51
|
Banerjee S, Sadler PJ. Transfer hydrogenation catalysis in cells. RSC Chem Biol 2021; 2:12-29. [PMID: 34458774 PMCID: PMC8341873 DOI: 10.1039/d0cb00150c] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 10/10/2020] [Indexed: 12/13/2022] Open
Abstract
Hydrogenation reactions in biology are usually carried out by enzymes with nicotinamide adenine dinucleotide (NAD(P)H) or flavin mononucleotide (FAMH2)/flavinadenine dinucleotide (FADH2) as cofactors and hydride sources. Industrial scale chemical transfer hydrogenation uses small molecules such as formic acid or alcohols (e.g. propanol) as hydride sources and transition metal complexes as catalysts. We focus here on organometallic half-sandwich RuII and OsII η6-arene complexes and RhIII and IrIII η5-Cp x complexes which catalyse hydrogenation of biomolecules such as pyruvate and quinones in aqueous media, and generate biologically important species such as H2 and H2O2. Organometallic catalysts can achieve enantioselectivity, and moreover can be active in living cells, which is surprising on account of the variety of poisons present. Such catalysts can induce reductive stress using formate as hydride source or oxidative stress by accepting hydride from NAD(P)H. In some cases, photocatalytic redox reactions can be induced by light absorption at metal or flavin centres. These artificial transformations can interfere in biochemical pathways in unusual ways, and are the basis for the design of metallodrugs with novel mechanisms of action.
Collapse
Affiliation(s)
- Samya Banerjee
- Department of Chemistry, University of Warwick, Gibbet Hill Road Coventry CV4 7AL UK
| | - Peter J Sadler
- Department of Chemistry, University of Warwick, Gibbet Hill Road Coventry CV4 7AL UK
| |
Collapse
|
52
|
Pamplona R, Jové M, Mota-Martorell N, Barja G. Is the NDUFV2 subunit of the hydrophilic complex I domain a key determinant of animal longevity? FEBS J 2021; 288:6652-6673. [PMID: 33455045 DOI: 10.1111/febs.15714] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 12/02/2020] [Accepted: 01/14/2021] [Indexed: 12/18/2022]
Abstract
Complex I, a component of the electron transport chain, plays a central functional role in cell bioenergetics and the biology of free radicals. The structural and functional N module of complex I is one of the main sites of the generation of free radicals. The NDUFV2 subunit/N1a cluster is a component of this module. Furthermore, the rate of free radical production is linked to animal longevity. In this review, we explore the hypothesis that NDUFV2 is the only conserved core subunit designed with a regulatory function to ensure correct electron transfer and free radical production, that low gene expression and protein abundance of the NDUFV2 subunit is an evolutionary adaptation needed to achieve a longevity phenotype, and that these features are determinants of the lower free radical generation at the mitochondrial level and a slower rate of aging of long-lived animals.
Collapse
Affiliation(s)
- Reinald Pamplona
- Department of Experimental Medicine, University of Lleida-Lleida Biomedical Research Institute (UdL-IRBLleida), Lleida, Spain
| | - Mariona Jové
- Department of Experimental Medicine, University of Lleida-Lleida Biomedical Research Institute (UdL-IRBLleida), Lleida, Spain
| | - Natalia Mota-Martorell
- Department of Experimental Medicine, University of Lleida-Lleida Biomedical Research Institute (UdL-IRBLleida), Lleida, Spain
| | - Gustavo Barja
- Department of Genetics, Physiology and Microbiology, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
53
|
Ramaccini D, Montoya-Uribe V, Aan FJ, Modesti L, Potes Y, Wieckowski MR, Krga I, Glibetić M, Pinton P, Giorgi C, Matter ML. Mitochondrial Function and Dysfunction in Dilated Cardiomyopathy. Front Cell Dev Biol 2021; 8:624216. [PMID: 33511136 PMCID: PMC7835522 DOI: 10.3389/fcell.2020.624216] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 12/16/2020] [Indexed: 12/14/2022] Open
Abstract
Cardiac tissue requires a persistent production of energy in order to exert its pumping function. Therefore, the maintenance of this function relies on mitochondria that represent the “powerhouse” of all cardiac activities. Mitochondria being one of the key players for the proper functioning of the mammalian heart suggests continual regulation and organization. Mitochondria adapt to cellular energy demands via fusion-fission events and, as a proof-reading ability, undergo mitophagy in cases of abnormalities. Ca2+ fluxes play a pivotal role in regulating all mitochondrial functions, including ATP production, metabolism, oxidative stress balance and apoptosis. Communication between mitochondria and others organelles, especially the sarcoplasmic reticulum is required for optimal function. Consequently, abnormal mitochondrial activity results in decreased energy production leading to pathological conditions. In this review, we will describe how mitochondrial function or dysfunction impacts cardiac activities and the development of dilated cardiomyopathy.
Collapse
Affiliation(s)
- Daniela Ramaccini
- University of Hawaii Cancer Center, Honolulu, HI, United States.,Department of Medical Sciences, University of Ferrara, Ferrara, Italy.,Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, Ferrara, Italy
| | | | - Femke J Aan
- University of Hawaii Cancer Center, Honolulu, HI, United States
| | - Lorenzo Modesti
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy.,Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, Ferrara, Italy
| | - Yaiza Potes
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Mariusz R Wieckowski
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Irena Krga
- Center of Research Excellence in Nutrition and Metabolism, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Marija Glibetić
- Center of Research Excellence in Nutrition and Metabolism, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Paolo Pinton
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy.,Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, Ferrara, Italy.,Maria Cecilia Hospital, GVM Care & Research, Cotignola, Italy
| | - Carlotta Giorgi
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy.,Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, Ferrara, Italy
| | | |
Collapse
|
54
|
Liu Y, Nie X, Zhu J, Wang T, Li Y, Wang Q, Sun Z. NDUFA4L2 in smooth muscle promotes vascular remodeling in hypoxic pulmonary arterial hypertension. J Cell Mol Med 2021; 25:1221-1237. [PMID: 33340241 PMCID: PMC7812284 DOI: 10.1111/jcmm.16193] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 11/10/2020] [Accepted: 11/21/2020] [Indexed: 12/12/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterized by a progressive increase in pulmonary vascular resistance and obliterative pulmonary vascular remodelling (PVR). The imbalance between the proliferation and apoptosis of pulmonary artery smooth muscle cells (PASMCs) is an important cause of PVR leading to PAH. Mitochondria play a key role in the production of hypoxia-induced pulmonary hypertension (HPH). However, there are still many issues worth studying in depth. In this study, we demonstrated that NADH dehydrogenase (ubiquinone) 1 alpha subcomplex 4 like 2 (NDUFA4L2) was a proliferation factor and increased in vivo and in vitro through various molecular biology experiments. HIF-1α was an upstream target of NDUFA4L2. The plasma levels of 4-hydroxynonene (4-HNE) were increased both in PAH patients and hypoxic PAH model rats. Knockdown of NDUFA4L2 decreased the levels of malondialdehyde (MDA) and 4-HNE in human PASMCs in hypoxia. Elevated MDA and 4-HNE levels might be associated with excessive ROS generation and increased expression of 5-lipoxygenase (5-LO) in hypoxia, but this effect was blocked by siNDUFA4L2. Further research found that p38-5-LO was a downstream signalling pathway of PASMCs proliferation induced by NDUFA4L2. Up-regulated NDUFA4L2 plays a critical role in the development of HPH, which mediates ROS production and proliferation of PASMCs, suggesting NDUFA4L2 as a potential new therapeutic target for PAH.
Collapse
MESH Headings
- Aldehydes/metabolism
- Animals
- Arachidonate 5-Lipoxygenase/metabolism
- Cell Hypoxia
- Cell Proliferation
- Disease Models, Animal
- Electron Transport Complex I/genetics
- Electron Transport Complex I/metabolism
- Endothelial Cells/metabolism
- Gene Expression Regulation
- Gene Silencing
- Humans
- Hypoxia/complications
- Hypoxia/physiopathology
- Male
- Malondialdehyde/metabolism
- Models, Biological
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/physiopathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Oxidation-Reduction
- Oxygen Consumption
- Pulmonary Arterial Hypertension/complications
- Pulmonary Arterial Hypertension/metabolism
- Pulmonary Arterial Hypertension/pathology
- Pulmonary Arterial Hypertension/physiopathology
- Pulmonary Artery/pathology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats, Wistar
- Reactive Oxygen Species/metabolism
- Vascular Remodeling/genetics
- p38 Mitogen-Activated Protein Kinases/metabolism
- Rats
Collapse
Affiliation(s)
- Yun Liu
- Department of Pharmacy, The First People's Hospital of Lianyungang, Lianyungang, China
- Department of Pharmacy, The Affiliated Lianyungang Hospital of Xuzhou Medical University/The First People's Hospital of Lianyungang, Lianyungang, China
| | - Xiaowei Nie
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Shenzhen, China
- Lung Transplant Group, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Jinquan Zhu
- Department of Pharmacy, The First People's Hospital of Lianyungang, Lianyungang, China
| | - Tianyan Wang
- Department of Pharmacy, The First People's Hospital of Lianyungang, Lianyungang, China
| | - Yanli Li
- Department of Pharmacy, The First People's Hospital of Lianyungang, Lianyungang, China
| | - Qian Wang
- Department of Anesthesiology, Children's Hospital of Soochow University, Suzhou, China
| | - Zengxian Sun
- Department of Pharmacy, The First People's Hospital of Lianyungang, Lianyungang, China
- Department of Pharmacy, The Affiliated Lianyungang Hospital of Xuzhou Medical University/The First People's Hospital of Lianyungang, Lianyungang, China
| |
Collapse
|
55
|
Pasquale V, Ducci G, Campioni G, Ventrici A, Assalini C, Busti S, Vanoni M, Vago R, Sacco E. Profiling and Targeting of Energy and Redox Metabolism in Grade 2 Bladder Cancer Cells with Different Invasiveness Properties. Cells 2020; 9:cells9122669. [PMID: 33322565 PMCID: PMC7764708 DOI: 10.3390/cells9122669] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/07/2020] [Accepted: 12/08/2020] [Indexed: 12/14/2022] Open
Abstract
Bladder cancer is one of the most prevalent deadly diseases worldwide. Grade 2 tumors represent a good window of therapeutic intervention, whose optimization requires high resolution biomarker identification. Here we characterize energy metabolism and cellular properties associated with spreading and tumor progression of RT112 and 5637, two Grade 2 cancer cell lines derived from human bladder, representative of luminal-like and basal-like tumors, respectively. The two cell lines have similar proliferation rates, but only 5637 cells show efficient lateral migration. In contrast, RT112 cells are more prone to form spheroids. RT112 cells produce more ATP by glycolysis and OXPHOS, present overall higher metabolic plasticity and are less sensitive than 5637 to nutritional perturbation of cell proliferation and migration induced by treatment with 2-deoxyglucose and metformin. On the contrary, spheroid formation is less sensitive to metabolic perturbations in 5637 than RT112 cells. The ability of metformin to reduce, although with different efficiency, cell proliferation, sphere formation and migration in both cell lines, suggests that OXPHOS targeting could be an effective strategy to reduce the invasiveness of Grade 2 bladder cancer cells.
Collapse
Affiliation(s)
- Valentina Pasquale
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (V.P.); (G.D.); (G.C.); (A.V.); (S.B.)
- SYSBIO-ISBE-IT-Candidate National Node of Italy for ISBE, Research Infrastructure for Systems Biology Europe, 20126 Milan, Italy
| | - Giacomo Ducci
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (V.P.); (G.D.); (G.C.); (A.V.); (S.B.)
- SYSBIO-ISBE-IT-Candidate National Node of Italy for ISBE, Research Infrastructure for Systems Biology Europe, 20126 Milan, Italy
| | - Gloria Campioni
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (V.P.); (G.D.); (G.C.); (A.V.); (S.B.)
- SYSBIO-ISBE-IT-Candidate National Node of Italy for ISBE, Research Infrastructure for Systems Biology Europe, 20126 Milan, Italy
| | - Adria Ventrici
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (V.P.); (G.D.); (G.C.); (A.V.); (S.B.)
| | - Chiara Assalini
- Urological Research Institute, Division of Experimental Oncology, IRCCS San Raffaele Hospital, 20132 Milan, Italy;
| | - Stefano Busti
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (V.P.); (G.D.); (G.C.); (A.V.); (S.B.)
- SYSBIO-ISBE-IT-Candidate National Node of Italy for ISBE, Research Infrastructure for Systems Biology Europe, 20126 Milan, Italy
| | - Marco Vanoni
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (V.P.); (G.D.); (G.C.); (A.V.); (S.B.)
- SYSBIO-ISBE-IT-Candidate National Node of Italy for ISBE, Research Infrastructure for Systems Biology Europe, 20126 Milan, Italy
- Correspondence: (M.V.); (R.V.); (E.S.); Tel.: +39-02-6448-3525 (M.V.); +39-02-2643-5664 (R.V.); +39-02-6448-3379 (E.S.)
| | - Riccardo Vago
- Urological Research Institute, Division of Experimental Oncology, IRCCS San Raffaele Hospital, 20132 Milan, Italy;
- Università Vita-Salute San Raffaele, 20132 Milan, Italy
- Correspondence: (M.V.); (R.V.); (E.S.); Tel.: +39-02-6448-3525 (M.V.); +39-02-2643-5664 (R.V.); +39-02-6448-3379 (E.S.)
| | - Elena Sacco
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (V.P.); (G.D.); (G.C.); (A.V.); (S.B.)
- SYSBIO-ISBE-IT-Candidate National Node of Italy for ISBE, Research Infrastructure for Systems Biology Europe, 20126 Milan, Italy
- Correspondence: (M.V.); (R.V.); (E.S.); Tel.: +39-02-6448-3525 (M.V.); +39-02-2643-5664 (R.V.); +39-02-6448-3379 (E.S.)
| |
Collapse
|
56
|
Seco-Cervera M, González-Cabo P, Pallardó FV, Romá-Mateo C, García-Giménez JL. Thioredoxin and Glutaredoxin Systems as Potential Targets for the Development of New Treatments in Friedreich's Ataxia. Antioxidants (Basel) 2020; 9:antiox9121257. [PMID: 33321938 PMCID: PMC7763308 DOI: 10.3390/antiox9121257] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 12/11/2022] Open
Abstract
The thioredoxin family consists of a small group of redox proteins present in all organisms and composed of thioredoxins (TRXs), glutaredoxins (GLRXs) and peroxiredoxins (PRDXs) which are found in the extracellular fluid, the cytoplasm, the mitochondria and in the nucleus with functions that include antioxidation, signaling and transcriptional control, among others. The importance of thioredoxin family proteins in neurodegenerative diseases is gaining relevance because some of these proteins have demonstrated an important role in the central nervous system by mediating neuroprotection against oxidative stress, contributing to mitochondrial function and regulating gene expression. Specifically, in the context of Friedreich’s ataxia (FRDA), thioredoxin family proteins may have a special role in the regulation of Nrf2 expression and function, in Fe-S cluster metabolism, controlling the expression of genes located at the iron-response element (IRE) and probably regulating ferroptosis. Therefore, comprehension of the mechanisms that closely link thioredoxin family proteins with cellular processes affected in FRDA will serve as a cornerstone to design improved therapeutic strategies.
Collapse
Affiliation(s)
- Marta Seco-Cervera
- Centre for Biomedical Research on Rare Diseases (CIBERER), 46010 Valencia, Spain; (M.S.-C.); (P.G.-C.); (F.V.P.)
- Department of Physiology, Faculty of Medicine and Dentistry, Universitat de València (UV), 46010 Valencia, Spain
- Biomedical Research Institute INCLIVA, 46010 Valencia, Spain
| | - Pilar González-Cabo
- Centre for Biomedical Research on Rare Diseases (CIBERER), 46010 Valencia, Spain; (M.S.-C.); (P.G.-C.); (F.V.P.)
- Department of Physiology, Faculty of Medicine and Dentistry, Universitat de València (UV), 46010 Valencia, Spain
- Biomedical Research Institute INCLIVA, 46010 Valencia, Spain
| | - Federico V. Pallardó
- Centre for Biomedical Research on Rare Diseases (CIBERER), 46010 Valencia, Spain; (M.S.-C.); (P.G.-C.); (F.V.P.)
- Department of Physiology, Faculty of Medicine and Dentistry, Universitat de València (UV), 46010 Valencia, Spain
- Biomedical Research Institute INCLIVA, 46010 Valencia, Spain
| | - Carlos Romá-Mateo
- Centre for Biomedical Research on Rare Diseases (CIBERER), 46010 Valencia, Spain; (M.S.-C.); (P.G.-C.); (F.V.P.)
- Department of Physiology, Faculty of Medicine and Dentistry, Universitat de València (UV), 46010 Valencia, Spain
- Biomedical Research Institute INCLIVA, 46010 Valencia, Spain
- Correspondence: (C.R.-M.); (J.L.G.-G.); Tel.: +34-963-864-646 (C.R.-M. & J.L.G.-G.)
| | - José Luis García-Giménez
- Centre for Biomedical Research on Rare Diseases (CIBERER), 46010 Valencia, Spain; (M.S.-C.); (P.G.-C.); (F.V.P.)
- Department of Physiology, Faculty of Medicine and Dentistry, Universitat de València (UV), 46010 Valencia, Spain
- Biomedical Research Institute INCLIVA, 46010 Valencia, Spain
- Correspondence: (C.R.-M.); (J.L.G.-G.); Tel.: +34-963-864-646 (C.R.-M. & J.L.G.-G.)
| |
Collapse
|
57
|
Mitochondrial Translocation of DJ-1 Is Mediated by Grp75: Implication in Cardioprotection of Resveratrol Against Hypoxia/Reoxygenation-Induced Oxidative Stress. J Cardiovasc Pharmacol 2020; 75:305-313. [PMID: 32040033 DOI: 10.1097/fjc.0000000000000805] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Resveratrol (Res) was recently reported to ameliorate hypoxia/reoxygenation (H/R)-caused oxidative stress in H9c2 cardiomyocytes through promoting the mitochondrial translocation of DJ-1 protein and subsequently preserving the activity of mitochondrial complex I. However, it is noteworthy that DJ-1 possesses no mitochondria-targeting sequence. Therefore, how Res induces DJ-1 mitochondrial translocation is an important and interesting question for further exploration. Glucose-regulated protein 75 (Grp75), whose N-terminus contains a 51-amino acid long mitochondrial-targeting signal peptide, is a cytoprotective chaperone that partakes in mitochondrial import of several proteins. Here, the contribution of Grp75 to mitochondrial import of DJ-1 by Res was investigated in a cellular model of H/R. Our results showed that Res upregulated the expression of DJ-1 protein, enhanced the interaction of DJ-1 and Grp75, and promoted DJ-1 translocation to mitochondria from cytosol in H9c2 cardiomyocytes undergoing H/R. Importantly, knockdown of Grp75 markedly reduced the interaction of DJ-1 with Grp75 and subsequent DJ-1 mitochondrial translocation induced by Res. Furthermore, Res pretreatment promoted the association of DJ-1 with ND1 and NDUFA4 subunits of complex I, preserved the activity of complex I, decreased mitochondria-derived reactive oxygen species production, and eventually ameliorated H/R-caused oxidative stress damage. Intriguingly, these effects were largely prevented also by small interfering RNA targeting Grp75. Overall, these results suggested that Grp75 interacts with DJ-1 to facilitate its translocation from cytosol to mitochondria, which is required for Res-mediated preservation of mitochondria complex I and cardioprotection from H/R-caused oxidative stress injury.
Collapse
|
58
|
Yu W, Wang S, Cao D, Rui H, Liu C, Sheng Y, Sun Y, Zhang J, Xu J, Jiang D. Insight into an Oxidative DNA-Cleaving DNAzyme: Multiple Cofactors, the Catalytic Core Map and a Highly Efficient Variant. iScience 2020; 23:101555. [PMID: 33083724 PMCID: PMC7522124 DOI: 10.1016/j.isci.2020.101555] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 08/24/2020] [Accepted: 09/09/2020] [Indexed: 02/07/2023] Open
Abstract
An oxidative DNA-cleaving DNAzyme (PL) employs a double-cofactor model “X/Cu2+” for catalysis. Herein, we verified that reduced nicotinamide adenine dinucleotide (NADH), flavin mononucleotide, cysteine, dithiothreitol, catechol, resorcinol, hydroquinone, phloroglucinol, o-phenylenediamine, 3,3′,5,5'-tetramethylbenzidine, and hydroxylamine acted as cofactor X. According to their structural similarities or fluorescence property, we further confirmed that reduced nicotinamide adenine dinucleotide phosphate (NADPH), 2-mercaptoethanol, dopamine, chlorogenic acid, resveratrol, and 5-carboxyfluorescein also functioned as cofactor X. Superoxide anions might be the commonality behind these cofactors. We subsequently determined the conservative change of individual nucleotides in the catalytic core under four different cofactor X. The nucleotides A4 and C5 are highly conserved, whereas the conservative levels of other nucleotides are dependent on the types of cofactor X. Moreover, we observed that the minor change in the PL's secondary structure affects electrophoretic mobility. Finally, we characterized a highly efficient variant T3G and converted its double-cofactor NADH/Cu2+ to sole-cofactor NADH. An oxidative cleavage DNAzyme works with various cofactor X Catalytic nucleotide conservation fluctuates with different cofactor X The PL DNAzyme's minor secondary structure change affects electrophoretic mobility Double-cofactor model of the variant T3G can be converted to sole-cofactor model
Collapse
Affiliation(s)
- Wenqian Yu
- Key Lab for Molecular Enzymology & Engineering of the Ministry of Education, School of Life Sciences, Jilin University, 2699# Qianjin Street, Changchun 130012, China
| | - Shijin Wang
- Key Lab for Molecular Enzymology & Engineering of the Ministry of Education, School of Life Sciences, Jilin University, 2699# Qianjin Street, Changchun 130012, China
| | - Dongling Cao
- Key Lab for Molecular Enzymology & Engineering of the Ministry of Education, School of Life Sciences, Jilin University, 2699# Qianjin Street, Changchun 130012, China
| | - Hongyue Rui
- Key Lab for Molecular Enzymology & Engineering of the Ministry of Education, School of Life Sciences, Jilin University, 2699# Qianjin Street, Changchun 130012, China
| | - Chengcheng Liu
- Key Lab for Molecular Enzymology & Engineering of the Ministry of Education, School of Life Sciences, Jilin University, 2699# Qianjin Street, Changchun 130012, China
| | - Yongjie Sheng
- Key Lab for Molecular Enzymology & Engineering of the Ministry of Education, School of Life Sciences, Jilin University, 2699# Qianjin Street, Changchun 130012, China
| | - Yanhong Sun
- Key Lab for Molecular Enzymology & Engineering of the Ministry of Education, School of Life Sciences, Jilin University, 2699# Qianjin Street, Changchun 130012, China
| | - Jin Zhang
- Key Lab for Molecular Enzymology & Engineering of the Ministry of Education, School of Life Sciences, Jilin University, 2699# Qianjin Street, Changchun 130012, China
| | - Jiacui Xu
- College of Animal Sciences, Jilin University, 5333# Xi'an Road, Changchun 130062, China
- Corresponding author
| | - Dazhi Jiang
- Key Lab for Molecular Enzymology & Engineering of the Ministry of Education, School of Life Sciences, Jilin University, 2699# Qianjin Street, Changchun 130012, China
- Corresponding author
| |
Collapse
|
59
|
Hypoxia-mediated regulation of mitochondrial transcription factors in renal epithelial cells: implications for hypertensive renal physiology. Hypertens Res 2020; 44:154-167. [PMID: 32917968 DOI: 10.1038/s41440-020-00539-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/19/2020] [Accepted: 07/23/2020] [Indexed: 02/07/2023]
Abstract
Kidneys have a high resting metabolic rate and low partial pressure of oxygen due to enhanced mitochondrial oxygen consumption and ATP production needed for active solute transport. Heightened mitochondrial activity leads to progressively increasing hypoxia from the renal cortex to the renal medulla. Renal hypoxia is prominent in hypertensive rats due to increased sodium reabsorption within the nephrons, which demands higher energy production by oxidative phosphorylation (OXPHOS). Consequently, spontaneously hypertensive rats (SHR) display greater oxygen deficiency (hypoxia) than normotensive Wistar Kyoto rats (WKY). Here, we sought to investigate the expression of key proteins for mitochondrial biogenesis in SHR and WKY, and study the regulation of mitochondrial transcription factors (mtTFs) under in vitro hypoxic conditions in renal epithelial cells. We report that renal expressions of hypoxia-inducible factor-1-alpha (HIF-1α), peroxisome proliferator-activated receptor-gamma coactivator-1-alpha (PGC-1α), mtTFs, and OXPHOS proteins are elevated in SHR compared to WKY. In addition, our experiments in cultured kidney cells demonstrate that acute hypoxia augments the expression of these genes. Furthermore, we show that the transcripts of HIF-1α and mtTFs are positively correlated in various human tissues. We reveal, for the first time to our knowledge, that HIF-1α transactivates mtTF genes by direct interaction with their promoters in rat kidney epithelial cells (NRK-52E) under acute hypoxia. Concomitant increases in the mitochondrial DNA and RNA, and OXPHOS proteins are observed. Taken together, this study suggests that hypoxia within the renal epithelial cells may enhance mitochondrial function to meet the energy demand in proximal tubular cells during prehypertensive stages in kidneys of young SHR.
Collapse
|
60
|
Grivennikova VG, Gladyshev GV, Vinogradov AD. Deactivation of mitochondrial NADH:ubiquinone oxidoreductase (respiratory complex I): Extrinsically affecting factors. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2020; 1861:148207. [DOI: 10.1016/j.bbabio.2020.148207] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 03/13/2020] [Accepted: 04/14/2020] [Indexed: 12/14/2022]
|
61
|
Riquelme SA, Lozano C, Moustafa AM, Liimatta K, Tomlinson KL, Britto C, Khanal S, Gill SK, Narechania A, Azcona-Gutiérrez JM, DiMango E, Saénz Y, Planet P, Prince A. CFTR-PTEN-dependent mitochondrial metabolic dysfunction promotes Pseudomonas aeruginosa airway infection. Sci Transl Med 2020; 11:11/499/eaav4634. [PMID: 31270271 DOI: 10.1126/scitranslmed.aav4634] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 02/12/2019] [Accepted: 06/12/2019] [Indexed: 12/12/2022]
Abstract
Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) is a tumor suppressor best known for regulating cell proliferation and metabolism. PTEN forms a complex with the cystic fibrosis (CF) transmembrane conductance regulator (CFTR) at the plasma membrane, and this complex is known to be functionally impaired in CF. Here, we demonstrated that the combined effect of PTEN and CFTR dysfunction stimulates mitochondrial activity, resulting in excessive release of succinate and reactive oxygen species. This environment promoted the colonization of the airway by Pseudomonas aeruginosa, bacteria that preferentially metabolize succinate, and stimulated an anti-inflammatory host response dominated by immune-responsive gene 1 (IRG1) and itaconate. The recruitment of myeloid cells induced by these strains was inefficient in clearing the infection and increased numbers of phagocytes accumulated under CFTR-PTEN axis dysfunction. This central metabolic defect in mitochondrial function due to impaired PTEN activity contributes to P. aeruginosa infection in CF.
Collapse
Affiliation(s)
| | - Carmen Lozano
- Area de Microbiología Molecular, Centro de Investigación Biomédica de la Rioja (CIBIR), Microbiología Molecular, Logroño, LG 26006, Spain
| | - Ahmed M Moustafa
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania and Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Kalle Liimatta
- Department of Pediatrics, Columbia University, New York, NY 10032, USA
| | - Kira L Tomlinson
- Department of Pediatrics, Columbia University, New York, NY 10032, USA
| | - Clemente Britto
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Sara Khanal
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Simren K Gill
- Department of Pediatrics, Columbia University, New York, NY 10032, USA
| | | | - Jose M Azcona-Gutiérrez
- Departamento de Diagnóstico Biomédico. Laboratorio de Microbiología, Hospital San Pedro, Logroño, LG 26006, Spain
| | - Emily DiMango
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Yolanda Saénz
- Area de Microbiología Molecular, Centro de Investigación Biomédica de la Rioja (CIBIR), Microbiología Molecular, Logroño, LG 26006, Spain
| | - Paul Planet
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania and Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Alice Prince
- Department of Pediatrics, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
62
|
Globally Abundant " Candidatus Udaeobacter" Benefits from Release of Antibiotics in Soil and Potentially Performs Trace Gas Scavenging. mSphere 2020; 5:5/4/e00186-20. [PMID: 32641424 PMCID: PMC7343977 DOI: 10.1128/msphere.00186-20] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Verrucomicrobia affiliated with "Candidatus Udaeobacter" belong to the most abundant soil bacteria worldwide. Although the synthesis of antibiotics presumably evolved in soil, and environmental pollution with antimicrobials increases, the impact of these complex molecules on "Ca Udaeobacter" remains to be elucidated. In this study, we demonstrate that "Ca. Udaeobacter" representatives residing in grassland as well as forest soil ecosystems show multidrug resistance and even take advantage of antibiotics release. Soils treated with up to six different antibiotics exhibited a higher "Ca. Udaeobacter" abundance than corresponding controls after 3, 8, and 20 days of incubation. In this context, we provide evidence that "Ca. Udaeobacter" representatives may utilize nutrients which are released due to antibiotic-driven lysis of other soil microbes and thereby reduce energetically expensive synthesis of required biomolecules. Moreover, genomic analysis revealed the presence of genes conferring resistance to multiple classes of antibiotics and indicated that "Ca. Udaeobacter" representatives most likely oxidize the trace gas H2 to generate energy. This energy might be required for long-term persistence in terrestrial habitats, as already suggested for other dominant soil bacteria. Our study illustrates, for the first time, that globally abundant "Ca. Udaeobacter" benefits from release of antibiotics, which confers advantages over other soil bacteria and represents a so-far overlooked fundamental lifestyle feature of this poorly characterized verrucomicrobial genus. Furthermore, our study suggests that "Ca. Udaeobacter" representatives can utilize H2 as an alternative electron donor.IMPORTANCE Soil bacteria have been investigated for more than a century, but one of the most dominant terrestrial groups on Earth, "Candidatus Udaeobacter," remains elusive and largely unexplored. Its natural habitat is considered a major reservoir of antibiotics, which directly or indirectly impact phylogenetically diverse microorganisms. Here, we found that "Ca. Udaeobacter" representatives exhibit multidrug resistance and not only evade harmful effects of antimicrobials but even benefit from antibiotic pressure in soil. Therefore, "Ca. Udaeobacter" evidently affects the composition of soil resistomes worldwide and might represent a winner of rising environmental pollution with antimicrobials. In addition, our study indicates that "Ca. Udaeobacter" representatives utilize H2 and thereby contribute to global hydrogen cycling. The here-reported findings provide insights into elementary lifestyle features of "Ca. Udaeobacter," potentially contributing to its successful global dissemination.
Collapse
|
63
|
Panov J, Simchi L, Feuermann Y, Kaphzan H. Bioinformatics Analyses of the Transcriptome Reveal Ube3a-Dependent Effects on Mitochondrial-Related Pathways. Int J Mol Sci 2020; 21:ijms21114156. [PMID: 32532103 PMCID: PMC7312912 DOI: 10.3390/ijms21114156] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/07/2020] [Accepted: 06/08/2020] [Indexed: 12/14/2022] Open
Abstract
The UBE3A gene encodes the ubiquitin E3-ligase protein, UBE3A, which is implicated in severe neurodevelopmental disorders. Lack of UBE3A expression results in Angelman syndrome, while UBE3A overexpression, due to genomic 15q duplication, results in autism. The cellular roles of UBE3A are not fully understood, yet a growing body of evidence indicates that these disorders involve mitochondrial dysfunction and increased oxidative stress. We utilized bioinformatics approaches to delineate the effects of murine Ube3a deletion on the expression of mitochondrial-related genes and pathways. For this, we generated an mRNA sequencing dataset from mouse embryonic fibroblasts (MEFs) in which both alleles of Ube3a gene were deleted and their wild-type controls. Since oxidative stress and mitochondrial dysregulation might not be exhibited in the resting baseline state, we also activated mitochondrial functioning in the cells of these two genotypes using TNFα application. Transcriptomes of the four groups of MEFs, Ube3a+/+ and Ube3a-/-, with or without the application of TNFα, were analyzed using various bioinformatics tools and machine learning approaches. Our results indicate that Ube3a deletion affects the gene expression profiles of mitochondrial-associated pathways. We further confirmed these results by analyzing other publicly available human transcriptome datasets of Angelman syndrome and 15q duplication syndrome.
Collapse
|
64
|
Systems Analysis of NADH Dehydrogenase Mutants Reveals Flexibility and Limits of Pseudomonas taiwanensis VLB120's Metabolism. Appl Environ Microbiol 2020; 86:AEM.03038-19. [PMID: 32245760 DOI: 10.1128/aem.03038-19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 03/18/2020] [Indexed: 12/14/2022] Open
Abstract
Obligate aerobic organisms rely on a functional electron transport chain for energy conservation and NADH oxidation. Because of this essential requirement, the genes of this pathway are likely constitutively and highly expressed to avoid a cofactor imbalance and energy shortage under fluctuating environmental conditions. We here investigated the essentiality of the three NADH dehydrogenases of the respiratory chain of the obligate aerobe Pseudomonas taiwanensis VLB120 and the impact of the knockouts of corresponding genes on its physiology and metabolism. While a mutant lacking all three NADH dehydrogenases seemed to be nonviable, the single or double knockout mutant strains displayed no, or only a weak, phenotype. Only the mutant deficient in both type 2 dehydrogenases showed a clear phenotype with biphasic growth behavior and a strongly reduced growth rate in the second phase. In-depth analyses of the metabolism of the generated mutants, including quantitative physiological experiments, transcript analysis, proteomics, and enzyme activity assays revealed distinct responses to type 2 and type 1 dehydrogenase deletions. An overall high metabolic flexibility enables P. taiwanensis to cope with the introduced genetic perturbations and maintain stable phenotypes, likely by rerouting of metabolic fluxes. This metabolic adaptability has implications for biotechnological applications. While the phenotypic robustness is favorable in large-scale applications with inhomogeneous conditions, the possible versatile redirecting of carbon fluxes upon genetic interventions can thwart metabolic engineering efforts.IMPORTANCE While Pseudomonas has the capability for high metabolic activity and the provision of reduced redox cofactors important for biocatalytic applications, exploitation of this characteristic might be hindered by high, constitutive activity of and, consequently, competition with the NADH dehydrogenases of the respiratory chain. The in-depth analysis of NADH dehydrogenase mutants of Pseudomonas taiwanensis VLB120 presented here provides insight into the phenotypic and metabolic response of this strain to these redox metabolism perturbations. This high degree of metabolic flexibility needs to be taken into account for rational engineering of this promising biotechnological workhorse toward a host with a controlled and efficient supply of redox cofactors for product synthesis.
Collapse
|
65
|
Langlois JP, Millette G, Guay I, Dubé-Duquette A, Chamberland S, Jacques PÉ, Rodrigue S, Bouarab K, Marsault É, Malouin F. Bactericidal Activity of the Bacterial ATP Synthase Inhibitor Tomatidine and the Combination of Tomatidine and Aminoglycoside Against Persistent and Virulent Forms of Staphylococcus aureus. Front Microbiol 2020; 11:805. [PMID: 32431678 PMCID: PMC7216300 DOI: 10.3389/fmicb.2020.00805] [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: 12/13/2019] [Accepted: 04/03/2020] [Indexed: 12/15/2022] Open
Abstract
Tomatidine (TO), a steroid alkaloid, exerts a strong bactericidal activity on the infection-persistent phenotype of Staphylococcus aureus, the small-colony variant (SCV), with a minimal inhibitory concentration (MIC) of 0.06 μg/ml. Also, the combination of TO to an aminoglycoside (AMG) shows a strong synergistic effect against prototypical (WT) S. aureus (MIC 0.06 μg/ml), which is otherwise unaffected by TO alone (MIC > 128 μg/ml). We have recently established that the ATP synthase (subunit AtpE) was the molecular target of TO and that TO reduces the production of ATP in S. aureus. The purpose of this study was to understand how TO and the TO-AMG combination exert bactericidal activities against S. aureus SCV and WT strains, respectively. The impact of TO and of the TO-gentamicin (GEN) combination on the membrane potential and generation of reactive oxygen species (ROS) were determined using florescent probes. GEN uptake in WT was assessed in the presence of TO. Virulence of SCV and WT strains as well as of in vitro-selected mutants showing resistance to TO or the TO-GEN combination was evaluated in a murine thigh infection model. TO causes a reduction in membrane potential in both WT and SCV, but significant amounts of ROS are only produced in SCVs. Besides, the presence of TO improves the uptake of GEN by the WT strain and the combination TO-GEN generated 2.5-folds more ROS in WT, compared to that induced by GEN alone. Under anaerobic conditions, WT adopts a fermentative slow-growth phenotype and becomes susceptible to TO even if used alone. In vivo, TO- or TO-GEN-resistant strains were significantly altered in their ability to colonize tissues. These results shed light on the mechanism of action of TO and its synergy with AMGs against S. aureus WT. TO bactericidal activity against SCVs is attributable to both a critical drop in the membrane potential accompanied by a substantial ROS production. In the WT, TO helps GEN uptake and ROS is also important for the synergy. Acquiring resistance to TO significantly impairs virulence. The residual ATP synthase activity of SCVs might represent the Achilles’ heel of persistent S. aureus.
Collapse
Affiliation(s)
- Jean-Philippe Langlois
- Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Guillaume Millette
- Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Isabelle Guay
- Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Alexis Dubé-Duquette
- Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Suzanne Chamberland
- Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Pierre-Étienne Jacques
- Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Sébastien Rodrigue
- Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Kamal Bouarab
- Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Éric Marsault
- Département de Pharmacologie et Physiologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - François Malouin
- Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
66
|
Awasthi K, Chang FL, Hsieh PY, Hsu HY, Ohta N. Characterization of endogenous fluorescence in nonsmall lung cancerous cells: A comparison with nonmalignant lung normal cells. JOURNAL OF BIOPHOTONICS 2020; 13:e201960210. [PMID: 32067342 DOI: 10.1002/jbio.201960210] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/11/2020] [Accepted: 02/13/2020] [Indexed: 06/10/2023]
Abstract
Monitoring fluorescence properties of endogenous fluorophores such as nicotinamide adenine dinucleotide (NADH) and flavin adenine dinucleotide (FAD) in normal and cancerous cells provide substantial information noninvasively on biochemical and biophysical aspects of metabolic dysfunction of cancerous cells. Time-resolved spectral profiles and fluorescence lifetime images of NADH and FAD were obtained in human lung nonsmall carcinomas (H661 and A549) and normal lung cells (MRC-5). Both fluorophores show the fast and slowly decaying emission components upon pulsed excitation, and fluorescence spectra of NADH and FAD show blue- and red-shifts, respectively, during their decay. All identified lifetime components of NADH and FAD were found to be shorter in cancerous cells than in normal cells, no matter how they were measured under different extra-cellular conditions (cells suspended in cuvette and cells attached on glass substrate), indicating that the changes in metabolism likely altered the subcellular milieu and potentially also affected the interaction of NADH and FAD with enzymes to which these cofactors were bound. The intensity ratio of NADH and FAD of cancerous cells was also shown to be larger than that of normal cells.
Collapse
Affiliation(s)
- Kamlesh Awasthi
- Department of Applied Chemistry and Institute of Molecular Science, National Chiao Tung University, Hsinchu, Taiwan
- Center for Emergent Functional Matter Science, National Chiao Tung University, Hsinchu, Taiwan
| | - Feng-Lin Chang
- Department of Applied Chemistry and Institute of Molecular Science, National Chiao Tung University, Hsinchu, Taiwan
| | - Pei-Ying Hsieh
- Center for Emergent Functional Matter Science, National Chiao Tung University, Hsinchu, Taiwan
| | - Hsin-Yun Hsu
- Department of Applied Chemistry and Institute of Molecular Science, National Chiao Tung University, Hsinchu, Taiwan
- Center for Emergent Functional Matter Science, National Chiao Tung University, Hsinchu, Taiwan
| | - Nobuhiro Ohta
- Department of Applied Chemistry and Institute of Molecular Science, National Chiao Tung University, Hsinchu, Taiwan
- Center for Emergent Functional Matter Science, National Chiao Tung University, Hsinchu, Taiwan
| |
Collapse
|
67
|
A salvage pathway maintains highly functional respiratory complex I. Nat Commun 2020; 11:1643. [PMID: 32242014 PMCID: PMC7118099 DOI: 10.1038/s41467-020-15467-7] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 03/05/2020] [Indexed: 02/07/2023] Open
Abstract
Regulation of the turnover of complex I (CI), the largest mitochondrial respiratory chain complex, remains enigmatic despite huge advancement in understanding its structure and the assembly. Here, we report that the NADH-oxidizing N-module of CI is turned over at a higher rate and largely independently of the rest of the complex by mitochondrial matrix protease ClpXP, which selectively removes and degrades damaged subunits. The observed mechanism seems to be a safeguard against the accumulation of dysfunctional CI arising from the inactivation of the N-module subunits due to attrition caused by its constant activity under physiological conditions. This CI salvage pathway maintains highly functional CI through a favorable mechanism that demands much lower energetic cost than de novo synthesis and reassembly of the entire CI. Our results also identify ClpXP activity as an unforeseen target for therapeutic interventions in the large group of mitochondrial diseases characterized by the CI instability. Maintenance and quality control of the mitochondrial respiratory chain complexes responsible for bulk energy production are unclear. Here, the authors show that the mitochondrial protease ClpXP is required for the rapid turnover of the core N-module of respiratory complex I, which happens independently of other modules in the complex.
Collapse
|
68
|
PGC-1 α, Inflammation, and Oxidative Stress: An Integrative View in Metabolism. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:1452696. [PMID: 32215168 PMCID: PMC7085407 DOI: 10.1155/2020/1452696] [Citation(s) in RCA: 325] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 02/20/2020] [Indexed: 02/07/2023]
Abstract
Peroxisome proliferator-activated receptor-γ coactivator (PGC)-1α is a transcriptional coactivator described as a master regulator of mitochondrial biogenesis and function, including oxidative phosphorylation and reactive oxygen species detoxification. PGC-1α is highly expressed in tissues with high energy demands, and it is clearly associated with the pathogenesis of metabolic syndrome and its principal complications including obesity, type 2 diabetes mellitus, cardiovascular disease, and hepatic steatosis. We herein review the molecular pathways regulated by PGC-1α, which connect oxidative stress and mitochondrial metabolism with inflammatory response and metabolic syndrome. PGC-1α regulates the expression of mitochondrial antioxidant genes, including manganese superoxide dismutase, catalase, peroxiredoxin 3 and 5, uncoupling protein 2, thioredoxin 2, and thioredoxin reductase and thus prevents oxidative injury and mitochondrial dysfunction. Dysregulation of PGC-1α alters redox homeostasis in cells and exacerbates inflammatory response, which is commonly accompanied by metabolic disturbances. During inflammation, low levels of PGC-1α downregulate mitochondrial antioxidant gene expression, induce oxidative stress, and promote nuclear factor kappa B activation. In metabolic syndrome, which is characterized by a chronic low grade of inflammation, PGC-1α dysregulation modifies the metabolic properties of tissues by altering mitochondrial function and promoting reactive oxygen species accumulation. In conclusion, PGC-1α acts as an essential node connecting metabolic regulation, redox control, and inflammatory pathways, and it is an interesting therapeutic target that may have significant benefits for a number of metabolic diseases.
Collapse
|
69
|
Wu D, Ma Y, Cao Y, Zhang T. Mitochondrial toxicity of nanomaterials. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 702:134994. [PMID: 31715400 DOI: 10.1016/j.scitotenv.2019.134994] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/11/2019] [Accepted: 10/14/2019] [Indexed: 05/11/2023]
Abstract
In recent years, nanomaterials have been widely applied in electronics, food, biomedicine and other fields, resulting in increased human exposure and consequent research focus on their biological and toxic effects. Mitochondria, the main target organelle for nanomaterials (NM), play a critical role in their toxic activities. Several studies to date have shown that nanomaterials cause alterations in mitochondrial morphology, mitochondrial membrane potential, opening of the mitochondrial permeability transition pore (MPTP) and mitochondrial respiratory function, and promote cytochrome C release. An earlier mitochondrial toxicity study of NMs additionally reported induction of mitochondrial dynamic changes. Here, we have reviewed the mitochondrial toxicity of NMs and provided a scientific basis for the contribution of mitochondria to the toxicological effects of different NMs along with approaches to reduce mitochondrial and, consequently, overall toxicity of NMs.
Collapse
Affiliation(s)
- Daming Wu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Ying Ma
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Yuna Cao
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Ting Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China.
| |
Collapse
|
70
|
Molecular Mechanism Underlying Hypoxic Preconditioning-Promoted Mitochondrial Translocation of DJ-1 in Hypoxia/Reoxygenation H9c2 Cells. Molecules 2019; 25:molecules25010071. [PMID: 31878239 PMCID: PMC6983240 DOI: 10.3390/molecules25010071] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/17/2019] [Accepted: 12/20/2019] [Indexed: 01/06/2023] Open
Abstract
DJ-1 was recently reported to be involved in the cardioprotection of hypoxic preconditioning (HPC) against hypoxia/reoxygenation (H/R)-induced oxidative stress damage, by preserving mitochondrial complex I activity and, subsequently, inhibiting mitochondrial reactive oxygen species (ROS) generation. However, the molecular mechanism by which HPC enables mitochondrial translocation of DJ-1, which has no mitochondria-targeting sequence, to preserve mitochondrial complex I, is largely unknown. In this study, co-immunoprecipitation data showed that DJ-1 was associated with glucose-regulated protein 75 (Grp75), and this association was significantly enhanced after HPC. Immunofluorescence imaging and Western blot analysis showed that HPC substantially enhanced the translocation of DJ-1 from cytosol to mitochondria in H9c2 cells subjected to H/R, which was mimicked by DJ-1 overexpression induced by pFlag-DJ-1 transfection. Importantly, knockdown of Grp75 markedly reduced the mitochondrial translocation of DJ-1 induced by HPC and pFlag-DJ-1 transfection. Moreover, HPC promoted the association of DJ-1 with mitochondrial complex I subunits ND1 and NDUFA4, improved complex I activity, and inhibited mitochondria-derived ROS production and subsequent oxidative stress damage after H/R, which was also mimicked by pFlag-DJ-1 transfection. Intriguingly, these effects of HPC and pFlag-DJ-1 transfection were also prevented by Grp75 knockdown. In conclusion, these results indicated that HPC promotes the translocation of DJ-1 from cytosol to mitochondria in a Grp75-dependent manner and Grp75 is required for DJ-1-mediated protection of HPC on H/R-induced mitochondrial complex I defect and subsequent oxidative stress damage.
Collapse
|
71
|
Pohl EE, Jovanovic O. The Role of Phosphatidylethanolamine Adducts in Modification of the Activity of Membrane Proteins under Oxidative Stress. Molecules 2019; 24:E4545. [PMID: 31842328 PMCID: PMC6943717 DOI: 10.3390/molecules24244545] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 12/11/2022] Open
Abstract
Reactive oxygen species (ROS) and their derivatives, reactive aldehydes (RAs), have been implicated in the pathogenesis of many diseases, including metabolic, cardiovascular, and inflammatory disease. Understanding how RAs can modify the function of membrane proteins is critical for the design of therapeutic approaches in the above-mentioned pathologies. Over the last few decades, direct interactions of RA with proteins have been extensively studied. Yet, few studies have been performed on the modifications of membrane lipids arising from the interaction of RAs with the lipid amino group that leads to the formation of adducts. It is even less well understood how various multiple adducts affect the properties of the lipid membrane and those of embedded membrane proteins. In this short review, we discuss a crucial role of phosphatidylethanolamine (PE) and PE-derived adducts as mediators of RA effects on membrane proteins. We propose potential PE-mediated mechanisms that explain the modulation of membrane properties and the functions of membrane transporters, channels, receptors, and enzymes. We aim to highlight this new area of research and to encourage a more nuanced investigation of the complex nature of the new lipid-mediated mechanism in the modification of membrane protein function under oxidative stress.
Collapse
Affiliation(s)
- Elena E. Pohl
- Institute of Physiology, Pathophysiology and Biophysics, Department of Biomedical Sciences, University of Veterinary Medicine, Vienna A-1210, Austria
| | - Olga Jovanovic
- Institute of Physiology, Pathophysiology and Biophysics, Department of Biomedical Sciences, University of Veterinary Medicine, Vienna A-1210, Austria
| |
Collapse
|
72
|
Abstract
Using an unbiased genetic screen, To et al. map genes that enhance or suppress growth defects in response to different mitochondrial inhibitors to model mitochondrial disease. The findings have novel implications for the interconnectivity of bioenergetic pathways, and suggest a provocative strategy to treat primary mitochondrial disorders.
Collapse
|
73
|
Trisolini L, Gambacorta N, Gorgoglione R, Montaruli M, Laera L, Colella F, Volpicella M, De Grassi A, Pierri CL. FAD/NADH Dependent Oxidoreductases: From Different Amino Acid Sequences to Similar Protein Shapes for Playing an Ancient Function. J Clin Med 2019; 8:jcm8122117. [PMID: 31810296 PMCID: PMC6947548 DOI: 10.3390/jcm8122117] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 11/11/2019] [Accepted: 11/18/2019] [Indexed: 12/29/2022] Open
Abstract
Flavoprotein oxidoreductases are members of a large protein family of specialized dehydrogenases, which include type II NADH dehydrogenase, pyridine nucleotide-disulphide oxidoreductases, ferredoxin-NAD+ reductases, NADH oxidases, and NADH peroxidases, playing a crucial role in the metabolism of several prokaryotes and eukaryotes. Although several studies have been performed on single members or protein subgroups of flavoprotein oxidoreductases, a comprehensive analysis on structure-function relationships among the different members and subgroups of this great dehydrogenase family is still missing. Here, we present a structural comparative analysis showing that the investigated flavoprotein oxidoreductases have a highly similar overall structure, although the investigated dehydrogenases are quite different in functional annotations and global amino acid composition. The different functional annotation is ascribed to their participation in species-specific metabolic pathways based on the same biochemical reaction, i.e., the oxidation of specific cofactors, like NADH and FADH2. Notably, the performed comparative analysis sheds light on conserved sequence features that reflect very similar oxidation mechanisms, conserved among flavoprotein oxidoreductases belonging to phylogenetically distant species, as the bacterial type II NADH dehydrogenases and the mammalian apoptosis-inducing factor protein, until now retained as unique protein entities in Bacteria/Fungi or Animals, respectively. Furthermore, the presented computational analyses will allow consideration of FAD/NADH oxidoreductases as a possible target of new small molecules to be used as modulators of mitochondrial respiration for patients affected by rare diseases or cancer showing mitochondrial dysfunction, or antibiotics for treating bacterial/fungal/protista infections.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Anna De Grassi
- Correspondence: (A.D.G.); or (C.L.P.); Tel.: +39-080-544-3614 (A.D.G. & C.L.P.); Fax: +39-080-544-2770 (A.D.G. & C.L.P.)
| | - Ciro Leonardo Pierri
- Correspondence: (A.D.G.); or (C.L.P.); Tel.: +39-080-544-3614 (A.D.G. & C.L.P.); Fax: +39-080-544-2770 (A.D.G. & C.L.P.)
| |
Collapse
|
74
|
Abstract
Complexes I to IV, with the exception of Complex II, are redox-driven proton pumps that convert redox energy of oxygen reduction to proton gradient across the mitochondrial or bacterial membrane; in turn, the created electrochemical gradient drives the adenosine triphosphate synthesis in the cells by utilizing complex V of the chain. Here we address a general question of the efficiency of such enzymes, considering them as molecular machines that couple endergonic and exergonic reactions and converting one form of free energy into another. One well-known example of the efficiency is given by Carnot's theorem for heat engines. Here we extend the concept to respiratory enzymes and specifically focus on the proton pumping by Complex I of the respiratory chain, nicotinamide adenine dinucleotide dehydrogenase. To discuss the efficiency issues, we develop a model of enzyme kinetics, which generalizes the Michaelis-Menten model. Our model includes several substrates and products and, in general, can be considered as Generalized Michaelis-Menten Kinetic model. The model might be useful for describing complex enzyme kinetics, regardless of the efficiency issues that are addressed in this paper.
Collapse
Affiliation(s)
- Alexei A Stuchebrukhov
- Department of Chemistry , University of California at Davis , Davis , California 95616 , United States
| |
Collapse
|
75
|
Müller TE, Nunes MEM, Rodrigues NR, Fontana BD, Hartmann DD, Franco JL, Rosemberg DB. Neurochemical mechanisms underlying acute and chronic ethanol-mediated responses in zebrafish: The role of mitochondrial bioenergetics. Neurochem Int 2019; 131:104584. [PMID: 31654679 DOI: 10.1016/j.neuint.2019.104584] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 10/02/2019] [Accepted: 10/18/2019] [Indexed: 11/18/2022]
Abstract
Ethanol (EtOH) is a socially-accepted drug, whose consumption is a risk factor for non-intentional injuries, development of pathologies, and addiction. In the brain, EtOH affects redox signaling and increases reactive oxygen species (ROS) production after acute and chronic exposures. Here, using a high-resolution respirometry assay, we investigated whether changes in mitochondrial bioenergetics play a role in both acute and chronic EtOH-mediated neurochemical responses in zebrafish. For the first time, we showed that acute and chronic EtOH exposures differently affect brain mitochondrial function. Acutely, EtOH stimulated mitochondrial respiration through increased baseline state, CI-mediated OXPHOS, OXPHOS capacity, OXPHOS coupling efficiency, bioenergetic efficiency, and ROX/ETS ratio. Conversely, EtOH chronically decreased baseline respiration, complex I- and II-mediated ETS, as well as increased ROX state and ROX/ETS ratio, which are associated with ROS formation. Overall, we observed that changes in mitochondrial bioenergetics play a role, at least partially, in both acute and chronic effects of EtOH in the zebrafish brain. Moreover, our findings reinforce the face, predictive, and construct validities of zebrafish models to explore the neurochemical bases involved in alcohol abuse and alcoholism.
Collapse
Affiliation(s)
- Talise E Müller
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS, 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS, 97105-900, Brazil.
| | - Mauro E M Nunes
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS, 97105-900, Brazil; Oxidative Stress and Cell Signaling Research Group, Interdisciplinary Center for Biotechnology Research - CIPBIOTEC, Campus São Gabriel, Universidade Federal do Pampa, São Gabriel, RS, 97300-000, Brazil
| | - Nathane R Rodrigues
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS, 97105-900, Brazil; Oxidative Stress and Cell Signaling Research Group, Interdisciplinary Center for Biotechnology Research - CIPBIOTEC, Campus São Gabriel, Universidade Federal do Pampa, São Gabriel, RS, 97300-000, Brazil
| | - Barbara D Fontana
- Brain and Behaviour Laboratory, School of Pharmacy and Biomedical Sciences, University of Portsmouth, UK
| | - Diane D Hartmann
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS, 97105-900, Brazil
| | - Jeferson L Franco
- Oxidative Stress and Cell Signaling Research Group, Interdisciplinary Center for Biotechnology Research - CIPBIOTEC, Campus São Gabriel, Universidade Federal do Pampa, São Gabriel, RS, 97300-000, Brazil
| | - Denis B Rosemberg
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS, 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS, 97105-900, Brazil; The International Zebrafish Neuroscience Research Consortium (ZNRC), 309 Palmer Court, Slidell, LA, 70458, USA.
| |
Collapse
|
76
|
Tauffenberger A, Fiumelli H, Almustafa S, Magistretti PJ. Lactate and pyruvate promote oxidative stress resistance through hormetic ROS signaling. Cell Death Dis 2019; 10:653. [PMID: 31506428 PMCID: PMC6737085 DOI: 10.1038/s41419-019-1877-6] [Citation(s) in RCA: 156] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 07/17/2019] [Accepted: 08/01/2019] [Indexed: 12/11/2022]
Abstract
L-lactate was long considered a glycolytic by-product but is now being recognized as a signaling molecule involved in cell survival. In this manuscript, we report the role of L-lactate in stress resistance and cell survival mechanisms using neuroblastoma cells (SH-SY5Y) as well as the C. elegans model. We observed that L-lactate promotes cellular defense mechanisms, including Unfolded Protein Response (UPR) and activation of nuclear factor erythroid 2-related factor 2 (NRF2), by promoting a mild Reactive Oxygen Species (ROS) burst. This increase in ROS triggers antioxidant defenses and pro-survival pathways, such as PI3K/AKT and Endoplasmic Reticulum (ER) chaperones. These results contribute to the understanding of the molecular mechanisms involved in beneficial effects of L-lactate, involving mild ROS burst, leading to activation of unfolded protein responses and detoxification mechanisms. We present evidence that this hormetic mechanism induced by L-lactate protects against oxidative stress in vitro and in vivo. This work contributes to the identification of molecular mechanisms, which could serve as targets for future therapeutic approaches for cell protection and aging-related disorders.
Collapse
Affiliation(s)
- Arnaud Tauffenberger
- Laboratory for Cellular Imaging and Energetics, Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia.
| | - Hubert Fiumelli
- Laboratory for Cellular Imaging and Energetics, Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Salam Almustafa
- Laboratory for Cellular Imaging and Energetics, Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
- Imam Abdulrahman bin Faisal University, Dammam, Saudi Arabia
| | - Pierre J Magistretti
- Laboratory for Cellular Imaging and Energetics, Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia.
| |
Collapse
|
77
|
Marshall KD, Klutho PJ, Song L, Krenz M, Baines CP. The novel cyclophilin-D-interacting protein FASTKD1 protects cells against oxidative stress-induced cell death. Am J Physiol Cell Physiol 2019; 317:C584-C599. [PMID: 31268778 DOI: 10.1152/ajpcell.00471.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Opening of the mitochondrial permeability transition (MPT) pore leads to necrotic cell death. Excluding cyclophilin D (CypD), the makeup of the MPT pore remains conjecture. The purpose of these experiments was to identify novel MPT modulators by analyzing proteins that associate with CypD. We identified Fas-activated serine/threonine phosphoprotein kinase domain-containing protein 1 (FASTKD1) as a novel CypD interactor. Overexpression of FASTKD1 protected mouse embryonic fibroblasts (MEFs) against oxidative stress-induced reactive oxygen species (ROS) production and cell death, whereas depletion of FASTKD1 sensitized them. However, manipulation of FASTKD1 levels had no effect on MPT responsiveness, Ca2+-induced cell death, or antioxidant capacity. Moreover, elevated FASTKD1 levels still protected against oxidative stress in CypD-deficient MEFs. FASTKD1 overexpression decreased Complex-I-dependent respiration and ΔΨm in MEFs, effects that were abrogated in CypD-null cells. Additionally, overexpression of FASTKD1 in MEFs induced mitochondrial fragmentation independent of CypD, activation of Drp1, and inhibition of autophagy/mitophagy, whereas knockdown of FASTKD1 had the opposite effect. Manipulation of FASTKD1 expression also modified oxidative stress-induced caspase-3 cleavage yet did not alter apoptotic death. Finally, the effects of FASTKD1 overexpression on oxidative stress-induced cell death and mitochondrial morphology were recapitulated in cultured cardiac myocytes. Together, these data indicate that FASTKD1 supports mitochondrial homeostasis and plays a critical protective role against oxidant-induced death.
Collapse
Affiliation(s)
- Kurt D Marshall
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Paula J Klutho
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Lihui Song
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Maike Krenz
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri.,Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Christopher P Baines
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri.,Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| |
Collapse
|
78
|
Song J, Yang X, Yan LJ. Role of pseudohypoxia in the pathogenesis of type 2 diabetes. HYPOXIA 2019; 7:33-40. [PMID: 31240235 PMCID: PMC6560198 DOI: 10.2147/hp.s202775] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 04/23/2019] [Indexed: 12/14/2022]
Abstract
Type 2 diabetes is caused by persistent high blood glucose, which is known as diabetic hyperglycemia. This hyperglycemic situation, when not controlled, can overproduce NADH and lower nicotinamide adenine dinucleotide (NAD), thereby creating NADH/NAD redox imbalance and leading to cellular pseudohypoxia. In this review, we discussed two major enzymatic systems that are activated by diabetic hyperglycemia and are involved in creation of this pseudohypoxic condition. One system is aldose reductase in the polyol pathway, and the other is poly (ADP ribose) polymerase. While aldose reductase drives overproduction of NADH, PARP could in contrast deplete NAD. Therefore, activation of the two pathways underlies the major mechanisms of NADH/NAD redox imbalance and diabetic pseudohypoxia. Consequently, reductive stress occurs, followed by oxidative stress and eventual cell death and tissue dysfunction. Additionally, fructose formed in the polyol pathway can also cause metabolic syndrome such as hypertension and nonalcoholic fatty liver disease. Moreover, pseudohypoxia can also lower sirtuin protein contents and induce protein acetylation which can impair protein function. Finally, we discussed the possibility of using nicotinamide riboside, an NAD precursor, as a promising therapeutic agent for restoring NADH/NAD redox balance and for preventing the occurrence of diabetic pseudohypoxia.
Collapse
Affiliation(s)
- Jing Song
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX, USA.,School of Public Health, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Xiaojuan Yang
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX, USA.,Department of Geriatrics, First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Liang-Jun Yan
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX, USA
| |
Collapse
|
79
|
Bunik VI. Redox-Driven Signaling: 2-Oxo Acid Dehydrogenase Complexes as Sensors and Transmitters of Metabolic Imbalance. Antioxid Redox Signal 2019; 30:1911-1947. [PMID: 30187773 DOI: 10.1089/ars.2017.7311] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
SIGNIFICANCE This article develops a holistic view on production of reactive oxygen species (ROS) by 2-oxo acid dehydrogenase complexes. Recent Advances: Catalytic and structural properties of the complexes and their components evolved to minimize damaging effects of side reactions, including ROS generation, simultaneously exploiting the reactions for homeostatic signaling. CRITICAL ISSUES Side reactions of the complexes, characterized in vitro, are analyzed in view of protein interactions and conditions in vivo. Quantitative data support prevalence of the forward 2-oxo acid oxidation over the backward NADH oxidation in feeding physiologically significant ROS production by the complexes. Special focus on interactions between the active sites within 2-oxo acid dehydrogenase complexes highlights the central relevance of the complex-bound thiyl radicals in regulation of and signaling by complex-generated ROS. The thiyl radicals arise when dihydrolipoyl residues of the complexes regenerate FADH2 from the flavin semiquinone coproduced with superoxide anion radical in 1e- oxidation of FADH2 by molecular oxygen. FUTURE DIRECTIONS Interaction of 2-oxo acid dehydrogenase complexes with thioredoxins (TRXs), peroxiredoxins, and glutaredoxins mediates scavenging of the thiyl radicals and ROS generated by the complexes, underlying signaling of disproportional availability of 2-oxo acids, CoA, and NAD+ in key metabolic branch points through thiol/disulfide exchange and medically important hypoxia-inducible factor, mammalian target of rapamycin (mTOR), poly (ADP-ribose) polymerase, and sirtuins. High reactivity of the coproduced ROS and thiyl radicals to iron/sulfur clusters and nitric oxide, peroxynitrite reductase activity of peroxiredoxins and transnitrosylating function of thioredoxin, implicate the side reactions of 2-oxo acid dehydrogenase complexes in nitric oxide-dependent signaling and damage.
Collapse
Affiliation(s)
- Victoria I Bunik
- 1 Belozersky Institute of Physicochemical Biology, Lomonosov Moscow State University, Moscow, Russian Federation.,2 Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russian Federation
| |
Collapse
|
80
|
Yu Z, Yu Z, Chen Z, Yang L, Ma M, Lu S, Wang C, Teng C, Nie Y. Zinc chelator TPEN induces pancreatic cancer cell death through causing oxidative stress and inhibiting cell autophagy. J Cell Physiol 2019; 234:20648-20661. [PMID: 31054150 DOI: 10.1002/jcp.28670] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 03/07/2019] [Accepted: 03/14/2019] [Indexed: 12/15/2022]
Abstract
The essential trace element zinc (Zn) is widely required in cellular functions, and abnormal Zn homeostasis causes a variety of health problems including immunodeficiency and sensory dysfunctions. Previous studies had shown that Zn availability was also important for tumor growth and progression. The aim of the present study was to investigate the potential mechanisms of N,N,N,N-Tetrakis(2-pyridylmethyl)-ethylenediamine (TPEN) (a membrane permeable zinc chelator) induced pancreatic cancer cell death. The text of inductively coupled plasma-mass spectrometry (ICP-MS) showed in human pancreatic cancer samples that the zinc content in cancer was higher than that in adjacent tissues. The pancreatic cancer cell lines Panc-1, 8988T, BxPc-3, and L3.6 were used in this study. Our results indicated that TPEN markedly induced cell death, via increasing reactive oxygen species (ROS) and restraining autophagy. Our data also indicated that TPEN-stimulated mitochondrial metabolism produced much ROS. Meanwhile, TPEN reduced the levels of glutathione (GSH) and triggered ROS outbreak, which were the main causes of cell death. In addition, cell autophagy was significantly depressed in Panc-1 cells treated by TPEN, which was due to the ability of disrupting lysosomal by TPEN. Thus, we thought zinc depletion by TPEN was a potential therapeutic strategy for pancreatic cancer.
Collapse
Affiliation(s)
- Zhen Yu
- College of Life Science, Northeast Forestry University, Harbin, China
| | - Ze Yu
- College of Life Science, Northeast Forestry University, Harbin, China
| | - ZhenBao Chen
- College of Life Science, Northeast Forestry University, Harbin, China
| | - Lin Yang
- College of Life Science, Northeast Forestry University, Harbin, China
| | - MingJun Ma
- College of Life Science, Northeast Forestry University, Harbin, China
| | - ShouNan Lu
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - ChunSheng Wang
- College of Life Science, Northeast Forestry University, Harbin, China
| | - ChunBo Teng
- College of Life Science, Northeast Forestry University, Harbin, China
| | - YuZhe Nie
- College of Life Science, Northeast Forestry University, Harbin, China
| |
Collapse
|
81
|
Quinone-thioether metabolites of hydroquinone play a dual role in promoting a vicious cycle of ROS generation: in vitro and in silico insights. Arch Toxicol 2019; 93:1297-1309. [PMID: 30976846 DOI: 10.1007/s00204-019-02443-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 04/09/2019] [Indexed: 02/06/2023]
Abstract
Humans are exposed to hydroquinone (HQ) via diet, smoking, occupation, and even via inhalation of polluted air. Given its preferential distribution in kidney and liver, the impact of biotransformation on the nephrotoxicity and hepatotoxicity of HQ was evaluated. Indeed, HQ and its metabolites, benzoquinone, and quinone-thioethers (50, 100, 200, and 400 μM) all induced ROS-dependent cell death in both HK-2, a human kidney proximal epithelial cell line, and THLE-2, a human liver epithelial cell line, in a concentration-dependent manner. For a deeper insight into the biological mechanism of ROS stimulation, the bioinformatics database was reviewed. Intriguingly, 163 proteins were currently reported to form co-crystal complex with benzoquinone analogs, a large proportion of which are closely related to ROS generation. After a thorough assessment of the interaction affinity and binding energy, three key mitochondrial proteins that are particularly involved in electric transport, namely, cytochrome BC1, succinate dehydrogenase, and sulfide:quinone oxidoreductase, were highlighted for further verification. Their binding affinity and the action pattern were explored and validated by molecular docking and molecular dynamics simulations. Remarkably, quinone-thioether metabolites of HQ afforded high affinity to the above proteins that purportedly cause a surge in the generation of ROS. Therefore, HQ can be further converted into quinone-thioethers, which on one hand can function as substrates for redox cycling, and on the other hand may afford high affinity with key proteins evolved in mitochondrial electron transport system, leading to a vicious cycle of ROS generation. The combined data provide a prospective insight into the mechanisms of ROS motivation, expanding HQ-mediated toxicology profiles.
Collapse
|
82
|
Thomas LW, Stephen JM, Esposito C, Hoer S, Antrobus R, Ahmed A, Al-Habib H, Ashcroft M. CHCHD4 confers metabolic vulnerabilities to tumour cells through its control of the mitochondrial respiratory chain. Cancer Metab 2019; 7:2. [PMID: 30886710 PMCID: PMC6404347 DOI: 10.1186/s40170-019-0194-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 02/05/2019] [Indexed: 12/15/2022] Open
Abstract
Background Tumour cells rely on glycolysis and mitochondrial oxidative phosphorylation (OXPHOS) to survive. Thus, mitochondrial OXPHOS has become an increasingly attractive area for therapeutic exploitation in cancer. However, mitochondria are required for intracellular oxygenation and normal physiological processes, and it remains unclear which mitochondrial molecular mechanisms might provide therapeutic benefit. Previously, we discovered that coiled-coil-helix-coiled-coil-helix domain-containing protein 4 (CHCHD4) is critical for regulating intracellular oxygenation and required for the cellular response to hypoxia (low oxygenation) in tumour cells through molecular mechanisms that we do not yet fully understand. Overexpression of CHCHD4 in human cancers correlates with increased tumour progression and poor patient survival. Results Here, we show that elevated CHCHD4 expression provides a proliferative and metabolic advantage to tumour cells in normoxia and hypoxia. Using stable isotope labelling with amino acids in cell culture (SILAC) and analysis of the whole mitochondrial proteome, we show that CHCHD4 dynamically affects the expression of a broad range of mitochondrial respiratory chain subunits from complex I-V, including multiple subunits of complex I (CI) required for complex assembly that are essential for cell survival. We found that loss of CHCHD4 protects tumour cells from respiratory chain inhibition at CI, while elevated CHCHD4 expression in tumour cells leads to significantly increased sensitivity to CI inhibition, in part through the production of mitochondrial reactive oxygen species (ROS). Conclusions Our study highlights an important role for CHCHD4 in regulating tumour cell metabolism and reveals that CHCHD4 confers metabolic vulnerabilities to tumour cells through its control of the mitochondrial respiratory chain and CI biology.
Collapse
Affiliation(s)
- Luke W. Thomas
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0AH UK
| | - Jenna M. Stephen
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0AH UK
| | - Cinzia Esposito
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0AH UK
- Present address: Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Simon Hoer
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0XY UK
| | - Robin Antrobus
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0XY UK
| | - Afshan Ahmed
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0AH UK
- Present address: AstraZeneca Ltd., Cambridge, UK
| | - Hasan Al-Habib
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0AH UK
| | - Margaret Ashcroft
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0AH UK
| |
Collapse
|
83
|
Zhang M, Zhang Y, Li Z, Zhang C, Tan X, Liu X, Wan C, Yang X, Lee DJ. Anaerobic co-digestion of food waste/excess sludge: substrates - products transformation and role of NADH as an indicator. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2019; 232:197-206. [PMID: 30472563 DOI: 10.1016/j.jenvman.2018.11.087] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 11/06/2018] [Accepted: 11/19/2018] [Indexed: 06/09/2023]
Abstract
The process of anaerobic co-digestion is vital importance to resource recovery from organic solid wastes such as food waste and municipal sludge. However, its application is hindered by the limited understanding on the complex substrates-products transformation reactions and mechanisms therein. In this study, food waste (FW) and excess sludge (ES) from municipal wastewater treatment were mixed at various ratios (ES/FW 5:0, 4:1, 2:1, 1:1, 1:2, 1:4, w/w), and the co-digestion process was studied in a batch test. The consumption of substrates including soluble proteins and carbohydrates, the variation in the intermediates such as various volatile fatty acids, and the production of hydrogen and methane gases were monitored. The results suggested that 4:1 was likely the optimal ratio where substrates were consumed and biogas generated efficiently, whereas 1:2 and 1:4 caused severe inhibition. Fermentation of ES alone produced mainly acetic and propionic acid, while the addition of FW led to butyric acid type fermentation. Intermediates in the fermentation liquid were tentatively identified, and the levels of NADH quantified using 3D-excitation/emission fluorescence spectrometry. One class of the intermediates, tryptophan-like proteins were correlated to the butyric acid accumulation in ES/FW mixtures, and NADH level was proposed as an indicator of VFAs production activities.
Collapse
Affiliation(s)
- Min Zhang
- Department of Environmental Science and Engineering, Fudan University, Shanghai, 200433, China
| | - Yi Zhang
- Department of Environmental Science and Engineering, Fudan University, Shanghai, 200433, China
| | - Zhengwen Li
- Department of Environmental Science and Engineering, Fudan University, Shanghai, 200433, China
| | - Chen Zhang
- Shanghai Municipal Engineering Design General Institute, Shanghai, 200092, China
| | - Xuejun Tan
- Shanghai Municipal Engineering Design General Institute, Shanghai, 200092, China
| | - Xiang Liu
- Department of Environmental Science and Engineering, Fudan University, Shanghai, 200433, China.
| | - Chunli Wan
- Department of Environmental Science and Engineering, Fudan University, Shanghai, 200433, China.
| | - Xue Yang
- Shanghai Municipal Engineering Design General Institute, Shanghai, 200092, China
| | - Duu-Jong Lee
- Department of Chemical Engineering, National Taiwan University, Taipei, 106, Taiwan
| |
Collapse
|
84
|
Bellomo F, Signorile A, Tamma G, Ranieri M, Emma F, De Rasmo D. Impact of atypical mitochondrial cyclic-AMP level in nephropathic cystinosis. Cell Mol Life Sci 2018; 75:3411-3422. [PMID: 29549422 PMCID: PMC11105431 DOI: 10.1007/s00018-018-2800-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 02/20/2018] [Accepted: 03/14/2018] [Indexed: 02/06/2023]
Abstract
Nephropathic cystinosis (NC) is a rare disease caused by mutations in the CTNS gene encoding for cystinosin, a lysosomal transmembrane cystine/H+ symporter, which promotes the efflux of cystine from lysosomes to cytosol. NC is the most frequent cause of Fanconi syndrome (FS) in young children, the molecular basis of which is not well established. Proximal tubular cells have very high metabolic rate due to the active transport of many solutes. Not surprisingly, mitochondrial disorders are often characterized by FS. A similar mechanism may also apply to NC. Because cAMP has regulatory properties on mitochondrial function, we have analyzed cAMP levels and mitochondrial targets in CTNS-/- conditionally immortalized proximal tubular epithelial cells (ciPTEC) carrying the classical homozygous 57-kb deletion (delCTNS-/-) or with compound heterozygous loss-of-function mutations (mutCTNS-/-). Compared to wild-type cells, cystinotic cells had significantly lower mitochondrial cAMP levels (delCTNS-/- ciPTEC by 56% ± 10.5, P < 0.0001; mutCTNS-/- by 26% ± 4.3, P < 0.001), complex I and V activities, mitochondrial membrane potential, and SIRT3 protein levels, which were associated with increased mitochondrial fragmentation. Reduction of complex I and V activities was associated with lower expression of part of their subunits. Treatment with the non-hydrolysable cAMP analog 8-Br-cAMP restored mitochondrial potential and corrected mitochondria morphology. Treatment with cysteamine, which reduces the intra-lysosomal cystine, was able to restore mitochondrial cAMP levels, as well as most other abnormal mitochondrial findings. These observations were validated in CTNS-silenced HK-2 cells, indicating a pivotal role of mitochondrial cAMP in the proximal tubular dysfunction observed in NC.
Collapse
Affiliation(s)
- Francesco Bellomo
- Laboratory of Nephrology, Department of Rare Diseases, Bambino Gesù Children's Hospital, Viale di S. Paolo, 15, 00149, Rome, Italy.
| | - Anna Signorile
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari "Aldo Moro", Policlinico, Piazza G. Cesare, 11, 70124, Bari, Italy
| | - Grazia Tamma
- Department of Bioscience, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", Bari, Italy
| | - Marianna Ranieri
- Department of Bioscience, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", Bari, Italy
| | - Francesco Emma
- Laboratory of Nephrology, Department of Rare Diseases, Bambino Gesù Children's Hospital, Viale di S. Paolo, 15, 00149, Rome, Italy
- Division of Nephrology, Department of Pediatric Subspecialties, Bambino Gesù Children's Hospital, Rome, Italy
| | - Domenico De Rasmo
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari "Aldo Moro", Policlinico, Piazza G. Cesare, 11, 70124, Bari, Italy.
- Institute of Biomembrane, Bioenergetics and Molecular Biotechnology (IBIOM), National Research Council (CNR), Bari, Italy.
| |
Collapse
|
85
|
Diaz-Morales N, Lopez-Domenech S, Iannantuoni F, Lopez-Gallardo E, Sola E, Morillas C, Rocha M, Ruiz-Pesini E, Victor VM. Mitochondrial DNA Haplogroup JT is Related to Impaired Glycaemic Control and Renal Function in Type 2 Diabetic Patients. J Clin Med 2018; 7:jcm7080220. [PMID: 30115863 PMCID: PMC6111716 DOI: 10.3390/jcm7080220] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 08/13/2018] [Accepted: 08/14/2018] [Indexed: 12/25/2022] Open
Abstract
The association between mitochondrial DNA (mtDNA) haplogroup and risk of type 2 diabetes (T2D) is undetermined and controversial. This study aims to evaluate the impact of the main mtDNA haplogroups on glycaemic control and renal function in a Spanish population of 303 T2D patients and 153 healthy controls. Anthropometrical and metabolic parameters were assessed and mtDNA haplogroup was determined in each individual. Distribution of the different haplogroups was similar in diabetic and healthy populations and, as expected, T2D patients showed poorer glycaemic control and renal function than controls. T2D patients belonging to the JT haplogroup (polymorphism m.4216T>C) displayed statistically significant higher levels of fasting glucose and HbA1c than those of the other haplogroups, suggesting a poorer glycaemic control. Furthermore, diabetic patients with the JT haplogroup showed a worse kidney function than those with other haplogroups, evident by higher levels of serum creatinine, lower estimated glomerular filtration rate (eGFR), and slightly higher (although not statistically significant) urinary albumin-to-creatinine ratio. Our results suggest that JT haplogroup (in particular, change at position 4216 of the mtDNA) is associated with poorer glycaemic control in T2D, which can trigger the development of diabetic nephropathy.
Collapse
Affiliation(s)
- Noelia Diaz-Morales
- Service of Endocrinology, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain.
| | - Sandra Lopez-Domenech
- Service of Endocrinology, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain.
| | - Francesca Iannantuoni
- Service of Endocrinology, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain.
| | - Ester Lopez-Gallardo
- Department of Biochemistry and Molecular and Cell Biology, University of Zaragoza, 50013 Zaragoza, Spain.
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón), 50013 Zaragoza, Spain.
- Centro de Investigaciones Biomédicas En Red de Enfermedades Raras (CIBERER), 50013 Zaragoza, Spain.
| | - Eva Sola
- Service of Endocrinology, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain.
| | - Carlos Morillas
- Service of Endocrinology, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain.
| | - Milagros Rocha
- Service of Endocrinology, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain.
- CIBERehd-Department of Pharmacology and Physiology, University of Valencia, 46010 Valencia, Spain.
| | - Eduardo Ruiz-Pesini
- Department of Biochemistry and Molecular and Cell Biology, University of Zaragoza, 50013 Zaragoza, Spain.
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón), 50013 Zaragoza, Spain.
- Centro de Investigaciones Biomédicas En Red de Enfermedades Raras (CIBERER), 50013 Zaragoza, Spain.
- Fundación ARAID, 50018 Zaragoza, Spain.
| | - Victor M Victor
- Service of Endocrinology, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain.
- CIBERehd-Department of Pharmacology and Physiology, University of Valencia, 46010 Valencia, Spain.
| |
Collapse
|
86
|
Gladyshev GV, Grivennikova VG, Vinogradov AD. FMN site-independent energy-linked reverse electron transfer in mitochondrial respiratory complex I. FEBS Lett 2018; 592:2213-2219. [PMID: 29851085 DOI: 10.1002/1873-3468.13117] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 05/16/2018] [Accepted: 05/17/2018] [Indexed: 11/06/2022]
Abstract
A simple assay procedure for measuring ATP-dependent reverse electron transfer from ubiquinol to hexaammineruthenium (III) (HAR) catalyzed by mitochondrial respiratory complex I is introduced. The specific activity of the enzyme in this reaction and its sensitivity to the standard inhibitors and uncoupling are the same as with other well-known electron acceptors, NAD+ and ferricyanide. In contrast to the reactions with these acceptors, the energy-dependent HAR reduction is not inhibited by NADH-OH, the specific inhibitor of NADH-binding site. These results suggest that a catalytically competent electron connection exists between HAR and a redox component of complex I that is different from flavin mononucleotide bound at the substrate-binding site.
Collapse
Affiliation(s)
- Grigory V Gladyshev
- Department of Biochemistry, School of Biology, Moscow State University, Russia
| | - Vera G Grivennikova
- Department of Biochemistry, School of Biology, Moscow State University, Russia
| | - Andrei D Vinogradov
- Department of Biochemistry, School of Biology, Moscow State University, Russia
| |
Collapse
|
87
|
The Oncojanus Paradigm of Respiratory Complex I. Genes (Basel) 2018; 9:genes9050243. [PMID: 29735924 PMCID: PMC5977183 DOI: 10.3390/genes9050243] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 04/09/2018] [Accepted: 05/03/2018] [Indexed: 02/07/2023] Open
Abstract
Mitochondrial respiratory function is now recognized as a pivotal player in all the aspects of cancer biology, from tumorigenesis to aggressiveness and chemotherapy resistance. Among the enzymes that compose the respiratory chain, by contributing to energy production, redox equilibrium and oxidative stress, complex I assumes a central role. Complex I defects may arise from mutations in mitochondrial or nuclear DNA, in both structural genes or assembly factors, from alteration of the expression levels of its subunits, or from drug exposure. Since cancer cells have a high-energy demand and require macromolecules for proliferation, it is not surprising that severe complex I defects, caused either by mutations or treatment with specific inhibitors, prevent tumor progression, while contributing to resistance to certain chemotherapeutic agents. On the other hand, enhanced oxidative stress due to mild complex I dysfunction drives an opposite phenotype, as it stimulates cancer cell proliferation and invasiveness. We here review the current knowledge on the contribution of respiratory complex I to cancer biology, highlighting the double-edged role of this metabolic enzyme in tumor progression, metastasis formation, and response to chemotherapy.
Collapse
|
88
|
Shumaev KB, Dudylina AL, Ivanova MV, Pugachenko IS, Ruuge EK. Dinitrosyl iron complexes: Formation and antiradical action in heart mitochondria. Biofactors 2018; 44:237-244. [PMID: 29469215 DOI: 10.1002/biof.1418] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 01/25/2018] [Indexed: 02/02/2023]
Abstract
Mitochondria are widely known as a major source of reactive oxygen and nitrogen species for the cardiovascular system. Numerous studies established that superoxide anion radical production by heart mitochondria is only slightly suppressed under conditions of deep hypoxia, but is completely blocked under anoxia. It was found also that dinitrosyl iron complexes (DNIC) compare favourably with other physiologically active derivatives of nitric oxide (NO). DNIC with glutathione effectively scavenge superoxide radicals generated by mitochondria at different partial pressures of oxygen. Under conditions of simulated hypoxia, the synthesis of thiol-containing DNIC takes place in mitochondria and is concomitant with a significant decrease in the concentration of NO metabolites at the reoxygenation step. Free NO required for DNIC synthesis is generated in the reaction of S-nitrosothiols with superoxide or during single-electron oxidation of the nitroxyl radical (HNO) by coenzyme Q. Plausible mechanisms of antiradical effects of DNIC and their protective role in oxidative stress induced by hypoxia/reoxygenation of myocardial tissues are considered. © 2018 BioFactors, 44(3):237-244, 2018.
Collapse
Affiliation(s)
- Konstantin B Shumaev
- A.N. Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, 119071, Russia
- National Medical Research Centre for Cardiology, Moscow, 121552, Russia
| | - Arina L Dudylina
- Faculty of Physics, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Marina V Ivanova
- National Medical Research Centre for Cardiology, Moscow, 121552, Russia
| | - Igor S Pugachenko
- A.N. Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, 119071, Russia
| | - Enno K Ruuge
- National Medical Research Centre for Cardiology, Moscow, 121552, Russia
- Faculty of Physics, Lomonosov Moscow State University, Moscow, 119234, Russia
| |
Collapse
|
89
|
Ning P, Zheng Z, Aweya JJ, Yao D, Li S, Ma H, Wang F, Zhang Y. Litopenaeus vannamei notch affects lipopolysaccharides induced reactive oxygen species. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 81:74-82. [PMID: 29155012 DOI: 10.1016/j.dci.2017.11.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 11/14/2017] [Accepted: 11/14/2017] [Indexed: 06/07/2023]
Abstract
Notch signaling pathway was originally discovered in the development stage of drosophila but has recently been found to play essential roles in innate immunity. Most previous studies on Notch have focused on mammals, whereas, in this study, we employed the shrimp Litopenaeus vannamei as a model to study the functions of Notch in invertebrate innate immune system. Our results showed that LvNotch was highly expressed in hemocytes and could be strongly induced by lipopolysaccharides (LPS) injection. Small interfering RNA (siRNA)-mediated knockdown of LvNotch could significantly increase LPS induced L. vannamei mortality, which might be due to the fact that LPS induced ROS was greatly enhanced in LvNotch knockdown shrimps. Further, quantitative polymerase chain reaction (qPCR) analysis revealed that LvNotch could affect the expression of multiple genes, including dorsal, relish, anti-lipopolysaccharide factor 1 (ALF1), ALF3 and NADH dehydrogenases which were upregulated, and Hypoxia-inducible factor (HIF, α/β) which were downregulated in LPS treated shrimps. In summary, LvNotch is important in the control of inflammation-induced ROS production in shrimp.
Collapse
Affiliation(s)
- Pei Ning
- Department of Biology and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China
| | - Zhihong Zheng
- Department of Biology and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China
| | - Jude Juventus Aweya
- Department of Biology and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China
| | - Defu Yao
- Department of Biology and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China
| | - Shengkang Li
- Department of Biology and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China
| | - Hongyu Ma
- Department of Biology and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China
| | - Fan Wang
- Department of Biology and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China.
| | - Yueling Zhang
- Department of Biology and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China.
| |
Collapse
|
90
|
Angelova PR, Abramov AY. Role of mitochondrial ROS in the brain: from physiology to neurodegeneration. FEBS Lett 2018; 592:692-702. [PMID: 29292494 DOI: 10.1002/1873-3468.12964] [Citation(s) in RCA: 504] [Impact Index Per Article: 72.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 12/17/2017] [Accepted: 12/21/2017] [Indexed: 12/14/2022]
Abstract
Mitochondria are key cell organelles in that they are responsible for energy production and control many processes from signalling to cell death. The function of the mitochondrial electron transport chain is coupled with the production of reactive oxygen species (ROS) in the form of superoxide anion or hydrogen peroxide. As a result of the constant production of ROS, mitochondria are protected by highly efficient antioxidant systems. The rapidly changing levels of ROS in mitochondria, coupled with multiple essential cellular functions, make ROS apt for physiological signalling. Thus, mutations, environmental toxins and chronic ischaemic conditions could affect the mitochondrial redox balance and lead to the development of pathology. In long-living and non-mitotic cells such as neurons, oxidative stress induced by overproduction of mitochondrial ROS or impairment of the antioxidant defence results in a dysfunction of mitochondria and initiation of the cell death cascade. Mitochondrial ROS overproduction and changes in mitochondrial redox homeostasis have been shown to be involved in both a number of neurological conditions and a majority of neurodegenerative diseases. Here, we summarise the involvement of mitochondrial ROS in the mechanism of neuronal loss of major neurodegenerative disorders.
Collapse
Affiliation(s)
- Plamena R Angelova
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Andrey Y Abramov
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| |
Collapse
|
91
|
Carmo C, Naia L, Lopes C, Rego AC. Mitochondrial Dysfunction in Huntington’s Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1049:59-83. [DOI: 10.1007/978-3-319-71779-1_3] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
92
|
Koshikawa N, Akimoto M, Hayashi JI, Nagase H, Takenaga K. Association of predicted pathogenic mutations in mitochondrial ND genes with distant metastasis in NSCLC and colon cancer. Sci Rep 2017; 7:15535. [PMID: 29138417 PMCID: PMC5686070 DOI: 10.1038/s41598-017-15592-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 10/31/2017] [Indexed: 11/17/2022] Open
Abstract
Cancer cells have more mutations in their mitochondrial DNA (mtDNA) than do normal cells, and pathogenic mutations in the genes encoding mitochondrial NADH dehydrogenase (ND) subunits have been found to enhance the invasive and metastatic ability of various tumour cells in animal experiments. However, it is unknown whether single-nucleotide variants (SNVs) of the ND genes that decrease complex I activity are involved in distant metastasis in human clinical samples. Here, we demonstrated the enhancement of the distant metastasis of Lewis lung carcinoma cells by the ND6 13885insC mutation, which is accompanied by the overexpression of metastasis-related genes, metabolic reprogramming, the enhancement of tumour angiogenesis and the acquisition of resistance to stress-induced cell death. We then sequenced ND genes in primary tumour lesions with or without distant metastases as well as metastatic tumour lesions from 115 patients with non-small cell lung cancer (NSCLC) and colon cancer, and we subsequently selected 14 SNVs with the potential to decrease complex I activity. Intriguingly, a significant correlation was observed (P < 0.05 by Chi-square test) between the incidence of the selected mutations and distant metastasis. Thus, these results strongly suggest that pathogenic ND gene mutations participate in enhancing distant metastasis in human cancers.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Animals
- Carcinoma, Lewis Lung/genetics
- Carcinoma, Lewis Lung/pathology
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/secondary
- Cell Line, Tumor
- Colonic Neoplasms/genetics
- Colonic Neoplasms/secondary
- DNA, Mitochondrial/genetics
- Female
- Genes, Mitochondrial/genetics
- Humans
- Lung Neoplasms/genetics
- Lung Neoplasms/pathology
- Male
- Mice, Inbred C57BL
- Middle Aged
- Mitochondria/genetics
- Mutation
- NADH Dehydrogenase/genetics
- Neoplasm Metastasis
- Polymorphism, Single Nucleotide
Collapse
Affiliation(s)
- Nobuko Koshikawa
- Laboratory of Cancer Genetics, Chiba Cancer Center Research Institute, 666-2 Nitona, Chuoh-ku, Chiba, 260-8717, Japan
| | - Miho Akimoto
- Department of Life Science, Shimane University Faculty of Medicine, 89-1 Enya, Izumo, Shimane, 693-8501, Japan
- Department of Biochemistry, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan
| | - Jun-Ichi Hayashi
- University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8572, Japan
| | - Hiroki Nagase
- Laboratory of Cancer Genetics, Chiba Cancer Center Research Institute, 666-2 Nitona, Chuoh-ku, Chiba, 260-8717, Japan
| | - Keizo Takenaga
- Department of Life Science, Shimane University Faculty of Medicine, 89-1 Enya, Izumo, Shimane, 693-8501, Japan.
| |
Collapse
|
93
|
Metformin ameliorates arsenic trioxide hepatotoxicity via inhibiting mitochondrial complex I. Cell Death Dis 2017; 8:e3159. [PMID: 29095437 PMCID: PMC5775401 DOI: 10.1038/cddis.2017.482] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 08/19/2017] [Accepted: 08/22/2017] [Indexed: 12/11/2022]
Abstract
Arsenic trioxide (ATO) is a well-accepted chemotherapy agent in managing promyelocytic leukemia. ATO often causes severe health hazards such as hepatotoxicity, dermatosis, neurotoxicity, nephrotoxicity and cardiotoxicity. The production of reactive oxygen species, (ROS) play a significant role in ATO-induced hepatotoxicity. The oral hypoglycemic drug, metformin, is considered to be a potential novel agent for chemoprevention in the treatment of cancer. Moreover, metformin has also been shown to have hepatoprotective effects. In the present study, we demonstrated that metformin protected normal hepatocytes from ATO-induced apoptotic cell death in vitro and in vivo. Gene expression screening revealed that glucose metabolism might be related to the metformin-induced protective effect on ATO-treated AML12 cells. The metformin-promoted or induced glycolysis was not responsible for the protection of AML12 cells from ATO-induced apoptotic cell death. Instead, metformin increased the intracellular NADH/NAD+ ratio by inhibiting mitochondrial respiratory chain complex I, further decreasing the intracellular ROS induced by ATO. Treatment with low glucose or rotenone, a mitochondrial respiratory chain complex I inhibitor, also protected AML12 cells from ATO-induced apoptotic cell death. We show for the first time that metformin protects the hepatocyte from ATO by regulating the mitochondrial function. With its properties of chemoprevention, chemosensitization and the amelioration of liver damage, metformin has great prospects for clinical application other than type 2 diabetes mellitus (T2DM).
Collapse
|
94
|
Wu J, Luo X, Thangthaeng N, Sumien N, Chen Z, Rutledge MA, Jing S, Forster MJ, Yan LJ. Pancreatic mitochondrial complex I exhibits aberrant hyperactivity in diabetes. Biochem Biophys Rep 2017; 11:119-129. [PMID: 28868496 PMCID: PMC5580358 DOI: 10.1016/j.bbrep.2017.07.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 07/13/2017] [Accepted: 07/18/2017] [Indexed: 12/28/2022] Open
Abstract
It is well established that NADH/NAD+ redox balance is heavily perturbed in diabetes, and the NADH/NAD+ redox imbalance is a major source of oxidative stress in diabetic tissues. In mitochondria, complex I is the only site for NADH oxidation and NAD+ regeneration and is also a major site for production of mitochondrial reactive oxygen species (ROS). Yet how complex I responds to the NADH/NAD+ redox imbalance and any potential consequences of such response in diabetic pancreas have not been investigated. We report here that pancreatic mitochondrial complex I showed aberrant hyperactivity in either type 1 or type 2 diabetes. Further studies focusing on streptozotocin (STZ)-induced diabetes indicate that complex I hyperactivity could be attenuated by metformin. Moreover, complex I hyperactivity was accompanied by increased activities of complexes II to IV, but not complex V, suggesting that overflow of NADH via complex I in diabetes could be diverted to ROS production. Indeed in diabetic pancreas, ROS production and oxidative stress increased and mitochondrial ATP production decreased, which can be attributed to impaired pancreatic mitochondrial membrane potential that is responsible for increased cell death. Additionally, cellular defense systems such as glucose 6-phosphate dehydrogenase, sirtuin 3, and NQO1 were found to be compromised in diabetic pancreas. Our findings point to the direction that complex I aberrant hyperactivity in pancreas could be a major source of oxidative stress and β cell failure in diabetes. Therefore, inhibiting pancreatic complex I hyperactivity and attenuating its ROS production by various means in diabetes might serve as a promising approach for anti-diabetic therapies.
Collapse
Affiliation(s)
- Jinzi Wu
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, United States
| | - Xiaoting Luo
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, United States
- Department of Biochemistry and Molecular Biology, Gannan Medical University, Ganzhou, Jiangxi Province 341000, China
| | - Nopporn Thangthaeng
- Center for Neuroscience Discovery, Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX 76107, United States
| | - Nathalie Sumien
- Center for Neuroscience Discovery, Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX 76107, United States
| | - Zhenglan Chen
- Center for Neuroscience Discovery, Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX 76107, United States
| | - Margaret A. Rutledge
- Center for Neuroscience Discovery, Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX 76107, United States
| | - Siqun Jing
- College of Life Sciences and Technology, Xinjiang University, Urumqi, Xinjiang 830046, China
| | - Michael J. Forster
- Center for Neuroscience Discovery, Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX 76107, United States
| | - Liang-Jun Yan
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, United States
| |
Collapse
|
95
|
Pinegin B, Vorobjeva N, Pashenkov M, Chernyak B. The role of mitochondrial ROS in antibacterial immunity. J Cell Physiol 2017; 233:3745-3754. [PMID: 28771715 DOI: 10.1002/jcp.26117] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 08/01/2017] [Indexed: 12/26/2022]
Abstract
Reactive oxygen species (ROS) are essential participants of various innate immune cell responses against microorganisms and are also involved in many cellular regulatory pathways. It was believed that the main pool of ROS in the innate immune cells is generated by the NADPH oxidase enzymatic complex. However, it was discovered recently that mitochondrial ROS (mtROS) are equally important for the functioning of the immune system. mtROS play an important role in the development of the antimicrobial innate immune responses. The present mini-review summarizes the most recent data on the role of mtROS in the antibacterial immunity. The principles of mtROS formation and possible mechanisms of their generation under the activation of innate immunity are highlighted in this review. We also speculate on the possibilities of using activators of mtROS production in clinical practice.
Collapse
Affiliation(s)
- Boris Pinegin
- Laboratory of Clinical Immunology, National Research Center "Institute of Immunology" of the Federal Medical-Biological Agency, Moscow, Russia
| | - Nina Vorobjeva
- Department of Immunology, Biological Faculty, Lomonosov Moscow State University, Moscow, Russia
| | - Mikhail Pashenkov
- Laboratory of Clinical Immunology, National Research Center "Institute of Immunology" of the Federal Medical-Biological Agency, Moscow, Russia
| | - Boris Chernyak
- Department of Bioenergetics, A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
96
|
Reduction of the off-pathway iron-sulphur cluster N1a of Escherichia coli respiratory complex I restrains NAD + dissociation. Sci Rep 2017; 7:8754. [PMID: 28821859 PMCID: PMC5562879 DOI: 10.1038/s41598-017-09345-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 07/25/2017] [Indexed: 12/24/2022] Open
Abstract
Respiratory complex I couples the electron transfer from NADH to ubiquinone with the translocation of protons across the membrane. The reaction starts with NADH oxidation by a flavin cofactor followed by transferring the electrons through a chain of seven iron-sulphur clusters to quinone. An eighth cluster called N1a is located proximally to flavin, but on the opposite side of the chain of clusters. N1a is strictly conserved although not involved in the direct electron transfer to quinone. Here, we show that the NADH:ferricyanide oxidoreductase activity of E. coli complex I is strongly diminished when the reaction is initiated by an addition of ferricyanide instead of NADH. This effect is significantly less pronounced in a variant containing N1a with a 100 mV more negative redox potential. Detailed kinetic analysis revealed that the reduced activity is due to a lower dissociation constant of bound NAD+. Thus, reduction of N1a induces local structural rearrangements of the protein that stabilise binding of NAD+. The variant features a considerably enhanced production of reactive oxygen species indicating that bound NAD+ represses this process.
Collapse
|
97
|
Gawthrop PJ. Bond Graph Modeling of Chemiosmotic Biomolecular Energy Transduction. IEEE Trans Nanobioscience 2017; 16:177-188. [PMID: 28252411 DOI: 10.1109/tnb.2017.2674683] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Engineering systems modeling and analysis based on the bond graph approach has been applied to biomolecular systems. In this context, the notion of a Faraday-equivalent chemical potential is introduced which allows chemical potential to be expressed in an analogous manner to electrical volts thus allowing engineering intuition to be applied to biomolecular systems. Redox reactions, and their representation by half-reactions, are key components of biological systems which involve both electrical and chemical domains. A bond graph interpretation of redox reactions is given which combines bond graphs with the Faraday-equivalent chemical potential. This approach is particularly relevant when the biomolecular system implements chemoelectrical transduction - for example chemiosmosis within the key metabolic pathway of mitochondria: oxidative phosphorylation. An alternative way of implementing computational modularity using bond graphs is introduced and used to give a physically based model of the mitochondrial electron transport chain To illustrate the overall approach, this model is analyzed using the Faraday-equivalent chemical potential approach and engineering intuition is used to guide affinity equalisation: a energy based analysis of the mitochondrial electron transport chain.
Collapse
|
98
|
Fuhrmann DC, Brüne B. Mitochondrial composition and function under the control of hypoxia. Redox Biol 2017; 12:208-215. [PMID: 28259101 PMCID: PMC5333533 DOI: 10.1016/j.redox.2017.02.012] [Citation(s) in RCA: 412] [Impact Index Per Article: 51.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 01/31/2017] [Accepted: 02/18/2017] [Indexed: 12/13/2022] Open
Abstract
Hypoxia triggers several mechanisms to adapt cells to a low oxygen environment. Mitochondria are major consumers of oxygen and a potential source of reactive oxygen species (ROS). In response to hypoxia they exchange or modify distinct subunits of the respiratory chain and adjust their metabolism, especially lowering the citric acid cycle. Intermediates of the citric acid cycle participate in regulating hypoxia inducible factors (HIF), the key mediators of adaptation to hypoxia. Here we summarize how hypoxia conditions mitochondria with consequences for ROS-production and the HIF-pathway. Hypoxia provokes changes in mitochondrial morphology, metabolism, and respiration. Hypoxia calls forth changes in redox signaling. HIF-signaling is linked to mitochondrial metabolism and ROS formation. Hypoxia adjusts ETC complex formation, activity, respiration, and ROS formation.
Collapse
Affiliation(s)
- Dominik C Fuhrmann
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; Project Group Translational Medicine and Pharmacology TMP, Fraunhofer Institute for Molecular Biology and Applied Ecology, 60596 Frankfurt, Germany.
| |
Collapse
|
99
|
Zakharova VV, Pletjushkina OY, Zinovkin RA, Popova EN, Chernyak BV. Mitochondria-Targeted Antioxidants and Uncouplers of Oxidative Phosphorylation in Treatment of the Systemic Inflammatory Response Syndrome (SIRS). J Cell Physiol 2016; 232:904-912. [DOI: 10.1002/jcp.25626] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 09/28/2016] [Indexed: 02/06/2023]
Affiliation(s)
- Vlada V. Zakharova
- Belozersky Institute of Physico-Chemical Biology; Lomonosov Moscow State University; Moscow Russia
- Faculty of Bioengineering and Bioinformatics; Lomonosov Moscow State University; Moscow Russia
| | - Olga Yu. Pletjushkina
- Belozersky Institute of Physico-Chemical Biology; Lomonosov Moscow State University; Moscow Russia
| | - Roman A. Zinovkin
- Belozersky Institute of Physico-Chemical Biology; Lomonosov Moscow State University; Moscow Russia
| | - Ekaterina N. Popova
- Belozersky Institute of Physico-Chemical Biology; Lomonosov Moscow State University; Moscow Russia
| | - Boris V. Chernyak
- Belozersky Institute of Physico-Chemical Biology; Lomonosov Moscow State University; Moscow Russia
| |
Collapse
|
100
|
Letts JA, Degliesposti G, Fiedorczuk K, Skehel M, Sazanov LA. Purification of Ovine Respiratory Complex I Results in a Highly Active and Stable Preparation. J Biol Chem 2016; 291:24657-24675. [PMID: 27672209 DOI: 10.1074/jbc.m116.735142] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 09/15/2016] [Indexed: 01/06/2023] Open
Abstract
NADH-ubiquinone oxidoreductase (complex I) is the largest (∼1 MDa) and the least characterized complex of the mitochondrial electron transport chain. Because of the ease of sample availability, previous work has focused almost exclusively on bovine complex I. However, only medium resolution structural analyses of this complex have been reported. Working with other mammalian complex I homologues is a potential approach for overcoming these limitations. Due to the inherent difficulty of expressing large membrane protein complexes, screening of complex I homologues is limited to large mammals reared for human consumption. The high sequence identity among these available sources may preclude the benefits of screening. Here, we report the characterization of complex I purified from Ovis aries (ovine) heart mitochondria. All 44 unique subunits of the intact complex were identified by mass spectrometry. We identified differences in the subunit composition of subcomplexes of ovine complex I as compared with bovine, suggesting differential stability of inter-subunit interactions within the complex. Furthermore, the 42-kDa subunit, which is easily lost from the bovine enzyme, remains tightly bound to ovine complex I. Additionally, we developed a novel purification protocol for highly active and stable mitochondrial complex I using the branched-chain detergent lauryl maltose neopentyl glycol. Our data demonstrate that, although closely related, significant differences exist between the biochemical properties of complex I prepared from ovine and bovine mitochondria and that ovine complex I represents a suitable alternative target for further structural studies.
Collapse
Affiliation(s)
- James A Letts
- From the Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria
| | - Gianluca Degliesposti
- the Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 0QH, United Kingdom, and
| | - Karol Fiedorczuk
- From the Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria,; the Medical Research Council Mitochondrial Biology Unit, Cambridge CB2 0XY, United Kingdom
| | - Mark Skehel
- the Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 0QH, United Kingdom, and
| | - Leonid A Sazanov
- From the Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria,.
| |
Collapse
|