1
|
Cereghetti G, Kissling VM, Koch LM, Arm A, Schmidt CC, Thüringer Y, Zamboni N, Afanasyev P, Linsenmeier M, Eichmann C, Kroschwald S, Zhou J, Cao Y, Pfizenmaier DM, Wiegand T, Cadalbert R, Gupta G, Boehringer D, Knowles TPJ, Mezzenga R, Arosio P, Riek R, Peter M. An evolutionarily conserved mechanism controls reversible amyloids of pyruvate kinase via pH-sensing regions. Dev Cell 2024; 59:1876-1891.e7. [PMID: 38788715 DOI: 10.1016/j.devcel.2024.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 10/15/2023] [Accepted: 04/26/2024] [Indexed: 05/26/2024]
Abstract
Amyloids are known as irreversible aggregates associated with neurodegenerative diseases. However, recent evidence shows that a subset of amyloids can form reversibly and fulfill essential cellular functions. Yet, the molecular mechanisms regulating functional amyloids and distinguishing them from pathological aggregates remain unclear. Here, we investigate the conserved principles of amyloid reversibility by studying the essential metabolic enzyme pyruvate kinase (PK) in yeast and human cells. We demonstrate that yeast PK (Cdc19) and human PK (PKM2) form reversible amyloids through a pH-sensitive amyloid core. Stress-induced cytosolic acidification promotes aggregation via protonation of specific glutamate (yeast) or histidine (human) residues within the amyloid core. Mutations mimicking protonation cause constitutive PK aggregation, while non-protonatable PK mutants remain soluble even upon stress. Physiological PK aggregation is coupled to metabolic rewiring and glycolysis arrest, causing severe growth defects when misregulated. Our work thus identifies an evolutionarily conserved, potentially widespread mechanism regulating functional amyloids during stress.
Collapse
Affiliation(s)
- Gea Cereghetti
- Institute of Biochemistry, Department of Biology, ETH Zürich, 8093 Zürich, Switzerland; Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, CB2 1EW Cambridge, UK.
| | - Vera M Kissling
- Institute of Biochemistry, Department of Biology, ETH Zürich, 8093 Zürich, Switzerland; Particles-Biology Interactions Laboratory, Department of Materials Meet Life, Empa, 9014 St. Gallen, Switzerland
| | - Lisa M Koch
- Institute of Biochemistry, Department of Biology, ETH Zürich, 8093 Zürich, Switzerland
| | - Alexandra Arm
- Institute of Biochemistry, Department of Biology, ETH Zürich, 8093 Zürich, Switzerland
| | - Claudia C Schmidt
- Institute of Biochemistry, Department of Biology, ETH Zürich, 8093 Zürich, Switzerland
| | - Yannik Thüringer
- Institute of Biochemistry, Department of Biology, ETH Zürich, 8093 Zürich, Switzerland
| | - Nicola Zamboni
- Institute of Molecular Systems Biology, Department of Biology, ETH Zürich, 8093 Zürich, Switzerland
| | - Pavel Afanasyev
- Cryo-EM Knowledge Hub (CEMK), ETH Zurich, 8093 Zürich, Switzerland
| | - Miriam Linsenmeier
- Institute for Chemical and Bioengineering, Department of Chemistry and Applied Biosciences, ETH Zürich, 8093 Zürich, Switzerland
| | - Cédric Eichmann
- Laboratory of Physical Chemistry, Department of Chemistry and Applied Biosciences, ETH Zürich, 8093 Zürich, Switzerland
| | - Sonja Kroschwald
- Institute of Biochemistry, Department of Biology, ETH Zürich, 8093 Zürich, Switzerland
| | - Jiangtao Zhou
- Department of Health Sciences & Technology, ETH Zürich, 8092 Zürich, Switzerland
| | - Yiping Cao
- Department of Health Sciences & Technology, ETH Zürich, 8092 Zürich, Switzerland
| | - Dorota M Pfizenmaier
- Institute of Biochemistry, Department of Biology, ETH Zürich, 8093 Zürich, Switzerland
| | - Thomas Wiegand
- Laboratory of Physical Chemistry, Department of Chemistry and Applied Biosciences, ETH Zürich, 8093 Zürich, Switzerland; Max Planck Institute for Chemical Energy Conversion, 45470 Mülheim an der Ruhr, Germany; Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, 52074 Aachen, Germany
| | - Riccardo Cadalbert
- Laboratory of Physical Chemistry, Department of Chemistry and Applied Biosciences, ETH Zürich, 8093 Zürich, Switzerland
| | - Govind Gupta
- Particles-Biology Interactions Laboratory, Department of Materials Meet Life, Empa, 9014 St. Gallen, Switzerland
| | | | - Tuomas P J Knowles
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, CB2 1EW Cambridge, UK
| | - Raffaele Mezzenga
- Department of Health Sciences & Technology, ETH Zürich, 8092 Zürich, Switzerland
| | - Paolo Arosio
- Institute for Chemical and Bioengineering, Department of Chemistry and Applied Biosciences, ETH Zürich, 8093 Zürich, Switzerland
| | - Roland Riek
- Laboratory of Physical Chemistry, Department of Chemistry and Applied Biosciences, ETH Zürich, 8093 Zürich, Switzerland
| | - Matthias Peter
- Institute of Biochemistry, Department of Biology, ETH Zürich, 8093 Zürich, Switzerland.
| |
Collapse
|
2
|
Chen S, Zou Y, Song C, Cao K, Cai K, Wu Y, Zhang Z, Geng D, Sun W, Ouyang N, Zhang N, Li Z, Sun G, Zhang Y, Sun Y, Zhang Y. The role of glycolytic metabolic pathways in cardiovascular disease and potential therapeutic approaches. Basic Res Cardiol 2023; 118:48. [PMID: 37938421 PMCID: PMC10632287 DOI: 10.1007/s00395-023-01018-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/09/2023]
Abstract
Cardiovascular disease (CVD) is a major threat to human health, accounting for 46% of non-communicable disease deaths. Glycolysis is a conserved and rigorous biological process that breaks down glucose into pyruvate, and its primary function is to provide the body with the energy and intermediate products needed for life activities. The non-glycolytic actions of enzymes associated with the glycolytic pathway have long been found to be associated with the development of CVD, typically exemplified by metabolic remodeling in heart failure, which is a condition in which the heart exhibits a rapid adaptive response to hypoxic and hypoxic conditions, occurring early in the course of heart failure. It is mainly characterized by a decrease in oxidative phosphorylation and a rise in the glycolytic pathway, and the rise in glycolysis is considered a hallmark of metabolic remodeling. In addition to this, the glycolytic metabolic pathway is the main source of energy for cardiomyocytes during ischemia-reperfusion. Not only that, the auxiliary pathways of glycolysis, such as the polyol pathway, hexosamine pathway, and pentose phosphate pathway, are also closely related to CVD. Therefore, targeting glycolysis is very attractive for therapeutic intervention in CVD. However, the relationship between glycolytic pathway and CVD is very complex, and some preclinical studies have confirmed that targeting glycolysis does have a certain degree of efficacy, but its specific role in the development of CVD has yet to be explored. This article aims to summarize the current knowledge regarding the glycolytic pathway and its key enzymes (including hexokinase (HK), phosphoglucose isomerase (PGI), phosphofructokinase-1 (PFK1), aldolase (Aldolase), phosphoglycerate metatase (PGAM), enolase (ENO) pyruvate kinase (PKM) lactate dehydrogenase (LDH)) for their role in cardiovascular diseases (e.g., heart failure, myocardial infarction, atherosclerosis) and possible emerging therapeutic targets.
Collapse
Affiliation(s)
- Shuxian Chen
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Yuanming Zou
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Chunyu Song
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Kexin Cao
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Kexin Cai
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Yanjiao Wu
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Zhaobo Zhang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Danxi Geng
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Wei Sun
- Department of Thyroid Surgery, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
| | - Nanxiang Ouyang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
| | - Naijin Zhang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
- Institute of Health Sciences, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning Province, People's Republic of China.
- Key Laboratory of Reproductive and Genetic Medicine, China Medical University, National Health Commission, 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning Province, People's Republic of China.
| | - Zhao Li
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
| | - Guozhe Sun
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
| | - Yixiao Zhang
- Department of Urology Surgery, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning Province, People's Republic of China.
| | - Yingxian Sun
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
- Institute of Health Sciences, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning Province, People's Republic of China.
| | - Ying Zhang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
- Institute of Health Sciences, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning Province, People's Republic of China.
| |
Collapse
|
3
|
IgSF11-mediated phosphorylation of pyruvate kinase M2 regulates osteoclast differentiation and prevents pathological bone loss. Bone Res 2023; 11:17. [PMID: 36928396 PMCID: PMC10020456 DOI: 10.1038/s41413-023-00251-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 01/04/2023] [Accepted: 02/12/2023] [Indexed: 03/18/2023] Open
Abstract
Osteoclasts are primary bone-resorbing cells, and receptor-activated NF-kB ligand (RANKL) stimulation is the key driver of osteoclast differentiation. During late-stage differentiation, osteoclasts become multinucleated and enlarged (so-called "maturation"), suggesting their need to adapt to changing metabolic demands and a substantial increase in size. Here, we demonstrate that immunoglobulin superfamily 11 (IgSF11), which is required for osteoclast differentiation through an association with the postsynaptic scaffolding protein PSD-95, regulates osteoclast differentiation by controlling the activity of pyruvate kinase M isoform 2 (PKM2). By using a system that directly induces the activation of IgSF11 in a controlled manner, we identified PKM2 as a major IgSF11-induced tyrosine-phosphorylated protein. IgSF11 activates multiple Src family tyrosine kinases (SFKs), including c-Src, Fyn, and HcK, which phosphorylate PKM2 and thereby inhibit PKM2 activity. Consistently, IgSF11-deficient cells show higher PKM2 activity and defective osteoclast differentiation. Furthermore, inhibiting PKM2 activities with the specific inhibitor Shikonin rescues the impaired osteoclast differentiation in IgSF11-deficient cells, and activating PKM2 with the specific activator TEPP46 suppresses osteoclast differentiation in wild-type cells. Moreover, PKM2 activation further suppresses osteoclastic bone loss without affecting bone formation in vivo. Taken together, these results show that IgSF11 controls osteoclast differentiation through PKM2 activity, which is a metabolic switch necessary for optimal osteoclast maturation.
Collapse
|
4
|
Sych K, Nold SP, Pfeilschifter J, Vutukuri R, Meisterknecht J, Wittig I, Frank S, Goren I. Expression of PKM2 in wound keratinocytes is coupled to angiogenesis during skin repair in vivo and in HaCaT keratinocytes in vitro. J Mol Med (Berl) 2023; 101:151-169. [PMID: 36633604 PMCID: PMC9977898 DOI: 10.1007/s00109-022-02280-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 01/13/2023]
Abstract
An injured skin is rapidly restored in a manner of wound healing. We have previously shown that intact insulin signaling and glucose uptake are fundamental to proper wound closure. Consequently, under exacerbated inflammation, compromised insulin action and glucose uptake lead to impaired healing. However, in spite of the increased attention to cell metabolism during tissue regeneration, metabolic mediators that govern cellular and physiological processes throughout skin repair remained largely elusive. Through assessment of mRNA using real-time PCR and protein blot analysis, we report that healing of cutaneous wounds comprise a boosted expression of genes involved in glycolysis, oxidative phosphorylation, pentose phosphate shunt, and glutamine anaplerosis. We further focused on the functional role of pyruvate kinase M (PKM) isoenzymes that catalyze the final and rate-limiting step of glycolysis. Whereas the expression of the metabolic constitutively active Pkm1 isozyme remained almost unchanged, Pkm2 is augmented during the inflammatory phase of healing. The immunohistochemistry and RNA in situ hybridization analysis showed a confined Pkm2 expression to keratinocytes of the hyperproliferative epithelium and, to a lesser extent, infiltrating neutrophils and monocytes as well as later on in macrophages. Notably, the expression of Pkm2 in keratinocytes facing the wound bed side colocalized with VEGF expression. The in vitro knockdown of PKM2 in HaCaT keratinocytes using small interfering (si) RNA confirmed an acute role for PKM2 in facilitating the complete induction of VEGF mRNA and protein expression in keratinocytes; this function is mainly HIF-1α independent. KEY MESSAGES: • Wound healing involves activation of glycolysis, oxidative phosphorylation, pentos-phosphate shunt, and replenishment of tri-carboxylic acid (TCA) cycle through glutamine anaplerosis. • The pyruvate kinase M2 (PKM2) isoform is upregulated during the inflammatory phase of cutaneous healing, mainly in keratinocytes of hyperproliferative epithelia. • In vivo, the expression of VEGF in wound keratinocytes is colocalized with PKM2. • PKM2 is required for full induction of VEGF in HaCaT keratinocytes in vitro.
Collapse
Affiliation(s)
- Khrystyna Sych
- Pharmazentrum Frankfurt/ZAFES, General Pharmacology and Toxicology, Faculty of Medicine, Goethe-University, Frankfurt, Frankfurt am Main, Germany
| | - Simon P Nold
- Pharmazentrum Frankfurt/ZAFES, General Pharmacology and Toxicology, Faculty of Medicine, Goethe-University, Frankfurt, Frankfurt am Main, Germany
| | - Josef Pfeilschifter
- Pharmazentrum Frankfurt/ZAFES, General Pharmacology and Toxicology, Faculty of Medicine, Goethe-University, Frankfurt, Frankfurt am Main, Germany
| | - Rajkumar Vutukuri
- Pharmazentrum Frankfurt/ZAFES, General Pharmacology and Toxicology, Faculty of Medicine, Goethe-University, Frankfurt, Frankfurt am Main, Germany
| | - Jana Meisterknecht
- Functional Proteomics, Institute for Cardiovascular Physiology, Goethe-University, Frankfurt, Theodor-Stern-Kai 7, D-60590, Frankfurt, Frankfurt am Main, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhein-Main, Frankfurt am Main, Germany
| | - Ilka Wittig
- Functional Proteomics, Institute for Cardiovascular Physiology, Goethe-University, Frankfurt, Theodor-Stern-Kai 7, D-60590, Frankfurt, Frankfurt am Main, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhein-Main, Frankfurt am Main, Germany
| | - Stefan Frank
- Pharmazentrum Frankfurt/ZAFES, General Pharmacology and Toxicology, Faculty of Medicine, Goethe-University, Frankfurt, Frankfurt am Main, Germany
| | - Itamar Goren
- Pharmazentrum Frankfurt/ZAFES, General Pharmacology and Toxicology, Faculty of Medicine, Goethe-University, Frankfurt, Frankfurt am Main, Germany.
| |
Collapse
|
5
|
Esen I, Jiemy WF, van Sleen Y, Bijzet J, de Jong DM, Nienhuis PH, Slart RHJA, Heeringa P, Boots AMH, Brouwer E. Plasma Pyruvate Kinase M2 as a marker of vascular inflammation in giant cell arteritis. Rheumatology (Oxford) 2022; 61:3060-3070. [PMID: 34730794 PMCID: PMC9258600 DOI: 10.1093/rheumatology/keab814] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 10/29/2021] [Indexed: 11/28/2022] Open
Abstract
OBJECTIVES GCA is a large vessel vasculitis in which metabolically active immune cells play an important role. GCA diagnosis is based on CRP/ESR and temporal artery biopsies (TABs), in combination with 18F-fluorodeoxyglucose ([18F]FDG)-PET/CT relying on enhanced glucose uptake by glycolytic macrophages. Here, we studied circulating Pyruvate Kinase M2 (PKM2), a glycolytic enzyme, as a possible systemic marker of vessel wall inflammation in GCA. METHODS Immunohistochemical detection of PKM2 was performed on inflamed (n = 12) and non-inflamed (n = 4) TABs from GCA patients and non-GCA (n = 9) patients. Dimeric PKM2 levels were assessed in plasma of GCA patients (n = 44), age-matched healthy controls (n = 41), metastatic melanoma patients (n = 7) and infection controls (n = 11). CRP, ESR and macrophage markers calprotectin and YKL-40 were correlated with plasma PKM2 levels. To detect the cellular source of plasma PKM2 in tissue, double IF staining was performed on inflamed GCA TABs. [18F]FDG-PET scans of 23 GCA patients were analysed and maximum standard uptake values and target to background ratios were calculated. RESULTS PKM2 is abundantly expressed in TABs of GCA patients. Dimeric PKM2 plasma levels were elevated in GCA and correlated with CRP, ESR, calprotectin and YKL-40 levels. Elevated plasma PKM2 levels were downmodulated by glucocorticoid treatment. PKM2 was detected in both macrophages and T cells at the site of vascular inflammation. Circulating PKM2 levels correlated with average target to background ratios PET scores. CONCLUSION Elevated plasma PKM2 levels reflect active vessel inflammation in GCA and may assist in disease diagnosis and in disease monitoring.
Collapse
Affiliation(s)
- Idil Esen
- Department of Rheumatology and Clinical Immunology
| | | | | | - Johan Bijzet
- Department of Rheumatology and Clinical Immunology
| | | | - Pieter H Nienhuis
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen
| | - Riemer H J A Slart
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen
- Department of Biomedical Photonic Imaging, Faculty of Science and Technology, University of Twente, Enschede
| | - Peter Heeringa
- Department of Pathology and Medical Biology, University of Groningen, Groningen, The Netherlands
| | | | | |
Collapse
|
6
|
Davidson SM, Schmidt DR, Heyman JE, O'Brien JP, Liu AC, Israelsen WJ, Dayton TL, Sehgal R, Bronson RT, Freinkman E, Mak HH, Fanelli GN, Malstrom S, Bellinger G, Carracedo A, Pandolfi PP, Courtney KD, Jha A, DePinho RA, Horner JW, Thomas CJ, Cantley LC, Loda M, Vander Heiden MG. Pyruvate Kinase M1 Suppresses Development and Progression of Prostate Adenocarcinoma. Cancer Res 2022; 82:2403-2416. [PMID: 35584006 PMCID: PMC9256808 DOI: 10.1158/0008-5472.can-21-2352] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 04/19/2022] [Accepted: 05/11/2022] [Indexed: 01/07/2023]
Abstract
SIGNIFICANCE Differential expression of PKM1 and PKM2 impacts prostate tumorigenesis and suggests a potential therapeutic vulnerability in prostate cancer.
Collapse
Affiliation(s)
- Shawn M. Davidson
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Broad Institute of MIT and Harvard University, Cambridge, Massachusetts.,Corresponding Authors: Matthew G. Vander Heiden, Koch Institute/Biology, Massachusetts Institute of Technology, Cambridge, MA 02139. E-mail: ; and Shawn M. Davidson,
| | - Daniel R. Schmidt
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Broad Institute of MIT and Harvard University, Cambridge, Massachusetts.,Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Julia E. Heyman
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - James P. O'Brien
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Amy C. Liu
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - William J. Israelsen
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Talya L. Dayton
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | | | - Roderick T. Bronson
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | | | - Howard H. Mak
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Giuseppe Nicolò Fanelli
- Weill Cornell Medical College, New York, New York.,Division of Pathology, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Scott Malstrom
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Gary Bellinger
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | | | | | | | | | | | | | - Craig J. Thomas
- National Center for Advancing Translational Sciences, NIH, Bethesda, Maryland
| | - Lewis C. Cantley
- Beth Israel Deaconess Medical Center, Boston, Massachusetts.,Weill Cornell Medical College, New York, New York
| | - Massimo Loda
- Weill Cornell Medical College, New York, New York.,Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Matthew G. Vander Heiden
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Broad Institute of MIT and Harvard University, Cambridge, Massachusetts.,Dana-Farber Cancer Institute, Boston, Massachusetts.,Corresponding Authors: Matthew G. Vander Heiden, Koch Institute/Biology, Massachusetts Institute of Technology, Cambridge, MA 02139. E-mail: ; and Shawn M. Davidson,
| |
Collapse
|
7
|
Xia Y, Wang S, Song C, Luo R. Spatiotemporal feedforward between PKM2 tetramers and mTORC1 prompts mTORC1 activation. Phys Biol 2022; 19. [PMID: 35613602 DOI: 10.1088/1478-3975/ac7372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/25/2022] [Indexed: 11/11/2022]
Abstract
Most mammalian cells couple glucose availability to anabolic processes via the mTORC1 pathway. However, the mechanism by which fluctuations in glucose availability are rapidly translated into mTORC1 signals remains elusive. Here, we show that cells rapidly respond to changes in glucose availability through the spatial coupling of mTORC1 and tetramers of the key glycolytic enzyme pyruvate kinase M2 (PKM2) on lysosomal surfaces in the late G1/S phases. The lysosomal localization of PKM2 tetramers enables rapid increases in local ATP concentrations around lysosomes to activate mTORC1, while bypassing the need to elevate global ATP levels in the entire cell. In essence, this spatial coupling establishes a feedforward loop to enable mTORC1 to rapidly sense and respond to changes in glucose availability. We further demonstrate that this mechanism ensures robust cell proliferation upon fluctuating glucose availability. Thus, we present mechanistic insights into the rapid response of the mTORC1 pathway to changes in glucose availability. The underlying mechanism may be applicable to the control of other cellular processes.
Collapse
Affiliation(s)
- Yu Xia
- Fudan University, Rm A601# Life Science Building Fudan University, Yangpu, Shanghai, , Shanghai, 200433, CHINA
| | - ShuMing Wang
- Fudan University, Rm A608# Life Science Building, Fudan University, Yangpu, Shanghai, Shanghai, Shanghai, 200433, CHINA
| | - Chunbo Song
- Fudan University, #Rm 519# Life Science Building, Fudan University, Shanghai, Shanghai, 200433, CHINA
| | - Ruoyu Luo
- School of Life Science, Fudan University, 601# Rm, Building of School of Life Science, 2005#,Songhu Rd, Shanghai, Shanghai, 200433, CHINA
| |
Collapse
|
8
|
The Role of PKM2 in the Regulation of Mitochondrial Function: Focus on Mitochondrial Metabolism, Oxidative Stress, Dynamic, and Apoptosis. PKM2 in Mitochondrial Function. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7702681. [PMID: 35571239 PMCID: PMC9106463 DOI: 10.1155/2022/7702681] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 03/16/2022] [Indexed: 02/05/2023]
Abstract
The M2 isoform of pyruvate kinase (PKM2) is one isoform of pyruvate kinase (PK). PKM2 is expressed at high levels during embryonic development and tumor progression and is subject to complex allosteric regulation. PKM2 is a special glycolytic enzyme that regulates the final step of glycolysis; the role of PKM2 in the metabolism, survival, and apoptosis of cancer cells has received increasing attention. Mitochondria are directly or indirectly involved in the regulation of energy metabolism, susceptibility to oxidative stress, and cell death; however, the role of PKM2 in mitochondrial functions remains unclear. Herein, we review the related mechanisms of the role of PKM2 in the regulation of mitochondrial functions from the aspects of metabolism, reactive oxygen species (ROS), dynamic, and apoptosis, which can be highlighted as a target for the clinical management of cardiovascular and metabolic diseases.
Collapse
|
9
|
Xia Y, Wang X, Liu Y, Shapiro E, Lepor H, Tang MS, Sun TT, Wu XR. PKM2 Is Essential for Bladder Cancer Growth and Maintenance. Cancer Res 2022; 82:571-585. [PMID: 34903602 PMCID: PMC8857058 DOI: 10.1158/0008-5472.can-21-0403] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 08/30/2021] [Accepted: 12/02/2021] [Indexed: 11/16/2022]
Abstract
Pyruvate kinase M2 (PKM2) has been shown to promote tumorigenesis by facilitating the Warburg effect and enhancing the activities of oncoproteins. However, this paradigm has recently been challenged by studies in which the absence of PKM2 failed to inhibit and instead accelerated tumorigenesis in mouse models. These results seem inconsistent with the fact that most human tumors overexpress PKM2. To further elucidate the role of PKM2 in tumorigenesis, we investigated the effect of PKM2 knockout in oncogenic HRAS-driven urothelial carcinoma. While PKM2 ablation in mouse urothelial cells did not affect tumor initiation, it impaired the growth and maintenance of HRAS-driven tumors. Chemical inhibition of PKM2 recapitulated these effects. Both conditions substantially reduced complex formation of PKM2 with STAT3, their nuclear translocation, and HIF1α- and VEGF-related angiogenesis. The reduction in nuclear STAT3 in the absence of PKM2 also correlated with decreased autophagy and increased apoptosis. Time-controlled, inducible PKM2 overexpression in simple urothelial hyperplasia did not trigger tumorigenesis, while overexpression of PKM2, but not PKM1, in nodular urothelial hyperplasia with angiogenesis strongly accelerated tumorigenesis. Finally, in human patients, PKM2 was overexpressed in low-grade nonmuscle-invasive and high-grade muscle-invasive bladder cancer. Based on these data, PKM2 is not required for tumor initiation but is essential for tumor growth and maintenance by enhancing angiogenesis and metabolic addiction. The PKM2-STAT3-HIF1α/VEGF signaling axis may play a critical role in bladder cancer and may serve as an actionable therapeutic target. SIGNIFICANCE Genetic manipulation and pharmacologic inhibition of PKM2 in mouse urothelial lesions highlight its essential role in promoting angiogenesis and metabolic addiction, events indispensable for tumor growth and maintenance.
Collapse
MESH Headings
- Active Transport, Cell Nucleus/genetics
- Animals
- Apoptosis/genetics
- Autophagy/genetics
- Carcinogenesis/genetics
- Carcinoma, Transitional Cell/blood supply
- Carcinoma, Transitional Cell/genetics
- Carcinoma, Transitional Cell/metabolism
- Cell Line, Tumor
- Cell Proliferation/genetics
- Gene Expression Regulation, Neoplastic
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Male
- Mice, Knockout
- Mice, Transgenic
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Proto-Oncogene Proteins p21(ras)/genetics
- Proto-Oncogene Proteins p21(ras)/metabolism
- Pyruvate Kinase/genetics
- Pyruvate Kinase/metabolism
- STAT3 Transcription Factor/genetics
- STAT3 Transcription Factor/metabolism
- Urinary Bladder Neoplasms/genetics
- Urinary Bladder Neoplasms/metabolism
- Urinary Bladder Neoplasms/pathology
- Vascular Endothelial Growth Factor A/genetics
- Vascular Endothelial Growth Factor A/metabolism
- Mice
Collapse
Affiliation(s)
- Yong Xia
- Department of Urology, New York University School of Medicine, New York, NY 10016
| | - Xing Wang
- Department of Urology, New York University School of Medicine, New York, NY 10016
- Veterans Affairs New York Harbor Healthcare System, Manhattan Campus, New York, NY 10010
| | - Yan Liu
- Department of Urology, New York University School of Medicine, New York, NY 10016
- Veterans Affairs New York Harbor Healthcare System, Manhattan Campus, New York, NY 10010
| | - Ellen Shapiro
- Department of Urology, New York University School of Medicine, New York, NY 10016
| | - Herbert Lepor
- Department of Urology, New York University School of Medicine, New York, NY 10016
| | - Moon-shong Tang
- Department of Environmental Medicine, New York University School of Medicine, New York, NY 10016
| | - Tung-Tien Sun
- Department of Urology, New York University School of Medicine, New York, NY 10016
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016
| | - Xue-Ru Wu
- Department of Urology, New York University School of Medicine, New York, NY 10016
- Department of Pathology, New York University School of Medicine, New York, NY 10016
- Veterans Affairs New York Harbor Healthcare System, Manhattan Campus, New York, NY 10010
| |
Collapse
|
10
|
Understanding the Role of Metalloproteinases and Their Inhibitors in Periodontology. Clin Rev Bone Miner Metab 2022. [DOI: 10.1007/s12018-021-09281-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
11
|
Tang D, Sandoval W, Liu P, Lam C, Snedecor B, Misaghi S. Preventing pyruvate kinase muscle expression in Chinese hamster ovary cells curbs lactogenic behavior by altering glycolysis, gating pyruvate generation, and increasing pyruvate flux into the TCA cycle. Biotechnol Prog 2021; 37:e3193. [PMID: 34288605 DOI: 10.1002/btpr.3193] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/09/2021] [Accepted: 07/15/2021] [Indexed: 11/07/2022]
Abstract
Deletion of the pyruvate kinase muscle (PKM) gene, which is involved in conversion of phosphoenolpyruvate to pyruvate, has been shown to curb lactogenic behavior in Chinese hamster ovary (CHO) cells. This study describes the generation of pyruvate kinase muscle isoforms 1 and 2 knockout (PKM-KO) and pyruvate kinase muscle isoform-1 knockout (PKM1-KO) CHO host cells to understand metabolic shifts that reduce lactate secretion in these cells. Glucose and amino acids uptake levels in wild-type (WT), PKM-KO, and PKM1-KO stable cell lines, expressing two different antibodies, were analyzed in 14-day fed-batch production assays using different vessels. PKM-KO and PKM1-KO cells consumed more glucose per cell, altered amino acids metabolism, had higher flux of pyruvate into the tricarboxylic acid (TCA) cycle, and as previously shown reduced lactate secretion levels compared with the WT cells. Additionally, both PKM-KO and PKM1-KO cells had higher specific productivity and lower cell growth rates compared with the WT cells. Our findings suggest that rewiring the flux of pyruvate to the TCA cycle by deletion of PKM or PKM1 reduced cell growth and increased specific productivity in CHO cells. Overall, PKM1-KO cells had similar product quality and comparable or better titers relative to the WT cells, hence, targeted deletion of this isoform for curbing lactogenic behavior in CHO cells is suggested.
Collapse
Affiliation(s)
- Danming Tang
- Cell Culture and Bioprocess Operations Department, Genentech Inc., South San Francisco, California, USA
| | - Wendy Sandoval
- Department of Microchemistry, Proteomics and Lipidomics, Genentech Inc., South San Francisco, California, USA
| | - Peter Liu
- Department of Microchemistry, Proteomics and Lipidomics, Genentech Inc., South San Francisco, California, USA
| | - Cynthia Lam
- Cell Culture and Bioprocess Operations Department, Genentech Inc., South San Francisco, California, USA
| | - Brad Snedecor
- Cell Culture and Bioprocess Operations Department, Genentech Inc., South San Francisco, California, USA
| | - Shahram Misaghi
- Cell Culture and Bioprocess Operations Department, Genentech Inc., South San Francisco, California, USA
| |
Collapse
|
12
|
Wiese EK, Hitosugi S, Loa ST, Sreedhar A, Andres-Beck LG, Kurmi K, Pang YP, Karnitz LM, Gonsalves WI, Hitosugi T. Enzymatic activation of pyruvate kinase increases cytosolic oxaloacetate to inhibit the Warburg effect. Nat Metab 2021; 3:954-968. [PMID: 34226744 PMCID: PMC8316326 DOI: 10.1038/s42255-021-00424-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 06/04/2021] [Indexed: 11/30/2022]
Abstract
Pharmacological activation of the glycolytic enzyme PKM2 or expression of the constitutively active PKM1 isoform in cancer cells results in decreased lactate production, a phenomenon known as the PKM2 paradox in the Warburg effect. Here we show that oxaloacetate (OAA) is a competitive inhibitor of human lactate dehydrogenase A (LDHA) and that elevated PKM2 activity increases de novo synthesis of OAA through glutaminolysis, thereby inhibiting LDHA in cancer cells. We also show that replacement of human LDHA with rabbit LDHA, which is relatively resistant to OAA inhibition, eliminated the paradoxical correlation between the elevated PKM2 activity and the decreased lactate concentration in cancer cells treated with a PKM2 activator. Furthermore, rabbit LDHA-expressing tumours, compared to human LDHA-expressing tumours in mice, displayed resistance to the PKM2 activator. These findings describe a mechanistic explanation for the PKM2 paradox by showing that OAA accumulates and inhibits LDHA following PKM2 activation.
Collapse
Affiliation(s)
- Elizabeth K Wiese
- Molecular Pharmacology and Experimental Therapeutics Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Sadae Hitosugi
- Division of Oncology Research, Mayo Clinic, Rochester, MN, USA
| | - Sharon T Loa
- Division of Oncology Research, Mayo Clinic, Rochester, MN, USA
| | | | - Lindsey G Andres-Beck
- Molecular Pharmacology and Experimental Therapeutics Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Kiran Kurmi
- Division of Oncology Research, Mayo Clinic, Rochester, MN, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Yuan-Ping Pang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Larry M Karnitz
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
- Division of Oncology Research, Mayo Clinic, Rochester, MN, USA
| | | | - Taro Hitosugi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA.
- Division of Oncology Research, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
13
|
Patel CB, Beinat C, Xie Y, Chang E, Gambhir SS. Tumor treating fields (TTFields) impairs aberrant glycolysis in glioblastoma as evaluated by [ 18F]DASA-23, a non-invasive probe of pyruvate kinase M2 (PKM2) expression. Neoplasia 2021; 23:58-67. [PMID: 33221711 PMCID: PMC7689378 DOI: 10.1016/j.neo.2020.11.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 12/27/2022]
Abstract
Despite the anti-proliferative and survival benefits from tumor treating fields (TTFields) in human glioblastoma (hGBM), little is known about the effects of this form of alternating electric fields therapy on the aberrant glycolysis of hGBM. [18F]FDG is the most common radiotracer in cancer metabolic imaging, but its utility in hGBM is impaired due to high glucose uptake in normal brain tissue. With TTFields, radiochemistry, Western blot, and immunofluorescence microscopy, we identified pyruvate kinase M2 (PKM2) as a biomarker of hGBM response to therapeutic TTFields. We used [18F]DASA-23, a novel radiotracer that measures PKM2 expression and which has been shown to be safe in humans, to detect a shift away from hGBM aberrant glycolysis in response to TTFields. Compared to unexposed hGBM, [18F]DASA-23 uptake was reduced in hGBM exposed to TTFields (53%, P< 0.05) or temozolomide chemotherapy (33%, P > 0.05) for 3 d. A 6-d TTFields exposure resulted in a 31% reduction (P = 0.043) in 60-min uptake of [18F]DASA-23. [18F]DASA-23 was retained after a 10 but not 30-min wash-out period. Compared to [18F]FDG, [18F]DASA-23 demonstrated a 4- to 9-fold greater uptake, implying an improved tumor-to-background ratio. Furthermore, compared to no-TTFields exposure, a 6-d TTFields exposure caused a 35% reduction in [18F]DASA-23 30-min uptake compared to only an 8% reduction in [18F]FDG 30-min uptake. Quantitative Western blot analysis and qualitative immunofluorescence for PKM2 confirmed the TTFields-induced reduction in PKM2 expression. This is the first study to demonstrate that TTFields impairs hGBM aberrant glycolytic metabolism through reduced PKM2 expression, which can be non-invasively detected by the [18F]DASA-23 radiotracer.
Collapse
Affiliation(s)
- Chirag B Patel
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA; Division of Adult Neuro-Oncology, Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.
| | - Corinne Beinat
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yuanyang Xie
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Edwin Chang
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Sanjiv S Gambhir
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA; Departments of Bioengineering and Materials Science & Engineering, Stanford University, Stanford, CA, USA
| |
Collapse
|
14
|
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: 11] [Impact Index Per Article: 2.2] [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
|
15
|
Verbrugge SAJ, Gehlert S, Stadhouders LEM, Jacko D, Aussieker T, M. J. de Wit G, Vogel ISP, Offringa C, Schönfelder M, Jaspers RT, Wackerhage H. PKM2 Determines Myofiber Hypertrophy In Vitro and Increases in Response to Resistance Exercise in Human Skeletal Muscle. Int J Mol Sci 2020; 21:E7062. [PMID: 32992783 PMCID: PMC7583908 DOI: 10.3390/ijms21197062] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 12/22/2022] Open
Abstract
Nearly 100 years ago, Otto Warburg investigated the metabolism of growing tissues and discovered that tumors reprogram their metabolism. It is poorly understood whether and how hypertrophying muscle, another growing tissue, reprograms its metabolism too. Here, we studied pyruvate kinase muscle (PKM), which can be spliced into two isoforms (PKM1, PKM2). This is of interest, because PKM2 redirects glycolytic flux towards biosynthetic pathways, which might contribute to muscle hypertrophy too. We first investigated whether resistance exercise changes PKM isoform expression in growing human skeletal muscle and found that PKM2 abundance increases after six weeks of resistance training, whereas PKM1 decreases. Second, we determined that Pkm2 expression is higher in fast compared to slow fiber types in rat skeletal muscle. Third, by inducing hypertrophy in differentiated C2C12 cells and by selectively silencing Pkm1 and/or Pkm2 with siRNA, we found that PKM2 limits myotube growth. We conclude that PKM2 contributes to hypertrophy in C2C12 myotubes and indicates a changed metabolic environment within hypertrophying human skeletal muscle fibers. PKM2 is preferentially expressed in fast muscle fibers and may partly contribute to the increased potential for hypertrophy in fast fibers.
Collapse
Affiliation(s)
- Sander A. J. Verbrugge
- Department for Sport and Health Sciences, Technical University of Munich, Georg-Brauchle-Ring 60/62, 80992 München/Munich, Germany; (S.A.J.V.); (M.S.)
| | - Sebastian Gehlert
- Department for the Biosciences of Sports, Institute of Sports Science, University of Hildesheim, Universitätsplatz 1, 31141 Hildesheim, Germany
- Department for Molecular and Cellular Sports Medicine, German Sport University Cologne, 50933 Cologne, Germany; (D.J.); (T.A.)
| | - Lian E. M. Stadhouders
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands; (L.E.M.S.); (G.M.J.d.W.); (I.S.P.V.); (C.O.)
| | - Daniel Jacko
- Department for Molecular and Cellular Sports Medicine, German Sport University Cologne, 50933 Cologne, Germany; (D.J.); (T.A.)
| | - Thorben Aussieker
- Department for Molecular and Cellular Sports Medicine, German Sport University Cologne, 50933 Cologne, Germany; (D.J.); (T.A.)
| | - Gerard M. J. de Wit
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands; (L.E.M.S.); (G.M.J.d.W.); (I.S.P.V.); (C.O.)
| | - Ilse S. P. Vogel
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands; (L.E.M.S.); (G.M.J.d.W.); (I.S.P.V.); (C.O.)
| | - Carla Offringa
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands; (L.E.M.S.); (G.M.J.d.W.); (I.S.P.V.); (C.O.)
| | - Martin Schönfelder
- Department for Sport and Health Sciences, Technical University of Munich, Georg-Brauchle-Ring 60/62, 80992 München/Munich, Germany; (S.A.J.V.); (M.S.)
| | - Richard T. Jaspers
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands; (L.E.M.S.); (G.M.J.d.W.); (I.S.P.V.); (C.O.)
| | - Henning Wackerhage
- Department for Sport and Health Sciences, Technical University of Munich, Georg-Brauchle-Ring 60/62, 80992 München/Munich, Germany; (S.A.J.V.); (M.S.)
| |
Collapse
|
16
|
Molecular mechanisms of interplay between autophagy and metabolism in cancer. Life Sci 2020; 259:118184. [PMID: 32763290 DOI: 10.1016/j.lfs.2020.118184] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 07/26/2020] [Accepted: 07/28/2020] [Indexed: 12/19/2022]
Abstract
Autophagy is an essential mechanism of cellular degradation, a way to protect the cells under stress conditions, such as deprivation of nutrients, growth factors and cellular damage. However, in normal physiology autophagy plays a significant role in cancer cells. Current research is in progress to understand how autophagy and cancer cells go hand in hand to support cancer cell progression. The important aspect in cancer and autophagy is the interdependence of autophagy in the survival and progression of cancer cells. Autophagy is known to be a major cause of chemotherapeutic resistance in various cancer cell types. Therefore, inhibition of autophagy as an effective therapeutic approach is being actively studied and tested in clinical studies. Multiple metabolic pathways are linked with autophagy that could potentially be a significant target for chemotherapeutic strategy. The comprehension of the interconnection of autophagy with cancer metabolism can pave a novel findings for effective combinatorial therapeutic strategies.
Collapse
|
17
|
Liu VM, Howell AJ, Hosios AM, Li Z, Israelsen WJ, Heiden MGV. Cancer-associated mutations in human pyruvate kinase M2 impair enzyme activity. FEBS Lett 2020; 594:646-664. [PMID: 31642061 PMCID: PMC7042059 DOI: 10.1002/1873-3468.13648] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 09/28/2019] [Accepted: 10/15/2019] [Indexed: 12/18/2022]
Abstract
Mammalian pyruvate kinase catalyzes the final step of glycolysis, and its M2 isoform (PKM2) is widely expressed in proliferative tissues. Mutations in PKM2 are found in some human cancers; however, the effects of these mutations on enzyme activity and regulation are unknown. Here, we characterized five cancer-associated PKM2 mutations, occurring at various locations on the enzyme, with respect to substrate kinetics and activation by the allosteric activator fructose-1,6-bisphosphate (FBP). The mutants exhibit reduced maximal velocity, reduced substrate affinity, and/or altered activation by FBP. The kinetic parameters of five additional PKM2 mutants that have been used to study enzyme function or regulation also demonstrate the deleterious effects of mutations on PKM2 function. Our findings indicate that PKM2 is sensitive to many amino acid changes and support the hypothesis that decreased PKM2 activity is selected for in rapidly proliferating cells.
Collapse
Affiliation(s)
- Vivian M. Liu
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States
- Harvard-MIT Health Sciences and Technology Division, Harvard Medical School, Boston, MA 02115, United States
| | - Andrea J. Howell
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States
| | - Aaron M. Hosios
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States
| | - Zhaoqi Li
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States
| | - William J. Israelsen
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Matthew G. Vander Heiden
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States
- Dana-Farber Cancer Institute, Boston, MA 02115, United States
| |
Collapse
|
18
|
Rihan M, Nalla LV, Dharavath A, Shard A, Kalia K, Khairnar A. Pyruvate Kinase M2: a Metabolic Bug in Re-Wiring the Tumor Microenvironment. CANCER MICROENVIRONMENT : OFFICIAL JOURNAL OF THE INTERNATIONAL CANCER MICROENVIRONMENT SOCIETY 2019; 12:149-167. [PMID: 31183810 PMCID: PMC6937361 DOI: 10.1007/s12307-019-00226-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 05/17/2019] [Indexed: 12/16/2022]
Abstract
Metabolic reprogramming is a newly emerged hallmark of cancer attaining a recent consideration as an essential factor for the progression and endurance of cancer cells. A prime event of this altered metabolism is increased glucose uptake and discharge of lactate into the cells surrounding constructing a favorable tumor niche. Several oncogenic factors help in promoting this consequence including, pyruvate kinase M2 (PKM2) a rate-limiting enzyme of glycolysis in tumor metabolism via exhibiting its low pyruvate kinase activity and nuclear moon-lightening functions to increase the synthesis of lactate and macromolecules for tumor proliferation. Not only its role in cancer cells but also its role in the tumor microenvironment cells has to be understood for developing the small molecules against it which is lacking with the literature till date. Therefore, in this present review, the role of PKM2 with respect to various tumor niche cells will be clarified. Further, it highlights the updated list of therapeutics targeting PKM2 pre-clinically and clinically with their added limitations. This upgraded understanding of PKM2 may provide a pace for the reader in developing chemotherapeutic strategies for better clinical survival with limited resistance.
Collapse
Affiliation(s)
- Mohd Rihan
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Palaj, Gandhinagar, Gujarat, -382355, India
| | - Lakshmi Vineela Nalla
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Palaj, Gandhinagar, Gujarat, -382355, India
| | - Anil Dharavath
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Palaj, Gandhinagar, Gujarat, -382355, India
| | - Amit Shard
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Palaj, Gandhinagar, Gujarat, -382355, India.
| | - Kiran Kalia
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gujarat, India
| | - Amit Khairnar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Palaj, Gandhinagar, Gujarat, -382355, India.
| |
Collapse
|
19
|
Yang H, Zhu R, Zhao X, Liu L, Zhou Z, Zhao L, Liang B, Ma W, Zhao J, Liu J, Huang G. Sirtuin-mediated deacetylation of hnRNP A1 suppresses glycolysis and growth in hepatocellular carcinoma. Oncogene 2019; 38:4915-4931. [PMID: 30858544 DOI: 10.1038/s41388-019-0764-z] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 01/25/2019] [Accepted: 02/16/2019] [Indexed: 01/06/2023]
Abstract
Tumor cells undergo a metabolic shift in order to adapt to the altered microenvironment, although the underlying mechanisms have not been fully explored. HnRNP A1 is involved in the alternative splicing of the pyruvate kinase (PK) mRNA, allowing tumor cells to specifically produce the PKM2 isoform. We found that the acetylation status of hnRNP A1 in hepatocellular carcinoma (HCC) cells was dependent on glucose availability, which affected the PKM2-dependent glycolytic pathway. In the glucose-starved HCC cells, SIRT1 and SIRT6, members of deacetylase sirtuin family, were highly expressed and deacetylated hnRNP A1 after direct binding. We identified four lysine residues in hnRNP A1 that were deacetylated by SIRT1 and SIRT6, resulting in significant inhibition of glycolysis in HCC cells. Deacetylated hnRNP A1 reduced PKM2 and increased PKM1 alternative splicing in HCC cells under normal glucose conditions, thereby reducing the metabolic activity of PK and the non-metabolic PKM2-β-catenin signaling pathway. However, under glucose starvation, the low levels of acetylated hnRNP A1 reduced HCC cell metabolism to adapt to the nutrient deficiency. Taken together, sirtuin-mediated hnRNP A1 deacetylation inhibits HCC cell proliferation and tumorigenesis in a PKM2-dependent manner. These findings point to the metabolic reprogramming induced by hnRNP A1 acetylation in order to adapt to the nutritional status of the tumor microenvironment.
Collapse
Affiliation(s)
- Hao Yang
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
- Department of Nuclear Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Rongxuan Zhu
- Department of Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Xiaoping Zhao
- Department of Nuclear Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Liu Liu
- Department of Nuclear Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Zhaoli Zhou
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Li Zhao
- Department of Nuclear Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Beibei Liang
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Wenjing Ma
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Jian Zhao
- International Joint Cancer Institute, The Second Military Medical University, Shanghai, 200433, China
| | - Jianjun Liu
- Department of Nuclear Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| | - Gang Huang
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China.
- Department of Nuclear Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| |
Collapse
|
20
|
Yang Q, Liu R, Yu Q, Bi Y, Liu G. Metabolic regulation of inflammasomes in inflammation. Immunology 2019; 157:95-109. [PMID: 30851192 DOI: 10.1111/imm.13056] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 02/25/2019] [Accepted: 03/05/2019] [Indexed: 12/21/2022] Open
Abstract
Inflammasome activation and subsequent inflammatory cytokine secretion are essential for innate immune defence against multiple stimuli and are regarded as a link to adaptive immune responses. Dysfunction of inflammasome activation has been discovered at the onset or progression of infectious diseases, autoimmune diseases and cancer, all of which are also associated with metabolic factors. Furthermore, many studies concerning the metabolic regulation of inflammasome activation have emerged in recent years, especially regarding the activity of the NLRP3 inflammasome under metabolic reprogramming. In this review, we discuss the molecular mechanisms of the interactions between metabolic pathways and inflammasome activation, which exerts further important effects on various diseases.
Collapse
Affiliation(s)
- Qiuli Yang
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Ruichen Liu
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Qing Yu
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Yujing Bi
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Guangwei Liu
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, China
| |
Collapse
|
21
|
Fu J, Xiong Z, Huang C, Li J, Yang W, Han Y, Paiboonrungruan C, Major MB, Chen KN, Kang X, Chen X. Hyperactivity of the transcription factor Nrf2 causes metabolic reprogramming in mouse esophagus. J Biol Chem 2019; 294:327-340. [PMID: 30409900 PMCID: PMC6322899 DOI: 10.1074/jbc.ra118.005963] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 11/03/2018] [Indexed: 12/17/2022] Open
Abstract
Mutations in the genes encoding nuclear factor (erythroid-derived 2)-like 2 (NRF2), Kelch-like ECH-associated protein 1 (KEAP1), and cullin 3 (CUL3) are commonly observed in human esophageal squamous cell carcinoma (ESCC) and result in activation of the NRF2 signaling pathway. Moreover, hyperactivity of the transcription factor Nrf2 has been found to cause esophageal hyperproliferation and hyperkeratosis in mice. However, the underlying mechanism is unclear. In this study, we aimed to understand the molecular mechanisms of esophageal hyperproliferation in mice due to hyperactive Nrf2. Esophageal tissues were obtained from genetically modified mice that differed in the status of the Nrf2 gene and genes in the same pathway (Nrf2-/-, Keap1-/-, K5Cre;Pkm2fl/fl;Keap1-/-, and WT) and analyzed for metabolomic profiles, Nrf2 ChIP-seq, and gene expression. We found that hyperactive Nrf2 causes metabolic reprogramming and up-regulation of metabolic genes in the mouse esophagus. One of the glycolysis genes encoding pyruvate kinase M2 (Pkm2) was not only differentially up-regulated, but also glycosylated and oligomerized, resulting in increased ATP biosynthesis. However, constitutive knockout of Pkm2 failed to inhibit this esophageal phenotype in vivo, and this failure may have been due to compensation by Pkm1 up-regulation. Transient inhibition of NRF2 or glycolysis inhibited the growth of human ESCC cells in which NRF2 is hyperactive in vitro In summary, hyperactive Nrf2 causes metabolic reprogramming in the mouse esophagus through its transcriptional regulation of metabolic genes. Blocking glycolysis transiently inhibits cell proliferation and may therefore have therapeutically beneficial effects on NRF2high ESCC in humans.
Collapse
Affiliation(s)
- Junsheng Fu
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, Fujian Province 350002, China; Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina 27707
| | - Zhaohui Xiong
- Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina 27707
| | - Caizhi Huang
- Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina 27707
| | - Jing Li
- Department of Thoracic Surgery, Ningxia Medical University General Hospital, Yinchuan, Ningxia 750004, China
| | - Wenjun Yang
- Key Laboratory of Fertility Preservation and Maintenance (Ministry of Education), Cancer Institute of the General Hospital, Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Yuning Han
- Department of Thoracic Surgery, Ningxia Medical University General Hospital, Yinchuan, Ningxia 750004, China
| | - Chorlada Paiboonrungruan
- Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina 27707
| | - Michael B Major
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Ke-Neng Chen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), The First Department of Thoracic Surgery, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Xiaozheng Kang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), The First Department of Thoracic Surgery, Peking University Cancer Hospital and Institute, Beijing 100142, China.
| | - Xiaoxin Chen
- Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina 27707; Center for Esophageal Disease and Swallowing, Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina, Chapel Hill, North Carolina 27599.
| |
Collapse
|
22
|
Papa S, Choy PM, Bubici C. The ERK and JNK pathways in the regulation of metabolic reprogramming. Oncogene 2018; 38:2223-2240. [PMID: 30487597 PMCID: PMC6398583 DOI: 10.1038/s41388-018-0582-8] [Citation(s) in RCA: 263] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 09/24/2018] [Accepted: 10/23/2018] [Indexed: 12/13/2022]
Abstract
Most tumor cells reprogram their glucose metabolism as a result of mutations in oncogenes and tumor suppressors, leading to the constitutive activation of signaling pathways involved in cell growth. This metabolic reprogramming, known as aerobic glycolysis or the Warburg effect, allows tumor cells to sustain their fast proliferation and evade apoptosis. Interfering with oncogenic signaling pathways that regulate the Warburg effect in cancer cells has therefore become an attractive anticancer strategy. However, evidence for the occurrence of the Warburg effect in physiological processes has also been documented. As such, close consideration of which signaling pathways are beneficial targets and the effect of their inhibition on physiological processes are essential. The MAPK/ERK and MAPK/JNK pathways, crucial for normal cellular responses to extracellular stimuli, have recently emerged as key regulators of the Warburg effect during tumorigenesis and normal cellular functions. In this review, we summarize our current understanding of the roles of the ERK and JNK pathways in controlling the Warburg effect in cancer and discuss their implication in controlling this metabolic reprogramming in physiological processes and opportunities for targeting their downstream effectors for therapeutic purposes.
Collapse
Affiliation(s)
- Salvatore Papa
- Cell Signaling and Cancer Laboratory, Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James' University Hospital, Beckett Street, Leeds, UK.
| | - Pui Man Choy
- Cell Signaling and Cancer Laboratory, Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, St James' University Hospital, Beckett Street, Leeds, UK.,Department of Research & Development, hVIVO PLC, Biopark, Broadwater Road, Welwyn Garden City, UK
| | - Concetta Bubici
- College of Health and Life Sciences, Department of Life Sciences, Institute of Environment, Health and Societies, Division of Biosciences, Brunel University London, Uxbridge, UK. .,Department of Medicine, Faculty of Medicine, Imperial College London, London, UK.
| |
Collapse
|
23
|
Wiese EK, Hitosugi T. Tyrosine Kinase Signaling in Cancer Metabolism: PKM2 Paradox in the Warburg Effect. Front Cell Dev Biol 2018; 6:79. [PMID: 30087897 PMCID: PMC6066570 DOI: 10.3389/fcell.2018.00079] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 07/03/2018] [Indexed: 12/31/2022] Open
Abstract
The Warburg Effect, or aerobic glycolysis, is one of the major metabolic alterations observed in cancer. Hypothesized to increase a cell's proliferative capacity via regenerating NAD+, increasing the pool of glycolytic biosynthetic intermediates, and increasing lactate production that affects the tumor microenvironment, the Warburg Effect is important for the growth and proliferation of tumor cells. The mechanisms by which a cell acquires the Warburg Effect phenotype are regulated by the expression of numerous oncogenes, including oncogenic tyrosine kinases. Oncogenic tyrosine kinases play a significant role in phosphorylating and regulating the activity of numerous metabolic enzymes. Tyrosine phosphorylation of glycolytic enzymes increases the activities of a majority of glycolytic enzymes, thus promoting increased glycolytic rate and tumor cell proliferation. Paradoxically however, tyrosine phosphorylation of pyruvate kinase M2 isoform (PKM2) results in decreased PKM2 activity, and this decrease in PKM2 activity promotes the Warburg Effect. Furthermore, recent studies have shown that PKM2 is also able to act as a protein kinase using phosphoenolpyruvate (PEP) as a substrate to promote tumorigenesis. Therefore, numerous recent studies have investigated both the role of the classical and non-canonical activity of PKM2 in promoting the Warburg Effect and tumor growth, which raise further interesting questions. In this review, we will summarize these recent advances revealing the importance of tyrosine kinases in the regulation of the Warburg Effect as well as the role of PKM2 in the promotion of tumor growth.
Collapse
Affiliation(s)
- Elizabeth K Wiese
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States.,Molecular Pharmacology and Experimental Therapeutics Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | - Taro Hitosugi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States.,Division of Oncology Research, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|