1
|
Jin C, Hu W, Wang Y, Wu H, Zeng S, Ying M, Hu X. Deciphering the interaction between PKM2 and the built-in thermodynamic properties of the glycolytic pathway in cancer cells. J Biol Chem 2024; 300:107648. [PMID: 39121998 PMCID: PMC11402776 DOI: 10.1016/j.jbc.2024.107648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/24/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
Most cancer cells exhibit high glycolysis rates under conditions of abundant oxygen. Maintaining a stable glycolytic rate is critical for cancer cell growth as it ensures sufficient conversion of glucose carbons to energy, biosynthesis, and redox balance. Here we deciphered the interaction between PKM2 and the thermodynamic properties of the glycolytic pathway. Knocking down or knocking out PKM2 induced a thermodynamic equilibration in the glycolytic pathway, characterized by the reciprocal changes of the Gibbs free energy (ΔG) of the reactions catalyzed by PFK1 and PK, leading to a less exergonic PFK1-catalyzed reaction and a more exergonic PK-catalyzed reaction. The changes in the ΔGs of the two reactions cause the accumulation of intermediates, including the substrate PEP (the substrate of PK), in the segment between PFK1 and PK. The increased concentration of PEP in turn increased PK activity in the glycolytic pathway. Thus, the interaction between PKM2 and the thermodynamic properties of the glycolytic pathway maintains the reciprocal relationship between PK concentration and its substrate PEP concentration, by which, PK activity in the glycolytic pathway can be stabilized and effectively counteracts the effect of PKM2 KD or KO on glycolytic rate. In line with our previous reports, this study further validates the roles of the thermodynamics of the glycolytic pathway in stabilizing glycolysis in cancer cells. Deciphering the interaction between glycolytic enzymes and the thermodynamics of the glycolytic pathway will promote a better understanding of the flux control of glycolysis in cancer cells.
Collapse
Affiliation(s)
- Chengmeng Jin
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Province Key Laboratory of Molecular Biology in Medical Sciences, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, Zhejiang, China; Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China
| | - Wei Hu
- Center for Nutrition & Food Sciences, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, Saint Lucia, Queensland, Australia
| | - Yuqi Wang
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Hao Wu
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Province Key Laboratory of Molecular Biology in Medical Sciences, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, Zhejiang, China; Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China
| | - Siying Zeng
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Minfeng Ying
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Province Key Laboratory of Molecular Biology in Medical Sciences, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, Zhejiang, China; Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China
| | - Xun Hu
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Province Key Laboratory of Molecular Biology in Medical Sciences, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, Zhejiang, China; Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
2
|
Choithramani A, Das R, Bothra G, Patel Vatsa P, Muthukumar V, Bhuvana BKS, Kapoor S, Moola D, Chowdhury MG, Mandoli A, Shard A. Targeted suppression of oral squamous cell carcinoma by pyrimidine-tethered quinoxaline derivatives. RSC Med Chem 2024; 15:2729-2744. [PMID: 39149105 PMCID: PMC11324040 DOI: 10.1039/d4md00042k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 05/17/2024] [Indexed: 08/17/2024] Open
Abstract
Oral cancer (OC) stands as a prominent cause of global mortality. Despite numerous efforts in recent decades, the efficacy of novel therapies to extend the lifespan of OC patients remains disappointingly low. Consequently, the demand for innovative therapeutic agents has become all the more pressing. In this context, we present our work on the design and synthesis of twenty-five novel quinoxaline-tethered imidazopyri(mi)dine derivatives. This was followed by comprehensive investigations into the impact of these molecules on the OC cell line. The in vitro cytotoxicity studies performed in CAL-27 and normal oral epithelial (NOE) cell lines revealed that some of the synthesized molecules like 12d have potent antiproliferative activity specifically towards OC cells with an IC50 of 0.79 μM and show negligible cytotoxicity over NOE cells. Further, 12d arrested cell growth in the S phase of the cell cycle and induced cell death by early apoptosis. The in silico studies validated that 12d binds to the activator binding site on pyruvate kinase M2 (PKM2) overexpressed in OC while the lactate dehydrogenase (LDH)-coupled enzyme assay established 12d as a potent PKM2 activator with an AC50 of 0.6 nM. Hence, this study provides fruitful evidence for the designed compounds as anticancer agents against OC.
Collapse
Affiliation(s)
- Asmita Choithramani
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A) Opposite Airforce Station, Palaj Gandhinagar Gujarat - 382355 India
| | - Rudradip Das
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A) Opposite Airforce Station, Palaj Gandhinagar Gujarat - 382355 India
| | - Gourav Bothra
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A) Opposite Airforce Station, Palaj Gandhinagar Gujarat - 382355 India
| | - Priyanka Patel Vatsa
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A) Opposite Airforce Station, Palaj Gandhinagar Gujarat - 382355 India
| | - Venkatesh Muthukumar
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A) Opposite Airforce Station, Palaj Gandhinagar Gujarat - 382355 India
| | - Bombothu Kavya Sai Bhuvana
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A) Opposite Airforce Station, Palaj Gandhinagar Gujarat - 382355 India
| | - Saumya Kapoor
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A) Opposite Airforce Station, Palaj Gandhinagar Gujarat - 382355 India
| | - Deepshika Moola
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A) Opposite Airforce Station, Palaj Gandhinagar Gujarat - 382355 India
| | - Moumita Ghosh Chowdhury
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A) Opposite Airforce Station, Palaj Gandhinagar Gujarat - 382355 India
| | - Amit Mandoli
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A) Opposite Airforce Station, Palaj Gandhinagar Gujarat - 382355 India
| | - Amit Shard
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A) Opposite Airforce Station, Palaj Gandhinagar Gujarat - 382355 India
| |
Collapse
|
3
|
Wu B, Liang Z, Lan H, Teng X, Wang C. The role of PKM2 in cancer progression and its structural and biological basis. J Physiol Biochem 2024; 80:261-275. [PMID: 38329688 DOI: 10.1007/s13105-024-01007-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 01/10/2024] [Indexed: 02/09/2024]
Abstract
Pyruvate kinase M2 (PKM2), a subtype of pyruvate kinase (PK), has been shown to play an important role in the development of cancer. It regulates the last step of glycolytic pathway. PKM2 has both pyruvate kinase and protein kinase activity, and the conversion of these two functions of PKM2 depends on the mutual change of dimer and tetramer. The dimerization of PKM2 can promote the proliferation and growth of tumor cells, so inhibiting the dimerization of PKM2 is essential to curing cancer. The aggregation of PKM2 is regulated by both endogenous and exogenous cofactors as well as post-translational modification (PTM). Although there are many studies on the different aggregation of PKM2 in the process of tumor development, there are few summaries in recent years. In this review, we first introduce the role of PKM2 in various biological processes of tumor growth. Then, we summarize the aggregation regulation mechanism of PKM2 by various endogenous cofactors such as Fructose-1, 6-diphosphate (FBP), various amino acids, and post-translational modification (PTMs). Finally, the related inhibitors and agonists of PKM2 are summarized to provide reference for regulating PKM2 aggregation in the treatment of cancer in the future.
Collapse
Affiliation(s)
- Bingxin Wu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Zuhui Liang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Huan Lan
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Xiaojun Teng
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Caiyan Wang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| |
Collapse
|
4
|
Dinkova-Kostova AT, Hakomäki H, Levonen AL. Electrophilic metabolites targeting the KEAP1/NRF2 partnership. Curr Opin Chem Biol 2024; 78:102425. [PMID: 38241876 DOI: 10.1016/j.cbpa.2024.102425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/03/2024] [Accepted: 01/03/2024] [Indexed: 01/21/2024]
Abstract
Numerous electrophilic metabolites are formed during cellular activity, particularly under conditions of oxidative, inflammatory and metabolic stress. Among them are lipid oxidation and nitration products, and compounds derived from amino acid and central carbon metabolism. Here we focus on one cellular target of electrophiles, the Kelch-like ECH associated protein 1 (KEAP1)/nuclear factor erythroid 2 p45-related factor 2 (NRF2) partnership. Many of these reactive compounds modify C151, C273 and/or C288 within KEAP1. Other types of modifications include S-lactoylation of C273, N-succinylation of K131, and formation of methylimidazole intermolecular crosslink between two KEAP1 monomers. Modified KEAP1 relays the initial signal to transcription factor NRF2 and its downstream targets, the ultimate effectors that provide means for detoxification, adaptation and survival. Thus, by non-enzymatically covalently modifying KEAP1, the electrophilic metabolites discussed here serve as chemical signals connecting metabolism with stress responses.
Collapse
Affiliation(s)
- Albena T Dinkova-Kostova
- Jacqui Wood Cancer Centre, Division of Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, UK; Department of Pharmacology and Molecular Sciences and Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Henriikka Hakomäki
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Anna-Liisa Levonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
5
|
Chintaluri C, Vogels TP. Metabolically regulated spiking could serve neuronal energy homeostasis and protect from reactive oxygen species. Proc Natl Acad Sci U S A 2023; 120:e2306525120. [PMID: 37988463 PMCID: PMC10691349 DOI: 10.1073/pnas.2306525120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 10/11/2023] [Indexed: 11/23/2023] Open
Abstract
So-called spontaneous activity is a central hallmark of most nervous systems. Such non-causal firing is contrary to the tenet of spikes as a means of communication, and its purpose remains unclear. We propose that self-initiated firing can serve as a release valve to protect neurons from the toxic conditions arising in mitochondria from lower-than-baseline energy consumption. To demonstrate the viability of our hypothesis, we built a set of models that incorporate recent experimental results indicating homeostatic control of metabolic products-Adenosine triphosphate (ATP), adenosine diphosphate (ADP), and reactive oxygen species (ROS)-by changes in firing. We explore the relationship of metabolic cost of spiking with its effect on the temporal patterning of spikes and reproduce experimentally observed changes in intrinsic firing in the fruitfly dorsal fan-shaped body neuron in a model with ROS-modulated potassium channels. We also show that metabolic spiking homeostasis can produce indefinitely sustained avalanche dynamics in cortical circuits. Our theory can account for key features of neuronal activity observed in many studies ranging from ion channel function all the way to resting state dynamics. We finish with a set of experimental predictions that would confirm an integrated, crucial role for metabolically regulated spiking and firmly link metabolic homeostasis and neuronal function.
Collapse
Affiliation(s)
- Chaitanya Chintaluri
- Institute of Science and Technology Austria, KlosterneuburgA-3400, Austria
- Centre for Neural Circuits and Behaviour, Department of Physiology, Anatomy and Genetics, University of Oxford, OxfordOX13SR, United Kingdom
| | - Tim P. Vogels
- Institute of Science and Technology Austria, KlosterneuburgA-3400, Austria
| |
Collapse
|
6
|
Ko Y, Hong M, Lee S, Kumar M, Ibrahim L, Nutsch K, Stanton C, Sondermann P, Sandoval B, Bulos ML, Iaconelli J, Chatterjee AK, Wiseman RL, Schultz PG, Bollong MJ. S-lactoyl modification of KEAP1 by a reactive glycolytic metabolite activates NRF2 signaling. Proc Natl Acad Sci U S A 2023; 120:e2300763120. [PMID: 37155889 PMCID: PMC10193962 DOI: 10.1073/pnas.2300763120] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 04/05/2023] [Indexed: 05/10/2023] Open
Abstract
KEAP1 (Kelch-like ECH-associated protein), a cytoplasmic repressor of the oxidative stress responsive transcription factor Nuclear factor erythroid 2-related factor 2 (NRF2), senses the presence of electrophilic agents by modification of its sensor cysteine residues. In addition to xenobiotics, several reactive metabolites have been shown to covalently modify key cysteines on KEAP1, although the full repertoire of these molecules and their respective modifications remain undefined. Here, we report the discovery of sAKZ692, a small molecule identified by high-throughput screening that stimulates NRF2 transcriptional activity in cells by inhibiting the glycolytic enzyme pyruvate kinase. sAKZ692 treatment promotes the buildup of glyceraldehyde 3-phosphate, a metabolite which leads to S-lactate modification of cysteine sensor residues of KEAP1, resulting in NRF2-dependent transcription. This work identifies a posttranslational modification of cysteine derived from a reactive central carbon metabolite and helps further define the complex relationship between metabolism and the oxidative stress-sensing machinery of the cell.
Collapse
Affiliation(s)
- Yeonjin Ko
- Department of Chemistry, The Scripps Research Institute, San Diego, CA92037
| | - Mannkyu Hong
- Department of Chemistry, The Scripps Research Institute, San Diego, CA92037
| | - Seungbeom Lee
- Department of Chemistry, The Scripps Research Institute, San Diego, CA92037
| | - Manoj Kumar
- Calibr, A Division of Scripps Research, San Diego, CA92037
| | - Lara Ibrahim
- Department of Chemistry, The Scripps Research Institute, San Diego, CA92037
- Department of Molecular Medicine, The Scripps Research Institute, San Diego, CA92037
| | - Kayla Nutsch
- Department of Chemistry, The Scripps Research Institute, San Diego, CA92037
| | - Caroline Stanton
- Department of Chemistry, The Scripps Research Institute, San Diego, CA92037
- Department of Molecular Medicine, The Scripps Research Institute, San Diego, CA92037
| | - Phillip Sondermann
- Department of Chemistry, The Scripps Research Institute, San Diego, CA92037
| | - Braddock Sandoval
- Department of Chemistry, The Scripps Research Institute, San Diego, CA92037
| | - Maya L. Bulos
- Department of Chemistry, The Scripps Research Institute, San Diego, CA92037
| | - Jonathan Iaconelli
- Department of Chemistry, The Scripps Research Institute, San Diego, CA92037
| | | | - R. Luke Wiseman
- Department of Molecular Medicine, The Scripps Research Institute, San Diego, CA92037
| | - Peter G. Schultz
- Department of Chemistry, The Scripps Research Institute, San Diego, CA92037
- Calibr, A Division of Scripps Research, San Diego, CA92037
| | - Michael J. Bollong
- Department of Chemistry, The Scripps Research Institute, San Diego, CA92037
| |
Collapse
|
7
|
Swint-Kruse L, Dougherty LL, Page B, Wu T, O’Neil PT, Prasannan CB, Timmons C, Tang Q, Parente DJ, Sreenivasan S, Holyoak T, Fenton AW. PYK-SubstitutionOME: an integrated database containing allosteric coupling, ligand affinity and mutational, structural, pathological, bioinformatic and computational information about pyruvate kinase isozymes. Database (Oxford) 2023; 2023:baad030. [PMID: 37171062 PMCID: PMC10176505 DOI: 10.1093/database/baad030] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 03/29/2023] [Accepted: 04/11/2023] [Indexed: 05/13/2023]
Abstract
Interpreting changes in patient genomes, understanding how viruses evolve and engineering novel protein function all depend on accurately predicting the functional outcomes that arise from amino acid substitutions. To that end, the development of first-generation prediction algorithms was guided by historic experimental datasets. However, these datasets were heavily biased toward substitutions at positions that have not changed much throughout evolution (i.e. conserved). Although newer datasets include substitutions at positions that span a range of evolutionary conservation scores, these data are largely derived from assays that agglomerate multiple aspects of function. To facilitate predictions from the foundational chemical properties of proteins, large substitution databases with biochemical characterizations of function are needed. We report here a database derived from mutational, biochemical, bioinformatic, structural, pathological and computational studies of a highly studied protein family-pyruvate kinase (PYK). A centerpiece of this database is the biochemical characterization-including quantitative evaluation of allosteric regulation-of the changes that accompany substitutions at positions that sample the full conservation range observed in the PYK family. We have used these data to facilitate critical advances in the foundational studies of allosteric regulation and protein evolution and as rigorous benchmarks for testing protein predictions. We trust that the collected dataset will be useful for the broader scientific community in the further development of prediction algorithms. Database URL https://github.com/djparente/PYK-DB.
Collapse
Affiliation(s)
- Liskin Swint-Kruse
- Department of Biochemistry and Molecular Biology, The University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - Larissa L Dougherty
- Department of Biochemistry and Molecular Biology, The University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - Braelyn Page
- Department of Biochemistry and Molecular Biology, The University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - Tiffany Wu
- Department of Biochemistry and Molecular Biology, The University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - Pierce T O’Neil
- Department of Biochemistry and Molecular Biology, The University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - Charulata B Prasannan
- Department of Biochemistry and Molecular Biology, The University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - Cody Timmons
- Chemistry Department, Southwestern Oklahoma State University, 100 Campus Dr., Weatherford, OK 73096, USA
| | - Qingling Tang
- Department of Biochemistry and Molecular Biology, The University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - Daniel J Parente
- Department of Biochemistry and Molecular Biology, The University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
- Department of Family Medicine and Community Health, The University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - Shwetha Sreenivasan
- Department of Biochemistry and Molecular Biology, The University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - Todd Holyoak
- Department of Biochemistry and Molecular Biology, The University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
- Department of Biology, University of Waterloo, 200 University Ave. W, Waterloo, ON N2L 3G1, Canada
| | - Aron W Fenton
- Department of Biochemistry and Molecular Biology, The University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| |
Collapse
|
8
|
Hicks KG, Cluntun AA, Schubert HL, Hackett SR, Berg JA, Leonard PG, Ajalla Aleixo MA, Zhou Y, Bott AJ, Salvatore SR, Chang F, Blevins A, Barta P, Tilley S, Leifer A, Guzman A, Arok A, Fogarty S, Winter JM, Ahn HC, Allen KN, Block S, Cardoso IA, Ding J, Dreveny I, Gasper WC, Ho Q, Matsuura A, Palladino MJ, Prajapati S, Sun P, Tittmann K, Tolan DR, Unterlass J, VanDemark AP, Vander Heiden MG, Webb BA, Yun CH, Zhao P, Wang B, Schopfer FJ, Hill CP, Nonato MC, Muller FL, Cox JE, Rutter J. Protein-metabolite interactomics of carbohydrate metabolism reveal regulation of lactate dehydrogenase. Science 2023; 379:996-1003. [PMID: 36893255 PMCID: PMC10262665 DOI: 10.1126/science.abm3452] [Citation(s) in RCA: 42] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 02/07/2023] [Indexed: 03/11/2023]
Abstract
Metabolic networks are interconnected and influence diverse cellular processes. The protein-metabolite interactions that mediate these networks are frequently low affinity and challenging to systematically discover. We developed mass spectrometry integrated with equilibrium dialysis for the discovery of allostery systematically (MIDAS) to identify such interactions. Analysis of 33 enzymes from human carbohydrate metabolism identified 830 protein-metabolite interactions, including known regulators, substrates, and products as well as previously unreported interactions. We functionally validated a subset of interactions, including the isoform-specific inhibition of lactate dehydrogenase by long-chain acyl-coenzyme A. Cell treatment with fatty acids caused a loss of pyruvate-lactate interconversion dependent on lactate dehydrogenase isoform expression. These protein-metabolite interactions may contribute to the dynamic, tissue-specific metabolic flexibility that enables growth and survival in an ever-changing nutrient environment.
Collapse
Affiliation(s)
- Kevin G Hicks
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Ahmad A Cluntun
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Heidi L Schubert
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | | | - Jordan A Berg
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Paul G Leonard
- Core for Biomolecular Structure and Function, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Institute for Applied Cancer Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mariana A Ajalla Aleixo
- Laboratório de Cristalografia de Proteinas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Youjia Zhou
- School of Computing, University of Utah, Salt Lake City, UT, USA
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, UT, USA
| | - Alex J Bott
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Sonia R Salvatore
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Fei Chang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Aubrie Blevins
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Paige Barta
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Samantha Tilley
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Aaron Leifer
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Andrea Guzman
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Ajak Arok
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Sarah Fogarty
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
- Howard Hughes Medical Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Jacob M Winter
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Hee-Chul Ahn
- Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang, The Republic of Korea
| | - Karen N Allen
- Department of Chemistry, Boston University, Boston, MA, USA
| | - Samuel Block
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Iara A Cardoso
- Laboratório de Cristalografia de Proteinas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Jianping Ding
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Shanghai, China
| | - Ingrid Dreveny
- Biodiscovery Institute, School of Pharmacy, University of Nottingham, Nottingham, UK
| | | | - Quinn Ho
- Department of Biology, Boston University, Boston, MA, USA
| | - Atsushi Matsuura
- Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang, The Republic of Korea
| | - Michael J Palladino
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sabin Prajapati
- Department of Molecular Enzymology, Göttingen Center of Molecular Biosciences, University of Göttingen, Göttingen, Germany
- Department of Structural Dynamics, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Pengkai Sun
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Shanghai, China
| | - Kai Tittmann
- Department of Molecular Enzymology, Göttingen Center of Molecular Biosciences, University of Göttingen, Göttingen, Germany
- Department of Structural Dynamics, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Dean R Tolan
- Department of Biology, Boston University, Boston, MA, USA
| | - Judith Unterlass
- Department of Oncology and Pathology, Karolinska Institute, Stockholm, Sweden
| | - Andrew P VanDemark
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Matthew G Vander Heiden
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Bradley A Webb
- Department of Biochemistry, West Virginia University, Morgantown, WV, USA
| | - Cai-Hong Yun
- Department of Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Pengkai Zhao
- Department of Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Bei Wang
- School of Computing, University of Utah, Salt Lake City, UT, USA
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, UT, USA
| | - Francisco J Schopfer
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Pittsburgh, PA, USA
- Pittsburgh Liver Research Center, Pittsburgh, PA, USA
- Center for Metabolism and Mitochondrial Medicine, Pittsburgh, PA, USA
| | - Christopher P Hill
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Maria Cristina Nonato
- Laboratório de Cristalografia de Proteinas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Florian L Muller
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James E Cox
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Jared Rutter
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
- Howard Hughes Medical Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| |
Collapse
|
9
|
Nandi S, Dey M. Identification of residues involved in allosteric signal transmission from amino acid binding site of pyruvate kinase muscle isoform 2. PLoS One 2023; 18:e0282508. [PMID: 36897854 PMCID: PMC10004559 DOI: 10.1371/journal.pone.0282508] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 02/16/2023] [Indexed: 03/11/2023] Open
Abstract
PKM2 is a rate-limiting enzyme in the glycolytic process and is involved in regulating tumor proliferation. Several amino acids (AAs) such as Asn, Asp, Val, and Cys have been shown to bind to the AA binding pocket of PKM2 and modulate its oligomeric state, substrate binding affinity, and activity. Although previous studies have attributed that the main chain and side chain of bound AAs are responsible for initiating signal to regulate PKM2, the signal transduction pathway remains elusive. To identify the residues involved in signal transfer process, N70 and N75 located at two ends of a β strand connecting the active site and AA binding pocket were altered. Biochemical studies of these variants with various AA ligands (Asn, Asp, Val, and Cys), illustrate that N70 and N75, along with β1 connecting these residues are part of the signal transduction pathway between the AA binding pocket and the active site. The results demonstrate that mutation of N70 to D prevents the transfer of the inhibitory signal mediated by Val and Cys, whereas N75 to L alteration blocks the activating signal initiated by Asn and Asp. Taken together, this study confirms that N70 is one of the residues responsible for transmitting the inhibitory signal and N75 is involved in the activation signal flow.
Collapse
Affiliation(s)
- Suparno Nandi
- Department of Chemistry, The University of Iowa, Iowa City, IA, United States of America
| | - Mishtu Dey
- Department of Chemistry, The University of Iowa, Iowa City, IA, United States of America
| |
Collapse
|
10
|
Marciniec K, Rzepka Z, Chrobak E, Boryczka S, Latocha M, Wrześniok D, Beberok A. Design, Synthesis and Biological Evaluation of Quinoline-8-Sulfonamides as Inhibitors of the Tumor Cell-Specific M2 Isoform of Pyruvate Kinase: Preliminary Study. Molecules 2023; 28:molecules28062509. [PMID: 36985481 PMCID: PMC10058396 DOI: 10.3390/molecules28062509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 02/28/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
Cancer cells need to carefully regulate their metabolism to keep them growing and dividing under the influence of different nutrients and oxygen levels. Muscle isoform 2 of pyruvate kinase (PKM2) is a key glycolytic enzyme involved in the generation of ATP and is critical for cancer metabolism. PKM2 is expressed in many human tumors and is regulated by complex mechanisms that promote tumor growth and proliferation. Therefore, it is considered an attractive therapeutic target for modulating tumor metabolism. Various modulators regulate PKM2, shifting it between highly active and less active states. In the presented work, a series of 8-quinolinesulfonamide derivatives of PKM2 modulators were designed using molecular docking and molecular dynamics techniques. New compounds were synthesized using the copper-catalyzed azide-alkyne cycloaddition (CuAAC) reaction. Compound 9a was identified in in silico studies as a potent modulator of muscle isoform 2 of pyruvate kinase. The results obtained from in vitro experiments confirmed the ability of compound 9a to reduce the intracellular pyruvate level in A549 lung cancer cells with simultaneous impact on cancer cell viability and cell-cycle phase distribution. Moreover, compound 9a exhibited more cytotoxicity on cancer cells than normal cells, pointing to high selectivity in the mode of action. These findings indicate that the introduction of another quinolinyl fragment to the modulator molecule may have a significant impact on pyruvate levels in cancer cells and provides further directions for future research to find novel analogs suitable for clinical applications in cancer treatment.
Collapse
Affiliation(s)
- Krzysztof Marciniec
- Department of Organic Chemistry, Medical University of Silesia, Jagiellońska 4, 41-200 Sosnowiec, Poland
- Correspondence:
| | - Zuzanna Rzepka
- Department of Pharmaceutical Chemistry, Jagiellońska 4, 41-200 Sosnowiec, Poland
| | - Elwira Chrobak
- Department of Organic Chemistry, Medical University of Silesia, Jagiellońska 4, 41-200 Sosnowiec, Poland
| | - Stanisław Boryczka
- Department of Organic Chemistry, Medical University of Silesia, Jagiellońska 4, 41-200 Sosnowiec, Poland
| | - Małgorzata Latocha
- Department of Molecular Biology, Jagiellońska 4, 41-200 Sosnowiec, Poland
| | - Dorota Wrześniok
- Department of Pharmaceutical Chemistry, Jagiellońska 4, 41-200 Sosnowiec, Poland
| | - Artur Beberok
- Department of Pharmaceutical Chemistry, Jagiellońska 4, 41-200 Sosnowiec, Poland
| |
Collapse
|
11
|
Ellagic Acid and Its Metabolites as Potent and Selective Allosteric Inhibitors of Liver Pyruvate Kinase. Nutrients 2023; 15:nu15030577. [PMID: 36771285 PMCID: PMC9919951 DOI: 10.3390/nu15030577] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/14/2023] [Accepted: 01/18/2023] [Indexed: 01/25/2023] Open
Abstract
Liver pyruvate kinase (PKL) has recently emerged as a new target for non-alcoholic fatty liver disease (NAFLD), and inhibitors of this enzyme could represent a new therapeutic option. However, this breakthrough is complicated by selectivity issues since pyruvate kinase exists in four different isoforms. In this work, we report that ellagic acid (EA) and its derivatives, present in numerous fruits and vegetables, can inhibit PKL potently and selectively. Several polyphenolic analogues of EA were synthesized and tested to identify the chemical features responsible for the desired activity. Molecular modelling studies suggested that this inhibition is related to the stabilization of the PKL inactive state. This unique inhibition mechanism could potentially herald the development of new therapeutics for NAFLD.
Collapse
|
12
|
Jiang C, Zhao X, Jeong T, Kang JY, Park JH, Kim IS, Kim HS. Novel Specific Pyruvate Kinase M2 Inhibitor, Compound 3h, Induces Apoptosis and Autophagy through Suppressing Akt/mTOR Signaling Pathway in LNCaP Cells. Cancers (Basel) 2022; 15:cancers15010265. [PMID: 36612260 PMCID: PMC9818605 DOI: 10.3390/cancers15010265] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 12/27/2022] [Accepted: 12/27/2022] [Indexed: 01/03/2023] Open
Abstract
Pyruvate kinase M2 (PKM2) is a key enzyme involved in the regulation of glycolysis. Although PKM2 is overexpressed in various tumor tissues, its functional role in cancer chemotherapy remains unexplored. In this study, we investigated the anticancer activity of a new PKM2 inhibitor, compound 3h, through the cell metabolism and associated signaling pathways in prostate cancer cells. To evaluate the molecular basis of specific PKM2 inhibitors, the interactions of compounds 3h and 3K with the PKM2 protein were assessed via molecular docking. We found that, compared to compound 3K, compound 3h exhibited a higher binding affinity for PKM2. Moreover, compound 3h significantly inhibited the pyruvate kinase activity and PKM2 expression. Cytotoxicity and colony formation assays revealed the potent anticancer activity of compound 3h against LNCaP cells. Compound 3h significantly increased the apoptotic and autophagic cell death in LNCaP cells. In addition, compound 3h induced AMPK activation along with the inhibition of the mTOR/p70S6K pathway. Furthermore, compound 3h significantly inhibited glycolysis and mitochondrial respiration, as determined by analyzing the extracellular acidification rate (ECAR) and oxygen consumption rate (OCR) production. Our results revealed that compound 3h caused apoptotic and autophagic cell death in LNCaP cells by inhibiting cancer cell metabolism. Therefore, blocking glycolytic pathways using specific PKM2 inhibitors can target cancer cell metabolism in PKM2-overexpressed prostate cancer cells.
Collapse
Affiliation(s)
- Chunxue Jiang
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Xiaodi Zhao
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Taejoo Jeong
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Ju Young Kang
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jae Hyeon Park
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - In Su Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Hyung Sik Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Correspondence: ; Tel.: +82-31-290-7789; Fax: +82-31-290-7748
| |
Collapse
|
13
|
Zhu J, Chen H, Le Y, Guo J, Liu Z, Dou X, Lu D. Salvianolic acid A regulates pyroptosis of endothelial cells via directly targeting PKM2 and ameliorates diabetic atherosclerosis. Front Pharmacol 2022; 13:1009229. [PMID: 36425580 PMCID: PMC9679534 DOI: 10.3389/fphar.2022.1009229] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 10/13/2022] [Indexed: 11/09/2023] Open
Abstract
Rescuing endothelial cells from pyroptotic cell death emerges as a potential therapeutic strategy to combat diabetic atherosclerosis. Salvianolic acid A (SAA) is a major water-soluble phenolic acid in the Salvia miltiorrhiza Bunge, which has been used in traditional Chinese medicine (TCM) and health food products for a long time. This study investigated whether SAA-regulated pyruvate kinase M2 (PKM2) functions to protect endothelial cells. In streptozotocin (STZ)-induced diabetic ApoE-/- mice subjected to a Western diet, SAA attenuated atherosclerotic plaque formation and inhibited pathological changes in the aorta. In addition, SAA significantly prevented NLRP3 inflammasome activation and pyroptosis of endothelial cells in the diabetic atherosclerotic aortic sinus or those exposed to high glucose. Mechanistically, PKM2 was verified to be the main target of SAA. We further revealed that SAA directly interacts with PKM2 at its activator pocket, inhibits phosphorylation of Y105, and hinders the nuclear translocation of PKM2. Also, SAA consistently decreased high glucose-induced overproduction of lactate and partially lactate-dependent phosphorylation of PKR (a regulator of the NLRP3 inflammasome). Further assay on Phenylalanine (PKM2 activity inhibitor) proved that SAA exhibits the function in high glucose-induced pyroptosis of endothelial cells dependently on PKM2 regulation. Furthermore, an assay on c16 (inhibitor of PKR activity) with co-phenylalanine demonstrated that the regulation of the phosphorylated PKR partially drives PKM2-dependent SAA modulation of cell pyroptosis. Therefore, this article reports on the novel function of SAA in the pyroptosis of endothelial cells and diabetic atherosclerosis, which provides important insights into immunometabolism reprogramming that is important for diabetic cardiovascular disease complications therapy.
Collapse
Affiliation(s)
- Ji Zhu
- The Third School of Clinical Medicine (School of Rehabilitation Medicine), Zhejiang Chinese Medical University, Hangzhou, China
- The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Hang Chen
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yifei Le
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jianan Guo
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhijun Liu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaobing Dou
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Dezhao Lu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
14
|
Data-driven and model-guided systematic framework for media development in CHO cell culture. Metab Eng 2022; 73:114-123. [DOI: 10.1016/j.ymben.2022.07.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 06/20/2022] [Accepted: 07/01/2022] [Indexed: 11/21/2022]
|
15
|
Arora S, Joshi G, Chaturvedi A, Heuser M, Patil S, Kumar R. A Perspective on Medicinal Chemistry Approaches for Targeting Pyruvate Kinase M2. J Med Chem 2022; 65:1171-1205. [PMID: 34726055 DOI: 10.1021/acs.jmedchem.1c00981] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The allosteric regulation of pyruvate kinase M2 (PKM2) affects the switching of the PKM2 protein between the high-activity and low-activity states that allow ATP and lactate production, respectively. PKM2, in its low catalytic state (dimeric form), is chiefly active in metabolically energetic cells, including cancer cells. More recently, PKM2 has emerged as an attractive target due to its role in metabolic dysfunction and other interrelated conditions. PKM2 (dimer) activity can be inhibited by modulating PKM2 dimer-tetramer dynamics using either PKM2 inhibitors that bind at the ATP binding active site of PKM2 (dimer) or PKM2 activators that bind at the allosteric site of PKM2, thus activating PKM2 from the dimer formation to the tetrameric formation. The present perspective focuses on medicinal chemistry approaches to design and discover PKM2 inhibitors and activators and further provides a scope for the future design of compounds targeting PKM2 with better efficacy and selectivity.
Collapse
Affiliation(s)
- Sahil Arora
- Laboratory for Drug Design and Synthesis, Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda 151401, India
| | - Gaurav Joshi
- Laboratory for Drug Design and Synthesis, Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda 151401, India
- School of Pharmacy, Graphic Era Hill University, Dehradun, Uttarakhand 248171, India
| | - Anuhar Chaturvedi
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover 30625, Germany
| | - Michael Heuser
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover 30625, Germany
| | - Santoshkumar Patil
- Discovery Services, Syngene International Ltd., Biocon Park, SEZ, Bommasandra Industrial Area-Phase-IV, Bommasandra-Jigani Link Road, Bengaluru, Karnataka 560099, India
| | - Raj Kumar
- Laboratory for Drug Design and Synthesis, Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda 151401, India
| |
Collapse
|
16
|
Celebi M, Inan T, Kurkcuoglu O, Akten ED. Potential allosteric sites captured in glycolytic enzymes via residue-based network models: Phosphofructokinase, glyceraldehyde-3-phosphate dehydrogenase and pyruvate kinase. Biophys Chem 2021; 280:106701. [PMID: 34736071 DOI: 10.1016/j.bpc.2021.106701] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/12/2021] [Accepted: 10/19/2021] [Indexed: 01/05/2023]
Abstract
Likelihood of new allosteric sites for glycolytic enzymes, phosphofructokinase (PFK), glyceraldehyde-3-phosphate dehydrogenase (GADPH) and pyruvate kinase (PK) was evaluated for bacterial, parasitic and human species. Allosteric effect of a ligand binding at a site was revealed on the basis of low-frequency normal modes via Cα-harmonic residue network model. In bacterial PFK, perturbation of the proposed allosteric site outperformed the known allosteric one, producing a high amount of stabilization or reduced dynamics, on all catalytic regions. Another proposed allosteric spot at the dimer interface in parasitic PFK exhibited major stabilization effect on catalytic regions. In parasitic GADPH, the most desired allosteric response was observed upon perturbation of its tunnel region which incorporated key residues for functional regulation. Proposed allosteric site in bacterial PK produced a satisfactory allosteric response on all catalytic regions, whereas in human and parasitic PKs, a partial inhibition was observed. Residue network model based solely on contact topology identified the 'hub residues' with high betweenness tracing plausible allosteric communication pathways between distant functional sites. For both bacterial PFK and PK, proposed sites accommodated hub residues twice as much as the known allosteric site. Tunnel region in parasitic GADPH with the strongest allosteric effect among species, incorporated the highest number of hub residues. These results clearly suggest a one-to-one correspondence between the degree of allosteric effect and the number of hub residues in that perturbation site, which increases the likelihood of its allosteric nature.
Collapse
Affiliation(s)
- Metehan Celebi
- Graduate Program of Computational Biology and Bioinformatics, Graduate School of Science and Engineering, Kadir Has University, Istanbul, Turkey
| | - Tugce Inan
- Department of Chemical Engineering, Istanbul Technical University, Istanbul, Turkey
| | - Ozge Kurkcuoglu
- Department of Chemical Engineering, Istanbul Technical University, Istanbul, Turkey
| | - Ebru Demet Akten
- Department of Bioinformatics and Genetics, Faculty of Engineering and Natural Sciences, Kadir Has University, Istanbul, Turkey.
| |
Collapse
|
17
|
Yang GJ, Wu J, Leung CH, Ma DL, Chen J. A review on the emerging roles of pyruvate kinase M2 in anti-leukemia therapy. Int J Biol Macromol 2021; 193:1499-1506. [PMID: 34740687 DOI: 10.1016/j.ijbiomac.2021.10.213] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/28/2021] [Accepted: 10/28/2021] [Indexed: 12/12/2022]
Abstract
Glycolysis is an important step in respiration and provides energy for cellular processes. Pyruvate kinase M2 (PKM2), a key rate-limiting enzyme of glycolysis, plays an important role in tumor cell metabolism and proliferation. It is also specifically overexpressed in leukemia cells and contributes to leukemic proliferation, differentiation, and drug resistance through both aerobic glycolysis and non-metabolic pathways. In this review, the functions and regulatory roles of PKM2 are firstly introduced. Then, the molecular mechanisms of PKM2 in leukemogenesis are summarized. Next, reported PKM2 modulators and their anti-leukemia mechanisms are described. Finally, the current challenges and the potential opportunities of PKM2 inhibitors or agonists in leukemia therapy are discussed.
Collapse
Affiliation(s)
- Guan-Jun Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo 315211, Zhejiang, China; Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao SAR, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China
| | - Jia Wu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao SAR, China
| | - Chung-Hang Leung
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao SAR, China; Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, 999078, Macao SAR, China.
| | - Dik-Lung Ma
- Department of Chemistry, Hong Kong Baptist University, Kowloon 999077, Hong Kong, China.
| | - Jiong Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo 315211, Zhejiang, China; Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao SAR, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, China.
| |
Collapse
|
18
|
Coassolo S, Davidson I. Regulation of glycolysis and cancer cell proliferation by PKM2 citrullination. Mol Cell Oncol 2021; 8:1927446. [PMID: 34616867 DOI: 10.1080/23723556.2021.1927446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Conversion of peptidyl-arginine to peptidyl citrulline, known as citrullination, is a post-translational protein modification catalyzed by the PADI (Protein Arginine Deiminase) family of enzymes. PADI1 and PADI3 catalyze citrullination of arginine 106 in the glycolytic enzyme pyruvate kinase M2 modulating its allosteric regulation, glycolysis and cancer cell proliferation.
Collapse
Affiliation(s)
- Sébastien Coassolo
- Equipe Labélisée Ligue Contre le Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch Cedex, France.,Centre National de la Recherche Scientifique, UMR7104. Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U1258. Illkirch, France.,Université de Strasbourg, Strasbourg, France
| | - Irwin Davidson
- Equipe Labélisée Ligue Contre le Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch Cedex, France.,Centre National de la Recherche Scientifique, UMR7104. Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U1258. Illkirch, France.,Université de Strasbourg, Strasbourg, France
| |
Collapse
|
19
|
Lee YB, Min JK, Kim JG, Cap KC, Islam R, Hossain AJ, Dogsom O, Hamza A, Mahmud S, Choi DR, Kim YS, Koh YH, Kim HA, Chung WS, Suh SW, Park JB. Multiple functions of pyruvate kinase M2 in various cell types. J Cell Physiol 2021; 237:128-148. [PMID: 34311499 DOI: 10.1002/jcp.30536] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/28/2021] [Accepted: 07/13/2021] [Indexed: 02/06/2023]
Abstract
Glucose metabolism is a mechanism by which energy is produced in form of adenosine triphosphate (ATP) by mitochondria and precursor metabolites are supplied to enable the ultimate enrichment of mature metabolites in the cell. Recently, glycolytic enzymes have been shown to have unconventional but important functions. Among these enzymes, pyruvate kinase M2 (PKM2) plays several roles including having conventional metabolic enzyme activity, and also being a transcriptional regulator and a protein kinase. Compared with the closely related PKM1, PKM2 is highly expressed in cancer cells and embryos, whereas PKM1 is dominant in mature, differentiated cells. Posttranslational modifications such as phosphorylation and acetylation of PKM2 change its cellular functions. In particular, PKM2 can translocate to the nucleus, where it regulates the transcription of many target genes. It is notable that PKM2 also acts as a protein kinase to phosphorylate several substrate proteins. Besides cancer cells and embryonic cells, astrocytes also highly express PKM2, which is crucial for lactate production via expression of lactate dehydrogenase A (LDHA), while mature neurons predominantly express PKM1. The lactate produced in cancer cells promotes tumor progress and that in astrocytes can be supplied to neurons and may act as a major source for neuronal ATP energy production. Thereby, we propose that PKM2 along with its different posttranslational modifications has specific purposes for a variety of cell types, performing unique functions.
Collapse
Affiliation(s)
- Yoon-Beom Lee
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Jung K Min
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Jae-Gyu Kim
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, Republic of Korea.,Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Kim Cuong Cap
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, Republic of Korea.,eLmed Inc. #3419, Hallym University, Chuncheon, Kangwon-do, Republic of Korea.,Institute of Research and Development, Duy Tan University, Danang, Vietnam
| | - Rokibul Islam
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, Republic of Korea.,Department of Biotechnology and Genetic Engineering, Faculty of Biological Science, Islamic University, Kushtia, Bangladesh
| | - Abu J Hossain
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Oyungerel Dogsom
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, Republic of Korea.,Department of Biology, School of Bio-Medicine, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Amir Hamza
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Shohel Mahmud
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, Republic of Korea.,National Institute of Biotechnology, Ganakbari, Savar, Dhaka, Bangladesh
| | - Dae R Choi
- Department of Internal Medicine, Chuncheon Sacred Heart Hospital, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Yong-Sun Kim
- Ilsong Institute of Life Science, Hallym University, Seoul, Republic of Korea
| | - Young-Ho Koh
- Ilsong Institute of Life Science, Hallym University, Seoul, Republic of Korea
| | - Hyun-A Kim
- Department of Internal Medicine, Hallym Sacred Heart Hospital, College of Medicine, Hallym University, Ahnyang, Republic of Korea
| | - Won-Suk Chung
- Department of Biological Science, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Sang W Suh
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Jae-Bong Park
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, Republic of Korea.,Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon, Republic of Korea.,eLmed Inc. #3419, Hallym University, Chuncheon, Kangwon-do, Republic of Korea
| |
Collapse
|
20
|
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: 13] [Impact Index Per Article: 4.3] [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
|
21
|
Michels PAM, Villafraz O, Pineda E, Alencar MB, Cáceres AJ, Silber AM, Bringaud F. Carbohydrate metabolism in trypanosomatids: New insights revealing novel complexity, diversity and species-unique features. Exp Parasitol 2021; 224:108102. [PMID: 33775649 DOI: 10.1016/j.exppara.2021.108102] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/13/2021] [Accepted: 03/18/2021] [Indexed: 12/16/2022]
Abstract
The human pathogenic trypanosomatid species collectively called the "TriTryp parasites" - Trypanosoma brucei, Trypanosoma cruzi and Leishmania spp. - have complex life cycles, with each of these parasitic protists residing in a different niche during their successive developmental stages where they encounter diverse nutrients. Consequently, they adapt their metabolic network accordingly. Yet, throughout the life cycles, carbohydrate metabolism - involving the glycolytic, gluconeogenic and pentose-phosphate pathways - always plays a central role in the biology of these parasites, whether the available carbon and free energy sources are saccharides, amino acids or lipids. In this paper, we provide an updated review of the carbohydrate metabolism of the TriTryps, highlighting new data about this metabolic network, the interconnection of its pathways and the compartmentalisation of its enzymes within glycosomes, cytosol and mitochondrion. Differences in the expression of the branches of the metabolic network between the successive life-cycle stages of each of these parasitic trypanosomatids are discussed, as well as differences between them. Recent structural and kinetic studies have revealed unique regulatory mechanisms for some of the network's key enzymes with important species-specific variations. Furthermore, reports of multiple post-translational modifications of trypanosomal glycolytic enzymes suggest that additional mechanisms for stage- and/or environmental cues that regulate activity are operational in the parasites. The detailed comparison of the carbohydrate metabolism of the TriTryps has thus revealed multiple differences and a greater complexity, including for the reduced metabolic network in bloodstream-form T. brucei, than previously appreciated. Although these parasites are related, share many cytological and metabolic features and are grouped within a single taxonomic family, the differences highlighted in this review reflect their separate evolutionary tracks from a common ancestor to the extant organisms. These differences are indicative of their adaptation to the different insect vectors and niches occupied in their mammalian hosts.
Collapse
Affiliation(s)
- Paul A M Michels
- Centre for Immunity, Infection and Evolution and Centre for Translational and Chemical Biology, School of Biological Sciences, The University of Edinburgh, Edinburgh, United Kingdom.
| | - Oriana Villafraz
- Laboratoire de Microbiologie Fondamentale et Pathogénicité (MFP), Université de Bordeaux, CNRS UMR-5234, France
| | - Erika Pineda
- Laboratoire de Microbiologie Fondamentale et Pathogénicité (MFP), Université de Bordeaux, CNRS UMR-5234, France
| | - Mayke B Alencar
- Laboratory of Biochemistry of Tryps, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, 05508-000, Brazil
| | - Ana J Cáceres
- Laboratorio de Enzimología de Parásitos, Departamento de Biología, Facultad de Ciencias, Universidad de Los Andes, Mérida, 5101, Venezuela.
| | - Ariel M Silber
- Laboratory of Biochemistry of Tryps, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, 05508-000, Brazil.
| | - Frédéric Bringaud
- Laboratoire de Microbiologie Fondamentale et Pathogénicité (MFP), Université de Bordeaux, CNRS UMR-5234, France.
| |
Collapse
|
22
|
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: 26] [Impact Index Per Article: 8.7] [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
|
23
|
Pasquale V, Ducci G, Campioni G, Ventrici A, Assalini C, Busti S, Vanoni M, Vago R, Sacco E. Profiling and Targeting of Energy and Redox Metabolism in Grade 2 Bladder Cancer Cells with Different Invasiveness Properties. Cells 2020; 9:cells9122669. [PMID: 33322565 PMCID: PMC7764708 DOI: 10.3390/cells9122669] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/07/2020] [Accepted: 12/08/2020] [Indexed: 12/14/2022] Open
Abstract
Bladder cancer is one of the most prevalent deadly diseases worldwide. Grade 2 tumors represent a good window of therapeutic intervention, whose optimization requires high resolution biomarker identification. Here we characterize energy metabolism and cellular properties associated with spreading and tumor progression of RT112 and 5637, two Grade 2 cancer cell lines derived from human bladder, representative of luminal-like and basal-like tumors, respectively. The two cell lines have similar proliferation rates, but only 5637 cells show efficient lateral migration. In contrast, RT112 cells are more prone to form spheroids. RT112 cells produce more ATP by glycolysis and OXPHOS, present overall higher metabolic plasticity and are less sensitive than 5637 to nutritional perturbation of cell proliferation and migration induced by treatment with 2-deoxyglucose and metformin. On the contrary, spheroid formation is less sensitive to metabolic perturbations in 5637 than RT112 cells. The ability of metformin to reduce, although with different efficiency, cell proliferation, sphere formation and migration in both cell lines, suggests that OXPHOS targeting could be an effective strategy to reduce the invasiveness of Grade 2 bladder cancer cells.
Collapse
Affiliation(s)
- Valentina Pasquale
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (V.P.); (G.D.); (G.C.); (A.V.); (S.B.)
- SYSBIO-ISBE-IT-Candidate National Node of Italy for ISBE, Research Infrastructure for Systems Biology Europe, 20126 Milan, Italy
| | - Giacomo Ducci
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (V.P.); (G.D.); (G.C.); (A.V.); (S.B.)
- SYSBIO-ISBE-IT-Candidate National Node of Italy for ISBE, Research Infrastructure for Systems Biology Europe, 20126 Milan, Italy
| | - Gloria Campioni
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (V.P.); (G.D.); (G.C.); (A.V.); (S.B.)
- SYSBIO-ISBE-IT-Candidate National Node of Italy for ISBE, Research Infrastructure for Systems Biology Europe, 20126 Milan, Italy
| | - Adria Ventrici
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (V.P.); (G.D.); (G.C.); (A.V.); (S.B.)
| | - Chiara Assalini
- Urological Research Institute, Division of Experimental Oncology, IRCCS San Raffaele Hospital, 20132 Milan, Italy;
| | - Stefano Busti
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (V.P.); (G.D.); (G.C.); (A.V.); (S.B.)
- SYSBIO-ISBE-IT-Candidate National Node of Italy for ISBE, Research Infrastructure for Systems Biology Europe, 20126 Milan, Italy
| | - Marco Vanoni
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (V.P.); (G.D.); (G.C.); (A.V.); (S.B.)
- SYSBIO-ISBE-IT-Candidate National Node of Italy for ISBE, Research Infrastructure for Systems Biology Europe, 20126 Milan, Italy
- Correspondence: (M.V.); (R.V.); (E.S.); Tel.: +39-02-6448-3525 (M.V.); +39-02-2643-5664 (R.V.); +39-02-6448-3379 (E.S.)
| | - Riccardo Vago
- Urological Research Institute, Division of Experimental Oncology, IRCCS San Raffaele Hospital, 20132 Milan, Italy;
- Università Vita-Salute San Raffaele, 20132 Milan, Italy
- Correspondence: (M.V.); (R.V.); (E.S.); Tel.: +39-02-6448-3525 (M.V.); +39-02-2643-5664 (R.V.); +39-02-6448-3379 (E.S.)
| | - Elena Sacco
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (V.P.); (G.D.); (G.C.); (A.V.); (S.B.)
- SYSBIO-ISBE-IT-Candidate National Node of Italy for ISBE, Research Infrastructure for Systems Biology Europe, 20126 Milan, Italy
- Correspondence: (M.V.); (R.V.); (E.S.); Tel.: +39-02-6448-3525 (M.V.); +39-02-2643-5664 (R.V.); +39-02-6448-3379 (E.S.)
| |
Collapse
|
24
|
Chinopoulos C. From Glucose to Lactate and Transiting Intermediates Through Mitochondria, Bypassing Pyruvate Kinase: Considerations for Cells Exhibiting Dimeric PKM2 or Otherwise Inhibited Kinase Activity. Front Physiol 2020; 11:543564. [PMID: 33335484 PMCID: PMC7736077 DOI: 10.3389/fphys.2020.543564] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 11/02/2020] [Indexed: 12/14/2022] Open
Abstract
A metabolic hallmark of many cancers is the increase in glucose consumption coupled to excessive lactate production. Mindful that L-lactate originates only from pyruvate, the question arises as to how can this be sustained in those tissues where pyruvate kinase activity is reduced due to dimerization of PKM2 isoform or inhibited by oxidative/nitrosative stress, posttranslational modifications or mutations, all widely reported findings in the very same cells. Hereby 17 pathways connecting glucose to lactate bypassing pyruvate kinase are reviewed, some of which transit through the mitochondrial matrix. An additional 69 converging pathways leading to pyruvate and lactate, but not commencing from glucose, are also examined. The minor production of pyruvate and lactate by glutaminolysis is scrutinized separately. The present review aims to highlight the ways through which L-lactate can still be produced from pyruvate using carbon atoms originating from glucose or other substrates in cells with kinetically impaired pyruvate kinase and underscore the importance of mitochondria in cancer metabolism irrespective of oxidative phosphorylation.
Collapse
|
25
|
Jain R, Özgümüş T, Jensen TM, du Plessis E, Keindl M, Møller CL, Falhammar H, Nyström T, Catrina SB, Jörneskog G, Jessen LE, Forsblom C, Haukka JK, Groop PH, Rossing P, Groop L, Eliasson M, Eliasson B, Brismar K, Al-Majdoub M, Nilsson PM, Taskinen MR, Ferrannini E, Spégel P, Berg TJ, Lyssenko V. Liver nucleotide biosynthesis is linked to protection from vascular complications in individuals with long-term type 1 diabetes. Sci Rep 2020; 10:11561. [PMID: 32665614 PMCID: PMC7360755 DOI: 10.1038/s41598-020-68130-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 05/29/2020] [Indexed: 12/11/2022] Open
Abstract
Identification of biomarkers associated with protection from developing diabetic complications is a prerequisite for an effective prevention and treatment. The aim of the present study was to identify clinical and plasma metabolite markers associated with freedom from vascular complications in people with very long duration of type 1 diabetes (T1D). Individuals with T1D, who despite having longer than 30 years of diabetes duration never developed major macro- or microvascular complications (non-progressors; NP) were compared with those who developed vascular complications within 25 years from diabetes onset (rapid progressors; RP) in the Scandinavian PROLONG (n = 385) and DIALONG (n = 71) cohorts. The DIALONG study also included 75 healthy controls. Plasma metabolites were measured using gas and/or liquid chromatography coupled to mass spectrometry. Lower hepatic fatty liver indices were significant common feature characterized NPs in both studies. Higher insulin sensitivity and residual ß-cell function (C-peptide) were also associated with NPs in PROLONG. Protection from diabetic complications was associated with lower levels of the glycolytic metabolite pyruvate and APOCIII in PROLONG, and with lower levels of thiamine monophosphate and erythritol, a cofactor and intermediate product in the pentose phosphate pathway as well as higher phenylalanine, glycine and serine in DIALONG. Furthermore, T1D individuals showed elevated levels of picolinic acid as compared to the healthy individuals. The present findings suggest a potential beneficial shunting of glycolytic substrates towards the pentose phosphate and one carbon metabolism pathways to promote nucleotide biosynthesis in the liver. These processes might be linked to higher insulin sensitivity and lower liver fat content, and might represent a mechanism for protection from vascular complications in individuals with long-term T1D.
Collapse
Affiliation(s)
- Ruchi Jain
- Department of Clinical Science/Diabetes and Endocrinology, Lund University Diabetes Centre, 205 02, Malmö, Sweden
| | - Türküler Özgümüş
- Department of Clinical Science, Center for Diabetes Research, University of Bergen, 5032, Bergen, Norway
| | - Troels Mygind Jensen
- Research Unit for General Practice, Danish Aging Research Center, University of Southern Denmark, Odense, Denmark.,Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Elsa du Plessis
- Department of Clinical Science, Center for Diabetes Research, University of Bergen, 5032, Bergen, Norway
| | - Magdalena Keindl
- Department of Clinical Science, Center for Diabetes Research, University of Bergen, 5032, Bergen, Norway
| | | | - Henrik Falhammar
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden.,Department of Endocrinology, Metabolism and Diabetes, Karolinska University Hospital, Stockholm, Sweden
| | - Thomas Nyström
- Department of Clinical Science and Education, Division of Internal Medicine, Unit for Diabetes Research, Karolinska Institute, South Hospital, Stockholm, Sweden
| | - Sergiu-Bogdan Catrina
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden.,Department of Endocrinology, Metabolism and Diabetes, Karolinska University Hospital, Stockholm, Sweden.,Center for Diabetes, Academica Specialist Centrum, Stockholm, Sweden
| | - Gun Jörneskog
- Department of Clinical Sciences, Division of Internal Medicine, Karolinska Institute, Danderyd University Hospital, Stockholm, Sweden
| | - Leon Eyrich Jessen
- Department of Health Technology, Section for Bioinformatics, Technical University of Denmark, Lyngby, Denmark
| | - Carol Forsblom
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Biomedicum Helsinki, Helsinki, Finland.,Research Programs for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jani K Haukka
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Biomedicum Helsinki, Helsinki, Finland.,Research Programs for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Per-Henrik Groop
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Biomedicum Helsinki, Helsinki, Finland.,Research Programs for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Peter Rossing
- Steno Diabetes Center Copenhagen, Gentofte, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Leif Groop
- Department of Clinical Science/Diabetes and Endocrinology, Lund University Diabetes Centre, 205 02, Malmö, Sweden.,Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland
| | - Mats Eliasson
- Department of Public Health and Clinical Medicine, Sunderby Research Unit, Umeå University, Umeå, Sweden
| | - Björn Eliasson
- Department of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Kerstin Brismar
- Department of Endocrinology, Metabolism and Diabetes, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Rolf Luft Center for Diabetes Research, Karolinska Institutet, Stockholm, Sweden.,Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Mahmoud Al-Majdoub
- Department of Clinical Science/Diabetes and Endocrinology, Lund University Diabetes Centre, 205 02, Malmö, Sweden
| | - Peter M Nilsson
- Department of Clinical Science/Diabetes and Endocrinology, Lund University Diabetes Centre, 205 02, Malmö, Sweden
| | - Marja-Riitta Taskinen
- Research Program Unit, Clinical and Molecular Metabolism, University of Helsinki, Helsinki, Finland
| | | | - Peter Spégel
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, 223 62, Lund, Sweden
| | - Tore Julsrud Berg
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Endocrinology, Oslo University Hospital, Oslo, Norway
| | - Valeriya Lyssenko
- Department of Clinical Science/Diabetes and Endocrinology, Lund University Diabetes Centre, 205 02, Malmö, Sweden. .,Department of Clinical Science, Center for Diabetes Research, University of Bergen, 5032, Bergen, Norway.
| |
Collapse
|
26
|
Nandi S, Dey M. Biochemical and structural insights into how amino acids regulate pyruvate kinase muscle isoform 2. J Biol Chem 2020; 295:5390-5403. [PMID: 32144209 PMCID: PMC7170521 DOI: 10.1074/jbc.ra120.013030] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/03/2020] [Indexed: 12/12/2022] Open
Abstract
Pyruvate kinase muscle isoform 2 (PKM2) is a key glycolytic enzyme involved in ATP generation and critical for cancer metabolism. PKM2 is expressed in many human cancers and is regulated by complex mechanisms that promote tumor growth and proliferation. Therefore, it is considered an attractive therapeutic target for modulating tumor metabolism. Various stimuli allosterically regulate PKM2 by cycling it between highly active and less active states. Several small molecules activate PKM2 by binding to its intersubunit interface. Serine and cysteine serve as an activator and inhibitor of PKM2, respectively, by binding to its amino acid (AA)-binding pocket, which therefore represents a potential druggable site. Despite binding similarly to PKM2, how cysteine and serine differentially regulate this enzyme remains elusive. Using kinetic analyses, fluorescence binding, X-ray crystallography, and gel filtration experiments with asparagine, aspartate, and valine as PKM2 ligands, we examined whether the differences in the side-chain polarity of these AAs trigger distinct allosteric responses in PKM2. We found that Asn (polar) and Asp (charged) activate PKM2 and that Val (hydrophobic) inhibits it. The results also indicate that both Asn and Asp can restore the activity of Val-inhibited PKM2. AA-bound crystal structures of PKM2 displayed distinctive interactions within the binding pocket, causing unique allosteric effects in the enzyme. These structure-function analyses of AA-mediated PKM2 regulation shed light on the chemical requirements in the development of mechanism-based small-molecule modulators targeting the AA-binding pocket of PKM2 and provide broader insights into the regulatory mechanisms of complex allosteric enzymes.
Collapse
Affiliation(s)
- Suparno Nandi
- Department of Chemistry, University of Iowa, Iowa City, Iowa 52242
| | - Mishtu Dey
- Department of Chemistry, University of Iowa, Iowa City, Iowa 52242.
| |
Collapse
|
27
|
Liu VM, Howell AJ, Hosios AM, Li Z, Israelsen WJ, Heiden MGV. Cancer-associated mutations in human pyruvate kinase M2 impair enzyme activity. FEBS Lett 2020; 594:646-664. [PMID: 31642061 PMCID: PMC7042059 DOI: 10.1002/1873-3468.13648] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 09/28/2019] [Accepted: 10/15/2019] [Indexed: 12/18/2022]
Abstract
Mammalian pyruvate kinase catalyzes the final step of glycolysis, and its M2 isoform (PKM2) is widely expressed in proliferative tissues. Mutations in PKM2 are found in some human cancers; however, the effects of these mutations on enzyme activity and regulation are unknown. Here, we characterized five cancer-associated PKM2 mutations, occurring at various locations on the enzyme, with respect to substrate kinetics and activation by the allosteric activator fructose-1,6-bisphosphate (FBP). The mutants exhibit reduced maximal velocity, reduced substrate affinity, and/or altered activation by FBP. The kinetic parameters of five additional PKM2 mutants that have been used to study enzyme function or regulation also demonstrate the deleterious effects of mutations on PKM2 function. Our findings indicate that PKM2 is sensitive to many amino acid changes and support the hypothesis that decreased PKM2 activity is selected for in rapidly proliferating cells.
Collapse
Affiliation(s)
- Vivian M. Liu
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States
- Harvard-MIT Health Sciences and Technology Division, Harvard Medical School, Boston, MA 02115, United States
| | - Andrea J. Howell
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States
| | - Aaron M. Hosios
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States
| | - Zhaoqi Li
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States
| | - William J. Israelsen
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Matthew G. Vander Heiden
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States
- Dana-Farber Cancer Institute, Boston, MA 02115, United States
| |
Collapse
|
28
|
Rashid S, Pho KB, Mesbahi H, MacNeil LT. Nutrient Sensing and Response Drive Developmental Progression in Caenorhabditis elegans. Bioessays 2020; 42:e1900194. [PMID: 32003906 DOI: 10.1002/bies.201900194] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 12/22/2019] [Indexed: 12/18/2022]
Abstract
In response to nutrient limitation, many animals, including Caenorhabditis elegans, slow or arrest their development. This process requires mechanisms that sense essential nutrients and induce appropriate responses. When faced with nutrient limitation, C. elegans can induce both short and long-term survival strategies, including larval arrest, decreased developmental rate, and dauer formation. To select the most advantageous strategy, information from many different sensors must be integrated into signaling pathways, including target of rapamycin (TOR) and insulin, that regulate developmental progression. Here, how nutrient information is sensed and integrated into developmental decisions that determine developmental rate and progression in C. elegans is reviewed.
Collapse
Affiliation(s)
- Sabih Rashid
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, L8S 4K1, Ontario, Canada
| | - Kim B Pho
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, L8S 4K1, Ontario, Canada
| | - Hiva Mesbahi
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, L8S 4K1, Ontario, Canada
| | - Lesley T MacNeil
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, L8S 4K1, Ontario, Canada.,Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, L8S 4K1, Ontario, Canada.,Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, L8S 4K1, Ontario, Canada
| |
Collapse
|
29
|
Bortoluzzi VT, Brust L, Preissler T, de Franceschi ID, Wannmacher CMD. Creatine plus pyruvate supplementation prevents oxidative stress and phosphotransfer network disturbances in the brain of rats subjected to chemically-induced phenylketonuria. Metab Brain Dis 2019; 34:1649-1660. [PMID: 31352540 DOI: 10.1007/s11011-019-00472-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 07/14/2019] [Indexed: 12/13/2022]
Abstract
Phenylketonuria (PKU) is the most common inborn error of amino acid metabolism. Usually diagnosed within the first month of birth, it is essential that the patient strictly follow the dietary restriction of natural protein intake. Otherwise, PKU impacts the development of the brain severely and may result in microcephaly, epilepsy, motor deficits, intellectual disability, and psychiatric and behavioral disorders. The neuropathology associated with PKU includes defects of myelination, insufficient synthesis of monoamine neurotransmitters, amino acid imbalance across the blood-brain barrier, and involves intermediary metabolic pathways supporting energy homeostasis and antioxidant defenses in the brain. Considering that the production of reactive oxygen species (ROS) is inherent to energy metabolism, we investigated the association of creatine+pyruvate (Cr + Pyr), both energy substrates with antioxidants properties, as a possible treatment to mitigate oxidative stress and phosphotransfer network impairment elicited in the brain of young Wistar rats by chemically-induced PKU. We induced PKU through the administration of α-methyl-L-phenylalanine and phenylalanine for 7 days, with and without Cr + Pyr supplementation, until postpartum day 14. The cotreatment with Cr + Pyr administered concurrently with PKU induction prevented ROS formation and part of the alterations observed in antioxidants defenses and phosphotransfer network enzymes in the cerebral cortex, hippocampus, and cerebellum. If such prevention also occurs in PKU patients, supplementing the phenylalanine-restricted diet with antioxidants and energetic substrates might be beneficial to these patients.
Collapse
Affiliation(s)
- Vanessa Trindade Bortoluzzi
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600-Anexo, Porto Alegre, RS, CEP 90.035-003, Brazil.
| | - Letícia Brust
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600-Anexo, Porto Alegre, RS, CEP 90.035-003, Brazil
| | - Thales Preissler
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600-Anexo, Porto Alegre, RS, CEP 90.035-003, Brazil
| | - Itiane Diehl de Franceschi
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600-Anexo, Porto Alegre, RS, CEP 90.035-003, Brazil
| | - Clovis Milton Duval Wannmacher
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600-Anexo, Porto Alegre, RS, CEP 90.035-003, Brazil
| |
Collapse
|
30
|
The molecular mechanisms of LncRNA-correlated PKM2 in cancer metabolism. Biosci Rep 2019; 39:220807. [PMID: 31654067 PMCID: PMC6851521 DOI: 10.1042/bsr20192453] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/27/2019] [Accepted: 10/01/2019] [Indexed: 12/17/2022] Open
Abstract
Reprogrammed metabolism is an important hallmark of cancer cells. Pyruvate kinase (PK) is one of the major rate-limiting enzymes in glucose metabolism. The M2 isoform of PK (PKM2), is considered to be an important marker of metabolic reprogramming and one of the key enzymes. Recently, through the continuous development of genome-wide analysis and functional studies, accumulating evidence has demonstrated that long non-coding RNAs (LncRNAs) play vital regulatory roles in cancer progression by acting as either potential oncogenes or tumor suppressors. Furthermore, several studies have shown that up-regulation of PKM2 in cancer tissues is associated with LncRNAs expression and patient survival. Thus, scientists have begun to unveil the mechanism of LncRNA-associated PKM2 in cancer metabolic progression. Based on these novel findings, in this mini-review, we summarize the detailed molecular mechanisms of LncRNA related to PKM2 in cancer metabolism. We expect that this work will promote a better understanding of the molecular mechanisms of PKM2, and provide a profound potential for targeting PKM2 to treat tumors.
Collapse
|
31
|
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: 88] [Impact Index Per Article: 17.6] [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
|
32
|
Zeng L, Yu Y, Cai X, Xie S, Chen J, Zhong L, Zhang Y. Differences in Serum Amino Acid Phenotypes Among Patients with Diabetic Nephropathy, Hypertensive Nephropathy, and Chronic Nephritis. Med Sci Monit 2019; 25:7235-7242. [PMID: 31557143 PMCID: PMC6778409 DOI: 10.12659/msm.915735] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Background We assessed levels of circulating amino acids in different etiologies of chronic kidney disease (CKD) and the association of amino acids with risk factors of CKD progression. Material/Methods High-performance liquid chromatography-based analysis was used to determine amino acid profiles in patients with diabetic nephropathy (DN, n=20), hypertensive nephropathy (HN, n=26), and chronic nephritis (CN, n=33), and in healthy controls (HC, n=25). Results All 3 types of CKD patients displayed decreased serum levels of serine, glycine, GABA, and tryptophan compared with healthy controls. Moreover, serine and tryptophan were positively correlated with glucose in DN cohorts. Total cholesterol was positively correlated with tryptophan levels in the DN cohort and negatively correlated with serine levels in the CN cohort. In the HN cohort, glycine was negatively correlated with triglyceride levels, and systolic blood pressure (SBP) was negatively correlated with GABA levels. Conclusions Patients with different etiologies of CKD have significantly different amino acids profiles, and this indicates specific supplementary nutritional needs in CKD patients.
Collapse
Affiliation(s)
- Li Zeng
- Department of Nephrology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China (mainland).,Chongqing Key Laboratory of Ultrasound Molecular Imaging, Ultrasound Department of Second Affiliated Hospital of Chongqing Medical University, Chongqing, China (mainland)
| | - Yuan Yu
- Department of Nephrology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China (mainland)
| | - Xi Cai
- Department of Nephrology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China (mainland)
| | - Shuqin Xie
- Department of Nephrology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China (mainland)
| | - Jianwei Chen
- Department of Nephrology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China (mainland)
| | - Ling Zhong
- Department of Nephrology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China (mainland)
| | - Ying Zhang
- Department of Nephrology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China (mainland)
| |
Collapse
|
33
|
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: 2.0] [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
|
34
|
Pyruvate Kinase Regulates the Pentose-Phosphate Pathway in Response to Hypoxia in Mycobacterium tuberculosis. J Mol Biol 2019; 431:3690-3705. [PMID: 31381898 DOI: 10.1016/j.jmb.2019.07.033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 07/25/2019] [Accepted: 07/27/2019] [Indexed: 11/21/2022]
Abstract
In response to the stress of infection, Mycobacterium tuberculosis (Mtb) reprograms its metabolism to accommodate nutrient and energetic demands in a changing environment. Pyruvate kinase (PYK) is an essential glycolytic enzyme in the phosphoenolpyruvate-pyruvate-oxaloacetate node that is a central switch point for carbon flux distribution. Here we show that the competitive binding of pentose monophosphate inhibitors or the activator glucose 6-phosphate (G6P) to MtbPYK tightly regulates the metabolic flux. Intriguingly, pentose monophosphates were found to share the same binding site with G6P. The determination of a crystal structure of MtbPYK with bound ribose 5-phosphate (R5P), combined with biochemical analyses and molecular dynamic simulations, revealed that the allosteric inhibitor pentose monophosphate increases PYK structural dynamics, weakens the structural network communication, and impairs substrate binding. G6P, on the other hand, primes and activates the tetramer by decreasing protein flexibility and strengthening allosteric coupling. Therefore, we propose that MtbPYK uses these differences in conformational dynamics to up- and down-regulate enzymic activity. Importantly, metabolome profiling in mycobacteria reveals a significant increase in the levels of pentose monophosphate during hypoxia, which provides insights into how PYK uses dynamics of the tetramer as a competitive allosteric mechanism to retard glycolysis and facilitate metabolic reprogramming toward the pentose-phosphate pathway for achieving redox balance and an anticipatory metabolic response in Mtb.
Collapse
|
35
|
Macpherson JA, Theisen A, Masino L, Fets L, Driscoll PC, Encheva V, Snijders AP, Martin SR, Kleinjung J, Barran PE, Fraternali F, Anastasiou D. Functional cross-talk between allosteric effects of activating and inhibiting ligands underlies PKM2 regulation. eLife 2019; 8:e45068. [PMID: 31264961 PMCID: PMC6636998 DOI: 10.7554/elife.45068] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 07/01/2019] [Indexed: 12/18/2022] Open
Abstract
Several enzymes can simultaneously interact with multiple intracellular metabolites, however, how the allosteric effects of distinct ligands are integrated to coordinately control enzymatic activity remains poorly understood. We addressed this question using, as a model system, the glycolytic enzyme pyruvate kinase M2 (PKM2). We show that the PKM2 activator fructose 1,6-bisphosphate (FBP) alone promotes tetramerisation and increases PKM2 activity, but addition of the inhibitor L-phenylalanine (Phe) prevents maximal activation of FBP-bound PKM2 tetramers. We developed a method, AlloHubMat, that uses eigenvalue decomposition of mutual information derived from molecular dynamics trajectories to identify residues that mediate FBP-induced allostery. Experimental mutagenesis of these residues identified PKM2 variants in which activation by FBP remains intact but cannot be attenuated by Phe. Our findings reveal residues involved in FBP-induced allostery that enable the integration of allosteric input from Phe and provide a paradigm for the coordinate regulation of enzymatic activity by simultaneous allosteric inputs.
Collapse
Affiliation(s)
- Jamie A Macpherson
- Cancer Metabolism LaboratoryThe Francis Crick InstituteLondonUnited Kingdom
- Randall Centre for Cell and Molecular BiophysicsKing’s College LondonLondonUnited Kingdom
| | - Alina Theisen
- Michael Barber Centre for Collaborative Mass Spectrometry, Manchester Institute of Biotechnology, School of ChemistryUniversity of ManchesterManchesterUnited Kingdom
| | - Laura Masino
- Structural Biology Science Technology PlatformThe Francis Crick InstituteLondonUnited Kingdom
| | - Louise Fets
- Cancer Metabolism LaboratoryThe Francis Crick InstituteLondonUnited Kingdom
| | - Paul C Driscoll
- Metabolomics Science Technology PlatformThe Francis Crick InstituteLondonUnited Kingdom
| | - Vesela Encheva
- Proteomics Science Technology PlatformThe Francis Crick InstituteLondonUnited Kingdom
| | - Ambrosius P Snijders
- Proteomics Science Technology PlatformThe Francis Crick InstituteLondonUnited Kingdom
| | - Stephen R Martin
- Structural Biology Science Technology PlatformThe Francis Crick InstituteLondonUnited Kingdom
| | - Jens Kleinjung
- Computational Biology Science Technology PlatformThe Francis Crick InstituteLondonUnited Kingdom
| | - Perdita E Barran
- Michael Barber Centre for Collaborative Mass Spectrometry, Manchester Institute of Biotechnology, School of ChemistryUniversity of ManchesterManchesterUnited Kingdom
| | - Franca Fraternali
- Randall Centre for Cell and Molecular BiophysicsKing’s College LondonLondonUnited Kingdom
| | | |
Collapse
|
36
|
A critical review of the role of M 2PYK in the Warburg effect. Biochim Biophys Acta Rev Cancer 2019; 1871:225-239. [PMID: 30708038 PMCID: PMC6525063 DOI: 10.1016/j.bbcan.2019.01.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 01/14/2019] [Accepted: 01/14/2019] [Indexed: 12/17/2022]
Abstract
It is becoming generally accepted in recent literature that the Warburg effect in cancer depends on inhibition of M2PYK, the pyruvate kinase isozyme most commonly expressed in tumors. We remain skeptical. There continues to be a general lack of solid experimental evidence for the underlying idea that a bottle neck in aerobic glycolysis at the level of M2PYK results in an expanded pool of glycolytic intermediates (which are thought to serve as building blocks necessary for proliferation and growth of cancer cells). If a bottle neck at M2PYK exists, then the remarkable increase in lactate production by cancer cells is a paradox, particularly since a high percentage of the carbons of lactate originate from glucose. The finding that pyruvate kinase activity is invariantly increased rather than decreased in cancer undermines the logic of the M2PYK bottle neck, but is consistent with high lactate production. The "inactive" state of M2PYK in cancer is often described as a dimer (with reduced substrate affinity) that has dissociated from an active tetramer of M2PYK. Although M2PYK clearly dissociates easier than other isozymes of pyruvate kinase, it is not clear that dissociation of the tetramer occurs in vivo when ligands are present that promote tetramer formation. Furthermore, it is also not clear whether the dissociated dimer retains any activity at all. A number of non-canonical functions for M2PYK have been proposed, all of which can be challenged by the finding that not all cancer cell types are dependent on M2PYK expression. Additional in-depth studies of the Warburg effect and specifically of the possible regulatory role of M2PYK in the Warburg effect are needed.
Collapse
|
37
|
Petchampai N, Murillo-Solano C, Isoe J, Pizarro JC, Scaraffia PY. Distinctive regulatory properties of pyruvate kinase 1 from Aedes aegypti mosquitoes. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2019; 104:82-90. [PMID: 30578824 PMCID: PMC6814295 DOI: 10.1016/j.ibmb.2018.12.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 12/17/2018] [Accepted: 12/17/2018] [Indexed: 05/17/2023]
Abstract
Female Aedes aegypti mosquitoes are vectors of arboviruses that cause diseases of public health significance. The discovery of new metabolic targets is crucial for improving mosquito control strategies. We recently demonstrated that glucose oxidation supports ammonia detoxification in A. aegypti. Pyruvate kinase (PK, EC 2.7.1.40) catalyzes the last step of the glycolytic pathway. In most organisms, one or more allosteric effectors control PK activity. However, the kinetic properties and structure of PK in mosquitoes have not been previously reported. In this study, two alternatively spliced mRNA variants (AaPK1 and AaPK2) that code for PKs were identified in the A. aegypti genome. The AaPK1 mRNA variant, which encodes a 529 amino acid protein with an estimated molecular weight of ∼57 kDa, was cloned. The protein was expressed in Escherichia coli and purified. The AaPK1 kinetic properties were identified. The recombinant protein was also crystallized and its 3D structure determined. We found that alanine, glutamine, proline, serine and fructose-1-phosphate displayed a classic allosteric activation on AaPK1. Ribulose-5-phosphate acted as an allosteric inhibitor of AaPK1 but its inhibitory effect was reversed by alanine, glutamine, proline and serine. Additionally, the allosteric activation of AaPK1 by amino acids was weakened by fructose-1,6-bisphosphate, whereas the allosteric activation of AaPK1 by alanine and serine was diminished by glucose-6-phosphate. The AaPK1 structure shows the presence of fructose-1,6-bisphosphate in the allosteric site. Together, our results reveal that specific amino acids and phosphorylated sugars tightly regulate conformational dynamics and catalytic changes of AaPK1. The distinctive AaPK1 allosteric properties support a complex role for this enzyme within mosquito metabolism.
Collapse
Affiliation(s)
- Natthida Petchampai
- Department of Tropical Medicine, Vector-Borne Infectious Disease Research Center, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Claribel Murillo-Solano
- Department of Tropical Medicine, Vector-Borne Infectious Disease Research Center, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Jun Isoe
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, AZ, 85721, USA
| | - Juan C Pizarro
- Department of Tropical Medicine, Vector-Borne Infectious Disease Research Center, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, 70112, USA.
| | - Patricia Y Scaraffia
- Department of Tropical Medicine, Vector-Borne Infectious Disease Research Center, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, 70112, USA.
| |
Collapse
|
38
|
Liu Y, Zhang Z, Wang J, Chen C, Tang X, Zhu J, Liu J. Metabolic reprogramming results in abnormal glycolysis in gastric cancer: a review. Onco Targets Ther 2019; 12:1195-1204. [PMID: 30863087 PMCID: PMC6389007 DOI: 10.2147/ott.s189687] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The Warburg effect in tumor cells involves the uptake of high levels of glucose, enhanced glycolysis, and the metabolism of pyruvate to lactic acid rather than oxidative phos-phorylation to generate energy under aerobic conditions. This effect is closely related to the occurrence, invasion, metastasis, drug resistance, and poor prognosis of gastric cancer (GC). Current research has further demonstrated that the Warburg effect in GC cells is not only mediated by the glycolysis pathway, but also includes roles for mitochondria, noncoding RNAs, and other proteins that do not directly regulate metabolism. As a result, changes in the glycolysis pathway not only lead to abnormal glucose metabolism, but they also affect mitochondrial functions, cellular processes such as apoptosis and cell cycle regulation, and the metabolism of lipids and amino acids. In this review, we discuss metabolic reprogramming in GC based on glycolysis, a possible link between glucose metabolism, lipid metabolism, and amino acid metabolism, and we clarify the role of mitochondria. We also examine recent studies of metabolic inhibitors in GC.
Collapse
Affiliation(s)
- Yuanda Liu
- Department of Gastrointestinal Surgery, The Second Hospital of Jilin University, Changchun 130041, China, ;
| | - Ze Zhang
- Department of General Surgery, The First Hospital of Jilin University, Changchun 130021, China
| | - Junyang Wang
- Department of Gastrointestinal Surgery, The Second Hospital of Jilin University, Changchun 130041, China, ;
| | - Chao Chen
- Department of Gastrointestinal Surgery, The Second Hospital of Jilin University, Changchun 130041, China, ;
| | - Xiaohuan Tang
- Department of Gastrointestinal Surgery, The Second Hospital of Jilin University, Changchun 130041, China, ;
| | - Jiaming Zhu
- Department of Gastrointestinal Surgery, The Second Hospital of Jilin University, Changchun 130041, China, ;
| | - Jingjing Liu
- Department of Gastrointestinal Surgery, The Second Hospital of Jilin University, Changchun 130041, China, ;
| |
Collapse
|
39
|
Redox regulation of pyruvate kinase M2 by cysteine oxidation and S-nitrosation. Biochem J 2018; 475:3275-3291. [PMID: 30254098 PMCID: PMC6208296 DOI: 10.1042/bcj20180556] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 09/21/2018] [Accepted: 09/24/2018] [Indexed: 01/09/2023]
Abstract
We show here that the M2 isoform of human pyruvate kinase (M2PYK) is susceptible to nitrosation and oxidation, and that these modifications regulate enzyme activity by preventing the formation of the active tetrameric form. The biotin-switch assay carried out on M1 and M2 isoforms showed that M2PYK is sensitive to nitrosation and that Cys326 is highly susceptible to redox modification. Structural and enzymatic studies have been carried out on point mutants for three cysteine residues (Cys424, Cys358, and Cys326) to characterise their potential roles in redox regulation. Nine cysteines are conserved between M2PYK and M1PYK. Cys424 is the only cysteine unique to M2PYK. C424S, C424A, and C424L showed a moderate effect on enzyme activity with 80, 100, and 140% activity, respectively, compared with M2PYK. C358 had been previously identified from in vivo studies to be the favoured target for oxidation. Our characterised mutant showed that this mutation stabilises tetrameric M2PYK, suggesting that the in vivo resistance to oxidation for the Cys358Ser mutation is due to stabilisation of the tetrameric form of the enzyme. In contrast, the Cys326Ser mutant exists predominantly in monomeric form. A biotin-switch assay using this mutant also showed a significant reduction in biotinylation of M2PYK, confirming that this is a major target for nitrosation and probably oxidation. Our results show that the sensitivity of M2PYK to oxidation and nitrosation is regulated by its monomer–tetramer equilibrium. In the monomer state, residues (in particular C326) are exposed to oxidative modifications that prevent reformation of the active tetrameric form.
Collapse
|