1
|
Hansman DS, Du J, Casson RJ, Peet DJ. Eye on the horizon: The metabolic landscape of the RPE in aging and disease. Prog Retin Eye Res 2024; 104:101306. [PMID: 39433211 DOI: 10.1016/j.preteyeres.2024.101306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/10/2024] [Accepted: 10/10/2024] [Indexed: 10/23/2024]
Abstract
To meet the prodigious bioenergetic demands of the photoreceptors, glucose and other nutrients must traverse the retinal pigment epithelium (RPE), a polarised monolayer of cells that lie at the interface between the outer retina and the choroid, the principal vascular layer of the eye. Recent investigations have revealed a metabolic ecosystem in the outer retina where the photoreceptors and RPE engage in a complex exchange of sugars, amino acids, and other metabolites. Perturbation of this delicate metabolic balance has been identified in the aging retina, as well as in age-related macular degeneration (AMD), the leading cause of blindness in the Western world. Also common in the aging and diseased retina are elevated levels of cytokines, oxidative stress, advanced glycation end-products, increased growth factor signalling, and biomechanical stress - all of which have been associated with metabolic dysregulation in non-retinal cell types and tissues. Herein, we outline the role of these factors in retinal homeostasis, aging, and disease. We discuss their effects on glucose, mitochondrial, lipid, and amino acid metabolism in tissues and cell types outside the retina, highlighting the signalling pathways through which they induce these changes. Lastly, we discuss promising avenues for future research investigating the roles of these pathological conditions on retinal metabolism, potentially offering novel therapeutic approaches to combat age-related retinal disease.
Collapse
Affiliation(s)
- David S Hansman
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia.
| | - Jianhai Du
- Department of Ophthalmology and Visual Sciences, Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV 26506, USA
| | - Robert J Casson
- Discipline of Ophthalmology and Visual Science, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Daniel J Peet
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
2
|
Brogyanyi T, Kejík Z, Veselá K, Dytrych P, Hoskovec D, Masařik M, Babula P, Kaplánek R, Přibyl T, Zelenka J, Ruml T, Vokurka M, Martásek P, Jakubek M. Iron chelators as mitophagy agents: Potential and limitations. Biomed Pharmacother 2024; 179:117407. [PMID: 39265234 DOI: 10.1016/j.biopha.2024.117407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/26/2024] [Accepted: 09/02/2024] [Indexed: 09/14/2024] Open
Abstract
Mitochondrial autophagy (mitophagy) is very important process for the maintenance of cellular homeostasis, functionality and survival. Its dysregulation is associated with high risk and progression numerous serious diseases (e.g., oncological, neurodegenerative and cardiovascular ones). Therefore, targeting mitophagy mechanisms is very hot topic in the biological and medicinal research. The interrelationships between the regulation of mitophagy and iron homeostasis are now becoming apparent. In short, mitochondria are central point for the regulation of iron homeostasis, but change in intracellular cheatable iron level can induce/repress mitophagy. In this review, relationships between iron homeostasis and mitophagy are thoroughly discussed and described. Also, therapeutic applicability of mitophagy chelators in the context of individual diseases is comprehensively and critically evaluated.
Collapse
Affiliation(s)
- Tereza Brogyanyi
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic; Institute of Pathological Physiology, First Faculty of Medicine, Charles University in Prague, U Nemocnice 5, 1, Prague 28 53, Czech Republic
| | - Zdeněk Kejík
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic
| | - Kateřina Veselá
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic
| | - Petr Dytrych
- 1st Department of Surgery-Department of Abdominal, Thoracic Surgery and Traumatology, First Faculty of Medicine, Charles University and General University Hospital, U Nemocnice 2, Prague 121 08, Czech Republic
| | - David Hoskovec
- 1st Department of Surgery-Department of Abdominal, Thoracic Surgery and Traumatology, First Faculty of Medicine, Charles University and General University Hospital, U Nemocnice 2, Prague 121 08, Czech Republic
| | - Michal Masařik
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic; Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno CZ-625 00, Czech Republic; Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic
| | - Petr Babula
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno CZ-625 00, Czech Republic
| | - Robert Kaplánek
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic
| | - Tomáš Přibyl
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Prague, Prague 166 28, Czech Republic
| | - Jaroslav Zelenka
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Prague, Prague 166 28, Czech Republic
| | - Tomáš Ruml
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Prague, Prague 166 28, Czech Republic
| | - Martin Vokurka
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University in Prague, U Nemocnice 5, 1, Prague 28 53, Czech Republic
| | - Pavel Martásek
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic
| | - Milan Jakubek
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic.
| |
Collapse
|
3
|
Grayson C, Faerman B, Koufos O, Mailloux RJ. Fatty acid oxidation drives mitochondrial hydrogen peroxide production by α-ketoglutarate dehydrogenase. J Biol Chem 2024; 300:107159. [PMID: 38479602 PMCID: PMC10997840 DOI: 10.1016/j.jbc.2024.107159] [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: 12/27/2023] [Revised: 02/29/2024] [Accepted: 03/06/2024] [Indexed: 04/07/2024] Open
Abstract
In the present study, we examined the mitochondrial hydrogen peroxide (mH2O2) generating capacity of α-ketoglutarate dehydrogenase (KGDH) and compared it to components of the electron transport chain using liver mitochondria isolated from male and female C57BL6N mice. We show for the first time there are some sex dimorphisms in the production of mH2O2 by electron transport chain complexes I and III when mitochondria are fueled with different substrates. However, in our investigations into these sex effects, we made the unexpected and compelling discovery that 1) KGDH serves as a major mH2O2 supplier in male and female liver mitochondria and 2) KGDH can form mH2O2 when liver mitochondria are energized with fatty acids but only when malate is used to prime the Krebs cycle. Surprisingly, 2-keto-3-methylvaleric acid (KMV), a site-specific inhibitor for KGDH, nearly abolished mH2O2 generation in both male and female liver mitochondria oxidizing palmitoyl-carnitine. KMV inhibited mH2O2 production in liver mitochondria from male and female mice oxidizing myristoyl-, octanoyl-, or butyryl-carnitine as well. S1QEL 1.1 (S1) and S3QEL 2 (S3), compounds that inhibit reactive oxygen species generation by complexes I and III, respectively, without interfering with OxPhos and respiration, had a negligible effect on the rate of mH2O2 production when pyruvate or acyl-carnitines were used as fuels. However, inclusion of KMV in reaction mixtures containing S1 and/or S3 almost abolished mH2O2 generation. Together, our findings suggest KGDH is the main mH2O2 generator in liver mitochondria, even when fatty acids are used as fuel.
Collapse
Affiliation(s)
- Cathryn Grayson
- The School of Human Nutrition, Faculty of Agricultural and Environmental Sciences, McGill University, Quebec, Canada
| | - Ben Faerman
- The School of Human Nutrition, Faculty of Agricultural and Environmental Sciences, McGill University, Quebec, Canada
| | - Olivia Koufos
- The School of Human Nutrition, Faculty of Agricultural and Environmental Sciences, McGill University, Quebec, Canada
| | - Ryan J Mailloux
- The School of Human Nutrition, Faculty of Agricultural and Environmental Sciences, McGill University, Quebec, Canada.
| |
Collapse
|
4
|
Ding L, Liu Y, Wang L, Yang Y. Distinguishing the responsive mechanisms of fluorescent probes to hydrogen peroxide, proteins, and DNA/RNA. Phys Chem Chem Phys 2024; 26:7765-7771. [PMID: 38372974 DOI: 10.1039/d4cp00082j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
The responsive mechanisms of cationic quinolinium-vinyl-N,N-dimethylaniline boronate (QVD-B) derivative probes to hydrogen peroxide (H2O2), proteins and DNA/RNA are theoretically investigated in this study. The potential energy curves of QVD-B scanned on a dihedral angle (N+-C-CC) in the first singlet (S1) state exhibit large torsional energy barriers. Additionally, the energy of the lowest unoccupied molecular orbital (LUMO) of an acceptor moiety (-3.14 eV) is lower than that of a donor moiety (-1.13 eV) in QVD-B. This demonstrates that photoinduced electron transfer (PET) quenches the fluorescence of QVD-B, as opposed to the previous report of intramolecular single-bond rotation. After reacting with H2O2, the reaction product of quinoline-vinyl-N,N-dimethylaniline (QVD) turns off the PET pathway and turns on the fluorescence at 550 nm, which is consistent with the experimental results (580 nm). Among the possible configurations of QVD-B that forms with proteins and DNA, the calculated fluorescence values of corresponding twisted QVD-B-P (638 nm) and QVD-B-D (686 nm) are consistent with the experimental results (632 and 688 nm). The frontier molecular orbital and electron-hole analysis show that the charge transfer distance follows the order of QVD (1.88 Å) < QVD-B-P (4.49 Å) < QVD-B-D (6.39 Å), which induces the fluorescence red-shifts of QVD-B-P and QVD-B-D compared to that of QVD. The multi-detection mechanism of the fluorescent probe QVD-B is attributed to PET progress and different degrees of local charge transfer after photoexcitation.
Collapse
Affiliation(s)
- Lina Ding
- School of Chemistry and Chemical Engineering, School of Physics, Henan Normal University, Xinxiang 453007, P. R. China.
| | - Yang Liu
- School of Chemistry and Chemical Engineering, School of Physics, Henan Normal University, Xinxiang 453007, P. R. China.
| | - Liang Wang
- School of Chemistry and Chemical Engineering, School of Physics, Henan Normal University, Xinxiang 453007, P. R. China.
| | - Yonggang Yang
- School of Chemistry and Chemical Engineering, School of Physics, Henan Normal University, Xinxiang 453007, P. R. China.
| |
Collapse
|
5
|
Scasny A, Alibayov B, Khan F, Rao SJ, Murin L, Jop Vidal AG, Smith P, Li W, Edwards K, Warncke K, Vidal JE. Oxidation of hemoproteins by Streptococcus pneumoniae collapses the cell cytoskeleton and disrupts mitochondrial respiration leading to the cytotoxicity of human lung cells. Microbiol Spectr 2024; 12:e0291223. [PMID: 38084982 PMCID: PMC10783075 DOI: 10.1128/spectrum.02912-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 11/06/2023] [Indexed: 12/21/2023] Open
Abstract
IMPORTANCE Streptococcus pneumoniae (Spn) colonizes the lungs, killing millions every year. During its metabolism, Spn produces abundant amounts of hydrogen peroxide. When produced in the lung parenchyma, Spn-hydrogen peroxide (H2O2) causes the death of lung cells, and details of the mechanism are studied here. We found that Spn-H2O2 targets intracellular proteins, resulting in the contraction of the cell cytoskeleton and disruption of mitochondrial function, ultimately contributing to cell death. Intracellular proteins targeted by Spn-H2O2 included cytochrome c and, surprisingly, a protein of the cell cytoskeleton, beta-tubulin. To study the details of oxidative reactions, we used, as a surrogate model, the oxidation of another hemoprotein, hemoglobin. Using the surrogate model, we specifically identified a highly reactive radical whose creation was catalyzed by Spn-H2O2. In sum, we demonstrated that the oxidation of intracellular targets by Spn-H2O2 plays an important role in the cytotoxicity caused by Spn, thus providing new targets for interventions.
Collapse
Affiliation(s)
- Anna Scasny
- Department of Cell and Molecular Biology, School of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Center for Immunology and Microbial Research, School of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Babek Alibayov
- Department of Cell and Molecular Biology, School of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Center for Immunology and Microbial Research, School of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Faidad Khan
- Department of Cell and Molecular Biology, School of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Center for Immunology and Microbial Research, School of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Shambavi J. Rao
- Department of Otolaryngology-Head and Neck Surgery, The Ohio State School of Medicine, The Ohio State Wexner Medical Center, Columbus, Ohio, USA
| | - Landon Murin
- Department of Cell and Molecular Biology, School of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Center for Immunology and Microbial Research, School of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Ana G. Jop Vidal
- Department of Cell and Molecular Biology, School of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Center for Immunology and Microbial Research, School of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Perriann Smith
- Mississippi INBRE Research Scholar, University of Southern Mississippi, Jackson, Mississippi, USA
| | - Wei Li
- Department of Physics, Emory University, Atlanta, Georgia, USA
| | - Kristin Edwards
- Department of Cell and Molecular Biology, School of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Kurt Warncke
- Department of Physics, Emory University, Atlanta, Georgia, USA
| | - Jorge E. Vidal
- Department of Cell and Molecular Biology, School of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Center for Immunology and Microbial Research, School of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
6
|
Flensted-Jensen M, Kleis-Olsen AS, Hassø RK, Lindtofte S, Corral Pérez J, Ortega-Gómez S, Larsen S. Combined changes in temperature and pH mimicking exercise result in decreased efficiency in muscle mitochondria. J Appl Physiol (1985) 2024; 136:79-88. [PMID: 37969081 DOI: 10.1152/japplphysiol.00293.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 11/07/2023] [Accepted: 11/10/2023] [Indexed: 11/17/2023] Open
Abstract
It is well known that exercise efficiency declines at intensities above the lactate threshold, yet the underlying mechanisms are poorly understood. Some have suggested it is due to a decline in mitochondrial efficiency, but this is difficult to examine in vivo. Therefore, the aim of the current study was to examine how changes in temperature and pH, mimicking those that occur during exercise, affect mitochondrial efficiency in skeletal muscle mitochondria. This study was performed on quadriceps muscle of 20 wild-type mice. Muscle tissue was dissected and either permeabilized (n = 10) or homogenized for isolation of mitochondria (n = 10), and oxidative phosphorylation capacity and P/O ratio were assessed using high-resolution respirometry. Samples from each muscle were analyzed in both normal physiological conditions (37°C, pH 7.4), decreased pH (6.8), increased temperature (40°C), and a combination of both. The combination of increased temperature and decreased pH resulted in a significantly lower P/O ratio, mirrored by an increase in leak respiration and a decrease in respiratory control ratio (RCR), in isolated mitochondria. In permeabilized fibers, RCR and leak were relatively unaffected, though a main effect of temperature was observed. Oxidative phosphorylation capacity was unaffected by changes in pH and temperature in both isolated mitochondria and permeabilized fibers. These results indicate that exercise-like changes in temperature and pH lead to impaired mitochondrial efficiency. These findings offer some degree of support to the concept of decreased mitochondrial efficiency during exercise, and may have implications for the assessment of mitochondrial function related to exercise.NEW & NOTEWORTHY To the best of our knowledge, this is the first study to examine the effects of combined changes in temperature and pH, mimicking intramuscular alterations during exercise. Our findings suggest that mitochondrial efficiency is impaired during exercise of moderate to high intensity, which could be a possible mechanism contributing to the decline in exercise efficiency at intensities above the lactate threshold.
Collapse
Affiliation(s)
- Mathias Flensted-Jensen
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ann-Sofie Kleis-Olsen
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rasmus Kinimond Hassø
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Søren Lindtofte
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Juan Corral Pérez
- MOVE-IT Research Group, Department of Physical Education, Faculty of Education Sciences, University of Cádiz, Cádiz, Spain
- ExPhy Research Group, Department of Physical Education, Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Universidad de Cádiz, Cádiz, Spain
| | - Sonia Ortega-Gómez
- MOVE-IT Research Group, Department of Physical Education, Faculty of Education Sciences, University of Cádiz, Cádiz, Spain
- Biomedical Research and Innovation Institute of Cádiz (INiBICA) Research Unit, Puerta del Mar University Hospital, Cádiz, Spain
| | - Steen Larsen
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
- Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
7
|
Hernandez-Baixauli J, Chomiciute G, Tracey H, Mora I, Cortés-Espinar AJ, Ávila-Román J, Abasolo N, Palacios-Jordan H, Foguet-Romero E, Suñol D, Galofré M, Alcaide-Hidalgo JM, Baselga-Escudero L, del Bas JM, Mulero M. Exploring Metabolic and Gut Microbiome Responses to Paraquat Administration in Male Wistar Rats: Implications for Oxidative Stress. Antioxidants (Basel) 2024; 13:67. [PMID: 38247491 PMCID: PMC10812659 DOI: 10.3390/antiox13010067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/22/2023] [Accepted: 12/27/2023] [Indexed: 01/23/2024] Open
Abstract
In this study, we examined the metabolic and gut microbiome responses to paraquat (PQ) in male Wistar rats, focusing on oxidative stress effects. Rats received a single intraperitoneal injection of PQ at 15 and 30 mg/kg, and various oxidative stress parameters (i.e., MDA, SOD, ROS, 8-isoprostanes) were assessed after three days. To explore the omic profile, GC-qTOF and UHPLC-qTOF were performed to assess the plasma metabolome; 1H-NMR was used to assess the urine metabolome; and shotgun metagenomics sequencing was performed to study the gut microbiome. Our results revealed reductions in body weight and tissue changes, particularly in the liver, were observed, suggesting a systemic effect of PQ. Elevated lipid peroxidation and reactive oxygen species levels in the liver and plasma indicated the induction of oxidative stress. Metabolic profiling revealed changes in the tricarboxylic acid cycle, accumulation of ketone body, and altered levels of key metabolites, such as 3-hydroxybutyric acid and serine, suggesting intricate links between energy metabolism and redox reactions. Plasma metabolomic analysis revealed alterations in mitochondrial metabolism, nicotinamide metabolism, and tryptophan degradation. The gut microbiome showed shifts, with higher PQ doses influencing microbial populations (e.g., Escherichia coli and Akkermansia muciniphila) and metagenomic functions (pyruvate metabolism, fermentation, nucleotide and amino acid biosynthesis). Overall, this study provides comprehensive insights into the complex interplay between PQ exposure, metabolic responses, and gut microbiome dynamics. These findings enhance our understanding of the mechanisms behind oxidative stress-induced metabolic alterations and underscore the connections between xenobiotic exposure, gut microbiota, and host metabolism.
Collapse
Affiliation(s)
- Julia Hernandez-Baixauli
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain; (J.H.-B.); (G.C.); (H.T.); (J.M.A.-H.); (L.B.-E.)
- Laboratory of Metabolism and Obesity, Vall d’Hebron-Institut de Recerca, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Gertruda Chomiciute
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain; (J.H.-B.); (G.C.); (H.T.); (J.M.A.-H.); (L.B.-E.)
| | - Harry Tracey
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain; (J.H.-B.); (G.C.); (H.T.); (J.M.A.-H.); (L.B.-E.)
- Department of Medical Sciences, School of Medicine, University of Girona, 17004 Girona, Spain
- School of Science, RMIT University, Bundoora, VIC 3000, Australia
| | - Ignasi Mora
- Brudy Technology S.L., 08006 Barcelona, Spain;
| | - Antonio J. Cortés-Espinar
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, 43007 Tarragona, Spain;
| | - Javier Ávila-Román
- Molecular and Applied Pharmacology Group (FARMOLAP), Department of Pharmacology, Universidad de Sevilla, 41012 Sevilla, Spain;
| | - Nerea Abasolo
- Eurecat, Centre Tecnològic de Catalunya, Centre for Omic Sciences (COS), Joint Unit Universitat Rovira i Virgili-EURECAT, 43204 Reus, Spain; (N.A.); (H.P.-J.); (E.F.-R.)
| | - Hector Palacios-Jordan
- Eurecat, Centre Tecnològic de Catalunya, Centre for Omic Sciences (COS), Joint Unit Universitat Rovira i Virgili-EURECAT, 43204 Reus, Spain; (N.A.); (H.P.-J.); (E.F.-R.)
| | - Elisabet Foguet-Romero
- Eurecat, Centre Tecnològic de Catalunya, Centre for Omic Sciences (COS), Joint Unit Universitat Rovira i Virgili-EURECAT, 43204 Reus, Spain; (N.A.); (H.P.-J.); (E.F.-R.)
| | - David Suñol
- Eurecat, Centre Tecnològic de Catalunya, Digital Health, 08005 Barcelona, Spain; (D.S.); (M.G.)
| | - Mar Galofré
- Eurecat, Centre Tecnològic de Catalunya, Digital Health, 08005 Barcelona, Spain; (D.S.); (M.G.)
| | - Juan María Alcaide-Hidalgo
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain; (J.H.-B.); (G.C.); (H.T.); (J.M.A.-H.); (L.B.-E.)
| | - Laura Baselga-Escudero
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain; (J.H.-B.); (G.C.); (H.T.); (J.M.A.-H.); (L.B.-E.)
| | - Josep M. del Bas
- Eurecat, Centre Tecnològic de Catalunya, Àrea Biotecnologia, 43204 Reus, Spain
| | - Miquel Mulero
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, 43007 Tarragona, Spain;
| |
Collapse
|
8
|
Mousavi-Aghdas SA, Farashi E, Naderi N. Iron Dyshomeostasis and Mitochondrial Function in the Failing Heart: A Review of the Literature. Am J Cardiovasc Drugs 2024; 24:19-37. [PMID: 38157159 DOI: 10.1007/s40256-023-00619-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/20/2023] [Indexed: 01/03/2024]
Abstract
Cardiac contraction and relaxation require a substantial amount of energy provided by the mitochondria. The failing heart is adenosine triphosphate (ATP)- and creatine-depleted. Studies have found iron is involved in almost every aspect of mitochondrial function, and previous studies have shown myocardial iron deficiency in heart failure (HF). Many clinicians advocated intravenous iron repletion for HF patients meeting the conventional criteria for systemic iron deficiency. While clinical trials showed improved quality of life, iron repletion failed to significantly impact survival or significant cardiovascular adverse events. There is evidence that in HF, labile iron is trapped inside the mitochondria causing oxidative stress and lipid peroxidation. There is also compelling preclinical evidence demonstrating the detrimental effects of both iron overload and depletion on cardiomyocyte function. We reviewed the mechanisms governing myocardial and mitochondrial iron content. Mitochondrial dynamics (i.e., fusion, fission, mitophagy) and the role of iron were also investigated. Ferroptosis, as an important regulated cell death mechanism involved in cardiomyocyte loss, was reviewed along with agents used to manipulate it. The membrane stability and iron content of mitochondria can be altered by many agents. Some studies are showing promising improvement in the cardiomyocyte function after iron chelation by deferiprone; however, whether the in vitro and in vivo findings will be reflected on on clinical grounds is still unclear. Finally, we briefly reviewed the clinical trials on intravenous iron repletion. There is a need for more well-simulated animal studies to shed light on the safety and efficacy of chelation agents and pave the road for clinical studies.
Collapse
Affiliation(s)
- Seyed Ali Mousavi-Aghdas
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Rajaie Cardiovascular, Medical, and Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ebrahim Farashi
- Department of Cardiothoracic Surgery, Imam Reza Medical Research & Training Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
- Rajaie Cardiovascular, Medical, and Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Nasim Naderi
- Department of Cardiothoracic Surgery, Imam Reza Medical Research & Training Hospital, Tabriz University of Medical Sciences, Tabriz, Iran.
- Rajaie Cardiovascular, Medical, and Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Chalifoux O, Faerman B, Mailloux RJ. Mitochondrial hydrogen peroxide production by pyruvate dehydrogenase and α-ketoglutarate dehydrogenase in oxidative eustress and oxidative distress. J Biol Chem 2023; 299:105399. [PMID: 37898400 PMCID: PMC10692731 DOI: 10.1016/j.jbc.2023.105399] [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: 07/26/2023] [Revised: 10/06/2023] [Accepted: 10/16/2023] [Indexed: 10/30/2023] Open
Abstract
Pyruvate dehydrogenase (PDH) and α-ketoglutarate dehydrogenase (KGDH) are vital entry points for monosaccharides and amino acids into the Krebs cycle and thus integral for mitochondrial bioenergetics. Both complexes produce mitochondrial hydrogen peroxide (mH2O2) and are deactivated by electrophiles. Here, we provide an update on the role of PDH and KGDH in mitochondrial redox balance and their function in facilitating metabolic reprogramming for the propagation of oxidative eustress signals in hepatocytes and how defects in these pathways can cause liver diseases. PDH and KGDH are known to account for ∼45% of the total mH2O2 formed by mitochondria and display rates of production several-fold higher than the canonical source complex I. This mH2O2 can also be formed by reverse electron transfer (RET) in vivo, which has been linked to metabolic dysfunctions that occur in pathogenesis. However, the controlled emission of mH2O2 from PDH and KGDH has been proposed to be fundamental for oxidative eustress signal propagation in several cellular contexts. Modification of PDH and KGDH with protein S-glutathionylation (PSSG) and S-nitrosylation (PSNO) adducts serves as a feedback inhibitor for mH2O2 production in response to glutathione (GSH) pool oxidation. PSSG and PSNO adduct formation also reprogram the Krebs cycle to generate metabolites vital for interorganelle and intercellular signaling. Defects in the redox modification of PDH and KGDH cause the over generation of mH2O2, resulting in oxidative distress and metabolic dysfunction-associated fatty liver disease (MAFLD). In aggregate, PDH and KGDH are essential platforms for emitting and receiving oxidative eustress signals.
Collapse
Affiliation(s)
- Olivia Chalifoux
- Faculty of Agricultural and Environmental Sciences, The School of Human Nutrition, McGill University, Ste.-Anne-de-Bellevue, Quebec, Canada
| | - Ben Faerman
- Faculty of Agricultural and Environmental Sciences, The School of Human Nutrition, McGill University, Ste.-Anne-de-Bellevue, Quebec, Canada
| | - Ryan J Mailloux
- Faculty of Agricultural and Environmental Sciences, The School of Human Nutrition, McGill University, Ste.-Anne-de-Bellevue, Quebec, Canada.
| |
Collapse
|
10
|
Davis MS, Bayly WM, Hansen CM, Barrett MR, Blake CA. Effects of hyperthermia and acidosis on mitochondrial production of reactive oxygen species. Am J Physiol Regul Integr Comp Physiol 2023; 325:R725-R734. [PMID: 37811714 DOI: 10.1152/ajpregu.00177.2023] [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: 07/18/2023] [Revised: 09/06/2023] [Accepted: 10/06/2023] [Indexed: 10/10/2023]
Abstract
Exercise is associated with the development of oxidative stress, but the specific source and mechanism of production of pro-oxidant chemicals during exercise has not been confirmed. We used equine skeletal muscle mitochondria to test the hypothesis that hyperthermia and acidosis affect mitochondrial oxygen consumption and production of reactive oxygen species (ROS). Skeletal muscle biopsies were obtained at rest, after an acute episode of fatiguing exercise, and after a 9-wk conditioning program to increase aerobic fitness. Mitochondrial oxygen consumption and ROS production were measured simultaneously using high-resolution respirometry. Both hyperthermia and acidosis increased nonphosphorylating (LEAK) respiration (5.8× and 3.0×, respectively, P < 0.001) and decreased efficiency of oxidative phosphorylation. The combined effects of hyperthermia and acidosis resulted in large decreases in phosphorylating respiration, further decreasing oxidative phosphorylation efficiency from 97% to 86% (P < 0.01). Increased aerobic fitness reduced the effects of acidosis on LEAK respiration. Hyperthermia increased and acidosis decreased ROS production (2× and 0.23×, respectively, P < 0.001). There was no effect of acute exercise, but an aerobic conditioning program was associated with increased ROS production during both nonphosphorylating and phosphorylating respiration. Hyperthermia increased the ratio of ROS production to O2 consumption during phosphorylating respiration, suggesting that high-temperature impaired transfer of energy through the electron transfer system despite relatively low mitochondrial membrane potential. These data support the role of skeletal muscle mitochondria in the development of exercise-induced oxidative stress, particularly during forms of exercise that result in prolonged hyperthermia without acidosis.NEW & NOTEWORTHY The results of this study provide evidence for the role of mitochondria-derived ROS in the development of systemic oxidative stress during exercise as well as skeletal muscle diseases such as exertional rhabdomyolysis.
Collapse
Affiliation(s)
- Michael S Davis
- Department of Physiological Sciences, Oklahoma State University, Stillwater, Oklahoma, United States
| | - Warwick M Bayly
- Department of Veterinary Clinical Sciences, Washington State University, Pullman, Washington, United States
| | - Cristina M Hansen
- Department of Veterinary Medicine, University of Alaska Fairbanks, Fairbanks, Alaska, United States
| | - Montana R Barrett
- Department of Physiological Sciences, Oklahoma State University, Stillwater, Oklahoma, United States
| | - Cara A Blake
- Central Hospital for Veterinary Medicine, North Haven, Connecticut, United States
| |
Collapse
|
11
|
Decker ST, Matias AA, Cuadra AE, Bannon ST, Madden JP, Erol ME, Serviente C, Fenelon K, Layec G. Tissue-specific mitochondrial toxicity of cigarette smoke concentrate: consequence to oxidative phosphorylation. Am J Physiol Heart Circ Physiol 2023; 325:H1088-H1098. [PMID: 37712922 PMCID: PMC10907033 DOI: 10.1152/ajpheart.00199.2023] [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: 04/10/2023] [Revised: 09/11/2023] [Accepted: 09/12/2023] [Indexed: 09/16/2023]
Abstract
Cigarette smoke exposure is a well-known risk factor for developing numerous chronic health conditions, including pulmonary disease and cardiometabolic disorders. However, the cellular mechanisms mediating the toxicity of cigarette smoke in extrapulmonary tissues are still poorly understood. Therefore, the purpose of this study was to characterize the acute dose-dependent toxicity of cigarette smoke on mitochondrial metabolism by determining the susceptibility and sensitivity of mitochondrial respiration from murine skeletal (gastrocnemius and soleus) and cardiac muscles, as well as the aorta to cigarette smoke concentrate (CSC). In all tissues, exposure to CSC inhibited tissue-specific respiration capacity, measured by high-resolution respirometry, according to a biphasic pattern. With a break point of 451 ± 235 μg/mL, the aorta was the least susceptible to CSC-induced mitochondrial respiration inhibition compared with the gastrocnemius (151 ± 109 μg/mL; P = 0.008, d = 2.3), soleus (211 ± 107 μg/mL; P = 0.112; d = 1.7), and heart (94 ± 51 μg/mL; P < 0.001; d = 2.6) suggesting an intrinsic resistance of the vascular smooth muscle mitochondria to cigarette smoke toxicity. In contrast, the cardiac muscle was the most susceptible and sensitive to the effects of CSC, demonstrating the greatest decline in tissue-specific respiration with increasing CSC concentration (P < 0.001, except the soleus). However, when normalized to citrate synthase activity to account for differences in mitochondrial content, cardiac fibers' sensitivity to cigarette smoke inhibition was no longer significantly different from both fast-twitch gastrocnemius and slow-twitch soleus muscle fibers, thus suggesting similar mitochondrial phenotypes. Collectively, these findings established the acute dose-dependent toxicity of cigarette smoke on oxidative phosphorylation in permeabilized tissues involved in the development of smoke-related cardiometabolic diseases.NEW & NOTEWORTHY Despite numerous investigations into the mechanisms underlying cigarette smoke-induced mitochondrial dysfunction, no studies have investigated the tissue-specific mitochondrial toxicity to cigarette smoke. We demonstrate that, while aorta is least sensitive and susceptible to cigarette smoke-induced toxicity, the degree of cigarette smoke-induced toxicity in striated muscle depends on the tissue-specific mitochondrial content. We conclude that while the mitochondrial content influences cigarette smoke-induced toxicity in striated muscles, aorta is intrinsically protected against cigarette smoke-induced mitochondrial toxicity.
Collapse
Affiliation(s)
- Stephen T Decker
- Department of Kinesiology, University of Massachusetts Amherst, Amherst, Massachusetts, United States
| | - Alexs A Matias
- Department of Kinesiology, University of Massachusetts Amherst, Amherst, Massachusetts, United States
| | - Adolfo E Cuadra
- Department of Biology, University of Massachusetts Amherst, Amherst, Massachusetts, United States
| | - Sean T Bannon
- Department of Kinesiology, University of Massachusetts Amherst, Amherst, Massachusetts, United States
| | - Jack P Madden
- Department of Kinesiology, University of Massachusetts Amherst, Amherst, Massachusetts, United States
| | - M Enes Erol
- Department of Kinesiology, University of Massachusetts Amherst, Amherst, Massachusetts, United States
| | - Corinna Serviente
- Department of Kinesiology, University of Massachusetts Amherst, Amherst, Massachusetts, United States
- Institute for Applied Life Science, University of Massachusetts Amherst, Amherst, Massachusetts, United States
| | - Karine Fenelon
- Institute for Applied Life Science, University of Massachusetts Amherst, Amherst, Massachusetts, United States
- Department of Biology, University of Massachusetts Amherst, Amherst, Massachusetts, United States
| | - Gwenael Layec
- Department of Kinesiology, University of Massachusetts Amherst, Amherst, Massachusetts, United States
- Institute for Applied Life Science, University of Massachusetts Amherst, Amherst, Massachusetts, United States
| |
Collapse
|
12
|
Scasny A, Alibayov B, Khan F, Rao SJ, Murin L, Jop Vidal AG, Smith P, Wei L, Edwards K, Warncke K, Vidal JE. Oxidation of hemoproteins by Streptococcus pneumoniae collapses the cell cytoskeleton and disrupts mitochondrial respiration leading to cytotoxicity of human lung cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.07.544089. [PMID: 37333138 PMCID: PMC10274756 DOI: 10.1101/2023.06.07.544089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Streptococcus pneumoniae (Spn) causes pneumonia that kills millions through acute toxicity and invasion of the lung parenchyma. During aerobic respiration, Spn releases hydrogen peroxide (Spn-H 2 O 2 ), as a by-product of enzymes SpxB and LctO, and causes cell death with signs of both apoptosis and pyroptosis by oxidizing unknown cell targets. Hemoproteins are molecules essential for life and prone to oxidation by H 2 O 2 . We recently demonstrated that during infection-mimicking conditions, Spn-H 2 O 2 oxidizes the hemoprotein hemoglobin (Hb), releasing toxic heme. In this study, we investigated details of the molecular mechanism(s) by which the oxidation of hemoproteins by Spn-H 2 O 2 causes human lung cell death. Spn strains, but not H 2 O 2 -deficient SpnΔ spxB Δ lctO strains caused time-dependent cell cytotoxicity characterized by the rearrangement of the actin, the loss of the microtubule cytoskeleton and nuclear contraction. Disruption of the cell cytoskeleton correlated with the presence of invasive pneumococci and an increase of intracellular reactive oxygen species. In cell culture, the oxidation of Hb or cytochrome c (Cyt c ) caused DNA degradation and mitochondrial dysfunction from inhibition of complex I-driven respiration, which was cytotoxic to human alveolar cells. Oxidation of hemoproteins resulted in the creation of a radical, which was identified as a protein derived side chain tyrosyl radical by using electron paramagnetic resonance (EPR). Thus, we demonstrate that Spn invades lung cells, releasing H 2 O 2 that oxidizes hemoproteins, including Cyt c , catalyzing the formation of a tyrosyl side chain radical on Hb and causing mitochondrial disruption, that ultimately leads to the collapse of the cell cytoskeleton.
Collapse
|
13
|
Wang Z, Zhao D, Qin S, Shi Z, Li X, Wang Y, Shao Y. Effects of Dietary Supplementation with Iron in Breeding Pigeons on the Blood Iron Status, Tissue Iron Content, and Full Expression of Iron-Containing Enzymes of Squabs. Biol Trace Elem Res 2022:10.1007/s12011-022-03530-x. [PMID: 36542305 DOI: 10.1007/s12011-022-03530-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022]
Abstract
This study was aimed at investigating the effects of diet iron levels on the blood iron status, tissue iron content, mRNA levels, and the activity of iron-containing enzymes in different tissues of squabs. A total of 120 pairs of healthy Silver Feather King parental pigeons with similar average body weight and egg production were randomly divided into 5 groups with 8 replicates and 3 pairs of pigeons per replicate. The five groups of breeding pigeons were fed an iron-unsupplemented basal diet and basal diet supplemented with 75, 150, 300, and 600 mg iron/kg, respectively. The diets were fed in the form of granular feed based on corn, soybean meal, wheat, and sorghum. A broken line model was used for regression analysis. The results showed that plasma iron (PI), serum ferritin, iron contents in crop milk and liver, liver catalase (CAT) activity, and heart succinate dehydrogenase (SDH) activity were affected by iron levels (P < 0.05). And PI, serum ferritin, iron content in crop milk, and heart SDH activity increased quadratically (P < 0.05), but the iron content and CAT activity in the liver decreased quadratically (P < 0.005) as dietary iron level increased. According to the broken-line model of serum ferritin fitting (P < 0.002), the optimal dietary iron level of breeding pigeons was estimated to be 193 mg/kg. In conclusion, serum ferritin is a sensitive index to evaluate the iron requirement of the breeding pigeon with two squabs, and the recommended iron supplemental level is 193 mg/kg.
Collapse
Affiliation(s)
- Zheng Wang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, 100097, China
| | - Dongdong Zhao
- Faculty of Animal Science and Technology, Gansu Agricultural University, No. 1 Yingmen Village, Anning District, Lanzhou, 730070, Gansu Province, China
| | - Shizhen Qin
- Faculty of Animal Science and Technology, Gansu Agricultural University, No. 1 Yingmen Village, Anning District, Lanzhou, 730070, Gansu Province, China
| | - Zhaoguo Shi
- Faculty of Animal Science and Technology, Gansu Agricultural University, No. 1 Yingmen Village, Anning District, Lanzhou, 730070, Gansu Province, China
| | - Xing Li
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, 100097, China
| | - Yangyang Wang
- Faculty of Animal Science and Technology, Gansu Agricultural University, No. 1 Yingmen Village, Anning District, Lanzhou, 730070, Gansu Province, China
| | - Yuxin Shao
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, 100097, China.
| |
Collapse
|
14
|
Staples JF, Mathers KE, Duffy BM. Mitochondrial Metabolism in Hibernation: Regulation and Implications. Physiology (Bethesda) 2022; 37:0. [PMID: 35658625 DOI: 10.1152/physiol.00006.2022] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Hibernators rapidly and reversibly suppress mitochondrial respiration and whole animal metabolism. Posttranslational modifications likely regulate these mitochondrial changes, which may help conserve energy in winter. These modifications are affected by reactive oxygen species (ROS), so suppressing mitochondrial ROS production may also be important for hibernators, just as it is important for surviving ischemia-reperfusion injury.
Collapse
Affiliation(s)
- James F Staples
- Department of Biology, University of Western Ontario, London, Ontario, Canada
| | - Katherine E Mathers
- Department of Biology, University of Western Ontario, London, Ontario, Canada
| | - Brynne M Duffy
- Department of Biology, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
15
|
Chemistry of Hydrogen Peroxide Formation and Elimination in Mammalian Cells, and Its Role in Various Pathologies. STRESSES 2022. [DOI: 10.3390/stresses2030019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Hydrogen peroxide (H2O2) is a compound involved in some mammalian reactions and processes. It modulates and signals the redox metabolism of cells by acting as a messenger together with hydrogen sulfide (H2S) and the nitric oxide radical (•NO), activating specific oxidations that determine the metabolic response. The reaction triggered determines cell survival or apoptosis, depending on which downstream metabolic pathways are activated. There are several ways to produce H2O2 in cells, and cellular systems tightly control its concentration. At the cellular level, the accumulation of hydrogen peroxide can trigger inflammation and even apoptosis, and when its concentration in the blood reaches toxic levels, it can lead to bioenergetic failure. This review summarizes existing research from a chemical perspective on the role of H2O2 in various enzymatic pathways and how this biochemistry leads to physiological or pathological responses.
Collapse
|
16
|
Jung P, Ha E, Zhang M, Fall C, Hwang M, Taylor E, Stetkevich S, Bhanot A, Wilson CG, Figueroa JD, Obenaus A, Bragg S, Tone B, Eliamani S, Holshouser B, Blood AB, Liu T. Neuroprotective role of nitric oxide inhalation and nitrite in a Neonatal Rat Model of Hypoxic-Ischemic Injury. PLoS One 2022; 17:e0268282. [PMID: 35544542 PMCID: PMC9094545 DOI: 10.1371/journal.pone.0268282] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 04/26/2022] [Indexed: 12/01/2022] Open
Abstract
Background There is evidence from various models of hypoxic-ischemic injury (HII) that nitric oxide (NO) is protective. We hypothesized that either inhaled NO (iNO) or nitrite would alleviate brain injury in neonatal HII via modulation of mitochondrial function. Methods We tested the effects of iNO and nitrite on the Rice-Vannucci model of HII in 7-day-old rats. Brain mitochondria were isolated for flow cytometry, aconitase activity, electron paramagnetic resonance, and Seahorse assays. Results Pretreatment of pups with iNO decreased survival in the Rice-Vannucci model of HII, while iNO administered post-insult did not. MRI analysis demonstrated that pre-HII iNO at 40 ppm and post-HII iNO at 20 ppm decreased the brain lesion sizes from 6.3±1.3% to 1.0±0.4% and 1.8±0.8%, respectively. Intraperitoneal nitrite at 0.165 μg/g improved neurobehavioral performance but was harmful at higher doses and had no effect on brain infarct size. NO reacted with complex IV at the heme a3 site, decreased the oxidative stress of mitochondria challenged with anoxia and reoxygenation, and suppressed mitochondrial oxygen respiration. Conclusions This study suggests that iNO administered following neonatal HII may be neuroprotective, possibly via its modulation of mitochondrial function.
Collapse
Affiliation(s)
- Peter Jung
- Division of Neonatology, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
| | - Euntaik Ha
- Division of Neonatology, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
| | - Meijuan Zhang
- Division of Neonatology, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
| | - Carolyn Fall
- Division of Neonatology, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
| | - Mindy Hwang
- Division of Neonatology, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
| | - Emily Taylor
- Division of Neonatology, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
| | - Samuel Stetkevich
- Division of Neonatology, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
| | - Aditi Bhanot
- Division of Neonatology, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
| | - Christopher G. Wilson
- Division of Neonatology, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
- Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
| | - Johnny D. Figueroa
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
| | - Andre Obenaus
- Department of Pediatrics, School of Medicine, University of California, Irvine, CA, United States of America
| | - Shannon Bragg
- Division of Neonatology, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
| | - Beatriz Tone
- Division of Neonatology, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
| | - Saburi Eliamani
- Center for Imaging Research, Department of Radiology, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
| | - Barbara Holshouser
- Center for Imaging Research, Department of Radiology, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
| | - Arlin B. Blood
- Division of Neonatology, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
- Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
| | - Taiming Liu
- Division of Neonatology, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
- * E-mail:
| |
Collapse
|
17
|
Hernández-Cuervo H, Soundararajan R, Sidramagowda Patil S, Breitzig M, Alleyn M, Galam L, Lockey R, Uversky VN, Kolliputi N. BMI1 Silencing Induces Mitochondrial Dysfunction in Lung Epithelial Cells Exposed to Hyperoxia. Front Physiol 2022; 13:814510. [PMID: 35431986 PMCID: PMC9005903 DOI: 10.3389/fphys.2022.814510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 02/04/2022] [Indexed: 11/17/2022] Open
Abstract
Acute Lung Injury (ALI), characterized by bilateral pulmonary infiltrates that restrict gas exchange, leads to respiratory failure. It is caused by an innate immune response with white blood cell infiltration of the lungs, release of cytokines, an increase in reactive oxygen species (ROS), oxidative stress, and changes in mitochondrial function. Mitochondrial alterations, changes in respiration, ATP production and the unbalancing fusion and fission processes are key events in ALI pathogenesis and increase mitophagy. Research indicates that BMI1 (B cell-specific Moloney murine leukemia virus integration site 1), a protein of the Polycomb repressive complex 1, is a cell cycle and survival regulator that plays a role in mitochondrial function. BMI1-silenced cultured lung epithelial cells were exposed to hyperoxia to determine the role of BMI1 in mitochondrial metabolism. Its expression significantly decreases in human lung epithelial cells (H441) following hyperoxic insult, as determined by western blot, Qrt-PCR, and functional analysis. This decrease correlates with an increase in mitophagy proteins, PINK1, Parkin, and DJ1; an increase in the expression of tumor suppressor PTEN; changes in the expression of mitochondrial biomarkers; and decreases in the oxygen consumption rate (OCR) and tricarboxylic acid enzyme activity. Our bioinformatics analysis suggested that the BMI1 multifunctionality is determined by its high level of intrinsic disorder that defines the ability of this protein to bind to numerous cellular partners. These results demonstrate a close relationship between BMI1 expression and mitochondrial health in hyperoxia-induced acute lung injury (HALI) and indicate that BMI1 is a potential therapeutic target to treat ALI and Acute Respiratory Distress Syndrome.
Collapse
Affiliation(s)
- Helena Hernández-Cuervo
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Ramani Soundararajan
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Sahebgowda Sidramagowda Patil
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Mason Breitzig
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
- Division of Epidemiology, Department of Public Health Sciences, College of Medicine, Pennsylvania State University, Hershey, PA, United States
| | - Matthew Alleyn
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Lakshmi Galam
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Richard Lockey
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Vladimir N. Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Narasaiah Kolliputi
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
- *Correspondence: Narasaiah Kolliputi,
| |
Collapse
|
18
|
Spyropoulos F, Sorrentino A, van der Reest J, Yang P, Waldeck-Weiermair M, Steinhorn B, Eroglu E, Saeedi Saravi SS, Yu P, Haigis M, Christou H, Michel T. Metabolomic and transcriptomic signatures of chemogenetic heart failure. Am J Physiol Heart Circ Physiol 2022; 322:H451-H465. [PMID: 35089810 PMCID: PMC8896991 DOI: 10.1152/ajpheart.00628.2021] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The failing heart is characterized by elevated levels of reactive oxygen species. We have developed an animal model of heart failure induced by chemogenetic production of oxidative stress in the heart using a recombinant adeno-associated virus (AAV9) expressing yeast d-amino acid oxidase (DAAO) targeted to cardiac myocytes. When DAAO-infected animals are fed the DAAO substrate d-alanine, the enzyme generates hydrogen peroxide (H2O2) in the cardiac myocytes, leading to dilated cardiomyopathy. However, the underlying mechanisms of oxidative stress-induced heart failure remain incompletely understood. Therefore, we investigated the effects of chronic oxidative stress on the cardiac transcriptome and metabolome. Rats infected with recombinant cardiotropic AAV9 expressing DAAO or control AAV9 were treated for 7 wk with d-alanine to stimulate chemogenetic H2O2 production by DAAO and generate dilated cardiomyopathy. After hemodynamic assessment, left and right ventricular tissues were processed for RNA sequencing and metabolomic profiling. DAAO-induced dilated cardiomyopathy was characterized by marked changes in the cardiac transcriptome and metabolome both in the left and right ventricle. Downregulated transcripts are related to energy metabolism and mitochondrial function, accompanied by striking alterations in metabolites involved in cardiac energetics, redox homeostasis, and amino acid metabolism. Upregulated transcripts are involved in cytoskeletal organization and extracellular matrix. Finally, we noted increased metabolite levels of antioxidants glutathione and ascorbate. These findings provide evidence that chemogenetic generation of oxidative stress leads to a robust heart failure model with distinct transcriptomic and metabolomic signatures and set the basis for understanding the underlying pathophysiology of chronic oxidative stress in the heart.NEW & NOTEWORTHY We have developed a "chemogenetic" heart failure animal model that recapitulates a central feature of human heart failure: increased cardiac redox stress. We used a recombinant DAAO enzyme to generate H2O2 in cardiomyocytes, leading to cardiomyopathy. Here we report striking changes in the cardiac metabolome and transcriptome following chemogenetic heart failure, similar to changes observed in human heart failure. Our findings help validate chemogenetic approaches for the discovery of novel therapeutic targets in heart failure.
Collapse
Affiliation(s)
- Fotios Spyropoulos
- 1Department of Pediatric Newborn Medicine, Brigham and
Women’s Hospital, Harvard Medical School, Boston, Massachusetts,2Cardiovascular Division, Department of Medicine, Brigham
and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Andrea Sorrentino
- 2Cardiovascular Division, Department of Medicine, Brigham
and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Peiran Yang
- 2Cardiovascular Division, Department of Medicine, Brigham
and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Markus Waldeck-Weiermair
- 2Cardiovascular Division, Department of Medicine, Brigham
and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Benjamin Steinhorn
- 2Cardiovascular Division, Department of Medicine, Brigham
and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Emrah Eroglu
- 2Cardiovascular Division, Department of Medicine, Brigham
and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Seyed Soheil Saeedi Saravi
- 2Cardiovascular Division, Department of Medicine, Brigham
and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Paul Yu
- 2Cardiovascular Division, Department of Medicine, Brigham
and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Marcia Haigis
- 3Department of Cell Biology, Harvard Medical School, Boston, Massachusetts
| | - Helen Christou
- 1Department of Pediatric Newborn Medicine, Brigham and
Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Thomas Michel
- 2Cardiovascular Division, Department of Medicine, Brigham
and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
19
|
Homocysteine-Thiolactone Modulates Gating of Mitochondrial Voltage-Dependent Anion Channel (VDAC) and Protects It from Induced Oxidative Stress. J Membr Biol 2022; 255:79-97. [PMID: 35103807 DOI: 10.1007/s00232-022-00215-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 01/10/2022] [Indexed: 12/26/2022]
Abstract
The gating of the Voltage-Dependent Anion Channel (VDAC) is linked to oxidative stress through increased generation of mitochondrial ROS with increasing mitochondrial membrane potential (ΔΨm). It has been already reported that H2O2 increases the single-channel conductance of VDAC on a bilayer lipid membrane. On the other hand, homocysteine (Hcy) has been reported to induce mitochondria-mediated cell death. It is argued that the thiol-form of homocysteine, HTL could be the plausible molecule responsible for the alteration in the function of proteins, such as VDAC. It is hypothesized that HTL interacts with VDAC that causes functional abnormalities. An investigation was undertaken to study the interaction of HTL with VDAC under H2O2 induced oxidative stress through biophysical and electrophysiological methods. Fluorescence spectroscopic studies indicate that HTL interacts with VDAC, but under induced oxidative stress the effect is prevented partially. Similarly, bilayer electrophysiology studies suggest that HTL shows a reduction in VDAC single-channel conductance, but the effects are partially prevented under an oxidative environment. Gly172 and His181 are predicted through bioinformatics tools to be the most plausible binding residues of HTL in Rat VDAC. The binding of HTL and H2O2 with VDAC appears to be cooperative as per our analysis of experimental data in the light of the Hill-Langmuir equation. The binding energies are estimated to be - 4.7 kcal mol-1 and - 2.8 kcal mol-1, respectively. The present in vitro studies suggest that when mitochondrial VDAC is under oxidative stress, the effects of amino acid metabolites like HTL are suppressed.
Collapse
|
20
|
The cardiac-enriched microprotein mitolamban regulates mitochondrial respiratory complex assembly and function in mice. Proc Natl Acad Sci U S A 2022; 119:2120476119. [PMID: 35101990 PMCID: PMC8833175 DOI: 10.1073/pnas.2120476119] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2021] [Indexed: 12/12/2022] Open
Abstract
Microproteins are a growing class of versatile small proteins previously overlooked by standard gene annotation methods due to their small size. Here we characterize mitolamban as a cardiac-enriched inner mitochondrial membrane–localized microprotein, which interacts with complex III of the electron transport chain and contributes to complex III assembly and function. Mitolamban gene deletion in mice leads to a reduction in complex III activity and metabolic perturbations in the heart that are consistent with complex III deficiency, as well as altered complex III assembly into respiratory supercomplexes. These findings define a functional role for mitolamban in the heart and highlight the importance of microproteins in regulating mitochondrial function and cardiomyocyte biology. Emerging evidence indicates that a subset of RNA molecules annotated as noncoding contain short open reading frames that code for small functional proteins called microproteins, which have largely been overlooked due to their small size. To search for cardiac-expressed microproteins, we used a comparative genomics approach and identified mitolamban (Mtlbn) as a highly conserved 47-amino acid transmembrane protein that is abundantly expressed in the heart. Mtlbn localizes specifically to the inner mitochondrial membrane where it interacts with subunits of complex III of the electron transport chain and with mitochondrial respiratory supercomplexes. Genetic deletion of Mtlbn in mice altered complex III assembly dynamics and reduced complex III activity. Unbiased metabolomic analysis of heart tissue from Mtlbn knockout mice further revealed an altered metabolite profile consistent with deficiencies in complex III activity. Cardiac-specific Mtlbn overexpression in transgenic (TG) mice induced cardiomyopathy with histological, biochemical, and ultrastructural pathologic features that contributed to premature death. Metabolomic analysis and biochemical studies indicated that hearts from Mtlbn TG mice exhibited increased oxidative stress and mitochondrial dysfunction. These findings reveal Mtlbn as a cardiac-expressed inner mitochondrial membrane microprotein that contributes to mitochondrial electron transport chain activity through direct association with complex III and the regulation of its assembly and function.
Collapse
|
21
|
Surabhi S, Jachmann LH, Lalk M, Hammerschmidt S, Methling K, Siemens N. Bronchial Epithelial Cells Accumulate Citrate Intracellularly in Response to Pneumococcal Hydrogen Peroxide. ACS Infect Dis 2021; 7:2971-2978. [PMID: 34623132 DOI: 10.1021/acsinfecdis.1c00372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Community-acquired pneumonia is an infection of the lower respiratory tract caused by various viral and bacterial pathogens, including influenza A virus (IAV), Streptococcus pneumoniae, and Staphylococcus aureus. To understand the disease pathology, it is important to delineate host metabolic responses to an infection. In this study, metabolome profiling of mono- and coinfected human bronchial epithelial cells was performed. We show that IAV and S. aureus silently survive within the cells with almost negligible effects on the host metabolome. In contrast, S. pneumoniae significantly altered various host pathways such as glycolysis, tricarboxylic acid cycle, and amino acid metabolism. Intracellular citrate accumulation was the most prominent signature of pneumococcal infections and was directly attributed to the action of pneumococci-derived hydrogen peroxide. No coinfection specific metabolome signatures were observed.
Collapse
Affiliation(s)
- Surabhi Surabhi
- Department of Molecular Genetics and Infection Biology, University of Greifswald, D-17489 Greifswald, Germany
| | - Lana H. Jachmann
- Department of Molecular Genetics and Infection Biology, University of Greifswald, D-17489 Greifswald, Germany
| | - Michael Lalk
- Institute of Biochemistry, University of Greifswald, D-17489 Greifswald, Germany
| | - Sven Hammerschmidt
- Department of Molecular Genetics and Infection Biology, University of Greifswald, D-17489 Greifswald, Germany
| | - Karen Methling
- Institute of Biochemistry, University of Greifswald, D-17489 Greifswald, Germany
| | - Nikolai Siemens
- Department of Molecular Genetics and Infection Biology, University of Greifswald, D-17489 Greifswald, Germany
| |
Collapse
|
22
|
Foo BJA, Eu JQ, Hirpara JL, Pervaiz S. Interplay between Mitochondrial Metabolism and Cellular Redox State Dictates Cancer Cell Survival. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:1341604. [PMID: 34777681 PMCID: PMC8580634 DOI: 10.1155/2021/1341604] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 09/30/2021] [Accepted: 10/04/2021] [Indexed: 02/06/2023]
Abstract
Mitochondria are the main powerhouse of the cell, generating ATP through the tricarboxylic acid cycle (TCA) and oxidative phosphorylation (OXPHOS), which drives myriad cellular processes. In addition to their role in maintaining bioenergetic homeostasis, changes in mitochondrial metabolism, permeability, and morphology are critical in cell fate decisions and determination. Notably, mitochondrial respiration coupled with the passage of electrons through the electron transport chain (ETC) set up a potential source of reactive oxygen species (ROS). While low to moderate increase in intracellular ROS serves as secondary messenger, an overwhelming increase as a result of either increased production and/or deficient antioxidant defenses is detrimental to biomolecules, cells, and tissues. Since ROS and mitochondria both regulate cell fate, attention has been drawn to their involvement in the various processes of carcinogenesis. To that end, the link between a prooxidant milieu and cell survival and proliferation as well as a switch to mitochondrial OXPHOS associated with recalcitrant cancers provide testimony for the remarkable metabolic plasticity as an important hallmark of cancers. In this review, the regulation of cell redox status by mitochondrial metabolism and its implications for cancer cell fate will be discussed followed by the significance of mitochondria-targeted therapies for cancer.
Collapse
Affiliation(s)
- Brittney Joy-Anne Foo
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, Singapore
| | - Jie Qing Eu
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, Singapore
- Cancer Science Institute, NUS, Singapore, Singapore
| | | | - Shazib Pervaiz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, NUS, Singapore, Singapore
- NUS Medicine Healthy Longevity Program, Yong Loo Lin School of Medicine, NUS, Singapore, Singapore
- Integrative Sciences and Engineering Program, NUS Graduate School, NUS, Singapore, Singapore
- National University Cancer Institute, National University Health System, Singapore, Singapore
- Faculté de Médicine, Université de Paris, Paris, France
| |
Collapse
|
23
|
Gujarati NA, Leonardo AR, Vasquez JM, Guo Y, Frimpong BO, Fozilov E, Revelo MP, Daehn IS, He JC, Bogenhagen D, Mallipattu SK. Loss of Functional SCO2 Attenuates Oxidative Stress in Diabetic Kidney Disease. Diabetes 2021; 71:db210316. [PMID: 34702781 PMCID: PMC8763871 DOI: 10.2337/db21-0316] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 10/18/2021] [Indexed: 11/13/2022]
Abstract
Increased oxidative stress in glomerular endothelial cells (GEnCs) contributes to early diabetic kidney disease (DKD). While mitochondrial respiratory complex IV activity is reduced in DKD, it remains unclear whether this is a driver or a consequence of oxidative stress in GEnCs. Synthesis of cytochrome C oxidase 2 (SCO2), a key metallochaperone in the electron transport chain, is critical to the biogenesis and assembly of subunits required for functional respiratory complex IV activity. Here, we investigated the effects of Sco2 hypomorphs (Sco2 KO/KI , Sco2 KI/KI ), with a functional loss of SCO2, in the progression of DKD using a murine model of Type II Diabetes Mellitus, db/db mice. Diabetic Sco2 KO/KI and Sco2 KI/KI hypomorphs exhibited a reduction in complex IV activity, but an improvement in albuminuria, serum creatinine, and histomorphometric evidence of early DKD as compared to db/db mice. Single-nucleus RNA sequencing with gene set enrichment analysis of differentially expressed genes in the endothelial cluster of Sco2 KO/KI ;db/db mice demonstrated an increase in genes involved in VEGF-VEGFR2 signaling and reduced oxidative stress as compared to db/db mice. These data suggest that reduced complex IV activity due to a loss of functional SCO2 might be protective in GEnCs in early DKD.
Collapse
Affiliation(s)
- Nehaben A Gujarati
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, NY
| | - Alexandra R Leonardo
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, NY
| | - Jessica M Vasquez
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, NY
| | - Yiqing Guo
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, NY
| | - Bismark O Frimpong
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, NY
| | - Elbek Fozilov
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, NY
| | - Monica P Revelo
- Department of Pathology, University of Utah, Salt Lake City, UT
| | - Ilse S Daehn
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - John C He
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Daniel Bogenhagen
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY
| | - Sandeep K Mallipattu
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, NY
- Renal Section, Northport VA Medical Center, Northport, NY
| |
Collapse
|
24
|
Huang J, Co HKC, Lee Y, Wu C, Chen S. Multistability maintains redox homeostasis in human cells. Mol Syst Biol 2021; 17:e10480. [PMID: 34612597 PMCID: PMC8493564 DOI: 10.15252/msb.202110480] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 09/11/2021] [Accepted: 09/14/2021] [Indexed: 01/22/2023] Open
Abstract
Cells metabolize nutrients through a complex metabolic and signaling network that governs redox homeostasis. At the core of this, redox regulatory network is a mutually inhibitory relationship between reduced glutathione and reactive oxygen species (ROS)-two opposing metabolites that are linked to upstream nutrient metabolic pathways (glucose, cysteine, and glutamine) and downstream feedback loops of signaling pathways (calcium and NADPH oxidase). We developed a nutrient-redox model of human cells to understand system-level properties of this network. Combining in silico modeling and ROS measurements in individual cells, we show that ROS dynamics follow a switch-like, all-or-none response upon glucose deprivation at a threshold that is approximately two orders of magnitude lower than its physiological concentration. We also confirm that this ROS switch can be irreversible and exhibits hysteresis, a hallmark of bistability. Our findings evidence that bistability modulates redox homeostasis in human cells and provide a general framework for quantitative investigations of redox regulation in humans.
Collapse
Affiliation(s)
- Jo‐Hsi Huang
- Department of Chemical and Systems BiologyStanford University School of MedicineStanfordCAUSA
| | - Hannah KC Co
- Institute of Molecular BiologyAcademia SinicaTaipeiTaiwan
- Molecular and Cell BiologyTaiwan International Graduate ProgramAcademia Sinica and Graduate Institute of Life ScienceNational Defense Medical CenterTaipeiTaiwan
| | - Yi‐Chen Lee
- Institute of Molecular BiologyAcademia SinicaTaipeiTaiwan
| | - Chia‐Chou Wu
- Institute of Molecular BiologyAcademia SinicaTaipeiTaiwan
| | - Sheng‐hong Chen
- Institute of Molecular BiologyAcademia SinicaTaipeiTaiwan
- Molecular and Cell BiologyTaiwan International Graduate ProgramAcademia Sinica and Graduate Institute of Life ScienceNational Defense Medical CenterTaipeiTaiwan
- Genome and Systems Biology Degree ProgramAcademia Sinica and National Taiwan UniversityTaipeiTaiwan
| |
Collapse
|
25
|
Weissman D, Maack C. Redox signaling in heart failure and therapeutic implications. Free Radic Biol Med 2021; 171:345-364. [PMID: 34019933 DOI: 10.1016/j.freeradbiomed.2021.05.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/17/2021] [Accepted: 05/03/2021] [Indexed: 12/13/2022]
Abstract
Heart failure is a growing health burden worldwide characterized by alterations in excitation-contraction coupling, cardiac energetic deficit and oxidative stress. While current treatments are mostly limited to antagonization of neuroendocrine activation, more recent data suggest that also targeting metabolism may provide substantial prognostic benefit. However, although in a broad spectrum of preclinical models, oxidative stress plays a causal role for the development and progression of heart failure, no treatment that targets reactive oxygen species (ROS) directly has entered the clinical arena yet. In the heart, ROS derive from various sources, such as NADPH oxidases, xanthine oxidase, uncoupled nitric oxide synthase and mitochondria. While mitochondria are the primary source of ROS in the heart, communication between different ROS sources may be relevant for physiological signalling events as well as pathologically elevated ROS that deteriorate excitation-contraction coupling, induce hypertrophy and/or trigger cell death. Here, we review the sources of ROS in the heart, the modes of pathological activation of ROS formation as well as therapeutic approaches that may target ROS specifically in mitochondria.
Collapse
Affiliation(s)
- David Weissman
- Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Würzburg, Germany
| | - Christoph Maack
- Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Würzburg, Germany; Department of Internal Medicine 1, University Clinic Würzburg, Würzburg, Germany.
| |
Collapse
|
26
|
Abstract
Oxidative stress and reactive oxygen species (ROS) are central to many physiological and pathophysiological processes. However, due to multiple technical challenges, it is hard to capture a comprehensive readout of the cell, involving both biochemical and functional status. We addressed this problem by developing a fully parallelized workflow for metabolomics (providing absolute quantities for > 100 metabolites including TCA cycle, pentose phosphate pathway, purine metabolism, glutathione metabolism, cysteine and methionine metabolism, glycolysis and gluconeogenesis) and live cell imaging microscopy. The correlative imaging strategy was applied to study morphological and metabolic adaptation of cancer cells upon short-term hydrogen peroxide (H2O2) exposure in vitro. The combination provided rich metabolic information at the endpoint of exposure together with imaging of mitochondrial effects. As a response, superoxide concentrations were elevated with a strong mitochondrial localization, and multi-parametric image analysis revealed a shift towards fragmentation. In line with this, metabolism reflected both the impaired mitochondrial function and shifts to support the first-line cellular defense and compensate for energy loss. The presented workflow combining high-end technologies demonstrates the applicability for the study of short-term oxidative stress, but it can be suitable for the in-depth study of various short-term oxidative and other cellular stress-related phenomena.
Collapse
|
27
|
Pravda J. Sepsis: Evidence-based pathogenesis and treatment. World J Crit Care Med 2021; 10:66-80. [PMID: 34316443 PMCID: PMC8291008 DOI: 10.5492/wjccm.v10.i4.66] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/13/2021] [Accepted: 06/02/2021] [Indexed: 02/06/2023] Open
Abstract
Sepsis can develop during the body’s response to a critical illness leading to multiple organ failure, irreversible shock, and death. Sepsis has been vexing health care providers for centuries due to its insidious onset, generalized metabolic dysfunction, and lack of specific therapy. A common factor underlying sepsis is the characteristic hypermetabolic response as the body ramps up every physiological system in its fight against the underlying critical illness. A hypermetabolic response requires supraphysiological amounts of energy, which is mostly supplied via oxidative phosphorylation generated ATP. A by-product of oxidative phosphorylation is hydrogen peroxide (H2O2), a toxic, membrane-permeable oxidizing agent that is produced in far greater amounts during a hypermetabolic state. Continued production of mitochondrial H2O2 can overwhelm cellular reductive (antioxidant) capacity leading to a build-up within cells and eventual diffusion into the bloodstream. H2O2 is a metabolic poison that can inhibit enzyme systems leading to organ failure, microangiopathic dysfunction, and irreversible septic shock. The toxic effects of H2O2 mirror the clinical and laboratory abnormalities observed in sepsis, and toxic levels of blood H2O2 have been reported in patients with septic shock. This review provides evidence to support a causal role for H2O2 in the pathogenesis of sepsis, and an evidence-based therapeutic intervention to reduce H2O2 levels in the body and restore redox homeostasis, which is necessary for normal organ function and vascular responsiveness.
Collapse
Affiliation(s)
- Jay Pravda
- Inflammatory Disease Research Centre, Therashock LLC, Palm Beach Gardens, FL 33410, United States
| |
Collapse
|
28
|
Brain Insulin Resistance: Focus on Insulin Receptor-Mitochondria Interactions. Life (Basel) 2021; 11:life11030262. [PMID: 33810179 PMCID: PMC8005009 DOI: 10.3390/life11030262] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/12/2021] [Accepted: 03/16/2021] [Indexed: 02/07/2023] Open
Abstract
Current hypotheses implicate insulin resistance of the brain as a pathogenic factor in the development of Alzheimer’s disease and other dementias, Parkinson’s disease, type 2 diabetes, obesity, major depression, and traumatic brain injury. A variety of genetic, developmental, and metabolic abnormalities that lead to disturbances in the insulin receptor signal transduction may underlie insulin resistance. Insulin receptor substrate proteins are generally considered to be the node in the insulin signaling system that is critically involved in the development of insulin insensitivity during metabolic stress, hyperinsulinemia, and inflammation. Emerging evidence suggests that lower activation of the insulin receptor (IR) is another common, while less discussed, mechanism of insulin resistance in the brain. This review aims to discuss causes behind the diminished activation of IR in neurons, with a focus on the functional relationship between mitochondria and IR during early insulin signaling and the related roles of oxidative stress, mitochondrial hypometabolism, and glutamate excitotoxicity in the development of IR insensitivity to insulin.
Collapse
|
29
|
Ilyich T, Kovalenia T, Lapshina E, Stępniak A, Palecz B, Zavodnik I. Thermodynamic parameters and mitochondrial effects of supramolecular complexes of quercetin with β-cyclodextrins. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2020.115184] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
30
|
Clementi ME, Maulucci G, Bianchetti G, Pizzoferrato M, Sampaolese B, Tringali G. Cytoprotective Effects of Punicalagin on Hydrogen-Peroxide-Mediated Oxidative Stress and Mitochondrial Dysfunction in Retinal Pigment Epithelium Cells. Antioxidants (Basel) 2021; 10:antiox10020192. [PMID: 33572785 PMCID: PMC7911437 DOI: 10.3390/antiox10020192] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/22/2021] [Accepted: 01/23/2021] [Indexed: 02/07/2023] Open
Abstract
The retinal pigment epithelium (RPE) is a densely pigmented, monostratified epithelium that provides metabolic and functional support to the outer segments of photoreceptors. Endogenous or exogenous oxidative stimuli determine a switch from physiological to pathological conditions, characterized by an increase of intracellular levels of reactive oxygen species (ROS). Accumulating evidence has elucidated that punicalagin (PUN), the major ellagitannin in pomegranate, is a potent antioxidant in several cell types. The present study aimed to investigate the protective effect of PUN on mitochondrial dysfunction associated with hydrogen peroxide (H2O2)-induced oxidative stress. For this purpose, we used a human RPE cell line (ARPE-19) exposed to H2O2 for 24 h. The effects of PUN pre-treatment (24 h) were examined on cell viability, mitochondrial ROS levels, mitochondrial membrane potential, and respiratory chain complexes, then finally on caspase-3 enzymatic activity. The results showed that supplementation with PUN: (a) significantly increased cell viability; (b) kept the mitochondrial membrane potential (ΔΨm) at healthy levels and limited ROS production; (c) preserved the activity of respiratory complexes; (d) reduced caspase-3 activity. In conclusion, due to its activity in helping mitochondrial functions, reducing oxidative stress, and subsequent induction of cellular apoptosis, PUN might be considered a useful nutraceutical agent in the treatment of oxidation-associated disorders of RPE.
Collapse
Affiliation(s)
- Maria Elisabetta Clementi
- Institute of Chemical Sciences and Technologies “Giulio Natta” (SCITEC)—CNR, L.go F. Vito 1, 00168 Rome, Italy;
- Correspondence: (M.E.C.); (G.T.); Tel.: +39-063-015-4215 (M.E.C.); +39-063-015-4367 (G.T.)
| | - Giuseppe Maulucci
- Biophysics Section, Neuroscience Department, Università Cattolica Del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; (G.M.); (G.B.)
- Fondazione Policlinico Universitario A, Gemelli IRCSS, 00168 Rome, Italy;
| | - Giada Bianchetti
- Biophysics Section, Neuroscience Department, Università Cattolica Del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; (G.M.); (G.B.)
- Fondazione Policlinico Universitario A, Gemelli IRCSS, 00168 Rome, Italy;
| | - Michela Pizzoferrato
- Fondazione Policlinico Universitario A, Gemelli IRCSS, 00168 Rome, Italy;
- Pharmacology Section, Department of Health Care Surveillance and Bioethics, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy
| | - Beatrice Sampaolese
- Institute of Chemical Sciences and Technologies “Giulio Natta” (SCITEC)—CNR, L.go F. Vito 1, 00168 Rome, Italy;
| | - Giuseppe Tringali
- Fondazione Policlinico Universitario A, Gemelli IRCSS, 00168 Rome, Italy;
- Pharmacology Section, Department of Health Care Surveillance and Bioethics, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy
- Correspondence: (M.E.C.); (G.T.); Tel.: +39-063-015-4215 (M.E.C.); +39-063-015-4367 (G.T.)
| |
Collapse
|
31
|
Kim SY, Zhang X, Schiattarella GG, Altamirano F, Ramos TAR, French KM, Jiang N, Szweda PA, Evers BM, May HI, Luo X, Li H, Szweda LI, Maracaja-Coutinho V, Lavandero S, Gillette TG, Hill JA. Epigenetic Reader BRD4 (Bromodomain-Containing Protein 4) Governs Nucleus-Encoded Mitochondrial Transcriptome to Regulate Cardiac Function. Circulation 2020; 142:2356-2370. [PMID: 33113340 PMCID: PMC7736324 DOI: 10.1161/circulationaha.120.047239] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 10/23/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND BET (bromodomain and extraterminal) epigenetic reader proteins, in particular BRD4 (bromodomain-containing protein 4), have emerged as potential therapeutic targets in a number of pathological conditions, including cancer and cardiovascular disease. Small-molecule BET protein inhibitors such as JQ1 have demonstrated efficacy in reversing cardiac hypertrophy and heart failure in preclinical models. Yet, genetic studies elucidating the biology of BET proteins in the heart have not been conducted to validate pharmacological findings and to unveil potential pharmacological side effects. METHODS By engineering a cardiomyocyte-specific BRD4 knockout mouse, we investigated the role of BRD4 in cardiac pathophysiology. We performed functional, transcriptomic, and mitochondrial analyses to evaluate BRD4 function in developing and mature hearts. RESULTS Unlike pharmacological inhibition, loss of BRD4 protein triggered progressive declines in myocardial function, culminating in dilated cardiomyopathy. Transcriptome analysis of BRD4 knockout mouse heart tissue identified early and specific disruption of genes essential to mitochondrial energy production and homeostasis. Functional analysis of isolated mitochondria from these hearts confirmed that BRD4 ablation triggered significant changes in mitochondrial electron transport chain protein expression and activity. Computational analysis identified candidate transcription factors participating in the BRD4-regulated transcriptome. In particular, estrogen-related receptor α, a key nuclear receptor in metabolic gene regulation, was enriched in promoters of BRD4-regulated mitochondrial genes. CONCLUSIONS In aggregate, we describe a previously unrecognized role for BRD4 in regulating cardiomyocyte mitochondrial homeostasis, observing that its function is indispensable to the maintenance of normal cardiac function.
Collapse
MESH Headings
- Animals
- Cardiomyopathy, Dilated/genetics
- Cardiomyopathy, Dilated/metabolism
- Cardiomyopathy, Dilated/pathology
- Cardiomyopathy, Dilated/physiopathology
- Cell Nucleus/genetics
- Cell Nucleus/metabolism
- Cell Nucleus/pathology
- Electron Transport Chain Complex Proteins/genetics
- Electron Transport Chain Complex Proteins/metabolism
- Energy Metabolism/genetics
- Epigenesis, Genetic
- Estrogen Receptor alpha/genetics
- Estrogen Receptor alpha/metabolism
- Gene Expression Profiling
- Heart Failure/genetics
- Heart Failure/metabolism
- Heart Failure/pathology
- Heart Failure/physiopathology
- Mice, Knockout
- Mitochondria, Heart/genetics
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/pathology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Transcriptome
- Ventricular Dysfunction, Left/genetics
- Ventricular Dysfunction, Left/metabolism
- Ventricular Dysfunction, Left/pathology
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Function, Left/genetics
Collapse
Affiliation(s)
- Soo Young Kim
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX, USA
| | - Xin Zhang
- Institute of Model Animal, Wuhan University, Wuhan, China
| | - Gabriele G. Schiattarella
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX, USA
| | - Francisco Altamirano
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX, USA
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX
| | - Thais A. R. Ramos
- Advanced Center for Chronic Disease, Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, University of Chile, Santiago, Chile
- Bioinformatics Multidisciplinary Environment, Digital Metropolis Institute, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Kristin M. French
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX, USA
| | - Nan Jiang
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX, USA
| | - Pamela A. Szweda
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX, USA
| | - Bret M. Evers
- Department of Pathology, University of Texas Southwestern, Dallas, TX, USA
| | - Herman I. May
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX, USA
| | - Xiang Luo
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX, USA
| | - Hongliang Li
- Institute of Model Animal, Wuhan University, Wuhan, China
| | - Luke I. Szweda
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX, USA
| | - Vinicius Maracaja-Coutinho
- Advanced Center for Chronic Disease, Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, University of Chile, Santiago, Chile
- Bioinformatics Multidisciplinary Environment, Digital Metropolis Institute, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Sergio Lavandero
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX, USA
- Advanced Center for Chronic Disease, Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, University of Chile, Santiago, Chile
| | - Thomas G. Gillette
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX, USA
| | - Joseph A. Hill
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX, USA
- Department of Molecular Biology, University of Texas Southwestern, Dallas, TX, USA
| |
Collapse
|
32
|
Xu W, Jain MK, Zhang L. Molecular link between circadian clocks and cardiac function: a network of core clock, slave clock, and effectors. Curr Opin Pharmacol 2020; 57:28-40. [PMID: 33189913 DOI: 10.1016/j.coph.2020.10.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/27/2020] [Accepted: 10/06/2020] [Indexed: 02/08/2023]
Abstract
The circadian rhythm has a strong influence on both cardiac physiology and disease in humans. Preclinical studies primarily using tissue-specific transgenic mouse models have contributed to our understanding of the molecular mechanism of the circadian clock in the cardiovascular system. The core clock driven by CLOCK:BMAL1 complex functions as a universal timing machinery that primarily sets the pace in all mammalian cell types. In one specific cell or tissue type, core clock may control a secondary transcriptional oscillator, conceptualized as slave clock, which confers the oscillatory expression of tissue-specific effectors. Here, we discuss a core clock-slave clock-effectors network, which links the molecular clock to cardiac function.
Collapse
Affiliation(s)
- Weiyi Xu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mukesh K Jain
- Case Cardiovascular Research Institute, Department of Medicine, Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, USA; School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Lilei Zhang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
33
|
Tatarkova Z, de Baaij JHF, Grendar M, Aschenbach JR, Racay P, Bos C, Sponder G, Hoenderop JGJ, Röntgen M, Turcanova Koprusakova M, Kolisek M. Dietary Mg 2+ Intake and the Na +/Mg 2+ Exchanger SLC41A1 Influence Components of Mitochondrial Energetics in Murine Cardiomyocytes. Int J Mol Sci 2020; 21:E8221. [PMID: 33153064 PMCID: PMC7663288 DOI: 10.3390/ijms21218221] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 10/27/2020] [Indexed: 01/02/2023] Open
Abstract
Cardiomyocytes are among the most energy-intensive cell types. Interplay between the components of cellular magnesium (Mg) homeostasis and energy metabolism in cardiomyocytes is poorly understood. We have investigated the effects of dietary Mg content and presence/functionality of the Na+/Mg2+ exchanger SLC41A1 on enzymatic functions of selected constituents of the Krebs cycle and complexes of the electron transport chain (ETC). The activities of aconitate hydratase (ACON), isocitrate dehydrogenase (ICDH), α-ketoglutarate dehydrogenase (KGDH), and ETC complexes CI-CV have been determined in vitro in mitochondria isolated from hearts of wild-type (WT) and Slc41a1-/- mice fed a diet with either normal or low Mg content. Our data demonstrate that both, the type of Mg diet and the Slc41a1 genotype largely impact on the activities of enzymes of the Krebs cycle and ETC. Moreover, a compensatory effect of Slc41a1-/- genotype on the effect of low Mg diet on activities of the tested Krebs cycle enzymes has been identified. A machine-learning analysis identified activities of ICDH, CI, CIV, and CV as common predictors of the type of Mg diet and of CII as suitable predictor of Slc41a1 genotype. Thus, our data delineate the effect of dietary Mg content and of SLC41A1 functionality on the energy-production in cardiac mitochondria.
Collapse
Affiliation(s)
- Zuzana Tatarkova
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4D, 036 01 Martin, Slovakia; (Z.T.); (P.R.)
| | - Jeroen H. F. de Baaij
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6500HB Nijmegen, The Netherlands; (J.H.F.d.B.); (C.B.); (J.G.J.H.)
| | - Marian Grendar
- Biomedical Center Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4D, 036 01 Martin, Slovakia;
| | - Jörg R. Aschenbach
- Institute of Veterinary Physiology, Freie Universität Berlin, Oertzenweg 19b, 14163 Berlin, Germany; (J.R.A.); (G.S.)
| | - Peter Racay
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4D, 036 01 Martin, Slovakia; (Z.T.); (P.R.)
- Biomedical Center Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4D, 036 01 Martin, Slovakia;
| | - Caro Bos
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6500HB Nijmegen, The Netherlands; (J.H.F.d.B.); (C.B.); (J.G.J.H.)
| | - Gerhard Sponder
- Institute of Veterinary Physiology, Freie Universität Berlin, Oertzenweg 19b, 14163 Berlin, Germany; (J.R.A.); (G.S.)
| | - Joost G. J. Hoenderop
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6500HB Nijmegen, The Netherlands; (J.H.F.d.B.); (C.B.); (J.G.J.H.)
| | - Monika Röntgen
- Leibniz Institute for Farm Animal Biology, Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany;
| | | | - Martin Kolisek
- Biomedical Center Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4D, 036 01 Martin, Slovakia;
| |
Collapse
|
34
|
Goodchild CG, DuRant SE. Fluorescent Heme Degradation Products Are Biomarkers of Oxidative Stress and Linked to Impaired Membrane Integrity in Avian Red Blood Cells. Physiol Biochem Zool 2020; 93:129-139. [PMID: 32027232 DOI: 10.1086/707920] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Oxidative stress is generally understood to be an important mediator of life-history traits, yet the specific relationships between oxidative stress and life-history traits have been difficult to describe because there is often a lack of covariation among biomarkers of oxidative stress. For instance, although oxidative damage to red blood cell (RBC) membranes can lead to pathological conditions (i.e., anemia), in some cases there is not a clear relationship between lipid oxidation and RBC membrane resistance to pro-oxidants. Alternatively, oxidative damage to hemoglobin may be an indirect mechanism contributing to RBC membrane damage. To better understand the mechanisms contributing to oxidative damage and probe new approaches to measuring oxidative stress, we used a series of in vitro and in vivo procedures in zebra finches (Taeniopygia guttata) to explore (1) whether avian RBCs exposed to a pro-oxidant generate fluorescent heme degradation products (HDPs), (2) whether HDPs interact with RBC membranes, and (3) whether HDPs are linked to impaired RBC integrity. We found that finch RBCs exposed in vitro to hydrogen peroxide produced fluorescent HDPs and HDPs associated with RBC membranes. Exposure to hydrogen peroxide also caused a reduction in hemoglobin and an increase in percent methemoglobin (a hemoglobin oxidation product), further indicating hemoglobin degradation. Moreover, HDP fluorescence correlated with impaired membrane integrity and erythrocyte osmotic fragility in vivo. This study suggests that reactive oxygen species may indirectly impair RBC membrane integrity via hemoglobin degradation products that associate with RBC membranes and that HDPs may be an inexpensive and logistically simple tool for measuring oxidative stress.
Collapse
|
35
|
Boardman NT, Migally B, Pileggi C, Parmar GS, Xuan JY, Menzies K, Harper ME. Glutaredoxin-2 and Sirtuin-3 deficiencies impair cardiac mitochondrial energetics but their effects are not additive. Biochim Biophys Acta Mol Basis Dis 2020; 1867:165982. [PMID: 33002579 DOI: 10.1016/j.bbadis.2020.165982] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 09/06/2020] [Accepted: 09/24/2020] [Indexed: 02/07/2023]
Abstract
Altered redox biology and oxidative stress have been implicated in the progression of heart failure. Glutaredoxin-2 (GRX2) is a glutathione-dependent oxidoreductase and catalyzes the reversible deglutathionylation of mitochondrial proteins. Sirtuin-3 (SIRT3) is a class III histone deacetylase and regulates lysine acetylation in mitochondria. Both GRX2 and SIRT3 are considered as key in the protection against oxidative damage in the myocardium. Knockout of either contributes to adverse heart pathologies including hypertrophy, hypertension, and cardiac dysfunction. Here, we created and characterized a GRX2 and SIRT3 double-knockout mouse model, hypothesizing that their deletions would have an additive effect on oxidative stress, and exacerbate mitochondrial function and myocardial structural remodeling. Wildtype, single-gene knockout (Sirt3-/-, Grx2-/-), and double-knockout mice (Grx2-/-/Sirt3-/-) were compared in heart weight, histology, mitochondrial respiration and H2O2 production. Overall, the hearts from Grx2-/-/Sirt3-/- mice displayed increased fibrosis and hypertrophy versus wildtype. In the Grx2-/- and the Sirt3-/- we observed changes in mitochondrial oxidative capacity, however this was associated with elevated H2O2 emission only in the Sirt3-/-. Similar changes were observed but not worsened in hearts from Grx2-/-/Sirt3-/- mice, suggesting that these changes were not additive. In human myocardium, using genetic and histopathological data from the human Genotype-Tissue Expression consortium, we confirmed that SIRT3 expression correlates inversely with heart pathology. Altogether, GRX2 and SIRT3 are important in the control of cardiac mitochondrial redox and oxidative processes, but their combined absence does not exacerbate effects, consistent with the overall conclusion that they function together in the complex redox and antioxidant systems in the heart.
Collapse
Affiliation(s)
- Neoma T Boardman
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Department of Medical Biology, Faculty of Health Sciences, UiT-Arctic University of Norway, Tromsø, Norway
| | - Baher Migally
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Chantal Pileggi
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Gaganvir S Parmar
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Jian Ying Xuan
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Keir Menzies
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Mary-Ellen Harper
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
36
|
Kedaigle AJ, Fraenkel E, Atwal RS, Wu M, Gusella JF, MacDonald ME, Kaye JA, Finkbeiner S, Mattis VB, Tom CM, Svendsen C, King AR, Chen Y, Stocksdale JT, Lim RG, Casale M, Wang PH, Thompson LM, Akimov SS, Ratovitski T, Arbez N, Ross CA. Bioenergetic deficits in Huntington's disease iPSC-derived neural cells and rescue with glycolytic metabolites. Hum Mol Genet 2020; 29:1757-1771. [PMID: 30768179 PMCID: PMC7372552 DOI: 10.1093/hmg/ddy430] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 12/09/2018] [Accepted: 12/11/2018] [Indexed: 12/14/2022] Open
Abstract
Altered cellular metabolism is believed to be an important contributor to pathogenesis of the neurodegenerative disorder Huntington's disease (HD). Research has primarily focused on mitochondrial toxicity, which can cause death of the vulnerable striatal neurons, but other aspects of metabolism have also been implicated. Most previous studies have been carried out using postmortem human brain or non-human cells. Here, we studied bioenergetics in an induced pluripotent stem cell-based model of the disease. We found decreased adenosine triphosphate (ATP) levels in HD cells compared to controls across differentiation stages and protocols. Proteomics data and multiomics network analysis revealed normal or increased levels of mitochondrial messages and proteins, but lowered expression of glycolytic enzymes. Metabolic experiments showed decreased spare glycolytic capacity in HD neurons, while maximal and spare respiratory capacities driven by oxidative phosphorylation were largely unchanged. ATP levels in HD neurons could be rescued with addition of pyruvate or late glycolytic metabolites, but not earlier glycolytic metabolites, suggesting a role for glycolytic deficits as part of the metabolic disturbance in HD neurons. Pyruvate or other related metabolic supplements could have therapeutic benefit in HD.
Collapse
Affiliation(s)
| | - Amanda J Kedaigle
- Computational and Systems Biology Graduate Program and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ernest Fraenkel
- Computational and Systems Biology Graduate Program and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ranjit S Atwal
- Center for Genomic Medicine, Massachusetts General Hospital, Simches Research Building, Cambridge Street, Boston, MA, USA
| | - Min Wu
- Center for Genomic Medicine, Massachusetts General Hospital, Simches Research Building, Cambridge Street, Boston, MA, USA
| | - James F Gusella
- Center for Genomic Medicine, Massachusetts General Hospital, Simches Research Building, Cambridge Street, Boston, MA, USA
| | - Marcy E MacDonald
- Center for Genomic Medicine, Massachusetts General Hospital, Simches Research Building, Cambridge Street, Boston, MA, USA
| | - Julia A Kaye
- Gladstone Institutes and Taube/Koret Center of Neurodegenerative Disease Research, Roddenberry Stem Cell Research Program, Departments of Neurology and Physiology, University of California, San Francisco, CA, USA
| | - Steven Finkbeiner
- Gladstone Institutes and Taube/Koret Center of Neurodegenerative Disease Research, Roddenberry Stem Cell Research Program, Departments of Neurology and Physiology, University of California, San Francisco, CA, USA
| | - Virginia B Mattis
- Board of Governors Regenerative Medicine Institute and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Colton M Tom
- Board of Governors Regenerative Medicine Institute and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Clive Svendsen
- Board of Governors Regenerative Medicine Institute and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Alvin R King
- Department of Psychiatry and Human Behavior, Department of Neurobiology and Behavior, Department of Medicine, Sue and Bill Gross Stem Cell Center and UCI MIND, University of California, Irvine, CA, USA
| | - Yumay Chen
- Department of Psychiatry and Human Behavior, Department of Neurobiology and Behavior, Department of Medicine, Sue and Bill Gross Stem Cell Center and UCI MIND, University of California, Irvine, CA, USA
| | - Jennifer T Stocksdale
- Department of Psychiatry and Human Behavior, Department of Neurobiology and Behavior, Department of Medicine, Sue and Bill Gross Stem Cell Center and UCI MIND, University of California, Irvine, CA, USA
| | - Ryan G Lim
- Department of Psychiatry and Human Behavior, Department of Neurobiology and Behavior, Department of Medicine, Sue and Bill Gross Stem Cell Center and UCI MIND, University of California, Irvine, CA, USA
| | - Malcolm Casale
- Department of Psychiatry and Human Behavior, Department of Neurobiology and Behavior, Department of Medicine, Sue and Bill Gross Stem Cell Center and UCI MIND, University of California, Irvine, CA, USA
| | - Ping H Wang
- Department of Psychiatry and Human Behavior, Department of Neurobiology and Behavior, Department of Medicine, Sue and Bill Gross Stem Cell Center and UCI MIND, University of California, Irvine, CA, USA
| | - Leslie M Thompson
- Department of Psychiatry and Human Behavior, Department of Neurobiology and Behavior, Department of Medicine, Sue and Bill Gross Stem Cell Center and UCI MIND, University of California, Irvine, CA, USA
| | - Sergey S Akimov
- Division of Neurobiology, Departments of Psychiatry, Neurology, Pharmacology, and Neuroscience, Johns Hopkins University School of Medicine, North Wolfe Street, Baltimore, MA, USA
| | - Tamara Ratovitski
- Division of Neurobiology, Departments of Psychiatry, Neurology, Pharmacology, and Neuroscience, Johns Hopkins University School of Medicine, North Wolfe Street, Baltimore, MA, USA
| | - Nicolas Arbez
- Division of Neurobiology, Departments of Psychiatry, Neurology, Pharmacology, and Neuroscience, Johns Hopkins University School of Medicine, North Wolfe Street, Baltimore, MA, USA
| | - Christopher A Ross
- Division of Neurobiology, Departments of Psychiatry, Neurology, Pharmacology, and Neuroscience, Johns Hopkins University School of Medicine, North Wolfe Street, Baltimore, MA, USA
| |
Collapse
|
37
|
Yang G, Liu Z, Zhang R, Tian X, Chen J, Han G, Liu B, Han X, Fu Y, Hu Z, Zhang Z. A Multi‐responsive Fluorescent Probe Reveals Mitochondrial Nucleoprotein Dynamics with Reactive Oxygen Species Regulation through Super‐resolution Imaging. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202005959] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Guanqing Yang
- School of Chemistry and Chemical Engineering and Institute of Physical Science and Information Technology Anhui University Hefei Anhui 230601 China
| | - Zhengjie Liu
- School of Chemistry and Chemical Engineering and Institute of Physical Science and Information Technology Anhui University Hefei Anhui 230601 China
| | - Ruilong Zhang
- School of Chemistry and Chemical Engineering and Institute of Physical Science and Information Technology Anhui University Hefei Anhui 230601 China
- Key Laboratory of Structure and Functional Regulation of Hybrid Materials (Anhui University) Ministry of Education Hefei Anhui 230601 China
| | - Xiaohe Tian
- School of Chemistry and Chemical Engineering and Institute of Physical Science and Information Technology Anhui University Hefei Anhui 230601 China
| | - Juan Chen
- School of Chemistry and Chemical Engineering and Institute of Physical Science and Information Technology Anhui University Hefei Anhui 230601 China
| | - Guangmei Han
- School of Chemistry and Chemical Engineering and Institute of Physical Science and Information Technology Anhui University Hefei Anhui 230601 China
| | - Bianhua Liu
- Institute of Intelligent Machines Chinese Academy of Sciences Hefei Anhui 230031 China
| | - Xinya Han
- School of Chemistry and Chemical Engineering Anhui University of Technology Ma'anshan Anhui 243032 China
| | - Yao Fu
- Department of Chemistry University of Science and Technology of China Hefei Anhui 230026 China
| | - Zhangjun Hu
- Department of Physics, Chemistry and Biology Linköping University Linköping 58183 Sweden
| | - Zhongping Zhang
- School of Chemistry and Chemical Engineering and Institute of Physical Science and Information Technology Anhui University Hefei Anhui 230601 China
- Key Laboratory of Structure and Functional Regulation of Hybrid Materials (Anhui University) Ministry of Education Hefei Anhui 230601 China
- Institute of Intelligent Machines Chinese Academy of Sciences Hefei Anhui 230031 China
| |
Collapse
|
38
|
Defective endoplasmic reticulum-mitochondria contacts and bioenergetics in SEPN1-related myopathy. Cell Death Differ 2020; 28:123-138. [PMID: 32661288 PMCID: PMC7853070 DOI: 10.1038/s41418-020-0587-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 06/11/2020] [Accepted: 06/25/2020] [Indexed: 01/19/2023] Open
Abstract
SEPN1-related myopathy (SEPN1-RM) is a muscle disorder due to mutations of the SEPN1 gene, which is characterized by muscle weakness and fatigue leading to scoliosis and life-threatening respiratory failure. Core lesions, focal areas of mitochondria depletion in skeletal muscle fibers, are the most common histopathological lesion. SEPN1-RM underlying mechanisms and the precise role of SEPN1 in muscle remained incompletely understood, hindering the development of biomarkers and therapies for this untreatable disease. To investigate the pathophysiological pathways in SEPN1-RM, we performed metabolic studies, calcium and ATP measurements, super-resolution and electron microscopy on in vivo and in vitro models of SEPN1 deficiency as well as muscle biopsies from SEPN1-RM patients. Mouse models of SEPN1 deficiency showed marked alterations in mitochondrial physiology and energy metabolism, suggesting that SEPN1 controls mitochondrial bioenergetics. Moreover, we found that SEPN1 was enriched at the mitochondria-associated membranes (MAM), and was needed for calcium transients between ER and mitochondria, as well as for the integrity of ER-mitochondria contacts. Consistently, loss of SEPN1 in patients was associated with alterations in body composition which correlated with the severity of muscle weakness, and with impaired ER-mitochondria contacts and low ATP levels. Our results indicate a role of SEPN1 as a novel MAM protein involved in mitochondrial bioenergetics. They also identify a systemic bioenergetic component in SEPN1-RM and establish mitochondria as a novel therapeutic target. This role of SEPN1 contributes to explain the fatigue and core lesions in skeletal muscle as well as the body composition abnormalities identified as part of the SEPN1-RM phenotype. Finally, these results point out to an unrecognized interplay between mitochondrial bioenergetics and ER homeostasis in skeletal muscle. They could therefore pave the way to the identification of biomarkers and therapeutic drugs for SEPN1-RM and for other disorders in which muscle ER-mitochondria cross-talk are impaired.
Collapse
|
39
|
Yang G, Liu Z, Zhang R, Tian X, Chen J, Han G, Liu B, Han X, Fu Y, Hu Z, Zhang Z. A Multi-responsive Fluorescent Probe Reveals Mitochondrial Nucleoprotein Dynamics with Reactive Oxygen Species Regulation through Super-resolution Imaging. Angew Chem Int Ed Engl 2020; 59:16154-16160. [PMID: 32573047 DOI: 10.1002/anie.202005959] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 06/11/2020] [Indexed: 11/06/2022]
Abstract
Understanding the biomolecular interactions in a specific organelle has been a long-standing challenge because it requires super-resolution imaging to resolve the spatial locations and dynamic interactions of multiple biomacromolecules. Two key difficulties are the scarcity of suitable probes for super-resolution nanoscopy and the complications that arise from the use of multiple probes. Herein, we report a quinolinium derivative probe that is selectively enriched in mitochondria and switches on in three different fluorescence modes in response to hydrogen peroxide (H2 O2 ), proteins, and nucleic acids, enabling the visualization of mitochondrial nucleoprotein dynamics. STED nanoscopy reveals that the proteins localize at mitochondrial cristae and largely fuse with nucleic acids to form nucleoproteins, whereas increasing H2 O2 level leads to disassociation of nucleic acid-protein complexes.
Collapse
Affiliation(s)
- Guanqing Yang
- School of Chemistry and Chemical Engineering and Institute of Physical Science and Information Technology, Anhui University, Hefei, Anhui, 230601, China
| | - Zhengjie Liu
- School of Chemistry and Chemical Engineering and Institute of Physical Science and Information Technology, Anhui University, Hefei, Anhui, 230601, China
| | - Ruilong Zhang
- School of Chemistry and Chemical Engineering and Institute of Physical Science and Information Technology, Anhui University, Hefei, Anhui, 230601, China.,Key Laboratory of Structure and Functional Regulation of Hybrid Materials (Anhui University), Ministry of Education, Hefei, Anhui, 230601, China
| | - Xiaohe Tian
- School of Chemistry and Chemical Engineering and Institute of Physical Science and Information Technology, Anhui University, Hefei, Anhui, 230601, China
| | - Juan Chen
- School of Chemistry and Chemical Engineering and Institute of Physical Science and Information Technology, Anhui University, Hefei, Anhui, 230601, China
| | - Guangmei Han
- School of Chemistry and Chemical Engineering and Institute of Physical Science and Information Technology, Anhui University, Hefei, Anhui, 230601, China
| | - Bianhua Liu
- Institute of Intelligent Machines, Chinese Academy of Sciences, Hefei, Anhui, 230031, China
| | - Xinya Han
- School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, Anhui, 243032, China
| | - Yao Fu
- Department of Chemistry, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Zhangjun Hu
- Department of Physics, Chemistry and Biology, Linköping University, Linköping, 58183, Sweden
| | - Zhongping Zhang
- School of Chemistry and Chemical Engineering and Institute of Physical Science and Information Technology, Anhui University, Hefei, Anhui, 230601, China.,Key Laboratory of Structure and Functional Regulation of Hybrid Materials (Anhui University), Ministry of Education, Hefei, Anhui, 230601, China.,Institute of Intelligent Machines, Chinese Academy of Sciences, Hefei, Anhui, 230031, China
| |
Collapse
|
40
|
Pandey RP, Kumar S, Ahmad S, Vibhuti A, Raj VS, Verma AK, Sharma P, Leal E. Use Chou's 5-steps rule to evaluate protective efficacy induced by antigenic proteins of Mycobacterium tuberculosis encapsulated in chitosan nanoparticles. Life Sci 2020; 256:117961. [PMID: 32534039 DOI: 10.1016/j.lfs.2020.117961] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The study focuses on whether antigenic proteins encapsulated in biopolymeric nanoparticles can augment protective efficacy. Chitosan nanoparticles (ChN) were prepared by ionic gelation method and Culture Filtrate Proteins (CFP) - CFP-10 and CFP-21 of Mycobacterium tuberculosis (Mtb) were encapsulated in ChN. The binding efficiency of nanoparticles with CFP-10 and CFP-21 proteins was confirmed by UV-Spectrophotometer. The efficacy of nanoparticles-encapsulated antigenic proteins administered intraperitoneal against Mtb aerosol infection was evaluated in Balb/c mice. Protection study was done by bacterial counts [CFU]. CFP-10 and CFP-21 proteins primed cells demonstrated a Th1 bias T cell response in an ex vivo assay. ChN-CFP10 and ChN-CFP21 nanoparticles have both protective and therapeutic potential against Mtb. In the group of mice immunized with CHN-CFP-10 the number of colonies reduced significantly from day 15 to day 60. ChN-CFP-21 showed maximum protection in ChN-CFP-21 immunized mice. ChN-CFP-10 and ChN-CFP-21 clearly showed enhanced protection against Mtb.
Collapse
Affiliation(s)
- Ramendra Pati Pandey
- Centre for Drug Design Discovery and Development (C4D), SRM University, Delhi-NCR, Rajiv Gandhi Education City, Sonepat 131 029, Haryana, India
| | - Santosh Kumar
- ICGEB (International Centre For Genetic Engineering And Biotechnology), New Delhi 110067, India
| | - Saheem Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Ha'il, Ha'il, 55476, Saudi Arabia
| | - Arpana Vibhuti
- Centre for Drug Design Discovery and Development (C4D), SRM University, Delhi-NCR, Rajiv Gandhi Education City, Sonepat 131 029, Haryana, India.
| | - V Samuel Raj
- Centre for Drug Design Discovery and Development (C4D), SRM University, Delhi-NCR, Rajiv Gandhi Education City, Sonepat 131 029, Haryana, India.
| | - Anita Kamra Verma
- Nano-Biotech Laboratory, Department of Zoology, Kirori Mal College, University of Delhi, New Delhi 110003, India
| | - Pawan Sharma
- ICGEB (International Centre For Genetic Engineering And Biotechnology), New Delhi 110067, India
| | - Elcio Leal
- Institute of Biological Sciences, Federal University of Para, Para 66075-000, Brazil.
| |
Collapse
|
41
|
Garabadu D, Agrawal N, Sharma A, Sharma S. Mitochondrial metabolism: a common link between neuroinflammation and neurodegeneration. Behav Pharmacol 2020; 30:642-652. [PMID: 31625975 DOI: 10.1097/fbp.0000000000000505] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Neurodegenerative disorders have been considered as a growing health concern for decades. Increasing risk of neurodegenerative disorders creates a socioeconomic burden to both patients and care givers. Mitochondria are organelle that are involved in both neuroinflammation and neurodegeneration. There are few reports on the effect of mitochondrial metabolism on the progress of neurodegeneration and neuroinflammation. Therefore, the present review summarizes the potential contribution of mitochondrial metabolic pathways in the pathogenesis of neuroinflammation and neurodegeneration. Mitochondrial pyruvate metabolism plays a critical role in the pathogenesis of neurodegenerative disorders such as Parkinson's disease and Alzheimer's disease. However, there its potential contribution in other neurodegenerative disorders is as yet unproven. The mitochondrial pyruvate carrier and pyruvate dehydrogenase can modulate mitochondrial pyruvate metabolism to attenuate neuroinflammation and neurodegeneration. Further, it has been observed that the mitochondrial citric acid cycle can regulate the pathogenesis of neuroinflammation and neurodegeneration. Additional research should be undertaken to target tricarboxylic acid cycle enzymes to minimize the progress of neuroinflammation and neurodegeneration. It has also been observed that the mitochondrial urea cycle can potentially contribute to the progression of neurodegenerative disorders. Therefore, targeting this pathway may control the mitochondrial dysfunction-induced neuroinflammation and neurodegeneration. Furthermore, the mitochondrial malate-aspartate shuttle could be another target to control mitochondrial dysfunction-induced neuroinflammation and neurodegeneration in neurodegenerative disorders.
Collapse
Affiliation(s)
- Debapriya Garabadu
- Division of Pharmacology, Institute of Pharmaceutical Research, GLA University, Mathura, India
| | | | | | | |
Collapse
|
42
|
Pott DM, Vallarino JG, Osorio S. Metabolite Changes during Postharvest Storage: Effects on Fruit Quality Traits. Metabolites 2020; 10:metabo10050187. [PMID: 32397309 PMCID: PMC7281412 DOI: 10.3390/metabo10050187] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 05/06/2020] [Indexed: 12/13/2022] Open
Abstract
Metabolic changes occurring in ripe or senescent fruits during postharvest storage lead to a general deterioration in quality attributes, including decreased flavor and ‘off-aroma’ compound generation. As a consequence, measures to reduce economic losses have to be taken by the fruit industry and have mostly consisted of storage at cold temperatures and the use of controlled atmospheres or ripening inhibitors. However, the biochemical pathways and molecular mechanisms underlying fruit senescence in commercial storage conditions are still poorly understood. In this sense, metabolomic platforms, enabling the profiling of key metabolites responsible for organoleptic and health-promoting traits, such as volatiles, sugars, acids, polyphenols and carotenoids, can be a powerful tool for further understanding the biochemical basis of postharvest physiology and have the potential to play a critical role in the identification of the pathways affected by fruit senescence. Here, we provide an overview of the metabolic changes during postharvest storage, with special attention to key metabolites related to fruit quality. The potential use of metabolomic approaches to yield metabolic markers useful for chemical phenotyping or even storage and marketing decisions is highlighted.
Collapse
Affiliation(s)
| | - José G. Vallarino
- Correspondence: (J.G.V.); (S.O.); Tel.: +34-952134271 (J.G.V. & S.O.)
| | - Sonia Osorio
- Correspondence: (J.G.V.); (S.O.); Tel.: +34-952134271 (J.G.V. & S.O.)
| |
Collapse
|
43
|
Pravda J. Hydrogen peroxide and disease: towards a unified system of pathogenesis and therapeutics. Mol Med 2020; 26:41. [PMID: 32380940 PMCID: PMC7204068 DOI: 10.1186/s10020-020-00165-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 04/07/2020] [Indexed: 02/07/2023] Open
Abstract
Although the immune response has a prominent role in the pathophysiology of ulcerative colitis, sepsis, and systemic lupus erythematosus, a primary immune causation has not been established to explain the pathogenesis of these diseases. However, studies have reported significantly elevated levels of colonic epithelial hydrogen peroxide (a known colitic agent) in ulcerative colitis prior to the appearance of colitis. And patients with sepsis are reported to have toxic levels of blood hydrogen peroxide, whose pathologic effects mirror the laboratory and clinical abnormalities observed in sepsis. More recently, evidence supports a causal role for cellular hydrogen peroxide (a potent apoptotic agent) in the enhanced apoptosis believed to be the driving force behind auto-antigenic exposure and chronic immune activation in systemic lupus erythematosus. The different biological properties of hydrogen peroxide exert distinct pathologic effects depending on the site of accumulation within the body resulting in a unique disease patho-phenotype. On a cellular level, the build-up of hydrogen peroxide triggers apoptosis resulting in systemic lupus erythematosus, on a tissue level (colonic epithelium) excess hydrogen peroxide leads to inflammation and ulcerative colitis, and on a systemic level the pathologic effects of toxic concentrations of blood hydrogen peroxide result in bioenergetic failure and microangiopathic dysfunction leading to multiple organ failure and circulatory shock, characteristic of advanced sepsis. The aim of this paper is to provide a unified evidence-based common causal role for hydrogen peroxide in the pathogenesis of ulcerative colitis, sepsis, and systemic lupus erythematosus. Based on this new theory of pathogenesis, a novel evidence-based treatment of sepsis is also discussed.
Collapse
|
44
|
Guo X, Park JE, Gallart-Palau X, Sze SK. Oxidative Damage to the TCA Cycle Enzyme MDH1 Dysregulates Bioenergetic Enzymatic Activity in the Aged Murine Brain. J Proteome Res 2020; 19:1706-1717. [DOI: 10.1021/acs.jproteome.9b00861] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Xue Guo
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Jung Eun Park
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Xavier Gallart-Palau
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Siu Kwan Sze
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| |
Collapse
|
45
|
Sunil B, Rajsheel P, Aswani V, Bapatla RB, Talla SK, Raghavendra AS. Photosynthesis is sensitive to nitric oxide and respiration sensitive to hydrogen peroxide: Studies with pea mesophyll protoplasts. JOURNAL OF PLANT PHYSIOLOGY 2020; 246-247:153133. [PMID: 32065920 DOI: 10.1016/j.jplph.2020.153133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 01/27/2020] [Accepted: 02/01/2020] [Indexed: 05/27/2023]
Abstract
Reports on the effect of nitric oxide (NO) or reactive oxygen species (ROS) on photosynthesis and respiration in leaf tissues are intriguing; therefore, the effects of exogenous addition of sodium nitroprusside (SNP, releases NO) or H2O2 on the photosynthetic O2 evolution and respiratory O2 uptake by mesophyll protoplasts in pea (Pisum sativum) were evaluated in the present study. Low concentrations of SNP or H2O2 were used to minimize nonspecific effects. The effects of NO or H2O2 on respiration and photosynthesis were different. The presence of NO decreased the rate of photosynthesis but caused a marginal stimulation of dark respiration. Conversely, externally administered H2O2 drastically decreased the rate of respiration but only slightly decreased photosynthesis. The PS I activity was more sensitive to NO than PS II. On the other hand, 100 μM H2O2 had no effect on the photochemical reactions of either PS I or PS II. The sensitivity of photosynthesis to antimycin A or SHAM (reflecting the interplay between chloroplasts and mitochondria) was not affected by NO. By contrast, H2O2 markedly decreased the sensitivity of photosynthesis to antimycin A and SHAM. It can be concluded that chloroplasts are the primary targets of NO, while mitochondria are the primary targets of ROS in plant cells. We propose that H2O2 can be an important signal to modulate the crosstalk between chloroplasts and mitochondria.
Collapse
Affiliation(s)
- Bobba Sunil
- Department of Plant Sciences, School of Life Sciences, University of Hyderabad, Hyderabad 500046, India
| | - Pidakala Rajsheel
- Department of Plant Sciences, School of Life Sciences, University of Hyderabad, Hyderabad 500046, India
| | - Vetcha Aswani
- Department of Plant Sciences, School of Life Sciences, University of Hyderabad, Hyderabad 500046, India
| | - Ramesh B Bapatla
- Department of Plant Sciences, School of Life Sciences, University of Hyderabad, Hyderabad 500046, India
| | - Sai K Talla
- Department of Plant Sciences, School of Life Sciences, University of Hyderabad, Hyderabad 500046, India
| | - Agepati S Raghavendra
- Department of Plant Sciences, School of Life Sciences, University of Hyderabad, Hyderabad 500046, India.
| |
Collapse
|
46
|
Liao PC, Franco-Iborra S, Yang Y, Pon LA. Live cell imaging of mitochondrial redox state in mammalian cells and yeast. Methods Cell Biol 2020; 155:295-319. [PMID: 32183963 DOI: 10.1016/bs.mcb.2019.11.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
The redox state of mitochondria is determined by the levels of reducing and oxidizing species in the organelle, which reflects mitochondrial metabolic activity and overall fitness. Mitochondria are also the primary endogenous source of reactive oxygen species (ROS). This chapter describes methods to measure the mitochondrial superoxide levels and the redox state of the organelle in mammalian cells and yeast. We describe the use of dihydroethidium (DHE) and MitoSOX (a derivative of dihydroethidium bound to a lipophilic cation) to detect mitochondrial superoxide in yeast and mammalian cells, respectively. We also describe the use of genetically encoded fluorescent biosensors for quantitative analysis of mitochondrial NADPH levels (iNap) in mammalian cells and mitochondrial redox state (mito-roGFP) in yeast.
Collapse
Affiliation(s)
- Pin-Chao Liao
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States
| | - Sandra Franco-Iborra
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States
| | - Yi Yang
- Synthetic Biology and Biotechnology Laboratory, Key State Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, Shanghai, China
| | - Liza A Pon
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States.
| |
Collapse
|
47
|
Shin D, Lee J, You JH, Kim D, Roh JL. Dihydrolipoamide dehydrogenase regulates cystine deprivation-induced ferroptosis in head and neck cancer. Redox Biol 2020; 30:101418. [PMID: 31931284 PMCID: PMC6957841 DOI: 10.1016/j.redox.2019.101418] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/16/2019] [Accepted: 12/29/2019] [Indexed: 12/19/2022] Open
Abstract
Ferroptosis is a new form of regulated cell death driven by iron-dependent lipid peroxidation. Glutaminolysis and tricarboxylic acid cycle are involved in ferroptosis, but the underlying metabolic process remains unclear. We examined the role of dihydrolipoamide dehydrogenase (DLD) in ferroptosis induction in head and neck cancer (HNC). The effects of cystine deprivation or sulfasalazine treatment and of DLD gene silencing/overexpression were tested on HNC cell lines and mouse tumor xenograft models. These effects were analyzed with regard to cell death, lipid reactive oxygen species (ROS) and mitochondrial iron production, mitochondrial membrane potential, mRNA/protein expression, and α-ketoglutarate dehydrogenase (KGDH)/succinate/aconitase activities. Cystine deprivation induced ferroptosis via glutaminolysis. Cystine deprivation or import inhibition using sulfasalazine induced cancer cell death and increased lipid ROS and mitochondrial iron levels, which had been significantly decreased by short-interfering RNA (siRNA) or short hairpin RNA (shRNA) targeting DLD (P < 0.01) but not by dihydrolipoyl succinyltransferase. The same results were noted in an in vivo mouse model transplanted with vector or shDLD-transduced HN9 cells. After cystine deprivation or sulfasalazine treatment, mitochondrial membrane potential, mitochondrial free iron level, KGDH activity, and succinate content significantly increased (P < 0.001), which had been blocked by DLD siRNA or shRNA and were consequently rescued by resistant DLD cDNA. Cystine deprivation caused iron starvation response and mitochondrial iron accumulation for Fenton reaction and ferroptosis. Our data suggest a close association of DLD with cystine deprivation- or import inhibition-induced ferroptosis.
Collapse
Affiliation(s)
- Daiha Shin
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Republic of Korea; Western Seoul Center, Korea Basic Science Institute, Seoul, Republic of Korea
| | - Jaewang Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Republic of Korea
| | - Ji Hyeon You
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Republic of Korea
| | - Doyeon Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Republic of Korea
| | - Jong-Lyel Roh
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Republic of Korea.
| |
Collapse
|
48
|
Dridi H, Yehya M, Barsotti R, Reiken S, Angebault C, Jung B, Jaber S, Marks AR, Lacampagne A, Matecki S. Mitochondrial oxidative stress induces leaky ryanodine receptor during mechanical ventilation. Free Radic Biol Med 2020; 146:383-391. [PMID: 31756525 DOI: 10.1016/j.freeradbiomed.2019.11.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 10/28/2019] [Accepted: 11/15/2019] [Indexed: 12/28/2022]
Abstract
RATIONALE Ventilator-induced diaphragm dysfunction (VIDD) increases morbidity and mortality in critical care patients. Although VIDD has been associated with mitochondrial oxidative stress and calcium homeostasis impairment, the underling mechanisms are still unknown. We hypothesized that diaphragmatic mitochondrial oxidative stress causes remodeling of the ryanodine receptor (RyR1)/calcium release channel, contributing to sarcoplasmic reticulum (SR) Ca2+ leak, proteolysis and VIDD. METHOD In mice diaphragms mechanically ventilated for short (6 h) and long (12 h) period, we assessed mitochondrial ROS production, mitochondrial aconitase activity as a marker of mitochondrial oxidative stress, RyR1 remodeling and function, Ca2+ dependent proteolysis, TGFβ1 and STAT3 pathway, muscle fibers cross-sectional area, and diaphragm specific force production, with or without the mitochondrial targeted anti-oxidant peptide d-Arg-2', 6'-dimethyltyrosine-Lys-Phe-NH2 (SS31). MEASUREMENTS AND MAIN RESULTS 6 h of mechanical ventilation (MV) resulted in increased mitochondrial ROS production, reduction of mitochondrial aconitase activity, increased oxidation, S-nitrosylation, S-glutathionylation and Ser-2844 phosphorylation of RyR1, depletion of stabilizing subunit calstabin1 from RyR1, increased SR Ca2+ leak. Preventing mROS production by SS31 treatment does not affect the TGFβ1 and STAT3 activation, which suggests that mitochondrial oxidative stress is a downstream pathway to TGFβ1 and STAT3, early involved in VIDD. This is further supported by the fact that SS-31 rescue all the other described cellular events and diaphragm contractile dysfunction induced by MV, while SS20, an analog of SS31 lacking antioxidant properties, failed to prevent these cellular events and the contractile dysfunction. Similar results were found in ventilated for 12 h. Moreover, SS31 treatment prevented calpain1 activity and diaphragm atrophy observed after 12 h of MV. This study emphasizes that mitochondrial oxidative stress during 6 h-MV contributes to SR Ca2+ leak via RyR1 remodeling, and diaphragm weakness, while longer periods of MV (12 h) were also associated with increased Ca2+-dependent proteolysis and diaphragm atrophy.
Collapse
Affiliation(s)
- Haikel Dridi
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology Columbia University College of Physicians and Surgeons, New York, USA
| | - Mohamad Yehya
- PhyMedExp, Montpellier University, INSERM, CNRS, CHRU Montpellier, 34295, Montpellier, France
| | - Robert Barsotti
- Department of Biomedical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, PA, USA
| | - Steven Reiken
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology Columbia University College of Physicians and Surgeons, New York, USA
| | - Claire Angebault
- PhyMedExp, Montpellier University, INSERM, CNRS, CHRU Montpellier, 34295, Montpellier, France
| | - Boris Jung
- PhyMedExp, Montpellier University, INSERM, CNRS, CHRU Montpellier, 34295, Montpellier, France; Medical Intensive Care Unit, Montpellier University and Montpellier University Health Care Center, 34295, Montpellier, France
| | - Samir Jaber
- PhyMedExp, Montpellier University, INSERM, CNRS, CHRU Montpellier, 34295, Montpellier, France; St Eloi Department of Anesthesiology and Critical Care Medicine, Montpellier University and Montpellier University Health Care Center, 34295, Montpellier, France
| | - Andrew R Marks
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology Columbia University College of Physicians and Surgeons, New York, USA
| | - Alain Lacampagne
- PhyMedExp, Montpellier University, INSERM, CNRS, CHRU Montpellier, 34295, Montpellier, France.
| | - Stephan Matecki
- PhyMedExp, Montpellier University, INSERM, CNRS, CHRU Montpellier, 34295, Montpellier, France; Arnaud de Villeneuve Physiological Department, Montpellier University and Montpellier University Health Care Center, 34295, Montpellier, France.
| |
Collapse
|
49
|
Zhang X, Lee MD, Wilson C, McCarron JG. Hydrogen peroxide depolarizes mitochondria and inhibits IP 3-evoked Ca 2+ release in the endothelium of intact arteries. Cell Calcium 2019; 84:102108. [PMID: 31715384 PMCID: PMC6891240 DOI: 10.1016/j.ceca.2019.102108] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 08/30/2019] [Accepted: 10/30/2019] [Indexed: 12/30/2022]
Abstract
H2O2 is produced by several cell processes including mitochondria and may act as an intracellular messenger and cell-cell signalling molecule. Spontaneous local Ca2+ signals and IP3-evoked Ca2+ increases were inhibited by H2O2. H2O2 suppression of IP3-evoked Ca2+ signalling may be mediated by mitochondria via a decrease in the mitochondrial membrane potential. H2O2-induced mitochondrial depolarization and inhibition of IP3-evoked Ca2+ release, may protect mitochondria from Ca2+ overload during IP3-linked Ca2+ signals.
Hydrogen peroxide (H2O2) is a mitochondrial-derived reactive oxygen species (ROS) that regulates vascular signalling transduction, vasocontraction and vasodilation. Although the physiological role of ROS in endothelial cells is acknowledged, the mechanisms underlying H2O2 regulation of signalling in native, fully-differentiated endothelial cells is unresolved. In the present study, the effects of H2O2 on Ca2+ signalling were investigated in the endothelium of intact rat mesenteric arteries. Spontaneous local Ca2+ signals and acetylcholine evoked Ca2+ increases were inhibited by H2O2. H2O2 inhibition of acetylcholine-evoked Ca2+ signals was reversed by catalase. H2O2 exerts its inhibition on the IP3 receptor as Ca2+ release evoked by photolysis of caged IP3 was supressed by H2O2. H2O2 suppression of IP3-evoked Ca2+ signalling may be mediated by mitochondria. H2O2 depolarized mitochondria membrane potential. Acetylcholine-evoked Ca2+ release was inhibited by depolarisation of the mitochondrial membrane potential by the uncoupler carbonyl cyanide 3-chlorophenylhydrazone (CCCP) or complex 1 inhibitor, rotenone. We propose that the suppression of IP3-evoked Ca2+ release by H2O2 arises from the decrease in mitochondrial membrane potential. These results suggest that mitochondria may protect themselves against Ca2+ overload during IP3-linked Ca2+ signals by a H2O2 mediated negative feedback depolarization of the organelle and inhibition of IP3-evoked Ca2+ release.
Collapse
Affiliation(s)
- Xun Zhang
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Matthew D Lee
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Calum Wilson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - John G McCarron
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK.
| |
Collapse
|
50
|
Lu M, Lan Y, Xiao J, Song M, Chen C, Liang C, Huang Q, Cao Y, Ho CT. Capsaicin Ameliorates the Redox Imbalance and Glucose Metabolism Disorder in an Insulin-Resistance Model via Circadian Clock-Related Mechanisms. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:10089-10096. [PMID: 31423784 DOI: 10.1021/acs.jafc.9b04016] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Circadian rhythms are closely associated with metabolic homeostasis. Metabolic disorders can be alleviated by many bioactive components through controlling of clock gene expressions. Capsaicin has been demonstrated with many beneficial effects including anti-obesity and anti-insulin resistance activities, yet whether the rhythmic expression of circadian clock genes are involved in the regulation of redox imbalance and glucose metabolism disorder by capsaicin remains unclear. In this work, the insulin resistance was induced in HepG2 cells by treatment of glucosamine. Glucose uptake levels, reactive oxygen species, H2O2 production, and mitochondrial membrane potential (MMP) were measured with/without capsaicin cotreatment. The mRNA and protein expressions of core circadian clock genes were evaluated by RT-qPCR and western blot analysis. Our study revealed that circadian misalignment could be ameliorated by capsaicin. The glucosamine-induced cellular redox imbalance and glucose metabolism disorder were ameliorated by capsaicin in a Bmal1-dependent manner.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Qingrong Huang
- Department of Food Science , Rutgers University , New Brunswick , New Jersey 08901 , United States
| | | | - Chi-Tang Ho
- Department of Food Science , Rutgers University , New Brunswick , New Jersey 08901 , United States
| |
Collapse
|