phospho-STAT3, STAT3, proliferating cell nuclear antigen (PCNA) and Cyclin D1 were evaluated by western blot. The distribution of PKM2 was verified by immunofluorescence staining. The degree of injured region was assessed by hematoxylin and eosin (HE) staining. The proliferation of liver cells was tested by Immunohistochemistry. KEY FINDINGS The nuclear accumulation of PKM2 increased in the liver treated with CCl4, but treatment with ML-265 significantly suppressed CCl4-induced nuclear accumulation of PKM2. In addition, treatment with ML-265 suppressed the level of cyclin D1 and proliferating cell nuclear antigen (PCNA), reduced the count of Ki67-positive hepatocytes, and expanded the damaged region in histological examination. Meanwhile, treatment with ML-265 suppressed the phosphorylation of nuclear signal transducer and activator of transcription 3 (STAT3). Inhibition of STAT3 by stattic made the same effects as ML-265. SIGNIFICANCE These data uncovered the role of nuclear PKM2 in liver regeneration and the pro-proliferation effects of nuclear PKM2 may be through targeting its downstream transcription factor STAT3.
Collapse
Affiliation(s)
- Kai Hu
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China; Department of Histology and Embryology, Chongqing Medical University, Chongqing, China
| | - Juanjuan Xu
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Kerui Fan
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Dan Zhou
- Department of Pathology, Fuling Center Hospital of Chongqing City, Chongqing, China
| | - Longjiang Li
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Li Tang
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Xianwen Peng
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Li Zhang
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China.
| | - Yaping Wang
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
142
|
Targeting immunometabolism as an anti-inflammatory strategy. Cell Res 2020; 30:300-314. [PMID: 32132672 PMCID: PMC7118080 DOI: 10.1038/s41422-020-0291-z] [Citation(s) in RCA: 338] [Impact Index Per Article: 67.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 02/02/2020] [Indexed: 12/14/2022] Open
Abstract
The growing field of immunometabolism has taught us how metabolic cellular reactions and processes not only provide a means to generate ATP and biosynthetic precursors, but are also a way of controlling immunity and inflammation. Metabolic reprogramming of immune cells is essential for both inflammatory as well as anti-inflammatory responses. Four anti-inflammatory therapies, DMF, Metformin, Methotrexate and Rapamycin all work by affecting metabolism and/or regulating or mimicking endogenous metabolites with anti-inflammatory effects. Evidence is emerging for the targeting of specific metabolic events as a strategy to limit inflammation in different contexts. Here we discuss these recent developments and speculate on the prospect of targeting immunometabolism in the effort to develop novel anti-inflammatory therapeutics. As accumulating evidence for roles of an intricate and elaborate network of metabolic processes, including lipid, amino acid and nucleotide metabolism provides key focal points for developing new therapies, we here turn our attention to glycolysis and the TCA cycle to provide examples of how metabolic intermediates and enzymes can provide potential novel therapeutic targets.
Collapse
|
143
|
Zahra K, Dey T, Ashish, Mishra SP, Pandey U. Pyruvate Kinase M2 and Cancer: The Role of PKM2 in Promoting Tumorigenesis. Front Oncol 2020; 10:159. [PMID: 32195169 PMCID: PMC7061896 DOI: 10.3389/fonc.2020.00159] [Citation(s) in RCA: 318] [Impact Index Per Article: 63.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 01/29/2020] [Indexed: 12/17/2022] Open
Abstract
Pyruvate kinase plays a pivotal role in regulating cell metabolism. The final and rate-limiting step of glycolysis is the conversion of Phosphoenolpyruvate (PEP) to Pyruvate, which is catalyzed by Pyruvate Kinase. There are four isomeric, tissue-specific forms of Pyruvate Kinase found in mammals: PKL, PKR, PKM1, and PKM2. PKM1 and PKM2 are formed bya single mRNA transcript of the PKM gene by alternative splicing. The oligomers of PKM2 exist in high activity tetramer and low activity dimer forms. The dimer PKM2 regulates the rate-limiting step of glycolysis that shifts the glucose metabolism from the normal respiratory chain to lactate production in tumor cells. Besides its role as a metabolic regulator, it also acts as protein kinase, which contributes to tumorigenesis. This review is focused on the metabolic role of pyruvate kinase M2 in normal cells vs. cancerous cells and its regulation at the transcriptional level. The review also highlights the role of PKM2 as a potential diagnostic marker and as a therapeutic target in cancer treatment.
Collapse
Affiliation(s)
- Kulsoom Zahra
- Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Tulika Dey
- Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Ashish
- Department of Anatomy, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Surendra Pratap Mishra
- Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Uma Pandey
- Department of Obstetrics and Gynecology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
144
|
Nie H, Ju H, Fan J, Shi X, Cheng Y, Cang X, Zheng Z, Duan X, Yi W. O-GlcNAcylation of PGK1 coordinates glycolysis and TCA cycle to promote tumor growth. Nat Commun 2020; 11:36. [PMID: 31911580 PMCID: PMC6946671 DOI: 10.1038/s41467-019-13601-8] [Citation(s) in RCA: 219] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 11/11/2019] [Indexed: 01/14/2023] Open
Abstract
Many cancer cells display enhanced glycolysis and suppressed mitochondrial metabolism. This phenomenon, known as the Warburg effect, is critical for tumor development. However, how cancer cells coordinate glucose metabolism through glycolysis and the mitochondrial tricarboxylic acid (TCA) cycle is largely unknown. We demonstrate here that phosphoglycerate kinase 1 (PGK1), the first ATP-producing enzyme in glycolysis, is reversibly and dynamically modified with O-linked N-acetylglucosamine (O-GlcNAc) at threonine 255 (T255). O-GlcNAcylation activates PGK1 activity to enhance lactate production, and simultaneously induces PGK1 translocation into mitochondria. Inside mitochondria, PGK1 acts as a kinase to inhibit pyruvate dehydrogenase (PDH) complex to reduce oxidative phosphorylation. Blocking T255 O-GlcNAcylation of PGK1 decreases colon cancer cell proliferation, suppresses glycolysis, enhances the TCA cycle, and inhibits tumor growth in xenograft models. Furthermore, PGK1 O-GlcNAcylation levels are elevated in human colon cancers. This study highlights O-GlcNAcylation as an important signal for coordinating glycolysis and the TCA cycle to promote tumorigenesis.
Collapse
Affiliation(s)
- Hao Nie
- MOE Key Laboratory of Biosystems Homeostasis & Protection, College of Life Sciences; The First Affiliated Hospital, School of Medicine, Zhejiang University, 310058, Hangzhou, China
| | - Haixing Ju
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, 310022, Hangzhou, China
| | - Jiayi Fan
- MOE Key Laboratory of Biosystems Homeostasis & Protection, College of Life Sciences; The First Affiliated Hospital, School of Medicine, Zhejiang University, 310058, Hangzhou, China
| | - Xiaoliu Shi
- MOE Key Laboratory of Biosystems Homeostasis & Protection, College of Life Sciences; The First Affiliated Hospital, School of Medicine, Zhejiang University, 310058, Hangzhou, China
| | - Yaxian Cheng
- MOE Key Laboratory of Biosystems Homeostasis & Protection, College of Life Sciences; The First Affiliated Hospital, School of Medicine, Zhejiang University, 310058, Hangzhou, China
| | - Xiaohui Cang
- Division of Medical Genetics and Genomics, The Children's Hospital, School of Medicine, Zhejiang University, 310058, Hangzhou, China
| | - Zhiguo Zheng
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, 310022, Hangzhou, China
| | - Xiaotao Duan
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, 100850, Beijing, China
| | - Wen Yi
- MOE Key Laboratory of Biosystems Homeostasis & Protection, College of Life Sciences; The First Affiliated Hospital, School of Medicine, Zhejiang University, 310058, Hangzhou, China.
| |
Collapse
|
145
|
Singh JP, Qian K, Lee JS, Zhou J, Han X, Zhang B, Ong Q, Ni W, Jiang M, Ruan HB, Li MD, Zhang K, Ding Z, Lee P, Singh K, Wu J, Herzog RI, Kaech S, Wendel HG, Yates JR, Han W, Sherwin RS, Nie Y, Yang X. O-GlcNAcase targets pyruvate kinase M2 to regulate tumor growth. Oncogene 2020; 39:560-573. [PMID: 31501520 PMCID: PMC7107572 DOI: 10.1038/s41388-019-0975-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Revised: 05/12/2019] [Accepted: 06/18/2019] [Indexed: 01/24/2023]
Abstract
Cancer cells are known to adopt aerobic glycolysis in order to fuel tumor growth, but the molecular basis of this metabolic shift remains largely undefined. O-GlcNAcase (OGA) is an enzyme harboring O-linked β-N-acetylglucosamine (O-GlcNAc) hydrolase and cryptic lysine acetyltransferase activities. Here, we report that OGA is upregulated in a wide range of human cancers and drives aerobic glycolysis and tumor growth by inhibiting pyruvate kinase M2 (PKM2). PKM2 is dynamically O-GlcNAcylated in response to changes in glucose availability. Under high glucose conditions, PKM2 is a target of OGA-associated acetyltransferase activity, which facilitates O-GlcNAcylation of PKM2 by O-GlcNAc transferase (OGT). O-GlcNAcylation inhibits PKM2 catalytic activity and thereby promotes aerobic glycolysis and tumor growth. These studies define a causative role for OGA in tumor progression and reveal PKM2 O-GlcNAcylation as a metabolic rheostat that mediates exquisite control of aerobic glycolysis.
Collapse
Affiliation(s)
- Jay Prakash Singh
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06519, USA
- Department of Comparative Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06519, USA
| | - Kevin Qian
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06519, USA
- Department of Comparative Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06519, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06519, USA
| | - Jeong-Sang Lee
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06519, USA
- Department of Comparative Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06519, USA
| | - Jinfeng Zhou
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Xuemei Han
- Department of Chemical Physiology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Bichen Zhang
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06519, USA
- Department of Comparative Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06519, USA
| | - Qunxiang Ong
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06519, USA
- Department of Comparative Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06519, USA
- Singapore Bioimaging Consortium, Singapore, Singapore
| | - Weiming Ni
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06519, USA
- Department of Comparative Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06519, USA
| | - Mingzuo Jiang
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Hai-Bin Ruan
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06519, USA
- Department of Comparative Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06519, USA
| | - Min-Dian Li
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06519, USA
- Department of Comparative Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06519, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06519, USA
| | - Kaisi Zhang
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06519, USA
- Department of Comparative Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06519, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06519, USA
| | - Zhaobing Ding
- Singapore Bioimaging Consortium, Singapore, Singapore
| | - Philip Lee
- Singapore Bioimaging Consortium, Singapore, Singapore
| | - Kamini Singh
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Jing Wu
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06519, USA
- Department of Comparative Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06519, USA
- School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China
| | - Raimund I Herzog
- Department of Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06519, USA
| | - Susan Kaech
- Department of Immunobiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06519, USA
| | - Hans-Guido Wendel
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - John R Yates
- Department of Chemical Physiology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Weiping Han
- Singapore Bioimaging Consortium, Singapore, Singapore
| | - Robert S Sherwin
- Department of Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06519, USA
| | - Yongzhan Nie
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Xiaoyong Yang
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06519, USA.
- Department of Comparative Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06519, USA.
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06519, USA.
| |
Collapse
|
146
|
Zhang L, Bailleul J, Yazal T, Dong K, Sung D, Dao A, Gosa L, Nathanson D, Bhat K, Duhachek-Muggy S, Alli C, Dratver MB, Pajonk F, Vlashi E. PK-M2-mediated metabolic changes in breast cancer cells induced by ionizing radiation. Breast Cancer Res Treat 2019; 178:75-86. [PMID: 31372790 PMCID: PMC6790295 DOI: 10.1007/s10549-019-05376-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 07/23/2019] [Indexed: 12/31/2022]
Abstract
PURPOSE Radiotherapy (RT) constitutes an important part of breast cancer treatment. However, triple negative breast cancers (TNBC) exhibit remarkable resistance to most therapies, including RT. Developing new ways to radiosensitize TNBC cells could result in improved patient outcomes. The M2 isoform of pyruvate kinase (PK-M2) is believed to be responsible for the re-wiring of cancer cell metabolism after oxidative stress. The aim of the study was to determine the effect of ionizing radiation (IR) on PK-M2-mediated metabolic changes in TNBC cells, and their survival. In addition, we determine the effect of PK-M2 activators on breast cancer stem cells, a radioresistant subpopulation of breast cancer stem cells. METHODS Glucose uptake, lactate production, and glutamine consumption were assessed. The cellular localization of PK-M2 was evaluated by western blot and confocal microscopy. The small molecule activator of PK-M2, TEPP46, was used to promote its pyruvate kinase function. Finally, effects on cancer stem cell were evaluated via sphere forming capacity. RESULTS Exposure of TNBC cells to IR increased their glucose uptake and lactate production. As expected, PK-M2 expression levels also increased, especially in the nucleus, although overall pyruvate kinase activity was decreased. PK-M2 nuclear localization was shown to be associated with breast cancer stem cells, and activation of PK-M2 by TEPP46 depleted this population. CONCLUSIONS Radiotherapy can induce metabolic changes in TNBC cells, and these changes seem to be mediated, at least in part by PK-M2. Importantly, our results show that activators of PK-M2 can deplete breast cancer stem cells in vitro. This study supports the idea of combining PK-M2 activators with radiation to enhance the effect of radiotherapy in resistant cancers, such as TNBC.
Collapse
Affiliation(s)
- Le Zhang
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA, 90095-1714, USA
| | - Justine Bailleul
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA, 90095-1714, USA
| | - Taha Yazal
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA, 90095-1714, USA
| | - Kevin Dong
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA, 90095-1714, USA
| | - David Sung
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA, 90095-1714, USA
| | - Amy Dao
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA, 90095-1714, USA
| | - Laura Gosa
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - David Nathanson
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA
| | - Kruttika Bhat
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA, 90095-1714, USA
| | - Sara Duhachek-Muggy
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA, 90095-1714, USA
| | - Claudia Alli
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA, 90095-1714, USA
| | - Milana Bochkur Dratver
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA, 90095-1714, USA
| | - Frank Pajonk
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA, 90095-1714, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA
| | - Erina Vlashi
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA, 90095-1714, USA.
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
147
|
Li, J, Wang, T, Xia J, Yao W, Huang F. Enzymatic and nonenzymatic protein acetylations control glycolysis process in liver diseases. FASEB J 2019; 33:11640-11654. [PMID: 31370704 PMCID: PMC6902721 DOI: 10.1096/fj.201901175r] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 07/09/2019] [Indexed: 12/12/2022]
Abstract
Impaired glycolysis has pathologic effects on the occurrence and progression of liver diseases, and it appears that glycolysis is increased to different degrees in different liver diseases. As an important post-translational modification, reversible lysine acetylation regulates almost all cellular processes, including glycolysis. Lysine acetylation can occur enzymatically with acetyltransferases or nonenzymatically with acetyl-coenzyme A. Accompanied by the progression of liver diseases, there seems to be a temporal and spatial variation between enzymatic and nonenzymatic acetylations in the regulation of glycolysis. Here, we summarize the most recent findings on the functions and targets of acetylation in controlling glycolysis in the different stages of liver diseases. In addition, we discuss the differences and causes between enzymatic and nonenzymatic acetylations in regulating glycolysis throughout the progression of liver diseases. Then, we review these new discoveries to provide the potential implications of these findings for therapeutic interventions in liver diseases.-Li, J., Wang, T., Xia, J., Yao, W., Huang, F. Enzymatic and nonenzymatic protein acetylations control glycolysis process in liver diseases.
Collapse
Affiliation(s)
- Juan Li,
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Tongxin Wang,
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Jun Xia
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Weilei Yao
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Feiruo Huang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
148
|
Alquraishi M, Puckett DL, Alani DS, Humidat AS, Frankel VD, Donohoe DR, Whelan J, Bettaieb A. Pyruvate kinase M2: A simple molecule with complex functions. Free Radic Biol Med 2019; 143:176-192. [PMID: 31401304 PMCID: PMC6848794 DOI: 10.1016/j.freeradbiomed.2019.08.007] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/29/2019] [Accepted: 08/07/2019] [Indexed: 12/31/2022]
Abstract
Pyruvate kinase M2 is a critical enzyme that regulates cell metabolism and growth under different physiological conditions. In its metabolic role, pyruvate kinase M2 catalyzes the last glycolytic step which converts phosphoenolpyruvate to pyruvate with the generation of ATP. Beyond this metabolic role in glycolysis, PKM2 regulates gene expression in the nucleus, phosphorylates several essential proteins that regulate major cell signaling pathways, and contribute to the redox homeostasis of cancer cells. The expression of PKM2 has been demonstrated to be significantly elevated in several types of cancer, and the overall inflammatory response. The unusual pattern of PKM2 expression inspired scientists to investigate the unrevealed functions of PKM2 and the therapeutic potential of targeting PKM2 in cancer and other disorders. Therefore, the purpose of this review is to discuss the mechanistic and therapeutic potential of targeting PKM2 with the focus on cancer metabolism, redox homeostasis, inflammation, and metabolic disorders. This review highlights and provides insight into the metabolic and non-metabolic functions of PKM2 and its relevant association with health and disease.
Collapse
Affiliation(s)
- Mohammed Alquraishi
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN, 37996-0840, USA
| | - Dexter L Puckett
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN, 37996-0840, USA
| | - Dina S Alani
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN, 37996-0840, USA
| | - Amal S Humidat
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN, 37996-0840, USA
| | - Victoria D Frankel
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN, 37996-0840, USA
| | - Dallas R Donohoe
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN, 37996-0840, USA
| | - Jay Whelan
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN, 37996-0840, USA
| | - Ahmed Bettaieb
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN, 37996-0840, USA; Department of Biochemistry, Cellular and Molecular Biology, University of Tennessee, Knoxville, TN, 37996-0840, USA; Graduate School of Genome Science and Technology, University of Tennessee, Knoxville, TN, 37996-0840, USA.
| |
Collapse
|
149
|
The Molecular Effects of Sulforaphane and Capsaicin on Metabolism upon Androgen and Tip60 Activation of Androgen Receptor. Int J Mol Sci 2019; 20:ijms20215384. [PMID: 31671779 PMCID: PMC6861939 DOI: 10.3390/ijms20215384] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 10/21/2019] [Accepted: 10/24/2019] [Indexed: 12/20/2022] Open
Abstract
Androgen receptor (AR) stimulators, such as androgen and Tip60, play a pivotal role in prostatic carcinogenesis as androgen receptor signaling is critical for the growth and transformation of the prostate gland. Moreover, androgen and Tip60 promotes HIF-1α activation, involved in metabolic reprogramming by increasing glycolysis, a hallmark in cancer initiation and development. In this study we evaluated the effect of androgen and Tip60 stimulus in AR pathway activation and HIF-1α stabilization, in terms of proliferation and cell metabolism in androgen-sensitive LNCaP cells. The protective role of the bioactive compounds sulforaphane and capsaicin against the effect of these stimuli leading to pro-carcinogenic features was also addressed. Sulforaphane and capsaicin decreased nuclear AR, prostate specific antigen and Bcl-XL levels, and cell proliferation induced by androgen and Tip60 in LNCaP cells. These bioactive compounds prevented the increase in glycolysis, hexokinase and pyruvate kinase activity, and reduced HIF-1α stabilization induced by androgen and Tip60 in LNCaP cells. The protective role of sulforaphane and capsaicin on prostate cancer may rely on mechanisms involving the inhibition of Tip60, AR and HIF-1α effects.
Collapse
|
150
|
Srivastava D, Nandi S, Dey M. Mechanistic and Structural Insights into Cysteine-Mediated Inhibition of Pyruvate Kinase Muscle Isoform 2. Biochemistry 2019; 58:3669-3682. [PMID: 31386812 DOI: 10.1021/acs.biochem.9b00349] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Cancer cells regulate key enzymes in the glycolytic pathway to control the glycolytic flux, which is necessary for their growth and proliferation. One of the enzymes is pyruvate kinase muscle isoform 2 (PKM2), which is allosterically regulated by various small molecules. Using detailed biochemical and kinetic studies, we demonstrate that cysteine inhibits wild-type (wt) PKM2 by shifting from an active tetramer to a mixture of a tetramer and a less active dimer/monomer equilibrium and that the inhibition is dependent on cysteine concentration. The cysteine-mediated PKM2 inhibition is reversed by fructose 1,6-bisphosphate, an allosteric activator of PKM2. Furthermore, kinetic studies using two dimeric PKM2 variants, S437Y PKM2 and G415R PKM2, show that the reversal is caused by the tetramerization of wtPKM2. The crystal structure of the wtPKM2-Cys complex was determined at 2.25 Å, which showed that cysteine is held to the amino acid binding site via its main chain groups, similar to that observed for phenylalanine, alanine, serine, and tryptophan. Notably, ligand binding studies using fluorescence and isothermal titration calorimetry show that the presence of phosphoenolpyruvate alters the binding affinities of amino acids for wtPKM2 and vice versa, thereby unravelling the existence of a functionally bidirectional coupling between the amino acid binding site and the active site of wtPKM2.
Collapse
Affiliation(s)
- Dhiraj Srivastava
- Department of Chemistry , The University of Iowa , Iowa City , Iowa 52242 , United States
| | - Suparno Nandi
- Department of Chemistry , The University of Iowa , Iowa City , Iowa 52242 , United States
| | - Mishtu Dey
- Department of Chemistry , The University of Iowa , Iowa City , Iowa 52242 , United States
| |
Collapse
|