151
|
Keppel M, Hünnefeld M, Filipchyk A, Viets U, Davoudi CF, Krüger A, Mack C, Pfeifer E, Polen T, Baumgart M, Bott M, Frunzke J. HrrSA orchestrates a systemic response to heme and determines prioritization of terminal cytochrome oxidase expression. Nucleic Acids Res 2020; 48:6547-6562. [PMID: 32453397 PMCID: PMC7337898 DOI: 10.1093/nar/gkaa415] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 04/26/2020] [Accepted: 05/05/2020] [Indexed: 01/02/2023] Open
Abstract
Heme is a multifaceted molecule. While serving as a prosthetic group for many important proteins, elevated levels are toxic to cells. The complexity of this stimulus has shaped bacterial network evolution. However, only a small number of targets controlled by heme-responsive regulators have been described to date. Here, we performed chromatin affinity purification and sequencing to provide genome-wide insights into in vivo promoter occupancy of HrrA, the response regulator of the heme-regulated two-component system HrrSA of Corynebacterium glutamicum. Time-resolved profiling revealed dynamic binding of HrrA to more than 200 different genomic targets encoding proteins associated with heme biosynthesis, the respiratory chain, oxidative stress response and cell envelope remodeling. By repression of the extracytoplasmic function sigma factor sigC, which activates the cydABCD operon, HrrA prioritizes the expression of genes encoding the cytochrome bc1-aa3 supercomplex. This is also reflected by a significantly decreased activity of the cytochrome aa3 oxidase in the ΔhrrA mutant. Furthermore, our data reveal that HrrA also integrates the response to heme-induced oxidative stress by activating katA encoding the catalase. These data provide detailed insights in the systemic strategy that bacteria have evolved to respond to the versatile signaling molecule heme.
Collapse
Affiliation(s)
- Marc Keppel
- Institute of Bio- und Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Max Hünnefeld
- Institute of Bio- und Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Andrei Filipchyk
- Institute of Bio- und Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Ulrike Viets
- Institute of Bio- und Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Cedric-Farhad Davoudi
- Institute of Bio- und Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Aileen Krüger
- Institute of Bio- und Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Christina Mack
- Institute of Bio- und Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Eugen Pfeifer
- Microbial Evolutionary Genomics, Institute Pasteur, 75015 Paris, France
| | - Tino Polen
- Institute of Bio- und Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Meike Baumgart
- Institute of Bio- und Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Michael Bott
- Institute of Bio- und Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Julia Frunzke
- Institute of Bio- und Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich, 52425 Jülich, Germany
| |
Collapse
|
152
|
Moon SJ, Dong W, Stephanopoulos GN, Sikes HD. Oxidative pentose phosphate pathway and glucose anaplerosis support maintenance of mitochondrial NADPH pool under mitochondrial oxidative stress. Bioeng Transl Med 2020; 5:e10184. [PMID: 33005744 PMCID: PMC7510474 DOI: 10.1002/btm2.10184] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 08/21/2020] [Accepted: 08/21/2020] [Indexed: 12/16/2022] Open
Abstract
Mitochondrial NADPH protects cells against mitochondrial oxidative stress by serving as an electron donor to antioxidant defense systems. However, due to technical challenges, it still remains unknown as to the pool size of mitochondrial NADPH, its dynamics, and NADPH/NADP+ ratio. Here, we have systemically modulated production rates of H2O2 in mitochondria and assessed mitochondrial NADPH metabolism using iNap sensors, 13C glucose isotopic tracers, and a mathematical model. Using sensors, we observed decreases in mitochondrial NADPH caused by excessive generation of mitochondrial H2O2, whereas the cytosolic NADPH was maintained upon perturbation. We further quantified the extent of mitochondrial NADPH/NADP+ based on the mathematical analysis. Utilizing 13C glucose isotopic tracers, we found increased activity in the pentose phosphate pathway (PPP) accompanied small decreases in the mitochondrial NADPH pool, whereas larger decreases induced both PPP activity and glucose anaplerosis. Thus, our integrative and quantitative approach provides insight into mitochondrial NADPH metabolism during mitochondrial oxidative stress.
Collapse
Affiliation(s)
- Sun Jin Moon
- Department of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Wentao Dong
- Department of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | | | - Hadley D. Sikes
- Department of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| |
Collapse
|
153
|
Bierman JC, Laughlin T, Tamura M, Hulette BC, Mack CE, Sherrill JD, Tan CY, Morenc M, Bellanger S, Oblong JE. Niacinamide mitigates SASP‐related inflammation induced by environmental stressors in human epidermal keratinocytes and skin. Int J Cosmet Sci 2020; 42:501-511. [DOI: 10.1111/ics.12651] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/08/2020] [Accepted: 07/09/2020] [Indexed: 12/14/2022]
Affiliation(s)
| | | | | | | | | | | | - Christina Y.R. Tan
- Skin Research Institute of Singapore A*STAR 8A Biomedical Grove, #06‐06 Immunos Singapore138648Singapore
| | - Malgorzata Morenc
- Skin Research Institute of Singapore A*STAR 8A Biomedical Grove, #06‐06 Immunos Singapore138648Singapore
| | - Sophie Bellanger
- Skin Research Institute of Singapore A*STAR 8A Biomedical Grove, #06‐06 Immunos Singapore138648Singapore
| | | |
Collapse
|
154
|
Pinheiro MJ, Bonturi N, Belouah I, Miranda EA, Lahtvee PJ. Xylose Metabolism and the Effect of Oxidative Stress on Lipid and Carotenoid Production in Rhodotorula toruloides: Insights for Future Biorefinery. Front Bioeng Biotechnol 2020; 8:1008. [PMID: 32974324 PMCID: PMC7466555 DOI: 10.3389/fbioe.2020.01008] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 07/31/2020] [Indexed: 12/04/2022] Open
Abstract
The use of cell factories to convert sugars from lignocellulosic biomass into chemicals in which oleochemicals and food additives, such as carotenoids, is essential for the shift toward sustainable processes. Rhodotorula toruloides is a yeast that naturally metabolises a wide range of substrates, including lignocellulosic hydrolysates, and converts them into lipids and carotenoids. In this study, xylose, the main component of hemicellulose, was used as the sole substrate for R. toruloides, and a detailed physiology characterisation combined with absolute proteomics and genome-scale metabolic models was carried out to understand the regulation of lipid and carotenoid production. To improve these productions, oxidative stress was induced by hydrogen peroxide and light irradiation and further enhanced by adaptive laboratory evolution. Based on the online measurements of growth and CO2 excretion, three distinct growth phases were identified during batch cultivations. Majority of the intracellular flux estimations showed similar trends with the measured protein levels and demonstrated improved NADPH regeneration, phosphoketolase activity and reduced β-oxidation, correlating with increasing lipid yields. Light irradiation resulted in 70% higher carotenoid and 40% higher lipid content compared to the optimal growth conditions. The presence of hydrogen peroxide did not affect the carotenoid production but culminated in the highest lipid content of 0.65 g/gDCW. The adapted strain showed improved fitness and 2.3-fold higher carotenoid content than the parental strain. This work presents a holistic view of xylose conversion into microbial oil and carotenoids by R. toruloides, in a process toward renewable and cost-effective production of these molecules.
Collapse
Affiliation(s)
- Marina Julio Pinheiro
- Institute of Technology, University of Tartu, Tartu, Estonia
- Department of Materials and Bioprocess Engineering, School of Chemical Engineering, University of Campinas, Campinas, Brazil
| | | | - Isma Belouah
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Everson Alves Miranda
- Department of Materials and Bioprocess Engineering, School of Chemical Engineering, University of Campinas, Campinas, Brazil
| | | |
Collapse
|
155
|
Walls JF, Subleski JJ, Palmieri EM, Gonzalez-Cotto M, Gardiner CM, McVicar DW, Finlay DK. Metabolic but not transcriptional regulation by PKM2 is important for natural killer cell responses. eLife 2020; 9:59166. [PMID: 32812866 PMCID: PMC7467725 DOI: 10.7554/elife.59166] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 08/15/2020] [Indexed: 12/25/2022] Open
Abstract
Natural Killer (NK) cells have an important role in immune responses to viruses and tumours. Integrating changes in signal transduction pathways and cellular metabolism is essential for effective NK cells responses. The glycolytic enzyme Pyruvate Kinase Muscle 2 (PKM2) has described roles in regulating glycolytic flux and signal transduction, particularly gene transcription. While PKM2 expression is robustly induced in activated NK cells, mice lacking PKM2 in NK cells showed no defect in NK cell metabolism, transcription or antiviral responses to MCMV infection. NK cell metabolism was maintained due to compensatory PKM1 expression in PKM2-null NK cells. To further investigate the role of PKM2, we used TEPP-46, which increases PKM2 catalytic activity while inhibiting any PKM2 signalling functions. NK cells activated with TEPP-46 had reduced effector function due to TEPP-46-induced increases in oxidative stress. Overall, PKM2-regulated glycolytic metabolism and redox status, not transcriptional control, facilitate optimal NK cells responses.
Collapse
Affiliation(s)
- Jessica F Walls
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,Laboratory of Cancer Immunometabolism, National Cancer Institute, Frederick, United States
| | - Jeff J Subleski
- Laboratory of Cancer Immunometabolism, National Cancer Institute, Frederick, United States
| | - Erika M Palmieri
- Laboratory of Cancer Immunometabolism, National Cancer Institute, Frederick, United States
| | - Marieli Gonzalez-Cotto
- Laboratory of Cancer Immunometabolism, National Cancer Institute, Frederick, United States
| | - Clair M Gardiner
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Daniel W McVicar
- Laboratory of Cancer Immunometabolism, National Cancer Institute, Frederick, United States
| | - David K Finlay
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,School of Pharmacy and Pharmaceutical Sciences, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
156
|
Hayes JD, Dinkova-Kostova AT, Tew KD. Oxidative Stress in Cancer. Cancer Cell 2020; 38:167-197. [PMID: 32649885 PMCID: PMC7439808 DOI: 10.1016/j.ccell.2020.06.001] [Citation(s) in RCA: 1241] [Impact Index Per Article: 310.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 04/29/2020] [Accepted: 05/29/2020] [Indexed: 12/13/2022]
Abstract
Contingent upon concentration, reactive oxygen species (ROS) influence cancer evolution in apparently contradictory ways, either initiating/stimulating tumorigenesis and supporting transformation/proliferation of cancer cells or causing cell death. To accommodate high ROS levels, tumor cells modify sulfur-based metabolism, NADPH generation, and the activity of antioxidant transcription factors. During initiation, genetic changes enable cell survival under high ROS levels by activating antioxidant transcription factors or increasing NADPH via the pentose phosphate pathway (PPP). During progression and metastasis, tumor cells adapt to oxidative stress by increasing NADPH in various ways, including activation of AMPK, the PPP, and reductive glutamine and folate metabolism.
Collapse
Affiliation(s)
- John D Hayes
- Division of Cellular Medicine, Jacqui Wood Cancer Centre, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK, Scotland.
| | - Albena T Dinkova-Kostova
- Division of Cellular Medicine, Jacqui Wood Cancer Centre, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK, Scotland; Department of Pharmacology and Molecular Sciences and Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Kenneth D Tew
- Department of Cell and Molecular Pharmacology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
157
|
Yang HC, Stern A, Chiu DTY. G6PD: A hub for metabolic reprogramming and redox signaling in cancer. Biomed J 2020; 44:285-292. [PMID: 33097441 PMCID: PMC8358196 DOI: 10.1016/j.bj.2020.08.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 06/11/2020] [Accepted: 08/02/2020] [Indexed: 12/12/2022] Open
Abstract
Metabolic hubs play a major role in the initiation and development of cancer. Oncogenic signaling pathways drive metabolic reprogramming and alter redox homeostasis. G6PD has potential oncogenic activity and it plays a pivotal role in cell proliferation, survival and stress responses. Aberrant activation of G6PD via metabolic reprogramming alters NADPH levels, leading to an antioxidant or a pro-oxidant environment which can either enhance DNA oxidative damage and genomic instability or initiate oncogenic signaling. Nutrient deprivation can rewire metabolism, which leads to mutations that determine a cancer cell's fate. Deregulated G6PD status and oxidative stress form a vicious cycle, which paves the way for cancer progression. This review aims to update and focus the potential role of G6PD in metabolic reprogramming and redox signaling in cancer.
Collapse
Affiliation(s)
- Hung-Chi Yang
- Department of Medical Laboratory Science and Biotechnology, Yuanpei University of Medical Technology, Hsinchu, Taiwan.
| | - Arnold Stern
- New York University School of Medicine, New York, NY, USA
| | - Daniel Tsun-Yee Chiu
- Research Center for Chinese Herbal Medicine, Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan; Department of Pediatric Hematology/Oncology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.
| |
Collapse
|
158
|
Greene E, Cauble R, Dhamad AE, Kidd MT, Kong B, Howard SM, Castro HF, Campagna SR, Bedford M, Dridi S. Muscle Metabolome Profiles in Woody Breast-(un)Affected Broilers: Effects of Quantum Blue Phytase-Enriched Diet. Front Vet Sci 2020; 7:458. [PMID: 32851035 PMCID: PMC7417653 DOI: 10.3389/fvets.2020.00458] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/22/2020] [Indexed: 12/21/2022] Open
Abstract
Woody breast (WB) myopathy is significantly impacting modern broilers and is imposing a huge economic burden on the poultry industry worldwide. Yet, its etiology is not fully defined. In a previous study, we have shown that hypoxia and the activation of its upstream mediators (AKT/PI3K/mTOR) played a key role in WB myopathy, and supplementation of quantum blue (QB) can help to reduce WB severity via modulation of hypoxia-related pathways. To gain further insights, we undertook here a metabolomics approach to identify key metabolite signatures and outline their most enriched biological functions. Ultra performance liquid chromatography coupled with high resolution mass spectrometry (UPLC-HRMS) identified a total of 108 known metabolites. Of these, mean intensity differences at P < 0.05 were found in 60 metabolites with 42 higher and 18 lower in WB-affected compared to unaffected muscles. Multivariate analysis and Partial Least Squares Discriminant analysis (PLS-DA) scores plot displayed different clusters when comparing metabolites profile from affected and unaffected tissues and from moderate (MOD) and severe (SEV) WB muscles indicating that unique metabolite profiles are present for the WB-affected and unaffected muscles. To gain biologically related molecule networks, a stringent pathway analyses was conducted using IPA knowledge-base. The top 10 canonical pathways generated, using a fold-change -1.5 and 1.5 cutoff, with the 50 differentially abundant-metabolites were purine nucleotide degradation and de novo biosynthesis, sirtuin signaling pathway, citrulline-nitric oxide cycle, salvage pathways of pyrimidine DNA, IL-1 signaling, iNOS, Angiogenesis, PI3K/AKT signaling, and oxidative phosphorylation. The top altered bio-functions in term of molecular and cellular functions in WB-affected tissues included cellular development, cellular growth and proliferation, cellular death and survival, small molecular biochemistry, inflammatory response, free radical scavenging, cell signaling and cell-to-cell interaction, cell cycles, and lipid, carbohydrate, amino acid, and nucleic acid metabolisms. The top disorder functions identified were organismal injury and abnormalities, cancer, skeletal and muscular disorders, connective tissue disorders, and inflammatory diseases. Breast tissues from birds fed with high dose (2,000 FTU) of QB phytase exhibited 22 metabolites with significantly different levels compared to the control group with a clear cluster using PLS-DA analysis. Of these 22 metabolites, 9 were differentially abundant between WB-affected and unaffected muscles. Taken together, this study determined many metabolic signatures and disordered pathways, which could be regarded as new routes for discovering potential mechanisms of WB myopathy.
Collapse
Affiliation(s)
- Elizabeth Greene
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Reagan Cauble
- Department of Animal Sciences, University of Arkansas, Fayetteville, AR, United States
| | - Ahmed E Dhamad
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Michael T Kidd
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Byungwhi Kong
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Sara M Howard
- Biological and Small Molecule Mass Spectrometry Core, Department of Chemistry, University of Tennessee, Knoxville, Knoxville, TN, United States
| | - Hector F Castro
- Biological and Small Molecule Mass Spectrometry Core, Department of Chemistry, University of Tennessee, Knoxville, Knoxville, TN, United States
| | - Shawn R Campagna
- Biological and Small Molecule Mass Spectrometry Core, Department of Chemistry, University of Tennessee, Knoxville, Knoxville, TN, United States
| | | | - Sami Dridi
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
| |
Collapse
|
159
|
Margaritelis NV, Paschalis V, Theodorou AA, Kyparos A, Nikolaidis MG. Redox basis of exercise physiology. Redox Biol 2020; 35:101499. [PMID: 32192916 PMCID: PMC7284946 DOI: 10.1016/j.redox.2020.101499] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/20/2020] [Accepted: 03/05/2020] [Indexed: 12/15/2022] Open
Abstract
Redox reactions control fundamental processes of human biology. Therefore, it is safe to assume that the responses and adaptations to exercise are, at least in part, mediated by redox reactions. In this review, we are trying to show that redox reactions are the basis of exercise physiology by outlining the redox signaling pathways that regulate four characteristic acute exercise-induced responses (muscle contractile function, glucose uptake, blood flow and bioenergetics) and four chronic exercise-induced adaptations (mitochondrial biogenesis, muscle hypertrophy, angiogenesis and redox homeostasis). Based on our analysis, we argue that redox regulation should be acknowledged as central to exercise physiology.
Collapse
Affiliation(s)
- N V Margaritelis
- Department of Physical Education and Sport Science at Serres, Aristotle University of Thessaloniki, Thessaloniki, Greece; Dialysis Unit, 424 General Military Hospital of Thessaloniki, Thessaloniki, Greece.
| | - V Paschalis
- School of Physical Education and Sport Science, National and Kapodistrian University of Athens, Athens, Greece
| | - A A Theodorou
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus
| | - A Kyparos
- Department of Physical Education and Sport Science at Serres, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - M G Nikolaidis
- Department of Physical Education and Sport Science at Serres, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| |
Collapse
|
160
|
Wang T, Jian Z, Baskys A, Yang J, Li J, Guo H, Hei Y, Xian P, He Z, Li Z, Li N, Long Q. MSC-derived exosomes protect against oxidative stress-induced skin injury via adaptive regulation of the NRF2 defense system. Biomaterials 2020; 257:120264. [PMID: 32791387 DOI: 10.1016/j.biomaterials.2020.120264] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 06/27/2020] [Accepted: 07/26/2020] [Indexed: 12/21/2022]
Abstract
Oxidative stress is a major cause of skin injury induced by damaging stimuli such as UV radiation. Currently, owing to their immunomodulatory properties, mesenchymal stem cell-derived exosomes (MSC-Exo), as a nanotherapeutic agent, have attracted considerable attention. Here, we investigated the therapeutic effects of MSC-Exo on oxidative injury in H2O2-stimulated epidermal keratinocytes and UV-irradiated wild type and nuclear factor-erythroid 2-related factor-2 (Nrf2) knocked down cell and animal models. Our findings showed that MSC-Exo treatment reduced reactive oxygen species generation, DNA damage, aberrant calcium signaling, and mitochondrial changes in H2O2-stimulated keratinocytes or UV-irradiated mice skin. Exosome therapy also improved antioxidant capacities shown by increased ferric ion reducing antioxidant power and glutathione peroxidase or superoxide dismutase activities in oxidative stress-induced cell and skin injury. In addition, it alleviated cellular and histological responses to inflammation and oxidation in cell or animal models. Furthermore, the NRF2 signaling pathway was involved in the antioxidation activity of MSC-Exo, while Nrf2 knockdown attenuated the antioxidant capacities of MSC-Exo in vitro and in vivo, suggesting that these effects are partially mediated by the NRF2 signaling pathway. These results indicate that MSC-Exo can repair oxidative stress-induced skin injury via adaptive regulation of the NRF2 defense system. Thus, MSC-Exo may be used as a potential dermatological nanotherapeutic agent for treating oxidative stress-induced skin diseases or disorders.
Collapse
Affiliation(s)
- Tian Wang
- Mini-invasive Neurosurgery and Translational Medical Center, Xi'an Central Hospital, Xi'an Jiaotong University, No. 161, West 5th Road, Xincheng District, Xi'an, 710003, PR China
| | - Zhe Jian
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No.17 Changle West Road, Xi'an, 710032, PR China
| | - Andrius Baskys
- Mini-invasive Neurosurgery and Translational Medical Center, Xi'an Central Hospital, Xi'an Jiaotong University, No. 161, West 5th Road, Xincheng District, Xi'an, 710003, PR China; Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, 91766, USA
| | - Junle Yang
- Mini-invasive Neurosurgery and Translational Medical Center, Xi'an Central Hospital, Xi'an Jiaotong University, No. 161, West 5th Road, Xincheng District, Xi'an, 710003, PR China
| | - Jianying Li
- Mini-invasive Neurosurgery and Translational Medical Center, Xi'an Central Hospital, Xi'an Jiaotong University, No. 161, West 5th Road, Xincheng District, Xi'an, 710003, PR China
| | - Hua Guo
- Mini-invasive Neurosurgery and Translational Medical Center, Xi'an Central Hospital, Xi'an Jiaotong University, No. 161, West 5th Road, Xincheng District, Xi'an, 710003, PR China
| | - Yue Hei
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No.17 Changle West Road, Xi'an, 710032, PR China
| | - Panpan Xian
- Mini-invasive Neurosurgery and Translational Medical Center, Xi'an Central Hospital, Xi'an Jiaotong University, No. 161, West 5th Road, Xincheng District, Xi'an, 710003, PR China
| | - Zhongzheng He
- Mini-invasive Neurosurgery and Translational Medical Center, Xi'an Central Hospital, Xi'an Jiaotong University, No. 161, West 5th Road, Xincheng District, Xi'an, 710003, PR China
| | - Zhengyu Li
- Mini-invasive Neurosurgery and Translational Medical Center, Xi'an Central Hospital, Xi'an Jiaotong University, No. 161, West 5th Road, Xincheng District, Xi'an, 710003, PR China
| | - Namiao Li
- Mini-invasive Neurosurgery and Translational Medical Center, Xi'an Central Hospital, Xi'an Jiaotong University, No. 161, West 5th Road, Xincheng District, Xi'an, 710003, PR China
| | - Qianfa Long
- Mini-invasive Neurosurgery and Translational Medical Center, Xi'an Central Hospital, Xi'an Jiaotong University, No. 161, West 5th Road, Xincheng District, Xi'an, 710003, PR China.
| |
Collapse
|
161
|
Xu K, He Z, Chen M, Wang N, Zhang D, Yang L, Xu Z, Xu H. HIF-1α regulates cellular metabolism, and Imatinib resistance by targeting phosphogluconate dehydrogenase in gastrointestinal stromal tumors. Cell Death Dis 2020; 11:586. [PMID: 32719331 PMCID: PMC7385157 DOI: 10.1038/s41419-020-02768-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022]
Abstract
The pentose phosphate pathway (PPP) plays a critical role in maintaining cellular redox homeostasis in tumor cells and macromolecule biosynthesis. Upregulation of the PPP has been shown in several types of tumor. However, how the PPP is regulated to confer selective growth advantages on drug resistant tumor cells is not well understood. Here we show a metabolic shift from tricarboxylic acid cycle (TCA) to PPP after a long period induction of Imatinib (IM). One of the rate-limiting enzymes of the PPP-phosphogluconate dehydrogenase (PGD), is dramatically upregulated in gastrointestinal stromal tumors (GISTs) and GIST cell lines resistant to Imatinib (IM) compared with sensitive controls. Functional studies revealed that the overexpression of PGD in resistant GIST cell lines promoted cell proliferation and suppressed cell apoptosis. Mechanistic analyses suggested that the protein level of hypoxia inducible factor-1α (HIF-1α) increased during long time stimulation of reactive oxygen species (ROS) produced by IM. Importantly, we further demonstrated that HIF-1α also had positive correlation with PGD, resulting in the change of metabolic pathway, and ultimately causing drug resistance in GIST. Our findings show that long term use of IM alters the metabolic phenotype of GIST through ROS and HIF-1α, and this may contribute to IM resistance. Our work offers preclinical proof of metabolic target as an effective strategy for the treatment of drug resistance in GIST.
Collapse
Affiliation(s)
- Kangjing Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medical University, Nanjing, 211166, China
| | - Zhongyuan He
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medical University, Nanjing, 211166, China
| | - Ming Chen
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medical University, Nanjing, 211166, China
| | - Nuofan Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medical University, Nanjing, 211166, China
| | - Diancai Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medical University, Nanjing, 211166, China
| | - Li Yang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medical University, Nanjing, 211166, China
| | - Zekuan Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China. .,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medical University, Nanjing, 211166, China.
| | - Hao Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China. .,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medical University, Nanjing, 211166, China.
| |
Collapse
|
162
|
Stucki D, Steinhausen J, Westhoff P, Krahl H, Brilhaus D, Massenberg A, Weber APM, Reichert AS, Brenneisen P, Stahl W. Endogenous Carbon Monoxide Signaling Modulates Mitochondrial Function and Intracellular Glucose Utilization: Impact of the Heme Oxygenase Substrate Hemin. Antioxidants (Basel) 2020; 9:antiox9080652. [PMID: 32717801 PMCID: PMC7465082 DOI: 10.3390/antiox9080652] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/17/2020] [Accepted: 07/19/2020] [Indexed: 12/22/2022] Open
Abstract
Stress-inducible heme oxygenase-1 (HO-1) catalyzes the oxidative cleavage of heme yielding biliverdin, ferrous iron, and carbon monoxide (CO). Heme oxygenase activity has been attributed to antioxidant defense via the redox cycling system of biliverdin and bilirubin. There is increasing evidence that CO is a gaseous signaling molecule and plays a role in the regulation of energy metabolism. Inhibitory effects of CO on the respiratory chain are well established, but the implication of such a process on the cellular stress response is not well understood. By means of extracellular flux analyses and isotopic tracing, we studied the effects of CO, either released from the CO donor CORM-401 or endogenously produced by heme oxygenases, on the respiratory chain and glucose metabolism. CORM-401 was thereby used as a tool to mimic endogenous CO production by heme oxygenases. In the long term (>60 min), CORM-401-derived CO exposure inhibited mitochondrial respiration, which was compensated by increased glycolysis accompanied by a loss of the ATP production rate and an increase in proton leakage. This effect pattern was likewise observed after endogenous CO production by heme oxygenases. However, in the present setting, these effects were only observed when sufficient substrate for heme oxygenases (hemin) was provided. Modulation of the HO-1 protein level was less important. The long-term influence of CO on glucose metabolism via glycolysis was preceded by a short-term response (<30 min) of the cells to CO. Stable isotope-labeling experiments and metabolic flux analysis revealed a short-term shift of glucose consumption from glycolysis to the pentose phosphate pathway (PPP) along with an increase in reactive oxygen species (ROS) generation. Overall, we suggest that signaling by endogenous CO stimulates the rapid formation of reduction equivalents (NADPH) via the PPP, and plays an additional role in antioxidant defense, e.g., via feed-forward stimulation of the bilirubin/biliverdin redox cycling system.
Collapse
Affiliation(s)
- David Stucki
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University Düsseldorf, D-40001 Düsseldorf, Germany; (D.S.); (J.S.); (H.K.); (A.M.); (A.S.R.); (P.B.)
| | - Julia Steinhausen
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University Düsseldorf, D-40001 Düsseldorf, Germany; (D.S.); (J.S.); (H.K.); (A.M.); (A.S.R.); (P.B.)
| | - Philipp Westhoff
- Plant Metabolism and Metabolomics Laboratory, Cluster of Excellence on Plant Sciences (CEPLAS), Heinrich Heine University Düsseldorf, D-40001 Düsseldorf, Germany; (P.W.); (D.B.)
| | - Heide Krahl
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University Düsseldorf, D-40001 Düsseldorf, Germany; (D.S.); (J.S.); (H.K.); (A.M.); (A.S.R.); (P.B.)
| | - Dominik Brilhaus
- Plant Metabolism and Metabolomics Laboratory, Cluster of Excellence on Plant Sciences (CEPLAS), Heinrich Heine University Düsseldorf, D-40001 Düsseldorf, Germany; (P.W.); (D.B.)
| | - Annika Massenberg
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University Düsseldorf, D-40001 Düsseldorf, Germany; (D.S.); (J.S.); (H.K.); (A.M.); (A.S.R.); (P.B.)
| | - Andreas P. M. Weber
- Institute of Plant Biochemistry, Cluster of Excellence on Plant Sciences (CEPLAS), Heinrich Heine University Düsseldorf, D-40001 Düsseldorf, Germany;
| | - Andreas S. Reichert
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University Düsseldorf, D-40001 Düsseldorf, Germany; (D.S.); (J.S.); (H.K.); (A.M.); (A.S.R.); (P.B.)
| | - Peter Brenneisen
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University Düsseldorf, D-40001 Düsseldorf, Germany; (D.S.); (J.S.); (H.K.); (A.M.); (A.S.R.); (P.B.)
| | - Wilhelm Stahl
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University Düsseldorf, D-40001 Düsseldorf, Germany; (D.S.); (J.S.); (H.K.); (A.M.); (A.S.R.); (P.B.)
- Correspondence: ; Tel.: +49-211-811-2711
| |
Collapse
|
163
|
Imaging of metabolic activity adaptations to UV stress, drugs and differentiation at cellular resolution in skin and skin equivalents - Implications for oxidative UV damage. Redox Biol 2020; 37:101583. [PMID: 32713735 PMCID: PMC7767734 DOI: 10.1016/j.redox.2020.101583] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 05/12/2020] [Accepted: 05/13/2020] [Indexed: 12/29/2022] Open
Abstract
The epidermis is a multi-layered epithelium that consists mainly of keratinocytes which proliferate in its basal layer and then differentiate to form the stratum corneum, the skin's ultimate barrier to the environment. During differentiation keratinocyte function, chemical composition, physical properties, metabolism and secretion are profoundly changed. Extrinsic or intrinsic stressors, like ultraviolet (UV) radiation thus may differently affect the epidermal keratinocytes, depending on differentiation stage. Exposure to UV elicits the DNA damage responses, activation of pathways which detoxify or repair damage or induction of programmed cell death when the damage was irreparable. Recently, rapid diversion of glucose flux into the pentose phosphate pathway (PPP) was discovered as additional mechanism by which cells rapidly generate reduction equivalents and precursors for nucleotides - both being in demand after UV damage. There is however little known about the correlation of such metabolic activity with differentiation state, cell damage and tissue localization of epidermal cells. We developed a method to correlate the activity of G6PD, the first and rate-limiting enzyme of this metabolic UV response, at cellular resolution to cell type, differentiation state, and cell damage in human skin and in organotypic reconstructed epidermis. We thereby could verify rapid activation of G6PD as an immediate UVB response not only in basal but also in differentiating epidermal keratinocytes and found increased activity in cells which initiated DNA damage responses. When keratinocytes had been UVB irradiated before organotypic culture, their distribution within the skin equivalent was abnormal and the G6PD activity was reduced compared to neighboring cells. Finally, we found that the anti-diabetic and potential anti-aging drug metformin strongly induced G6PD activity throughout reconstructed epidermis. Activation of the protective pentose phosphate pathway may be useful to enhance the skin's antioxidant defense systems and DNA damage repair capacity on demand.
Collapse
|
164
|
Grove RA, Madhavan D, Boone CHT, Braga CP, Papackova Z, Kyllo H, Samson K, Simeone K, Simeone T, Helikar T, Hanson CK, Adamec J. Aberrant energy metabolism and redox balance in seizure onset zones of epileptic patients. J Proteomics 2020; 223:103812. [PMID: 32418907 PMCID: PMC10588813 DOI: 10.1016/j.jprot.2020.103812] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 04/13/2020] [Accepted: 05/05/2020] [Indexed: 12/16/2022]
Abstract
Epilepsy is a disorder that affects around 1% of the population. Approximately one third of patients do not respond to anti-convulsant drugs treatment. To understand the underlying biological processes involved in drug resistant epilepsy (DRE), a combination of proteomics strategies was used to compare molecular differences and enzymatic activities in tissue implicated in seizure onset to tissue with no abnormal activity within patients. Label free quantitation identified 17 proteins with altered abundance in the seizure onset zone as compared to tissue with normal activity. Assessment of oxidative protein damage by protein carbonylation identified additional 11 proteins with potentially altered function in the seizure onset zone. Pathway analysis revealed that most of the affected proteins are involved in energy metabolism and redox balance. Further, enzymatic assays showed significantly decreased activity of transketolase indicating a disruption of the Pentose Phosphate Pathway and diversion of intermediates into purine metabolic pathway, resulting in the generation of the potentially pro-convulsant metabolites. Altogether, these findings suggest that imbalance in energy metabolism and redox balance, pathways critical to proper neuronal function, play important roles in neuronal network hyperexcitability and can be used as a primary target for potential therapeutic strategies to combat DRE. SIGNIFICANCE: Epileptic seizures are some of the most difficult to treat neurological disorders. Up to 40% of patients with epilepsy are resistant to first- and second-line anticonvulsant therapy, a condition that has been classified as refractory epilepsy. One potential therapy for this patient population is the ketogenic diet (KD), which has been proven effective against multiple refractory seizure types However, compliance with the KD is extremely difficult, and carries severe risks, including ketoacidosis, renal failure, and dangerous electrolyte imbalances. Therefore, identification of pathways disruptions or shortages can potentially uncover cellular targets for anticonvulsants, leading to a personalized treatment approach depending on a patient's individual metabolic signature.
Collapse
Affiliation(s)
- Ryan A Grove
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, United States of America
| | - Deepak Madhavan
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States of America
| | - Cory H T Boone
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, United States of America
| | - Camila Pereira Braga
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, United States of America
| | - Zuzana Papackova
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, CZ, Czech Republic; Czech University of Life Science Prague, Faculty of Agrobiology-Food and Natural Recourses, Department of Veterinary Science, Prague, CZ, Czech Republic
| | - Hannah Kyllo
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States of America
| | - Kaeli Samson
- Department of Pharmacology, Creighton University School of Medicine, Omaha, NE 68178, United States of America
| | - Kristina Simeone
- Department of Pharmacology, Creighton University School of Medicine, Omaha, NE 68178, United States of America
| | - Timothy Simeone
- Department of Pharmacology, Creighton University School of Medicine, Omaha, NE 68178, United States of America
| | - Tomas Helikar
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, United States of America
| | - Corrine K Hanson
- College of Allied Health Professions, University of Nebraska Medical Center, Omaha, NE, 68198, United States of America
| | - Jiri Adamec
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, United States of America.
| |
Collapse
|
165
|
Johnson LA. APOE and metabolic dysfunction in Alzheimer's disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 154:131-151. [PMID: 32739002 DOI: 10.1016/bs.irn.2020.02.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The strongest genetic risk factor for sporadic Alzheimer's disease (AD) is carriage of the E4 allele of APOE. Metabolic dysfunction also increases risk of dementia and AD. Facing a need for effective therapies and an aging global population, studies aimed at uncovering new therapeutic targets for AD have become critical. Insight into the biology underlying the effects of E4 and metabolic impairment on the brain may lead to novel therapies to reduce AD risk. An understudied hallmark of both AD patients and E4 individuals is a common metabolic impairment-cerebral glucose hypometabolism. This is a robust and replicated finding in humans, and begins decades prior to cognitive decline. Possession of E4 also appears to alter several other aspects of cerebral glucose metabolism, fatty acid metabolism, and management of oxidative stress through the pentose phosphate pathway. A critical knowledge gap in AD is the mechanism by which APOE alters cerebral metabolism and clarification as to its relevance to AD risk. Facing a need for effective therapies, studies aimed at uncovering new therapeutic targets have become critical. One such approach is to gain a better understanding of the metabolic mechanisms that may underlie E4-associated cognitive dysfunction and AD risk.
Collapse
Affiliation(s)
- Lance A Johnson
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY, United States; Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY, United States.
| |
Collapse
|
166
|
Dan X, Babbar M, Moore A, Wechter N, Tian J, Mohanty J, Croteau DL, Bohr VA. DNA damage invokes mitophagy through a pathway involving Spata18. Nucleic Acids Res 2020; 48:6611-6623. [PMID: 32453416 PMCID: PMC7337932 DOI: 10.1093/nar/gkaa393] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 04/30/2020] [Accepted: 05/04/2020] [Indexed: 12/31/2022] Open
Abstract
Mitochondria are vital for cellular energy supply and intracellular signaling after stress. Here, we aimed to investigate how mitochondria respond to acute DNA damage with respect to mitophagy, which is an important mitochondrial quality control process. Our results show that mitophagy increases after DNA damage in primary fibroblasts, murine neurons and Caenorhabditis elegans neurons. Our results indicate that modulation of mitophagy after DNA damage is independent of the type of DNA damage stimuli used and that the protein Spata18 is an important player in this process. Knockdown of Spata18 suppresses mitophagy, disturbs mitochondrial Ca2+ homeostasis, affects ATP production, and attenuates DNA repair. Importantly, mitophagy after DNA damage is a vital cellular response to maintain mitochondrial functions and DNA repair.
Collapse
Affiliation(s)
- Xiuli Dan
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Mansi Babbar
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Anthony Moore
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Noah Wechter
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Jingyan Tian
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Joy G Mohanty
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Deborah L Croteau
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Vilhelm A Bohr
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
- Danish Center for Healthy Aging, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
167
|
Li Z, Hou M, Qiu Y, Zhao B, Nie H, Su S. Changes in Antioxidant Enzymes Activity and Metabolomic Profiles in the Guts of Honey Bee ( Apis mellifera) Larvae Infected with Ascosphaera apis. INSECTS 2020; 11:insects11070419. [PMID: 32640515 PMCID: PMC7412215 DOI: 10.3390/insects11070419] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/03/2020] [Accepted: 07/03/2020] [Indexed: 01/26/2023]
Abstract
The fungus Ascosphaera apis, an obligate fungal pathogen of honey bee brood, causes chalkbrood disease in honey bee larvae worldwide. Biological characteristics of the fungal pathogen and the molecular interactions between A. apis and honey bees have been studied extensively. However, little is known about the effects of A. apis infection on antioxidant enzyme activities and metabolic profiles of the gut of honey bee larvae. In this study, sandwich enzyme-linked immunosorbent assay and LC-MS based untargeted metabolomic analysis were employed to determine the changes in the specific activities of antioxidant enzymes and the metabolomic profiles in gut tissues of A. apis-infected larvae (105 A. apis spores per larva) and controls. Results showed that specific activities of superoxide dismutase, catalase and glutathione S-transferase were significantly higher in the guts of the control larvae than in the guts of the A. apis-infected larvae. The metabolomic data revealed that levels of 28 and 52 metabolites were significantly higher and lower, respectively, in the guts of A. apis-infected larvae than in the guts of control larvae. The 5-oxo-ETE level in the infected larvae was two times higher than that in the control larvae. Elevated 5-oxo-ETE levels may act as a potential metabolic biomarker for chalkbrood disease diagnosis, suggesting that A. apis infection induced obvious oxidative stress in the honey bee larvae. The levels of metabolites such as taurine, docosahexaenoic acid, and L-carnitine involved in combating oxidative stress were significantly decreased in the gut of A. apis-infected larvae. Overall, our results suggest that A. apis infection may compromise the ability of infected larvae to cope with oxidative stress, providing new insight into changing patterns of physiological responses to A. apis infection in honey bee larvae by concurrent use of conventional biochemical assays and untargeted metabolomics.
Collapse
|
168
|
Zhang N, Zhang Z, He R, Li H, Ding S. GLAST-CreER T2 mediated deletion of GDNF increases brain damage and exacerbates long-term stroke outcomes after focal ischemic stroke in mouse model. Glia 2020; 68:2395-2414. [PMID: 32497340 DOI: 10.1002/glia.23848] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 05/07/2020] [Accepted: 05/07/2020] [Indexed: 01/01/2023]
Abstract
Focal ischemic stroke (FIS) is a leading cause of human death. Glial scar formation largely caused by reactive astrogliosis in peri-infarct region (PIR) is the hallmark of FIS. Glial cell-derived neurotrophic factor (GDNF) was originally isolated from a rat glioma cell-line supernatant and is a potent survival neurotrophic factor. Here, using CreERT2 -LoxP recombination technology, we generated inducible and astrocyte-specific GDNF conditional knockout (cKO), that is, GLAST-GDNF-/- cKO mice to investigate the effect of reactive astrocytes (RAs)-derived GDNF on neuronal death, brain damage, oxidative stress and motor function recovery after photothrombosis (PT)-induced FIS. Under non-ischemic conditions, we found that adult GLAST-GDNF-/- cKO mice exhibited significant lower numbers of Brdu+, Ki67+ cells, and DCX+ cells in the dentate gyrus (DG) in hippocampus than GDNF floxed (GDNFf/f ) control (Ctrl) mice, indicating endogenous astrocytic GDNF can promote adult neurogenesis. Under ischemic conditions, GLAST-GDNF-/- cKO mice had a significant increase in infarct volume, hippocampal damage and FJB+ degenerating neurons after PT as compared with the Ctrl mice. GLAST-GDNF-/- cKO mice also had lower densities of Brdu+ and Ki67+ cells in the PIR and exhibited larger behavioral deficits than the Ctrl mice. Mechanistically, GDNF deficiency in astrocytes increased oxidative stress through the downregulation of glucose-6-phosphate dehydrogenase (G6PD) in RAs. In summary, our study indicates that RAs-derived endogenous GDNF plays important roles in reducing brain damage and promoting brain recovery after FIS through neural regeneration and suggests that promoting anti-oxidant mechanism in RAs is a potential strategy in stroke therapy.
Collapse
Affiliation(s)
- Nannan Zhang
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| | - Zhe Zhang
- Department of Biomedical, Biological and Chemical Engineering, University of Missouri, Columbia, Missouri, USA
| | - Rui He
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| | - Hailong Li
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA.,Department of Biomedical, Biological and Chemical Engineering, University of Missouri, Columbia, Missouri, USA
| | - Shinghua Ding
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA.,Department of Biomedical, Biological and Chemical Engineering, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
169
|
A Translational In Vivo and In Vitro Metabolomic Study Reveals Altered Metabolic Pathways in Red Blood Cells of Type 2 Diabetes. J Clin Med 2020; 9:jcm9061619. [PMID: 32471219 PMCID: PMC7355709 DOI: 10.3390/jcm9061619] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/19/2020] [Accepted: 05/24/2020] [Indexed: 12/21/2022] Open
Abstract
Clinical parameters used in type 2 diabetes mellitus (T2D) diagnosis and monitoring such as glycosylated haemoglobin (HbA1c) are often unable to capture important information related to diabetic control and chronic complications. In order to search for additional biomarkers, we performed a pilot study comparing T2D patients with healthy controls matched by age, gender, and weight. By using 1H-nuclear magnetic resonance (NMR) based metabolomics profiling of red blood cells (RBCs), we found that the metabolic signature of RBCs in T2D subjects differed significantly from non-diabetic controls. Affected metabolites included glutathione, 2,3-bisphophoglycerate, inosinic acid, lactate, 6-phosphogluconate, creatine and adenosine triphosphate (ATP) and several amino acids such as leucine, glycine, alanine, lysine, aspartate, phenylalanine and tyrosine. These results were validated by an independent cohort of T2D and control patients. An analysis of the pathways in which these metabolites were involved showed that energetic and redox metabolism in RBCs were altered in T2D, as well as metabolites transported by RBCs. Taken together, our results revealed that the metabolic profile of RBCs can discriminate healthy controls from T2D patients. Further research is needed to determine whether metabolic fingerprint in RBC could be useful to complement the information obtained from HbA1c and glycemic variability as well as its potential role in the diabetes management.
Collapse
|
170
|
Dikova V, Vorhauser J, Geng A, Pelster B, Sandbichler AM. Metabolic interaction of hydrogen peroxide and hypoxia in zebrafish fibroblasts. Free Radic Biol Med 2020; 152:469-481. [PMID: 31740229 DOI: 10.1016/j.freeradbiomed.2019.11.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 11/12/2019] [Accepted: 11/12/2019] [Indexed: 11/26/2022]
Abstract
Cells require oxygen for aerobic metabolism, which may also result in the production of reactive oxygen species (ROS) as a by-product. Under low oxygen conditions, ROS formation has been reported to either increase or decrease. We addressed this physiological response for the first time in zebrafish embryonic fibroblasts (Z3) and used a hydrogen peroxide (H2O2)-specific fluorescent protein (roGFP2-Orp1) either targeted to the mitochondria or expressed in the cytosol. Microfluidic live-cell imaging measurements showed that oxygen deprivation in Z3 cells results in decreased or stable H2O2 levels within the mitochondria or the cytosol, respectively, and that the reductive shift recorded in the mitochondrial matrix is directly dependent on oxygen concentration. The response was accompanied by a transient increase in extracellular acidification rate (ECAR) and a lower cellular reducing potential as assessed by the viability stain alamarBlue. Complex I and III inhibition with Rotenone and Antimycin A led to H2O2 production under normoxia but these inhibitors were not able to avert the reductive shift under hypoxia. Only by system-wide inhibition of flavin-containing oxidases with Diphenyleneiodonium (DPI) were we able to decrease the reductive shift, while selective inhibition of NADPH oxidases with the inhibitor Apocynin had no effect on the hypoxia response. Since DPI also led to a strong increase in ECAR we found that, in order to keep the cytosolic H2O2 levels stable, glycolytic metabolism was of fundamental importance. According to our experiments with the glucose-6-phosphate dehydrogenase inhibitor 6-Aminonicotinamide, this was attributable to the pentose phosphate pathway producing reducing equivalents required for ROS degradation.
Collapse
Affiliation(s)
- Valentina Dikova
- Institute of Zoology and Center for Molecular Biosciences (CMBI), University of Innsbruck, Austria
| | - Julia Vorhauser
- Institute of Zoology and Center for Molecular Biosciences (CMBI), University of Innsbruck, Austria
| | - Anne Geng
- Institute of Zoology and Center for Molecular Biosciences (CMBI), University of Innsbruck, Austria
| | - Bernd Pelster
- Institute of Zoology and Center for Molecular Biosciences (CMBI), University of Innsbruck, Austria
| | | |
Collapse
|
171
|
TRIM21 and PHLDA3 negatively regulate the crosstalk between the PI3K/AKT pathway and PPP metabolism. Nat Commun 2020; 11:1880. [PMID: 32312982 PMCID: PMC7170963 DOI: 10.1038/s41467-020-15819-3] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 03/30/2020] [Indexed: 12/13/2022] Open
Abstract
PI3K/AKT signaling is known to regulate cancer metabolism, but whether metabolic feedback regulates the PI3K/AKT pathway is unclear. Here, we demonstrate the important reciprocal crosstalk between the PI3K/AKT signal and pentose phosphate pathway (PPP) branching metabolic pathways. PI3K/AKT activation stabilizes G6PD, the rate-limiting enzyme of the PPP, by inhibiting the newly identified E3 ligase TIRM21 and promotes the PPP. PPP metabolites, in turn, reinforce AKT activation and further promote cancer metabolic reprogramming by blocking the expression of the AKT inhibitor PHLDA3. Knockout of TRIM21 or PHLDA3 promotes crosstalk and cell proliferation. Importantly, PTEN null human cancer cells and in vivo murine models are sensitive to anti-PPP treatments, suggesting the importance of the PPP in maintaining AKT activation even in the presence of a constitutively activated PI3K pathway. Our study suggests that blockade of this reciprocal crosstalk mechanism may have a therapeutic benefit for cancers with PTEN loss or PI3K/AKT activation.
Collapse
|
172
|
Investigation of Heterologously Expressed Glucose-6-Phosphate Dehydrogenase Genes in a Yeast zwf1 Deletion. Microorganisms 2020; 8:microorganisms8040546. [PMID: 32283834 PMCID: PMC7232176 DOI: 10.3390/microorganisms8040546] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/03/2020] [Accepted: 04/08/2020] [Indexed: 12/31/2022] Open
Abstract
Glucose-6-phosphate dehydrogenase (G6PD) is a key enzyme of the oxidative part of the pentose phosphate pathway and serves as the major source of NADPH for metabolic reactions and oxidative stress response in pro- and eukaryotic cells. We here report on a strain of the model yeast Saccharomyces cerevisiae which lacks the G6PD-encoding ZWF1 gene and displays distinct growth retardation on rich and synthetic media, as well as a strongly reduced chronological lifespan. This strain was used as a recipient to introduce plasmid-encoded heterologous G6PD genes, synthesized in the yeast codon usage and expressed under the control of the native PFK2 promotor. Complementation of the hypersensitivity of the zwf1 mutant towards hydrogen peroxide to different degrees was observed for the genes from humans (HsG6PD1), the milk yeast Kluyveromyces lactis (KlZWF1), the bacteria Escherichia coli (EcZWF1) and Leuconostoc mesenteroides (LmZWF1), as well as the genes encoding three different plant G6PD isoforms from Arabidopsis thaliana (AtG6PD1, AtG6PD5, AtG6PD6). The plastidic AtG6PD1 isoform retained its redox-sensitive activity when produced in the yeast as a cytosolic enzyme, demonstrating the suitability of this host for determination of its physiological properties. Mutations precluding the formation of a disulfide bridge in AtG6PD1 abolished its redox-sensitivity but improved its capacity to complement the yeast zwf1 deletion. Given the importance of G6PD in human diseases and plant growth, this heterologous expression system offers a broad range of applications.
Collapse
|
173
|
Sundararaghavan A, Mukherjee A, Sahoo S, Suraishkumar GK. Mechanism of the oxidative stress‐mediated increase in lipid accumulation by the bacterium,R. opacusPD630: Experimental analysis and genome‐scale metabolic modeling. Biotechnol Bioeng 2020; 117:1779-1788. [DOI: 10.1002/bit.27330] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 02/22/2020] [Accepted: 03/09/2020] [Indexed: 12/13/2022]
Affiliation(s)
- Archanaa Sundararaghavan
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences buildingIndian Institute of Technology Madras Chennai India
| | | | - Swagatika Sahoo
- Department of Chemical Engineering and Initiative for Biological Systems EngineeringIndian Institute of Technology Madras Chennai India
| | - G. K. Suraishkumar
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences buildingIndian Institute of Technology Madras Chennai India
| |
Collapse
|
174
|
Held JM. Redox Systems Biology: Harnessing the Sentinels of the Cysteine Redoxome. Antioxid Redox Signal 2020; 32:659-676. [PMID: 31368359 PMCID: PMC7047077 DOI: 10.1089/ars.2019.7725] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 07/30/2019] [Accepted: 07/30/2019] [Indexed: 12/16/2022]
Abstract
Significance: Cellular redox processes are highly interconnected, yet not in equilibrium, and governed by a wide range of biochemical parameters. Technological advances continue refining how specific redox processes are regulated, but broad understanding of the dynamic interconnectivity between cellular redox modules remains limited. Systems biology investigates multiple components in complex environments and can provide integrative insights into the multifaceted cellular redox state. This review describes the state of the art in redox systems biology as well as provides an updated perspective and practical guide for harnessing thousands of cysteine sensors in the redoxome for multiparameter characterization of cellular redox networks. Recent Advances: Redox systems biology has been applied to genome-scale models and large public datasets, challenged common conceptions, and provided new insights that complement reductionist approaches. Advances in public knowledge and user-friendly tools for proteome-wide annotation of cysteine sentinels can now leverage cysteine redox proteomics datasets to provide spatial, functional, and protein structural information. Critical Issues: Careful consideration of available analytical approaches is needed to broadly characterize the systems-level properties of redox signaling networks and be experimentally feasible. The cysteine redoxome is an informative focal point since it integrates many aspects of redox biology. The mechanisms and redox modules governing cysteine redox regulation, cysteine oxidation assays, proteome-wide annotation of the biophysical and biochemical properties of individual cysteines, and their clinical application are discussed. Future Directions: Investigating the cysteine redoxome at a systems level will uncover new insights into the mechanisms of selectivity and context dependence of redox signaling networks.
Collapse
Affiliation(s)
- Jason M. Held
- Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri
- Department of Anesthesiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri
- Siteman Cancer Center, Washington University School of Medicine in St. Louis, St. Louis, Missouri
| |
Collapse
|
175
|
Vasconcelos e Sá J, Simão D, Terrasso AP, Silva MM, Brito C, Isidro IA, Alves PM, Carrondo MJT. Unveiling dynamic metabolic signatures in human induced pluripotent and neural stem cells. PLoS Comput Biol 2020; 16:e1007780. [PMID: 32298259 PMCID: PMC7188302 DOI: 10.1371/journal.pcbi.1007780] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 04/28/2020] [Accepted: 03/08/2020] [Indexed: 11/19/2022] Open
Abstract
Metabolism plays an essential role in cell fate decisions. However, the methods used for metabolic characterization and for finding potential metabolic regulators are still based on characterizing cellular metabolic steady-state which is dependent on the extracellular environment. In this work, we hypothesized that the response dynamics of intracellular metabolic pools to extracellular stimuli is controlled in a cell type-specific manner. We applied principles of process dynamics and control to human induced pluripotent stem cells (hiPSC) and human neural stem cells (hNSC) subjected to a sudden extracellular glutamine step. The fold-changes of steady-states and the transient profiles of metabolic pools revealed that dynamic responses were reproducible and cell type-specific. Importantly, many amino acids had conserved dynamics and readjusted their steady state concentration in response to the increased glutamine influx. Overall, we propose a novel methodology for systematic metabolic characterization and identification of potential metabolic regulators.
Collapse
Affiliation(s)
- João Vasconcelos e Sá
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Daniel Simão
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Ana P. Terrasso
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Marta M. Silva
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Catarina Brito
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Inês A. Isidro
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Paula M. Alves
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Manuel J. T. Carrondo
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| |
Collapse
|
176
|
Milanese C, Mastroberardino PG. A perspective on DNA damage-induced potentiation of the pentose phosphate shunt and reductive stress in chemoresistance. Mol Cell Oncol 2020; 7:1733383. [PMID: 32391425 PMCID: PMC7199736 DOI: 10.1080/23723556.2020.1733383] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 02/07/2020] [Accepted: 02/10/2020] [Indexed: 02/06/2023]
Abstract
Metabolic rearrangements and genome instability are two hallmarks of cancer. Recent evidence from our laboratory demonstrates that persistent DNA lesions hampering transcription may cause glucose rerouting through the pentose phosphate shunt and reductive stress. Here, we highlight the relevance of these findings for cancer and chemoresistance development.
Collapse
Affiliation(s)
- Chiara Milanese
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Pier G Mastroberardino
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, Netherlands.,Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
177
|
Guo J, Zhang Q, Su Y, Lu X, Wang Y, Yin M, Hu W, Wen W, Lei QY. Arginine methylation of ribose-5-phosphate isomerase A senses glucose to promote human colorectal cancer cell survival. SCIENCE CHINA-LIFE SCIENCES 2020; 63:1394-1405. [PMID: 32157557 DOI: 10.1007/s11427-019-1562-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 10/15/2019] [Indexed: 10/24/2022]
Abstract
Cancer cells remodel their metabolic network to adapt to variable nutrient availability. Pentose phosphate pathway (PPP) plays protective and biosynthetic roles by oxidizing glucose to generate reducing power and ribose. How cancer cells modulate PPP activity in response to glucose supply remains unclear. Here we show that ribose-5-phosphate isomerase A (RPIA), an enzyme in PPP, directly interacts with co-activator associated arginine methyltransferase 1 (CARM1) and is methylated at arginine 42 (R42). R42 methylation up-regulates the catalytic activity of RPIA. Furthermore, glucose deprivation strengthens the binding of CARM1 with RPIA to induce R42 hypermethylation. Insufficient glucose supply links to RPIA hypermethylation at R42, which increases oxidative PPP flux. RPIA methylation supports ROS clearance by enhancing NADPH production and fuels nucleic acid synthesis by increasing ribose supply. Importantly, RPIA methylation at R42 significantly potentiates colorectal cancer cell survival under glucose starvation. Collectively, RPIA methylation connects glucose availability to nucleotide synthesis and redox homeostasis.
Collapse
Affiliation(s)
- Jizheng Guo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Qixiang Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ying Su
- Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xiaochen Lu
- Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yiping Wang
- Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Miao Yin
- Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Weiguo Hu
- Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Wenyu Wen
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Qun-Ying Lei
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,State Key Laboratory of Medical Neurobiology Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
178
|
The Two-Species Model of transketolase explains donor substrate-binding, inhibition and heat-activation. Sci Rep 2020; 10:4148. [PMID: 32139871 PMCID: PMC7057962 DOI: 10.1038/s41598-020-61175-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 02/22/2020] [Indexed: 11/23/2022] Open
Abstract
We recently characterised a low-activity form of E. coli transketolase, TKlow, which also binds the cofactor thiamine pyrophosphate (TPP) with an affinity up to two-orders of magnitude lower than the previously known high TPP-affinity and high-activity form, TKhigh, in the presence of Mg2+. We observed previously that partial oxidation was responsible for increased TKhigh activity, while low-activity TKlow was unmodified. In the present study, the fluorescence-based cofactor-binding assay was adapted to detect binding of the β-hydroxypyruvate (HPA) donor substrate to wild-type transketolase and a variant, S385Y/D469T/R520Q, that is active towards aromatic aldehydes. Transketolase HPA affinity again revealed the two distinct forms of transketolase at a TKhigh:TKlow ratio that matched those observed previously via TPP binding to each variant. The HPA dissociation constant of TKlow was comparable to the substrate-inhibition dissociation constant, KiHPA, determined previously. We provide evidence that KiHPA is a convolution of binding to the low-activity TKlow-TKlow dimer, and the TKlow subunit of the partially-active TKhigh-TKlow mixed dimer, where HPA binding to the TKlow subunit of the mixed dimer results in inhibition of the active TKhigh subunit. Heat-activation of transketolase was similarly investigated and found to convert the TKlow subunit of the mixed dimer to have TKhigh-like properties, but without oxidation.
Collapse
|
179
|
Hamon MP, Gergondey R, L'honoré A, Friguet B. Mitochondrial Lon protease - depleted HeLa cells exhibit proteome modifications related to protein quality control, stress response and energy metabolism. Free Radic Biol Med 2020; 148:83-95. [PMID: 31904544 DOI: 10.1016/j.freeradbiomed.2019.12.039] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 12/23/2019] [Accepted: 12/24/2019] [Indexed: 12/20/2022]
Abstract
The ATP-dependent Lon protease is located in the mitochondrial matrix and oxidized proteins are among its primary targets for their degradation. Impairment of mitochondrial morphology and function together with apoptosis were observed in lung fibroblasts depleted for Lon expression while accumulation of carbonylated mitochondrial proteins has been reported for yeast and HeLa Lon deficient cells. In addition, age-related mitochondrial dysfunction has been associated with an impairment of Lon expression. Using a HeLa cell line stably transfected with an inducible shRNA directed against Lon, we have previously observed that Lon depletion results in a mild phenotype characterized by an increase of both production of reactive oxygen species and level of oxidized proteins (Bayot et al., 2014, Biochimie, 100: 38-47). In this study using the same cell line, we now show that Lon knockdown leads to modifications of the expression of a number of specific proteins involved in protein quality control, stress response and energy metabolism, as evidenced using a 2D gel-based proteomic approach, and to alteration of the mitochondrial network morphology. We also show that these effects are associated with decreased proliferation and can be modulated by culture conditions in galactose versus glucose containing medium.
Collapse
Affiliation(s)
- Marie-Paule Hamon
- Sorbonne Université, CNRS, INSERM, Institut de Biologie Paris-Seine, Biological Adaptation and Aging, B2A-IBPS, F-75005, Paris, France
| | - Rachel Gergondey
- Sorbonne Université, CNRS, INSERM, Institut de Biologie Paris-Seine, Biological Adaptation and Aging, B2A-IBPS, F-75005, Paris, France
| | - Aurore L'honoré
- Sorbonne Université, CNRS, INSERM, Institut de Biologie Paris-Seine, Biological Adaptation and Aging, B2A-IBPS, F-75005, Paris, France
| | - Bertrand Friguet
- Sorbonne Université, CNRS, INSERM, Institut de Biologie Paris-Seine, Biological Adaptation and Aging, B2A-IBPS, F-75005, Paris, France.
| |
Collapse
|
180
|
Sisakht M, Darabian M, Mahmoodzadeh A, Bazi A, Shafiee SM, Mokarram P, Khoshdel Z. The role of radiation induced oxidative stress as a regulator of radio-adaptive responses. Int J Radiat Biol 2020; 96:561-576. [PMID: 31976798 DOI: 10.1080/09553002.2020.1721597] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Purpose: Various sources of radiation including radiofrequency, electromagnetic radiation (EMR), low- dose X-radiation, low-level microwave radiation and ionizing radiation (IR) are indispensable parts of modern life. In the current review, we discussed the adaptive responses of biological systems to radiation with a focus on the impacts of radiation-induced oxidative stress (RIOS) and its molecular downstream signaling pathways.Materials and methods: A comprehensive search was conducted in Web of Sciences, PubMed, Scopus, Google Scholar, Embase, and Cochrane Library. Keywords included Mesh terms of "radiation," "electromagnetic radiation," "adaptive immunity," "oxidative stress," and "immune checkpoints." Manuscripts published up until December 2019 were included.Results: RIOS induces various molecular adaptors connected with adaptive responses in radiation exposed cells. One of these adaptors includes p53 which promotes various cellular signaling pathways. RIOS also activates the intrinsic apoptotic pathway by depolarization of the mitochondrial membrane potential and activating the caspase apoptotic cascade. RIOS is also involved in radiation-induced proliferative responses through interaction with mitogen-activated protein kinases (MAPks) including p38 MAPK, ERK, and c-Jun N-terminal kinase (JNK). Protein kinase B (Akt)/phosphoinositide 3-kinase (PI3K) signaling pathway has also been reported to be involved in RIOS-induced proliferative responses. Furthermore, RIOS promotes genetic instability by introducing DNA structural and epigenetic alterations, as well as attenuating DNA repair mechanisms. Inflammatory transcription factors including macrophage migration inhibitory factor (MIF), nuclear factor κB (NF-κB), and signal transducer and activator of transcription-3 (STAT-3) paly major role in RIOS-induced inflammation.Conclusion: In conclusion, RIOS considerably contributes to radiation induced adaptive responses. Other possible molecular adaptors modulating RIOS-induced responses are yet to be divulged in future studies.
Collapse
Affiliation(s)
- Mohsen Sisakht
- Department of Medical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Darabian
- Department of Radiology, Faculty of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Mahmoodzadeh
- Department of Medical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.,Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ali Bazi
- Faculty of Allied Medical Sciences, Zabol University of Medical Sciences, Zabol, Iran
| | - Sayed Mohammad Shafiee
- Department of Medical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Pooneh Mokarram
- Department of Medical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Khoshdel
- Department of Medical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
181
|
Cherkas A, Holota S, Mdzinarashvili T, Gabbianelli R, Zarkovic N. Glucose as a Major Antioxidant: When, What for and Why It Fails? Antioxidants (Basel) 2020; 9:antiox9020140. [PMID: 32033390 PMCID: PMC7070274 DOI: 10.3390/antiox9020140] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 02/01/2020] [Accepted: 02/03/2020] [Indexed: 02/07/2023] Open
Abstract
A human organism depends on stable glucose blood levels in order to maintain its metabolic needs. Glucose is considered to be the most important energy source, and glycolysis is postulated as a backbone pathway. However, when the glucose supply is limited, ketone bodies and amino acids can be used to produce enough ATP. In contrast, for the functioning of the pentose phosphate pathway (PPP) glucose is essential and cannot be substituted by other metabolites. The PPP generates and maintains the levels of nicotinamide adenine dinucleotide phosphate (NADPH) needed for the reduction in oxidized glutathione and protein thiols, the synthesis of lipids and DNA as well as for xenobiotic detoxification, regulatory redox signaling and counteracting infections. The flux of glucose into a PPP—particularly under extreme oxidative and toxic challenges—is critical for survival, whereas the glycolytic pathway is primarily activated when glucose is abundant, and there is lack of NADP+ that is required for the activation of glucose-6 phosphate dehydrogenase. An important role of glycogen stores in resistance to oxidative challenges is discussed. Current evidences explain the disruptive metabolic effects and detrimental health consequences of chronic nutritional carbohydrate overload, and provide new insights into the positive metabolic effects of intermittent fasting, caloric restriction, exercise, and ketogenic diet through modulation of redox homeostasis.
Collapse
Affiliation(s)
- Andriy Cherkas
- Department of Internal Medicine # 1, Lviv National Medical University, 79010 Lviv, Ukraine
- Correspondence:
| | - Serhii Holota
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Lviv National Medical University, 79010 Lviv, Ukraine;
- Department of Organic Chemistry and Pharmacy, Lesya Ukrainka Eastern European National University, 43025 Lutsk, Ukraine
| | - Tamaz Mdzinarashvili
- Institute of Medical and Applied Biophysics, I. Javakhishvili Tbilisi State University, 0128 Tbilisi, Georgia;
| | - Rosita Gabbianelli
- Unit of Molecular Biology, School of Pharmacy, University of Camerino, 62032 Camerino, Italy;
| | - Neven Zarkovic
- Laboratory for Oxidative Stress (LabOS), Institute “Rudjer Boskovic”, HR-10000 Zagreb, Croatia;
| |
Collapse
|
182
|
Matsuda F, Maeda K, Okahashi N. Computational data mining method for isotopomer analysis in the quantitative assessment of metabolic reprogramming. Sci Rep 2020; 10:286. [PMID: 31937835 PMCID: PMC6959353 DOI: 10.1038/s41598-019-57146-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 12/18/2019] [Indexed: 12/12/2022] Open
Abstract
Measurement of metabolic flux levels using stable isotope labeling has been successfully used to investigate metabolic redirection and reprogramming in living cells or tissues. The metabolic flux ratio between two reactions can be estimated from the 13C-labeling patterns of a few metabolites combined with the knowledge of atom mapping in the complicated metabolic network. However, it remains unclear whether an observed change in the labeling pattern of the metabolites is sufficient evidence of a shift in flux ratio between two metabolic states. In this study, a data analysis method was developed for the quantitative assessment of metabolic reprogramming. The Metropolis-Hastings algorithm was used with an in silico metabolic model to generate a probability distribution of metabolic flux levels under a condition in which the 13C-labeling pattern was observed. Reanalysis of literature data demonstrated that the developed method enables analysis of metabolic redirection using whole 13C-labeling pattern data. Quantitative assessment by Cohen’s effect size (d) enables a more detailed read-out of metabolic reprogramming information. The developed method will enable future applications of the metabolic isotopomer analysis to various targets, including cultured cells, whole tissues, and organs.
Collapse
Affiliation(s)
- Fumio Matsuda
- Department of Bioinformatic Engineering, Graduate School of Information Science and Technology, Osaka University, Osaka, Japan.
| | - Kousuke Maeda
- Department of Bioinformatic Engineering, Graduate School of Information Science and Technology, Osaka University, Osaka, Japan
| | - Nobuyuki Okahashi
- Department of Bioinformatic Engineering, Graduate School of Information Science and Technology, Osaka University, Osaka, Japan
| |
Collapse
|
183
|
A Genetic Screen To Identify Genes Influencing the Secondary Redox Couple NADPH/NADP + in the Yeast Saccharomyces cerevisiae. G3-GENES GENOMES GENETICS 2020; 10:371-378. [PMID: 31757928 PMCID: PMC6945034 DOI: 10.1534/g3.119.400606] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
NADPH is an important cofactor in the cell. In addition to its role in the biosynthesis of critical metabolites, it plays crucial roles in the regeneration of the reduced forms of glutathione, thioredoxins and peroxiredoxins. The enzymes and pathways that regulate NADPH are thus extremely important to understand, and yet are only partially understood. We have been interested in understanding how NADPH fluxes are altered in the cell. We describe here both an assay and a genetic screen that allows one to discern changes in NADPH levels. The screen exploits the secondary redox property of NADPH. At low levels of glutathione we show that the redox contributions of NADPH become critical for growth, and we have used this to develop a genetic screen for genes affecting NADPH homeostasis. The screen was validated in pathways that both directly (pentose phosphate pathway) and indirectly (glycolytic pathway) affect NADPH levels, and was then exploited to identify mitochondrial genes that affect NADPH homeostasis. A total of 239 mitochondrial gene knockouts were assayed using this screen. Among these, several genes were predicted to play a role in NADPH homeostasis. This included several new genes of unknown function, and others of poorly defined function. We examined two of these genes, FMP40 which encodes a protein required during oxidative stress and GOR1, glyoxylate reductase. Our studies throw new light on these proteins that appear to be major consumers of NADPH in the cell. The genetic screen is thus predicted to be an exceedingly useful tool for investigating NADPH homeostasis.
Collapse
|
184
|
ABA and sucrose co-regulate strawberry fruit ripening and show inhibition of glycolysis. Mol Genet Genomics 2019; 295:421-438. [PMID: 31807909 DOI: 10.1007/s00438-019-01629-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 11/25/2019] [Indexed: 12/21/2022]
Abstract
Abscisic acid (ABA) and sucrose play an important role in strawberry fruit ripening, but how ABA and sucrose co-regulate this ripening progress remains unclear. The intention of this study was to examine the effect of ABA and sucrose on strawberry fruit ripening and to evaluate the ABA/sucrose interaction mechanism on the strawberry fruit ripening process. Here, we report that there is an acute synergistic effect between ABA and sucrose in accelerating strawberry fruit ripening. The time frame of fruit development and ripening was shortened after the application of ABA, sucrose, and ABA + sucrose, but most of the major quality parameters in treated-ripe fruit, including fruit weight, total soluble solids, anthocyanin, ascorbic acid, the total phenolic content, lightness (L*), chroma (C*), and hue angle (h°) values were not affected. Meanwhile, the endogenous ABA and sucrose levels, and the expression of ABA and sucrose signaling genes and ripening-related genes, such as NCED1, NCED2, SnRK2.2, SuSy, MYB5, CEL1, and CEL2, was all significantly enhanced by ABA or sucrose treatment alone, but in particular, by the ABA + sucrose treatment. Therefore, improving the ripening regulation efficiency is one synergetic action of ABA/sucrose. Another synergetic action of ABA/sucrose shows that a short inhibition of glycolysis occurs during accelerated strawberry ripening. ABA and sucrose can induce higher accumulation of H2O2, leading to a transient decrease in glycolysis. Conversely, lower endogenous H2O2 levels caused by reduced glutathione (GSH) treatment resulted in a transient increase in glycolysis while delaying strawberry fruit ripening. Collectively, this study demonstrates that the ABA/sucrose interaction affects the ripening regulation efficiency and shows inhibition of glycolysis.
Collapse
|
185
|
Habashy WS, Milfort MC, Rekaya R, Aggrey SE. Cellular antioxidant enzyme activity and biomarkers for oxidative stress are affected by heat stress. INTERNATIONAL JOURNAL OF BIOMETEOROLOGY 2019; 63:1569-1584. [PMID: 31352522 DOI: 10.1007/s00484-019-01769-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 07/01/2019] [Accepted: 07/17/2019] [Indexed: 06/10/2023]
Abstract
Heat stress (HS) causes oxidative stress and cellular changes in an attempt to detoxify the harmful effects of reactive oxygen species (ROS). However, how ROS affect different organs in chickens under acute and chronic HS is relatively unknown. We investigated the cellular enzyme activity and biomarker changes in the liver and Pectoralis (P) major muscle in broiler chickens subjected to both acute and chronic HS. Forty-eight broiler chickens at 14 days old were randomly assigned to either 25 °C (control) or 35 °C (heat-stressed) for 12 days. Five birds per treatment at 1 and 12 days post-HS were euthanized, and the liver and P. major muscle were sampled. Superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), glutathione (GSH), glutathione reductase (GR), glutathione S-transferase (GST) activity as well as 8-hydroxy-2'-deoxyguanosine (8-OHdG), advanced glycation end product (AGE), malondialdehyde (MDA), and protein carbonyl (PCO) were analyzed as biomarkers for DNA, carbohydrate, lipid, and protein oxidation, respectively. The SOD, CAT, and GSH-GPx activity levels in the liver and the P. major muscle changed under HS; however, some of the changes were tissue-specific or dependent on the duration of the HS. There were increased liver 8-OHdG during chronic HS and also increased liver AGE levels during both acute and chronic HS indicating significant carbohydrate and DNA oxidations. In the P. major muscle, we observed significant increases in lipid peroxidation and protein oxidation which may reflect that this tissue is less resilient to oxidative damage under heat stress. We show that heat stress caused tissue-specific changes to levels of oxidation biomarkers in chicken.
Collapse
Affiliation(s)
- Walid S Habashy
- NutriGenomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA, 30602, USA
- Department of Animal and Poultry Production, Damanhour University, Damanhour, Egypt
| | - Marie C Milfort
- NutriGenomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA, 30602, USA
| | - Romdhane Rekaya
- Department of Animal and Dairy Science, University of Georgia, Athens, GA, 30602, USA
| | - Samuel E Aggrey
- NutriGenomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA, 30602, USA.
| |
Collapse
|
186
|
Glutathionylation primes soluble glyceraldehyde-3-phosphate dehydrogenase for late collapse into insoluble aggregates. Proc Natl Acad Sci U S A 2019; 116:26057-26065. [PMID: 31772010 DOI: 10.1073/pnas.1914484116] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Protein aggregation is a complex physiological process, primarily determined by stress-related factors revealing the hidden aggregation propensity of proteins that otherwise are fully soluble. Here we report a mechanism by which glycolytic glyceraldehyde-3-phosphate dehydrogenase of Arabidopsis thaliana (AtGAPC1) is primed to form insoluble aggregates by the glutathionylation of its catalytic cysteine (Cys149). Following a lag phase, glutathionylated AtGAPC1 initiates a self-aggregation process resulting in the formation of branched chains of globular particles made of partially misfolded and totally inactive proteins. GSH molecules within AtGAPC1 active sites are suggested to provide the initial destabilizing signal. The following removal of glutathione by the formation of an intramolecular disulfide bond between Cys149 and Cys153 reinforces the aggregation process. Physiological reductases, thioredoxins and glutaredoxins, could not dissolve AtGAPC1 aggregates but could efficiently contrast their growth. Besides acting as a protective mechanism against overoxidation, S-glutathionylation of AtGAPC1 triggers an unexpected aggregation pathway with completely different and still unexplored physiological implications.
Collapse
|
187
|
Wilkinson HC, Dalby PA. Novel insights into transketolase activation by cofactor binding identifies two native species subpopulations. Sci Rep 2019; 9:16116. [PMID: 31695144 PMCID: PMC6834573 DOI: 10.1038/s41598-019-52647-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 10/21/2019] [Indexed: 12/02/2022] Open
Abstract
Transketolase (TK) cofactor binding has been studied extensively over many years, yet certain mysteries remain, such as a lack of consensus on the cooperativity of thiamine pyrophosphate (TPP) binding into the two active sites, in the presence and absence of the divalent cation, Mg2+. Using a novel fluorescence-based assay, we determined directly the dissociation constants and cooperativity of TPP binding and provide the first comprehensive study over a broad range of cofactor concentrations. We confirmed the high-affinity dissociation constants and revealed a dependence of both the affinity and cooperativity of binding on [Mg2+], which explained the previous lack of consensus. A second, discrete and previously uncharacterised low-affinity TPP binding-site was also observed, and hence indicated the existence of two forms of TK with high- (TKhigh) and low-affinity (TKlow). The relative proportions of each dimer were independent of the monomer-dimer transition, as probed by analytical ultracentrifugation at various [TK]. Mass spectrometry revealed that chemical oxidation of TKlow led to the formation of TKhigh, which was 22-fold more active than TKlow. Finally, we propose a two-species model of transketolase activation that describes the interconversions between apo-/holo-TKhigh and TKlow, and the potential to significantly improve biocatalytic activity by populating only the most active form.
Collapse
Affiliation(s)
- Henry C Wilkinson
- Department of Biochemical Engineering, University College London, London, WC1E 6BT, UK
| | - Paul A Dalby
- Department of Biochemical Engineering, University College London, London, WC1E 6BT, UK.
| |
Collapse
|
188
|
Balcells C, Foguet C, Tarragó-Celada J, de Atauri P, Marin S, Cascante M. Tracing metabolic fluxes using mass spectrometry: Stable isotope-resolved metabolomics in health and disease. Trends Analyt Chem 2019. [DOI: 10.1016/j.trac.2018.12.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
189
|
Zhang L, Bailleul J, Yazal T, Dong K, Sung D, Dao A, Gosa L, Nathanson D, Bhat K, Duhachek-Muggy S, Alli C, Dratver MB, Pajonk F, Vlashi E. PK-M2-mediated metabolic changes in breast cancer cells induced by ionizing radiation. Breast Cancer Res Treat 2019; 178:75-86. [PMID: 31372790 PMCID: PMC6790295 DOI: 10.1007/s10549-019-05376-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 07/23/2019] [Indexed: 12/31/2022]
Abstract
PURPOSE Radiotherapy (RT) constitutes an important part of breast cancer treatment. However, triple negative breast cancers (TNBC) exhibit remarkable resistance to most therapies, including RT. Developing new ways to radiosensitize TNBC cells could result in improved patient outcomes. The M2 isoform of pyruvate kinase (PK-M2) is believed to be responsible for the re-wiring of cancer cell metabolism after oxidative stress. The aim of the study was to determine the effect of ionizing radiation (IR) on PK-M2-mediated metabolic changes in TNBC cells, and their survival. In addition, we determine the effect of PK-M2 activators on breast cancer stem cells, a radioresistant subpopulation of breast cancer stem cells. METHODS Glucose uptake, lactate production, and glutamine consumption were assessed. The cellular localization of PK-M2 was evaluated by western blot and confocal microscopy. The small molecule activator of PK-M2, TEPP46, was used to promote its pyruvate kinase function. Finally, effects on cancer stem cell were evaluated via sphere forming capacity. RESULTS Exposure of TNBC cells to IR increased their glucose uptake and lactate production. As expected, PK-M2 expression levels also increased, especially in the nucleus, although overall pyruvate kinase activity was decreased. PK-M2 nuclear localization was shown to be associated with breast cancer stem cells, and activation of PK-M2 by TEPP46 depleted this population. CONCLUSIONS Radiotherapy can induce metabolic changes in TNBC cells, and these changes seem to be mediated, at least in part by PK-M2. Importantly, our results show that activators of PK-M2 can deplete breast cancer stem cells in vitro. This study supports the idea of combining PK-M2 activators with radiation to enhance the effect of radiotherapy in resistant cancers, such as TNBC.
Collapse
Affiliation(s)
- Le Zhang
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA, 90095-1714, USA
| | - Justine Bailleul
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA, 90095-1714, USA
| | - Taha Yazal
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA, 90095-1714, USA
| | - Kevin Dong
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA, 90095-1714, USA
| | - David Sung
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA, 90095-1714, USA
| | - Amy Dao
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA, 90095-1714, USA
| | - Laura Gosa
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - David Nathanson
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA
| | - Kruttika Bhat
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA, 90095-1714, USA
| | - Sara Duhachek-Muggy
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA, 90095-1714, USA
| | - Claudia Alli
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA, 90095-1714, USA
| | - Milana Bochkur Dratver
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA, 90095-1714, USA
| | - Frank Pajonk
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA, 90095-1714, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA
| | - Erina Vlashi
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA, 90095-1714, USA.
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
190
|
Lee V, Gober MD, Bashir H, O'Day C, Blair IA, Mesaros C, Weng L, Huang A, Chen A, Tang R, Anagnos V, Li J, Roling S, Sagaityte E, Wang A, Lin C, Yeh C, Atillasoy C, Marshall C, Dentchev T, Ridky T, Seykora JT. Voriconazole enhances UV-induced DNA damage by inhibiting catalase and promoting oxidative stress. Exp Dermatol 2019; 29:29-38. [PMID: 31519066 DOI: 10.1111/exd.14038] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 08/02/2019] [Accepted: 08/30/2019] [Indexed: 12/17/2022]
Abstract
Cutaneous squamous cell carcinoma (cSCC) is the second most common form of skin cancer and is associated with cumulative UV exposure. Studies have shown that prolonged voriconazole use promotes cSCC formation; however, the biological mechanisms responsible for the increased incidence remain unclear. Here, we show that voriconazole directly increases oxidative stress in human keratinocytes and promotes UV-induced DNA damage as determined by comet assay, 8-oxoguanine immunofluorescence and mass spectrometry. Voriconazole treatment of human keratinocytes potentiates UV-induced apoptosis and activation of the p38 MAP kinase and 53BP1 UV stress response pathways. The p38 MAP kinase activation promoted by voriconazole exposure can be mitigated by pretreating keratinocytes with N-acetylcysteine. Voriconazole increases oxidative stress in keratinocytes by directly inhibiting catalase leading to lower intracellular NADPH levels and the triazole moieties in voriconazole are critical for inhibiting catalase. Furthermore, voriconazole is shown to promote UV-induced dysplasia in an in vivo model. Together, these data demonstrate that voriconazole potentiates oxidative stress in UV-irradiated keratinocytes through catalase inhibition. Use of antioxidants may mitigate the pro-oncogenic effects of voriconazole.
Collapse
Affiliation(s)
- Vivian Lee
- Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Ophthalmology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michael D Gober
- Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hasan Bashir
- Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Ophthalmology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Conor O'Day
- Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ian A Blair
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Clementina Mesaros
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Liwei Weng
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Andrew Huang
- Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Aaron Chen
- Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Rachel Tang
- Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Vince Anagnos
- Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - JiLon Li
- Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sophie Roling
- Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Emilija Sagaityte
- Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Andrew Wang
- Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Chenyan Lin
- Department of Ophthalmology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Christopher Yeh
- Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Cem Atillasoy
- Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Christine Marshall
- Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Tzvete Dentchev
- Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Todd Ridky
- Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - John T Seykora
- Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
191
|
Sugar as a therapeutic target for the cognitive restoration following traumatic brain injury. Curr Opin Neurol 2019; 32:815-821. [PMID: 31609736 DOI: 10.1097/wco.0000000000000752] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW This review aims to discuss examples of changes in glucose (sugar) metabolism after traumatic brain injury (TBI). It will attempt to provide an understanding of what changes in glucose metabolism mean for the injured brain. It will further identify potential therapeutic target(s) emanating from our growing understanding of glucose pathways and their roles in TBI. RECENT FINDINGS Although a significant fraction of glucose is utilized for the energy production in the brain, a small fraction is utilized in other, often ignored pathways. Recent studies have unraveled unexpected biological effects of glucose through these pathways, including redox regulation, genetic and epigenetic regulation, glycation of proteins, nucleotide synthesis and amino acid synthesis. SUMMARY A number of regulatory players in minor glucose metabolic pathways, such as folate and chondroitin sulfate proteoglycans, have recently been identified as potential targets to restore cognitive functions. Targeting of these players should be combined with the supplementation of alternative energy substrates to achieve the maximal cognitive restoration after TBI. This multimodal therapeutic strategy deserves testing in various models of TBI. VIDEO ABSTRACT Supplemental digital video content 1: Video that demonstrates an effective therapeutic strategy for the cognitive restoration after TBI. http://links.lww.com/CONR/A46.
Collapse
|
192
|
Xie K, Varatnitskaya M, Maghnouj A, Bader V, Winklhofer KF, Hahn S, Leichert LI. Activation leads to a significant shift in the intracellular redox homeostasis of neutrophil-like cells. Redox Biol 2019; 28:101344. [PMID: 31639650 PMCID: PMC6807386 DOI: 10.1016/j.redox.2019.101344] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 09/19/2019] [Accepted: 10/11/2019] [Indexed: 11/28/2022] Open
Abstract
Neutrophils produce a cocktail of oxidative species during the so-called oxidative burst to attack phagocytized bacteria. However, little is known about the neutrophils' redox homeostasis during the oxidative burst and there is currently no consensus about the interplay between oxidative species and cellular signaling, e.g. during the initiation of the production of neutrophil extracellular traps (NETs). Using the genetically encoded redox sensor roGFP2, expressed in the cytoplasm of the neutrophil-like cell line PLB-985, we saw that stimulation by both PMA and E. coli resulted in oxidation of the thiol residues in this probe. In contrast to the redox state of phagocytized bacteria, which completely breaks down, the neutrophils' cytoplasmic redox state switched from its intital -318 ± 6 mV to a new, albeit higher oxidized, steady state of -264 ± 5 mV in the presence of bacteria. This highly significant oxidation of the cytosol (p value = 7 × 10-5) is dependent on NOX2 activity, but independent of the most effective thiol oxidant produced in neutrophils, MPO-derived HOCl. While the shift in the intracellular redox potential is correlated with effective NETosis, it is, by itself not sufficient: Inhibition of MPO, while not affecting the cytosolic oxidation, significantly decreased NETosis. Furthermore, inhibition of PI3K, which abrogates cytosolic oxidation, did not fully prevent NETosis induced by phagocytosis of bacteria. Thus, we conclude that NET-formation is regulated in a multifactorial way, in part by changes of the cytosolic thiol redox homeostasis in neutrophils, depending on the circumstance under which the generation of NETs was initiated.
Collapse
Affiliation(s)
- Kaibo Xie
- Ruhr University Bochum, Institute of Biochemistry and Pathobiochemistry, Microbial Biochemistry, Bochum, Germany
| | - Marharyta Varatnitskaya
- Ruhr University Bochum, Institute of Biochemistry and Pathobiochemistry, Microbial Biochemistry, Bochum, Germany
| | - Abdelouahid Maghnouj
- Ruhr University Bochum, Department of Molecular Gastrointestinal Oncology, Bochum, Germany
| | - Verian Bader
- Ruhr University Bochum, Institute for Biochemistry and Pathobiochemistry, Molecular Cell Biology, Bochum, Germany
| | - Konstanze F Winklhofer
- Ruhr University Bochum, Institute for Biochemistry and Pathobiochemistry, Molecular Cell Biology, Bochum, Germany
| | - Stephan Hahn
- Ruhr University Bochum, Department of Molecular Gastrointestinal Oncology, Bochum, Germany
| | - Lars I Leichert
- Ruhr University Bochum, Institute of Biochemistry and Pathobiochemistry, Microbial Biochemistry, Bochum, Germany.
| |
Collapse
|
193
|
Christodoulou D, Kuehne A, Estermann A, Fuhrer T, Lang P, Sauer U. Reserve Flux Capacity in the Pentose Phosphate Pathway by NADPH Binding Is Conserved across Kingdoms. iScience 2019; 19:1133-1144. [PMID: 31536961 PMCID: PMC6831883 DOI: 10.1016/j.isci.2019.08.047] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 07/13/2019] [Accepted: 08/24/2019] [Indexed: 02/03/2023] Open
Abstract
All organisms evolved defense mechanisms to counteract oxidative stress and buildup of reactive oxygen species (ROS). To test whether a potentially conserved mechanism exists for the rapid response, we investigated immediate metabolic dynamics of Escherichia coli, yeast, and human dermal fibroblasts to oxidative stress that we found to be conserved between species. To elucidate the regulatory mechanisms that implement this metabolic response, we developed mechanistic kinetic models for each organism's central metabolism and systematically tested activation and inactivation of each irreversible reaction by each metabolite. This ensemble modeling predicts in vivo relevant metabolite-enzyme interactions based on their ability to quantitatively describe metabolite dynamics. All three species appear to inhibit their oxidative pentose phosphate pathway during normal growth by the redox cofactor NADPH and relieve this inhibition to increase the pathway flux for detoxification of ROS during stress, with the sole exception of yeast when exposed to high levels of stress.
Collapse
Affiliation(s)
- Dimitris Christodoulou
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland; Systems Biology Graduate School, Zurich 8057, Switzerland
| | - Andreas Kuehne
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland; Systems Biology Graduate School, Zurich 8057, Switzerland
| | | | - Tobias Fuhrer
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Paul Lang
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Uwe Sauer
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
194
|
Kim E, Cho Y, Chung W, Roe J. The role of Rsv1 in the transcriptional regulation of genes involved in sugar metabolism for long‐term survival. FEBS J 2019; 287:878-896. [DOI: 10.1111/febs.15052] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 07/10/2019] [Accepted: 08/28/2019] [Indexed: 12/31/2022]
Affiliation(s)
- Eun‐Jung Kim
- Laboratory of Molecular Microbiology School of Biological Sciences and Institute of Microbiology Seoul National University Korea
| | | | - Woo‐Hyun Chung
- College of Pharmacy Duksung Women's University Seoul Korea
| | - Jung‐Hye Roe
- Laboratory of Molecular Microbiology School of Biological Sciences and Institute of Microbiology Seoul National University Korea
| |
Collapse
|
195
|
Yang HC, Wu YH, Yen WC, Liu HY, Hwang TL, Stern A, Chiu DTY. The Redox Role of G6PD in Cell Growth, Cell Death, and Cancer. Cells 2019; 8:cells8091055. [PMID: 31500396 PMCID: PMC6770671 DOI: 10.3390/cells8091055] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 09/02/2019] [Accepted: 09/07/2019] [Indexed: 02/07/2023] Open
Abstract
The generation of reducing equivalent NADPH via glucose-6-phosphate dehydrogenase (G6PD) is critical for the maintenance of redox homeostasis and reductive biosynthesis in cells. NADPH also plays key roles in cellular processes mediated by redox signaling. Insufficient G6PD activity predisposes cells to growth retardation and demise. Severely lacking G6PD impairs embryonic development and delays organismal growth. Altered G6PD activity is associated with pathophysiology, such as autophagy, insulin resistance, infection, inflammation, as well as diabetes and hypertension. Aberrant activation of G6PD leads to enhanced cell proliferation and adaptation in many types of cancers. The present review aims to update the existing knowledge concerning G6PD and emphasizes how G6PD modulates redox signaling and affects cell survival and demise, particularly in diseases such as cancer. Exploiting G6PD as a potential drug target against cancer is also discussed.
Collapse
Affiliation(s)
- Hung-Chi Yang
- Department of Medical Laboratory Science and Biotechnology, Yuanpei University of Medical Technology, Hsinchu, Taiwan.
| | - Yi-Hsuan Wu
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan.
| | - Wei-Chen Yen
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
- Department of Medical Biotechnology and Laboratory Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Hui-Ya Liu
- Department of Medical Biotechnology and Laboratory Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Tsong-Long Hwang
- Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan.
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
- Chinese Herbal Medicine Research Team, Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan.
- Department of Anaesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan.
- Department of Chemical Engineering, Ming Chi University of Technology, New Taipei City, Taiwan.
- Research Center for Chinese Herbal Medicine, Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan.
| | - Arnold Stern
- New York University School of Medicine, New York, NY, USA.
| | - Daniel Tsun-Yee Chiu
- Department of Medical Biotechnology and Laboratory Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
- Research Center for Chinese Herbal Medicine, Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan.
- Department of Pediatric Hematology/Oncology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan.
- Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
196
|
Bohutskyi P, McClure RS, Hill EA, Nelson WC, Chrisler WB, Nuñez JR, Renslow RS, Charania MA, Lindemann SR, Beliaev AS. Metabolic effects of vitamin B12 on physiology, stress resistance, growth rate and biomass productivity of Cyanobacterium stanieri planktonic and biofilm cultures. ALGAL RES 2019. [DOI: 10.1016/j.algal.2019.101580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
197
|
Shimizu K, Matsuoka Y. Redox rebalance against genetic perturbations and modulation of central carbon metabolism by the oxidative stress regulation. Biotechnol Adv 2019; 37:107441. [PMID: 31472206 DOI: 10.1016/j.biotechadv.2019.107441] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 08/04/2019] [Accepted: 08/23/2019] [Indexed: 12/11/2022]
Abstract
The micro-aerophilic organisms and aerobes as well as yeast and higher organisms have evolved to gain energy through respiration (via oxidative phosphorylation), thereby enabling them to grow much faster than anaerobes. However, during respiration, reactive oxygen species (ROSs) are inherently (inevitably) generated, and threaten the cell's survival. Therefore, living organisms (or cells) must furnish the potent defense systems to keep such ROSs at harmless level, where the cofactor balance plays crucial roles. Namely, NADH is the source of energy generation (catabolism) in the respiratory chain reactions, through which ROSs are generated, while NADPH plays important roles not only for the cell synthesis (anabolism) but also for detoxifying ROSs. Therefore, the cell must rebalance the redox ratio by modulating the fluxes of the central carbon metabolism (CCM) by regulating the multi-level regulation machinery upon genetic perturbations and the change in the growth conditions. Here, we discuss about how aerobes accomplish such cofactor homeostasis against redox perturbations. In particular, we consider how single-gene mutants (including pgi, pfk, zwf, gnd and pyk mutants) modulate their metabolisms in relation to cofactor rebalance (and also by adaptive laboratory evolution). We also discuss about how the overproduction of NADPH (by the pathway gene mutation) can be utilized for the efficient production of useful value-added chemicals such as medicinal compounds, polyhydroxyalkanoates, and amino acids, all of which require NADPH in their synthetic pathways. We then discuss about the metabolic responses against oxidative stress, where αketoacids play important roles not only for the coordination between catabolism and anabolism, but also for detoxifying ROSs by non-enzymatic reactions, as well as for reducing the production of ROSs by repressing the activities of the TCA cycle and respiration (via carbon catabolite repression). Thus, we discuss about the mechanisms (basic strategies) that modulate the metabolism from respiration to respiro-fermentative metabolism causing overflow, based on the role of Pyk activity, affecting the NADPH production at the oxidative pentose phosphate (PP) pathway, and the roles of αketoacids for the change in the source of energy generation from the oxidative phosphorylation to the substrate level phosphorylation.
Collapse
Affiliation(s)
- Kazuyuki Shimizu
- Kyushu institute of Technology, Iizuka, Fukuoka 820-8502, Japan; Institute of Advanced Biosciences, Keio university, Tsuruoka, Yamagata 997-0017, Japan.
| | - Yu Matsuoka
- Kyushu institute of Technology, Iizuka, Fukuoka 820-8502, Japan.
| |
Collapse
|
198
|
Schmelter C, Fomo KN, Perumal N, Manicam C, Bell K, Pfeiffer N, Grus FH. Synthetic Polyclonal-Derived CDR Peptides as an Innovative Strategy in Glaucoma Therapy. J Clin Med 2019; 8:jcm8081222. [PMID: 31443184 PMCID: PMC6723090 DOI: 10.3390/jcm8081222] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 07/30/2019] [Accepted: 08/12/2019] [Indexed: 12/16/2022] Open
Abstract
The pathogenesis of glaucoma is strongly associated with the occurrence of autoimmune-mediated loss of retinal ganglion cells (RGCs) and additionally, recent evidence shows that specific antibody-derived signature peptides are significantly differentially expressed in sera of primary-open angle glaucoma patients (POAG) compared to healthy controls. Synthetically antibody-derived peptides can modulate various effector functions of the immune system and act as antimicrobial or antiviral molecules. In an ex vivo adolescent glaucoma model, this study, for the first time, demonstrates that polyclonal-derived complementarity-determining regions (CDRs) can significantly increase the survival rate of RGCs (p = 0.013). We subsequently performed affinity capture experiments that verified the mitochondrial serine protease HTRA2 (gene name: HTRA2) as a high-affinity retinal epitope target of CDR1 sequence motif ASGYTFTNYGLSWVR. Quantitative proteomic analysis of the CDR-treated retinal explants revealed increased expression of various anti-apoptotic and anti-oxidative proteins (e.g., VDAC2 and TXN) compared to untreated controls (p < 0.05) as well as decreased expression levels of cellular stress response markers (e.g., HSPE1 and HSP90AA1). Mitochondrial dysfunction, the protein ubiquitination pathway and oxidative phosphorylation were annotated as the most significantly affected signaling pathways and possibly can be traced back to the CDR-induced inhibition or modulation of the master regulator HTRA2. These findings emphasize the great potential of synthetic polyclonal-derived CDR peptides as therapeutic agents in future glaucoma therapy and provide an excellent basis for affinity-based biomarker discovery purposes.
Collapse
Affiliation(s)
- Carsten Schmelter
- Department of Experimental and Translational Ophthalmology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany
| | - Kristian Nzogang Fomo
- Department of Experimental and Translational Ophthalmology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany
| | - Natarajan Perumal
- Department of Experimental and Translational Ophthalmology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany
| | - Caroline Manicam
- Department of Experimental and Translational Ophthalmology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany
| | - Katharina Bell
- Department of Experimental and Translational Ophthalmology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany
| | - Norbert Pfeiffer
- Department of Experimental and Translational Ophthalmology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany
| | - Franz H Grus
- Department of Experimental and Translational Ophthalmology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany.
| |
Collapse
|
199
|
Conserved roles of glucose in suppressing reactive oxygen species-induced cell death and animal survival. Aging (Albany NY) 2019; 11:5726-5743. [PMID: 31403933 PMCID: PMC6710067 DOI: 10.18632/aging.102155] [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: 06/19/2019] [Accepted: 08/03/2019] [Indexed: 02/06/2023]
Abstract
Carbohydrate overconsumption increases blood glucose levels, which contributes to the development of various diseases including obesity and diabetes. It is generally believed that high glucose metabolism increases cellular reactive oxygen species (ROS) levels, damages insulin-secreting cells and leads to age-associated diabetic phenotypes. Here we find that in contrast, high glucose suppresses ROS production induced by paraquat in both mammalian cells and the round worm C. elegans. The role of glucose in suppressing ROS is further supported by glucose's ability to alleviate paraquat's toxicity on C. elegans development. Consistently, we find that the ROS-regulated transcription factor SKN-1 is inactivated by glucose. As a result, the ROS/SKN-1-dependent lifespan extension observed in paraquat-treated animals, mitochondrial respiration mutant isp-1 and germline-less mutant glp-1 are all suppressed by glucose. Our study reveals an unprecedented interaction of glucose with ROS, which could have significant impact on our current understanding of glucose- and ROS-related diseases.
Collapse
|
200
|
Olin-Sandoval V, Yu JSL, Miller-Fleming L, Alam MT, Kamrad S, Correia-Melo C, Haas R, Segal J, Peña Navarro DA, Herrera-Dominguez L, Méndez-Lucio O, Vowinckel J, Mülleder M, Ralser M. Lysine harvesting is an antioxidant strategy and triggers underground polyamine metabolism. Nature 2019; 572:249-253. [PMID: 31367038 PMCID: PMC6774798 DOI: 10.1038/s41586-019-1442-6] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Accepted: 06/28/2019] [Indexed: 11/25/2022]
Abstract
Both single and multicellular organisms depend on anti-stress mechanisms that enable them to deal with sudden changes in the environment, including exposure to heat and oxidants. Central to the stress response are dynamic changes in metabolism, such as the transition from the glycolysis to the pentose phosphate pathway-a conserved first-line response to oxidative insults1,2. Here we report a second metabolic adaptation that protects microbial cells in stress situations. The role of the yeast polyamine transporter Tpo1p3-5 in maintaining oxidant resistance is unknown6. However, a proteomic time-course experiment suggests a link to lysine metabolism. We reveal a connection between polyamine and lysine metabolism during stress situations, in the form of a promiscuous enzymatic reaction in which the first enzyme of the polyamine pathway, Spe1p, decarboxylates lysine and forms an alternative polyamine, cadaverine. The reaction proceeds in the presence of extracellular lysine, which is taken up by cells to reach concentrations up to one hundred times higher than those required for growth. Such extensive harvest is not observed for the other amino acids, is dependent on the polyamine pathway and triggers a reprogramming of redox metabolism. As a result, NADPH-which would otherwise be required for lysine biosynthesis-is channelled into glutathione metabolism, leading to a large increase in glutathione concentrations, lower levels of reactive oxygen species and increased oxidant tolerance. Our results show that nutrient uptake occurs not only to enable cell growth, but when the nutrient availability is favourable it also enables cells to reconfigure their metabolism to preventatively mount stress protection.
Collapse
Affiliation(s)
- Viridiana Olin-Sandoval
- Department of Biochemistry, University of Cambridge, Cambridge, UK
- Department of Nutrition Physiology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Jason Shu Lim Yu
- The Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, UK
| | - Leonor Miller-Fleming
- Department of Biochemistry, University of Cambridge, Cambridge, UK
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | | | - Stephan Kamrad
- The Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, UK
- Department of Genetics, Evolution & Environment, University College London, London, UK
| | - Clara Correia-Melo
- The Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, UK
| | - Robert Haas
- Department of Biochemistry, University of Cambridge, Cambridge, UK
- The Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, UK
| | - Joanna Segal
- The Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, UK
| | | | | | - Oscar Méndez-Lucio
- Facultad de Química, Departamento de Farmacia, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Jakob Vowinckel
- Department of Biochemistry, University of Cambridge, Cambridge, UK
- Biognosys AG, Schlieren, Switzerland
| | - Michael Mülleder
- Department of Biochemistry, University of Cambridge, Cambridge, UK
- The Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, UK
- Department of Biochemistry, Charité University Medicine, Berlin, Germany
| | - Markus Ralser
- Department of Biochemistry, University of Cambridge, Cambridge, UK.
- The Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, UK.
- Department of Biochemistry, Charité University Medicine, Berlin, Germany.
| |
Collapse
|