1
|
Fitisemanu FM, Padilla-Benavides T. Emerging perspectives of copper-mediated transcriptional regulation in mammalian cell development. Metallomics 2024; 16:mfae046. [PMID: 39375833 PMCID: PMC11503025 DOI: 10.1093/mtomcs/mfae046] [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: 06/23/2024] [Accepted: 10/04/2024] [Indexed: 10/09/2024]
Abstract
Copper (Cu) is a vital micronutrient necessary for proper development and function of mammalian cells and tissues. Cu mediates the function of redox active enzymes that facilitate metabolic processes and signaling pathways. Cu levels are tightly regulated by a network of Cu-binding transporters, chaperones, and small molecule ligands. Extensive research has focused on the mammalian Cu homeostasis (cuprostasis) network and pathologies, which result from mutations and perturbations. There are roles for Cu-binding proteins as transcription factors (Cu-TFs) and regulators that mediate metal homeostasis through the activation or repression of genes associated with Cu handling. Emerging evidence suggests that Cu and some Cu-TFs may be involved in the regulation of targets related to development-expanding the biological roles of Cu-binding proteins. Cu and Cu-TFs are implicated in embryonic and tissue-specific development alongside the mediation of the cellular response to oxidative stress and hypoxia. Cu-TFs are also involved in the regulation of targets implicated in neurological disorders, providing new biomarkers and therapeutic targets for diseases such as Parkinson's disease, prion disease, and Friedreich's ataxia. This review provides a critical analysis of the current understanding of the role of Cu and cuproproteins in transcriptional regulation.
Collapse
|
2
|
Lin H, Wang L, Jiang X, Wang J. Glutathione dynamics in subcellular compartments and implications for drug development. Curr Opin Chem Biol 2024; 81:102505. [PMID: 39053236 DOI: 10.1016/j.cbpa.2024.102505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/30/2024] [Accepted: 07/08/2024] [Indexed: 07/27/2024]
Abstract
Glutathione (GSH) is a pivotal tripeptide antioxidant essential for maintaining cellular redox homeostasis and regulating diverse cellular processes. Subcellular compartmentalization of GSH underscores its multifaceted roles across various organelles including the cytosol, mitochondria, endoplasmic reticulum, and nucleus, each exhibiting distinct regulatory mechanisms. Perturbations in GSH dynamics contribute to pathophysiological conditions, emphasizing the clinical significance of understanding its intricate regulation. This review consolidates current knowledge on subcellular GSH dynamics, highlighting its implications in drug development, particularly in covalent drug design and antitumor strategies targeting intracellular GSH levels. Challenges and future directions in deciphering subcellular GSH dynamics are discussed, advocating for innovative methodologies to advance our comprehension and facilitate the development of precise therapeutic interventions based on GSH modulation.
Collapse
Affiliation(s)
- Hanfeng Lin
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX 77030, USA; Center for NextGen Therapeutics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lingfei Wang
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xiqian Jiang
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Jin Wang
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX 77030, USA; Center for NextGen Therapeutics, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
3
|
Wang J, Choi WG, Nguyen NK, Liu D, Kim SH, Lim D, Hwang BK, Jwa NS. Cytoplasmic Ca 2+ influx mediates iron- and reactive oxygen species-dependent ferroptotic cell death in rice immunity. FRONTIERS IN PLANT SCIENCE 2024; 15:1339559. [PMID: 38756966 PMCID: PMC11096502 DOI: 10.3389/fpls.2024.1339559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 04/17/2024] [Indexed: 05/18/2024]
Abstract
Iron- and reactive oxygen species (ROS)-dependent ferroptosis occurs in plant cells. Ca2+ acts as a conserved key mediator to control plant immune responses. Here, we report a novel role of cytoplasmic Ca2+ influx regulating ferroptotic cell death in rice immunity using pharmacological approaches. High Ca2+ influx triggered iron-dependent ROS accumulation, lipid peroxidation, and subsequent hypersensitive response (HR) cell death in rice (Oryza sativa). During Magnaporthe oryzae infection, 14 different Ca2+ influx regulators altered Ca2+, ROS and Fe2+ accumulation, glutathione reductase (GR) expression, glutathione (GSH) depletion and lipid peroxidation, leading to ferroptotic cell death in rice. High Ca2+ levels inhibited the reduction of glutathione isulphide (GSSG) to GSH in vitro. Ca2+ chelation by ethylene glycol-bis (2-aminoethylether)-N, N, N', N'-tetra-acetic acid (EGTA) suppressed apoplastic Ca2+ influx in rice leaf sheaths during infection. Blocking apoplastic Ca2+ influx into the cytoplasm by Ca2+ chelation effectively suppressed Ca2+-mediated iron-dependent ROS accumulation and ferroptotic cell death. By contrast, acibenzolar-S-methyl (ASM), a plant defense activator, significantly enhanced Ca2+ influx, as well as ROS and iron accumulation to trigger ferroptotic cell death in rice. The cytoplasmic Ca2+ influx through calcium-permeable cation channels, including the putative resistosomes, could mediate iron- and ROS-dependent ferroptotic cell death under reduced GR expression levels in rice immune responses.
Collapse
Affiliation(s)
- Juan Wang
- Division of Integrative Bioscience and Biotechnology, College of Life Sciences, Sejong University, Seoul, Republic of Korea
| | - Won-Gyu Choi
- Department of Biochemistry and Molecular Biology, University of Nevada, Reno, NV, United States
| | - Nam Khoa Nguyen
- Division of Integrative Bioscience and Biotechnology, College of Life Sciences, Sejong University, Seoul, Republic of Korea
| | - Dongping Liu
- Division of Integrative Bioscience and Biotechnology, College of Life Sciences, Sejong University, Seoul, Republic of Korea
| | - Su-Hwa Kim
- Department of Biochemistry and Molecular Biology, University of Nevada, Reno, NV, United States
| | - Dongyeol Lim
- Department of Chemistry, College of Natural Sciences, Sejong University, Seoul, Republic of Korea
| | - Byung Kook Hwang
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Nam-Soo Jwa
- Division of Integrative Bioscience and Biotechnology, College of Life Sciences, Sejong University, Seoul, Republic of Korea
| |
Collapse
|
4
|
Rothman DL, Behar KL, Dienel GA. Mechanistic stoichiometric relationship between the rates of neurotransmission and neuronal glucose oxidation: Reevaluation of and alternatives to the pseudo-malate-aspartate shuttle model. J Neurochem 2024; 168:555-591. [PMID: 36089566 DOI: 10.1111/jnc.15619] [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: 11/12/2021] [Revised: 04/08/2022] [Accepted: 04/15/2022] [Indexed: 11/26/2022]
Abstract
The ~1:1 stoichiometry between the rates of neuronal glucose oxidation (CMRglc-ox-N) and glutamate (Glu)/γ-aminobutyric acid (GABA)-glutamine (Gln) neurotransmitter (NT) cycling between neurons and astrocytes (VNTcycle) has been firmly established. However, the mechanistic basis for this relationship is not fully understood, and this knowledge is critical for the interpretation of metabolic and brain imaging studies in normal and diseased brain. The pseudo-malate-aspartate shuttle (pseudo-MAS) model established the requirement for glycolytic metabolism in cultured glutamatergic neurons to produce NADH that is shuttled into mitochondria to support conversion of extracellular Gln (i.e., astrocyte-derived Gln in vivo) into vesicular neurotransmitter Glu. The evaluation of this model revealed that it could explain half of the 1:1 stoichiometry and it has limitations. Modifications of the pseudo-MAS model were, therefore, devised to address major knowledge gaps, that is, submitochondrial glutaminase location, identities of mitochondrial carriers for Gln and other model components, alternative mechanisms to transaminate α-ketoglutarate to form Glu and shuttle glutamine-derived ammonia while maintaining mass balance. All modified models had a similar 0.5 to 1.0 predicted mechanistic stoichiometry between VNTcycle and the rate of glucose oxidation. Based on studies of brain β-hydroxybutyrate oxidation, about half of CMRglc-ox-N may be linked to glutamatergic neurotransmission and localized in pre-synaptic structures that use pseudo-MAS type mechanisms for Glu-Gln cycling. In contrast, neuronal compartments that do not participate in transmitter cycling may use the MAS to sustain glucose oxidation. The evaluation of subcellular compartmentation of neuronal glucose metabolism in vivo is a critically important topic for future studies to understand glutamatergic and GABAergic neurotransmission.
Collapse
Affiliation(s)
- Douglas L Rothman
- Magnetic Resonance Research Center and Departments of Radiology and Biomedical Engineering, Yale University, New Haven, Connecticut, USA
| | - Kevin L Behar
- Magnetic Resonance Research Center and Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Gerald A Dienel
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Department of Cell Biology and Physiology, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| |
Collapse
|
5
|
Chen TH, Wang HC, Chang CJ, Lee SY. Mitochondrial Glutathione in Cellular Redox Homeostasis and Disease Manifestation. Int J Mol Sci 2024; 25:1314. [PMID: 38279310 PMCID: PMC10816320 DOI: 10.3390/ijms25021314] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/15/2024] [Accepted: 01/19/2024] [Indexed: 01/28/2024] Open
Abstract
Mitochondria are critical for providing energy to maintain cell viability. Oxidative phosphorylation involves the transfer of electrons from energy substrates to oxygen to produce adenosine triphosphate. Mitochondria also regulate cell proliferation, metastasis, and deterioration. The flow of electrons in the mitochondrial respiratory chain generates reactive oxygen species (ROS), which are harmful to cells at high levels. Oxidative stress caused by ROS accumulation has been associated with an increased risk of cancer, and cardiovascular and liver diseases. Glutathione (GSH) is an abundant cellular antioxidant that is primarily synthesized in the cytoplasm and delivered to the mitochondria. Mitochondrial glutathione (mGSH) metabolizes hydrogen peroxide within the mitochondria. A long-term imbalance in the ratio of mitochondrial ROS to mGSH can cause cell dysfunction, apoptosis, necroptosis, and ferroptosis, which may lead to disease. This study aimed to review the physiological functions, anabolism, variations in organ tissue accumulation, and delivery of GSH to the mitochondria and the relationships between mGSH levels, the GSH/GSH disulfide (GSSG) ratio, programmed cell death, and ferroptosis. We also discuss diseases caused by mGSH deficiency and related therapeutics.
Collapse
Affiliation(s)
- Tsung-Hsien Chen
- Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 60002, Taiwan;
| | - Hsiang-Chen Wang
- Department of Mechanical Engineering, National Chung Cheng University, Chiayi 62102, Taiwan;
| | - Chia-Jung Chang
- Division of Critical Care Medicine, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 60002, Taiwan
| | - Shih-Yu Lee
- Division of Critical Care Medicine, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 60002, Taiwan
| |
Collapse
|
6
|
Vašková J, Kočan L, Vaško L, Perjési P. Glutathione-Related Enzymes and Proteins: A Review. Molecules 2023; 28:molecules28031447. [PMID: 36771108 PMCID: PMC9919958 DOI: 10.3390/molecules28031447] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
The tripeptide glutathione is found in all eukaryotic cells, and due to the compartmentalization of biochemical processes, its synthesis takes place exclusively in the cytosol. At the same time, its functions depend on its transport to/from organelles and interorgan transport, in which the liver plays a central role. Glutathione is determined as a marker of the redox state in many diseases, aging processes, and cell death resulting from its properties and reactivity. It also uses other enzymes and proteins, which enables it to engage and regulate various cell functions. This paper approximates the role of these systems in redox and detoxification reactions such as conjugation reactions of glutathione-S-transferases, glyoxylases, reduction of peroxides through thiol peroxidases (glutathione peroxidases, peroxiredoxins) and thiol-disulfide exchange reactions catalyzed by glutaredoxins.
Collapse
Affiliation(s)
- Janka Vašková
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, 040 11 Košice, Slovakia
- Correspondence: (J.V.); (P.P.); Tel.: +42-155-234-3232 (J.V.)
| | - Ladislav Kočan
- Clinic of Anaesthesiology and Intensive Care Medicine, East Slovak Institute of Cardiovascular Disease, 040 11 Košice, Slovakia
| | - Ladislav Vaško
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, 040 11 Košice, Slovakia
| | - Pál Perjési
- Institute of Pharmaceutical Chemistry, University of Pécs, 7600 Pécs, Hungary
- Correspondence: (J.V.); (P.P.); Tel.: +42-155-234-3232 (J.V.)
| |
Collapse
|
7
|
Linseman DA, Winter AN, Wilkins HM. The 2-Oxoglutarate Carrier Is S-Nitrosylated in the Spinal Cord of G93A Mutant hSOD1 Mice Resulting in Disruption of Mitochondrial Glutathione Transport. Biomedicines 2022; 11:61. [PMID: 36672568 PMCID: PMC9855976 DOI: 10.3390/biomedicines11010061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/15/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Mitochondrial oxidative stress and dysfunction are strongly implicated in the pathogenesis of amyotrophic lateral sclerosis (ALS). Glutathione (GSH) is an endogenous antioxidant that exists as distinct cytosolic and mitochondrial pools. The status of the mitochondrial GSH pool is reliant on transport from the cytosol through the 2-oxoglutarate carrier (OGC), an inner membrane anion carrier. We have previously reported that the outer mitochondrial membrane protein, Bcl-2, directly binds GSH and is a key regulator of OGC-dependent mitochondrial GSH transport. Here, we show that G93A mutant SOD1 (Cu, Zn-superoxide dismutase) reduces the binding of GSH to Bcl-2 and disrupts mitochondrial GSH uptake in vitro. In the G93A mutant hSOD1 mouse model of ALS, mitochondrial GSH is significantly depleted in spinal cord of end-stage mice. Finally, we show that OGC is heavily S-nitrosylated in the spinal cord of end-stage mice and consequently, the GSH uptake capacity of spinal cord mitochondria isolated from these mutant mice is significantly diminished. Collectively, these findings suggest that spinal cord GSH depletion, particularly at the level of the mitochondria, plays a significant role in ALS pathogenesis induced by mutant SOD1. Furthermore, the depletion of mitochondrial GSH in the G93A mutant hSOD1 mouse model may be caused by the S-nitrosylation of OGC and the capacity of mutant SOD1 to disrupt the Bcl-2/GSH interaction, resulting in a disruption of mitochondrial GSH transport.
Collapse
Affiliation(s)
- Daniel A. Linseman
- Department of Biological Sciences, Knoebel Institute for Healthy Aging, University of Denver, Denver, CO 80208, USA
| | | | - Heather M. Wilkins
- Alzheimer’s Disease Research Center, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
8
|
Chronic Ouabain Prevents Radiation-Induced Reduction in the α2 Na,K-ATPase Function in the Rat Diaphragm Muscle. Int J Mol Sci 2022; 23:ijms231810921. [PMID: 36142836 PMCID: PMC9505176 DOI: 10.3390/ijms231810921] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/11/2022] [Accepted: 09/15/2022] [Indexed: 11/29/2022] Open
Abstract
The damaging effect of ionizing radiation (IR) on skeletal muscle Na,K-ATPase is an open field of research. Considering a therapeutic potential of ouabain, a specific ligand of the Na,K-ATPase, we tested its ability to protect against the IR-induced disturbances of Na,K-ATPase function in rat diaphragm muscle that co-expresses the α1 and α2 isozymes of this protein. Male Wistar rats (n = 26) were subjected to 6-day injections of vehicle (0.9% NaCl) or ouabain (1 µg/kg/day). On the fourth day of injections, rats were exposed to one-time total-body X-ray irradiation (10 Gy), or a sham irradiation. The isolated muscles were studied 72 h post-irradiation. IR decreased the electrogenic contribution of the α2 Na,K-ATPase without affecting its protein content, thereby causing sarcolemma depolarization. IR increased serum concentrations of ouabain, IL-6, and corticosterone, decreased lipid peroxidation, and changed cellular redox status. Chronic ouabain administration prevented IR-induced depolarization and loss of the α2 Na,K-ATPase electrogenic contribution without changing its protein content. This was accompanied with an elevation of ouabain concentration in circulation and with the lack of IR-induced suppression of lipid peroxidation. Given the crucial role of Na,K-ATPase in skeletal muscle performance, these findings may have therapeutic implications as countermeasures for IR-induced muscle pathology.
Collapse
|
9
|
Rohatgi N, Ghoshdastider U, Baruah P, Kulshrestha T, Skanderup AJ. A pan-cancer metabolic atlas of the tumor microenvironment. Cell Rep 2022; 39:110800. [PMID: 35545044 DOI: 10.1016/j.celrep.2022.110800] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/14/2021] [Accepted: 04/20/2022] [Indexed: 11/03/2022] Open
Abstract
Tumors are heterogeneous cellular environments with entwined metabolic dependencies. Here, we use a tumor transcriptome deconvolution approach to profile the metabolic states of cancer and non-cancer (stromal) cells in bulk tumors of 20 solid tumor types. We identify metabolic genes and processes recurrently altered in cancer cells across tumor types, highlighting pan-cancer upregulation of deoxythymidine triphosphate (dTTP) production. In contrast, the tryptophan catabolism rate-limiting enzymes IDO1 and TDO2 are highly overexpressed in stroma, raising the hypothesis that kynurenine-mediated suppression of antitumor immunity may be predominantly constrained by the stroma. Oxidative phosphorylation is the most upregulated metabolic process in cancer cells compared to both stromal cells and a large atlas of cancer cell lines, suggesting that the Warburg effect may be less pronounced in cancer cells in vivo. Overall, our analysis highlights fundamental differences in metabolic states of cancer and stromal cells inside tumors and establishes a pan-cancer resource to interrogate tumor metabolism.
Collapse
Affiliation(s)
- Neha Rohatgi
- Genome Institute of Singapore, Agency for Science Technology and Research, Singapore, Singapore
| | - Umesh Ghoshdastider
- Genome Institute of Singapore, Agency for Science Technology and Research, Singapore, Singapore
| | - Probhonjon Baruah
- Genome Institute of Singapore, Agency for Science Technology and Research, Singapore, Singapore
| | - Tanmay Kulshrestha
- Genome Institute of Singapore, Agency for Science Technology and Research, Singapore, Singapore
| | | |
Collapse
|
10
|
Nguyen NT, Nguyen TT, Park KS. Oxidative Stress Related to Plasmalemmal and Mitochondrial Phosphate Transporters in Vascular Calcification. Antioxidants (Basel) 2022; 11:antiox11030494. [PMID: 35326144 PMCID: PMC8944874 DOI: 10.3390/antiox11030494] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 02/26/2022] [Accepted: 02/28/2022] [Indexed: 12/04/2022] Open
Abstract
Inorganic phosphate (Pi) is essential for maintaining cellular function but excess of Pi leads to serious complications, including vascular calcification. Accumulating evidence suggests that oxidative stress contributes to the pathogenic progression of calcific changes. However, the molecular mechanism underlying Pi-induced reactive oxygen species (ROS) generation and its detrimental consequences remain unclear. Type III Na+-dependent Pi cotransporter, PiT-1/-2, play a significant role in Pi uptake of vascular smooth muscle cells. Pi influx via PiT-1/-2 increases the abundance of PiT-1/-2 and depolarization-activated Ca2+ entry due to its electrogenic properties, which may lead to Ca2+ and Pi overload and oxidative stress. At least four mitochondrial Pi transporters are suggested, among which the phosphate carrier (PiC) is known to be mainly involved in mitochondrial Pi uptake. Pi transport via PiC may induce hyperpolarization and superoxide generation, which may lead to mitochondrial dysfunction and endoplasmic reticulum stress, together with generation of cytosolic ROS. Increase in net influx of Ca2+ and Pi and their accumulation in the cytosol and mitochondrial matrix synergistically increases oxidative stress and osteogenic differentiation, which could be prevented by suppressing either Ca2+ or Pi overload. Therapeutic strategies targeting plasmalemmal and mitochondrial Pi transports can protect against Pi-induced oxidative stress and vascular calcification.
Collapse
Affiliation(s)
- Nhung Thi Nguyen
- Department of Physiology, Wonju College of Medicine, Yonsei University, Wonju 26426, Korea;
- Mitohormesis Research Center, Wonju College of Medicine, Yonsei University, Wonju 26426, Korea
- Medical Doctor Program, College of Health Sciences, VinUniversity, Hanoi 12406, Vietnam
| | - Tuyet Thi Nguyen
- Department of Physiology, Wonju College of Medicine, Yonsei University, Wonju 26426, Korea;
- Internal Medicine Residency Program, College of Health Sciences, VinUniversity, Hanoi 12406, Vietnam
- Correspondence: (T.T.N.); (K.-S.P.); Tel.: +84-247-108-9779 (T.T.N.); +82-33-741-0294 (K.-S.P.)
| | - Kyu-Sang Park
- Department of Physiology, Wonju College of Medicine, Yonsei University, Wonju 26426, Korea;
- Mitohormesis Research Center, Wonju College of Medicine, Yonsei University, Wonju 26426, Korea
- Correspondence: (T.T.N.); (K.-S.P.); Tel.: +84-247-108-9779 (T.T.N.); +82-33-741-0294 (K.-S.P.)
| |
Collapse
|
11
|
Fiorani M, De Matteis R, Canonico B, Blandino G, Mazzoli A, Montanari M, Guidarelli A, Cantoni O. Temporal correlation of morphological and biochemical changes with the recruitment of different mechanisms of reactive oxygen species formation during human SW872 cell adipogenic differentiation. Biofactors 2021; 47:837-851. [PMID: 34260117 PMCID: PMC8597007 DOI: 10.1002/biof.1769] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 06/29/2021] [Indexed: 12/22/2022]
Abstract
Human SW872 preadipocyte conversion to mature adipocytes is associated with time-dependent changes in differentiation markers' expression and with morphological changes accompanied by the accumulation of lipid droplets (LDs) as well as by increased mitochondriogenesis and mitochondrial membrane potential. Under identical conditions, the formation of reactive oxygen species (ROS) revealed with a general probe was significant at days 3 and 10 of differentiation and bearly detectable at day 6. NADPH oxidase (NOX)-2 activity determined with an immunocytochemical approach followed a very similar pattern. There was no evidence of mitochondrial ROS (mROS), as detected with a selective fluorescence probe, at days 3 and 6, possibly due to the triggering of the Nrf-2 antioxidant response. mROS were instead clearly detected at day 10, concomitantly with the accumulation of very large LDs, oxidation of both cardiolipin and thioredoxin 2, and decreased mitochondrial glutathione. In conclusion, the morphological and biochemical changes of differentiating SW872 cells are accompanied by the discontinuous formation of ROS derived from NOX-2, increasingly implicated in adipogenesis and adipose tissue dysfunction. In addition, mROS formation was significant only in the late phase of differentiation and was associated with mitochondrial dysfunction.
Collapse
Affiliation(s)
- Mara Fiorani
- Department of Biomolecular SciencesUniversity of Urbino Carlo BoUrbinoItaly
| | - Rita De Matteis
- Department of Biomolecular SciencesUniversity of Urbino Carlo BoUrbinoItaly
| | - Barbara Canonico
- Department of Biomolecular SciencesUniversity of Urbino Carlo BoUrbinoItaly
| | - Giulia Blandino
- Department of Biomolecular SciencesUniversity of Urbino Carlo BoUrbinoItaly
| | - Alessandro Mazzoli
- Department of Biomolecular SciencesUniversity of Urbino Carlo BoUrbinoItaly
| | - Mariele Montanari
- Department of Biomolecular SciencesUniversity of Urbino Carlo BoUrbinoItaly
| | - Andrea Guidarelli
- Department of Biomolecular SciencesUniversity of Urbino Carlo BoUrbinoItaly
| | - Orazio Cantoni
- Department of Biomolecular SciencesUniversity of Urbino Carlo BoUrbinoItaly
| |
Collapse
|
12
|
Diverse Roles of Mitochondria in Renal Injury from Environmental Toxicants and Therapeutic Drugs. Int J Mol Sci 2021; 22:ijms22084172. [PMID: 33920653 PMCID: PMC8073222 DOI: 10.3390/ijms22084172] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/05/2021] [Accepted: 04/14/2021] [Indexed: 01/19/2023] Open
Abstract
Mitochondria are well-known to function as the primary sites of ATP synthesis in most mammalian cells, including the renal proximal tubule. Other functions have also been associated with different mitochondrial activities, including the regulation of redox status and the initiation of mitophagy and apoptosis. Mechanisms for the membrane transport of glutathione (GSH) and various GSH-derived metabolites across the mitochondrial inner membrane of renal proximal tubular cells are critical determinants of these functions and may serve as pharmacological targets for potential therapeutic approaches. Specific interactions of reactive intermediates, derived from drug metabolism, with molecular components in mitochondria have been identified as early steps in diverse forms of chemically-induced nephrotoxicity. Applying this key observation, we developed a novel hypothesis regarding the identification of early, sensitive, and specific biomarkers of exposure to nephrotoxicants. The underlying concept is that upon exposure to a diverse array of environmental contaminants, as well as therapeutic drugs whose efficacy is limited by nephrotoxicity, renal mitochondria will release both high- and low-molecular-weight components into the urine or the extracellular medium in an in vitro model. The detection of these components may then serve as indicators of exposure before irreversible renal injury has occurred.
Collapse
|
13
|
Patten DA, McGuirk S, Anilkumar U, Antoun G, Gandhi K, Parmar G, Iqbal MA, Wong J, Richardson RB, St-Pierre J, Slack RS, Harper ME. Altered mitochondrial fusion drives defensive glutathione synthesis in cells able to switch to glycolytic ATP production. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1868:118854. [PMID: 32926942 DOI: 10.1016/j.bbamcr.2020.118854] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 09/08/2020] [Accepted: 09/09/2020] [Indexed: 01/05/2023]
Abstract
Mitochondria are highly dynamic organelles. Alterations in mitochondrial dynamics are causal or are linked to numerous neurodegenerative, neuromuscular, and metabolic diseases. It is generally thought that cells with altered mitochondrial structure are prone to mitochondrial dysfunction, increased reactive oxygen species generation and widespread oxidative damage. The objective of the current study was to investigate the relationship between mitochondrial dynamics and the master cellular antioxidant, glutathione (GSH). We reveal that mouse embryonic fibroblasts (MEFs) lacking the mitochondrial fusion machinery display elevated levels of GSH, which limits oxidative damage. Moreover, targeted metabolomics and 13C isotopic labeling experiments demonstrate that cells lacking the inner membrane fusion GTPase OPA1 undergo widespread metabolic remodeling altering the balance of citric acid cycle intermediates and ultimately favoring GSH synthesis. Interestingly, the GSH precursor and antioxidant n-acetylcysteine did not increase GSH levels in OPA1 KO cells, suggesting that cysteine is not limiting for GSH production in this context. Post-mitotic neurons were unable to increase GSH production in the absence of OPA1. Finally, the ability to use glycolysis for ATP production was a requirement for GSH accumulation following OPA1 deletion. Thus, our results demonstrate a novel role for mitochondrial fusion in the regulation of GSH synthesis, and suggest that cysteine availability is not limiting for GSH synthesis in conditions of mitochondrial fragmentation. These findings provide a possible explanation for the heightened sensitivity of certain cell types to alterations in mitochondrial dynamics.
Collapse
Affiliation(s)
- David A Patten
- University of Ottawa, Faculty of Medicine, Department of Biochemistry Microbiology and Immunology, Ottawa, ON, K1H 8M5, Canada; Canadian Nuclear Laboratories (CNL), Radiobiology and Health Branch, Chalk River Laboratories, Chalk River, ON K0J 1J0, Canada; Ottawa Institute of Systems Biology, Ottawa K1H 8M5, Canada
| | - Shawn McGuirk
- McGill University, Department of Biochemistry, Montreal, QC H3G 1Y6, Canada
| | - Ujval Anilkumar
- University of Ottawa, Faculty of Medicine, Department of Cellular and Molecular Medicine, Ottawa, ON K1H 8M5, Canada
| | - Ghadi Antoun
- University of Ottawa, Faculty of Medicine, Department of Biochemistry Microbiology and Immunology, Ottawa, ON, K1H 8M5, Canada
| | - Karan Gandhi
- University of Ottawa, Faculty of Medicine, Department of Biochemistry Microbiology and Immunology, Ottawa, ON, K1H 8M5, Canada
| | - Gaganvir Parmar
- University of Ottawa, Faculty of Medicine, Department of Biochemistry Microbiology and Immunology, Ottawa, ON, K1H 8M5, Canada; Ottawa Institute of Systems Biology, Ottawa K1H 8M5, Canada
| | - Mohamed Ariff Iqbal
- University of Ottawa, Faculty of Medicine, Department of Cellular and Molecular Medicine, Ottawa, ON K1H 8M5, Canada
| | - Jacob Wong
- University of Ottawa, Faculty of Medicine, Department of Cellular and Molecular Medicine, Ottawa, ON K1H 8M5, Canada
| | - Richard B Richardson
- Canadian Nuclear Laboratories (CNL), Radiobiology and Health Branch, Chalk River Laboratories, Chalk River, ON K0J 1J0, Canada; McGill Medical Physics Unit, Cedars Cancer Centre - Glen Site, Montreal, QC H4A 3J1, Canada
| | - Julie St-Pierre
- University of Ottawa, Faculty of Medicine, Department of Biochemistry Microbiology and Immunology, Ottawa, ON, K1H 8M5, Canada; Ottawa Institute of Systems Biology, Ottawa K1H 8M5, Canada; McGill University, Department of Biochemistry, Montreal, QC H3G 1Y6, Canada
| | - Ruth S Slack
- University of Ottawa, Faculty of Medicine, Department of Cellular and Molecular Medicine, Ottawa, ON K1H 8M5, Canada.
| | - Mary-Ellen Harper
- University of Ottawa, Faculty of Medicine, Department of Biochemistry Microbiology and Immunology, Ottawa, ON, K1H 8M5, Canada; Ottawa Institute of Systems Biology, Ottawa K1H 8M5, Canada.
| |
Collapse
|
14
|
Mills RH, Wozniak JM, Vrbanac A, Campeau A, Chassaing B, Gewirtz A, Knight R, Gonzalez DJ. Organ-level protein networks as a reference for the host effects of the microbiome. Genome Res 2020; 30:276-286. [PMID: 31992612 PMCID: PMC7050531 DOI: 10.1101/gr.256875.119] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 01/21/2020] [Indexed: 02/07/2023]
Abstract
Connections between the microbiome and health are rapidly emerging in a wide range of diseases. However, a detailed mechanistic understanding of how different microbial communities are influencing their hosts is often lacking. One method researchers have used to understand these effects are germ-free (GF) mouse models. Differences found within the organ systems of these model organisms may highlight generalizable mechanisms that microbiome dysbioses have throughout the host. Here, we applied multiplexed, quantitative proteomics on the brains, spleens, hearts, small intestines, and colons of conventionally raised and GF mice, identifying associations to colonization state in over 7000 proteins. Highly ranked associations were constructed into protein-protein interaction networks and visualized onto an interactive 3D mouse model for user-guided exploration. These results act as a resource for microbiome researchers hoping to identify host effects of microbiome colonization on a given organ of interest. Our results include validation of previously reported effects in xenobiotic metabolism, the innate immune system, and glutamate-associated proteins while simultaneously providing organism-wide context. We highlight organism-wide differences in mitochondrial proteins including consistent increases in NNT, a mitochondrial protein with essential roles in influencing levels of NADH and NADPH, in all analyzed organs of conventional mice. Our networks also reveal new associations for further exploration, including protease responses in the spleen, high-density lipoproteins in the heart, and glutamatergic signaling in the brain. In total, our study provides a resource for microbiome researchers through detailed tables and visualization of the protein-level effects of microbial colonization on several organ systems.
Collapse
Affiliation(s)
- Robert H Mills
- Department of Pharmacology, University of California, San Diego, California 92093, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, California 92093, USA
- Department of Pediatrics, and Department of Computer Science and Engineering, University of California, San Diego, California 92093, USA
- Center for Microbiome Innovation, University of California, San Diego, California 92093, USA
| | - Jacob M Wozniak
- Department of Pharmacology, University of California, San Diego, California 92093, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, California 92093, USA
| | - Alison Vrbanac
- Department of Pediatrics, and Department of Computer Science and Engineering, University of California, San Diego, California 92093, USA
| | - Anaamika Campeau
- Department of Pharmacology, University of California, San Diego, California 92093, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, California 92093, USA
| | - Benoit Chassaing
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia 30303, USA
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30303, USA
- INSERM, U1016, 75014 Paris, France
- Université de Paris, 75006 Paris, France
| | - Andrew Gewirtz
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia 30303, USA
| | - Rob Knight
- Department of Pediatrics, and Department of Computer Science and Engineering, University of California, San Diego, California 92093, USA
- Center for Microbiome Innovation, University of California, San Diego, California 92093, USA
| | - David J Gonzalez
- Department of Pharmacology, University of California, San Diego, California 92093, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, California 92093, USA
- Center for Microbiome Innovation, University of California, San Diego, California 92093, USA
| |
Collapse
|
15
|
Wang M, Lau LI, Sreekumar PG, Spee C, Hinton DR, Sadda SR, Kannan R. Characterization and Regulation of Carrier Proteins of Mitochondrial Glutathione Uptake in Human Retinal Pigment Epithelium Cells. Invest Ophthalmol Vis Sci 2019; 60:500-516. [PMID: 30707752 PMCID: PMC6360990 DOI: 10.1167/iovs.18-25686] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Purpose To characterize two mitochondrial membrane transporters 2-oxoglutarate (OGC) and dicarboxylate (DIC) in human RPE (hRPE) and to elucidate their role in the regulation of mitochondrial glutathione (mGSH) uptake and cell death in oxidative stress. Methods The localization of OGC and DIC proteins in confluent hRPE, polarized hRPE monolayers and mouse retina was assessed by immunoblotting and confocal microscopy. Time- and dose-dependent expression of the two carriers were determined after treatment of hRPE with H2O2, phenyl succinate (PS), and butyl malonate (BM), respectively, for 24 hours. The effect of inhibition of OGC and DIC on apoptosis (TUNEL), mGSH, and mtDNA was determined. Silencing of OGC by siRNA knockdown on RPE cell death was studied. Kinetics of caspase 3/7 activation with OGC and DIC inhibitors and effect of cotreatment with glutathione monoethyl ester (GSH-MEE) was determined using the IncuCyte live cell imaging. Results OGC and DIC are expressed in hRPE mitochondria and exhibited a time- and dose-dependent decrease with stress. Pharmacologic inhibition caused a decrease in OGC and DIC in mitochondria without changes in mtDNA and resulted in increased apoptosis and mGSH depletion. GSH-MEE prevented apoptosis through restoration of mGSH. OGC siRNA exacerbated apoptotic cell death in stressed RPE which was inhibited by increased mGSH from GSH-MEE cotreatment. Conclusions Characterization and mechanism of action of two carrier proteins of mGSH uptake in RPE are reported. Regulation of OGC and DIC will be of value in devising therapeutic strategies for retinal disorders such as AMD.
Collapse
Affiliation(s)
- Mo Wang
- Department of Ophthalmology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
- The Stephen J. Ryan Initiative for Macular Research (RIMR), Doheny Eye Institute, Los Angeles, California, United States
| | - Lin-Ing Lau
- The Stephen J. Ryan Initiative for Macular Research (RIMR), Doheny Eye Institute, Los Angeles, California, United States
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Parameswaran G Sreekumar
- The Stephen J. Ryan Initiative for Macular Research (RIMR), Doheny Eye Institute, Los Angeles, California, United States
| | - Christine Spee
- Department of Pathology and Ophthalmology, USC Roski Eye Institute, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States
| | - David R Hinton
- Department of Pathology and Ophthalmology, USC Roski Eye Institute, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States
| | - Srinivas R Sadda
- The Stephen J. Ryan Initiative for Macular Research (RIMR), Doheny Eye Institute, Los Angeles, California, United States
| | - Ram Kannan
- The Stephen J. Ryan Initiative for Macular Research (RIMR), Doheny Eye Institute, Los Angeles, California, United States
| |
Collapse
|
16
|
Punzi G, Porcelli V, Ruggiu M, Hossain MF, Menga A, Scarcia P, Castegna A, Gorgoglione R, Pierri CL, Laera L, Lasorsa FM, Paradies E, Pisano I, Marobbio CMT, Lamantea E, Ghezzi D, Tiranti V, Giannattasio S, Donati MA, Guerrini R, Palmieri L, Palmieri F, De Grassi A. SLC25A10 biallelic mutations in intractable epileptic encephalopathy with complex I deficiency. Hum Mol Genet 2019; 27:499-504. [PMID: 29211846 DOI: 10.1093/hmg/ddx419] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 11/29/2017] [Indexed: 01/10/2023] Open
Abstract
Mitochondrial diseases are a plethora of inherited neuromuscular disorders sharing defects in mitochondrial respiration, but largely different from one another for genetic basis and pathogenic mechanism. Whole exome sequencing was performed in a familiar trio (trio-WES) with a child affected by severe epileptic encephalopathy associated with respiratory complex I deficiency and mitochondrial DNA depletion in skeletal muscle. By trio-WES we identified biallelic mutations in SLC25A10, a nuclear gene encoding a member of the mitochondrial carrier family. Genetic and functional analyses conducted on patient fibroblasts showed that SLC25A10 mutations are associated with reduction in RNA quantity and aberrant RNA splicing, and to absence of SLC25A10 protein and its transporting function. The yeast SLC25A10 ortholog knockout strain showed defects in mitochondrial respiration and mitochondrial DNA content, similarly to what observed in the patient skeletal muscle, and growth susceptibility to oxidative stress. Albeit patient fibroblasts were depleted in the main antioxidant molecules NADPH and glutathione, transport assays demonstrated that SLC25A10 is unable to transport glutathione. Here, we report the first recessive mutations of SLC25A10 associated to an inherited severe mitochondrial neurodegenerative disorder. We propose that SLC25A10 loss-of-function causes pathological disarrangements in respiratory-demanding conditions and oxidative stress vulnerability.
Collapse
Affiliation(s)
- Giuseppe Punzi
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, 70125 Bari, Italy
| | - Vito Porcelli
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, 70125 Bari, Italy
| | - Matteo Ruggiu
- Department of Biological Sciences, St. John's University, Queens, NY 11439, USA
| | - Md F Hossain
- Department of Biological Sciences, St. John's University, Queens, NY 11439, USA
| | - Alessio Menga
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, 70125 Bari, Italy
| | - Pasquale Scarcia
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, 70125 Bari, Italy
| | - Alessandra Castegna
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, 70125 Bari, Italy
| | - Ruggiero Gorgoglione
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, 70125 Bari, Italy
| | - Ciro L Pierri
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, 70125 Bari, Italy
| | - Luna Laera
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, 70125 Bari, Italy.,Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, CNR, 70125 Bari, Italy
| | - Francesco M Lasorsa
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, CNR, 70125 Bari, Italy
| | - Eleonora Paradies
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, CNR, 70125 Bari, Italy
| | - Isabella Pisano
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, 70125 Bari, Italy
| | - Carlo M T Marobbio
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, 70125 Bari, Italy
| | - Eleonora Lamantea
- Unit of Molecular Neurogenetics, Foundation IRCCS Institute of Neurology "C. Besta", 20126 Milan, Italy
| | - Daniele Ghezzi
- Unit of Molecular Neurogenetics, Foundation IRCCS Institute of Neurology "C. Besta", 20126 Milan, Italy.,Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy
| | - Valeria Tiranti
- Unit of Molecular Neurogenetics, Foundation IRCCS Institute of Neurology "C. Besta", 20126 Milan, Italy
| | - Sergio Giannattasio
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, CNR, 70125 Bari, Italy
| | - Maria A Donati
- Department of Neuroscience, Children's Hospital "A. Meyer", 50139 Florence, Italy
| | - Renzo Guerrini
- Department of Neuroscience, Children's Hospital "A. Meyer", 50139 Florence, Italy.,IRCCS Stella Maris Foundation, 56128 Pisa, Italy
| | - Luigi Palmieri
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, 70125 Bari, Italy.,Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, CNR, 70125 Bari, Italy
| | - Ferdinando Palmieri
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, 70125 Bari, Italy.,Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, CNR, 70125 Bari, Italy
| | - Anna De Grassi
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, 70125 Bari, Italy
| |
Collapse
|
17
|
Prendecki M, Florczak-Wyspianska J, Kowalska M, Ilkowski J, Grzelak T, Bialas K, Wiszniewska M, Kozubski W, Dorszewska J. Biothiols and oxidative stress markers and polymorphisms of TOMM40 and APOC1 genes in Alzheimer's disease patients. Oncotarget 2018; 9:35207-35225. [PMID: 30443289 PMCID: PMC6219666 DOI: 10.18632/oncotarget.26184] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 09/01/2018] [Indexed: 12/16/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive disease, with frequently observed improper biothiols turnover, homocysteine (Hcy) and glutathione (GSH). GSH protects cells from oxidative stress and may be determined by 8-oxo-2’-deoxyguanosine (8-oxo2dG) level and its repair enzyme 8-oxoguanine DNA glycosylase (OGG1). The presence of unfavorable alleles, e.g., in APOE cluster, TOMM40 or APOC1 is known to facilitate the dementia onset under oxidative stress. The aim of the study was to analyze rs1052452, rs2075650 TOMM40 polymorphisms, rs4420638 APOC1, and their correlation with Hcy, GSH, 8-oxo2dG, OGG1 levels in plasma of AD patients and controls. We recruited 230 individuals: 88 AD, 80 controls without (UC), 62 controls with (RC) positive family history of AD. The TOMM40 genotype was determined by HRM and capillary electrophoresis, while APOC1 by HRM. The concentrations of OGG1, 8-oxo2dG were determined by ELISA, whereas Hcy, GSH by HPLC/EC. We showed that over 60% of AD patients had increased Hcy levels (p<0.01 vs. UC, p<0.001 vs. RC), while GSH (p<0.01 vs. UC), 8-oxo2dG (p<0.01 vs. UC, p<0.001 vs. RC) were reduced. Minor variants: rs10524523-L, rs4420638-G, rs2075650-G were significantly overrepresented in AD. For rs4420638-G, rs2075650-G variants, the association remained significant in APOE E4 non-carriers. The misbalance of analyzed biothiols, and 8-oxo2dG, OGG1 were more pronounced in carriers of major variants: rs10524523-S/VL, rs4420638-A, rs2075650-A. We showed, for the first time, that APOC1 and TOMM40 rs2075650 polymorphisms may be independent risk factors of developing AD, whose major variants are accompanied by disruption of biothiols metabolism and inefficient removal of DNA oxidation.
Collapse
Affiliation(s)
- Michal Prendecki
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, Poznan, Poland
| | | | - Marta Kowalska
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, Poznan, Poland
| | - Jan Ilkowski
- Department of Emergency Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Teresa Grzelak
- Division of Biology of Civilization-Linked Diseases, Department of Chemistry and Clinical Biochemistry, Poznan University of Medical Sciences, Poznan, Poland
| | - Katarzyna Bialas
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, Poznan, Poland
| | - Malgorzata Wiszniewska
- Faculty of Health Care, Stanislaw Staszic University of Applied Sciences in Pila, Pila, Poland.,Department of Neurology, Specialistic Hospital in Pila, Pila, Poland
| | - Wojciech Kozubski
- Chair and Department of Neurology, Poznan University of Medical Sciences, Poznan, Poland
| | - Jolanta Dorszewska
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
18
|
Hlouschek J, Ritter V, Wirsdörfer F, Klein D, Jendrossek V, Matschke J. Targeting SLC25A10 alleviates improved antioxidant capacity and associated radioresistance of cancer cells induced by chronic-cycling hypoxia. Cancer Lett 2018; 439:24-38. [PMID: 30205167 DOI: 10.1016/j.canlet.2018.09.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 08/22/2018] [Accepted: 09/03/2018] [Indexed: 01/17/2023]
Abstract
High tumor heterogeneity and increased therapy resistance acquired in a hypoxic tumor microenvironment remain major obstacles to successful radiotherapy. Others and we have shown that adaptation of cancer cells to cycling severe hypoxia and intermittent reoxygenation stress (chronic-cycling hypoxia) increases cellular antioxidant capacity thereby supporting resistance to chemotherapy and radiotherapy. Here we explored the involvement of antioxidant-associated mitochondrial transport-systems for maintenance of redox-homeostasis in adaptation to chronic-cycling hypoxia and associated radioresistance. Genetic or pharmacological inhibition of the mitochondrial dicarboxylate carrier (SLC25A10) or the oxoglutarate-carrier (SLC25A11) increased the cytotoxic effects of ionizing radiation (IR). But only targeting of SLC25A10 was effective in overcoming chronic-cycling hypoxia-induced enhanced death resistance in vitro and in vivo by disturbing increased antioxidant capacity. Furthermore, in silico analysis revealed that overexpression of SLC25A10 but not SLC25A11 is associated with reduced overall survival in lung- and breast-cancer patients. Our study reveals a role of SLC25A10 in supporting both, redox- and energy-homeostasis, ensuring radioresistance of cancer cells with tolerance to chronic-cycling hypoxia thereby proposing a novel strategy to overcome a mechanism of hypoxia-induced therapy resistance with potential clinical relevance regarding decreased patient survival.
Collapse
Affiliation(s)
- Julian Hlouschek
- Institute of Cell Biology (Cancer Research), University Hospital Essen, 45122, Essen, North Rhine-Westphalia, Germany
| | - Violetta Ritter
- Institute of Cell Biology (Cancer Research), University Hospital Essen, 45122, Essen, North Rhine-Westphalia, Germany
| | - Florian Wirsdörfer
- Institute of Cell Biology (Cancer Research), University Hospital Essen, 45122, Essen, North Rhine-Westphalia, Germany
| | - Diana Klein
- Institute of Cell Biology (Cancer Research), University Hospital Essen, 45122, Essen, North Rhine-Westphalia, Germany
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University Hospital Essen, 45122, Essen, North Rhine-Westphalia, Germany
| | - Johann Matschke
- Institute of Cell Biology (Cancer Research), University Hospital Essen, 45122, Essen, North Rhine-Westphalia, Germany.
| |
Collapse
|
19
|
Ravindran S, Ramachandran K, Kurian GA. Sodium thiosulfate mediated cardioprotection against myocardial ischemia-reperfusion injury is defunct in rat heart with co-morbidity of vascular calcification. Biochimie 2018; 147:80-88. [PMID: 29366934 DOI: 10.1016/j.biochi.2018.01.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 01/11/2018] [Indexed: 12/27/2022]
Abstract
Sodium thiosulfate (STS) has shown promising effects in amelioration of myocardial ischemia-reperfusion injury (IR) in a rat model and is clinically useful in the treatment of chronic kidney disease (CKD) associated calciphylaxis. As the prevalence of cardiac complications is higher in CKD, we tested the effectiveness of STS in a rat model of adenine-induced vascular calcification and subjected the heart to IR. We observed an increased infarct size (29%) by TTC staining, lactate dehydrogenase (54%) and creatine kinase (32%) release in the coronary perfusate and altered hemodynamics compared to a normal rat treated with STS and subjected to IR. As functional mitochondria are essential for preserving heart from the detrimental effects of IR, we found that calcification induced mitochondrial dysfunction (reduced RCR->80%, P/O ratio->30%, ΔΨ->10% and swelling- 27%), could not be restored efficiently by STS treatment. Therefore we used nicorandil (mitochondrial potassium channel opener) along with STS as a combination therapy to treat the diseased heart and found an improvement in cardioprotection against IR injury, compared to STS alone. Upon evaluating these hearts, we found that both the cardiac mitochondria namely interfibrillar and subsarcolemmal were functionally well preserved.
Collapse
Affiliation(s)
- Sriram Ravindran
- Vascular Biology Lab, SASTRA Deemed University, Thanjavur, 613401, India
| | | | - Gino A Kurian
- Vascular Biology Lab, SASTRA Deemed University, Thanjavur, 613401, India.
| |
Collapse
|
20
|
Baulies A, Montero J, Matías N, Insausti N, Terrones O, Basañez G, Vallejo C, Conde de La Rosa L, Martinez L, Robles D, Morales A, Abian J, Carrascal M, Machida K, Kumar DBU, Tsukamoto H, Kaplowitz N, Garcia-Ruiz C, Fernández-Checa JC. The 2-oxoglutarate carrier promotes liver cancer by sustaining mitochondrial GSH despite cholesterol loading. Redox Biol 2017; 14:164-177. [PMID: 28942194 PMCID: PMC5609874 DOI: 10.1016/j.redox.2017.08.022] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 08/21/2017] [Accepted: 08/24/2017] [Indexed: 12/15/2022] Open
Abstract
Cancer cells exhibit mitochondrial cholesterol (mt-cholesterol) accumulation, which contributes to cell death resistance by antagonizing mitochondrial outer membrane (MOM) permeabilization. Hepatocellular mt-cholesterol loading, however, promotes steatohepatitis, an advanced stage of chronic liver disease that precedes hepatocellular carcinoma (HCC), by depleting mitochondrial GSH (mGSH) due to a cholesterol-mediated impairment in mGSH transport. Whether and how HCC cells overcome the restriction of mGSH transport imposed by mt-cholesterol loading to support mGSH uptake remains unknown. Although the transport of mGSH is not fully understood, SLC25A10 (dicarboxylate carrier, DIC) and SLC25A11 (2-oxoglutarate carrier, OGC) have been involved in mGSH transport, and therefore we examined their expression and role in HCC. Unexpectedly, HCC cells and liver explants from patients with HCC exhibit divergent expression of these mitochondrial carriers, with selective OGC upregulation, which contributes to mGSH maintenance. OGC but not DIC downregulation by siRNA depleted mGSH levels and sensitized HCC cells to hypoxia-induced ROS generation and cell death as well as impaired cell growth in three-dimensional multicellular HCC spheroids, effects that were reversible upon mGSH replenishment by GSH ethyl ester, a membrane permeable GSH precursor. We also show that OGC regulates mitochondrial respiration and glycolysis. Moreover, OGC silencing promoted hypoxia-induced cardiolipin peroxidation, which reversed the inhibition of cholesterol on the permeabilization of MOM-like liposomes induced by Bax or Bak. Genetic OGC knockdown reduced the ability of tumor-initiating stem-like cells to induce liver cancer. These findings underscore the selective overexpression of OGC as an adaptive mechanism of HCC to provide adequate mGSH levels in the face of mt-cholesterol loading and suggest that OGC may be a novel therapeutic target for HCC treatment.
Collapse
Affiliation(s)
- Anna Baulies
- Department of Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, Consejo Superior de Investigaciones Científicas, 08036 Barcelona, Spain; Liver Unit and Hospital Clínic i Provincial, IDIBAPS, and Centro de Investigación Biomédica en Red (CIBERehd), Spain
| | - Joan Montero
- Department of Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, Consejo Superior de Investigaciones Científicas, 08036 Barcelona, Spain; Liver Unit and Hospital Clínic i Provincial, IDIBAPS, and Centro de Investigación Biomédica en Red (CIBERehd), Spain
| | - Nuria Matías
- Department of Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, Consejo Superior de Investigaciones Científicas, 08036 Barcelona, Spain; Liver Unit and Hospital Clínic i Provincial, IDIBAPS, and Centro de Investigación Biomédica en Red (CIBERehd), Spain
| | - Naroa Insausti
- Department of Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, Consejo Superior de Investigaciones Científicas, 08036 Barcelona, Spain; Liver Unit and Hospital Clínic i Provincial, IDIBAPS, and Centro de Investigación Biomédica en Red (CIBERehd), Spain
| | - Oihana Terrones
- Unidad de Biofísica (Centro Mixto Consejo Superior de Investigaciones Científicas-Universidad del País Vasco/Euskal Herriko Unibertsitatea), Universidad del País Vasco/Euskal Herriko Unibertsitatea, 48080 Bilbao, Spain
| | - Gorka Basañez
- Unidad de Biofísica (Centro Mixto Consejo Superior de Investigaciones Científicas-Universidad del País Vasco/Euskal Herriko Unibertsitatea), Universidad del País Vasco/Euskal Herriko Unibertsitatea, 48080 Bilbao, Spain
| | - Carmen Vallejo
- Department of Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, Consejo Superior de Investigaciones Científicas, 08036 Barcelona, Spain; Liver Unit and Hospital Clínic i Provincial, IDIBAPS, and Centro de Investigación Biomédica en Red (CIBERehd), Spain
| | - Laura Conde de La Rosa
- Department of Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, Consejo Superior de Investigaciones Científicas, 08036 Barcelona, Spain; Liver Unit and Hospital Clínic i Provincial, IDIBAPS, and Centro de Investigación Biomédica en Red (CIBERehd), Spain
| | - Laura Martinez
- Department of Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, Consejo Superior de Investigaciones Científicas, 08036 Barcelona, Spain; Liver Unit and Hospital Clínic i Provincial, IDIBAPS, and Centro de Investigación Biomédica en Red (CIBERehd), Spain
| | - David Robles
- Department of Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, Consejo Superior de Investigaciones Científicas, 08036 Barcelona, Spain; Liver Unit and Hospital Clínic i Provincial, IDIBAPS, and Centro de Investigación Biomédica en Red (CIBERehd), Spain
| | - Albert Morales
- Department of Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, Consejo Superior de Investigaciones Científicas, 08036 Barcelona, Spain
| | - Joaquin Abian
- CSIC/UAB Proteomics Laboratory, IIBB-CSIC, 08036 Barcelona, Spain
| | | | - Keigo Machida
- Southern California Research Center for ALPD and Cirrhosis, Los Angeles, CA, USA
| | - Dinesh B U Kumar
- Southern California Research Center for ALPD and Cirrhosis, Los Angeles, CA, USA
| | - Hidekazu Tsukamoto
- Southern California Research Center for ALPD and Cirrhosis, Los Angeles, CA, USA; Department of Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Neil Kaplowitz
- University of Southern California Research Center for Liver Diseases, Keck School of Medicine, USC, Los Angeles, CA, USA
| | - Carmen Garcia-Ruiz
- Department of Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, Consejo Superior de Investigaciones Científicas, 08036 Barcelona, Spain; Liver Unit and Hospital Clínic i Provincial, IDIBAPS, and Centro de Investigación Biomédica en Red (CIBERehd), Spain; Southern California Research Center for ALPD and Cirrhosis, Los Angeles, CA, USA; University of Southern California Research Center for Liver Diseases, Keck School of Medicine, USC, Los Angeles, CA, USA.
| | - José C Fernández-Checa
- Department of Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, Consejo Superior de Investigaciones Científicas, 08036 Barcelona, Spain; Liver Unit and Hospital Clínic i Provincial, IDIBAPS, and Centro de Investigación Biomédica en Red (CIBERehd), Spain; Southern California Research Center for ALPD and Cirrhosis, Los Angeles, CA, USA; University of Southern California Research Center for Liver Diseases, Keck School of Medicine, USC, Los Angeles, CA, USA.
| |
Collapse
|
21
|
Zhou X, Paredes JA, Krishnan S, Curbo S, Karlsson A. The mitochondrial carrier SLC25A10 regulates cancer cell growth. Oncotarget 2016; 6:9271-83. [PMID: 25797253 PMCID: PMC4496216 DOI: 10.18632/oncotarget.3375] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 02/11/2015] [Indexed: 12/13/2022] Open
Abstract
Dysregulation of cell metabolism is critical for the growth properties of cancer cells. The purpose of this study was to understand the role of substrate transport across the mitochondrial membrane to sustain the metabolic shift and redox defense in cancer cells. Mitochondrial carrier SLC25A10 is up-regulated in a variety of tumors and is involved in regulating intracellular levels of reactive oxygen species. We show that knockdown of SLC25A10 in A549 cells changed the growth properties to a less malignant phenotype and casued increased glutamine dependency and sensitivity to oxidative stress. The metabolic alteration was linked to an energy metabolic shift from glycolysis to mitochondrial oxidative phosphorylation illustrated by increased expression of glutamate dehydrogenase, decreased expression of lactate dehydrogenase due to down-regulation of hypoxia inducible factor 1α. We identified effects on NADPH production linked to the growth changes observed in SLC25A10 knockdown cells, demonstrated by decreased NADPH production in cells deprived of glutamine. The contribution of SLC25A10 to reprogram cell metabolism and to regulate cell growth suggests SLC25A10 as a novel target for anti-cancer strategies.
Collapse
Affiliation(s)
- Xiaoshan Zhou
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden
| | - João A Paredes
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden
| | - Shuba Krishnan
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden
| | - Sophie Curbo
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden
| | - Anna Karlsson
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden
| |
Collapse
|
22
|
Lash LH. Mitochondrial Glutathione in Diabetic Nephropathy. J Clin Med 2015; 4:1428-47. [PMID: 26239684 PMCID: PMC4519798 DOI: 10.3390/jcm4071428] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 06/25/2015] [Accepted: 06/26/2015] [Indexed: 01/05/2023] Open
Abstract
Although there are many etiologies for diabetic nephropathy (DN), one common characteristic of all cases involves mitochondrial oxidative stress and consequent bioenergetic dysfunction. As the predominant low-molecular-weight, intramitochondrial thiol reductant, the mitochondrial glutathione (mtGSH) pool plays important roles in how this organelle adapts to the chronic hyperglycemia and redox imbalances associated with DN. This review will summarize information about the processes by which this important GSH pool is regulated and how manipulation of these processes can affect mitochondrial and cellular function in the renal proximal tubule. Mitochondria in renal proximal tubular (PT) cells do not appear to synthesize GSH de novo but obtain it by transport from the cytoplasm. Two inner membrane organic anion carriers, the dicarboxylate carrier (DIC; Slc25a10) and 2-oxoglutarate carrier (OGC; Slc25a11) are responsible for this transport. Genetic modulation of DIC or OGC expression in vitro in PT cells from diabetic rats can alter mitochondrial function and susceptibility of renal PT cells to oxidants, with overexpression leading to reversion of bioenergetic conditions to a non-diabetic state and protection of cells from injury. These findings support the mtGSH carriers as potential therapeutic targets to correct the underlying metabolic disturbance in DN.
Collapse
Affiliation(s)
- Lawrence H Lash
- Department of Pharmacology, Wayne State University School of Medicine, 540 East Canfield Avenue, Detroit, MI 48201, USA.
| |
Collapse
|
23
|
Ribas V, García-Ruiz C, Fernández-Checa JC. Glutathione and mitochondria. Front Pharmacol 2014; 5:151. [PMID: 25024695 PMCID: PMC4079069 DOI: 10.3389/fphar.2014.00151] [Citation(s) in RCA: 352] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 06/10/2014] [Indexed: 12/16/2022] Open
Abstract
Glutathione (GSH) is the main non-protein thiol in cells whose functions are dependent on the redox-active thiol of its cysteine moiety that serves as a cofactor for a number of antioxidant and detoxifying enzymes. While synthesized exclusively in the cytosol from its constituent amino acids, GSH is distributed in different compartments, including mitochondria where its concentration in the matrix equals that of the cytosol. This feature and its negative charge at physiological pH imply the existence of specific carriers to import GSH from the cytosol to the mitochondrial matrix, where it plays a key role in defense against respiration-induced reactive oxygen species and in the detoxification of lipid hydroperoxides and electrophiles. Moreover, as mitochondria play a central strategic role in the activation and mode of cell death, mitochondrial GSH has been shown to critically regulate the level of sensitization to secondary hits that induce mitochondrial membrane permeabilization and release of proteins confined in the intermembrane space that once in the cytosol engage the molecular machinery of cell death. In this review, we summarize recent data on the regulation of mitochondrial GSH and its role in cell death and prevalent human diseases, such as cancer, fatty liver disease, and Alzheimer’s disease.
Collapse
Affiliation(s)
- Vicent Ribas
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona, Consejo Superior de Investigaciones Científicas (IIBB-CSIC) Barcelona, Spain ; Liver Unit, Hospital Clínic, Centre Esther Koplowitz, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)-Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd) Barcelona, Spain
| | - Carmen García-Ruiz
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona, Consejo Superior de Investigaciones Científicas (IIBB-CSIC) Barcelona, Spain ; Liver Unit, Hospital Clínic, Centre Esther Koplowitz, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)-Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd) Barcelona, Spain ; Research Center for Alcoholic Liver and Pancreatic Diseases and Cirrhosis, Keck School of Medicine, University of Southern California Los Angeles, CA, USA
| | - José C Fernández-Checa
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona, Consejo Superior de Investigaciones Científicas (IIBB-CSIC) Barcelona, Spain ; Liver Unit, Hospital Clínic, Centre Esther Koplowitz, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)-Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd) Barcelona, Spain ; Research Center for Alcoholic Liver and Pancreatic Diseases and Cirrhosis, Keck School of Medicine, University of Southern California Los Angeles, CA, USA
| |
Collapse
|
24
|
Wilkins HM, Brock S, Gray JJ, Linseman DA. Stable over-expression of the 2-oxoglutarate carrier enhances neuronal cell resistance to oxidative stress via Bcl-2-dependent mitochondrial GSH transport. J Neurochem 2014; 130:75-86. [PMID: 24606213 DOI: 10.1111/jnc.12709] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 03/03/2014] [Accepted: 03/04/2014] [Indexed: 11/29/2022]
Abstract
Mitochondrial glutathione (GSH) is a key endogenous antioxidant and its maintenance is critical for cell survival. Here, we generated stable NSC34 motor neuron-like cell lines over-expressing the mitochondrial GSH transporter, the 2-oxoglutarate carrier (OGC), to further elucidate the importance of mitochondrial GSH transport in determining neuronal resistance to oxidative stress. Two stable OGC cell lines displayed specific increases in mitochondrial GSH content and resistance to oxidative and nitrosative stressors, but not staurosporine. Inhibition of transport through OGC reduced levels of mitochondrial GSH and resensitized the stable cell lines to oxidative stress. The stable OGC cell lines displayed significant up-regulation of the anti-apoptotic protein, B cell lymphoma 2 (Bcl-2). This result was reproduced in parental NSC34 cells by chronic treatment with GSH monoethylester, which specifically increased mitochondrial GSH levels. Knockdown of Bcl-2 expression decreased mitochondrial GSH and resensitized the stable OGC cells to oxidative stress. Finally, endogenous OGC was co-immunoprecipitated with Bcl-2 from rat brain lysates in a GSH-dependent manner. These data are the first to show that increased mitochondrial GSH transport is sufficient to enhance neuronal resistance to oxidative stress. Moreover, sustained and specific enhancement of mitochondrial GSH leads to increased Bcl-2 expression, a required mechanism for the maintenance of increased mitochondrial GSH levels. Stable over-expression of the 2-oxoglutarate carrier (OGC) in a motor neuronal cell line induced a specific increase in mitochondrial GSH and markedly enhanced resistance to oxidative stress. Over-expression of OGC also induced Bcl-2 expression which was owing to the specific increase in mitochondrial GSH. Intriguingly, enhanced expression of Bcl-2 was required to sustain OGC-dependent GSH transport into the mitochondria. Thus, OGC and Bcl-2 work in a concerted manner to maintain the mitochondrial GSH pool which is crucial for neuronal survival.
Collapse
Affiliation(s)
- Heather M Wilkins
- Department of Biological Sciences and Eleanor Roosevelt Institute, University of Denver, Denver, Colorado, USA
| | | | | | | |
Collapse
|
25
|
Abstract
The mitochondrion relies on compartmentalization of certain enzymes, ions and metabolites for the sake of efficient metabolism. In order to fulfil its activities, a myriad of carriers are properly expressed, targeted and folded in the inner mitochondrial membrane. Among these carriers, the six-transmembrane-helix mitochondrial SLC25 (solute carrier family 25) proteins facilitate transport of solutes with disparate chemical identities across the inner mitochondrial membrane. Although their proper function replenishes building blocks needed for metabolic reactions, dysfunctional SLC25 proteins are involved in pathological states. It is the purpose of the present review to cover the current knowledge on the role of SLC25 transporters in health and disease.
Collapse
|
26
|
Abstract
Rapid advances in redox systems biology are creating new opportunities to understand complexities of human disease and contributions of environmental exposures. New understanding of thiol-disulfide systems have occurred during the past decade as a consequence of the discoveries that thiol and disulfide systems are maintained in kinetically controlled steady states displaced from thermodynamic equilibrium, that a widely distributed family of NADPH oxidases produces oxidants that function in cell signaling and that a family of peroxiredoxins utilize thioredoxin as a reductant to complement the well-studied glutathione antioxidant system for peroxide elimination and redox regulation. This review focuses on thiol/disulfide redox state in biologic systems and the knowledge base available to support development of integrated redox systems biology models to better understand the function and dysfunction of thiol-disulfide redox systems. In particular, central principles have emerged concerning redox compartmentalization and utility of thiol/disulfide redox measures as indicators of physiologic function. Advances in redox proteomics show that, in addition to functioning in protein active sites and cell signaling, cysteine residues also serve as redox sensors to integrate biologic functions. These advances provide a framework for translation of redox systems biology concepts to practical use in understanding and treating human disease. Biological responses to cadmium, a widespread environmental agent, are used to illustrate the utility of these advances to the understanding of complex pleiotropic toxicities.
Collapse
Affiliation(s)
- Young-Mi Go
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, GA 30322, USA
| | | |
Collapse
|
27
|
Wilkins HM, Kirchhof D, Manning E, Joseph JW, Linseman DA. Mitochondrial glutathione transport is a key determinant of neuronal susceptibility to oxidative and nitrosative stress. J Biol Chem 2013; 288:5091-101. [PMID: 23283974 DOI: 10.1074/jbc.m112.405738] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Mitochondrial oxidative stress significantly contributes to the underlying pathology of several devastating neurodegenerative disorders. Mitochondria are highly sensitive to the damaging effects of reactive oxygen and nitrogen species; therefore, these organelles are equipped with a number of free radical scavenging systems. In particular, the mitochondrial glutathione (GSH) pool is a critical antioxidant reserve that is derived entirely from the larger cytosolic pool via facilitated transport. The mechanism of mitochondrial GSH transport has not been extensively studied in the brain. However, the dicarboxylate (DIC) and 2-oxoglutarate (OGC) carriers localized to the inner mitochondrial membrane have been established as GSH transporters in liver and kidney. Here, we investigated the role of these carriers in protecting neurons from oxidative and nitrosative stress. Immunoblot analysis of DIC and OGC in primary cultures of rat cerebellar granule neurons (CGNs) and cerebellar astrocytes showed differential expression of these carriers, with CGNs expressing only DIC and astrocytes expressing both DIC and OGC. Consistent with these findings, butylmalonate specifically reduced mitochondrial GSH in CGNs, whereas both butylmalonate and phenylsuccinate diminished mitochondrial GSH in astrocytes. Moreover, preincubation with butylmalonate but not phenylsuccinate significantly enhanced susceptibility of CGNs to oxidative and nitrosative stressors. This increased vulnerability was largely prevented by incubation with cell-permeable GSH monoethylester but not malate. Finally, knockdown of DIC with adenoviral siRNA also rendered CGNs more susceptible to oxidative stress. These findings demonstrate that maintenance of the mitochondrial GSH pool via sustained mitochondrial GSH transport is essential to protect neurons from oxidative and nitrosative stress.
Collapse
Affiliation(s)
- Heather M Wilkins
- Department of Biological Sciences and Eleanor Roosevelt Institute, University of Denver, Denver, Colorado 80208, USA
| | | | | | | | | |
Collapse
|
28
|
Marí M, Morales A, Colell A, García-Ruiz C, Kaplowitz N, Fernández-Checa JC. Mitochondrial glutathione: features, regulation and role in disease. Biochim Biophys Acta Gen Subj 2012; 1830:3317-28. [PMID: 23123815 DOI: 10.1016/j.bbagen.2012.10.018] [Citation(s) in RCA: 142] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 10/12/2012] [Accepted: 10/23/2012] [Indexed: 02/07/2023]
Abstract
BACKGROUND Mitochondria are the powerhouse of mammalian cells and the main source of reactive oxygen species (ROS) associated with oxygen consumption. In addition, they also play a strategic role in controlling the fate of cells through regulation of death pathways. Mitochondrial ROS production fulfills a signaling role through regulation of redox pathways, but also contributes to mitochondrial damage in a number of pathological states. SCOPE OF REVIEW Mitochondria are exposed to the constant generation of oxidant species, and yet the organelle remains functional due to the existence of an armamentarium of antioxidant defense systems aimed to repair oxidative damage, of which mitochondrial glutathione (mGSH) is of particular relevance. Thus, the aim of the review is to cover the regulation of mGSH and its role in disease. MAJOR CONCLUSIONS Cumulating evidence over recent years has demonstrated the essential role for mGSH in mitochondrial physiology and disease. Despite its high concentration in the mitochondrial matrix, mitochondria lack the enzymes to synthesize GSH de novo, so that mGSH originates from cytosolic GSH via transport through specific mitochondrial carriers, which exhibit sensitivity to membrane dynamics. Depletion of mGSH sensitizes cells to stimuli leading to oxidative stress such as TNF, hypoxia or amyloid β-peptide, thereby contributing to disease pathogenesis. GENERAL SIGNIFICANCE Understanding the regulation of mGSH may provide novel insights to disease pathogenesis and toxicity and the opportunity to design therapeutic targets of intervention in cell death susceptibility and disease. This article is part of a Special Issue entitled Cellular functions of glutathione.
Collapse
Affiliation(s)
- Montserrat Marí
- Department of Cell Death and Proliferation, IIBB-CSIC, Barcelona, Spain.
| | | | | | | | | | | |
Collapse
|
29
|
Abstract
Dysregulation of glutathione homeostasis and alterations in glutathione-dependent enzyme activities are increasingly implicated in the induction and progression of neurodegenerative diseases, including Alzheimer’s, Parkinson’s and Huntington’s diseases, amyotrophic lateral sclerosis, and Friedreich’s ataxia. In this review background is provided on the steady-state synthesis, regulation, and transport of glutathione, with primary focus on the brain. A brief overview is presented on the distinct but vital roles of glutathione in cellular maintenance and survival, and on the functions of key glutathione-dependent enzymes. Major contributors to initiation and progression of neurodegenerative diseases are considered, including oxidative stress, protein misfolding, and protein aggregation. In each case examples of key regulatory mechanisms are identified that are sensitive to changes in glutathione redox status and/or in the activities of glutathione-dependent enzymes. Mechanisms of dysregulation of glutathione and/or glutathione-dependent enzymes are discussed that are implicated in pathogenesis of each neurodegenerative disease. Limitations in information or interpretation are identified, and possible avenues for further research are described with an aim to elucidating novel targets for therapeutic interventions. The pros and cons of administration of N-acetylcysteine or glutathione as therapeutic agents for neurodegenerative diseases, as well as the potential utility of serum glutathione as a biomarker, are critically evaluated.
Collapse
|
30
|
Circu ML, Aw TY. Glutathione and modulation of cell apoptosis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2012; 1823:1767-77. [PMID: 22732297 DOI: 10.1016/j.bbamcr.2012.06.019] [Citation(s) in RCA: 241] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Revised: 05/24/2012] [Accepted: 06/13/2012] [Indexed: 01/01/2023]
Abstract
Apoptosis is a highly organized form of cell death that is important for tissue homeostasis, organ development and senescence. To date, the extrinsic (death receptor mediated) and intrinsic (mitochondria derived) apoptotic pathways have been characterized in mammalian cells. Reduced glutathione, is the most prevalent cellular thiol that plays an essential role in preserving a reduced intracellular environment. glutathione protection of cellular macromolecules like deoxyribose nucleic acid proteins and lipids against oxidizing, environmental and cytotoxic agents, underscores its central anti-apoptotic function. Reactive oxygen and nitrogen species can oxidize cellular glutathione or induce its extracellular export leading to the loss of intracellular redox homeostasis and activation of the apoptotic signaling cascade. Recent evidence uncovered a novel role for glutathione involvement in apoptotic signaling pathways wherein post-translational S-glutathiolation of protein redox active cysteines is implicated in the potentiation of apoptosis. In the present review we focus on the key aspects of glutathione redox mechanisms associated with apoptotic signaling that includes: (a) changes in cellular glutathione redox homeostasis through glutathione oxidation or GSH transport in relation to the initiation or propagation of the apoptotic cascade, and (b) evidence for S-glutathiolation in protein modulation and apoptotic initiation.
Collapse
Affiliation(s)
- Magdalena L Circu
- Department of Molecular & Cellular Physiology, Louisiana University Health Sciences Center, Shreveport, LA 71130, USA
| | | |
Collapse
|
31
|
Abstract
Abstract
The tripeptide glutathione (GSH), comprised of the amino acids l-cysteine, glycine, and l-glutamate, is found in all cells of aerobic organisms and plays numerous, critical roles as an antioxidant and nucleophile in regulating cellular homeostasis and drug metabolism. GSH is synthesized exclusively in the cytoplasm of most cells by two ATP-dependent reactions. Despite this compartmentation, GSH is found in other subcellular compartments, including mitochondria. As the GSH molecule has a net negative charge at physiological pH, it cannot cross cellular membranes by diffusion. Rather, GSH is a substrate for a variety of anion and amino acid transporters. Two organic anion carriers in the inner membrane of renal mitochondria, the dicarboxylate carrier (DIC; Slc25a10) and the 2-oxoglutarate carrier (OGC; Slc25a11), are responsible for most of the transport of GSH from cytoplasm into mitochondrial matrix. Genetic manipulation of DIC and/or OGC expression in renal cell lines demonstrated the ability to produce sustained increases in mitochondrial GSH content, which then protected these cells from cytotoxicity due to several oxidants and mitochondrial toxicants. Several diseases and pathological states are associated with mitochondrial dysfunction and oxidative stress, suggesting that the mitochondrial GSH pool may be a therapeutic target. One such disease that is of particular concern for public health is diabetic nephropathy. Another chronic, pathological state that is associated with bioenergetic and redox changes is compensatory renal hypertrophy that results from reductions in functional renal mass. This review summarizes pathways of mitochondrial GSH transport and discusses studies on its manipulation in toxicological and pathological states.
Collapse
Affiliation(s)
- Lawrence H. Lash
- Department of Pharmacology, Wayne State University School of Medicine, 540 East Canfield Avenue, Detroit, MI 48201, USA. Fax: +01-313-577-6739; Tel: +01-313-577-0475
| |
Collapse
|
32
|
Circu ML, Aw TY. Intestinal redox biology and oxidative stress. Semin Cell Dev Biol 2012; 23:729-37. [PMID: 22484611 DOI: 10.1016/j.semcdb.2012.03.014] [Citation(s) in RCA: 204] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Revised: 03/13/2012] [Accepted: 03/14/2012] [Indexed: 12/19/2022]
Abstract
The intestinal epithelium sits at the interface between an organism and its luminal environment, and as such is prone to oxidative damage induced by luminal oxidants. Mucosal integrity is maintained by the luminal redox status of the glutathione/glutathione disulfide (GSH/GSSG) and cysteine/cystine (Cys/CySS) couples which also support luminal nutrient absorption, mucus fluidity, and a diverse microbiota. The epithelial layer is uniquely organized for rapid self-renewal that is achieved by the well-regulated processes of crypt stem cell proliferation and crypt-to-villus cell differentiation. The GSH/GSSG and Cys/CySS redox couples, known to modulate intestinal cell transition through proliferation, differentiation or apoptosis, could govern the regenerative potential of the mucosa. These two couples, together with that of the thioredoxin/thioredoxin disulfide (Trx/TrxSS) couple are the major intracellular redox systems, and it is proposed that they each function as distinctive redox control nodes or circuitry in the control of metabolic processes and networks of enzymatic reactions. Specificity of redox signaling is accomplished in part by subcellular compartmentation of the individual redox systems within the mitochondria, nucleus, endoplasmic reticulum, and cytosol wherein each defined redox environment is suited to the specific metabolic function within that compartment. Mucosal oxidative stress would result from the disruption of these unique redox control nodes, and the subsequent alteration in redox signaling can contribute to the development of degenerative pathologies of the intestine, such as inflammation and cancer.
Collapse
Affiliation(s)
- Magdalena L Circu
- Department of Molecular & Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| | | |
Collapse
|
33
|
Murphy MP. Mitochondrial thiols in antioxidant protection and redox signaling: distinct roles for glutathionylation and other thiol modifications. Antioxid Redox Signal 2012; 16:476-95. [PMID: 21954972 DOI: 10.1089/ars.2011.4289] [Citation(s) in RCA: 251] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
SIGNIFICANCE The mitochondrial matrix contains much of the machinery at the heart of metabolism. This compartment is also exposed to a high and continual flux of superoxide, hydrogen peroxide, and related reactive species. To protect mitochondria from these sources of oxidative damage, there is an integrated set of thiol systems within the matrix comprising the thioredoxin/peroxiredoxin/methionine sulfoxide reductase pathways and the glutathione/glutathione peroxidase/glutathione-S-transferase/glutaredoxin pathways that in conjunction with protein thiols prevent much of this oxidative damage. In addition, the changes in the redox state of many components of these mitochondrial thiol systems may transduce and relay redox signals within and through the mitochondrial matrix to modulate the activity of biochemical processes. RECENT ADVANCES Here, mitochondrial thiol systems are reviewed, and areas of uncertainty are pointed out, focusing on recent developments in our understanding of their roles. CRITICAL ISSUES The areas of particular focus are on the multiple, overlapping roles of mitochondrial thiols and on understanding how these thiols contribute to both antioxidant defenses and redox signaling. FUTURE DIRECTIONS Recent technical progress in the identification and quantification of thiol modifications by redox proteomics means that many of the questions raised about the multiple roles of mitochondrial thiols can now be addressed.
Collapse
|
34
|
Wilkins HM, Marquardt K, Lash LH, Linseman DA. Bcl-2 is a novel interacting partner for the 2-oxoglutarate carrier and a key regulator of mitochondrial glutathione. Free Radic Biol Med 2012; 52:410-9. [PMID: 22115789 PMCID: PMC3253244 DOI: 10.1016/j.freeradbiomed.2011.10.495] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Revised: 10/28/2011] [Accepted: 10/31/2011] [Indexed: 01/06/2023]
Abstract
Despite making up only a minor fraction of the total cellular glutathione, recent studies indicate that the mitochondrial glutathione pool is essential for cell survival. Selective depletion of mitochondrial glutathione is sufficient to sensitize cells to mitochondrial oxidative stress (MOS) and intrinsic apoptosis. Glutathione is synthesized exclusively in the cytoplasm and must be actively transported into mitochondria. Therefore, regulation of mitochondrial glutathione transport is a key factor in maintaining the antioxidant status of mitochondria. Bcl-2 resides in the outer mitochondrial membrane where it acts as a central regulator of the intrinsic apoptotic cascade. In addition, Bcl-2 displays an antioxidant-like function that has been linked experimentally to the regulation of cellular glutathione content. We have previously demonstrated a novel interaction between recombinant Bcl-2 and reduced glutathione (GSH), which was antagonized by either Bcl-2 homology-3 domain (BH3) mimetics or a BH3-only protein, recombinant Bim. These previous findings prompted us to investigate if this novel Bcl-2/GSH interaction might play a role in regulating mitochondrial glutathione transport. Incubation of primary cultures of cerebellar granule neurons (CGNs) with the BH3 mimetic HA14-1 induced MOS and caused specific depletion of the mitochondrial glutathione pool. Bcl-2 was coimmunoprecipitated with GSH after chemical cross-linking in CGNs and this Bcl-2/GSH interaction was antagonized by preincubation with HA14-1. Moreover, both HA14-1 and recombinant Bim inhibited GSH transport into isolated rat brain mitochondria. To further investigate a possible link between Bcl-2 function and mitochondrial glutathione transport, we next examined if Bcl-2 associated with the 2-oxoglutarate carrier (OGC), an inner mitochondrial membrane protein known to transport glutathione in liver and kidney. After cotransfection of CHO cells, Bcl-2 was coimmunoprecipitated with OGC and this novel interaction was significantly enhanced by glutathione monoethyl ester. Similarly, recombinant Bcl-2 interacted with recombinant OGC in the presence of GSH. Bcl-2 and OGC cotransfection in CHO cells significantly increased the mitochondrial glutathione pool. Finally, the ability of Bcl-2 to protect CHO cells from apoptosis induced by hydrogen peroxide was significantly attenuated by the OGC inhibitor phenylsuccinate. These data suggest that GSH binding by Bcl-2 enhances its affinity for the OGC. Bcl-2 and OGC appear to act in a coordinated manner to increase the mitochondrial glutathione pool and enhance resistance of cells to oxidative stress. We conclude that regulation of mitochondrial glutathione transport is a principal mechanism by which Bcl-2 suppresses MOS.
Collapse
Affiliation(s)
- Heather M. Wilkins
- Department of Biological Sciences and Eleanor Roosevelt Institute, University of Denver, Denver, CO, USA
| | - Kristin Marquardt
- Department of Biological Sciences and Eleanor Roosevelt Institute, University of Denver, Denver, CO, USA
| | - Lawrence H. Lash
- Department of Pharmacology, Wayne State University, Detroit, MI, USA
| | - Daniel A. Linseman
- Department of Biological Sciences and Eleanor Roosevelt Institute, University of Denver, Denver, CO, USA
- Research Service, Veterans Affairs Medical Center, Denver, CO, USA
- Division of Clinical Pharmacology and Toxicology, Department of Medicine and Neuroscience Program, University of Colorado Denver, Aurora, CO, USA
- Address correspondence to: Daniel Linseman, PhD, Department of Biological Sciences and Eleanor Roosevelt Institute, University of Denver, 2199 S. University Blvd., Denver, CO 80208; Tel.: (303) 871-5654; Fax: (303) 871-3471;
| |
Collapse
|
35
|
Amaral AU, Cecatto C, Busanello ENB, Ribeiro CAJ, Melo DR, Leipnitz G, Castilho RF, Wajner M. Ethylmalonic acid impairs brain mitochondrial succinate and malate transport. Mol Genet Metab 2012; 105:84-90. [PMID: 22133302 DOI: 10.1016/j.ymgme.2011.10.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Revised: 10/13/2011] [Accepted: 10/13/2011] [Indexed: 10/16/2022]
Abstract
Tissue accumulation and high urinary excretion of ethylmalonic acid (EMA) occur in ethylmalonic encephalopathy (EE) and short chain acyl-CoA dehydrogenase deficiency (SCADD). Although these autosomal recessive disorders are clinically characterized by neurological abnormalities, the mechanisms underlying the brain damage are poorly known. Considering that little is known about the neurotoxicity of EMA and that hyperlacticacidemia occurs in EE and SCADD, we evaluated the effects of this metabolite on important parameters of oxidative metabolism in isolated rat brain mitochondria. EMA inhibited either ADP-stimulated or uncoupled mitochondrial respiration supported by succinate and malate, but not by glutamate plus malate. In addition, EMA mildly stimulated oxygen consumption by succinate-respiring mitochondria in resting state. Methylmalonic acid (MMA), malonic acid (MA) and butylmalonic acid (BtMA) had a similar effect on ADP-stimulated or uncoupled respiration. Furthermore, EMA-, MMA- and BtMA-induced inhibitory effects on succinate oxidation were significantly minimized by nonselective permeabilization of the mitochondrial membranes by alamethicin, whereas MA inhibitory effect was not altered. In addition, MA was the only tested compound that reduced succinate dehydrogenase activity. We also observed that EMA markedly inhibited succinate and malate transport through the mitochondrial dicarboxylate carrier. Mitochondrial membrane potential was also reduced by EMA and MA, but not by MMA, using succinate as electron donor, whereas none of these compounds was able to alter the membrane potential using glutamate plus malate as electron donors. Taken together, our results strongly indicate that EMA impairs succinate and malate uptake through the mitochondrial dicarboxylate carrier.
Collapse
Affiliation(s)
- Alexandre Umpierrez Amaral
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal de Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
The intestinal tract, known for its capability for self-renew, represents the first barrier of defence between the organism and its luminal environment. The thiol/disulfide redox systems comprising the glutathione/glutathione disulfide (GSH/GSSG), cysteine/cystine (Cys/CySS) and reduced and oxidized thioredoxin (Trx/TrxSS) redox couples play important roles in preserving tissue redox homeostasis, metabolic functions, and cellular integrity. Control of the thiol-disulfide status at the luminal surface is essential for maintaining mucus fluidity and absorption of nutrients, and protection against chemical-induced oxidant injury. Within intestinal cells, these redox couples preserve an environment that supports physiological processes and orchestrates networks of enzymatic reactions against oxidative stress. In this review, we focus on the intestinal redox and antioxidant systems, their subcellular compartmentation, redox signalling and epithelial turnover, and contribution of luminal microbiota, key aspects that are relevant to understanding redox-dependent processes in gut biology with implications for degenerative digestive disorders, such as inflammation and cancer.
Collapse
Affiliation(s)
- Magdalena L Circu
- Department of Molecular & Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | | |
Collapse
|
37
|
Kelly-Aubert M, Trudel S, Fritsch J, Nguyen-Khoa T, Baudouin-Legros M, Moriceau S, Jeanson L, Djouadi F, Matar C, Conti M, Ollero M, Brouillard F, Edelman A. GSH monoethyl ester rescues mitochondrial defects in cystic fibrosis models. Hum Mol Genet 2011; 20:2745-59. [DOI: 10.1093/hmg/ddr173] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
38
|
Wang Y, Zhang SXL, Gozal D. Reactive oxygen species and the brain in sleep apnea. Respir Physiol Neurobiol 2010; 174:307-16. [PMID: 20833273 DOI: 10.1016/j.resp.2010.09.001] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2010] [Revised: 08/31/2010] [Accepted: 09/01/2010] [Indexed: 02/07/2023]
Abstract
Rodents exposed to intermittent hypoxia (IH), a model of obstructive sleep apnea (OSA), manifest impaired learning and memory and somnolence. Increased levels of reactive oxygen species (ROS), oxidative tissue damage, and apoptotic neuronal cell death are associated with the presence of IH-induced CNS dysfunction. Furthermore, treatment with antioxidants or overexpression of antioxidant enzymes is neuroprotective during IH. These findings mimic clinical cases of OSA and suggest that ROS may play a key causal role in OSA-induced neuropathology. Controlled production of ROS occurs in multiple subcellular compartments of normal cells and de-regulation of such processes may result in excessive ROS production. The mitochondrial electron transport chain, especially complexes I and III, and the NADPH oxidase in the cellular membrane are the two main sources of ROS in brain cells, although other systems, including xanthine oxidase, phospholipase A2, lipoxygenase, cyclooxygenase, and cytochrome P450, may all play a role. The initial evidence for NADPH oxidase and mitochondrial involvement in IH-induced ROS production and neuronal injury unquestionably warrants future research efforts.
Collapse
Affiliation(s)
- Yang Wang
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, The University of Chicago, Chicago, IL 60637, USA
| | | | | |
Collapse
|