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
|
Arwani RT, Tan SCL, Sundarapandi A, Goh WP, Liu Y, Leong FY, Yang W, Zheng XT, Yu Y, Jiang C, Ang YC, Kong L, Teo SL, Chen P, Su X, Li H, Liu Z, Chen X, Yang L, Liu Y. Stretchable ionic-electronic bilayer hydrogel electronics enable in situ detection of solid-state epidermal biomarkers. NATURE MATERIALS 2024; 23:1115-1122. [PMID: 38867019 DOI: 10.1038/s41563-024-01918-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 05/08/2024] [Indexed: 06/14/2024]
Abstract
Continuous and in situ detection of biomarkers in biofluids (for example, sweat) can provide critical health data but is limited by biofluid accessibility. Here we report a sensor design that enables in situ detection of solid-state biomarkers ubiquitously present on human skin. We deploy an ionic-electronic bilayer hydrogel to facilitate the sequential dissolution, diffusion and electrochemical reaction of solid-state analytes. We demonstrate continuous monitoring of water-soluble analytes (for example, solid lactate) and water-insoluble analytes (for example, solid cholesterol) with ultralow detection limits of 0.51 and 0.26 nmol cm-2, respectively. Additionally, the bilayer hydrogel electrochemical interface reduces motion artefacts by a factor of three compared with conventional liquid-sensing electrochemical interfaces. In a clinical study, solid-state epidermal biomarkers measured by our stretchable wearable sensors showed a high correlation with biomarkers in human blood and dynamically correlated with physiological activities. These results present routes to universal platforms for biomarker monitoring without the need for biofluid acquisition.
Collapse
Affiliation(s)
- Ruth Theresia Arwani
- Department of Biomedical Engineering (BME), National University of Singapore, Singapore, Singapore
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- The N.1 Institute for Health, National University of Singapore, Singapore, Singapore
- Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore, Singapore
| | - Sherwin Chong Li Tan
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Archana Sundarapandi
- Department of Biomedical Engineering (BME), National University of Singapore, Singapore, Singapore
- The N.1 Institute for Health, National University of Singapore, Singapore, Singapore
- Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore, Singapore
| | - Wei Peng Goh
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Yin Liu
- Institute of High-Performance Computing (IHPC), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Fong Yew Leong
- Institute of High-Performance Computing (IHPC), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Weifeng Yang
- Department of Biomedical Engineering (BME), National University of Singapore, Singapore, Singapore
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Donghua University, Shanghai, P. R. China
| | - Xin Ting Zheng
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Yong Yu
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Changyun Jiang
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Yuan Ching Ang
- Department of Biomedical Engineering (BME), National University of Singapore, Singapore, Singapore
| | - Lingxuan Kong
- School of Chemistry, Chemical Engineering and Biotechnology (CCEB), Nanyang Technological University, Singapore, Singapore
| | - Siew Lang Teo
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Peng Chen
- School of Chemistry, Chemical Engineering and Biotechnology (CCEB), Nanyang Technological University, Singapore, Singapore
- Institute for Digital Molecular Analytics and Science (IDMxS), Nanyang Technological University, Singapore, Singapore
| | - Xinyi Su
- Department of Ophthalmology, Yong Loo Lin School of Medicine, Singapore, Singapore
- Institute of Molecular Cell and Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Hongying Li
- Institute of High-Performance Computing (IHPC), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Zhuangjian Liu
- Institute of High-Performance Computing (IHPC), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Xiaodong Chen
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Innovative Center for Flexible Devices (iFLEX), Max Planck-NTU Joint Laboratory for Artificial Senses, School of Materials Science and Engineering (MSE), Nanyang Technological University, Singapore, Singapore
| | - Le Yang
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
- Department of Materials Science and Engineering (MSE), National University of Singapore, Singapore, Singapore.
| | - Yuxin Liu
- Department of Biomedical Engineering (BME), National University of Singapore, Singapore, Singapore.
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
- The N.1 Institute for Health, National University of Singapore, Singapore, Singapore.
- Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore, Singapore.
| |
Collapse
|
3
|
Ayaz S, Erşan T, Dilgin Y, Apak R. A new colorimetric lactate biosensor based on CUPRAC reagent using binary enzyme (lactate-pyruvate oxidases)-immobilized silanized magnetite nanoparticles. Mikrochim Acta 2024; 191:455. [PMID: 38980437 PMCID: PMC11233342 DOI: 10.1007/s00604-024-06531-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 06/27/2024] [Indexed: 07/10/2024]
Abstract
A novel optical lactate biosensor is presented that utilizes a colorimetric interaction between H2O2 liberated by a binary enzymatic reaction and bis(neocuproine)copper(II) complex ([Cu(Nc)2]2+) known as CUPRAC (cupric reducing antioxidant capacity) reagent. In the first step, lactate oxidase (LOx) and pyruvate oxidase (POx) were separately immobilized on silanized magnetite nanoparticles (SiO2@Fe3O4 NPs), and thus, 2 mol of H2O2 was released per 1 mol of the substrate due to a sequential enzymatic reaction of the mixture of LOx-SiO2@Fe3O4 and POx-SiO2@Fe3O4 NPs with lactate and pyruvate, respectively. In the second step, the absorbance at 450 nm of the yellow-orange [Cu(Nc)2]+ complex formed through the color reaction of enzymatically produced H2O2 with [Cu(Nc)2]2+ was recorded. The results indicate that the developed colorimetric binary enzymatic biosensor exhibits a broad linear range of response between 0.5 and 50.0 µM for lactate under optimal conditions with a detection limit of 0.17 µM. The fabricated biosensor did not respond to other saccharides, while the positive interferences of certain reducing compounds such as dopamine, ascorbic acid, and uric acid were minimized through their oxidative removal with a pre-oxidant (NaBiO3) before enzymatic and colorimetric reactions. The fabricated optical biosensor was applied to various samples such as artificial blood, artificial/real sweat, and cow milk. The high recovery values (close to 100%) achieved for lactate-spiked samples indicate an acceptable accuracy of this colorimetric biosensor in the determination of lactate in real samples. Due to the increase in H2O2 production with the bienzymatic lactate sensor, the proposed method displays double-fold sensitivity relative to monoenzymatic biosensors and involves a neat color reaction with cupric-neocuproine having a clear stoichiometry as opposed to the rather indefinite stoichiometry of analogous redox dye methods.
Collapse
Affiliation(s)
- Selen Ayaz
- Department of Chemistry, Faculty of Science, Çanakkale Onsekiz Mart University, Canakkale, 17020, Turkey
| | - Teslime Erşan
- Department of Chemistry, Faculty of Science, Çanakkale Onsekiz Mart University, Canakkale, 17020, Turkey
| | - Yusuf Dilgin
- Department of Chemistry, Faculty of Science, Çanakkale Onsekiz Mart University, Canakkale, 17020, Turkey.
| | - Reşat Apak
- Department of Chemistry, Faculty of Engineering, İstanbul University-Cerrahpaşa, Avcılar, 34320, Istanbul, Turkey.
- Turkish Academy of Sciences (TUBA), Bayraktar Neighborhood, Vedat Dalokay St. No: 112, Çankaya, 06690, Ankara, Turkey.
| |
Collapse
|
4
|
Lin J, Yuan P, Lin R, Xue X, Chen M, Xing L. A Self-Powered Lactate Sensor Based on the Piezoelectric Effect for Assessing Tumor Development. SENSORS (BASEL, SWITZERLAND) 2024; 24:2161. [PMID: 38610372 PMCID: PMC11014382 DOI: 10.3390/s24072161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 03/21/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024]
Abstract
The build-up of lactate in solid tumors stands as a crucial and early occurrence in malignancy development, and the concentration of lactate in the tumor microenvironment may be a more sensitive indicator for analyzing primary tumors. In this study, we designed a self-powered lactate sensor for the rapid analysis of tumor samples, utilizing the coupling between the piezoelectric effect and enzymatic reaction. This lactate sensor is fabricated using a ZnO nanowire array modified with lactate oxidase (LOx). The sensing process does not require an external power source or batteries. The device can directly output electric signals containing lactate concentration information when subjected to external forces. The lactate concentration detection upper limit of the sensor is at least 27 mM, with a limit of detection (LOD) of approximately 1.3 mM and a response time of around 10 s. This study innovatively applied self-powered technology to the in situ detection of the tumor microenvironment and used the results to estimate the growth period of the primary tumor. The availability of this application has been confirmed through biological experiments. Furthermore, the sensor data generated by the device offer valuable insights for evaluating the likelihood of remote tumor metastasis. This study may expand the research scope of self-powered technology in the field of medical diagnosis and offer a novel perspective on cancer diagnosis.
Collapse
Affiliation(s)
- Jiayan Lin
- School of Physics, University of Electronic Science and Technology of China, Chengdu 611731, China; (J.L.); (P.Y.); (R.L.); (X.X.)
| | - Pengcheng Yuan
- School of Physics, University of Electronic Science and Technology of China, Chengdu 611731, China; (J.L.); (P.Y.); (R.L.); (X.X.)
| | - Rui Lin
- School of Physics, University of Electronic Science and Technology of China, Chengdu 611731, China; (J.L.); (P.Y.); (R.L.); (X.X.)
| | - Xinyu Xue
- School of Physics, University of Electronic Science and Technology of China, Chengdu 611731, China; (J.L.); (P.Y.); (R.L.); (X.X.)
| | - Meihua Chen
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610041, China;
| | - Lili Xing
- School of Physics, University of Electronic Science and Technology of China, Chengdu 611731, China; (J.L.); (P.Y.); (R.L.); (X.X.)
| |
Collapse
|
5
|
Trujillo MN, Jennings EQ, Hoffman EA, Zhang H, Phoebe AM, Mastin GE, Kitamura N, Reisz JA, Megill E, Kantner D, Marcinkiewicz MM, Twardy SM, Lebario F, Chapman E, McCullough RL, D'Alessandro A, Snyder NW, Cusanovich DA, Galligan JJ. Lactoylglutathione promotes inflammatory signaling in macrophages through histone lactoylation. Mol Metab 2024; 81:101888. [PMID: 38307385 PMCID: PMC10869261 DOI: 10.1016/j.molmet.2024.101888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/25/2024] [Accepted: 01/26/2024] [Indexed: 02/04/2024] Open
Abstract
Chronic, systemic inflammation is a pathophysiological manifestation of metabolic disorders. Inflammatory signaling leads to elevated glycolytic flux and a metabolic shift towards aerobic glycolysis and lactate generation. This rise in lactate corresponds with increased generation of lactoylLys modifications on histones, mediating transcriptional responses to inflammatory stimuli. Lactoylation is also generated through a non-enzymatic S-to-N acyltransfer from the glyoxalase cycle intermediate, lactoylglutathione (LGSH). Here, we report a regulatory role for LGSH in mediating histone lactoylation and inflammatory signaling. In the absence of the primary LGSH hydrolase, glyoxalase 2 (GLO2), RAW264.7 macrophages display significant elevations in LGSH and histone lactoylation with a corresponding potentiation of the inflammatory response when exposed to lipopolysaccharides. An analysis of chromatin accessibility shows that lactoylation is associated with more compacted chromatin than acetylation in an unstimulated state; upon stimulation, however, regions of the genome associated with lactoylation become markedly more accessible. Lastly, we demonstrate a spontaneous S-to-S acyltransfer of lactate from LGSH to CoA, yielding lactoyl-CoA. This represents the first known mechanism for the generation of this metabolite. Collectively, these data suggest that LGSH, and not intracellular lactate, is the primary driving factor facilitating histone lactoylation and a major contributor to inflammatory signaling.
Collapse
Affiliation(s)
- Marissa N Trujillo
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
| | - Erin Q Jennings
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
| | - Emely A Hoffman
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
| | - Hao Zhang
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Aiden M Phoebe
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
| | - Grace E Mastin
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
| | - Naoya Kitamura
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
| | - Julie A Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver - Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Emily Megill
- Center for Metabolic Disease Research, Department of Cardiovascular Sciences, Temple University, Philadelphia, PA, USA
| | - Daniel Kantner
- Center for Metabolic Disease Research, Department of Cardiovascular Sciences, Temple University, Philadelphia, PA, USA
| | - Mariola M Marcinkiewicz
- Center for Metabolic Disease Research, Department of Cardiovascular Sciences, Temple University, Philadelphia, PA, USA
| | - Shannon M Twardy
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Felicidad Lebario
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
| | - Eli Chapman
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
| | - Rebecca L McCullough
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver - Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Nathaniel W Snyder
- Center for Metabolic Disease Research, Department of Cardiovascular Sciences, Temple University, Philadelphia, PA, USA
| | - Darren A Cusanovich
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA; Asthma and Airway Disease Research Center, University of Arizona, Tucson, AZ, USA
| | - James J Galligan
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA.
| |
Collapse
|
6
|
Martin SD, Connor T, Sanigorski A, McEwen KA, Henstridge DC, Nijagal B, De Souza D, Tull DL, Meikle PJ, Kowalski GM, Bruce CR, Gregorevic P, Febbraio MA, Collier FM, Walder KR, McGee SL. Class IIa HDACs inhibit cell death pathways and protect muscle integrity in response to lipotoxicity. Cell Death Dis 2023; 14:787. [PMID: 38040704 PMCID: PMC10692215 DOI: 10.1038/s41419-023-06319-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 11/13/2023] [Accepted: 11/16/2023] [Indexed: 12/03/2023]
Abstract
Lipotoxicity, the accumulation of lipids in non-adipose tissues, alters the metabolic transcriptome and mitochondrial metabolism in skeletal muscle. The mechanisms involved remain poorly understood. Here we show that lipotoxicity increased histone deacetylase 4 (HDAC4) and histone deacetylase 5 (HDAC5), which reduced the expression of metabolic genes and oxidative metabolism in skeletal muscle, resulting in increased non-oxidative glucose metabolism. This metabolic reprogramming was also associated with impaired apoptosis and ferroptosis responses, and preserved muscle cell viability in response to lipotoxicity. Mechanistically, increased HDAC4 and 5 decreased acetylation of p53 at K120, a modification required for transcriptional activation of apoptosis. Redox drivers of ferroptosis derived from oxidative metabolism were also reduced. The relevance of this pathway was demonstrated by overexpression of loss-of-function HDAC4 and HDAC5 mutants in skeletal muscle of obese db/db mice, which enhanced oxidative metabolic capacity, increased apoptosis and ferroptosis and reduced muscle mass. This study identifies HDAC4 and HDAC5 as repressors of skeletal muscle oxidative metabolism, which is linked to inhibition of cell death pathways and preservation of muscle integrity in response to lipotoxicity.
Collapse
Affiliation(s)
- Sheree D Martin
- Institute for Mental and Physical Heath and Clinical Translation (IMPACT) and Metabolic Research Unit, School of Medicine, Deakin University, Geelong, VIC, 3216, Australia
| | - Timothy Connor
- Institute for Mental and Physical Heath and Clinical Translation (IMPACT) and Metabolic Research Unit, School of Medicine, Deakin University, Geelong, VIC, 3216, Australia
| | - Andrew Sanigorski
- Institute for Mental and Physical Heath and Clinical Translation (IMPACT) and Metabolic Research Unit, School of Medicine, Deakin University, Geelong, VIC, 3216, Australia
| | - Kevin A McEwen
- Institute for Mental and Physical Heath and Clinical Translation (IMPACT) and Metabolic Research Unit, School of Medicine, Deakin University, Geelong, VIC, 3216, Australia
| | - Darren C Henstridge
- College of Health and Medicine, School of Health Sciences, University of Tasmania, Launceston, Australia
- Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
| | - Brunda Nijagal
- Metabolomics Australia, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - David De Souza
- Metabolomics Australia, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Dedreia L Tull
- Metabolomics Australia, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
| | - Greg M Kowalski
- Institute for Mental and Physical Heath and Clinical Translation (IMPACT) and Metabolic Research Unit, School of Medicine, Deakin University, Geelong, VIC, 3216, Australia
- Institute of Physical Activity and Nutrition (IPAN) and School of Exercise and Nutrition Sciences, Deakin University, Geelong, VIC, 3216, Australia
| | - Clinton R Bruce
- Institute of Physical Activity and Nutrition (IPAN) and School of Exercise and Nutrition Sciences, Deakin University, Geelong, VIC, 3216, Australia
| | - Paul Gregorevic
- Centre for Muscle Research, Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, Australia
| | - Mark A Febbraio
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | | | - Ken R Walder
- Institute for Mental and Physical Heath and Clinical Translation (IMPACT) and Metabolic Research Unit, School of Medicine, Deakin University, Geelong, VIC, 3216, Australia
| | - Sean L McGee
- Institute for Mental and Physical Heath and Clinical Translation (IMPACT) and Metabolic Research Unit, School of Medicine, Deakin University, Geelong, VIC, 3216, Australia.
| |
Collapse
|
7
|
Trujillo MN, Jennings EQ, Hoffman EA, Zhang H, Phoebe AM, Mastin GE, Kitamura N, Reisz JA, Megill E, Kantner D, Marcinkiewicz MM, Twardy SM, Lebario F, Chapman E, McCullough RL, D'Alessandro A, Snyder NW, Cusanovich DA, Galligan JJ. Lactoylglutathione promotes inflammatory signaling in macrophages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.10.561739. [PMID: 37873172 PMCID: PMC10592727 DOI: 10.1101/2023.10.10.561739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Chronic, systemic inflammation is a pathophysiological manifestation of metabolic disorders. Inflammatory signaling leads to elevated glycolytic flux and a metabolic shift towards aerobic glycolysis and lactate generation. This rise in lactate corresponds with increased generation of lactoylLys modifications on histones, mediating transcriptional responses to inflammatory stimuli. Lactoylation is also generated through a non-enzymatic S-to-N acyltransfer from the glyoxalase cycle intermediate, lactoylglutathione (LGSH). Here, we report a regulatory role for LGSH in inflammatory signaling. In the absence of the primary LGSH hydrolase, glyoxalase 2 (GLO2), RAW264.7 macrophages display significant elevations in LGSH, while demonstrating a potentiated inflammatory response when exposed to lipopolysaccharides, corresponding with a rise in histone lactoylation. Interestingly, our data demonstrate that lactoylation is associated with more compacted chromatin than acetylation in an unstimulated state, however, upon stimulation, regions of the genome associated with lactoylation become markedly more accessible. Lastly, we demonstrate a spontaneous S-to-S acyltransfer of lactate from LGSH to CoA, yielding lactoyl-CoA. This represents the first known mechanism for the generation of this metabolite. Collectively, these data suggest that LGSH, and not intracellular lactate, is a primary contributing factor facilitating the inflammatory response.
Collapse
|
8
|
Luo X, Peng Y, Fan X, Xie X, Jin Z, Zhang X. The Crosstalk and Clinical Implications of CircRNAs and Glucose Metabolism in Gastrointestinal Cancers. Cancers (Basel) 2023; 15:cancers15082229. [PMID: 37190158 DOI: 10.3390/cancers15082229] [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: 02/21/2023] [Revised: 04/05/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
The majority of glucose in tumor cells is converted to lactate despite the presence of sufficient oxygen and functional mitochondria, a phenomenon known as the "Warburg effect" or "aerobic glycolysis". Aerobic glycolysis supplies large amounts of ATP, raw material for macromolecule synthesis, and also lactate, thereby contributing to cancer progression and immunosuppression. Increased aerobic glycolysis has been identified as a key hallmark of cancer. Circular RNAs (circRNAs) are a type of endogenous single-stranded RNAs characterized by covalently circular structures. Accumulating evidence suggests that circRNAs influence the glycolytic phenotype of various cancers. In gastrointestinal (GI) cancers, circRNAs are related to glucose metabolism by regulating specific glycolysis-associated enzymes and transporters as well as some pivotal signaling pathways. Here, we provide a comprehensive review of glucose-metabolism-associated circRNAs in GI cancers. Furthermore, we also discuss the potential clinical prospects of glycolysis-associated circRNAs as diagnostic and prognostic biomarkers and therapeutic targets in GI cancers.
Collapse
Affiliation(s)
- Xiaonuan Luo
- Guangdong Provincial Key Laboratory of Genome Stability and Disease Prevention and Regional Immunity and Diseases, Department of Pathology, Basic Medicine School, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Yin Peng
- Guangdong Provincial Key Laboratory of Genome Stability and Disease Prevention and Regional Immunity and Diseases, Department of Pathology, Basic Medicine School, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Xinmin Fan
- Guangdong Provincial Key Laboratory of Genome Stability and Disease Prevention and Regional Immunity and Diseases, Department of Pathology, Basic Medicine School, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Xiaoxun Xie
- Department of Histology and Embryology, School of Pre-Clinical Medicine, Guangxi Medical University, Nanning 530021, China
| | - Zhe Jin
- Guangdong Provincial Key Laboratory of Genome Stability and Disease Prevention and Regional Immunity and Diseases, Department of Pathology, Basic Medicine School, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Xiaojing Zhang
- Guangdong Provincial Key Laboratory of Genome Stability and Disease Prevention and Regional Immunity and Diseases, Department of Pathology, Basic Medicine School, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| |
Collapse
|
9
|
Xu X, Xu R, Hou S, Kang Z, Lü C, Wang Q, Zhang W, Wang X, Xu P, Gao C, Ma C. A Selective Fluorescent l-Lactate Biosensor Based on an l-Lactate-Specific Transcription Regulator and Förster Resonance Energy Transfer. BIOSENSORS 2022; 12:1111. [PMID: 36551077 PMCID: PMC9775004 DOI: 10.3390/bios12121111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 11/21/2022] [Accepted: 11/28/2022] [Indexed: 06/17/2023]
Abstract
Selective detection of l-lactate levels in foods, clinical, and bacterial fermentation samples has drawn intensive attention. Many fluorescent biosensors based on non-stereoselective recognition elements have been developed for lactate detection. Herein, the allosteric transcription factor STLldR from Salmonella enterica serovar Typhimurium LT2 was identified to be stereo-selectively respond to l-lactate. Then, STLldR was combined with Förster resonance energy transfer (FRET) to construct a fluorescent l-lactate biosensor FILLac. FILLac was further optimized by truncating the N- and C-terminal amino acids of STLldR between cyan and yellow fluorescent proteins. The optimized biosensor FILLac10N0C exhibited a maximum emission ratio change (ΔRmax) of 33.47 ± 1.91%, an apparent dissociation constant (Kd) of 6.33 ± 0.79 μM, and a limit of detection of 0.68 μM. FILLac10N0C was applied in 96-well microplates to detect l-lactate in bacterial fermentation samples and commercial foods such as Jiaosu and yogurt. The quantitation results of FILLac10N0C exhibited good agreement with that of a commercial l-lactate biosensor SBA-40D bioanalyzer. Thus, the biosensor FILLac10N0C compatible with high-throughput detection may be a potential choice for quantitation of l-lactate in different biological samples.
Collapse
Affiliation(s)
- Xianzhi Xu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Rong Xu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Shuang Hou
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Zhaoqi Kang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Chuanjuan Lü
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Qian Wang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Wen Zhang
- Institute of Medical Sciences, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250033, China
| | - Xia Wang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Ping Xu
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Chao Gao
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Cuiqing Ma
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| |
Collapse
|
10
|
Rosendo Machado S, Qu J, Koopman WJH, Miesen P. The DEAD-box RNA helicase Dhx15 controls glycolysis and arbovirus replication in Aedes aegypti mosquito cells. PLoS Pathog 2022; 18:e1010694. [PMID: 36441781 PMCID: PMC9731432 DOI: 10.1371/journal.ppat.1010694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 12/08/2022] [Accepted: 11/11/2022] [Indexed: 11/30/2022] Open
Abstract
Aedes aegypti mosquitoes are responsible for the transmission of arthropod-borne (arbo)viruses including dengue and chikungunya virus (CHIKV) but in contrast to human hosts, arbovirus-infected mosquitoes are able to efficiently control virus replication to sub-pathological levels. Yet, our knowledge of the molecular interactions of arboviruses with their mosquito hosts is incomplete. Here, we aimed to identify and characterize novel host genes that control arbovirus replication in Aedes mosquitoes. RNA binding proteins (RBPs) are well-known to regulate immune signaling pathways in all kingdoms of life. We therefore performed a knockdown screen targeting 461 genes encoding predicted RBPs in Aedes aegypti Aag2 cells and identified 15 genes with antiviral activity against Sindbis virus. Amongst these, the three DEAD-box RNA helicases AAEL004419/Dhx15, AAEL008728, and AAEL004859 also acted as antiviral factors in dengue and CHIKV infections. Here, we explored the mechanism of Dhx15 in regulating an antiviral transcriptional response in mosquitoes by silencing Dhx15 in Aag2 cells followed by deep-sequencing of poly-A enriched RNAs. Dhx15 knockdown in uninfected and CHIKV-infected cells resulted in differential expression of 856 and 372 genes, respectively. Interestingly, amongst the consistently downregulated genes, glycolytic process was the most enriched gene ontology (GO) term as the expression of all core enzymes of the glycolytic pathway was reduced, suggesting that Dhx15 regulates glycolytic function. A decrease in lactate production indicated that Dhx15 silencing indeed functionally impaired glycolysis. Modified rates of glycolytic metabolism have been implicated in controlling the replication of several classes of viruses and strikingly, infection of Aag2 cells with CHIKV by itself also resulted in the decrease of several glycolytic genes. Our data suggests that Dhx15 regulates replication of CHIKV, and possibly other arboviruses, by controlling glycolysis in mosquito cells.
Collapse
Affiliation(s)
- Samara Rosendo Machado
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jieqiong Qu
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Werner J. H. Koopman
- Department of Pediatrics, Amalia Children’s Hospital, Radboud Institute for Molecular Life Sciences, Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Pascal Miesen
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- * E-mail:
| |
Collapse
|
11
|
Dias C, Fernandes E, Barbosa RM, Ledo A. A Platinized Carbon Fiber Microelectrode-Based Oxidase Biosensor for Amperometric Monitoring of Lactate in Brain Slices. SENSORS (BASEL, SWITZERLAND) 2022; 22:7011. [PMID: 36146360 PMCID: PMC9501957 DOI: 10.3390/s22187011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/12/2022] [Accepted: 09/14/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Direct and real-time monitoring of lactate in the extracellular space can help elucidate the metabolic and modulatory role of lactate in the brain. Compared to in vivo studies, brain slices allow the investigation of the neural contribution separately from the effects of cerebrovascular response and permit easy control of recording conditions. METHODS We have used a platinized carbon fiber microelectrode platform to design an oxidase-based microbiosensor for monitoring lactate in brain slices with high spatial and temporal resolution operating at 32 °C. Lactate oxidase (Aerococcus viridans) was immobilized by crosslinking with glutaraldehyde and a layer of polyurethane was added to extend the linear range. Selectivity was improved by electropolymerization of m-phenylenediamine and concurrent use of a null sensor. RESULTS The lactate microbiosensor exhibited high sensitivity, selectivity, and optimal analytical performance at a pH and temperature compatible with recording in hippocampal slices. Evaluation of operational stability under conditions of repeated use supports the suitability of this design for up to three repeated assays. CONCLUSIONS The microbiosensor displayed good analytical performance to monitor rapid changes in lactate concentration in the hippocampal tissue in response to potassium-evoked depolarization.
Collapse
Affiliation(s)
- Cândida Dias
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Eliana Fernandes
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Rui M. Barbosa
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ana Ledo
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| |
Collapse
|
12
|
Conejeros I, López-Osorio S, Zhou E, Velásquez ZD, Del Río MC, Burgos RA, Alarcón P, Chaparro-Gutiérrez JJ, Hermosilla C, Taubert A. Glycolysis, monocarboxylate transport, and purinergic signaling are key events in Eimeria bovis-induced NETosis. Front Immunol 2022; 13:842482. [PMID: 36032127 PMCID: PMC9403323 DOI: 10.3389/fimmu.2022.842482] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 07/14/2022] [Indexed: 12/15/2022] Open
Abstract
The protozoan parasite Eimeria bovis is the causative agent of bovine coccidiosis, an enteric disease of global importance that significantly affects cattle productivity. Previous studies showed that bovine NETosis—an important early host innate effector mechanism of polymorphonuclear neutrophil (PMN)—is elicited by E. bovis stages. So far, the metabolic requirements of E. bovis-triggered NET formation are unknown. We here studied early glycolytic and mitochondrial responses of PMN as well as the role of pH, distinct metabolic pathways, P2 receptor-mediated purinergic signaling, and monocarboxylate transporters 1 and 2 (MCT1, MCT2) in E. bovis sporozoite-induced NET formation. Seahorse-based experiments revealed a rapid induction of both neutrophil oxygen consumption rate (OCR) and early glycolytic responses, thereby reflecting immediate PMN activation and metabolic changes upon confrontation with sporozoites. The impact of these metabolic changes on NET formation was studied via chemical inhibition experiments targeting glycolysis and energy generation by the use of 2-fluor-2-deoxy-D-glucose (FDG), 6-diazo-5-oxo-L-norleucin (DON), sodium dichloroacetate (DCA), oxythiamine (OT), sodium oxamate (OXA), and oligomycin A (OmA) to block glycolysis, glutaminolysis, pyruvate dehydrogenase kinase, pyruvate dehydrogenase, lactate dehydrogenase, and mitochondrial ATP-synthase, respectively. Overall, sporozoite-induced NET formation was significantly diminished via PMN pretreatments with OmA and OXA, thereby indicating a key role of ATP- and lactate-mediated metabolic pathways. Consequently, we additionally studied the effects of extracellular pH, MCT1, MCT2, and purinergic receptor inhibitors (AR-C141900, AR-C155858, theobromine, and NF449, respectively). Pretreatment with the latter inhibitors led to blockage of sporozoite-triggered DNA release from exposed bovine PMN. This report provides first evidence on the pivotal role of carbohydrate-related metabolic pathways and purinergic receptors being involved in E. bovis sporozoite-induced NETosis.
Collapse
Affiliation(s)
- Iván Conejeros
- Institute of Parasitology, Justus -Liebig University Giessen, Giessen, Germany
- *Correspondence: Iván Conejeros,
| | - Sara López-Osorio
- Institute of Parasitology, Justus -Liebig University Giessen, Giessen, Germany
- CIBAV Research Group, Facultad de Ciencias Agrarias, Universidad de Antioquia (UdeA), Medellín, Colombia
| | - Ershun Zhou
- Institute of Parasitology, Justus -Liebig University Giessen, Giessen, Germany
- College of Life Sciences and Engineering, University of Foshan, Foshan, China
| | - Zahady D. Velásquez
- Institute of Parasitology, Justus -Liebig University Giessen, Giessen, Germany
| | - María Cristina Del Río
- Department of Animal Pathology, Faculty of Veterinary Medicine, Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Rafael Agustín Burgos
- Laboratory of Inflammation Pharmacology, Faculty of Veterinary Science, Institute of Pharmacology and Morphophysiology, Universidad Austral de Chile, Valdivia, Chile
| | - Pablo Alarcón
- Laboratory of Inflammation Pharmacology, Faculty of Veterinary Science, Institute of Pharmacology and Morphophysiology, Universidad Austral de Chile, Valdivia, Chile
| | | | - Carlos Hermosilla
- Institute of Parasitology, Justus -Liebig University Giessen, Giessen, Germany
| | - Anja Taubert
- Institute of Parasitology, Justus -Liebig University Giessen, Giessen, Germany
| |
Collapse
|
13
|
Lee H, Xu Y, Zhu X, Jang C, Choi W, Bae H, Wang W, He L, Jin S, Arany Z, Simons M. Endothelium-derived lactate is required for pericyte function and blood-brain barrier maintenance. EMBO J 2022; 41:e109890. [PMID: 35243676 PMCID: PMC9058541 DOI: 10.15252/embj.2021109890] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 02/01/2022] [Accepted: 02/03/2022] [Indexed: 01/05/2023] Open
Abstract
Endothelial cells differ from other cell types responsible for the formation of the vascular wall in their unusual reliance on glycolysis for most energy needs, which results in extensive production of lactate. We find that endothelium-derived lactate is taken up by pericytes, and contributes substantially to pericyte metabolism including energy generation and amino acid biosynthesis. Endothelial-pericyte proximity is required to facilitate the transport of endothelium-derived lactate into pericytes. Inhibition of lactate production in the endothelium by deletion of the glucose transporter-1 (GLUT1) in mice results in loss of pericyte coverage in the retina and brain vasculatures, leading to the blood-brain barrier breakdown and increased permeability. These abnormalities can be largely restored by oral lactate administration. Our studies demonstrate an unexpected link between endothelial and pericyte metabolisms and the role of endothelial lactate production in the maintenance of the blood-brain barrier integrity. In addition, our observations indicate that lactate supplementation could be a useful therapeutic approach for GLUT1 deficiency metabolic syndrome patients.
Collapse
Affiliation(s)
- Heon‐Woo Lee
- Yale Cardiovascular Research CenterSection of Cardiovascular MedicineDepartment of Internal MedicineYale University School of MedicineNew HavenCTUSA
| | - Yanying Xu
- Yale Cardiovascular Research CenterSection of Cardiovascular MedicineDepartment of Internal MedicineYale University School of MedicineNew HavenCTUSA
- Department of Cardiovascular MedicineXiangya HospitalCentral South UniversityChangshaChina
| | - Xiaolong Zhu
- Yale Cardiovascular Research CenterSection of Cardiovascular MedicineDepartment of Internal MedicineYale University School of MedicineNew HavenCTUSA
| | - Cholsoon Jang
- Department of Biological ChemistryUniversity of California IrvineIrvineCAUSA
| | - Woosoung Choi
- School of Life Sciences and Cell Logistics Research CenterGwangju Institute of Science and Technology (GIST)GwangjuKorea
| | - Hosung Bae
- Department of Biological ChemistryUniversity of California IrvineIrvineCAUSA
| | - Weiwei Wang
- W. M. Keck Biotechnology Resource LaboratoryYale University School of MedicineNew HavenCTUSA
| | - Liqun He
- Department of Immunology, Genetics and PathologyRudbeck LaboratoryUppsala UniversityUppsalaSweden
| | - Suk‐Won Jin
- Yale Cardiovascular Research CenterSection of Cardiovascular MedicineDepartment of Internal MedicineYale University School of MedicineNew HavenCTUSA
- School of Life Sciences and Cell Logistics Research CenterGwangju Institute of Science and Technology (GIST)GwangjuKorea
| | - Zoltan Arany
- Cardiovascular InstitutePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Michael Simons
- Yale Cardiovascular Research CenterSection of Cardiovascular MedicineDepartment of Internal MedicineYale University School of MedicineNew HavenCTUSA
- Department of Cell BiologyYale University School of MedicineNew HavenCTUSA
| |
Collapse
|
14
|
Tao J, Li C, Zheng Y, Wang F, Zhang M, Wu X, Chen Y, Zeng Q, Chen F, Fei W. Biological protein mediated ferroptotic tumor nanotherapeutics. J Mater Chem B 2021; 9:9262-9284. [PMID: 34730601 DOI: 10.1039/d1tb01289d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Ferroptosis, a cell death pathway involving iron-related generation of lipid hydroperoxides for achieving incredible tumor suppression, has reignited the hope of chemotherapy in tumor treatment in the past decade. With extensive research studies, various bioactive proteins and cellular pathways have been demonstrated to regulate the occurrence and development of ferroptosis. The gradually established ferroptotic regulatory network is conducive to find effective proteins from a holistic perspective and guides better designs for future ferroptotic tumor therapies. The first section of this review summarizes the recent advances in ferroptotic regulatory mechanisms of proteins and attempts to clarify their latent function in the ferroptotic regulatory network. Second, the existing protein-mediated ferroptotic tumor nanotherapeutic strategies were reviewed, including the protein-mediated iron supplement, cell membrane transporter inhibition, glutathione peroxidase 4 interference, glutathione depletion, bioenzyme-mediated reactive oxygen species generation, heat shock protein inhibition, and tumor-overexpressed protein-triggered drug release for ferroptotic therapy. Finally, the future expectations and challenges of ferroptotic tumor nanotherapeutics for clinical cancer therapy are highlighted.
Collapse
Affiliation(s)
- Jiaoyang Tao
- Department of Pharmacy, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Chaoqun Li
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Yongquan Zheng
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Fengmei Wang
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Meng Zhang
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Xiaodong Wu
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yue Chen
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Qingquan Zeng
- Eye Center, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Fengying Chen
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Weidong Fei
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| |
Collapse
|
15
|
Verma M, Naqvi TK, Tripathi SK, Kulkarni MM, Prasad NE, Dwivedi PK. Plasmonic Paper based Flexible SERS Biosensor for Highly Sensitive Detection of Lactic and Uric Acid. IEEE Trans Nanobioscience 2021; 21:294-300. [PMID: 34710047 DOI: 10.1109/tnb.2021.3124055] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Selective detection and quantification of biomarkers related to human diseases are essential for preventive healthcare. Surface-enhanced Raman scattering (SERS) spectroscopy is a powerful analytical tool offering high sensitivity. However, the success of this promising analytical tool relies on the ability to effectively fabricate SERS substrate. Herein we have demonstrated a plasmonic paper-based flexible substrate (PPFS) for SERS sensing. In situ growth of silver nanostructures (AgNS) on the paper-based substrate was achieved by using a simple one-step silver mirror reaction (SMR). FESEM and TEM results depicts that the increasing silver ion content influences the morphology (growth of multifacets), as well as size of AgNS. Further, the PPFS substrate was tested with Rhodamine-6G (Rh-6G) dye and an attomole sensitivity with a LOD of 4.54 x 10-18 M was achieved. Further, two biomarkers, lactic acid (LA) and uric acid (UA) were detected on the PPFS substrate, with μM and pM sensitivity, having LOD values of 0.6 x 10-6 and 0.3 x 10-12 M respectively. Above detection levels for UA on PPFS is two orders better than reported values, whereas for LA it is comparable with reported substrates. Finally, UA, LA and their mixtures were tested on PPFS and results compared with commercial substrate. The performance of PPFS were found better in all cases, thus, multifaceted AgNS paper based PPFS offers the potential to be used as a biosensor for detection of various biomarkers from body fluids, responsible for the detection of the critical disease for preventive health care.
Collapse
|
16
|
Mahan VL. Effects of lactate and carbon monoxide interactions on neuroprotection and neuropreservation. Med Gas Res 2021; 11:158-173. [PMID: 34213499 PMCID: PMC8374456 DOI: 10.4103/2045-9912.318862] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 08/21/2020] [Accepted: 10/23/2020] [Indexed: 11/04/2022] Open
Abstract
Lactate, historically considered a waste product of anerobic metabolism, is a metabolite in whole-body metabolism needed for normal central nervous system (CNS) functions and a potent signaling molecule and hormone in the CNS. Neuronal activity signals normally induce its formation primarily in astrocytes and production is dependent on anerobic and aerobic metabolisms. Functions are dependent on normal dynamic, expansive, and evolving CNS functions. Levels can change under normal physiologic conditions and with CNS pathology. A readily combusted fuel that is sshuttled throughout the body, lactate is used as an energy source and is needed for CNS hemostasis, plasticity, memory, and excitability. Diffusion beyond the neuron active zone impacts activity of neurons and astrocytes in other areas of the brain. Barriergenesis, function of the blood-brain barrier, and buffering between oxidative metabolism and glycolysis and brain metabolism are affected by lactate. Important to neuroprotection, presence or absence is associated with L-lactate and heme oxygenase/carbon monoxide (a gasotransmitter) neuroprotective systems. Effects of carbon monoxide on L-lactate affect neuroprotection - interactions of the gasotransmitter with L-lactate are important to CNS stability, which will be reviewed in this article.
Collapse
Affiliation(s)
- Vicki L. Mahan
- Department of Surgery and Pediatrics, Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
17
|
Zhu X, Jin C, Pan Q, Hu X. Determining the quantitative relationship between glycolysis and GAPDH in cancer cells exhibiting the Warburg effect. J Biol Chem 2021; 296:100369. [PMID: 33545174 PMCID: PMC7960551 DOI: 10.1016/j.jbc.2021.100369] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 01/29/2021] [Accepted: 02/01/2021] [Indexed: 11/04/2022] Open
Abstract
Previous studies have identified GAPDH as a promising target for treating cancer and modulating immunity because its inhibition reduces glycolysis in cells (cancer cells and immune cells) with the Warburg effect, a modified form of cellular metabolism found in cancer cells. However, the quantitative relationship between GAPDH and the aerobic glycolysis remains unknown. Here, using siRNA-mediated knockdown of GAPDH expression and iodoacetate-dependent inhibition of enzyme activity, we examined the quantitative relationship between GAPDH activity and glycolysis rate. We found that glycolytic rates were unaffected by the reduction of GAPDH activity down to 19% ± 4.8% relative to untreated controls. However, further reduction of GAPDH activity below this level caused proportional reductions in the glycolysis rate. GAPDH knockdown or inhibition also simultaneously increased the concentration of glyceraldehyde 3-phosphate (GA3P, the substrate of GAPDH). This increased GA3P concentration countered the effect of GAPDH knockdown or inhibition and stabilized the glycolysis rate by promoting GAPDH activity. Mechanistically, the intracellular GA3P concentration is controlled by the Gibbs free energy of the reactions upstream of GAPDH. The thermodynamic state of the reactions along the glycolysis pathway was only affected when GAPDH activity was reduced below 19% ± 4.8%. Doing so moved the reactions catalyzed by GAPDH + PGK1 (phosphoglycerate kinase 1, the enzyme immediate downstream of GAPDH) away from the near-equilibrium state, revealing an important biochemical basis to interpret the rate control of glycolysis by GAPDH. Collectively, we resolved the numerical relationship between GAPDH and glycolysis in cancer cells with the Warburg effect and interpreted the underlying mechanism.
Collapse
Affiliation(s)
- Xiaobing Zhu
- Cancer Institute (Key Laboratory for Cancer Intervention and Prevention, China National Ministry of Education, Zhejiang Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chengmeng Jin
- Cancer Institute (Key Laboratory for Cancer Intervention and Prevention, China National Ministry of Education, Zhejiang Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiangrong Pan
- Cancer Institute (Key Laboratory for Cancer Intervention and Prevention, China National Ministry of Education, Zhejiang Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xun Hu
- Cancer Institute (Key Laboratory for Cancer Intervention and Prevention, China National Ministry of Education, Zhejiang Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
18
|
Time-resolved non-invasive metabolomic monitoring of a single cancer spheroid by microfluidic NMR. Sci Rep 2021; 11:53. [PMID: 33420162 PMCID: PMC7794408 DOI: 10.1038/s41598-020-79693-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 11/27/2020] [Indexed: 02/07/2023] Open
Abstract
We present a quantitative study of the metabolic activity of a single spheroid culture of human cancer cells. NMR (nuclear magnetic resonance) spectroscopy is an ideal tool for observation of live systems due to its non-invasive nature. However, limited sensitivity has so far hindered its application in microfluidic culture systems. We have used an optimised micro-NMR platform to observe metabolic changes from a single spheroid. NMR spectra were obtained by directly inserting microfluidic devices containing spheroids ranging from 150 [Formula: see text]m to 300 [Formula: see text]m in diameter in 2.5 [Formula: see text]L of culture medium into a dedicated NMR probe. Metabolite concentrations were found to change linearly with time, with rates approximately proportional to the number of cells in the spheroid. The results demonstrate that quantitative monitoring of a single spheroid with [Formula: see text] 2500 cells is possible. A change in spheroid size by 600 cells leads to a clearly detectable change in the L-Lactic acid production rate ([Formula: see text]). The consumption of D-Glucose and production of L-Lactic acid were approximately 2.5 times slower in spheroids compared to monolayer culture of the same number of cells. Moreover, while cells in monolayer culture were found to produce L-Alanine and L-Glutamine, spheroids showed slight consumption in both cases.
Collapse
|
19
|
Zhou X, Zhao W, Wang M, Zhang S, Li Y, Hu W, Ren L, Luo S, Chen Z. Dual-Modal Therapeutic Role of the Lactate Oxidase-Embedded Hierarchical Porous Zeolitic Imidazolate Framework as a Nanocatalyst for Effective Tumor Suppression. ACS APPLIED MATERIALS & INTERFACES 2020; 12:32278-32288. [PMID: 32580547 DOI: 10.1021/acsami.0c05783] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The increasing evidence supports the fact that lactate in the tumor microenvironment (TME) plays a vital role in tumor proliferation, metastasis, and recurrence, which in turn is emerging as one of the most interesting molecular targets for tumor treatment. Here, hierarchical porous zeolitic imidazolate framework-8 (ZIF-8) as the nanocarrier is fabricated to simultaneously load lactate oxidase (LOD) and Fe3O4 nanoparticles (NPs), called LOD & Fe3O4@ZIF-8 NPs (LFZ NPs), for tumor therapy. On one hand, the sharp consumption of lactate in the TME by LOD will change the essential "soil" where tumor cells live so as to suppress tumor rapid growth. On the other hand, hydrogen peroxide (H2O2) is produced in the TME from the oxidation of lactate catalyzed by LOD and subsequently converted to highly toxic hydroxyl radicals (•OH) catalyzed by Fe3O4 NPs via Fenton-like reactions to kill tumor cells. Based on the endogenous catalysis, this dual-modal strategy of tumor therapy based on lactate is simple, safe, and effective, which deserves to be well concerned.
Collapse
Affiliation(s)
- Xi Zhou
- Department of Biomaterials, College of Materials, Xiamen University, Xiamen, Fujian 361005, P. R. China
| | - Wen Zhao
- Department of Biomaterials, College of Materials, Xiamen University, Xiamen, Fujian 361005, P. R. China
| | - Muxue Wang
- Department of Biomaterials, College of Materials, Xiamen University, Xiamen, Fujian 361005, P. R. China
| | - Shuai Zhang
- Department of Biomaterials, College of Materials, Xiamen University, Xiamen, Fujian 361005, P. R. China
| | - Yunhong Li
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance Research, School of Electronic Science and Engineering, Xiamen University, Xiamen, Fujian 361005, P. R. China
| | - Wenxin Hu
- Harvard College, Harvard University, 209 Dunster Mail Center, 945 Memorial Drive, Cambridge, Massachusetts 02138, United States
| | - Lei Ren
- Department of Biomaterials, College of Materials, Xiamen University, Xiamen, Fujian 361005, P. R. China
| | - Shenglin Luo
- State Key Laboratory of Trauma Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, P. R. China
| | - Zhiwei Chen
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance Research, School of Electronic Science and Engineering, Xiamen University, Xiamen, Fujian 361005, P. R. China
| |
Collapse
|
20
|
Jin C, Zhu X, Wu H, Wang Y, Hu X. Perturbation of phosphoglycerate kinase 1 (PGK1) only marginally affects glycolysis in cancer cells. J Biol Chem 2020; 295:6425-6446. [PMID: 32217690 DOI: 10.1074/jbc.ra119.012312] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 03/23/2020] [Indexed: 12/20/2022] Open
Abstract
Phosphoglycerate kinase 1 (PGK1) plays important roles in glycolysis, yet its forward reaction kinetics are unknown, and its role especially in regulating cancer cell glycolysis is unclear. Here, we developed an enzyme assay to measure the kinetic parameters of the PGK1-catalyzed forward reaction. The Km values for 1,3-bisphosphoglyceric acid (1,3-BPG, the forward reaction substrate) were 4.36 μm (yeast PGK1) and 6.86 μm (human PKG1). The Km values for 3-phosphoglycerate (3-PG, the reverse reaction substrate and a serine precursor) were 146 μm (yeast PGK1) and 186 μm (human PGK1). The V max of the forward reaction was about 3.5- and 5.8-fold higher than that of the reverse reaction for the human and yeast enzymes, respectively. Consistently, the intracellular steady-state concentrations of 3-PG were between 180 and 550 μm in cancer cells, providing a basis for glycolysis to shuttle 3-PG to the serine synthesis pathway. Using siRNA-mediated PGK1-specific knockdown in five cancer cell lines derived from different tissues, along with titration of PGK1 in a cell-free glycolysis system, we found that the perturbation of PGK1 had no effect or only marginal effects on the glucose consumption and lactate generation. The PGK1 knockdown increased the concentrations of fructose 1,6-bisphosphate, dihydroxyacetone phosphate, glyceraldehyde 3-phosphate, and 1,3-BPG in nearly equal proportions, controlled by the kinetic and thermodynamic states of glycolysis. We conclude that perturbation of PGK1 in cancer cells insignificantly affects the conversion of glucose to lactate in glycolysis.
Collapse
Affiliation(s)
- Chengmeng Jin
- Cancer Institute (Key Laboratory for Cancer Intervention and Prevention, China National Ministry of Education, Zhejiang Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Xiaobing Zhu
- Cancer Institute (Key Laboratory for Cancer Intervention and Prevention, China National Ministry of Education, Zhejiang Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Hao Wu
- Cancer Institute (Key Laboratory for Cancer Intervention and Prevention, China National Ministry of Education, Zhejiang Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Yuqi Wang
- Cancer Institute (Key Laboratory for Cancer Intervention and Prevention, China National Ministry of Education, Zhejiang Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Xun Hu
- Cancer Institute (Key Laboratory for Cancer Intervention and Prevention, China National Ministry of Education, Zhejiang Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| |
Collapse
|
21
|
Krycer JR, Quek LE, Francis D, Fazakerley DJ, Elkington SD, Diaz-Vegas A, Cooke KC, Weiss FC, Duan X, Kurdyukov S, Zhou PX, Tambar UK, Hirayama A, Ikeda S, Kamei Y, Soga T, Cooney GJ, James DE. Lactate production is a prioritized feature of adipocyte metabolism. J Biol Chem 2020; 295:83-98. [PMID: 31690627 PMCID: PMC6952601 DOI: 10.1074/jbc.ra119.011178] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 10/31/2019] [Indexed: 12/14/2022] Open
Abstract
Adipose tissue is essential for whole-body glucose homeostasis, with a primary role in lipid storage. It has been previously observed that lactate production is also an important metabolic feature of adipocytes, but its relationship to adipose and whole-body glucose disposal remains unclear. Therefore, using a combination of metabolic labeling techniques, here we closely examined lactate production of cultured and primary mammalian adipocytes. Insulin treatment increased glucose uptake and conversion to lactate, with the latter responding more to insulin than did other metabolic fates of glucose. However, lactate production did not just serve as a mechanism to dispose of excess glucose, because we also observed that lactate production in adipocytes did not solely depend on glucose availability and even occurred independently of glucose metabolism. This suggests that lactate production is prioritized in adipocytes. Furthermore, knocking down lactate dehydrogenase specifically in the fat body of Drosophila flies lowered circulating lactate and improved whole-body glucose disposal. These results emphasize that lactate production is an additional metabolic role of adipose tissue beyond lipid storage and release.
Collapse
Affiliation(s)
- James R Krycer
- School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia; Charles Perkins Centre, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Lake-Ee Quek
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales 2006, Australia; School of Mathematics and Statistics, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Deanne Francis
- School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia; Charles Perkins Centre, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Daniel J Fazakerley
- School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia; Charles Perkins Centre, The University of Sydney, Sydney, New South Wales 2006, Australia; Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, United Kingdom
| | - Sarah D Elkington
- School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia; Charles Perkins Centre, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Alexis Diaz-Vegas
- School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia; Charles Perkins Centre, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Kristen C Cooke
- School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia; Charles Perkins Centre, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Fiona C Weiss
- School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia; Charles Perkins Centre, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Xiaowen Duan
- School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia; Charles Perkins Centre, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Sergey Kurdyukov
- School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia; Charles Perkins Centre, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Ping-Xin Zhou
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9038; School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Uttam K Tambar
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9038
| | - Akiyoshi Hirayama
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0052, Japan; AMED-CREST, Japan Agency for Medical Research and Development (AMED), 1-7-1 Otemachi, Chiyoda-Ku, Tokyo 100-0004, Japan
| | - Satsuki Ikeda
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0052, Japan
| | - Yushi Kamei
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0052, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0052, Japan; AMED-CREST, Japan Agency for Medical Research and Development (AMED), 1-7-1 Otemachi, Chiyoda-Ku, Tokyo 100-0004, Japan
| | - Gregory J Cooney
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales 2006, Australia; Sydney Medical School, The University of Sydney, Sydney, New South Wales 2006, Australia.
| | - David E James
- School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia; Charles Perkins Centre, The University of Sydney, Sydney, New South Wales 2006, Australia; Sydney Medical School, The University of Sydney, Sydney, New South Wales 2006, Australia.
| |
Collapse
|
22
|
Yadav S, Pandey SK, Goel Y, Temre MK, Singh SM. Diverse Stakeholders of Tumor Metabolism: An Appraisal of the Emerging Approach of Multifaceted Metabolic Targeting by 3-Bromopyruvate. Front Pharmacol 2019; 10:728. [PMID: 31333455 PMCID: PMC6620530 DOI: 10.3389/fphar.2019.00728] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 06/05/2019] [Indexed: 12/14/2022] Open
Abstract
Malignant cells possess a unique metabolic machinery to endure unobstructed cell survival. It comprises several levels of metabolic networking consisting of 1) upregulated expression of membrane-associated transporter proteins, facilitating unhindered uptake of substrates; 2) upregulated metabolic pathways for efficient substrate utilization; 3) pH and redox homeostasis, conducive for driving metabolism; 4) tumor metabolism-dependent reconstitution of tumor growth promoting the external environment; 5) upregulated expression of receptors and signaling mediators; and 6) distinctive genetic and regulatory makeup to generate and sustain rearranged metabolism. This feat is achieved by a "battery of molecular patrons," which acts in a highly cohesive and mutually coordinated manner to bestow immortality to neoplastic cells. Consequently, it is necessary to develop a multitargeted therapeutic approach to achieve a formidable inhibition of the diverse arrays of tumor metabolism. Among the emerging agents capable of such multifaceted targeting of tumor metabolism, an alkylating agent designated as 3-bromopyruvate (3-BP) has gained immense research focus because of its broad spectrum and specific antineoplastic action. Inhibitory effects of 3-BP are imparted on a variety of metabolic target molecules, including transporters, metabolic enzymes, and several other crucial stakeholders of tumor metabolism. Moreover, 3-BP ushers a reconstitution of the tumor microenvironment, a reversal of tumor acidosis, and recuperative action on vital organs and systems of the tumor-bearing host. Studies have been conducted to identify targets of 3-BP and its derivatives and characterization of target binding for further optimization. This review presents a brief and comprehensive discussion about the current state of knowledge concerning various aspects of tumor metabolism and explores the prospects of 3-BP as a safe and effective antineoplastic agent.
Collapse
Affiliation(s)
| | | | | | | | - Sukh Mahendra Singh
- School of Biotechnology, Institute of Science, Banaras Hindu University, Varanasi, India
| |
Collapse
|
23
|
Schruf E, Schroeder V, Kuttruff CA, Weigle S, Krell M, Benz M, Bretschneider T, Holweg A, Schuler M, Frick M, Nicklin P, Garnett JP, Sobotta MC. Human lung fibroblast-to-myofibroblast transformation is not driven by an LDH5-dependent metabolic shift towards aerobic glycolysis. Respir Res 2019; 20:87. [PMID: 31072408 PMCID: PMC6507142 DOI: 10.1186/s12931-019-1058-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 04/24/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a fatal respiratory disease characterized by aberrant fibroblast activation and progressive fibrotic remodelling of the lungs. Though the exact pathophysiological mechanisms of IPF remain unknown, TGF-β1 is thought to act as a main driver of the disease by mediating fibroblast-to-myofibroblast transformation (FMT). Recent reports have indicated that a metabolic shift towards aerobic glycolysis takes place during FMT and that metabolic shifts can directly influence aberrant cell function. This has led to the hypothesis that inhibition of lactate dehydrogenase 5 (LDH5), an enzyme responsible for converting pyruvate into lactate, could constitute a therapeutic concept for IPF. METHODS In this study, we investigated the potential link between aerobic glycolysis and FMT using a potent LDH5 inhibitor (Compound 408, Genentech). Seahorse analysis was performed to determine the effect of Compound 408 on TGF-β1-driven glycolysis in WI-38 fibroblasts. TGF-β1-mediated FMT was measured by quantifying α-smooth muscle actin (α-SMA) and fibronectin in primary human lung fibroblasts following treatment with Compound 408. Lactate and pyruvate levels in the cell culture supernatant were assessed by LC-MS/MS. In addition to pharmacological LDH5 inhibition, the effect of siRNA-mediated knockdown of LDHA and LDHB on FMT was examined. RESULTS We show that treatment of lung fibroblasts with Compound 408 efficiently inhibits LDH5 and attenuates the TGF-β1-mediated metabolic shift towards aerobic glycolysis. Additionally, we demonstrate that LDH5 inhibition has no significant effect on TGF-β1-mediated FMT in primary human lung fibroblasts by analysing α-SMA fibre formation and fibronectin expression. CONCLUSIONS Our data strongly suggest that while LDH5 inhibition can prevent metabolic shifts in fibroblasts, it has no influence on FMT and therefore glycolytic dysregulation is unlikely to be the sole driver of FMT.
Collapse
Affiliation(s)
- Eva Schruf
- Immunology & Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Straße 65, 88397, Biberach an der Riss, Germany
| | - Victoria Schroeder
- Immunology & Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Straße 65, 88397, Biberach an der Riss, Germany
| | - Christian A Kuttruff
- Immunology & Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Straße 65, 88397, Biberach an der Riss, Germany.,Medicinal Chemistry, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Straße 65, 88397, Biberach an der Riss, Germany
| | - Sabine Weigle
- Immunology & Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Straße 65, 88397, Biberach an der Riss, Germany.,Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Straße 65, 88397, Biberach an der Riss, Germany
| | - Martin Krell
- Immunology & Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Straße 65, 88397, Biberach an der Riss, Germany
| | - Maryke Benz
- Immunology & Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Straße 65, 88397, Biberach an der Riss, Germany
| | - Tom Bretschneider
- Immunology & Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Straße 65, 88397, Biberach an der Riss, Germany
| | - Alexander Holweg
- Immunology & Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Straße 65, 88397, Biberach an der Riss, Germany
| | - Michael Schuler
- Immunology & Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Straße 65, 88397, Biberach an der Riss, Germany
| | - Manfred Frick
- Medicinal Chemistry, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Straße 65, 88397, Biberach an der Riss, Germany.,Institute of General Physiology, University of Ulm, Ulm, Germany
| | - Paul Nicklin
- Immunology & Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Straße 65, 88397, Biberach an der Riss, Germany
| | - James P Garnett
- Immunology & Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Straße 65, 88397, Biberach an der Riss, Germany.
| | - Mirko C Sobotta
- Immunology & Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Straße 65, 88397, Biberach an der Riss, Germany.
| |
Collapse
|
24
|
Qin Z, Xiang C, Zhong F, Liu Y, Dong Q, Li K, Shi W, Ding C, Qin L, He F. Transketolase (TKT) activity and nuclear localization promote hepatocellular carcinoma in a metabolic and a non-metabolic manner. J Exp Clin Cancer Res 2019; 38:154. [PMID: 30971297 PMCID: PMC6458711 DOI: 10.1186/s13046-019-1131-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 03/08/2019] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Metabolic reprogramming is one of the hallmarks of cancer cells. The pentose phosphate pathway (PPP), a branch of glycolysis, is an important metabolic pathway for the survival and biosynthesis of cancer cells. Transketolase (TKT) is a key enzyme in the non-oxidative phase of PPP. The mechanistic details of TKT in hepatocellular carcinoma (HCC) development remain unclear. METHODS TKT level and subcellular location were examined in HCC cell lines and tissue samples. We established the TKT overexpression and knocking-down stable cells in HCC cell lines. Proliferation, migration, viability and enzyme activity assays in vitro, tumor growth and metastasis assays in vivo were employed to test the effects of TKT on HCC development. GFP-tagged TKT truncations and mutants were used to locate the nuclear localization sequence (NLSs) of TKT. Cross-linking co-IP/MS was applied to identify the interaction proteins of nuclear TKT. RESULTS We showed that TKT increased the proliferation and migration of HCC cells, as well as the viability under oxidative stress in vitro and accelerated the growth and metastasis of HCC cells in vivo. We found as a key enzyme of PPP, TKT could promote the proliferation, cell cycle, migration and viability by regulating the metabolic flux. Moreover, it was firstly reported that unlike other key enzymes in PPP, TKT showed a strong nuclear localization in HCC cells. We found not only high TKT expression, but also its nuclear localization was a prediction for poor prognosis of HCC patients. We further identified the nuclear localization sequences (NLS) for TKT and demonstrated the NLS mutations decreased the pro-tumor function of TKT independent of the enzyme activity. Cross-linking Co-IP/MS showed that nuclear TKT interacted with kinases and transcriptional coregulators such as EGFR and MAPK3, which are associated with cell activation or stress response processes. EGF treatment significantly increased the viability and proliferation of HCC cells in the enzyme-inactivating mutation TKT-D155A overexpression cells but not in the NLS-D155A double mutant group, which could be blocked by EGFR inhibitor erlotinib treatment. CONCLUSIONS Our research suggests that in addition to the metabolic manner, TKT can promote the development of HCC in a non-metabolic manner via its nuclear localization and EGFR pathway.
Collapse
Affiliation(s)
- Zhaoyu Qin
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032 China
| | - Chan Xiang
- Department of Pathology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030 China
| | - Fan Zhong
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032 China
| | - Yang Liu
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032 China
| | - Qiongzhu Dong
- Department of Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, 200040 China
| | - Kai Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing, 102206 China
| | - Wenhao Shi
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing, 102206 China
| | - Chen Ding
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032 China
| | - Lunxiu Qin
- Department of Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, 200040 China
| | - Fuchu He
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032 China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing, 102206 China
| |
Collapse
|
25
|
HSIEH CL, LIN PY, AKITA T, MITA M, IDE T, LEE JA, HAMASE K. Development of a Three-Dimensional HPLC System for the Simultaneous Determination of Lactate and 3-Hydroxybutyrate Enantiomers in Mammalian Urine. CHROMATOGRAPHY 2019. [DOI: 10.15583/jpchrom.2018.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Chin-Ling HSIEH
- Graduate School of Pharmaceutical Sciences, Kyushu University
- School of Pharmacy, Taipei Medical University
| | - Po-Yeh LIN
- School of Pharmacy, Taipei Medical University
| | - Takeyuki AKITA
- Graduate School of Pharmaceutical Sciences, Kyushu University
| | | | - Tomomi IDE
- Graduate School of Medical Sciences, Kyushu University
| | - Jen-Ai LEE
- School of Pharmacy, Taipei Medical University
| | - Kenji HAMASE
- Graduate School of Pharmaceutical Sciences, Kyushu University
- School of Pharmacy, Taipei Medical University
| |
Collapse
|
26
|
Segarra-Mondejar M, Casellas-Díaz S, Ramiro-Pareta M, Müller-Sánchez C, Martorell-Riera A, Hermelo I, Reina M, Aragonés J, Martínez-Estrada OM, Soriano FX. Synaptic activity-induced glycolysis facilitates membrane lipid provision and neurite outgrowth. EMBO J 2018; 37:e97368. [PMID: 29615453 PMCID: PMC5920244 DOI: 10.15252/embj.201797368] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 02/21/2018] [Accepted: 03/05/2018] [Indexed: 12/22/2022] Open
Abstract
The formation of neurites is an important process affecting the cognitive abilities of an organism. Neurite growth requires the addition of new membranes, but the metabolic remodeling necessary to supply lipids for membrane expansion is poorly understood. Here, we show that synaptic activity, one of the most important inducers of neurite growth, transcriptionally regulates the expression of neuronal glucose transporter Glut3 and rate-limiting enzymes of glycolysis, resulting in enhanced glucose uptake and metabolism that is partly used for lipid synthesis. Mechanistically, CREB regulates the expression of Glut3 and Siah2, the latter and LDH activity promoting the normoxic stabilization of HIF-1α that regulates the expression of rate-limiting genes of glycolysis. The expression of dominant-negative HIF-1α or Glut3 knockdown blocks activity-dependent neurite growth in vitro while pharmacological inhibition of the glycolysis and specific ablation of HIF-1α in early postnatal mice impairs the neurite architecture. These results suggest that the manipulation of neuronal glucose metabolism could be used to treat some brain developmental disorders.
Collapse
Affiliation(s)
- Marc Segarra-Mondejar
- Celltec-UB, Department of Cell Biology, Physiology and Immunology, University of Barcelona, Barcelona, Spain
- Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Sergi Casellas-Díaz
- Celltec-UB, Department of Cell Biology, Physiology and Immunology, University of Barcelona, Barcelona, Spain
- Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Marina Ramiro-Pareta
- Celltec-UB, Department of Cell Biology, Physiology and Immunology, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine, University of Barcelona, Barcelona, Spain
| | - Claudia Müller-Sánchez
- Celltec-UB, Department of Cell Biology, Physiology and Immunology, University of Barcelona, Barcelona, Spain
| | - Alejandro Martorell-Riera
- Celltec-UB, Department of Cell Biology, Physiology and Immunology, University of Barcelona, Barcelona, Spain
| | - Ismaïl Hermelo
- Celltec-UB, Department of Cell Biology, Physiology and Immunology, University of Barcelona, Barcelona, Spain
| | - Manuel Reina
- Celltec-UB, Department of Cell Biology, Physiology and Immunology, University of Barcelona, Barcelona, Spain
| | - Julián Aragonés
- Research Unit, Hospital of La Princesa, Research Institute Princesa, Autonomous University of Madrid, Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, Carlos III Health Institute, Madrid, Spain
| | - Ofelia M Martínez-Estrada
- Celltec-UB, Department of Cell Biology, Physiology and Immunology, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine, University of Barcelona, Barcelona, Spain
| | - Francesc X Soriano
- Celltec-UB, Department of Cell Biology, Physiology and Immunology, University of Barcelona, Barcelona, Spain
- Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|