1
|
Krizanac M, Štancl P, Mass-Sanchez PB, Karlić R, Moeckel D, Lammers T, Asimakopoulos A, Weiskirchen R. The influence of perilipin 5 deficiency on gut microbiome profiles in murine metabolic dysfunction-associated fatty liver disease (MAFLD) and MAFLD-hepatocellular carcinoma. Front Cell Infect Microbiol 2024; 14:1443654. [PMID: 39469452 PMCID: PMC11513398 DOI: 10.3389/fcimb.2024.1443654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 09/20/2024] [Indexed: 10/30/2024] Open
Abstract
Introduction Metabolic dysfunction-associated fatty liver disease (MAFLD) has emerged as the leading cause of hepatocellular carcinoma (HCC) worldwide. Over the years, Perilipin 5 (PLIN5) has been recognized as a key regulator of both MAFLD and HCC development. In our previous studies we demonstrated that deficiency in Plin5 reduces the severity of MAFLD and HCC in mice. Interestingly, it has been established that patients with MAFLD and HCC exhibit various changes in their gut microbiome profiles. The gut microbiome itself has been shown to play a role in modulating carcinogenesis and the immune response against cancer. Methods Therefore, we conducted a study to investigate the alterations in fecal microbiome composition in wild type (WT) and Plin5-deficient (Plin5 -/-) mice models of MAFLD and MAFLD-induced HCC (MAFLD-HCC). We utilized 16S rRNA gene sequencing analysis to profile the composition of gut bacteria in fecal samples. Results Notably, we discovered that the absence of Plin5 alone is already associated with changes in gut microbiota composition. Moreover, feeding the mice a Western diet (WD) resulted in additional microbial alterations. Interestingly, Plin5 -/- animals exhibited an enrichment of the beneficial taxa Lactobacillus in both animal models. Discussion Our findings identify Plin5 as a major regulator of gut microbiota during the development of MAFLD and MAFLD-HCC.
Collapse
Affiliation(s)
- Marinela Krizanac
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Aachen, Germany
| | - Paula Štancl
- Bioinformatics Group, Division of Molecular Biology, Department of Biology, Faculty of Science, University of Zagreb, Zagreb, Croatia
| | - Paola Berenice Mass-Sanchez
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Aachen, Germany
| | - Rosa Karlić
- Bioinformatics Group, Division of Molecular Biology, Department of Biology, Faculty of Science, University of Zagreb, Zagreb, Croatia
| | - Diana Moeckel
- Institute for Experimental Molecular Imaging, RWTH Aachen, Aachen, Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, RWTH Aachen, Aachen, Germany
| | - Anastasia Asimakopoulos
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Aachen, Germany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Aachen, Germany
| |
Collapse
|
2
|
Azevedo LG, Sosa E, de Queiroz ATL, Barral A, Wheeler RJ, Nicolás MF, Farias LP, Do Porto DF, Ramos PIP. High-throughput prioritization of target proteins for development of new antileishmanial compounds. Int J Parasitol Drugs Drug Resist 2024; 25:100538. [PMID: 38669848 PMCID: PMC11068527 DOI: 10.1016/j.ijpddr.2024.100538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 03/11/2024] [Accepted: 04/04/2024] [Indexed: 04/28/2024]
Abstract
Leishmaniasis, a vector-borne disease, is caused by the infection of Leishmania spp., obligate intracellular protozoan parasites. Presently, human vaccines are unavailable, and the primary treatment relies heavily on systemic drugs, often presenting with suboptimal formulations and substantial toxicity, making new drugs a high priority for LMIC countries burdened by the disease, but a low priority in the agenda of most pharmaceutical companies due to unattractive profit margins. New ways to accelerate the discovery of new, or the repositioning of existing drugs, are needed. To address this challenge, our study aimed to identify potential protein targets shared among clinically-relevant Leishmania species. We employed a subtractive proteomics and comparative genomics approach, integrating high-throughput multi-omics data to classify these targets based on different druggability metrics. This effort resulted in the ranking of 6502 ortholog groups of protein targets across 14 pathogenic Leishmania species. Among the top 20 highly ranked groups, metabolic processes known to be attractive drug targets, including the ubiquitination pathway, aminoacyl-tRNA synthetases, and purine synthesis, were rediscovered. Additionally, we unveiled novel promising targets such as the nicotinate phosphoribosyltransferase enzyme and dihydrolipoamide succinyltransferases. These groups exhibited appealing druggability features, including less than 40% sequence identity to the human host proteome, predicted essentiality, structural classification as highly druggable or druggable, and expression levels above the 50th percentile in the amastigote form. The resources presented in this work also represent a comprehensive collection of integrated data regarding trypanosomatid biology.
Collapse
Affiliation(s)
- Lucas G Azevedo
- Center for Data and Knowledge Integration for Health (CIDACS), Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (Fiocruz Bahia), Salvador, Bahia, Brazil; Post-graduate Program in Biotechnology and Investigative Medicine, Instituto Gonçalo Moniz, Salvador, Bahia, Brazil.
| | - Ezequiel Sosa
- Universidad de Buenos Aires, Buenos Aires, Argentina.
| | - Artur T L de Queiroz
- Center for Data and Knowledge Integration for Health (CIDACS), Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (Fiocruz Bahia), Salvador, Bahia, Brazil; Post-graduate Program in Biotechnology and Investigative Medicine, Instituto Gonçalo Moniz, Salvador, Bahia, Brazil.
| | - Aldina Barral
- Laboratório de Medicina e Saúde Pública de Precisão (MeSP2), Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (Fiocruz Bahia), Salvador, Bahia, Brazil.
| | - Richard J Wheeler
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom.
| | - Marisa F Nicolás
- Laboratório Nacional de Computação Científica, Petrópolis, Rio de Janeiro, Brazil.
| | - Leonardo P Farias
- Post-graduate Program in Biotechnology and Investigative Medicine, Instituto Gonçalo Moniz, Salvador, Bahia, Brazil; Laboratório de Medicina e Saúde Pública de Precisão (MeSP2), Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (Fiocruz Bahia), Salvador, Bahia, Brazil.
| | | | - Pablo Ivan P Ramos
- Center for Data and Knowledge Integration for Health (CIDACS), Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (Fiocruz Bahia), Salvador, Bahia, Brazil; Post-graduate Program in Biotechnology and Investigative Medicine, Instituto Gonçalo Moniz, Salvador, Bahia, Brazil.
| |
Collapse
|
3
|
Kagan VE, Straub AC, Tyurina YY, Kapralov AA, Hall R, Wenzel SE, Mallampalli RK, Bayir H. Vitamin E/Coenzyme Q-Dependent "Free Radical Reductases": Redox Regulators in Ferroptosis. Antioxid Redox Signal 2024; 40:317-328. [PMID: 37154783 PMCID: PMC10890965 DOI: 10.1089/ars.2022.0154] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 03/10/2023] [Accepted: 04/08/2023] [Indexed: 05/10/2023]
Abstract
Significance: Lipid peroxidation and its products, oxygenated polyunsaturated lipids, act as essential signals coordinating metabolism and physiology and can be deleterious to membranes when they accumulate in excessive amounts. Recent Advances: There is an emerging understanding that regulation of polyunsaturated fatty acid (PUFA) phospholipid peroxidation, particularly of PUFA-phosphatidylethanolamine, is important in a newly discovered type of regulated cell death, ferroptosis. Among the most recently described regulatory mechanisms is the ferroptosis suppressor protein, which controls the peroxidation process due to its ability to reduce coenzyme Q (CoQ). Critical Issues: In this study, we reviewed the most recent data in the context of the concept of free radical reductases formulated in the 1980-1990s and focused on enzymatic mechanisms of CoQ reduction in different membranes (e.g., mitochondrial, endoplasmic reticulum, and plasma membrane electron transporters) as well as TCA cycle components and cytosolic reductases capable of recycling the high antioxidant efficiency of the CoQ/vitamin E system. Future Directions: We highlight the importance of individual components of the free radical reductase network in regulating the ferroptotic program and defining the sensitivity/tolerance of cells to ferroptotic death. Complete deciphering of the interactive complexity of this system may be important for designing effective antiferroptotic modalities. Antioxid. Redox Signal. 40, 317-328.
Collapse
Affiliation(s)
- Valerian E. Kagan
- Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Environmental Health and Pharmacology and Chemical Biology and University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Radiation Oncology and Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Adam C. Straub
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Yulia Y. Tyurina
- Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Environmental Health and Pharmacology and Chemical Biology and University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Alexandr A. Kapralov
- Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Environmental Health and Pharmacology and Chemical Biology and University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Robert Hall
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Sally E. Wenzel
- Department of Environmental Health and Pharmacology and Chemical Biology and University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rama K. Mallampalli
- Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Hülya Bayir
- Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, Children's Hospital Neuroscience Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pediatrics, Columbia University, New York, New York, USA
| |
Collapse
|
4
|
Novák LVF, Treitli SC, Pyrih J, Hałakuc P, Pipaliya SV, Vacek V, Brzoň O, Soukal P, Eme L, Dacks JB, Karnkowska A, Eliáš M, Hampl V. Genomics of Preaxostyla Flagellates Illuminates the Path Towards the Loss of Mitochondria. PLoS Genet 2023; 19:e1011050. [PMID: 38060519 PMCID: PMC10703272 DOI: 10.1371/journal.pgen.1011050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 11/03/2023] [Indexed: 12/18/2023] Open
Abstract
The notion that mitochondria cannot be lost was shattered with the report of an oxymonad Monocercomonoides exilis, the first eukaryote arguably without any mitochondrion. Yet, questions remain about whether this extends beyond the single species and how this transition took place. The Oxymonadida is a group of gut endobionts taxonomically housed in the Preaxostyla which also contains free-living flagellates of the genera Trimastix and Paratrimastix. The latter two taxa harbour conspicuous mitochondrion-related organelles (MROs). Here we report high-quality genome and transcriptome assemblies of two Preaxostyla representatives, the free-living Paratrimastix pyriformis and the oxymonad Blattamonas nauphoetae. We performed thorough comparisons among all available genomic and transcriptomic data of Preaxostyla to further decipher the evolutionary changes towards amitochondriality, endobiosis, and unstacked Golgi. Our results provide insights into the metabolic and endomembrane evolution, but most strikingly the data confirm the complete loss of mitochondria for all three oxymonad species investigated (M. exilis, B. nauphoetae, and Streblomastix strix), suggesting the amitochondriate status is common to a large part if not the whole group of Oxymonadida. This observation moves this unique loss to 100 MYA when oxymonad lineage diversified.
Collapse
Affiliation(s)
- Lukáš V. F. Novák
- Charles University, Faculty of Science, Department of Parasitology, BIOCEV, Vestec, Czech Republic
- Université de Bretagne Occidentale, CNRS, Unité Biologie et Ecologie des Ecosystèmes Marins Profonds BEEP, IUEM, Plouzané, France
| | - Sebastian C. Treitli
- Charles University, Faculty of Science, Department of Parasitology, BIOCEV, Vestec, Czech Republic
- RG Insect Gut Microbiology and Symbiosis, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Jan Pyrih
- Charles University, Faculty of Science, Department of Parasitology, BIOCEV, Vestec, Czech Republic
| | - Paweł Hałakuc
- Institute of Evolutionary Biology, Biological and Chemical Research Centre, Faculty of Biology, University of Warsaw, Poland
| | - Shweta V. Pipaliya
- Division of Infectious Diseases, Department of Medicine, University of Alberta, Edmonton, Canada
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland; Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Vojtěch Vacek
- Charles University, Faculty of Science, Department of Parasitology, BIOCEV, Vestec, Czech Republic
| | - Ondřej Brzoň
- Charles University, Faculty of Science, Department of Parasitology, BIOCEV, Vestec, Czech Republic
| | - Petr Soukal
- Charles University, Faculty of Science, Department of Parasitology, BIOCEV, Vestec, Czech Republic
| | - Laura Eme
- Ecology, Systematics, and Evolution Unit, Université Paris-Saclay, CNRS, Orsay, France
| | - Joel B. Dacks
- Division of Infectious Diseases, Department of Medicine, University of Alberta, Edmonton, Canada
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, České Budějovice, Czechia
| | - Anna Karnkowska
- Institute of Evolutionary Biology, Biological and Chemical Research Centre, Faculty of Biology, University of Warsaw, Poland
| | - Marek Eliáš
- University of Ostrava, Faculty of Science, Department of Biology and Ecology, Ostrava, Czech Republic
| | - Vladimír Hampl
- Charles University, Faculty of Science, Department of Parasitology, BIOCEV, Vestec, Czech Republic
| |
Collapse
|
5
|
Yan LJ, Wang Y. Roles of Dihydrolipoamide Dehydrogenase in Health and Disease. Antioxid Redox Signal 2023; 39:794-806. [PMID: 37276180 PMCID: PMC10615065 DOI: 10.1089/ars.2022.0181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 05/22/2023] [Accepted: 05/28/2023] [Indexed: 06/07/2023]
Abstract
Significance: Dihydrolipoamide dehydrogenase (DLDH) is a flavin-dependent disulfide oxidoreductase. The active form of DLDH is a stable homodimer, and its deficiencies have been linked to numerous metabolic disorders. A better understanding of redox and nonredox features of DLDH may reveal druggable targets for disease interventions or preventions. Recent Advances: In this article, the authors review the different roles of DLDH in selected pathological conditions, including its deficiency in humans, its role in stroke and neuroprotection, skin photoaging, Alzheimer's disease, and DLDH as a nondehydrogenating protein, and construction of genetically modified DLDH animal models for further studying the role of DLDH in specific pathological conditions. DLDH is also vulnerable to oxidative modifications in pathological conditions. Critical Issues: Novel animal models need to be constructed using gene knockdown techniques to investigate the redox- and nonredox roles of DLDH in related metabolic diseases. Specific small-molecule DLDH inhibitors need to be discovered. The relationship between modifications of specific amino acid residues in DLDH and given pathological conditions is an interesting area that remains to be comprehensively evaluated. Future Directions: Cell-specific or tissue-specific knockdown of DLDH creating specific pathological conditions will provide more insights into the mechanisms, whereby DLDH may have therapeutic values under a variety of pathological conditions. Antioxid. Redox Signal. 39, 794-806.
Collapse
Affiliation(s)
- Liang-Jun Yan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Yucheng Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|
6
|
Korczowska-Łącka I, Słowikowski B, Piekut T, Hurła M, Banaszek N, Szymanowicz O, Jagodziński PP, Kozubski W, Permoda-Pachuta A, Dorszewska J. Disorders of Endogenous and Exogenous Antioxidants in Neurological Diseases. Antioxidants (Basel) 2023; 12:1811. [PMID: 37891890 PMCID: PMC10604347 DOI: 10.3390/antiox12101811] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/19/2023] [Accepted: 09/27/2023] [Indexed: 10/29/2023] Open
Abstract
In diseases of the central nervous system, such as Alzheimer's disease (AD), Parkinson's disease (PD), stroke, amyotrophic lateral sclerosis (ALS), Huntington's disease (HD), and even epilepsy and migraine, oxidative stress load commonly surpasses endogenous antioxidative capacity. While oxidative processes have been robustly implicated in the pathogenesis of these diseases, the significance of particular antioxidants, both endogenous and especially exogenous, in maintaining redox homeostasis requires further research. Among endogenous antioxidants, enzymes such as catalase, superoxide dismutase, and glutathione peroxidase are central to disabling free radicals, thereby preventing oxidative damage to cellular lipids, proteins, and nucleic acids. Whether supplementation with endogenously occurring antioxidant compounds such as melatonin and glutathione carries any benefit, however, remains equivocal. Similarly, while the health benefits of certain exogenous antioxidants, including ascorbic acid (vitamin C), carotenoids, polyphenols, sulforaphanes, and anthocyanins are commonly touted, their clinical efficacy and effectiveness in particular neurological disease contexts need to be more robustly defined. Here, we review the current literature on the cellular mechanisms mitigating oxidative stress and comment on the possible benefit of the most common exogenous antioxidants in diseases such as AD, PD, ALS, HD, stroke, epilepsy, and migraine. We selected common neurological diseases of a basically neurodegenerative nature.
Collapse
Affiliation(s)
- Izabela Korczowska-Łącka
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland (M.H.)
| | - Bartosz Słowikowski
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (B.S.); (P.P.J.)
| | - Thomas Piekut
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland (M.H.)
| | - Mikołaj Hurła
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland (M.H.)
| | - Natalia Banaszek
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland (M.H.)
| | - Oliwia Szymanowicz
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland (M.H.)
| | - Paweł P. Jagodziński
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (B.S.); (P.P.J.)
| | - Wojciech Kozubski
- Chair and Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland
| | - Agnieszka Permoda-Pachuta
- Department of Psychiatry, Psychotherapy and Early Intervention, Medical University of Lublin, 20-059 Lublin, Poland
| | - Jolanta Dorszewska
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland (M.H.)
| |
Collapse
|
7
|
Granata C, Caruana NJ, Botella J, Jamnick NA, Huynh K, Kuang J, Janssen HA, Reljic B, Mellett NA, Laskowski A, Stait TL, Frazier AE, Coughlan MT, Meikle PJ, Thorburn DR, Stroud DA, Bishop DJ. High-intensity training induces non-stoichiometric changes in the mitochondrial proteome of human skeletal muscle without reorganisation of respiratory chain content. Nat Commun 2021; 12:7056. [PMID: 34862379 PMCID: PMC8642543 DOI: 10.1038/s41467-021-27153-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 10/26/2021] [Indexed: 12/28/2022] Open
Abstract
Mitochondrial defects are implicated in multiple diseases and aging. Exercise training is an accessible, inexpensive therapeutic intervention that can improve mitochondrial bioenergetics and quality of life. By combining multiple omics techniques with biochemical and in silico normalisation, we removed the bias arising from the training-induced increase in mitochondrial content to unearth an intricate and previously undemonstrated network of differentially prioritised mitochondrial adaptations. We show that changes in hundreds of transcripts, proteins, and lipids are not stoichiometrically linked to the overall increase in mitochondrial content. Our findings suggest enhancing electron flow to oxidative phosphorylation (OXPHOS) is more important to improve ATP generation than increasing the abundance of the OXPHOS machinery, and do not support the hypothesis that training-induced supercomplex formation enhances mitochondrial bioenergetics. Our study provides an analytical approach allowing unbiased and in-depth investigations of training-induced mitochondrial adaptations, challenging our current understanding, and calling for careful reinterpretation of previous findings.
Collapse
Affiliation(s)
- Cesare Granata
- Institute for Health and Sport (iHeS), Victoria University, Melbourne, VIC, 3011, Australia.
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia.
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University, 40225, Düsseldorf, Germany.
| | - Nikeisha J Caruana
- Institute for Health and Sport (iHeS), Victoria University, Melbourne, VIC, 3011, Australia
- Department of Biochemistry and Pharmacology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Javier Botella
- Institute for Health and Sport (iHeS), Victoria University, Melbourne, VIC, 3011, Australia
| | - Nicholas A Jamnick
- Institute for Health and Sport (iHeS), Victoria University, Melbourne, VIC, 3011, Australia
- Metabolic Research Unit, School of Medicine and Institute for Mental and Physical Health and Clinical Translation (iMPACT), Deakin University, Geelong, VIC, Australia
| | - Kevin Huynh
- Baker Heart & Diabetes Institute, Melbourne, VIC, 3004, Australia
| | - Jujiao Kuang
- Institute for Health and Sport (iHeS), Victoria University, Melbourne, VIC, 3011, Australia
| | - Hans A Janssen
- Institute for Health and Sport (iHeS), Victoria University, Melbourne, VIC, 3011, Australia
| | - Boris Reljic
- Department of Biochemistry and Pharmacology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, 3010, Australia
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, 3800, Melbourne, Australia
| | | | - Adrienne Laskowski
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Tegan L Stait
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, 3052, Australia
| | - Ann E Frazier
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, 3052, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Melinda T Coughlan
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
- Baker Heart & Diabetes Institute, Melbourne, VIC, 3004, Australia
| | - Peter J Meikle
- Baker Heart & Diabetes Institute, Melbourne, VIC, 3004, Australia
| | - David R Thorburn
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, 3052, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC, 3052, Australia
- Victorian Clinical Genetics Services, Royal Children's Hospital, Melbourne, VIC, 3052, Australia
| | - David A Stroud
- Department of Biochemistry and Pharmacology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, 3010, Australia.
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, 3052, Australia.
| | - David J Bishop
- Institute for Health and Sport (iHeS), Victoria University, Melbourne, VIC, 3011, Australia.
| |
Collapse
|
8
|
Yuan S, Schmidt HM, Wood KC, Straub AC. CoenzymeQ in cellular redox regulation and clinical heart failure. Free Radic Biol Med 2021; 167:321-334. [PMID: 33753238 DOI: 10.1016/j.freeradbiomed.2021.03.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/22/2021] [Accepted: 03/08/2021] [Indexed: 12/12/2022]
Abstract
Coenzyme Q (CoQ) is ubiquitously embedded in lipid bilayers of various cellular organelles. As a redox cycler, CoQ shuttles electrons between mitochondrial complexes and extramitochondrial reductases and oxidases. In this way, CoQ is crucial for maintaining the mitochondrial function, ATP synthesis, and redox homeostasis. Cardiomyocytes have a high metabolic rate and rely heavily on mitochondria to provide energy. CoQ levels, in both plasma and the heart, correlate with heart failure in patients, indicating that CoQ is critical for cardiac function. Moreover, CoQ supplementation in clinics showed promising results for treating heart failure. This review provides a comprehensive view of CoQ metabolism and its interaction with redox enzymes and reactive species. We summarize the clinical trials and applications of CoQ in heart failure and discuss the caveats and future directions to improve CoQ therapeutics.
Collapse
Affiliation(s)
- Shuai Yuan
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Heidi M Schmidt
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Katherine C Wood
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Adam C Straub
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
9
|
Reactive Oxygen Species and Their Involvement in Red Blood Cell Damage in Chronic Kidney Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6639199. [PMID: 33708334 PMCID: PMC7932781 DOI: 10.1155/2021/6639199] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/25/2021] [Accepted: 02/08/2021] [Indexed: 02/06/2023]
Abstract
Reactive oxygen species (ROS) released in cells are signaling molecules but can also modify signaling proteins. Red blood cells perform a major role in maintaining the balance of the redox in the blood. The main cytosolic protein of RBC is hemoglobin (Hb), which accounts for 95-97%. Most other proteins are involved in protecting the blood cell from oxidative stress. Hemoglobin is a major factor in initiating oxidative stress within the erythrocyte. RBCs can also be damaged by exogenous oxidants. Hb autoxidation leads to the generation of a superoxide radical, of which the catalyzed or spontaneous dismutation produces hydrogen peroxide. Both oxidants induce hemichrome formation, heme degradation, and release of free iron which is a catalyst for free radical reactions. To maintain the redox balance, appropriate antioxidants are present in the cytosol, such as superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), and peroxiredoxin 2 (PRDX2), as well as low molecular weight antioxidants: glutathione, ascorbic acid, lipoic acid, α-tocopherol, β-carotene, and others. Redox imbalance leads to oxidative stress and may be associated with overproduction of ROS and/or insufficient capacity of the antioxidant system. Oxidative stress performs a key role in CKD as evidenced by the high level of markers associated with oxidative damage to proteins, lipids, and DNA in vivo. In addition to the overproduction of ROS, a reduced antioxidant capacity is observed, associated with a decrease in the activity of SOD, GPx, PRDX2, and low molecular weight antioxidants. In addition, hemodialysis is accompanied by oxidative stress in which low-biocompatibility dialysis membranes activate phagocytic cells, especially neutrophils and monocytes, leading to a respiratory burst. This review shows the production of ROS under normal conditions and CKD and its impact on disease progression. Oxidative damage to red blood cells (RBCs) in CKD and their contribution to cardiovascular disease are also discussed.
Collapse
|
10
|
Okoye CN, Stevens D, Kamunde C. Modulation of mitochondrial site-specific hydrogen peroxide efflux by exogenous stressors. Free Radic Biol Med 2021; 164:439-456. [PMID: 33383085 DOI: 10.1016/j.freeradbiomed.2020.12.234] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/18/2020] [Accepted: 12/21/2020] [Indexed: 12/16/2022]
Abstract
Oxygen (O2) deprivation and metals are common environmental stressors and their exposure to aquatic organisms can induce oxidative stress by disrupting cellular reactive oxygen species (ROS) homeostasis. Mitochondria are a major source of ROS in the cell wherein a dozen sites located on enzymes of the electron transport system (ETS) and substrate oxidation produce superoxide anion radicals (O2˙‾) or hydrogen peroxide (H2O2). Sites located on ETS enzymes can generate ROS by forward electron transfer (FET) and reverse electron transfer (RET) reactions; however, knowledge of how exogenous stressors modulate site-specific ROS production is limited. We investigated the effects of anoxia-reoxygenation and cadmium (Cd) on H2O2 emission in fish liver mitochondria oxidizing glutamate-malate, succinate or palmitoylcarnitine-malate. We find that anoxia-reoxygenation attenuates H2O2 emission while the effect of Cd depends on the substrate, with monotonic responses for glutamate-malate and palmitoylcarnitine-malate, and a biphasic response for succinate. Anoxia-reoxygenation exerts a substrate-dependent inhibition of mitochondrial respiration which is more severe with palmitoylcarnitine-malate compared with succinate or glutamate-malate. Additionally, specific mitochondrial ROS-emitting sites were sequestered using blockers of electron transfer and the effects of anoxia-reoxygenation and Cd on H2O2 emission were evaluated. Here, we find that site-specific H2O2 emission capacities depend on the substrate and the direction of electron flow. Moreover, anoxia-reoxygenation alters site-specific H2O2 emission rates during succinate and glutamate-malate oxidation whereas Cd imposes monotonic or biphasic H2O2 emission responses depending on the substrate and site. Contrary to our expectation, anoxia-reoxygenation blunts the effect of Cd. These results suggest that the effect of exogenous stressors on mitochondrial oxidant production is governed by their impact on energy conversion reactions and mitochondrial redox poise. Moreover, direct increased ROS production seemingly does not explain the increased adverse effects associated with combined exposure of aquatic organisms to Cd and low dissolved oxygen levels.
Collapse
Affiliation(s)
- Chidozie N Okoye
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, PE, C1A 4P3, Canada; Department of Veterinary Obstetrics and Reproductive Diseases. Faculty of Veterinary Medicine, University of Nigeria, Nsukka, Nigeria
| | - Don Stevens
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, PE, C1A 4P3, Canada
| | - Collins Kamunde
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, PE, C1A 4P3, Canada.
| |
Collapse
|
11
|
Gazaryan IG, Shchedrina VA, Klyachko NL, Zakhariants AA, Kazakov SV, Brown AM. Zinc Switch in Pig Heart Lipoamide Dehydrogenase: Steady-State and Transient Kinetic Studies of the Diaphorase Reaction. BIOCHEMISTRY (MOSCOW) 2020; 85:908-919. [PMID: 33045951 DOI: 10.1134/s0006297920080064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Elevation of intracellular Zn2+ following ischemia contributes to cell death by affecting mitochondrial function. Zn2+ is a differential regulator of the mitochondrial enzyme lipoamide dehydrogenase (LADH) at physiological concentrations (Ka = 0.1 µM free zinc), inhibiting lipoamide and accelerating NADH dehydrogenase activities. These differential effects have been attributed to coordination of Zn2+ by LADH active-site cysteines. A detailed kinetic mechanism has now been developed for the diaphorase (NADH-dehydrogenase) reaction catalyzed by pig heart LADH using 2,6-dichlorophenol-indophenol (DCPIP) as a model quinone electron acceptor. Anaerobic stopped-flow experiments show that two-electron reduced LADH is 15-25-fold less active towards DCPIP reduction than four-electron reduced enzyme, or Zn2+-modified reduced LADH (the corresponding values of the rate constants are (6.5 ± 1.5) × 103 M-1·s-1, (9 ± 2) × 104 M-1·s-1, and (1.6 ± 0.5) × 105 M-1·s-1, respectively). Steady-state kinetic studies with different diaphorase substrates show that Zn2+ accelerates reaction rates exclusively for two-electron acceptors (duroquinone, DCPIP), but not for one-electron acceptors (benzoquinone, ubiquinone, ferricyanide). This implies that the two-electron reduced form of LADH, prevalent at low NADH levels, is a poor two-electron donor compared to the four-electron reduced or Zn2+-modified reduced LADH forms. These data suggest that zinc binding to the active-site thiols switches the enzyme from one- to two-electron donor mode. This zinc-activated switch has the potential to alter the ratio of superoxide and H2O2 generated by the LADH oxidase activity.
Collapse
Affiliation(s)
- I G Gazaryan
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY 10605, USA.,Department of Chemistry and Physical Sciences, Dyson College of Arts and Sciences, Pace University, Pleasantville, NY 10570, USA.,Department of Chemical Enzymology, Lomonosov Moscow State University, Moscow, 119899, Russia.,Bach Institute of Biochemistry, Federal Research Centre "Fundamentals of Biotechnology", Russian Academy of Sciences, Moscow, 119071, Russia
| | - V A Shchedrina
- Department of Chemical Enzymology, Lomonosov Moscow State University, Moscow, 119899, Russia
| | - N L Klyachko
- Department of Chemical Enzymology, Lomonosov Moscow State University, Moscow, 119899, Russia.,Derzhavin Tambov State University, Tambov, 392000, Russia
| | - A A Zakhariants
- Bach Institute of Biochemistry, Federal Research Centre "Fundamentals of Biotechnology", Russian Academy of Sciences, Moscow, 119071, Russia.,Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
| | - S V Kazakov
- Department of Chemistry and Physical Sciences, Dyson College of Arts and Sciences, Pace University, Pleasantville, NY 10570, USA
| | - A M Brown
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY 10605, USA.
| |
Collapse
|
12
|
Lee KH, Cha M, Lee BH. Neuroprotective Effect of Antioxidants in the Brain. Int J Mol Sci 2020; 21:ijms21197152. [PMID: 32998277 PMCID: PMC7582347 DOI: 10.3390/ijms21197152] [Citation(s) in RCA: 263] [Impact Index Per Article: 52.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/18/2020] [Accepted: 09/23/2020] [Indexed: 12/29/2022] Open
Abstract
The brain is vulnerable to excessive oxidative insults because of its abundant lipid content, high energy requirements, and weak antioxidant capacity. Reactive oxygen species (ROS) increase susceptibility to neuronal damage and functional deficits, via oxidative changes in the brain in neurodegenerative diseases. Overabundance and abnormal levels of ROS and/or overload of metals are regulated by cellular defense mechanisms, intracellular signaling, and physiological functions of antioxidants in the brain. Single and/or complex antioxidant compounds targeting oxidative stress, redox metals, and neuronal cell death have been evaluated in multiple preclinical and clinical trials as a complementary therapeutic strategy for combating oxidative stress associated with neurodegenerative diseases. Herein, we present a general analysis and overview of various antioxidants and suggest potential courses of antioxidant treatments for the neuroprotection of the brain from oxidative injury. This review focuses on enzymatic and non-enzymatic antioxidant mechanisms in the brain and examines the relative advantages and methodological concerns when assessing antioxidant compounds for the treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Kyung Hee Lee
- Department of Dental Hygiene, Division of Health Science, Dongseo University, Busan 47011, Korea;
| | - Myeounghoon Cha
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Korea;
| | - Bae Hwan Lee
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Korea;
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
- Correspondence: ; Tel.: +82-2-2228-1711
| |
Collapse
|
13
|
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
|
14
|
Takahashi T, Mine Y, Okamoto T. Extracellular coenzyme Q 10 (CoQ 10) is reduced to ubiquinol-10 by intact Hep G2 cells independent of intracellular CoQ 10 reduction. Arch Biochem Biophys 2019; 672:108067. [PMID: 31400302 DOI: 10.1016/j.abb.2019.108067] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 08/03/2019] [Accepted: 08/06/2019] [Indexed: 11/29/2022]
Abstract
Coenzyme Q10 (CoQ10) is an essential factor in the mitochondrial respiratory chain and is closely associated with ATP production in humans. It is known that orally administered CoQ10 in humans is rapidly reduced, and most is detected as a reduced form, ubiquinol-10 (CoQ10H2), in serum. However, the mechanism of exogenous CoQ10 reduction in vivo is unclear. Therefore, in order to clarify how CoQ10 is reduced to CoQ10H2, we conducted a study using human liver cancer cell line Hep G2 cells, which show strong intracellular CoQ10-reducing activity. When intact cells were incubated with CoQ10, the exogenously added CoQ10 was incorporated into the cells, time-, concentration-, and temperature-dependently, and 50-80% of that was detected as CoQ10H2. On the other hand, a part of the extracellular CoQ10 was also detected as CoQ10H2, and the amount was greater than that of the intracellular CoQ10H2. Furthermore, the CoQ10-loaded cells did not leak the intracellular CoQ10H2 (or CoQ10) to the outside of the cells, and modulation of the extracellular CoQ10H2 amount had little effect on the intracellular CoQ10 or CoQ10H2 contents, suggesting the existence of an individual mechanism of CoQ10 reduction inside and outside the cells. Moreover, intact cells could reduce CoQ10 in low-density lipoprotein to CoQ10H2. Therefore, we concluded that a novel CoQ10-reducing mechanism may exist in the plasma membrane, probably the outer surface, of Hep G2 cells, and it may work to reduce extracellular CoQ10 and/or maintain extracellular CoQ10H2.
Collapse
Affiliation(s)
- Takayuki Takahashi
- Laboratory of Biochemistry, Department of Health Science and Social Pharmacy, Faculty of Pharmaceutical Sciences, Kobe Gakuin University, 1-1-3 Minatojima, Chuo-ku, Kobe, 650-8586, Japan.
| | - Yukitoshi Mine
- Laboratory of Biochemistry, Department of Health Science and Social Pharmacy, Faculty of Pharmaceutical Sciences, Kobe Gakuin University, 1-1-3 Minatojima, Chuo-ku, Kobe, 650-8586, Japan
| | - Tadashi Okamoto
- Laboratory of Biochemistry, Department of Health Science and Social Pharmacy, Faculty of Pharmaceutical Sciences, Kobe Gakuin University, 1-1-3 Minatojima, Chuo-ku, Kobe, 650-8586, Japan
| |
Collapse
|
15
|
Yang X, Song J, Yan LJ. Chronic Inhibition of Mitochondrial Dihydrolipoamide Dehydrogenase (DLDH) as an Approach to Managing Diabetic Oxidative Stress. Antioxidants (Basel) 2019; 8:32. [PMID: 30717346 PMCID: PMC6406859 DOI: 10.3390/antiox8020032] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 01/21/2019] [Accepted: 01/28/2019] [Indexed: 12/27/2022] Open
Abstract
Mitochondrial dihydrolipoamide dehydrogenase (DLDH) is a redox enzyme involved in decarboxylation of pyruvate to form acetyl-CoA during the cascade of glucose metabolism and mitochondrial adenine triphosphate (ATP) production. Depending on physiological or pathophysiological conditions, DLDH can either enhance or attenuate the production of reactive oxygen species (ROS) and reactive nitrogen species. Recent research in our laboratory has demonstrated that inhibition of DLDH induced antioxidative responses and could serve as a protective approach against oxidative stress in stroke injury. In this perspective article, we postulated that chronic inhibition of DLDH could also attenuate oxidative stress in type 2 diabetes. We discussed DLDH-involving mitochondrial metabolic pathways and metabolic intermediates that could accumulate upon DLDH inhibition and their corresponding roles in abrogating oxidative stress in diabetes. We also discussed a couple of DLDH inhibitors that could be tested in animal models of type 2 diabetes. It is our belief that DLDH inhibition could be a novel approach to fighting type 2 diabetes.
Collapse
Affiliation(s)
- Xiaojuan Yang
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA.
| | - Jing Song
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA.
| | - Liang-Jun Yan
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA.
| |
Collapse
|
16
|
Laloo AE, Wei J, Wang D, Narayanasamy S, Vanwonterghem I, Waite D, Steen J, Kaysen A, Heintz-Buschart A, Wang Q, Schulz B, Nouwens A, Wilmes P, Hugenholtz P, Yuan Z, Bond PL. Mechanisms of Persistence of the Ammonia-Oxidizing Bacteria Nitrosomonas to the Biocide Free Nitrous Acid. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2018; 52:5386-5397. [PMID: 29620869 DOI: 10.1021/acs.est.7b04273] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Free nitrous acid (FNA) exerts a broad range of antimicrobial effects on bacteria, although susceptibility varies considerably among microorganisms. Among nitrifiers found in activated sludge of wastewater treatment processes (WWTPs), nitrite-oxidizing bacteria (NOB) are more susceptible to FNA compared to ammonia-oxidizing bacteria (AOB). This selective inhibition of NOB over AOB in WWTPs bypasses nitrate production and improves the efficiency and costs of the nitrogen removal process in both the activated sludge and anaerobic ammonium oxidation (Anammox) system. However, the molecular mechanisms governing this atypical tolerance of AOB to FNA have yet to be understood. Herein we investigate the varying effects of the antimicrobial FNA on activated sludge containing AOB and NOB using an integrated metagenomics and label-free quantitative sequential windowed acquisition of all theoretical fragment ion mass spectra (SWATH-MS) metaproteomic approach. The Nitrosomonas genus of AOB, on exposure to FNA, maintains internal homeostasis by upregulating a number of known oxidative stress enzymes, such as pteridine reductase and dihydrolipoyl dehydrogenase. Denitrifying enzymes were upregulated on exposure to FNA, suggesting the detoxification of nitrite to nitric oxide. Interestingly, proteins involved in stress response mechanisms, such as DNA and protein repair enzymes, phage prevention proteins, and iron transport proteins, were upregulated on exposure to FNA. In addition enzymes involved in energy generation were also upregulated on exposure to FNA. The total proteins specifically derived from the NOB genus Nitrobacter was low and, as such, did not allow for the elucidation of the response mechanism to FNA exposure. These findings give us an understanding of the adaptive mechanisms of tolerance within the AOB Nitrosomonas to the biocidal agent FNA.
Collapse
Affiliation(s)
- Andrew E Laloo
- Advanced Water Management Centre , The University of Queensland , St. Lucia , Brisbane , QLD 4072 , Australia
| | - Justin Wei
- Advanced Water Management Centre , The University of Queensland , St. Lucia , Brisbane , QLD 4072 , Australia
| | - Dongbo Wang
- College of Environmental Science and Engineering and Key Laboratory of Environmental Biology and Pollution Control, Ministry of Education , Hunan University , Changsa 410082 , China
| | - Shaman Narayanasamy
- Luxembourg Centre for Systems Biomedicine , Université du Luxembourg , L-4362 Esch-sur-Alzette , Luxembourg
| | - Inka Vanwonterghem
- Australian Centre for Ecogenomics (ACE), School of Chemistry and Molecular Bioscience , The University of Queensland , St. Lucia , Brisbane , QLD 4072 , Australia
| | - David Waite
- Australian Centre for Ecogenomics (ACE), School of Chemistry and Molecular Bioscience , The University of Queensland , St. Lucia , Brisbane , QLD 4072 , Australia
| | - Jason Steen
- Australian Centre for Ecogenomics (ACE), School of Chemistry and Molecular Bioscience , The University of Queensland , St. Lucia , Brisbane , QLD 4072 , Australia
| | - Anne Kaysen
- Luxembourg Centre for Systems Biomedicine , Université du Luxembourg , L-4362 Esch-sur-Alzette , Luxembourg
| | - Anna Heintz-Buschart
- Luxembourg Centre for Systems Biomedicine , Université du Luxembourg , L-4362 Esch-sur-Alzette , Luxembourg
| | - Qilin Wang
- Griffith School of Engineering & Centre for Clean Environment and Energy , Griffith University , Nathan , QLD 4111 , Australia
| | - Benjamin Schulz
- School of Chemistry and Molecular Biosciences , The University of Queensland , St. Lucia , Brisbane , QLD 4072 , Australia
| | - Amanda Nouwens
- School of Chemistry and Molecular Biosciences , The University of Queensland , St. Lucia , Brisbane , QLD 4072 , Australia
| | - Paul Wilmes
- Luxembourg Centre for Systems Biomedicine , Université du Luxembourg , L-4362 Esch-sur-Alzette , Luxembourg
| | - Philip Hugenholtz
- Australian Centre for Ecogenomics (ACE), School of Chemistry and Molecular Bioscience , The University of Queensland , St. Lucia , Brisbane , QLD 4072 , Australia
| | - Zhiguo Yuan
- Advanced Water Management Centre , The University of Queensland , St. Lucia , Brisbane , QLD 4072 , Australia
| | - Philip L Bond
- Advanced Water Management Centre , The University of Queensland , St. Lucia , Brisbane , QLD 4072 , Australia
| |
Collapse
|
17
|
Recombinant expression, characterization and application of a dihydrolipoamide dehydrogenase with diaphorase activity from Bacillus sphaericus. 3 Biotech 2017; 7:153. [PMID: 28597164 DOI: 10.1007/s13205-017-0763-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 02/02/2017] [Indexed: 10/19/2022] Open
Abstract
Diaphorases are flavin-containing enzymes with potential applications in biotransfomation reactions, biosensor design and in vitro diagnostic tests. In this communication, we describe recombinant expression, characterization and application of a lipoamide dehydrogenase (DLD) with diaphorase activity from a strain of Bacillus sphaericus. The DLD gene consisting of 1413 bp encoding a protein of 470 amino acids was expressed in Escherichia coli BL21 (DE3) and the recombinant enzyme was characterized. B. sphaericus DLD catalyzed the reduction of NAD+ by dihydrolipoamide and exhibited NADH-dependent diaphorase activity. The molecular weight of purified enzyme was about 50 kDa, and determined to be a monomeric protein. Diaphorase was active and stable from pH 7.0 to 9.0 with an optimal activity at pH 8.5. It showed its maximal activity at temperature of 30 °C and was almost stable at temperatures between 25 and 30 °C. Different metal ions and inhibitors showed no influence on the activity of target enzyme. The K m and V max values for NADH were estimated to be 0.33 mM and 200.0 U/ml, respectively. Moreover, recombinant B. sphaericus diaphorase exhibited considerable potential to be used as a component of diagnostic tests for the quantification of metabolites. In conclusion, considering the properties of diaphorase from B. sphaericus PAD-91, it can have potential application as a diagnostic enzyme.
Collapse
|
18
|
Suenobu T, Shibata S, Fukuzumi S. Catalytic Formation of Hydrogen Peroxide from Coenzyme NADH and Dioxygen with a Water-Soluble Iridium Complex and a Ubiquinone Coenzyme Analogue. Inorg Chem 2016; 55:7747-54. [PMID: 27403568 DOI: 10.1021/acs.inorgchem.6b01220] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
A ubiquinone coenzyme analogue (Q0: 2,3-dimethoxy-5-methyl-1,4-benzoquinone) was reduced by coenzyme NADH to yield the corresponding reduced form of Q0 (Q0H2) in the presence of a catalytic amount of a [C,N] cyclometalated organoiridium complex (1: [Ir(III)(Cp*)(4-(1H-pyrazol-1-yl-κN(2))benzoic acid-κC(3))(H2O)]2SO4) in water at ambient temperature as observed in the respiratory chain complex I (Complex I). In the catalytic cycle, the reduction of 1 by NADH produces the corresponding iridium hydride complex that in turn reduces Q0 to produce Q0H2. Q0H2 reduced dioxygen to yield hydrogen peroxide (H2O2) under slightly basic conditions. Catalytic generation of H2O2 was made possible in the reaction of O2 with NADH as the functional expression of NADH oxidase in white blood cells utilizing the redox cycle of Q0 as well as 1 for the first time in a nonenzymatic homogeneous reaction system.
Collapse
Affiliation(s)
- Tomoyoshi Suenobu
- Department of Material and Life Science, Graduate School of Engineering, Osaka University, ALCA and SENTAN, Japan Science and Technology , Suita, Osaka 565-0871, Japan
| | - Satoshi Shibata
- Department of Material and Life Science, Graduate School of Engineering, Osaka University, ALCA and SENTAN, Japan Science and Technology , Suita, Osaka 565-0871, Japan
| | - Shunichi Fukuzumi
- Department of Material and Life Science, Graduate School of Engineering, Osaka University, ALCA and SENTAN, Japan Science and Technology , Suita, Osaka 565-0871, Japan.,Department of Chemistry and Nano Science, Ewha Womans University , Seoul 120-750, Korea.,Faculty of Science and Engineering, Meijo University, ALCA and SENTAN, Japan Science and Technology Agency , Nagoya, Aichi 468-0073, Japan
| |
Collapse
|
19
|
Cao S, Liu Y, Wang H, Mao X, Chen J, Liu J, Xia Z, Zhang L, Liu X, Yu T. Ischemic postconditioning influences electron transport chain protein turnover in Langendorff-perfused rat hearts. PeerJ 2016; 4:e1706. [PMID: 26925330 PMCID: PMC4768691 DOI: 10.7717/peerj.1706] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 01/28/2016] [Indexed: 01/03/2023] Open
Abstract
Ischemia postconditioning (IPo) is a promising strategy in reducing myocardial ischemia reperfusion (I/R) injury (MIRI), but its specific molecular mechanism is incompletely understood. Langendorff-perfused isolated rat hearts were subjected to global I/R and received IPo in the absence or presence of the mitochondrial ATP-sensitive potassium channel (mitoKATP) blocker 5-hydroxydecanoate (5-HD). Myocardial mitochondria were extracted and mitochondrial comparative proteomics was analyzed. IPo significantly reduces post-ischemic myocardial infarction and improved cardiac function in I/R rat hearts, while 5-HD basically cancelled IPo's myocardial protective effect. Joint application of two-dimensional polyacrylamide gel electrophoresis (2DE) and MALDI-TOF MS identified eight differentially expressed proteins between groups. Expression of cardiac succinate dehydrogenase (ubiquinone) flavoprotein subunit (SDHA) increased more than two-fold after I/R, while IPo led to overexpression of dihydrolipoyl dehydrogenase (DLD), NADH dehydrogenase (ubiquinone) flavoprotein 1 and isoform CRA_b (NDUFV1). When the mitoKATP was blocked, MICOS complex subunit Mic60 (IMMT) and Stress-70 protein (Grp75) were over expressed, while DLDH, ATPase subunit A (ATPA) and rCG44606 were decreased. Seven of the differential proteins belong to electron transport chain (ETC) or metabolism regulating proteins, and five of them were induced by closing mitoKATP in I/R hearts. We thus conclude that IPo's myocardial protective effect relies on energy homeostasis regulation. DLD, SDHA, NDUFV1, Grp75, ATPA and rCG44606 may contribute to IPo's cardial protective effect.
Collapse
Affiliation(s)
- Song Cao
- Department of Anesthesiology, Zunyi Medical College, Zunyi, China; Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical College, Zunyi, China; Department of Pain Medicine, Affiliated Hospital of Zunyi Medical College, Zunyi, China
| | - Yun Liu
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical College, Zunyi, China; Research Center for Medicine & Biology, Zunyi Medical College, Zunyi, China
| | - Haiying Wang
- Department of Anesthesiology, Zunyi Medical College , Zunyi , China
| | - Xiaowen Mao
- Department of Anesthesiology, The University of Hong Kong , Hong Kong , China
| | - Jincong Chen
- Department of Anesthesiology, Zunyi Medical College , Zunyi , China
| | - Jiming Liu
- Department of Anesthesiology, Zunyi Medical College , Zunyi , China
| | - Zhengyuan Xia
- Department of Anesthesiology, The University of Hong Kong , Hong Kong , China
| | - Lin Zhang
- Department of Anesthesiology, Zunyi Medical College, Zunyi, China; Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical College, Zunyi, China
| | - Xingkui Liu
- Department of Anesthesiology, Zunyi Medical College , Zunyi , China
| | - Tian Yu
- Department of Anesthesiology, Zunyi Medical College, Zunyi, China; Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical College, Zunyi, China
| |
Collapse
|
20
|
Agarwal A, Sharma R, Durairajanayagam D, Ayaz A, Cui Z, Willard B, Gopalan B, Sabanegh E. Major protein alterations in spermatozoa from infertile men with unilateral varicocele. Reprod Biol Endocrinol 2015; 13:8. [PMID: 25890347 PMCID: PMC4383193 DOI: 10.1186/s12958-015-0007-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 02/11/2015] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND The etiology of varicocele, a common cause of male factor infertility, remains unclear. Proteomic changes responsible for the underlying pathology of unilateral varicocele have not been evaluated. The objective of this prospective study was to employ proteomic techniques and bioinformatic tools to identify and analyze proteins of interest in infertile men with unilateral varicocele. METHODS Spermatozoa from infertile men with unilateral varicocele (n=5) and from fertile men (control; n=5) were pooled in two groups respectively. Proteins were extracted and separated by 1-D SDS-PAGE. Bands were digested and identified on a LTQ-Orbitrap Elite hybrid mass spectrometer system. Bioinformatic analysis identified the pathways and functions of the differentially expressed proteins (DEP). RESULTS Sperm concentration, motility and morphology were lower, and reactive oxygen species levels were higher in unilateral varicocele patients compared to healthy controls. The total number of proteins identified were 1055, 1010 and 1042 in the fertile group, and 795, 713 and 763 proteins in the unilateral varicocele group. Of the 369 DEP between both groups, 120 proteins were unique to the fertile group and 38 proteins were unique to the unilateral varicocele group. Compared to the control group, 114 proteins were overexpressed while 97 proteins were underexpressed in the unilateral varicocele group. We have identified 29 proteins of interest that are involved in spermatogenesis and other fundamental reproductive events such as sperm maturation, acquisition of sperm motility, hyperactivation, capacitation, acrosome reaction and fertilization. The major functional pathways of the 359 DEP related to the unilateral varicocele group involve metabolism, disease, immune system, gene expression, signal transduction and apoptosis. Functional annotations showed that unilateral varicocele mostly affected small molecule biochemistry and post-translational modification proteins. Proteins expressed uniquely in the unilateral varicocele group were cysteine-rich secretory protein 2 precursor (CRISP2) and arginase-2 (ARG2). CONCLUSIONS The expression of these proteins of interest are altered and possibly functionally compromised in infertile men with unilateral varicocele. If validated, these proteins may lead to potential biomarker(s) and help better understand the mechanism involved in the pathophysiology of unilateral varicocele in infertile men.
Collapse
Affiliation(s)
- Ashok Agarwal
- Center for Reproductive Medicine, Glickman Urological & Kidney Institute, Cleveland Clinic, Mail Code X-11, 10681 Carnegie Avenue, Cleveland, OH, 44195, USA.
| | - Rakesh Sharma
- Center for Reproductive Medicine, Glickman Urological & Kidney Institute, Cleveland Clinic, Mail Code X-11, 10681 Carnegie Avenue, Cleveland, OH, 44195, USA.
| | - Damayanthi Durairajanayagam
- Center for Reproductive Medicine, Glickman Urological & Kidney Institute, Cleveland Clinic, Mail Code X-11, 10681 Carnegie Avenue, Cleveland, OH, 44195, USA.
| | - Ahmet Ayaz
- Center for Reproductive Medicine, Glickman Urological & Kidney Institute, Cleveland Clinic, Mail Code X-11, 10681 Carnegie Avenue, Cleveland, OH, 44195, USA.
| | - Zhihong Cui
- Center for Reproductive Medicine, Glickman Urological & Kidney Institute, Cleveland Clinic, Mail Code X-11, 10681 Carnegie Avenue, Cleveland, OH, 44195, USA.
| | - Belinda Willard
- Proteomics Research Core Services, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.
| | - Banu Gopalan
- Proteomics Research Core Services, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.
| | - Edmund Sabanegh
- Center for Reproductive Medicine, Glickman Urological & Kidney Institute, Cleveland Clinic, Mail Code X-11, 10681 Carnegie Avenue, Cleveland, OH, 44195, USA.
| |
Collapse
|
21
|
Grivennikova VG, Vinogradov AD. Mitochondrial production of reactive oxygen species. BIOCHEMISTRY (MOSCOW) 2014; 78:1490-511. [PMID: 24490736 DOI: 10.1134/s0006297913130087] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Numerous biochemical studies are aimed at elucidating the sources and mechanisms of formation of reactive oxygen species (ROS) because they are involved in cellular, organ-, and tissue-specific physiology. Mitochondria along with other cellular organelles of eukaryotes contribute significantly to ROS formation and utilization. This review is a critical account of the mitochondrial ROS production and methods for their registration. The physiological and pathophysiological significance of the mitochondrially produced ROS are discussed.
Collapse
Affiliation(s)
- V G Grivennikova
- Department of Biochemistry, Biological Faculty, Lomonosov Moscow State University, Moscow, 119991, Russia.
| | | |
Collapse
|
22
|
Thioredoxin system regulation in the central nervous system: experimental models and clinical evidence. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:590808. [PMID: 24723994 PMCID: PMC3958682 DOI: 10.1155/2014/590808] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 01/21/2014] [Accepted: 01/23/2014] [Indexed: 02/07/2023]
Abstract
The reactive oxygen species produced continuously during oxidative metabolism are generated at very high rates in the brain. Therefore, defending against oxidative stress is an essential task within the brain. An important cellular system against oxidative stress is the thioredoxin system (TS). TS is composed of thioredoxin, thioredoxin reductase, and NADPH. This review focuses on the evidence gathered in recent investigations into the central nervous system, specifically the different brain regions in which the TS is expressed. Furthermore, we address the conditions that modulate the thioredoxin system in both, animal models and the postmortem brains of human patients associated with the most common neurodegenerative disorders, in which the thioredoxin system could play an important part.
Collapse
|
23
|
Staniszewski K, Audi SH, Sepehr R, Jacobs ER, Ranji M. Surface fluorescence studies of tissue mitochondrial redox state in isolated perfused rat lungs. Ann Biomed Eng 2013; 41:827-36. [PMID: 23238793 PMCID: PMC3606690 DOI: 10.1007/s10439-012-0716-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Accepted: 11/28/2012] [Indexed: 12/24/2022]
Abstract
We designed a fiber-optic-based optoelectronic fluorometer to measure emitted fluorescence from the auto-fluorescent electron carriers NADH and FAD of the mitochondrial electron transport chain (ETC). The ratio of NADH to FAD is called the redox ratio (RR = NADH/FAD) and is an indicator of the oxidoreductive state of tissue. We evaluated the fluorometer by measuring the fluorescence intensities of NADH and FAD at the surface of isolated, perfused rat lungs. Alterations of lung mitochondrial metabolic state were achieved by the addition of rotenone (complex I inhibitor), potassium cyanide (KCN, complex IV inhibitor) and/or pentachlorophenol (PCP, uncoupler) into the perfusate recirculating through the lung. Rotenone- or KCN-containing perfusate increased RR by 21 and 30%, respectively. In contrast, PCP-containing perfusate decreased RR by 27%. These changes are consistent with the established effects of rotenone, KCN, and PCP on the redox status of the ETC. Addition of blood to perfusate quenched NADH and FAD signal, but had no effect on RR. This study demonstrates the capacity of fluorometry to detect a change in mitochondrial redox state in isolated perfused lungs, and suggests the potential of fluorometry for use in in vivo experiments to extract a sensitive measure of lung tissue health in real-time.
Collapse
Affiliation(s)
- Kevin Staniszewski
- Biophotonics Lab, Department of Electrical Engineering, University of Wisconsin Milwaukee, 3200 N Cramer St., Milwaukee, WI 53211
| | - Said H. Audi
- Department of Biomedical Engineering, Marquette University, 1515 West Wisconsin Avenue, Milwaukee, WI, 53233
| | - Reyhaneh Sepehr
- Biophotonics Lab, Department of Electrical Engineering, University of Wisconsin Milwaukee, 3200 N Cramer St., Milwaukee, WI 53211
| | - Elizabeth R. Jacobs
- Associate Chief of Staff, Research and Development, Clement J. Zablocki VA Medical Center, 5000 W. National Avenue Milwaukee, WI 5329 and Associate Dean Research, Medical College of Wisconsin
| | - Mahsa Ranji
- Biophotonics Lab, Department of Electrical Engineering, University of Wisconsin Milwaukee, 3200 N Cramer St., Milwaukee, WI 53211
| |
Collapse
|
24
|
Vaubel RA, Rustin P, Isaya G. Mutations in the dimer interface of dihydrolipoamide dehydrogenase promote site-specific oxidative damages in yeast and human cells. J Biol Chem 2011; 286:40232-45. [PMID: 21930696 PMCID: PMC3220568 DOI: 10.1074/jbc.m111.274415] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Revised: 09/13/2011] [Indexed: 12/31/2022] Open
Abstract
Dihydrolipoamide dehydrogenase (DLD) is a multifunctional protein well characterized as the E3 component of the pyruvate dehydrogenase and α-ketoglutarate dehydrogenase complexes. Previously, conditions predicted to destabilize the DLD dimer revealed that DLD could also function as a diaphorase and serine protease. However, the relevance of these cryptic activities remained undefined. We analyzed human DLD mutations linked to strikingly different clinical phenotypes, including E340K, D444V, R447G, and R460G in the dimer interface domain that are responsible for severe multisystem disorders of infancy and G194C in the NAD(+)-binding domain that is typically associated with milder presentations. In vitro, all of these mutations decreased to various degrees dihydrolipoamide dehydrogenase activity, whereas dimer interface mutations also enhanced proteolytic and/or diaphorase activity. Human DLD proteins carrying each individual mutation complemented fully the respiratory-deficient phenotype of yeast cells lacking endogenous DLD even when residual dihydrolipoamide dehydrogenase activity was as low as 21% of controls. However, under elevated oxidative stress, expression of DLD proteins with dimer interface mutations greatly accelerated the loss of respiratory function, resulting from enhanced oxidative damage to the lipoic acid cofactor of pyruvate dehydrogenase and α-ketoglutarate dehydrogenase and other mitochondrial targets. This effect was not observed with the G194C mutation or a mutation that disrupts the proteolytic active site of DLD. As in yeast, lipoic acid cofactor was damaged in human D444V-homozygous fibroblasts after exposure to oxidative stress. We conclude that the cryptic activities of DLD promote oxidative damage to neighboring molecules and thus contribute to the clinical severity of DLD mutations.
Collapse
Affiliation(s)
- Rachael A. Vaubel
- From the Departments of Pediatric and Adolescent Medicine and Biochemistry and Molecular Biology, Mayo Clinic, Rochester Minnesota 55905
| | - Pierre Rustin
- INSERM U676 Hôpital Robert Debré, F-75019 Paris, France, and
- Université Paris 7, Faculté de Médecine Denis Diderot, IFR02 Paris, France
| | - Grazia Isaya
- From the Departments of Pediatric and Adolescent Medicine and Biochemistry and Molecular Biology, Mayo Clinic, Rochester Minnesota 55905
| |
Collapse
|
25
|
Cai Y, Cao F, Wei K, Zhang G, Wu F. Genotypic dependent effect of exogenous glutathione on Cd-induced changes in proteins, ultrastructure and antioxidant defense enzymes in rice seedlings. JOURNAL OF HAZARDOUS MATERIALS 2011; 192:1056-66. [PMID: 21741764 DOI: 10.1016/j.jhazmat.2011.06.011] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Revised: 05/25/2011] [Accepted: 06/06/2011] [Indexed: 05/07/2023]
Abstract
Greenhouse hydroponic experiments were conducted using Cd-sensitive (cv. Xiushui63) and tolerant (Bing97252) rice genotypes to evaluate how different genotypes responded to Cd toxicity in presence of glutathione (GSH). Results showed that GSH alleviates Cd-toxicity, ameliorates Cd-induced damages on leaf/root ultrastructures. Nine proteins in roots were identified, using 2-DE coupled with mass spectrometry, whose expression were down-regulated in Xiushui63, up-regulated/unchanged in Bing97252 by Cd; coinstantaneously enhanced/unchanged in Cd+GSH over Cd alone treatment in both genotypes. They are l-ascorbate peroxidase, putative short-chain dehydrogenase/reductase, Glycolipid transfer protein, elongation factor, Os04g0652700, carbonic anhydrase, Os08g0374000, chitinase, and putative disease resistance response protein. Eight proteins in leaves with expression of increase in Bing97252 but down-regulate/unchange in Xiushui63, categorized as four groups of their functions: carbon metabolism, TCA cycle, photorespiration and RNA processing. Furthermore, we identified eight proteins with repressed expression in Cd-treated and up-regulated in Cd+GSH-treated rice leaves of Xiushui63.
Collapse
Affiliation(s)
- Yue Cai
- Department of Agronomy, College of Agriculture and Biotechnology, Zijingang Campus, Zhejiang University, Hangzhou 310058, PR China
| | | | | | | | | |
Collapse
|
26
|
Inhibitors of succinate: quinone reductase/Complex II regulate production of mitochondrial reactive oxygen species and protect normal cells from ischemic damage but induce specific cancer cell death. Pharm Res 2011; 28:2695-730. [PMID: 21863476 DOI: 10.1007/s11095-011-0566-7] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Accepted: 08/10/2011] [Indexed: 12/23/2022]
Abstract
Succinate:quinone reductase (SQR) of Complex II occupies a unique central point in the mitochondrial respiratory system as a major source of electrons driving reactive oxygen species (ROS) production. It is an ideal pharmaceutical target for modulating ROS levels in normal cells to prevent oxidative stress-induced damage or alternatively,increase ROS in cancer cells, inducing cell death.The value of drugs like diazoxide to prevent ROS production,protecting normal cells, whereas vitamin E analogues promote ROS in cancer cells to kill them is highlighted. As pharmaceuticals these agents may prevent degenerative disease and their modes of action are presently being fully explored. The evidence that SDH/Complex II is tightly coupled to the NADH/NAD+ ratio in all cells,impacted by the available supplies of Krebs cycle intermediates as essential NAD-linked substrates, and the NAD+-dependent regulation of SDH/Complex II are reviewed, as are links to the NAD+-dependent dehydrogenases, Complex I and the E3 dihiydrolipoamide dehydrogenase to produce ROS. This review collates and discusses diverse sources of information relating to ROS production in different biological systems, focussing on evidence for SQR as the main source of ROS production in mitochondria, particularly its relevance to protection from oxidative stress and to the mitochondrial-targeted anti cancer drugs (mitocans) as novel cancer therapies [corrected].
Collapse
|
27
|
Kettawan A, Kunthida C, Takahashi T, Kishi T, Chikazawa J, Sakata Y, Yano E, Watabe K, Yamamoto Y, Okamoto T. The quality control assessment of commercially available coenzyme q(10)-containing dietary and health supplements in Japan. J Clin Biochem Nutr 2011; 41:124-31. [PMID: 18193106 PMCID: PMC2170950 DOI: 10.3164/jcbn.2007017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2006] [Accepted: 01/19/2007] [Indexed: 12/05/2022] Open
Abstract
Coenzyme Q10 (CoQ10) has been widely commercially available in Japan as a dietary and health supplement since 2001 and is used for the prevention of lifestyle-related diseases induced by free radicals and aging. We evaluated CoQ10 supplements to ensure that these supplements can be used effectively and safely. Commercially available products were selected and assessed by the quality control tests specified in the Japanese Pharmacopoeia XV. When the disintegration time of CoQ10 supplements was measured, a few tested supplements did not completely disintegrate even after incubation in water for an hour at 37°C. In the content test, many samples were well controlled. However, a few supplements showed low recovery rates of CoQ10 as compared to manufacturer’s indicated contents. Among soft capsule and liquid supplements, the reduced form of CoQ10 (H2CoQ10), as well as the oxidized form, was detected by HPLC with electrochemical detector. The results for experimental formulated CoQ10 supplements demonstrated that H2CoQ10 was produced by the interaction of CoQ10 with vitamins E and/or C. From these results, we concluded that quality varied considerably among the many supplement brands containing CoQ10. Additionally, we also demonstrated that H2CoQ10 can be detected in some foods as well as in CoQ10 supplements.
Collapse
Affiliation(s)
- Aikkarach Kettawan
- Laboratory of Biochemistry, Department of Health Sciences and Social Pharmacy, Faculty of Pharmaceutical Sciences, Kobe Gakuin University, Kobe 651-2180, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Kettawan A, Takahashi T, Kongkachuichai R, Charoenkiatkul S, Kishi T, Okamoto T. Protective effects of coenzyme q(10) on decreased oxidative stress resistance induced by simvastatin. J Clin Biochem Nutr 2011; 40:194-202. [PMID: 18398496 PMCID: PMC2275764 DOI: 10.3164/jcbn.40.194] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2006] [Accepted: 10/25/2006] [Indexed: 11/29/2022] Open
Abstract
The effects of simvastatin, an inhibitor of 3-hydroxy-3-methylglutaryl CoA reductase (HMG-CoA reductase), on oxidative stress resistance and the protective effects of coenzyme Q (CoQ) were investigated. When simvastatin was administered orally to mice, the levels of oxidized and reduced CoQ9 and CoQ10 in serum, liver, and heart, decreased significantly when compared to those of control. The levels of thiobarbituric acid reactive substances induced by Fe2+-ascorbate in liver and heart mitochondria also increased significantly with simvastatin. Furthermore, cultured cardiac myocytes treated with simvastatin exhibited less resistance to oxidative stress, decreased time to the cessation of spontaneous beating in response to H2O2 addition, and decreased responsiveness to electrical field stimulation. These results suggested that oral administration of simvastatin suppresses the biosynthesis of CoQ, which shares the same biosynthesis pathway as cholesterol up to farnesyl pyrophosphate, thus compromising the physiological function of reduced CoQ, which possesses antioxidant activity. However, these undesirable effects induced by simvastatin were alleviated by coadministering CoQ10 with simvastatin to mice. Simvastatin also reduced the activity of NADPH-CoQ reductase, a biological enzyme that converts oxidized CoQ to the corresponding reduced CoQ, while CoQ10 administration improved it. These findings may also support the efficacy of coadministering CoQ10 with statins.
Collapse
Affiliation(s)
- Aikkarach Kettawan
- Laboratory of Biochemistry, Division of Health Sciences and Social Pharmacy, Faculty of Pharmaceutical Sciences, Kobe Gakuin University, Kobe 650-8586, Japan
| | | | | | | | | | | |
Collapse
|
29
|
Abstract
Lipoic acid [(R)-5-(1,2-dithiolan-3-yl)pentanoic acid] is an enzyme cofactor required for intermediate metabolism in free-living cells. Lipoic acid was discovered nearly 60 years ago and was shown to be covalently attached to proteins in several multicomponent dehydrogenases. Cells can acquire lipoate (the deprotonated charge form of lipoic acid that dominates at physiological pH) through either scavenging or de novo synthesis. Microbial pathogens implement these basic lipoylation strategies with a surprising variety of adaptations which can affect pathogenesis and virulence. Similarly, lipoylated proteins are responsible for effects beyond their classical roles in catalysis. These include roles in oxidative defense, bacterial sporulation, and gene expression. This review surveys the role of lipoate metabolism in bacterial, fungal, and protozoan pathogens and how these organisms have employed this metabolism to adapt to niche environments.
Collapse
|
30
|
Gibson GE, Starkov A, Blass JP, Ratan RR, Beal MF. Cause and consequence: mitochondrial dysfunction initiates and propagates neuronal dysfunction, neuronal death and behavioral abnormalities in age-associated neurodegenerative diseases. Biochim Biophys Acta Mol Basis Dis 2009; 1802:122-34. [PMID: 19715758 DOI: 10.1016/j.bbadis.2009.08.010] [Citation(s) in RCA: 187] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2009] [Revised: 08/14/2009] [Accepted: 08/17/2009] [Indexed: 12/31/2022]
Abstract
Age-related neurodegenerative diseases are associated with mild impairment of oxidative metabolism and accumulation of abnormal proteins. Within the cell, the mitochondria appears to be a dominant site for initiation and propagation of disease processes. Shifts in metabolism in response to mild metabolic perturbations may decrease the threshold for irreversible injury in response to ordinarily sublethal metabolic insults. Mild impairment of metabolism accrue from and lead to increased reactive oxygen species (ROS). Increased ROS change cell signaling via post-transcriptional and transcriptional changes. The cause and consequences of mild impairment of mitochondrial metabolism is one focus of this review. Many experiments in tissues from humans support the notion that oxidative modification of the alpha-ketoglutarate dehydrogenase complex (KGDHC) compromises neuronal energy metabolism and enhances ROS production in Alzheimer's Disease (AD). These data suggest that cognitive decline in AD derives from the selective tricarboxylic acid (TCA) cycle abnormalities. By contrast in Huntington's Disease (HD), a movement disorder with cognitive features distinct form AD, complex II+III abnormalities may dominate. These distinct mitochondrial abnormalities culminate in oxidative stress, energy dysfunction, and aberrant homeostasis of cytosolic calcium. Cytosolic calcium, elevations even only transiently, leads to hyperactivity of a number of enzymes. One calcium-activated enzyme with demonstrated pathophysiological import in HD and AD is transglutaminase (TGase). TGase is a crosslinking enzymes that can modulate transcription, inactivate metabolic enzymes, and cause aggregation of critical proteins. Recent data indicate that TGase can silence expression of genes involved in compensating for metabolic stress. Altogether, our results suggest that increasing KGDHC via inhibition of TGase or via a host of other strategies to be described would be effective therapeutic approaches in age-associated neurodegenerative diseases.
Collapse
Affiliation(s)
- Gary E Gibson
- Department of Neurology and Neuroscience, Weill Cornell Medical College of Cornell University at Burke Medical Research Institute, 785 Mamaroneck Avenue, White Plains, NY 10605, USA.
| | | | | | | | | |
Collapse
|
31
|
Yan LJ, Thangthaeng N, Forster MJ. Changes in dihydrolipoamide dehydrogenase expression and activity during postnatal development and aging in the rat brain. Mech Ageing Dev 2008; 129:282-90. [PMID: 18316113 PMCID: PMC2441877 DOI: 10.1016/j.mad.2008.01.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2007] [Revised: 12/19/2007] [Accepted: 01/23/2008] [Indexed: 02/06/2023]
Abstract
Brain energy metabolism is increased during postnatal development and diminished in neurodegenerative diseases linked to senescence. The objective of this study was to determine if these conditions could involve postnatal or senescence-related shifts in activity or expression of dihydrolipoamide dehydrogenase (DLDH), a key mitochondrial oxidoreductase. Rats ranging from 10 to 60 days of age were used in studies of postnatal development, whereas rats aged 5 or 30 months were used in the aging studies. The expression of DLDH was determined by Western blot analysis using anti-DLDH antibodies and DLDH diaphorase activity was measured by an in-gel activity staining method using nitroblue tetrazolium (NBT)/NADH. Activity of DLDH dehydrogenase was measured as NAD+ oxidation of dihydrolipoamide. When these measures were considered in separate groups of 10-, 20-, 30-, or 60-day-old rats, all three showed an increase between 10 and 20 days of age. However, dehydrogenase activity of DLDH showed a further, progressive increase from 20 days to adulthood, in the absence of any further change in DLDH expression or diaphorase activity. No age-related decline in DLDH activity or expression was evident over the period from 5 to 30 months of age. Moreover, aging did not render DLDH more susceptible to oxidative inactivation by mitochondria-generated reactive oxygen species (ROS). Taken together, results of the present study indicate that (1) brain DLDH expression and activity undergo independent postnatal maturational increases; (2) senescence does not confer any detectable change in the activity of DLDH or its susceptibility to inactivation by mitochondrial oxidative stress.
Collapse
Affiliation(s)
- Liang-Jun Yan
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA. <>
| | | | | |
Collapse
|
32
|
Batista AP, Kletzin A, Pereira MM. The dihydrolipoamide dehydrogenase from the crenarchaeon Acidianus ambivalens. FEMS Microbiol Lett 2008; 281:147-54. [DOI: 10.1111/j.1574-6968.2008.01082.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
33
|
Takahashi T, Okuno M, Okamoto T, Kishi T. NADPH-dependent coenzyme Q reductase is the main enzyme responsible for the reduction of non-mitochondrial CoQ in cells. Biofactors 2008; 32:59-70. [PMID: 19096101 DOI: 10.1002/biof.5520320108] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
We purified an NADPH-dependent coenzyme Q reductase (NADPH-CoQ reductase) in rat liver cytosol and compared its enzymatic properties with those of the other CoQ10 reductases such as NADPH: quinone acceptor oxidoreductase 1 (NQO1), lipoamide dehydrogenase, thioredoxine reductase and glutathione reductase. NADPH-CoQ reductase was the only enzyme that preferred NADPH to NADH as an electron donor and was also different from the other CoQ10 reductases in the sensitivities to its inhibitors and stimulators. Especially, Zn2+ was the most powerful inhibitor for NADPH-CoQ reductase, but CoQ10 reduction by the other CoQ10 reductases could not be inhibited by Zn2+. Furthermore, the reduction of the CoQ9 incorporated into HeLa cells was also inhibited by Zn2+ in the presence of pyrithione, a zinc ionophore. Moreover, NQO1 gene silencing in HeLa cells by transfection of a small interfering RNA resulted in lowering of both the NQO1 protein level and the NQO1 activity by about 75%. However, this transfection did not affect the NADPH-CoQ reductase activity and the reduction of CoQ9 incorporated into the cells. These results suggest that the NADPH-CoQ reductase located in cytosol may be the main enzyme responsible for the reduction of non-mitochondrial CoQ in cells.
Collapse
Affiliation(s)
- Takayuki Takahashi
- Laboratory of Biochemistry, Department of Health Sciences and Social Pharmacy, Faculty of Pharmaceutical Sciences, Kobe Gakuin University, Kobe, Japan.
| | | | | | | |
Collapse
|
34
|
Yan LJ, Yang SH, Shu H, Prokai L, Forster MJ. Histochemical staining and quantification of dihydrolipoamide dehydrogenase diaphorase activity using blue native PAGE. Electrophoresis 2007; 28:1036-45. [PMID: 17315258 DOI: 10.1002/elps.200600574] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Mammalian mitochondrial dihydrolipoamide dehydrogenase (DLDH, EC 1.8.1.4) catalyzes NAD(+)-dependent oxidation of dihydrolipoamide in vivo and can also act as a diaphorase catalyzing in vitro nicotinamide adenine dinucleotide (reduced form) (NADH)-dependent reduction of electron-accepting molecules such as ubiquinone and nitroblue tetrazolium (NBT). In this paper, we report a gel-based method for histochemical staining and quantification of DLDH diaphorase activity using blue native PAGE (BN-PAGE). Rat brain mitochondrial extracts, used as the source of DLDH, were resolved by nongradient BN-PAGE (9%), which was followed by diaphorase activity staining using NADH as the electron donor and NBT as the electron acceptor. It was shown that activity staining of DLDH diaphorase was both protein amount- and time-dependent. Moreover, this in-gel activity-staining method was demonstrated to be in good agreement with the conventional spectrophotometric method that measures DLDH dehydrogenase activity using dihydrolipoamide as the substrate. The method was applied to determine levels of DLDH diaphorase activity in several rat tissues other than the brain, and the results indicated a similar level of DLDH diaphorase activity for all the tissues examined. Finally, the effects of thiol-reactive reagents such as N-ethylmaleimide (NEM) and nitric oxide donors on DLDH diaphorase activity were evaluated, demonstrating that, with this method, DLDH diaphorase activity can be determined without having to remove these thiol-reactive reagents that may otherwise interfere with spectrophotometric measurement of DLDH dehydrogenase activity. The gel-based method can also be used as a means to isolate mitochondrial DLDH that is to be analyzed by mass spectral techniques in studying DLDH post-translational modifications.
Collapse
Affiliation(s)
- Liang-Jun Yan
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, TX 76107, USA.
| | | | | | | | | |
Collapse
|
35
|
Cabreiro F, Picot CR, Perichon M, Mary J, Friguet B, Petropoulos I. Identification of proteins undergoing expression level modifications in WI-38 SV40 fibroblasts overexpressing methionine sulfoxide reductase A. Biochimie 2007; 89:1388-95. [PMID: 17624653 DOI: 10.1016/j.biochi.2007.05.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2007] [Accepted: 05/18/2007] [Indexed: 01/15/2023]
Abstract
Methionine sulfoxide reductase A overexpressing WI-38 SV40 human fibroblasts have been previously shown to exhibit higher resistance to oxidative stress by decreasing intracellular reactive oxygen species content and oxidative damage to proteins [C.R. Picot, I. Petropoulos, M. Perichon, M. Moreau, C. Nizard, B. Friguet, Overexpression of MsrA protects WI-38 SV40 human fibroblasts against H(2)O(2)-mediated oxidative stress, Free Radic Biol Med 39 (2005) 1332-1341]. In order to get further insight into the molecular mechanisms underlying this resistance to oxidative stress, proteins that are differentially expressed in methionine sulfoxide reductase A overexpressing cells were identified by 2D gel and Western blot quantitative analyses. Five proteins were shown to be differentially expressed and were identified by mass spectrometry, some of them were related to either cellular protection against oxidative stress, apoptosis or premature ageing.
Collapse
Affiliation(s)
- F Cabreiro
- Laboratoire de Biologie et Biochimie Cellulaire du Vieillissement, EA 3106/IFR 117, Université Paris Diderot-Paris 7, 2 place Jussieu, Tour 33-23, 1(er) étage, CC 7128, 75251, Paris Cedex 05, France
| | | | | | | | | | | |
Collapse
|
36
|
Babady NE, Pang YP, Elpeleg O, Isaya G. Cryptic proteolytic activity of dihydrolipoamide dehydrogenase. Proc Natl Acad Sci U S A 2007; 104:6158-63. [PMID: 17404228 PMCID: PMC1851069 DOI: 10.1073/pnas.0610618104] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The mitochondrial enzyme, dihydrolipoamide dehydrogenase (DLD), is essential for energy metabolism across eukaryotes. Here, conditions known to destabilize the DLD homodimer enabled the mouse, pig, or human enzyme to function as a protease. A catalytic dyad (S456-E431) buried at the homodimer interface was identified. Serine protease inhibitors and an S456A or an E431A point mutation abolished the proteolytic activity, whereas other point mutations at the homodimer interface domain enhanced the proteolytic activity, causing partial or complete loss of DLD activity. In humans, mutations in the DLD homodimer interface have been linked to an atypical form of DLD deficiency. These findings reveal a previously unrecognized mechanism by which certain DLD mutations can simultaneously induce the loss of a primary metabolic activity and the gain of a moonlighting proteolytic activity. The latter could contribute to the metabolic derangement associated with DLD deficiency and represent a target for therapies of this condition.
Collapse
Affiliation(s)
- Ngolela Esther Babady
- Departments of *Pediatric and Adolescent Medicine and
- Biochemistry and Molecular Biology and
| | - Yuan-Ping Pang
- Computer-Aided Molecular Design Laboratory, Mayo Clinic College of Medicine, Rochester, MN 55905; and
| | - Orly Elpeleg
- Metabolic Disease Unit, Hadassah Hebrew University Medical Center, Jerusalem 91120, Israel
| | - Grazia Isaya
- Departments of *Pediatric and Adolescent Medicine and
- Biochemistry and Molecular Biology and
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
37
|
Reisch AS, Elpeleg O. Biochemical assays for mitochondrial activity: assays of TCA cycle enzymes and PDHc. Methods Cell Biol 2007; 80:199-222. [PMID: 17445696 DOI: 10.1016/s0091-679x(06)80010-5] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Ann Saada Reisch
- The Metabolic Disease Unit, Hadassah-Hebrew University Medical Centre, Jerusalem 91120, Israel
| | | |
Collapse
|
38
|
Ingle RA, Smith JAC, Sweetlove LJ. Responses to Nickel in the Proteome of the Hyperaccumulator Plant Alyssum lesbiacum. Biometals 2005; 18:627-41. [PMID: 16388402 DOI: 10.1007/s10534-005-2999-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2005] [Accepted: 09/12/2005] [Indexed: 01/30/2023]
Abstract
A proteomic analysis of the Ni hyperaccumulator plant Alyssum lesbiacum was carried out to identify proteins that may play a role in the exceptional degree of Ni tolerance and accumulation characteristic of this metallophyte. Of the 816 polypeptides detected in root tissue by 2D SDS-PAGE, eleven increased and one decreased in abundance relative to total protein after 6-week-old plants were transferred from a standard nutrient solution containing trace concentrations of Ni to a moderately high Ni treatment (0.3 mM NiSO4) for 48 h. These polypeptides were identified by tandem mass spectrometry and the majority were found to be involved in sulphur metabolism (consistent with a re-allocation of sulphur towards cysteine and glutathione), protection against reactive oxygen species, or heat-shock response. In contrast, very few polypeptides were found to change in abundance in root or shoot tissue after plants were exposed for 28 days to 0.03 mM NiSO4, a concentration representing the optimum for growth of this species but sufficient to lead to hyperaccumulation of Ni in the shoot. Under these conditions, constitutively expressed genes in this highly Ni-tolerant species may be sufficient to allow for effective chelation and sequestration of Ni without the need for additional protein synthesis.
Collapse
Affiliation(s)
- Robert A Ingle
- Department of Plant Sciences, University of Oxford, South Parks Road, Oxford, OX1 3RB, United Kingdom.
| | | | | |
Collapse
|
39
|
Whitworth KM, Agca C, Kim JG, Patel RV, Springer GK, Bivens NJ, Forrester LJ, Mathialagan N, Green JA, Prather RS. Transcriptional Profiling of Pig Embryogenesis by Using a 15-K Member Unigene Set Specific for Pig Reproductive Tissues and Embryos1. Biol Reprod 2005; 72:1437-51. [PMID: 15703372 DOI: 10.1095/biolreprod.104.037952] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Differential mRNA expression patterns were evaluated between germinal vesicle oocytes (pgvo), four-cell (p4civv), blastocyst (pblivv), and in vitro-produced four-cell (p4civp) and in vitro-produced blastocyst (pblivp) stage embryos to determine key transcripts responsible for early embryonic development in the pig. Five comparisons were made: pgvo to p4civv, p4civv to pblivv, pgvo to pblivv, p4civv to p4civp, and pblivv to pblivp. ANOVA (P < 0.05) was performed with the Benjamini and Hochberg false-discovery-rate multiple correction test on each comparison. A comparison of pgvo to p4civv, p4civv to pblivv, and pgvo to pblivv resulted in 3214, 1989, and 4528 differentially detected cDNAs, respectively. Real-time PCR analysis on seven transcripts showed an identical pattern of changes in expression as observed on the microarrays, while one transcript deviated at a single cell stage. There were 1409 and 1696 differentially detected cDNAs between the in vitro- and in vivo-produced embryos at the four-cell and blastocyst stages, respectively, without the Benjamini and Hochberg false-discovery-rate multiple correction test. Real-time polymerase chain reaction (PCR) analysis on four genes at the four-cell stage showed an identical pattern of gene expression as found on the microarrays. Real-time PCR analysis on four of five genes at the blastocyst stage showed an identical pattern of gene expression as found on the microarrays. Thus, only 1 of the 39 comparisons of the pattern of gene expression exhibited a major deviation between the microarray and the real-time PCR. These results illustrate the complex mechanisms involved in pig early embryonic development.
Collapse
Affiliation(s)
- K M Whitworth
- Department of Animal Science, University of Missouri-Columbia, Missouri 65211, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Center SA. Metabolic, antioxidant, nutraceutical, probiotic, and herbal therapies relating to the management of hepatobiliary disorders. Vet Clin North Am Small Anim Pract 2004; 34:67-172, vi. [PMID: 15032126 DOI: 10.1016/j.cvsm.2003.09.015] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Many nutraceuticals, conditionally essential nutrients, and botanical extracts have been proposed as useful in the management of liver disease. The most studied of these are addressed in terms of proposed mechanisms of action, benefits, hazards, and safe dosing recommendations allowed by current information. While this is an area of soft science, it is important to keep an open and tolerant mind, considering that many major treatment discoveries were in fact serendipitous accidents.
Collapse
Affiliation(s)
- Sharon A Center
- College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
41
|
Bae W, Chen X. Proteomic Study for the Cellular Responses to Cd2+ in Schizosaccharomyces pombe Through Amino Acid-coded Mass Tagging and Liquid Chromatography Tandem Mass Spectrometry. Mol Cell Proteomics 2004; 3:596-607. [PMID: 15004206 DOI: 10.1074/mcp.m300122-mcp200] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cadmium (Cd(2+)) is one of well-known toxic heavy metal ions. To gain a global understanding how Cd(2+) affects cells at the molecular level, we systematically studied the cellular response of the fission yeast Schizosaccharomyces pombe to Cd(2+) using our integrated proteomic strategy of amino acid-coded mass tagging (AACT) and liquid chromatography-tandem mass spectrometry. Our proteome-wide investigation unequivocally identified 1133 S. pombe proteins. Of which, the AACT-based quantitative analysis revealed 106 up-regulated and 55 down-regulated proteins on the Cd(2+) exposure. The most prevalent functional class in the up-regulated proteins, approximately 28% of our profile, was the proteins involved in protein biosynthesis, showing a time-dependent biphasic expression pattern characteristic with rapid initial induction and later repression. Most significantly, 27 proteins functionally classified as cell rescue and defense were up-regulated for oxygen and radical detoxification, heat shock response, and other stress response. Furthermore, the large precursor sequence coverage of our AACT approach allowed us to unequivocally identify and quantitate different isozymes for glutathione S-transferase, which have close similarity in their amino acid sequence. Our quantitative dataset also showed that 80% of the up-regulated proteins found in the S. pombe response were different from those in the Saccharomyces cerevisiae response. The function of some of the key identifications was validated through biochemical assays. It is very interesting that the induction of cysteine synthase expression was not observed in our study, although it has been proven as a critical enzyme to supply free cysteines for the enhancing synthesis of Cd(2+)-sequestering molecules such as glutathione and phytochelatins in plants and some yeasts. Our quantitative proteomic result instead suggested that, as an alternative mechanism for the detoxification of Cd(2+), S. pombe produced significantly higher level of inorganic sulfide to immobilize cellular Cd(2+) as a form of CdS nanocrystallites capped with glutathione and/or phytochelatins.
Collapse
Affiliation(s)
- Weon Bae
- B-2, MS M888, Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM 87545, USA
| | | |
Collapse
|
42
|
Björnstedt M, Nordman T, Olsson JM. Extramitochondrial reduction of ubiquinone by flavoenzymes. Methods Enzymol 2004; 378:131-8. [PMID: 15038962 DOI: 10.1016/s0076-6879(04)78008-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Affiliation(s)
- Mikael Björnstedt
- Department of Laboratory Medicine, Karolinska Institutet, Hudinge University Hospital, Stockholm, Sweden
| | | | | |
Collapse
|
43
|
Affiliation(s)
- M Flint Beal
- Department of Neurology and Neuroscience, Weill Medical College of Cornell University, New York, New York 10021, USA
| |
Collapse
|
44
|
Turunen M, Olsson J, Dallner G. Metabolism and function of coenzyme Q. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2004; 1660:171-99. [PMID: 14757233 DOI: 10.1016/j.bbamem.2003.11.012] [Citation(s) in RCA: 733] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Coenzyme Q (CoQ) is present in all cells and membranes and in addition to be a member of the mitochondrial respiratory chain it has also several other functions of great importance for the cellular metabolism. This review summarizes the findings available to day concerning CoQ distribution, biosynthesis, regulatory modifications and its participation in cellular metabolism. There are a number of indications that this lipid is not always functioning by its direct presence at the site of action but also using e.g. receptor expression modifications, signal transduction mechanisms and action through its metabolites. The biosynthesis of CoQ is studied in great detail in bacteria and yeast but only to a limited extent in animal tissues and therefore the informations available is restricted. However, it is known that the CoQ is compartmentalized in the cell with multiple sites of biosynthesis, breakdown and regulation which is the basis of functional specialization. Some regulatory mechanisms concerning amount and biosynthesis are established and nuclear transcription factors are partly identified in this process. Using appropriate ligands of nuclear receptors the biosynthetic rate can be increased in experimental system which raises the possibility of drug-induced upregulation of the lipid in deficiency. During aging and pathophysiological conditions the tissue concentration of CoQ is modified which influences cellular functions. In this case the extent of disturbances is dependent on the localization and the modified distribution of the lipid at cellular and membrane levels.
Collapse
Affiliation(s)
- Mikael Turunen
- Department of Biochemistry and Biophysics, Stockholm University, Arrhenius Laboratories for Natural Sciences, SE-106 91 Stockholm, Sweden.
| | | | | |
Collapse
|
45
|
Argyrou A, Blanchard JS. Flavoprotein Disulfide Reductases: Advances in Chemistry and Function. PROGRESS IN NUCLEIC ACID RESEARCH AND MOLECULAR BIOLOGY 2004; 78:89-142. [PMID: 15210329 DOI: 10.1016/s0079-6603(04)78003-4] [Citation(s) in RCA: 150] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The flavoprotein disulfide reductases represent a family of enzymes that show high sequence and structural homology. They catalyze the pyridine-nucleotide-dependent reduction of a variety of substrates, including disulfide-bonded substrates (lipoamide dehydrogenase, glutathione reductase and functional homologues, thioredoxin reductase, and alkylhydroperoxide reductase), mercuric ion (mercuric ion reductase), hydrogen peroxide (NADH peroxidase), molecular oxygen (NADH oxidase), and the reductive cleavage of a carbonyl-activated carbon-sulfur bond followed by carboxylation (2-ketopropyl-coenzyme-M carboxylase?oxidoreductase). They use at least one nonflavin redox center to transfer electrons from reduced pyridine nucleotide to their substrate through flavin adenine dinucleotide. The nature of the nonflavin redox center located adjacent to the flavin varies and three types have been identified: an enzymic disulfide (most commonly), an enzymic cysteine sulfenic acid (NADH peroxidase and NADH oxidase), and a mixed Cys-S-S-CoA disulfide (coenzyme A disulfide reductase). Selection of the particular nonflavin redox center and utilization of a second, or even a third, nonflavin redox center in some cases presumably represents the most efficient strategy for reduction of the individual substrate.
Collapse
Affiliation(s)
- Argyrides Argyrou
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | |
Collapse
|
46
|
Yamashoji S. Coenzyme Q1-catalyzed luminol chemiluminescent assay for rapid antimicrobial susceptibility testing of Mycobacterium bovis. Microbiol Immunol 2003; 47:191-8. [PMID: 12725288 DOI: 10.1111/j.1348-0421.2003.tb03386.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Coenzyme Q1 is herein proposed as the best catalyst among coenzymes Q and vitamins K for quinone-catalyzed luminol chemiluminescent assays applied to rapid determination of viability or rapid antimicrobial susceptibility tests of Mycobacterium bovis. Luminol chemiluminescence intensity (LCI) was determined 10 min after the incubation of M. bovis with coenzyme Q1, and was proportional to CFU (colony-forming unit)/ml in the range of 9,000 to 2,250,000. LCI depended on the the production of the superoxide anion (O2-) rather than H2O2 during a 10-min incubation of M. bovis with coenzyme Q1, as superoxide dismutase reduced LCI more effectively than catalase. The minimal inhibitory concentrations (MICs) of 10 kinds of antituberculous agents estimated on the basis of decrease in LCI after one or two days' cultivation were in good agreement with MICs determined by turbidity analysis, which requires upwards of 1 week to complete.
Collapse
Affiliation(s)
- Shiro Yamashoji
- Nikken Biomedical Laboratory, 23 Teigaien, Ohashibe, Kumiyama-cho, Kuze-gun, Kyoto 613-0046, Japan.
| |
Collapse
|
47
|
Xia L, Nordman T, Olsson JM, Damdimopoulos A, Björkhem-Bergman L, Nalvarte I, Eriksson LC, Arnér ESJ, Spyrou G, Björnstedt M. The mammalian cytosolic selenoenzyme thioredoxin reductase reduces ubiquinone. A novel mechanism for defense against oxidative stress. J Biol Chem 2003; 278:2141-6. [PMID: 12435734 DOI: 10.1074/jbc.m210456200] [Citation(s) in RCA: 160] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The selenoprotein thioredoxin reductase (TrxR1) is an essential antioxidant enzyme known to reduce many compounds in addition to thioredoxin, its principle protein substrate. Here we found that TrxR1 reduced ubiquinone-10 and thereby regenerated the antioxidant ubiquinol-10 (Q10), which is important for protection against lipid and protein peroxidation. The reduction was time- and dose-dependent, with an apparent K(m) of 22 microm and a maximal rate of about 12 nmol of reduced Q10 per milligram of TrxR1 per minute. TrxR1 reduced ubiquinone maximally at a physiological pH of 7.5 at similar rates using either NADPH or NADH as cofactors. The reduction of Q10 by mammalian TrxR1 was selenium dependent as revealed by comparison with Escherichia coli TrxR or selenium-deprived mutant and truncated mammalian TrxR forms. In addition, the rate of reduction of ubiquinone was significantly higher in homogenates from human embryo kidney 293 cells stably overexpressing thioredoxin reductase and was induced along with increasing cytosolic TrxR activity after the addition of selenite to the culture medium. These data demonstrate that the selenoenzyme thioredoxin reductase is an important selenium-dependent ubiquinone reductase and can explain how selenium and ubiquinone, by a combined action, may protect the cell from oxidative damage.
Collapse
Affiliation(s)
- Ling Xia
- Department of Microbiology, Pathology, and Immunology, Division of Pathology, F46, Karolinska Institutet, Huddinge University Hospital, SE-141 86 Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Nordman T, Xia L, Björkhem-Bergman L, Damdimopoulos A, Nalvarte I, Arnér ESJ, Spyrou G, Eriksson LC, Björnstedt M, Olsson JM. Regeneration of the antioxidant ubiquinol by lipoamide dehydrogenase, thioredoxin reductase and glutathione reductase. Biofactors 2003; 18:45-50. [PMID: 14695919 DOI: 10.1002/biof.5520180206] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Ubiquinol is a powerful antioxidant, which is oxidized in action and needs to be replaced or regenerated to be capable of a sustained effort. This article summarises current knowledge of extramitochondrial reduction of ubiquinone by three flavoenzymes, i.e. lipoamide dehydrogenase, glutathione reductase and thioredoxin reductase, belonging to the same pyridine nucleotide-disulfide oxidoreductase family. These three enzymes are the most efficient extramitochondrial ubiquinone reductases so far described. The reduction of ubiquinone by lipoamide dehydrogenase and glutathione reductase is potently stimulated by zinc and the highest rate of reduction is achieved at acidic pH and the rates are equal with either NADPH or NADH as co-factors. The most efficient ubiquinone reductases are mammalian cytosolic thioredoxin reductases, which are selenoenzymes with a number of biological functions. Reduction of ubiquinone by thioredoxin reductase is in contrast to the other two enzymes investigated, inhibited by zinc and shows a sharp physiological pH optimum at pH 7.5. Furthermore, the reaction is selenium dependent as revealed from experiments using truncated and mutant forms of the enzyme and also in a cellular context by selenium treatment of transfected thioredoxin reductase overexpressing stable cell lines. The reduction of ubiquinone by the three enzymes offers a multifunctional system for extramitochondrial regeneration of an important antioxidant.
Collapse
Affiliation(s)
- Tomas Nordman
- Department of Laboratory Medicine, F 46, Karolinska Institutet, Huddinge University Hospital, SE-141 86 Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Affiliation(s)
- M Flint Beal
- Department of Neurology and Neuroscience, New York Presbyterian Hospital, New York, NY 10021, USA.
| | | |
Collapse
|
50
|
Jonassen T, Marbois BN, Faull KF, Clarke CF, Larsen PL. Development and fertility in Caenorhabditis elegans clk-1 mutants depend upon transport of dietary coenzyme Q8 to mitochondria. J Biol Chem 2002; 277:45020-7. [PMID: 12324451 DOI: 10.1074/jbc.m204758200] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The Caenorhabditis elegans clk-1 mutants lack coenzyme Q(9) and instead accumulate the biosynthetic intermediate demethoxy-Q(9) (DMQ(9)). clk-1 animals grow to reproductive adults, albeit slowly, if supplied with Q(8)-containing Escherichia coli. However, if Q is withdrawn from the diet, clk-1 animals either arrest development as young larvae or become sterile adults depending upon the stage at the time of the withdrawal. To understand this stage-dependent response to a Q-less diet, the quinone content was determined during development of wild-type animals. The quinone content varies in the different developmental stages in wild-type fed Q(8)-replete E. coli. The amounts peak at the second larval stage, which coincides with the stage of arrest of clk-1 larvae fed a Q-less diet from hatching. Levels of the endogenously synthesized DMQ(9) are high in the clk-1(qm30)-arrested larvae and sterile adults fed Q-less food. Comparison of quinones from animals fed a Q-replete or a Q-less diet establishes that the Q(8) present is assimilated from the E. coli. Furthermore, this E. coli-specific Q(8) is present in mitochondria isolated from fertile clk-1(qm30) adults fed a Q-replete diet. These results suggest that the uptake and transport of dietary Q(8) to mitochondria prevent the arrest and sterility phenotypes of clk-1 mutants and that DMQ is not functionally equivalent to Q.
Collapse
Affiliation(s)
- Tanya Jonassen
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095-1569, USA
| | | | | | | | | |
Collapse
|