1
|
Li Q, Homilius M, Achilles E, Massey LK, Convey V, Ohlsson Å, Ljungvall I, Häggström J, Boler BV, Steiner P, Day S, MacRae CA, Oyama MA. Metabolic abnormalities and reprogramming in cats with naturally occurring hypertrophic cardiomyopathy. ESC Heart Fail 2024. [PMID: 39499136 DOI: 10.1002/ehf2.15135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/15/2024] [Accepted: 10/05/2024] [Indexed: 11/07/2024] Open
Abstract
BACKGROUND AND AIMS The heart is a metabolic organ rich in mitochondria. The failing heart reprograms to utilize different energy substrates, which increase its oxygen consumption. These adaptive changes contribute to increased oxidative stress. Hypertrophic cardiomyopathy (HCM) is a common heart condition, affecting approximately 15% of the general cat population. Feline HCM shares phenotypical and genotypical similarities with human HCM, but the disease mechanisms for both species are incompletely understood. Our goal was to characterize global changes in metabolome between healthy control cats and cats with different stages of HCM. METHODS Serum samples from 83 cats, the majority (70/83) of which were domestic shorthair and included 23 healthy control cats, 31 and 12 preclinical cats with American College of Veterinary Internal Medicine (ACVIM) stages B1 and B2, respectively, and 17 cats with history of clinical heart failure or arterial thromboembolism (ACVIM stage C), were collected for untargeted metabolomic analysis. Multiple linear regression adjusted for age, sex and body weight was applied to compare between control and across HCM groups. RESULTS Our study identified 1253 metabolites, of which 983 metabolites had known identities. Statistical analysis identified 167 metabolites that were significantly different among groups (adjusted P < 0.1). About half of the differentially identified metabolites were lipids, including glycerophospholipids, sphingolipids and cholesterol. Serum concentrations of free fatty acids, 3-hydroxy fatty acids and acylcarnitines were increased in HCM groups compared with control group. The levels of creatine phosphate and multiple Krebs cycle intermediates, including succinate, aconitate and α-ketoglutarate, also accumulated in the circulation of HCM cats. In addition, serum levels of nicotinamide and tryptophan, precursors for de novo NAD+ biosynthesis, were reduced in HCM groups versus control group. Glutathione metabolism was altered. Serum levels of cystine, the oxidized form of cysteine and cysteine-glutathione disulfide, were elevated in the HCM groups, indicative of heightened oxidative stress. Further, the level of ophthalmate, an endogenous glutathione analog and competitive inhibitor, was increased by more than twofold in HCM groups versus control group. Finally, several uremic toxins, including guanidino compounds and protein bound putrescine, accumulated in the circulation of HCM cats. CONCLUSIONS Our study provided evidence of deranged energy metabolism, altered glutathione homeostasis and impaired renal uremic toxin excretion. Altered lipid metabolism suggested perturbed structure and function of cardiac sarcolemma membrane and lipid signalling.
Collapse
Affiliation(s)
- Qinghong Li
- Nestlé Purina Research, St. Louis, Missouri, USA
| | - Max Homilius
- Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Erin Achilles
- Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Laura K Massey
- Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Victoria Convey
- Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Åsa Ohlsson
- Department of Animal Biosciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Ingrid Ljungvall
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Jens Häggström
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | | | | | - Sharlene Day
- Division of Cardiovascular Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Calum A MacRae
- Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Mark A Oyama
- Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
2
|
Kwon YH, Kim JA, Park YS, Kim JH, Choi CY. Effects of red-light irradiation and melatonininjection on the antioxidant capacity and occurrence of apoptosis in abalones (Haliotis discus hannai) subjected to thermal stress. Comp Biochem Physiol A Mol Integr Physiol 2024; 296:111689. [PMID: 38945429 DOI: 10.1016/j.cbpa.2024.111689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/10/2024] [Accepted: 06/27/2024] [Indexed: 07/02/2024]
Abstract
High ocean temperatures caused by global warming induce oxidative stress in aquatic organisms. Melatonin treatment and irradiation using red light-emitting diodes (LEDs) have been reported to reduce oxidative stress in a few aquatic organisms. However, the effects of red LED irradiation and melatonin injection on the antioxidant capacity and degree of apoptosis in abalones, which are nocturnal organisms, have not yet been reported. In this study, we compared the expression levels of antioxidant enzymes, total antioxidant capacity, and the degree of apoptosis in abalones subjected to red LED irradiation and melatonin treatment. The results revealed that at high water temperatures (25 °C), the mRNA expression levels of the superoxide dismutase (SOD) and glutathione peroxidase (GPx) genes and the antioxidant activity of SOD decreased in abalones in the red-LED irradiated and melatonin-treated groups compared with those in abalones in the control group. Although high water temperatures induced DNA damage in the abalone samples, the degree of apoptosis was lower in the red-LED irradiated and melatonin-treated groups than in the control group. Overall, the abalones in the melatonin-treated and red-LED irradiated groups showed reduced oxidative stress and increased antioxidant enzyme levels under thermal stress compared with those in the control group. Therefore, red LED irradiation is a promising alternative to melatonin treatment, which is difficult to administer continuously for a long time, for protecting abalones from oxidative stress.
Collapse
Affiliation(s)
- Young Hoon Kwon
- Department of Convergence Study on the Ocean Science and Technology, National Korea Maritime and Ocean University, Busan 49112, Republic of Korea; Division of Marine BioScience, National Korea Maritime and Ocean University, Busan 49112, Republic of Korea
| | - Jin A Kim
- Department of Convergence Study on the Ocean Science and Technology, National Korea Maritime and Ocean University, Busan 49112, Republic of Korea
| | - Young-Su Park
- Department of Nursing, Catholic University of Pusan, Busan 46252, Republic of Korea
| | - Jun-Hwan Kim
- Department of Aquatic Life Medicine, Jeju National University, Jeju 63243, Republic of Korea.
| | - Cheol Young Choi
- Department of Convergence Study on the Ocean Science and Technology, National Korea Maritime and Ocean University, Busan 49112, Republic of Korea; Division of Marine BioScience, National Korea Maritime and Ocean University, Busan 49112, Republic of Korea.
| |
Collapse
|
3
|
Zhang W, Ou Z, Tang T, Yang T, Li Y, Wu H, Li L, Liu M, Niu L, Zhu J. Up-regulated SLC25A39 promotes cell growth and metastasis via regulating ROS production in colorectal cancer. J Cancer 2024; 15:5841-5854. [PMID: 39308681 PMCID: PMC11414614 DOI: 10.7150/jca.98844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 08/30/2024] [Indexed: 09/25/2024] Open
Abstract
Background: The mitochondrial transporter SLC25A39 has been implicated in the import of mitochondrial glutathione (mGSH) from the cytoplasm, crucial for mitigating oxidative stress and preserving mitochondrial function. Despite the well-established involvement of mitochondria in cancer, the functional impact of SLC25A39 on CRC progression remains elusive. Methods: The mRNA and protein expressions were detected by PCR, immunohistochemistry, and Western blot, respectively. Cell activity, cell proliferation, colony formation, and apoptosis were measured by CCK8 assay, EdU incorporation assay, plated colony formation assay, and flow cytometry, respectively. Cell migration was detected by wound healing and transwell chamber assay. The tumor microenvironment (TME), immune checkpoint molecules, and drug sensitivity of CRC patients were investigated using R language, GraphPad Prism 8 and online databases. Results: Here, we report a significant upregulation of SLC25A39 expression in CRC. Functional assays revealed that overexpression of SLC25A39 promoted CRC cell proliferation and migration while inhibiting apoptosis. Conversely, SLC25A39 knockdown suppressed cell growth and migration while enhancing apoptosis in vitro. Additionally, reduced SLC25A39 expression attenuated tumor growth in xenograft models. Mechanistically, elevated SLC25A39 levels correlated with reduced reactive oxygen species (ROS) accumulation in CRC. Furthermore, bioinformatic analyses unveiled the high SLC25A39 levels was associated with decreased expression of immune checkpoints and reduced responsiveness to immunotherapy. Single-cell transcriptomic profiling identified diverse cellular expression patterns of SLC25A39 and related immune regulators. Lastly, drug sensitivity analysis indicated potential therapeutic avenues targeting SLC25A39 in CRC. Conclusion Our findings underscore the pivotal role of SLC25A39 in CRC progression and suggest its candidacy as a therapeutic target in CRC management.
Collapse
Affiliation(s)
- Wentao Zhang
- Department of Medical Cellular Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Zhigao Ou
- Department of Medical Cellular Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ting Tang
- Department of Medical Cellular Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Tian Yang
- Department of Medical Cellular Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yubo Li
- Department of Medical Cellular Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Hao Wu
- Department of Medical Cellular Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Li Li
- Department of Medical Cellular Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ming Liu
- Department of Medical Cellular Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Li Niu
- Department of Pathophysiology, School of Basic Medical Science, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jianjun Zhu
- Department of Medical Cellular Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
4
|
Liu J, Zuo S. Control of mitochondrial glutathione homeostasis by SLC25A39. Acta Biochim Biophys Sin (Shanghai) 2024; 56:1093-1095. [PMID: 38766696 PMCID: PMC11322872 DOI: 10.3724/abbs.2024072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 04/25/2024] [Indexed: 05/22/2024] Open
Affiliation(s)
- Jiao Liu
- Department of BiopharmaceuticsThe Province and Ministry Co-sponsored Collaborative Innovation Center for Medical EpigeneticsTianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and DiagnosticsSchool of PharmacyTianjin Medical UniversityTianjin300070China
- Department of PharmacologyTianjin Key Laboratory of Inflammatory BiologySchool of Basic Medical SciencesTianjin Medical UniversityTianjin300070China
| | - Shengkai Zuo
- Department of BiopharmaceuticsThe Province and Ministry Co-sponsored Collaborative Innovation Center for Medical EpigeneticsTianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and DiagnosticsSchool of PharmacyTianjin Medical UniversityTianjin300070China
| |
Collapse
|
5
|
Naidu AS, Wang CK, Rao P, Mancini F, Clemens RA, Wirakartakusumah A, Chiu HF, Yen CH, Porretta S, Mathai I, Naidu SAG. Precision nutrition to reset virus-induced human metabolic reprogramming and dysregulation (HMRD) in long-COVID. NPJ Sci Food 2024; 8:19. [PMID: 38555403 PMCID: PMC10981760 DOI: 10.1038/s41538-024-00261-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 03/15/2024] [Indexed: 04/02/2024] Open
Abstract
SARS-CoV-2, the etiological agent of COVID-19, is devoid of any metabolic capacity; therefore, it is critical for the viral pathogen to hijack host cellular metabolic machinery for its replication and propagation. This single-stranded RNA virus with a 29.9 kb genome encodes 14 open reading frames (ORFs) and initiates a plethora of virus-host protein-protein interactions in the human body. These extensive viral protein interactions with host-specific cellular targets could trigger severe human metabolic reprogramming/dysregulation (HMRD), a rewiring of sugar-, amino acid-, lipid-, and nucleotide-metabolism(s), as well as altered or impaired bioenergetics, immune dysfunction, and redox imbalance in the body. In the infectious process, the viral pathogen hijacks two major human receptors, angiotensin-converting enzyme (ACE)-2 and/or neuropilin (NRP)-1, for initial adhesion to cell surface; then utilizes two major host proteases, TMPRSS2 and/or furin, to gain cellular entry; and finally employs an endosomal enzyme, cathepsin L (CTSL) for fusogenic release of its viral genome. The virus-induced HMRD results in 5 possible infectious outcomes: asymptomatic, mild, moderate, severe to fatal episodes; while the symptomatic acute COVID-19 condition could manifest into 3 clinical phases: (i) hypoxia and hypoxemia (Warburg effect), (ii) hyperferritinemia ('cytokine storm'), and (iii) thrombocytosis (coagulopathy). The mean incubation period for COVID-19 onset was estimated to be 5.1 days, and most cases develop symptoms after 14 days. The mean viral clearance times were 24, 30, and 39 days for acute, severe, and ICU-admitted COVID-19 patients, respectively. However, about 25-70% of virus-free COVID-19 survivors continue to sustain virus-induced HMRD and exhibit a wide range of symptoms that are persistent, exacerbated, or new 'onset' clinical incidents, collectively termed as post-acute sequelae of COVID-19 (PASC) or long COVID. PASC patients experience several debilitating clinical condition(s) with >200 different and overlapping symptoms that may last for weeks to months. Chronic PASC is a cumulative outcome of at least 10 different HMRD-related pathophysiological mechanisms involving both virus-derived virulence factors and a multitude of innate host responses. Based on HMRD and virus-free clinical impairments of different human organs/systems, PASC patients can be categorized into 4 different clusters or sub-phenotypes: sub-phenotype-1 (33.8%) with cardiac and renal manifestations; sub-phenotype-2 (32.8%) with respiratory, sleep and anxiety disorders; sub-phenotype-3 (23.4%) with skeleto-muscular and nervous disorders; and sub-phenotype-4 (10.1%) with digestive and pulmonary dysfunctions. This narrative review elucidates the effects of viral hijack on host cellular machinery during SARS-CoV-2 infection, ensuing detrimental effect(s) of virus-induced HMRD on human metabolism, consequential symptomatic clinical implications, and damage to multiple organ systems; as well as chronic pathophysiological sequelae in virus-free PASC patients. We have also provided a few evidence-based, human randomized controlled trial (RCT)-tested, precision nutrients to reset HMRD for health recovery of PASC patients.
Collapse
Affiliation(s)
- A Satyanarayan Naidu
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA.
- N-terminus Research Laboratory, 232659 Via del Rio, Yorba Linda, CA, 92887, USA.
| | - Chin-Kun Wang
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA
- School of Nutrition, Chung Shan Medical University, 110, Section 1, Jianguo North Road, Taichung, 40201, Taiwan
| | - Pingfan Rao
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA
- College of Food and Bioengineering, Fujian Polytechnic Normal University, No.1, Campus New Village, Longjiang Street, Fuqing City, Fujian, China
| | - Fabrizio Mancini
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA
- President-Emeritus, Parker University, 2540 Walnut Hill Lane, Dallas, TX, 75229, USA
| | - Roger A Clemens
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA
- University of Southern California, Alfred E. Mann School of Pharmacy/D. K. Kim International Center for Regulatory & Quality Sciences, 1540 Alcazar St., CHP 140, Los Angeles, CA, 90089, USA
| | - Aman Wirakartakusumah
- International Union of Food Science and Technology (IUFoST), Guelph, ON, Canada
- IPMI International Business School Jakarta; South East Asian Food and Agriculture Science and Technology, IPB University, Bogor, Indonesia
| | - Hui-Fang Chiu
- Department of Chinese Medicine, Taichung Hospital, Ministry of Health & Well-being, Taichung, Taiwan
| | - Chi-Hua Yen
- Department of Family and Community Medicine, Chung Shan Medical University Hospital; School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Sebastiano Porretta
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA
- President, Italian Association of Food Technology (AITA), Milan, Italy
- Experimental Station for the Food Preserving Industry, Department of Consumer Science, Viale Tanara 31/a, I-43121, Parma, Italy
| | - Issac Mathai
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA
- Soukya International Holistic Health Center, Whitefield, Bengaluru, India
| | - Sreus A G Naidu
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA
- N-terminus Research Laboratory, 232659 Via del Rio, Yorba Linda, CA, 92887, USA
| |
Collapse
|
6
|
Shi X, DeCiucis M, Grabinska KA, Kanyo J, Liu A, Lam TT, Shen H. Dual regulation of SLC25A39 by AFG3L2 and iron controls mitochondrial glutathione homeostasis. Mol Cell 2024; 84:802-810.e6. [PMID: 38157846 PMCID: PMC10922821 DOI: 10.1016/j.molcel.2023.12.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/02/2023] [Accepted: 12/08/2023] [Indexed: 01/03/2024]
Abstract
Organelle transporters define metabolic compartmentalization, and how this metabolite transport process can be modulated is poorly explored. Here, we discovered that human SLC25A39, a mitochondrial transporter critical for mitochondrial glutathione uptake, is a short-lived protein under dual regulation at the protein level. Co-immunoprecipitation mass spectrometry and CRISPR knockout (KO) in mammalian cells identified that mitochondrial m-AAA protease AFG3L2 is responsible for degrading SLC25A39 through the matrix loop 1. SLC25A39 senses mitochondrial iron-sulfur cluster using four matrix cysteine residues and inhibits its degradation. SLC25A39 protein regulation is robust in developing and mature neurons. This dual transporter regulation, by protein quality control and metabolic sensing, allows modulating mitochondrial glutathione level in response to iron homeostasis, opening avenues for exploring regulation of metabolic compartmentalization. Neuronal SLC25A39 regulation connects mitochondrial protein quality control, glutathione, and iron homeostasis, which were previously unrelated biochemical features in neurodegeneration.
Collapse
Affiliation(s)
- Xiaojian Shi
- Cellular and Molecular Physiology Department, Yale School of Medicine, New Haven, CT, USA; Systems Biology Institute, Yale West Campus, West Haven, CT, USA
| | - Marisa DeCiucis
- Cellular and Molecular Physiology Department, Yale School of Medicine, New Haven, CT, USA; Systems Biology Institute, Yale West Campus, West Haven, CT, USA
| | - Kariona A Grabinska
- Cellular and Molecular Physiology Department, Yale School of Medicine, New Haven, CT, USA; Systems Biology Institute, Yale West Campus, West Haven, CT, USA
| | - Jean Kanyo
- Keck MS & Proteomics Resource, Yale School of Medicine, New Haven, CT, USA
| | - Adam Liu
- Amity High School, Woodbridge, CT, USA
| | - Tukiet T Lam
- Keck MS & Proteomics Resource, Yale School of Medicine, New Haven, CT, USA; Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
| | - Hongying Shen
- Cellular and Molecular Physiology Department, Yale School of Medicine, New Haven, CT, USA; Systems Biology Institute, Yale West Campus, West Haven, CT, USA.
| |
Collapse
|
7
|
Chen X, Gan B. SLC25A39 links mitochondrial GSH sensing with iron metabolism. Mol Cell 2024; 84:616-618. [PMID: 38364779 DOI: 10.1016/j.molcel.2023.12.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 12/21/2023] [Accepted: 12/21/2023] [Indexed: 02/18/2024]
Abstract
Two recent studies by Liu et al.1 in Science and Shi et al.2 in this issue of Molecular Cell identify a mitochondrial GSH-sensing mechanism that couples SLC25A39-mediated GSH import to iron metabolism, advancing our understanding of nutrient sensing within organelles.
Collapse
Affiliation(s)
- Xiong Chen
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Boyi Gan
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.
| |
Collapse
|
8
|
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: 25] [Impact Index Per Article: 25.0] [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
|
9
|
Gao R, Zhou D, Qiu X, Zhang J, Luo D, Yang X, Qian C, Liu Z. Cancer Therapeutic Potential and Prognostic Value of the SLC25 Mitochondrial Carrier Family: A Review. Cancer Control 2024; 31:10732748241287905. [PMID: 39313442 PMCID: PMC11439189 DOI: 10.1177/10732748241287905] [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: 05/23/2024] [Revised: 08/29/2024] [Accepted: 09/10/2024] [Indexed: 09/25/2024] Open
Abstract
Transporters of the solute carrier family 25 (SLC25) regulate the intracellular distribution and concentration of nucleotides, amino acids, dicarboxylates, and vitamins within the mitochondrial and cytoplasmic matrices. This mechanism involves changes in mitochondrial function, regulation of cellular metabolism, and the ability to provide energy. In this review, important members of the SLC25 family and their pathways affecting tumorigenesis and progression are elucidated, highlighting the diversity and complexity of these pathways. Furthermore, the significant potential of the members of SLC25 as both cancer therapeutic targets and biomarkers will be emphasized.
Collapse
Affiliation(s)
- Renzhuo Gao
- School of Queen Mary, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Dan Zhou
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xingpeng Qiu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Jiayi Zhang
- School of Queen Mary, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Daya Luo
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xiaohong Yang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Caiyun Qian
- Department of Blood Transfusion, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Zhuoqi Liu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
10
|
Hernandez-Baixauli J, Chomiciute G, Tracey H, Mora I, Cortés-Espinar AJ, Ávila-Román J, Abasolo N, Palacios-Jordan H, Foguet-Romero E, Suñol D, Galofré M, Alcaide-Hidalgo JM, Baselga-Escudero L, del Bas JM, Mulero M. Exploring Metabolic and Gut Microbiome Responses to Paraquat Administration in Male Wistar Rats: Implications for Oxidative Stress. Antioxidants (Basel) 2024; 13:67. [PMID: 38247491 PMCID: PMC10812659 DOI: 10.3390/antiox13010067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/22/2023] [Accepted: 12/27/2023] [Indexed: 01/23/2024] Open
Abstract
In this study, we examined the metabolic and gut microbiome responses to paraquat (PQ) in male Wistar rats, focusing on oxidative stress effects. Rats received a single intraperitoneal injection of PQ at 15 and 30 mg/kg, and various oxidative stress parameters (i.e., MDA, SOD, ROS, 8-isoprostanes) were assessed after three days. To explore the omic profile, GC-qTOF and UHPLC-qTOF were performed to assess the plasma metabolome; 1H-NMR was used to assess the urine metabolome; and shotgun metagenomics sequencing was performed to study the gut microbiome. Our results revealed reductions in body weight and tissue changes, particularly in the liver, were observed, suggesting a systemic effect of PQ. Elevated lipid peroxidation and reactive oxygen species levels in the liver and plasma indicated the induction of oxidative stress. Metabolic profiling revealed changes in the tricarboxylic acid cycle, accumulation of ketone body, and altered levels of key metabolites, such as 3-hydroxybutyric acid and serine, suggesting intricate links between energy metabolism and redox reactions. Plasma metabolomic analysis revealed alterations in mitochondrial metabolism, nicotinamide metabolism, and tryptophan degradation. The gut microbiome showed shifts, with higher PQ doses influencing microbial populations (e.g., Escherichia coli and Akkermansia muciniphila) and metagenomic functions (pyruvate metabolism, fermentation, nucleotide and amino acid biosynthesis). Overall, this study provides comprehensive insights into the complex interplay between PQ exposure, metabolic responses, and gut microbiome dynamics. These findings enhance our understanding of the mechanisms behind oxidative stress-induced metabolic alterations and underscore the connections between xenobiotic exposure, gut microbiota, and host metabolism.
Collapse
Affiliation(s)
- Julia Hernandez-Baixauli
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain; (J.H.-B.); (G.C.); (H.T.); (J.M.A.-H.); (L.B.-E.)
- Laboratory of Metabolism and Obesity, Vall d’Hebron-Institut de Recerca, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Gertruda Chomiciute
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain; (J.H.-B.); (G.C.); (H.T.); (J.M.A.-H.); (L.B.-E.)
| | - Harry Tracey
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain; (J.H.-B.); (G.C.); (H.T.); (J.M.A.-H.); (L.B.-E.)
- Department of Medical Sciences, School of Medicine, University of Girona, 17004 Girona, Spain
- School of Science, RMIT University, Bundoora, VIC 3000, Australia
| | - Ignasi Mora
- Brudy Technology S.L., 08006 Barcelona, Spain;
| | - Antonio J. Cortés-Espinar
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, 43007 Tarragona, Spain;
| | - Javier Ávila-Román
- Molecular and Applied Pharmacology Group (FARMOLAP), Department of Pharmacology, Universidad de Sevilla, 41012 Sevilla, Spain;
| | - Nerea Abasolo
- Eurecat, Centre Tecnològic de Catalunya, Centre for Omic Sciences (COS), Joint Unit Universitat Rovira i Virgili-EURECAT, 43204 Reus, Spain; (N.A.); (H.P.-J.); (E.F.-R.)
| | - Hector Palacios-Jordan
- Eurecat, Centre Tecnològic de Catalunya, Centre for Omic Sciences (COS), Joint Unit Universitat Rovira i Virgili-EURECAT, 43204 Reus, Spain; (N.A.); (H.P.-J.); (E.F.-R.)
| | - Elisabet Foguet-Romero
- Eurecat, Centre Tecnològic de Catalunya, Centre for Omic Sciences (COS), Joint Unit Universitat Rovira i Virgili-EURECAT, 43204 Reus, Spain; (N.A.); (H.P.-J.); (E.F.-R.)
| | - David Suñol
- Eurecat, Centre Tecnològic de Catalunya, Digital Health, 08005 Barcelona, Spain; (D.S.); (M.G.)
| | - Mar Galofré
- Eurecat, Centre Tecnològic de Catalunya, Digital Health, 08005 Barcelona, Spain; (D.S.); (M.G.)
| | - Juan María Alcaide-Hidalgo
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain; (J.H.-B.); (G.C.); (H.T.); (J.M.A.-H.); (L.B.-E.)
| | - Laura Baselga-Escudero
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain; (J.H.-B.); (G.C.); (H.T.); (J.M.A.-H.); (L.B.-E.)
| | - Josep M. del Bas
- Eurecat, Centre Tecnològic de Catalunya, Àrea Biotecnologia, 43204 Reus, Spain
| | - Miquel Mulero
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, 43007 Tarragona, Spain;
| |
Collapse
|
11
|
Shen R, Ardianto C, Celia C, Sidharta VM, Sasmita PK, Satriotomo I, Turana Y. Brain-derived neurotrophic factor interplay with oxidative stress: neuropathology approach in potential biomarker of Alzheimer's disease. Dement Neuropsychol 2023; 17:e20230012. [PMID: 38053647 PMCID: PMC10695442 DOI: 10.1590/1980-5764-dn-2023-0012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/05/2023] [Accepted: 07/30/2023] [Indexed: 12/07/2023] Open
Abstract
The aging population poses a serious challenge concerning an increased prevalence of Alzheimer's disease (AD) and its impact on global burden, morbidity, and mortality. Oxidative stress, as a molecular hallmark that causes susceptibility in AD, interplays to other AD-related neuropathology cascades and decreases the expression of central and circulation brain-derived neurotrophic factor (BDNF), an essential neurotrophin that serves as nerve development and survival, and synaptic plasticity in AD. By its significant correlation with the molecular and clinical progression of AD, BDNF can potentially be used as an objectively accurate biomarker for AD diagnosis and progressivity follow-up in future clinical practice. This comprehensive review highlights the oxidative stress interplay with BDNF in AD neuropathology and its potential use as an AD biomarker.
Collapse
Affiliation(s)
- Robert Shen
- Atma Jaya Catholic University of Indonesia, School of Medicine and Health Sciences, Jakarta, Indonesia
| | - Christian Ardianto
- Atma Jaya Catholic University of Indonesia, School of Medicine and Health Sciences, Jakarta, Indonesia
| | - Celia Celia
- Atma Jaya Catholic University of Indonesia, School of Medicine and Health Sciences, Jakarta, Indonesia
| | - Veronika Maria Sidharta
- Atma Jaya Catholic University of Indonesia, School of Medicine and Health Sciences, Jakarta, Indonesia
| | - Poppy Kristina Sasmita
- Atma Jaya Catholic University of Indonesia, School of Medicine and Health Sciences, Jakarta, Indonesia
| | - Irawan Satriotomo
- University of Florida, Gainesville, Department of Neurology, Florida, USA
- Satriotomo Foundation, Indonesia Neuroscience Institute, Jakarta, Indonesia
| | - Yuda Turana
- Atma Jaya Catholic University of Indonesia, School of Medicine and Health Sciences, Jakarta, Indonesia
| |
Collapse
|
12
|
Prange CJ, Hu X, Tang L. Smart chemistry for traceless release of anticancer therapeutics. Biomaterials 2023; 303:122353. [PMID: 37925794 DOI: 10.1016/j.biomaterials.2023.122353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 10/10/2023] [Accepted: 10/12/2023] [Indexed: 11/07/2023]
Abstract
In the design of delivery strategies for anticancer therapeutics, the controlled release of intact cargo at the destined tumor and metastasis locations is of particular importance. To this end, stimuli-responsive chemical linkers have been extensively investigated owing to their ability to respond to tumor-specific physiological stimuli, such as lowered pH, altered redox conditions, increased radical oxygen species and pathological enzymatic activities. To prevent premature action and off-target effects, anticancer therapeutics are chemically modified to be transiently inactivated, a strategy known as prodrug development. Prodrugs are reactivated upon stimuli-dependent release at the sites of interest. As most drugs and therapeutic proteins have the optimal activity when released from carriers in their native and original forms, traceless release mechanisms are increasingly investigated. In this review, we summarize the chemical toolkit for developing innovative traceless prodrug strategies for stimuli-responsive drug delivery and discuss the applications of these chemical modifications in anticancer treatment including cancer immunotherapy.
Collapse
Affiliation(s)
- Céline Jasmin Prange
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland; Institute of Chemical Sciences and Engineering, EPFL, Lausanne, CH-1015, Switzerland
| | - Xile Hu
- Institute of Chemical Sciences and Engineering, EPFL, Lausanne, CH-1015, Switzerland.
| | - Li Tang
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland; Institute of Materials Science & Engineering, EPFL, Lausanne, CH-1015, Switzerland.
| |
Collapse
|
13
|
Kim J, Gong YX, Jeong EM. Measuring Glutathione Regeneration Capacity in Stem Cells. Int J Stem Cells 2023; 16:356-362. [PMID: 37385637 PMCID: PMC10465335 DOI: 10.15283/ijsc23047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 04/25/2023] [Accepted: 05/06/2023] [Indexed: 07/01/2023] Open
Abstract
Glutathione (GSH) is a chief cellular antioxidant, affecting stem cell functions. The cellular GSH level is dynamically altered by the redox buffering system and transcription factors, including NRF2. Additionally, GSH is differentially regulated in each organelle. We previously reported a protocol for monitoring the real-time GSH levels in live stem cells using the reversible GSH sensor FreSHtracer. However, GSH-based stem cell analysis needs be comprehensive and organelle-specific. Hence, in this study, we demonstrate a detailed protocol to measure the GSH regeneration capacity (GRC) in living stem cells by measuring the intensities of the FreSHtracer and the mitochondrial GSH sensor MitoFreSHtracer using a high-content screening confocal microscope. This protocol typically analyses the GRC in approximately 4 h following the seeding of the cells onto plates. This protocol is simple and quantitative. With some minor modifications, it can be employed flexibly to measure the GRC for the whole-cell area or just the mitochondria in all adherent mammalian stem cells.
Collapse
Affiliation(s)
- Jihye Kim
- Department of Pharmacy, College of Pharmacy, Jeju Research Institute of Pharmaceutical Sciences, Jeju National University, Jeju, Korea
| | - Yi-Xi Gong
- Department of Pharmacy, College of Pharmacy, Jeju Research Institute of Pharmaceutical Sciences, Jeju National University, Jeju, Korea
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Bio-Health Materials Core-Facility Center and Practical Translational Research Center, Jeju National University, Jeju, Korea
| | - Eui Man Jeong
- Department of Pharmacy, College of Pharmacy, Jeju Research Institute of Pharmaceutical Sciences, Jeju National University, Jeju, Korea
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Bio-Health Materials Core-Facility Center and Practical Translational Research Center, Jeju National University, Jeju, Korea
| |
Collapse
|
14
|
Li F, Chu Q, Hu Z, Lu Z, Fang C, Han G, Fu Y, Li X. An Inter-Cooperative Biohybrid Platform to Enable Tumor Ablation and Immune Activation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207194. [PMID: 37314157 PMCID: PMC10427385 DOI: 10.1002/advs.202207194] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 05/03/2023] [Indexed: 06/15/2023]
Abstract
A biohybrid therapeutic system, consisting of responsive materials and living microorganisms with inter-cooperative effects, is designed and investigated for tumor treatment. In this biohybrid system, S2 O3 2- -intercalated CoFe layered double hydroxides (LDH) are integrated at the surface of Baker's yeasts. Under the tumor microenvironment, functional interactions between yeast and LDH are effectively triggered, resulting in S2 O3 2- release, H2 S production, and in-situ generation of highly catalytic agents. Meanwhile, the degradation of LDH in the tumor microenvironment induces the exposure of the surface antigen of yeast, leading to effective immune activation at the tumor site. By virtue of the inter-cooperative phenomena, this biohybrid system exhibits significant efficacy in tumor ablation and strong inhibition of recurrence. This study has potentially offered an alternative concept by utilizing the metabolism of living microorganisms and materials in exploring effective tumor therapeutics.
Collapse
Affiliation(s)
- Feiyu Li
- State Key Laboratory of Silicon and Advanced Semiconductor MaterialsSchool of Materials Science and EngineeringZhejiang UniversityHangzhou310027China
- ZJU‐Hangzhou Global Science and Technology Innovation CenterZhejiang UniversityHangzhou311215China
| | - Qiang Chu
- Tea Research InstituteCollege of Agriculture and BiotechnologyZhejiang UniversityHangzhou310058China
| | - Zefeng Hu
- State Key Laboratory of Silicon and Advanced Semiconductor MaterialsSchool of Materials Science and EngineeringZhejiang UniversityHangzhou310027China
- ZJU‐Hangzhou Global Science and Technology Innovation CenterZhejiang UniversityHangzhou311215China
| | - Zijie Lu
- State Key Laboratory of Silicon and Advanced Semiconductor MaterialsSchool of Materials Science and EngineeringZhejiang UniversityHangzhou310027China
| | - Chao Fang
- State Key Laboratory of Silicon and Advanced Semiconductor MaterialsSchool of Materials Science and EngineeringZhejiang UniversityHangzhou310027China
| | - Gaorong Han
- State Key Laboratory of Silicon and Advanced Semiconductor MaterialsSchool of Materials Science and EngineeringZhejiang UniversityHangzhou310027China
| | - Yike Fu
- State Key Laboratory of Silicon and Advanced Semiconductor MaterialsSchool of Materials Science and EngineeringZhejiang UniversityHangzhou310027China
- ZJU‐Hangzhou Global Science and Technology Innovation CenterZhejiang UniversityHangzhou311215China
| | - Xiang Li
- State Key Laboratory of Silicon and Advanced Semiconductor MaterialsSchool of Materials Science and EngineeringZhejiang UniversityHangzhou310027China
- ZJU‐Hangzhou Global Science and Technology Innovation CenterZhejiang UniversityHangzhou311215China
| |
Collapse
|
15
|
Meng L, Wu G. Recent advances in small molecules for improving mitochondrial disorders. RSC Adv 2023; 13:20476-20485. [PMID: 37435377 PMCID: PMC10331567 DOI: 10.1039/d3ra03313a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/03/2023] [Indexed: 07/13/2023] Open
Abstract
Mitochondrial disorders are observed in various human diseases, including rare genetic disorders and complex acquired pathologies. Recent advances in molecular biological techniques have dramatically expanded the understanding of multiple pathomechanisms involving mitochondrial disorders. However, the therapeutic methods for mitochondrial disorders are limited. For this reason, there is increasing interest in identifying safe and effective strategies to mitigate mitochondrial impairments. Small-molecule therapies hold promise for improving mitochondrial performance. This review focuses on the latest advances in developing bioactive compounds for treating mitochondrial disease, aiming to provide a broader perspective of fundamental studies that have been carried out to evaluate the effects of small molecules in regulating mitochondrial function. Novel-designed small molecules ameliorating mitochondrial functions are urgent for further research.
Collapse
Affiliation(s)
- Liying Meng
- Department of Central Laboratory and Mitochondrial Medicine Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University Qingdao China
| | - Guanzhao Wu
- Department of Central Laboratory and Mitochondrial Medicine Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University Qingdao China
| |
Collapse
|
16
|
Cuong Tran NK, Jeong JH, Sharma N, Doan Nguyen YN, Phi Tran HY, Dang DK, Park JH, Byun JK, Jin D, Xiaoyan Z, Ko SK, Nah SY, Kim HC, Shin EJ. Ginsenoside Re blocks Bay k-8644-induced neurotoxicity via attenuating mitochondrial dysfunction and PKCδ activation in the hippocampus of mice: Involvement of antioxidant potential. Food Chem Toxicol 2023:113869. [PMID: 37308051 DOI: 10.1016/j.fct.2023.113869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/01/2023] [Indexed: 06/14/2023]
Abstract
Although the anticonvulsant effects of ginsenosides are recognized, little is known about their effects on the convulsive behaviors induced by the activation of L-type Ca2+ channels. Here, we investigated whether ginsenoside Re (GRe) modulates excitotoxicity induced by the L-type Ca2+ channel activator Bay k-8644. GRe significantly attenuated Bay k-8644-induced convulsive behaviors and hippocampal oxidative stress in mice. GRe-mediated antioxidant potential was more pronounced in the mitochondrial fraction than cytosolic fraction. As L-type Ca2+ channels are thought to be targets of protein kinase C (PKC), we investigated the role of PKC under excitotoxic conditions. GRe attenuated Bay k-8644-induced mitochondrial dysfunction, PKCδ activation, and neuronal loss. The PKCδ inhibition and neuroprotection mediated by GRe were comparable to those by the ROS inhibitor N-acetylcysteine, the mitochondrial protectant cyclosporin A, the microglial inhibitor minocycline, or the PKCδ inhibitor rottlerin. Consistently, the GRe-mediated PKCδ inhibition and neuroprotection were counteracted by the mitochondrial toxin 3-nitropropionic acid or the PKC activator bryostatin-1. GRe treatment did not have additional effects on PKCδ gene knockout-mediated neuroprotection, suggesting that PKCδ is a molecular target of GRe. Collectively, our results suggest that GRe-mediated anticonvulsive/neuroprotective effects require the attenuation of mitochondrial dysfunction and altered redox status and inactivation of PKCδ.
Collapse
Affiliation(s)
- Ngoc Kim Cuong Tran
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea.
| | - Ji Hoon Jeong
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, College of Medicine, Chung-Ang University, Seoul, 06974, Republic of Korea.
| | - Naveen Sharma
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, College of Medicine, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Yen Nhi Doan Nguyen
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Hoang-Yen Phi Tran
- Physical Chemistry Department, University of Medicine and Pharmacy, Ho Chi Minh City, 760000, Viet Nam
| | - Duy-Khanh Dang
- Pharmacy Faculty, Can Tho University of Medicine and Pharmacy, Can Tho City, 900000, Viet Nam
| | - Jung Hoon Park
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Jae Kyung Byun
- Korea Society of Forest Environmental Research, Namyangju, 12106, Republic of Korea
| | - Dezhong Jin
- Department of Oriental Medical Food & Nutrition, Semyung University, Jecheon, 27316, Republic of Korea
| | - Zeng Xiaoyan
- Department of Oriental Medical Food & Nutrition, Semyung University, Jecheon, 27316, Republic of Korea
| | - Sung Kwon Ko
- Department of Oriental Medical Food & Nutrition, Semyung University, Jecheon, 27316, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, 05029, Republic of Korea
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea.
| | - Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea.
| |
Collapse
|
17
|
Zhen C, Li J, Liu J, Lyu Y, Xie L, Lv H. Phenethyl isothiocyanate induces oxidative cell death in osteosarcoma cells with regulation on mitochondrial network, function and metabolism. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166740. [PMID: 37142133 DOI: 10.1016/j.bbadis.2023.166740] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 04/11/2023] [Accepted: 04/27/2023] [Indexed: 05/06/2023]
Abstract
Phenethyl isothiocyanate (PEITC), a kind of isothiocyanate available in cruciferous vegetables, exhibits inhibitory effects on cancers. PEITC has been extensively recorded for its effect on regulation of redox status in cancer cells. Our previous studies revealed that PEITC induced ROS-dependent cell death in osteosarcoma. Mitochondria are the main sites for ROS generation and play significant role in deciding cell fate. To dissect the mechanism of PEITC's action on osteosarcoma cells, we detected changes on mitochondrial network, function and metabolism in K7M2 and 143B cells. Here, PEITC induced cytosolic, lipid and mitochondrial ROS production in osteosarcoma cells. It changed mitochondrial morphology from elongated to punctate network and decreased mitochondrial mass. Meantime, PEITC increased mitochondrial transmembrane potential in short time, decreased it with time prolonged, and later collapsed it in K7M2 cells, and reduced it in 143B cells. PEITC inhibited proliferation potential of osteosarcoma cells with damage on mitochondrial respiratory chain complexes. Further, PEITC-treated osteosarcoma cells experienced a sudden increase in ATP level, and later its content was decreased. Moreover, PEITC downregulated the expressions of mitochondrial respiratory chain complexes including COX IV, UQCR, SDHA and NDUFA9 in 143B cells and COX IV in K7M2 cells. At last, by using Rho 0 cells derived from K7M2 and 143B cells, we found that osteosarcoma cells that depleted mtDNA were less sensitive to PEITC-induced changes on cellular morphology, cytoskeleton filament, mitochondrial transmembrane potential and ROS generation. In conclusion, our study demonstrated that mitochondria may play important role in PEITC-induced oxidative cell death in osteosarcoma cells.
Collapse
Affiliation(s)
- Chenxiao Zhen
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China; Key Laboratory for Space Bioscience and Biotechnology, Northwestern Polytechnical University, Xi'an, China
| | - Jindou Li
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Junyu Liu
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China; Key Laboratory for Space Bioscience and Biotechnology, Northwestern Polytechnical University, Xi'an, China
| | - Yi Lyu
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China; Key Laboratory for Space Bioscience and Biotechnology, Northwestern Polytechnical University, Xi'an, China
| | - Li Xie
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China; Key Laboratory for Space Bioscience and Biotechnology, Northwestern Polytechnical University, Xi'an, China
| | - Huanhuan Lv
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China; Key Laboratory for Space Bioscience and Biotechnology, Northwestern Polytechnical University, Xi'an, China.
| |
Collapse
|
18
|
da Silva MC, Fabiano LC, da Costa Salomão KC, de Freitas PLZ, Neves CQ, Borges SC, de Souza Carvalho MDG, Breithaupt-Faloppa AC, de Thomaz AA, Dos Santos AM, Buttow NC. A Rodent Model of Human-Dose-Equivalent 5-Fluorouracil: Toxicity in the Liver, Kidneys, and Lungs. Antioxidants (Basel) 2023; 12:antiox12051005. [PMID: 37237871 DOI: 10.3390/antiox12051005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
5-Fluorouracil (5-FU) is a chemotherapy drug widely used to treat a range of cancer types, despite the recurrence of adverse reactions. Therefore, information on its side effects when administered at a clinically recommended dose is relevant. On this basis, we examined the effects of the 5-FU clinical treatment on the integrity of the liver, kidneys, and lungs of rats. For this purpose, 14 male Wistar rats were divided into treated and control groups and 5-FU was administered at 15 mg/kg (4 consecutive days), 6 mg/kg (4 alternate days), and 15 mg/kg on the 14th day. On the 15th day, blood, liver, kidney, and lung samples were collected for histological, oxidative stress, and inflammatory evaluations. We observed a reduction in the antioxidant markers and an increase in lipid hydroperoxides (LOOH) in the liver of treated animals. We also detected elevated levels of inflammatory markers, histological lesions, apoptotic cells, and aspartate aminotransferase. Clinical treatment with 5-FU did not promote inflammatory or oxidative alterations in the kidney samples; however, histological and biochemical changes were observed, including increased serum urea and uric acid. 5-FU reduces endogenous antioxidant defenses and increases LOOH levels in the lungs, suggesting oxidative stress. Inflammation and histopathological alterations were also detected. The clinical protocol of 5-FU promotes toxicity in the liver, kidneys, and lungs of healthy rats, resulting in different levels of histological and biochemical alterations. These results will be useful in the search for new adjuvants to attenuate the adverse effects of 5-FU in such organs.
Collapse
Affiliation(s)
- Mariana Conceição da Silva
- Biological Physics and Cell Signaling Laboratory, Institute of Biology, Department of Structural and Functional Biology, State University of Campinas, Campinas 13083-970, SP, Brazil
| | - Lilian Catarim Fabiano
- Department of Morphological Science, State University of Maringá, Maringá 87020-900, PR, Brazil
| | | | | | - Camila Quaglio Neves
- Department of Morphological Science, State University of Maringá, Maringá 87020-900, PR, Brazil
| | | | - Maria das Graças de Souza Carvalho
- Biological Physics and Cell Signaling Laboratory, Institute of Biology, Department of Structural and Functional Biology, State University of Campinas, Campinas 13083-970, SP, Brazil
| | - Ana Cristina Breithaupt-Faloppa
- Laboratório de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-904, SP, Brasil
| | - André Alexandre de Thomaz
- Quantum Electronic Department, Institute of Physics Gleb Wataghin, State University of Campinas, Campinas 13083-872, SP, Brazil
| | - Aline Mara Dos Santos
- Biological Physics and Cell Signaling Laboratory, Institute of Biology, Department of Structural and Functional Biology, State University of Campinas, Campinas 13083-970, SP, Brazil
| | - Nilza Cristina Buttow
- Department of Morphological Science, State University of Maringá, Maringá 87020-900, PR, Brazil
| |
Collapse
|
19
|
Liu A, Cai H, Xu Z, Li J, Weng X, Liao C, He J, Liu L, Wang Y, Qu J, Li H, Song J, Guo J. Multifunctional carbon dots for glutathione detection and Golgi imaging. Talanta 2023; 259:124520. [PMID: 37058943 DOI: 10.1016/j.talanta.2023.124520] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 03/30/2023] [Accepted: 03/31/2023] [Indexed: 04/16/2023]
Abstract
Glutathione (GSH) is present in almost every cell in the body and plays various integral roles in many biological processes. The Golgi apparatus is a eukaryotic organelle for the biosynthesis, intracellular distribution, and secretion of various macromolecules; however, the mechanism of GSH in the Golgi apparatus has not been fully elucidated. Here, specific and sensitive sulfur-nitrogen co-doped carbon dots (SNCDs) with orange-red fluorescence was synthesized for the detection of GSH in the Golgi apparatus. The SNCDs have a Stokes shift of 147 nm and excellent fluorescence stability, and they exhibited excellent selectivity and high sensitivity to GSH. The linear response of the SNCDs to GSH was in the range of 10-460 μM (LOD = 0.25 μΜ). More importantly, we used SNCDs with excellent optical properties and low cytotoxicity as probes, and successfully realized golgi imaging in HeLa cells and GSH detection at the same time.
Collapse
Affiliation(s)
- Aikun Liu
- State Key Laboratory of Radio Frequency Heterogeneous Integration(Shenzhen University); College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen, 518060, PR China
| | - Haojie Cai
- State Key Laboratory of Radio Frequency Heterogeneous Integration(Shenzhen University); College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen, 518060, PR China
| | - Zhibing Xu
- State Key Laboratory of Radio Frequency Heterogeneous Integration(Shenzhen University); College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen, 518060, PR China
| | - Jinlei Li
- State Key Laboratory of Radio Frequency Heterogeneous Integration(Shenzhen University); College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen, 518060, PR China
| | - Xiaoyu Weng
- State Key Laboratory of Radio Frequency Heterogeneous Integration(Shenzhen University); College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen, 518060, PR China
| | - Changrui Liao
- State Key Laboratory of Radio Frequency Heterogeneous Integration(Shenzhen University); College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen, 518060, PR China
| | - Jun He
- State Key Laboratory of Radio Frequency Heterogeneous Integration(Shenzhen University); College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen, 518060, PR China
| | - Liwei Liu
- State Key Laboratory of Radio Frequency Heterogeneous Integration(Shenzhen University); College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen, 518060, PR China
| | - Yiping Wang
- State Key Laboratory of Radio Frequency Heterogeneous Integration(Shenzhen University); College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen, 518060, PR China
| | - Junle Qu
- State Key Laboratory of Radio Frequency Heterogeneous Integration(Shenzhen University); College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen, 518060, PR China
| | - Hao Li
- State Key Laboratory of Radio Frequency Heterogeneous Integration(Shenzhen University); College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen, 518060, PR China.
| | - Jun Song
- State Key Laboratory of Radio Frequency Heterogeneous Integration(Shenzhen University); College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen, 518060, PR China.
| | - Jiaqing Guo
- State Key Laboratory of Radio Frequency Heterogeneous Integration(Shenzhen University); College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen, 518060, PR China.
| |
Collapse
|
20
|
Lund M, Heaton R, Hargreaves IP, Gregersen N, Olsen RKJ. Odd- and even-numbered medium-chained fatty acids protect against glutathione depletion in very long-chain acyl-CoA dehydrogenase deficiency. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159248. [PMID: 36356723 DOI: 10.1016/j.bbalip.2022.159248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 10/09/2022] [Accepted: 10/17/2022] [Indexed: 11/09/2022]
Abstract
Recent trials have reported the ability of triheptanoin to improve clinical outcomes for the severe symptoms associated with long-chain fatty acid oxidation disorders, including very long-chain acyl-CoA dehydrogenase (VLCAD) deficiency. However, the milder myopathic symptoms are still challenging to treat satisfactorily. Myopathic pathogenesis is multifactorial, but oxidative stress is an important component. We have previously shown that metabolic stress increases the oxidative burden in VLCAD-deficient cell lines and can deplete the antioxidant glutathione (GSH). We investigated whether medium-chain fatty acids provide protection against GSH depletion during metabolic stress in VLCAD-deficient fibroblasts. To investigate the effect of differences in anaplerotic capacity, we included both even-(octanoate) and odd-numbered (heptanoate) medium-chain fatty acids. Overall, we show that modulation of the concentration of medium-chain fatty acids in culture media affects levels of GSH retained during metabolic stress in VLCAD-deficient cell lines but not in controls. Lowered glutamine concentration in the culture media during metabolic stress led to GSH depletion and decreased viability in VLCAD deficient cells, which could be rescued by both heptanoate and octanoate in a dose-dependent manner. Unlike GSH levels, the levels of total thiols increased after metabolic stress exposure, the size of this increase was not affected by differences in cell culture medium concentrations of glutamine, heptanoate or octanoate. Addition of a PPAR agonist further exacerbated stress-related GSH-depletion and viability loss, requiring higher concentrations of fatty acids to restore GSH levels and cell viability. Both odd- and even-numbered medium-chain fatty acids efficiently protect VLCADdeficient cells against metabolic stress-induced antioxidant depletion.
Collapse
Affiliation(s)
- Martin Lund
- Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University and Aarhus University Hospital, Palle Juel-Jensens Boulevard 99, 8200 Aarhus, Denmark.
| | - Robert Heaton
- School of Pharmacy, Liverpool John Moore University, Byrom Street, Liverpool L3 3AF, United Kingdom
| | - Iain P Hargreaves
- School of Pharmacy, Liverpool John Moore University, Byrom Street, Liverpool L3 3AF, United Kingdom
| | - Niels Gregersen
- Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University and Aarhus University Hospital, Palle Juel-Jensens Boulevard 99, 8200 Aarhus, Denmark
| | - Rikke K J Olsen
- Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University and Aarhus University Hospital, Palle Juel-Jensens Boulevard 99, 8200 Aarhus, Denmark.
| |
Collapse
|
21
|
Basiouni S, Tellez-Isaias G, Latorre JD, Graham BD, Petrone-Garcia VM, El-Seedi HR, Yalçın S, El-Wahab AA, Visscher C, May-Simera HL, Huber C, Eisenreich W, Shehata AA. Anti-Inflammatory and Antioxidative Phytogenic Substances against Secret Killers in Poultry: Current Status and Prospects. Vet Sci 2023; 10:55. [PMID: 36669057 PMCID: PMC9866488 DOI: 10.3390/vetsci10010055] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 11/19/2022] [Accepted: 01/04/2023] [Indexed: 01/17/2023] Open
Abstract
Chronic stress is recognized as a secret killer in poultry. It is associated with systemic inflammation due to cytokine release, dysbiosis, and the so-called leaky gut syndrome, which mainly results from oxidative stress reactions that damage the barrier function of the cells lining the gut wall. Poultry, especially the genetically selected broiler breeds, frequently suffer from these chronic stress symptoms when exposed to multiple stressors in their growing environments. Since oxidative stress reactions and inflammatory damages are multi-stage and long-term processes, overshooting immune reactions and their down-stream effects also negatively affect the animal's microbiota, and finally impair its performance and commercial value. Means to counteract oxidative stress in poultry and other animals are, therefore, highly welcome. Many phytogenic substances, including flavonoids and phenolic compounds, are known to exert anti-inflammatory and antioxidant effects. In this review, firstly, the main stressors in poultry, such as heat stress, mycotoxins, dysbiosis and diets that contain oxidized lipids that trigger oxidative stress and inflammation, are discussed, along with the key transcription factors involved in the related signal transduction pathways. Secondly, the most promising phytogenic substances and their current applications to ameliorate oxidative stress and inflammation in poultry are highlighted.
Collapse
Affiliation(s)
- Shereen Basiouni
- Institute of Molecular Physiology, Johannes-Gutenberg University, 55128 Mainz, Germany
- Clinical Pathology Department, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh 13736, Egypt
| | - Guillermo Tellez-Isaias
- Department of Poultry Science, University of Arkansas Agricultural Experiment Station, Fayetteville, AR 72701, USA
| | - Juan D. Latorre
- Department of Poultry Science, University of Arkansas Agricultural Experiment Station, Fayetteville, AR 72701, USA
| | - Brittany D. Graham
- Department of Poultry Science, University of Arkansas Agricultural Experiment Station, Fayetteville, AR 72701, USA
| | - Victor M. Petrone-Garcia
- Facultad de Estudios Superiores Cuautitlan, Universidad Nacional Autonoma de Mexico (UNAM), Cuautitlan Izcalli 58190, Mexico
| | - Hesham R. El-Seedi
- Pharmacognosy Group, Department of Pharmaceutical Biosciences, Uppsala University, Biomedical Centre, SE 751 24 Uppsala, Sweden
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
- International Joint Research Laboratory of Intelligent Agriculture and Agri-Products Processing, Jiangsu Education Department, Jiangsu University, Nanjing 210024, China
| | - Sakine Yalçın
- Department of Animal Nutrition and Nutritional Diseases, Faculty of Veterinary Medicine, Ankara University (AU), 06110 Ankara, Turkey
| | - Amr Abd El-Wahab
- Institute for Animal Nutrition, University of Veterinary Medicine Hannover, Foundation, Bischofsholer Damm 15, 30173 Hanover, Germany
- Department of Nutrition and Nutritional Deficiency Diseases, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Christian Visscher
- Institute for Animal Nutrition, University of Veterinary Medicine Hannover, Foundation, Bischofsholer Damm 15, 30173 Hanover, Germany
| | - Helen L. May-Simera
- Institute of Molecular Physiology, Johannes-Gutenberg University, 55128 Mainz, Germany
| | - Claudia Huber
- Structural Biochemistry of Membranes, Bavarian NMR Center, Technical University of Munich (TUM), D-85747 Garching, Germany
| | - Wolfgang Eisenreich
- Structural Biochemistry of Membranes, Bavarian NMR Center, Technical University of Munich (TUM), D-85747 Garching, Germany
| | - Awad A. Shehata
- Avian and Rabbit Diseases Department, Faculty of Veterinary Medicine, University of Sadat City, Sadat City 32897, Egypt
- Research and Development Section, PerNaturam GmbH, An der Trift 8, 56290 Gödenroth, Germany
- Prophy-Institute for Applied Prophylaxis, 59159 Bönen, Germany
| |
Collapse
|
22
|
Potential Therapeutic Implication of Herbal Medicine in Mitochondria-Mediated Oxidative Stress-Related Liver Diseases. Antioxidants (Basel) 2022; 11:antiox11102041. [PMID: 36290765 PMCID: PMC9598588 DOI: 10.3390/antiox11102041] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/10/2022] [Accepted: 10/10/2022] [Indexed: 11/22/2022] Open
Abstract
Mitochondria are double-membrane organelles that play a role in ATP synthesis, calcium homeostasis, oxidation-reduction status, apoptosis, and inflammation. Several human disorders have been linked to mitochondrial dysfunction. It has been found that traditional therapeutic herbs are effective on alcoholic liver disease (ALD) and nonalcoholic fatty liver disease (NAFLD) which are leading causes of liver cirrhosis and hepatocellular carcinoma. The generation of reactive oxygen species (ROS) in response to oxidative stress is caused by mitochondrial dysfunction and is considered critical for treatment. The role of oxidative stress, lipid toxicity, and inflammation in NAFLD are well known. NAFLD is a chronic liver disease that commonly progresses to cirrhosis and chronic liver disease, and people with obesity, insulin resistance, diabetes, hyperlipidemia, and hypertension are at a higher risk of developing NAFLD. NAFLD is associated with a number of pathological factors, including insulin resistance, lipid metabolic dysfunction, oxidative stress, inflammation, apoptosis, and fibrosis. As a result, the improvement in steatosis and inflammation is enough to entice researchers to look into liver disease treatment. However, antioxidant treatment has not been very effective for liver disease. Additionally, it has been suggested that the beneficial effects of herbal medicines on immunity and inflammation are governed by various mechanisms for lipid metabolism and inflammation control. This review provided a summary of research on herbal medicines for the therapeutic implementation of mitochondria-mediated ROS production in liver disease as well as clinical applications through herbal medicine. In addition, the pathophysiology of common liver disorders such as ALD and NAFLD would be investigated in the role that mitochondria play in the process to open new therapeutic avenues in the management of patients with liver disease.
Collapse
|
23
|
Lian Y, Jiang R, Zhang Z, Lin Z, Wang N, Wang XD. Fully Reversible Ratiometric Nanosensors for Continuously Quantifying Mitochondrial Glutathione Concentration in Living Cells. Anal Chem 2022; 94:12570-12577. [PMID: 36074089 DOI: 10.1021/acs.analchem.2c00855] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Mitochondrial glutathione (mGSH) is both the cause of the oxidative damage and a mechanism for maintaining the redox homeostasis in mitochondria. To effectively measure mGSH dynamics in living cells, we have developed a new FRET-based nanosensor by immobilizing rhodamine B into dendritic mesoporous silica nanoparticles and installing GSH probes and mitochondria-targeting motifs onto the surface of nanoparticles. The result shows that these nanosensors show efficient FRET and a full reversibility and rapid response (<10 s) to GSH in the range of 0.5-20 mM, due to their unique nanostructure and well-overlapped spectra. The excellent photostability and low cytotoxicity make them an effective means for monitoring mGSH concentration in real time. When the mGSH nanosensors are used for quantitatively measuring mGSH variations under glucose deprivation stimulation in HeLa cells, they successfully prove themselves a useful tool for mitochondrial redox activity studies.
Collapse
Affiliation(s)
- Ying Lian
- Department of Chemistry, Fudan University, 200438 Shanghai, P.R. China
| | - Rui Jiang
- Department of Chemistry, Fudan University, 200438 Shanghai, P.R. China.,Human Phenome Institute, Fudan University, 200438 Shanghai, P.R. China
| | - Zeyu Zhang
- Department of Chemistry, Fudan University, 200438 Shanghai, P.R. China
| | - Zhenzhen Lin
- Department of Chemistry, Fudan University, 200438 Shanghai, P.R. China
| | - Nianhong Wang
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University; National Medical Center for Neurological Disorders, 200040 Shanghai, P.R. China
| | - Xu-Dong Wang
- Human Phenome Institute, Fudan University, 200438 Shanghai, P.R. China
| |
Collapse
|
24
|
Slc25a39 and Slc25a40 Expression in Mice with Bile Duct Ligation or Lipopolysaccharide Treatment. Int J Mol Sci 2022; 23:ijms23158573. [PMID: 35955707 PMCID: PMC9369313 DOI: 10.3390/ijms23158573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/28/2022] [Accepted: 07/30/2022] [Indexed: 02/01/2023] Open
Abstract
SLC25A39/40, involved in mitochondrial GSH (mGSH) import from the cytoplasm, is essential for protection against oxidative stress and mitochondrial dysfunction. We examined the effects of cholestasis, through bile duct ligation (BDL) and lipopolysaccharide (LPS)-induced inflammation in mice, on Slc25a39/40 expression. Additionally, we used human clear cell renal carcinoma (KMRC-1) cells to elucidate the mechanism of regulation of SLC25A39/40 expression in the kidneys after LPS treatment. BDL resulted in a decrease in Slc25a39 mRNA in the liver and a decrease in Slc25a39/40 mRNA and protein in the kidneys. Consequently, there was a significant decrease in mGSH levels in the kidneys of BDL mice compared with those in sham mice. LPS treatment resulted in increased Slc25a40 expression in the kidneys. In KMRC-1 cells, the combination treatment of LPS-RS or FPS-ZM1 with LPS suppressed the LPS-induced increase in SLC25A40, suggesting that SLC25A40 expression could be regulated by the signaling pathway via toll-like receptor 4 and the receptor for advanced glycation end products, respectively. Our findings contribute to understanding the role of mGSH in the maintenance of the mitochondrial redox state. To the best of our knowledge, this is the first study that demonstrates the changes in Slc25a39/40 expression in mice with cholestasis-associated renal injury and LPS-induced inflammation.
Collapse
|
25
|
Shi X, Reinstadler B, Shah H, To TL, Byrne K, Summer L, Calvo SE, Goldberger O, Doench JG, Mootha VK, Shen H. Combinatorial GxGxE CRISPR screen identifies SLC25A39 in mitochondrial glutathione transport linking iron homeostasis to OXPHOS. Nat Commun 2022; 13:2483. [PMID: 35513392 PMCID: PMC9072411 DOI: 10.1038/s41467-022-30126-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 04/18/2022] [Indexed: 12/18/2022] Open
Abstract
The SLC25 carrier family consists of 53 transporters that shuttle nutrients and co-factors across mitochondrial membranes. The family is highly redundant and their transport activities coupled to metabolic state. Here, we use a pooled, dual CRISPR screening strategy that knocks out pairs of transporters in four metabolic states - glucose, galactose, OXPHOS inhibition, and absence of pyruvate - designed to unmask the inter-dependence of these genes. In total, we screen 63 genes in four metabolic states, corresponding to 2016 single and pair-wise genetic perturbations. We recover 19 gene-by-environment (GxE) interactions and 9 gene-by-gene (GxG) interactions. One GxE interaction hit illustrates that the fitness defect in the mitochondrial folate carrier (SLC25A32) KO cells is genetically buffered in galactose due to a lack of substrate in de novo purine biosynthesis. GxG analysis highlights a buffering interaction between the iron transporter SLC25A37 (A37) and the poorly characterized SLC25A39 (A39). Mitochondrial metabolite profiling, organelle transport assays, and structure-guided mutagenesis identify A39 as critical for mitochondrial glutathione (GSH) import. Functional studies reveal that A39-mediated glutathione homeostasis and A37-mediated mitochondrial iron uptake operate jointly to support mitochondrial OXPHOS. Our work underscores the value of studying family-wide genetic interactions across different metabolic environments.
Collapse
Affiliation(s)
- Xiaojian Shi
- Cellular and Molecular Physiology Department, Yale School of Medicine, New Haven, CT, USA
- Systems Biology Institute, Yale West Campus, West Haven, CT, USA
| | - Bryn Reinstadler
- Howard Hughes Medical Institute and Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
| | - Hardik Shah
- Howard Hughes Medical Institute and Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
| | - Tsz-Leung To
- Howard Hughes Medical Institute and Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
| | - Katie Byrne
- Cellular and Molecular Physiology Department, Yale School of Medicine, New Haven, CT, USA
- Systems Biology Institute, Yale West Campus, West Haven, CT, USA
| | - Luanna Summer
- Cellular and Molecular Physiology Department, Yale School of Medicine, New Haven, CT, USA
- Systems Biology Institute, Yale West Campus, West Haven, CT, USA
| | - Sarah E Calvo
- Howard Hughes Medical Institute and Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
| | - Olga Goldberger
- Howard Hughes Medical Institute and Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | | | - Vamsi K Mootha
- Howard Hughes Medical Institute and Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
| | - Hongying Shen
- Cellular and Molecular Physiology Department, Yale School of Medicine, New Haven, CT, USA.
- Systems Biology Institute, Yale West Campus, West Haven, CT, USA.
| |
Collapse
|
26
|
Pourzand C, Albieri-Borges A, Raczek NN. Shedding a New Light on Skin Aging, Iron- and Redox-Homeostasis and Emerging Natural Antioxidants. Antioxidants (Basel) 2022; 11:471. [PMID: 35326121 PMCID: PMC8944509 DOI: 10.3390/antiox11030471] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/25/2022] [Accepted: 02/25/2022] [Indexed: 12/10/2022] Open
Abstract
Reactive oxygen species (ROS) are necessary for normal cell signaling and the antimicrobial defense of the skin. However excess production of ROS can disrupt the cellular redox balance and overwhelm the cellular antioxidant (AO) capacity, leading to oxidative stress. In the skin, oxidative stress plays a key role in driving both extrinsic and intrinsic aging. Sunlight exposure has also been a major contributor to extrinsic photoaging of the skin as its oxidising components disrupt both redox- and iron-homeostasis, promoting oxidative damage to skin cells and tissue constituents. Upon oxidative insults, the interplay between excess accumulation of ROS and redox-active labile iron (LI) and its detrimental consequences to the skin are often overlooked. In this review we have revisited the oxidative mechanisms underlying skin damage and aging by focussing on the concerted action of ROS and redox-active LI in the initiation and progression of intrinsic and extrinsic skin aging processes. Based on these, we propose to redefine the selection criteria for skin antiaging and photoprotective ingredients to include natural antioxidants (AOs) exhibiting robust redox-balancing and/or iron-chelating properties. This would promote the concept of natural-based or bio-inspired bifunctional anti-aging and photoprotective ingredients for skincare and sunscreen formulations with both AO and iron-chelating properties.
Collapse
Affiliation(s)
- Charareh Pourzand
- Medicines Design, Department of Pharmacy and Pharmacology, University of Bath, Bath BA2 7AY, UK
- Medicines Development, Centre for Therapeutic Innovation, University of Bath, Bath BA2 7AY, UK
| | - Andrea Albieri-Borges
- Research and Development, ASEA LLC., Pleasant Grove, UT 84062, USA; (A.A.-B.); (N.N.R.)
| | - Nico N. Raczek
- Research and Development, ASEA LLC., Pleasant Grove, UT 84062, USA; (A.A.-B.); (N.N.R.)
| |
Collapse
|
27
|
Sun Y, Yuan K, Mo X, Chen X, Deng Y, Liu C, Yuan Y, Nie J, Zhang Y. Tyndall-Effect-inspired assay with gold nanoparticles for the colorimetric discrimination and quantification of mercury ions and glutathione. Talanta 2022; 238:122999. [PMID: 34857332 DOI: 10.1016/j.talanta.2021.122999] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/23/2021] [Accepted: 10/25/2021] [Indexed: 11/15/2022]
Abstract
This work initially reports a new nanosening method for simple, sensitive, specific, visual detection of mercury (II) (Hg2+) and glutathione (GSH) using the Tyndall Effect (TE) of the same colloidal gold nanoparticle (GNP) probes for efficient colorimetric signaling amplification. For the TE-inspired assay (TEA) method, arginine (Arg) molecules are pre-modified on the GNPs' surfaces (Arg-GNPs). Upon the Hg2+ introduction, it can be specifically coordinated with the terminal -NH2 and -COOH groups of the Arg molecules to make the Arg-GNPs aggregate, producing a significantly-enhanced TE signal in the reaction solution after its irradiation by a 635-nm red laser pointer pen. On the other hand, the introduction of the GSH results in the production of the original Arg-GNPs' weak TE response, as it is able to bind such metal ion via mercury-thiol reactions to inhibit the above aggregation. Under the optimal conditions, the utility of the new TEA method is well demonstrated to quantitatively detect the Hg2+ and GSH with the aid of a smartphone as a portable TE reader during the linear concentration ranges of 50-3000 and 10-3000 nM, respectively. The detection limits for the Hg2+ and GSH are estimated to be as low as ∼3.5 and ∼0.3 nM, respectively. The recovery results obtained from the detection of Hg2+ in the complex tap and pond water samples and the assay of GSH in real human serum and urine samples are also satisfactory.
Collapse
Affiliation(s)
- Yao Sun
- Guangxi Key Laboratory of Electrochemical and Magnetochemical Function Materials, College of Chemistry and Bioengineering, Guilin University of Technology, 12 Jiangan Road, Guilin, 541004, PR China
| | - Kaijing Yuan
- Guangxi Key Laboratory of Electrochemical and Magnetochemical Function Materials, College of Chemistry and Bioengineering, Guilin University of Technology, 12 Jiangan Road, Guilin, 541004, PR China
| | - Xiaomei Mo
- Guangxi Key Laboratory of Electrochemical and Magnetochemical Function Materials, College of Chemistry and Bioengineering, Guilin University of Technology, 12 Jiangan Road, Guilin, 541004, PR China
| | - Xuejiang Chen
- Guangxi Key Laboratory of Electrochemical and Magnetochemical Function Materials, College of Chemistry and Bioengineering, Guilin University of Technology, 12 Jiangan Road, Guilin, 541004, PR China
| | - Yanan Deng
- Guangxi Key Laboratory of Electrochemical and Magnetochemical Function Materials, College of Chemistry and Bioengineering, Guilin University of Technology, 12 Jiangan Road, Guilin, 541004, PR China
| | - Chang Liu
- Guangxi Key Laboratory of Electrochemical and Magnetochemical Function Materials, College of Chemistry and Bioengineering, Guilin University of Technology, 12 Jiangan Road, Guilin, 541004, PR China
| | - Yali Yuan
- Guangxi Key Laboratory of Electrochemical and Magnetochemical Function Materials, College of Chemistry and Bioengineering, Guilin University of Technology, 12 Jiangan Road, Guilin, 541004, PR China
| | - Jinfang Nie
- Guangxi Key Laboratory of Electrochemical and Magnetochemical Function Materials, College of Chemistry and Bioengineering, Guilin University of Technology, 12 Jiangan Road, Guilin, 541004, PR China.
| | - Yun Zhang
- Guangxi Key Laboratory of Electrochemical and Magnetochemical Function Materials, College of Chemistry and Bioengineering, Guilin University of Technology, 12 Jiangan Road, Guilin, 541004, PR China.
| |
Collapse
|
28
|
Rodríguez-Graciani KM, Chapa-Dubocq XR, Ayala-Arroyo EJ, Chaves-Negrón I, Jang S, Chorna N, S. Maskrey T, Wipf P, Javadov S. Effects of Ferroptosis on the Metabolome in Cardiac Cells: The Role of Glutaminolysis. Antioxidants (Basel) 2022; 11:antiox11020278. [PMID: 35204160 PMCID: PMC8868370 DOI: 10.3390/antiox11020278] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/26/2022] [Accepted: 01/27/2022] [Indexed: 12/26/2022] Open
Abstract
Ferroptosis is a novel iron-dependent regulated cell death mechanism that affects cell metabolism; however, a detailed metabolomic analysis of ferroptotic cells is not yet available. Here, we elucidated the metabolome of H9c2 cardioblasts by gas chromatography-mass spectrometry during ferroptosis induced by RSL3, a GPX4 inhibitor, in the presence of ferrostatin-1 (a ferroptosis inhibitor), XJB-5-131 (a mitochondrial-targeted ROS scavenger), or TSM-1005-44 (a newly developed cellular ROS scavenger). Results demonstrated that RSL3 decreased the levels of amino acids involved in glutathione synthesis more than two-fold. In contrast, saturated fatty acids levels were markedly increased in RSL3-challenged cells, with no effects on unsaturated fatty acids. RSL3 significantly altered the levels of mitochondrial tricarboxylic acid cycle intermediates; isocitrate and 2-oxoglutarate were found to increase, whereas succinate was significantly decreased in RSL3-challenged cells. Ferrostatin-1, XJB-5-131, and TSM-1005-44 prevented RSL3-induced cell death and conserved the metabolomic profile of the cells. Since 2-oxoglutarate is involved in the regulation of ferroptosis, particularly through glutamine metabolism, we further assessed the role of glutaminolysis in ferroptosis in H9c2 cardioblasts. Genetic silencing of GLS1, which encodes the K-type mitochondrial glutaminase (glutaminase C), protected against ferroptosis in the early stage. In conclusion, our study demonstrates that RSL3-induced ferroptosis impairs the metabolome of H9c2 cardioblasts.
Collapse
Affiliation(s)
- Keishla M. Rodríguez-Graciani
- Department of Physiology, School of Medicine, University of Puerto Rico, San Juan, PR 00936, USA; (K.M.R.-G.); (X.R.C.-D.); (E.J.A.-A.); (I.C.-N.); (S.J.)
| | - Xavier R. Chapa-Dubocq
- Department of Physiology, School of Medicine, University of Puerto Rico, San Juan, PR 00936, USA; (K.M.R.-G.); (X.R.C.-D.); (E.J.A.-A.); (I.C.-N.); (S.J.)
| | - Esteban J. Ayala-Arroyo
- Department of Physiology, School of Medicine, University of Puerto Rico, San Juan, PR 00936, USA; (K.M.R.-G.); (X.R.C.-D.); (E.J.A.-A.); (I.C.-N.); (S.J.)
| | - Ivana Chaves-Negrón
- Department of Physiology, School of Medicine, University of Puerto Rico, San Juan, PR 00936, USA; (K.M.R.-G.); (X.R.C.-D.); (E.J.A.-A.); (I.C.-N.); (S.J.)
| | - Sehwan Jang
- Department of Physiology, School of Medicine, University of Puerto Rico, San Juan, PR 00936, USA; (K.M.R.-G.); (X.R.C.-D.); (E.J.A.-A.); (I.C.-N.); (S.J.)
| | - Nataliya Chorna
- Department of Biochemistry, School of Medicine, University of Puerto Rico, San Juan, PR 00936, USA;
| | - Taber S. Maskrey
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA 15260, USA; (T.S.M.); (P.W.)
| | - Peter Wipf
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA 15260, USA; (T.S.M.); (P.W.)
| | - Sabzali Javadov
- Department of Physiology, School of Medicine, University of Puerto Rico, San Juan, PR 00936, USA; (K.M.R.-G.); (X.R.C.-D.); (E.J.A.-A.); (I.C.-N.); (S.J.)
- Correspondence: ; Tel.: +1-787-758-2525 (ext. 2909)
| |
Collapse
|
29
|
Wang D, Ye J, Shi R, Zhao B, Liu Z, Lin W, Liu X. Dietary protein and amino acid restriction: Roles in metabolic health and aging-related diseases. Free Radic Biol Med 2022; 178:226-242. [PMID: 34890767 DOI: 10.1016/j.freeradbiomed.2021.12.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 12/03/2021] [Accepted: 12/04/2021] [Indexed: 12/13/2022]
Abstract
The prevalence of obesity is a worldwide phenomenon in all age groups and is associated with aging-related diseases such as type 2 diabetes, as well metabolic and cardiovascular diseases. The use of dietary restriction (DR) while avoiding malnutrition has many profound beneficial effects on aging and metabolic health, and dietary protein or specific amino acid (AA) restrictions, rather than overall calorie intake, are considered to play key roles in the effects of DR on host health. Whereas comprehensive reviews of the underlying mechanisms are limited, protein restriction and methionine (Met) restriction improve metabolic health and aging-related neurodegenerative diseases, and may be associated with FGF21, mTOR and autophagy, improved mitochondrial function and oxidative stress. Circulating branched-chain amino acids (BCAAs) are inversely correlated with metabolic health, and BCAAs and leucine (Leu) restriction promote metabolic homeostasis in rodents. Although tryptophan (Trp) restriction extends the lifespan of rodents, the Trp-restricted diet is reported to increase inflammation in aged mice, while severe Trp restriction has side effects such as anorexia. Furthermore, inadequate protein intake in the elderly increases the risk of muscle-centric health. Therefore, the restriction of specific AAs may be an effective and executable dietary manipulation for metabolic and aging-related health in humans, which warrants further investigation to elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Danna Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Jin Ye
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Renjie Shi
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Beita Zhao
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Zhigang Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Wei Lin
- Department of Neurosurgery, Xijing Institute of Clinical Neuroscience, Xijing Hospital, Air Force Medical University, Xi'an, Shanxi, China.
| | - Xuebo Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling, China.
| |
Collapse
|
30
|
Ting Hao W, Huang L, Pan W, Ren YL. Antioxidant glutathione inhibits inflammation in synovial fibroblasts via PTEN/PI3K/AKT pathway: An in vitro study. Arch Rheumatol 2021; 37:212-222. [PMID: 36017213 PMCID: PMC9377173 DOI: 10.46497/archrheumatol.2022.9109] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/23/2021] [Indexed: 11/29/2022] Open
Abstract
Objectives
In this study, we aimed to investigate whether glutathione (GSH) could decrease the secretion of reactive oxygen species (ROS), reduce inflammation, and modulate the phosphatase and tensin homolog deleted on chromosome 10/phosphatidylinositol 3-kinase/AKT (PTEN/PI3K/AKT) in synovial fibroblasts (SFs). Patients and methods
A total of 30 DBA/1J female mice were used in this study. The release of ROS in MH7A cells was examined using a ROS assay kit. The effects of GSH on the messenger ribonucleic acid (mRNA) expression and protein levels of inflammatory cytokines were determined via reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and enzyme-linked immunosorbent assay (ELISA) in mouse SFs and MH7A cells, respectively. The PTEN/PI3K/AKT pathway was investigated via Western blotting. The effects of buthionine-sulfoximine (BSO), as an inhibitor of GSH, on these molecules were examined. Results
The ROS were decreased after GSH treatment, and the mRNA levels of tumor necrosis factor-alpha (TNF-α), interleukin (IL)-1β, IL-6, matrix metalloproteinase (MMP)-1, MMP-3, were also significantly inhibited after GSH stimulation. However, the IL-10 levels were enhanced, and GSH increased the expression of PTEN. The GSH suppressed the activation of phosphorylated (p)-PI3K and p-AKT. The supplementation of the BSO restored the activation of PI3K/AKT pathway with a high production of ROS. The levels of TNF-α, IL-1β and IL-6 were also elevated, when the BSO was added. Conclusion
These findings suggest that GSH can act as an inflammatory suppressor by downregulating the PTEN/PI3K/AKT pathway in MH7A cells. These data indicated a novel function of GSH for improving the inflammation of RA SFs and may help to alleviate the pathological process of RA.
Collapse
Affiliation(s)
- Wen Ting Hao
- Department of Rheumatology and Immunology, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, China
| | - Lu Huang
- Department of Rheumatology and Immunology, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, China
| | - Wei Pan
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, Jiangsu
| | - Yi Le Ren
- Department of Rheumatology and Immunology, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
31
|
Chełchowska M, Gajewska J, Ambroszkiewicz J, Mazur J, Ołtarzewski M, Maciejewski TM. Influence of Oxidative Stress Generated by Smoking during Pregnancy on Glutathione Status in Mother-Newborn Pairs. Antioxidants (Basel) 2021; 10:antiox10121866. [PMID: 34942969 PMCID: PMC8698311 DOI: 10.3390/antiox10121866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/15/2021] [Accepted: 11/21/2021] [Indexed: 11/16/2022] Open
Abstract
Glutathione plays a key role in maintaining a physiological balance between prooxidants and antioxidants in the human body. Therefore, we examined the influence of maternal smoking as a source of oxidative stress measured by total oxidant capacity (TOC) on reduced glutathione (GSH), oxidized glutathione (GSSG), glutathione peroxidase (GPx-3), and reductase (GR) amount in maternal and umbilical cord blood in 110 (45 smoking and 65 non-smoking) mother-newborn pairs. Concentrations of glutathione status markers and TOC were evaluated by competitive inhibition enzyme immunoassay technique. Plasma TOC levels were significantly higher and the GSH/GSSG ratio, which is considered an index of the cell’s redox status, were significantly lower in smoking women and their offspring than in non-smoking pairs. Decreased GR levels were found in smoking mothers and their newborns compared with similar non-smoking groups. Although plasma GPx-3 concentrations were similar in both maternal groups, in the cord blood of newborns exposed to tobacco smoke in utero they were reduced compared with the levels observed in children of tobacco abstinent mothers. Oxidative stress generated by tobacco smoke impairs glutathione homeostasis in both the mother and the newborn. The severity of oxidative processes in the mother co-existing with the reduced potential of antioxidant systems may have a negative effect on the oxidative-antioxidant balance in the newborn.
Collapse
Affiliation(s)
- Magdalena Chełchowska
- Department of Screening Tests and Metabolic Diagnostics, Institute of Mother and Child, Kasprzaka 17a, 01-211 Warsaw, Poland; (J.G.); (J.A.); (M.O.)
- Correspondence: ; Tel./Fax: +48-2-2327-7260
| | - Joanna Gajewska
- Department of Screening Tests and Metabolic Diagnostics, Institute of Mother and Child, Kasprzaka 17a, 01-211 Warsaw, Poland; (J.G.); (J.A.); (M.O.)
| | - Jadwiga Ambroszkiewicz
- Department of Screening Tests and Metabolic Diagnostics, Institute of Mother and Child, Kasprzaka 17a, 01-211 Warsaw, Poland; (J.G.); (J.A.); (M.O.)
| | - Joanna Mazur
- Department of Humanization in Medicine and Sexology, Collegium Medicum University of Zielona Góra, 65-729 Zielona Góra, Poland;
| | - Mariusz Ołtarzewski
- Department of Screening Tests and Metabolic Diagnostics, Institute of Mother and Child, Kasprzaka 17a, 01-211 Warsaw, Poland; (J.G.); (J.A.); (M.O.)
| | - Tomasz M. Maciejewski
- Clinic of Obstetrics and Gynaecology, Institute of Mother and Child, Kasprzaka 17a, 01-211 Warsaw, Poland;
| |
Collapse
|
32
|
Wan XL, Li N, Chen YJ, Chen XS, Yang Z, Xu L, Yang HM, Wang ZY. Protective effects of lycopene on mitochondrial oxidative injury and dysfunction in the liver of aflatoxin B 1-exposed broilers. Poult Sci 2021; 100:101441. [PMID: 34547623 PMCID: PMC8456063 DOI: 10.1016/j.psj.2021.101441] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 08/10/2021] [Accepted: 08/15/2021] [Indexed: 11/23/2022] Open
Abstract
This study was conducted to investigate the effects of lycopene (LYC) on mitochondrial oxidative injury and dysfunction in the liver of aflatoxin B1 (AFB1)-exposed broilers. A total of 192 healthy 1-day-old male broilers were randomly divided into 3 groups with 8 replicates of 8 birds each. Birds in the 3 groups were fed basal diet (control), basal diet with 100 µg/kg AFB1, and basal diet with 100 µg/kg AFB1 and 200 mg/kg LYC, respectively. The experiment lasted 42 d. The results showed that AFB1 decreased average daily body weight gain (ADG), average daily feed intake, and gain to feed ratio (G :F) compared to the control group, the LYC supplementation increased ADG and G/F compared to AFB1 group (P < 0.05). Broilers in the AFB1 group had lower mitochondrial glutathione (mGSH) concentration and glutathione peroxidase (GSH-Px), manganese superoxide dismutase (MnSOD), and thioredoxin reductase activities, and higher hydrogen peroxide (H2O2) and reactive oxygen species (ROS) concentrations than the control group (P < 0.05). The LYC increased mGSH concentration and GSH-Px and MnSOD activities, and decreased H2O2 and ROS concentrations compared to AFB1 group (P < 0.05). Broilers fed the AFB1 diet showed increased mitochondrial swelling and decreased adenosine triphosphate concentration than the control group, and LYC had opposite effects (P < 0.05). The AFB1 decreased the activities of mitochondrial electron transfer chain (ETC) complexes I, II, III, and V, downregulated the mRNA expression levels of hepatic MnSOD, thioredoxin 2, thioredoxin reductase, peroxiredoxin-3, peroxisome proliferator-activated receptor γ coactivator 1α, nuclear respiratory factor 1, and mitochondrial transcription factor A compared with the control group (P < 0.05), and LYC increased activities of mitochondrial ETC complexes III and V, and upregulated mRNA expression levels of these genes in comparison to AFB1 group (P < 0.05). In conclusion, the LYC protected broilers from AFB1-induced liver mitochondrial oxidative injury and dysfunction by stimulating mitochondrial antioxidant capacity and maintaining mitochondrial biogenesis.
Collapse
Affiliation(s)
- X L Wan
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province, 225009, P. R. China
| | - N Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province, 225009, P. R. China
| | - Y J Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province, 225009, P. R. China
| | - X S Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province, 225009, P. R. China
| | - Z Yang
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu Province, 225009, P. R. China
| | - L Xu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province, 225009, P. R. China
| | - H M Yang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province, 225009, P. R. China
| | - Z Y Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province, 225009, P. R. China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu Province, 225009, P. R. China.
| |
Collapse
|
33
|
Shin EJ, Jeong JH, Nguyen BT, Sharma N, Nah SY, Chung YH, Lee Y, Byun JK, Nabeshima T, Ko SK, Kim HC. Ginsenoside Re Protects against Serotonergic Behaviors Evoked by 2,5-Dimethoxy-4-iodo-amphetamine in Mice via Inhibition of PKCδ-Mediated Mitochondrial Dysfunction. Int J Mol Sci 2021; 22:ijms22137219. [PMID: 34281274 PMCID: PMC8268959 DOI: 10.3390/ijms22137219] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/26/2021] [Accepted: 06/29/2021] [Indexed: 02/07/2023] Open
Abstract
It has been recognized that serotonin 2A receptor (5-HT2A) agonist 2,5-dimethoxy-4-iodo-amphetamine (DOI) impairs serotonergic homeostasis. However, the mechanism of DOI-induced serotonergic behaviors remains to be explored. Moreover, little is known about therapeutic interventions against serotonin syndrome, although evidence suggests that ginseng might possess modulating effects on the serotonin system. As ginsenoside Re (GRe) is well-known as a novel antioxidant in the nervous system, we investigated whether GRe modulates 5-HT2A receptor agonist DOI-induced serotonin impairments. We proposed that protein kinase Cδ (PKCδ) mediates serotonergic impairments. Treatment with GRe or 5-HT2A receptor antagonist MDL11939 significantly attenuated DOI-induced serotonergic behaviors (i.e., overall serotonergic syndrome behaviors, head twitch response, hyperthermia) by inhibiting mitochondrial translocation of PKCδ, reducing mitochondrial glutathione peroxidase activity, mitochondrial dysfunction, and mitochondrial oxidative stress in wild-type mice. These attenuations were in line with those observed upon PKCδ inhibition (i.e., pharmacologic inhibitor rottlerin or PKCδ knockout mice). Furthermore, GRe was not further implicated in attenuation mediated by PKCδ knockout in mice. Our results suggest that PKCδ is a therapeutic target for GRe against serotonergic behaviors induced by DOI.
Collapse
Affiliation(s)
- Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 24341, Korea; (E.-J.S.); (B.-T.N.); (N.S.)
| | - Ji Hoon Jeong
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, College of Medicine, Chung-Ang University, Seoul 06974, Korea;
| | - Bao-Trong Nguyen
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 24341, Korea; (E.-J.S.); (B.-T.N.); (N.S.)
| | - Naveen Sharma
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 24341, Korea; (E.-J.S.); (B.-T.N.); (N.S.)
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, College of Medicine, Chung-Ang University, Seoul 06974, Korea;
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory, Department of Physiology, College of Veterinary Medicine and Bio/Molecular Informatics Center, Konkuk University, Seoul 05029, Korea;
| | - Yoon Hee Chung
- Department of Anatomy, College of Medicine, Chung-Ang University, Seoul 06974, Korea;
| | - Yi Lee
- Department of Industrial Plant Science & Technology, Chungbuk National University, Chungju 28644, Korea;
| | - Jae Kyung Byun
- Korea Society of Forest Environmental Research, Namyanju 12106, Korea;
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Science, Toyoake 470-1192, Japan;
| | - Sung Kwon Ko
- Department of Oriental Medical Food and Nutrition, Semyung University, Jecheon 27136, Korea
- Correspondence: (S.K.K.); (H.-C.K.); Tel.: +82-33-250-6917 (H.-C.K.); Fax: +82-33-259-5631 (H.-C.K.)
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 24341, Korea; (E.-J.S.); (B.-T.N.); (N.S.)
- Correspondence: (S.K.K.); (H.-C.K.); Tel.: +82-33-250-6917 (H.-C.K.); Fax: +82-33-259-5631 (H.-C.K.)
| |
Collapse
|
34
|
Zhang W, Gao J, Lu L, Bold T, Li X, Wang S, Chang Z, Chen J, Kong X, Zheng Y, Zhang M, Tang J. Intracellular GSH/GST antioxidants system change as an earlier biomarker for toxicity evaluation of iron oxide nanoparticles. NANOIMPACT 2021; 23:100338. [PMID: 35559839 DOI: 10.1016/j.impact.2021.100338] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 06/14/2021] [Accepted: 06/18/2021] [Indexed: 05/14/2023]
Abstract
Glutathione (GSH) and glutathione-S-transferases (GSTs) are two frontlines of cellular defense against both acute and chronic toxicity of xenobiotics-induced oxidative stress. The contribution of GSH and GST enzymes to signaling pathways and the regulation of GSH homeostasis play a central role in the detoxification of numerous environmental toxins and impurities. Iron oxide nanoparticles stemmed from traffic exhaust, steel manufacturing, or welding as a potential environmental pollution can lead to adverse respiratory outcomes and aggravate the risk of chronic health conditions via persistent oxidative stress. In this work, two kinds of acute exposure experiments of iron oxide (Fe2O3 and Fe3O4) nanoparticles in cells and in vivo were conducted to evaluate the GSH levels and GST activity. Our current research presented Fe3O4 nanoparticles at lower concentrations (≤100 μg/ml) seem to be more toxic to the human bronchial epithelial cells as their consumption of GSH and decrease of GST activity. The catalysis activity of Fe3O4 nanoparticles per se may contribute to the intracellular GSH consumption along with inhibition of glutathione-S-transferase class mu 1 and P (GSTM1 and GSTP1) active site and expression decrease of GSTM1 and GSTP1. Accordingly, the GSH consumption and decrease in GST activity directed to the further lipid peroxidation regarded as an earlier marker for toxicity evaluation of iron oxide nanoparticles, and relevant intervention may be effective for prevention of respiratory exposure induced damage from iron oxide nanoparticles.
Collapse
Affiliation(s)
- Wanjun Zhang
- Departmental of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Jinling Gao
- Departmental of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China; Department of Infection Management Service, Dushu Lake Hospital Affiliated of Soochow University, Suzhou 215000, China
| | - Lin Lu
- Departmental of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Tsendmaa Bold
- Departmental of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Xin Li
- Departmental of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Shuo Wang
- Departmental of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Zhishang Chang
- Departmental of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Jing Chen
- Departmental of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Xiao Kong
- Departmental of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Yuxin Zheng
- Departmental of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Mingliang Zhang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300384, China.
| | - Jinglong Tang
- Departmental of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
35
|
Role of Glutathione in Chalcone Derivative Induced Apoptosis of Brugia malayi and its Possible Therapeutic Implication. Acta Parasitol 2021; 66:406-415. [PMID: 33037957 DOI: 10.1007/s11686-020-00291-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 09/28/2020] [Indexed: 10/22/2022]
Abstract
PURPOSE Oxidative stress is an essential component of innate response against microbes. The oxidative impact has a very subtle connection with apoptosis. Our previous work indicated presumptive evidence of apoptosis by the chalcone derivatives against the human lymphatic filarial parasite. Evidence suggests the involvement of glutathione-S-transferase (GST) in the mechanism of action of chalcone drugs. In the present study, we explored the implications of redox status in apoptosis of the parasite by this drug. RESULTS Treatment with the representative drug, 4t, significantly decreased GSH level and increased GST activity in the Brugia malayi microfilariae (Mf) in comparison to Mf without 4t treatment. Drug-induced loss of motility of the parasites was reversed by the treatment with GSH (41%) and NAC (19%). A significant fall in rGST activity was observed due to drug addition, which could be reversed by the addition of GSH co-substrate, but not with the re-addition of rGST, indicating a vital role of GSH. In silico study demonstrated a favorable drug-GST enzyme interaction. Oxidative stress was reflected by increased protein carbonylation and intracellular reactive oxygen species level, in the drug-treated parasite. Mitochondrial oxygen consumption was reduced by the drug, which was reversed on the addition of GSH. Mitochondrial dysfunction was confirmed by MTT and cytochrome c assay. Apoptosis was confirmed by the inhibition in PARP activity. CONCLUSION We conclude that the depletion of GSH by chalcone with concomitant mitochondrial dysfunction revealed a novel rationale of apoptosis in the parasite. Such a mechanism might have wide therapeutic implications.
Collapse
|
36
|
Sreekumar PG, Ferrington DA, Kannan R. Glutathione Metabolism and the Novel Role of Mitochondrial GSH in Retinal Degeneration. Antioxidants (Basel) 2021; 10:661. [PMID: 33923192 PMCID: PMC8146950 DOI: 10.3390/antiox10050661] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/18/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023] Open
Abstract
Glutathione (GSH) is present ubiquitously, and its role as a crucial cellular antioxidant in tissues, including the retina, is well established. GSH's antioxidant function arises from its ability to scavenge reactive oxygen species or to serve as an essential cofactor for GSH S-transferases and peroxidases. This review summarizes the general functions, retinal distribution, disorders linked to GSH deficiency, and the emerging role for mitochondrial GSH (mGSH) in retinal function. Though synthesized only in the cytosol, the presence of GSH in multiple cell organelles suggests the requirement for its active transport across organellar membranes. The localization and distribution of 2-oxoglutarate carrier (OGC) and dicarboxylate carrier (DIC), two recently characterized mitochondrial carrier proteins in RPE and retina, show that these transporters are highly expressed in human retinal pigment epithelium (RPE) cells and retinal layers, and their expression increases with RPE polarity in cultured cells. Depletion of mGSH levels via inhibition of the two transporters resulted in reduced mitochondrial bioenergetic parameters (basal respiration, ATP production, maximal respiration, and spare respiratory capacity) and increased RPE cell death. These results begin to reveal a critical role for mGSH in maintaining RPE bioenergetics and cell health. Thus, augmentation of mGSH pool under GSH-deficient conditions may be a valuable tool in treating retinal disorders, such as age-related macular degeneration and optic neuropathies, whose pathologies have been associated with mitochondrial dysfunction.
Collapse
Affiliation(s)
- Parameswaran G. Sreekumar
- The Stephen J. Ryan Initiative for Macular Research (RIMR), Doheny Eye Institute, Los Angeles, CA 90033, USA;
| | - Deborah A. Ferrington
- Department of Ophthalmology and Visual Neurosciences and Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Ram Kannan
- The Stephen J. Ryan Initiative for Macular Research (RIMR), Doheny Eye Institute, Los Angeles, CA 90033, USA;
- Stein Eye Institute, Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
37
|
Erlichman JS, Leiter JC. Complexity of the Nano-Bio Interface and the Tortuous Path of Metal Oxides in Biological Systems. Antioxidants (Basel) 2021; 10:antiox10040547. [PMID: 33915992 PMCID: PMC8066112 DOI: 10.3390/antiox10040547] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 03/11/2021] [Accepted: 03/23/2021] [Indexed: 01/12/2023] Open
Abstract
Metal oxide nanoparticles (NPs) have received a great deal of attention as potential theranostic agents. Despite extensive work on a wide variety of metal oxide NPs, few chemically active metal oxide NPs have received Food and Drug Administration (FDA) clearance. The clinical translation of metal oxide NP activity, which often looks so promising in preclinical studies, has not progressed as rapidly as one might expect. The lack of FDA approval for metal oxide NPs appears to be a consequence of the complex transformation of NP chemistry as any given NP passes through multiple extra- and intracellular environments and interacts with a variety of proteins and transport processes that may degrade or transform the chemical properties of the metal oxide NP. Moreover, the translational models frequently used to study these materials do not represent the final therapeutic environment well, and studies in reduced preparations have, all too frequently, predicted fundamentally different physico-chemical properties from the biological activity observed in intact organisms. Understanding the evolving pharmacology of metal oxide NPs as they interact with biological systems is critical to establish translational test systems that effectively predict future theranostic activity.
Collapse
Affiliation(s)
- Joseph S. Erlichman
- Department of Biology, St. Lawrence University, Canton, NY 13617, USA
- Correspondence: ; Tel.: +1-(315)-229-5639
| | - James C. Leiter
- White River Junction VA Medical Center, White River Junction, VT 05009, USA;
| |
Collapse
|
38
|
Lund M, Andersen KG, Heaton R, Hargreaves IP, Gregersen N, Olsen RKJ. Bezafibrate activation of PPAR drives disturbances in mitochondrial redox bioenergetics and decreases the viability of cells from patients with VLCAD deficiency. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166100. [PMID: 33549744 DOI: 10.1016/j.bbadis.2021.166100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 01/11/2021] [Accepted: 02/01/2021] [Indexed: 10/22/2022]
Abstract
Very long-chain acyl-CoA dehydrogenase (VLCAD) deficiency is the most common inborn long-chain fatty acid oxidation (FAO) disorder. VLCAD deficiency is characterized by distinct phenotypes. The severe phenotypes are potentially life-threatening and affect the heart or liver, with a comparatively milder phenotype characterized by myopathic symptoms. There is an unmet clinical need for effective treatment options for the myopathic phenotype. The molecular mechanisms driving the gradual decrease in mitochondrial function and associated alterations of muscle fibers are unclear. The peroxisome proliferator-activated receptor (PPAR) pan-agonist bezafibrate is a potent modulator of FAO and multiple other mitochondrial functions and has been proposed as a potential medication for myopathic cases of long-chain FAO disorders. In vitro experiments have demonstrated the ability of bezafibrate to increase VLCAD expression and activity. However, the outcome of small-scale clinical trials has been controversial. We found VLCAD deficient patient fibroblasts to have an increased oxidative stress burden and deranged mitochondrial bioenergetic capacity, compared to controls. Applying heat stress under fasting conditions to bezafibrate pretreated patient cells, caused a marked further increase of mitochondrial superoxide levels. Patient cells failed to maintain levels of the essential thiol peptide antioxidant glutathione and experienced a decrease in cellular viability. Our findings indicate that chronic PPAR activation is a plausible initiator of long-term pathogenesis in VLCAD deficiency. Our findings further implicate disruption of redox homeostasis as a key pathogenic mechanism in VLCAD deficiency and support the notion that a deranged thiol metabolism might be an important pathogenic factor in VLCAD deficiency.
Collapse
Affiliation(s)
- Martin Lund
- Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University and Aarhus University Hospital, Palle Juel-Jensens Boulevard 99, 8200 Aarhus, Denmark
| | - Kathrine G Andersen
- Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University and Aarhus University Hospital, Palle Juel-Jensens Boulevard 99, 8200 Aarhus, Denmark
| | - Robert Heaton
- School of Pharmacy, Liverpool John Moore University, Byrom Street, Liverpool L3 3AF, United Kingdom
| | - Iain P Hargreaves
- School of Pharmacy, Liverpool John Moore University, Byrom Street, Liverpool L3 3AF, United Kingdom
| | - Niels Gregersen
- Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University and Aarhus University Hospital, Palle Juel-Jensens Boulevard 99, 8200 Aarhus, Denmark
| | - Rikke K J Olsen
- Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University and Aarhus University Hospital, Palle Juel-Jensens Boulevard 99, 8200 Aarhus, Denmark.
| |
Collapse
|
39
|
Jardim FR, Almeida FJSD, Luckachaki MD, Oliveira MRD. Effects of sulforaphane on brain mitochondria: mechanistic view and future directions. J Zhejiang Univ Sci B 2021; 21:263-279. [PMID: 32253837 DOI: 10.1631/jzus.b1900614] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The organosulfur compound sulforaphane (SFN; C6H11NOS2) is a potent cytoprotective agent promoting antioxidant, anti-inflammatory, antiglycative, and antimicrobial effects in in vitro and in vivo experimental models. Mitochondria are the major site of adenosine triphosphate (ATP) production due to the work of the oxidative phosphorylation (OXPHOS) system. They are also the main site of reactive oxygen species (ROS) production in nucleated human cells. Mitochondrial impairment is central in several human diseases, including neurodegeneration and metabolic disorders. In this paper, we describe and discuss the effects and mechanisms of action by which SFN modulates mitochondrial function and dynamics in mammalian cells. Mitochondria-related pro-apoptotic effects promoted by SFN in tumor cells are also discussed. SFN may be considered a cytoprotective agent, at least in part, because of the effects this organosulfur agent induces in mitochondria. Nonetheless, there are certain points that should be addressed in further experiments, indicated here as future directions, which may help researchers in this field of research.
Collapse
Affiliation(s)
- Fernanda Rafaela Jardim
- Forensic Institute, Forensic Toxicology Division, Postmortem Toxicology Sector, CEP 90160-093, Porto Alegre, RS, Brazil
| | - Fhelipe Jolner Souza de Almeida
- Postgraduate Program in Health Sciences (PPGCS), Federal University of Mato Grosso (UFMT), CEP 78060-900, Cuiaba, MT, Brazil
| | | | - Marcos Roberto de Oliveira
- Postgraduate Program in Chemistry (PPGQ), Federal University of Mato Grosso (UFMT), CEP 78060-900, Cuiaba, MT, Brazil.,Department of Biochemistry Prof. "Tuiskon Dick", Federal University of Rio Grande do Sul (UFRGS), CEP 90035-000, Porto Alegre, RS, Brazil
| |
Collapse
|
40
|
Gerasimov NY, Ivanenko GF, Bobkova NV, Nevrova OV, Goloshchapov AN. Investigation of Changes in the Microviscosity of the Erythrocyte Membranes and Glutathione in the Plasma of Animals with an Experimental Pathology Type Alzheimer’s Diseases. RUSSIAN JOURNAL OF PHYSICAL CHEMISTRY B 2021. [DOI: 10.1134/s1990793121010176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
41
|
Carter JL, Hege K, Yang J, Kalpage HA, Su Y, Edwards H, Hüttemann M, Taub JW, Ge Y. Targeting multiple signaling pathways: the new approach to acute myeloid leukemia therapy. Signal Transduct Target Ther 2020; 5:288. [PMID: 33335095 PMCID: PMC7746731 DOI: 10.1038/s41392-020-00361-x] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 02/06/2023] Open
Abstract
Acute myeloid leukemia (AML) is the most common form of acute leukemia in adults and the second most common form of acute leukemia in children. Despite this, very little improvement in survival rates has been achieved over the past few decades. This is partially due to the heterogeneity of AML and the need for more targeted therapeutics than the traditional cytotoxic chemotherapies that have been a mainstay in therapy for the past 50 years. In the past 20 years, research has been diversifying the approach to treating AML by investigating molecular pathways uniquely relevant to AML cell proliferation and survival. Here we review the development of novel therapeutics in targeting apoptosis, receptor tyrosine kinase (RTK) signaling, hedgehog (HH) pathway, mitochondrial function, DNA repair, and c-Myc signaling. There has been an impressive effort into better understanding the diversity of AML cell characteristics and here we highlight important preclinical studies that have supported therapeutic development and continue to promote new ways to target AML cells. In addition, we describe clinical investigations that have led to FDA approval of new targeted AML therapies and ongoing clinical trials of novel therapies targeting AML survival pathways. We also describe the complexity of targeting leukemia stem cells (LSCs) as an approach to addressing relapse and remission in AML and targetable pathways that are unique to LSC survival. This comprehensive review details what we currently understand about the signaling pathways that support AML cell survival and the exceptional ways in which we disrupt them.
Collapse
Affiliation(s)
- Jenna L Carter
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI, USA.,MD/PhD Program, Wayne State University School of Medicine, Detroit, MI, USA
| | - Katie Hege
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jay Yang
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA.,Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Hasini A Kalpage
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| | - Yongwei Su
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA.,Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA.,National Engineering Laboratory for AIDS Vaccine, Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| | - Holly Edwards
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA.,Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Maik Hüttemann
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jeffrey W Taub
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI, USA. .,Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA. .,Division of Pediatric Hematology/Oncology, Children's Hospital of Michigan, Detroit, MI, USA. .,Department of Pediatrics, Wayne State University School of Medicine, Detroit, MI, USA.
| | - Yubin Ge
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI, USA. .,Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA. .,Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA.
| |
Collapse
|
42
|
Abstract
The testosterone decline is one of the potential causes of oxidative stress-induced anxiety and depressive behaviors, and cognitive impairment induces irreversible neuronal damage, which is not clearly understood. The orchidectomized rat model was used; the hippocampal neurons and anxiety behavior were analyzed. Adult male albino rats were divided into control and orchidectomy (ORX) groups, orchidectomy (ORX + T), and normal (Cont + T) groups. Testosterone propionate was used as a testosterone supplement. The anxiety and depressive-like behavior observed in ORX animals in the open field (OF) and elevated plus-maze experiments were effectively overturned in the ORX + T group. Studies on isolated hippocampus showed reduced antioxidant enzymes (SOD, CAT, and glutathione (GSH) compounds), increased lipid peroxidation (LPO), elevated caspase3, and reduced anti-apoptotic protein Bcl-2, and increased apoptotic nuclei in TUNEL staining of the hippocampus in the ORX rats. These observations indicate free radical-mediated neural damage. Testosterone presence promoted the antioxidant defense system and restored normal pyramidal neuron morphology in ORX + T. This study confirms that testosterone is indispensable in the normal adult hippocampus and deficiency seems to be a potential risk factor for neurodegenerative disorders. Besides, androgen appears to be a possible therapeutic strategy for treating depression/neurodegenerative diseases in aging men.
Collapse
Affiliation(s)
- Sakthi Jothi Muthu
- Department of Anatomy, Dr. Arcot Lakshmanasamy Mudaliar Postgraduate Institute of Basic Medical Sciences, University of Madras, Chennai, India
| | - Prakash Seppan
- Department of Anatomy, Dr. Arcot Lakshmanasamy Mudaliar Postgraduate Institute of Basic Medical Sciences, University of Madras, Chennai, India
| |
Collapse
|
43
|
Bjørklund G, Tinkov AA, Hosnedlová B, Kizek R, Ajsuvakova OP, Chirumbolo S, Skalnaya MG, Peana M, Dadar M, El-Ansary A, Qasem H, Adams JB, Aaseth J, Skalny AV. The role of glutathione redox imbalance in autism spectrum disorder: A review. Free Radic Biol Med 2020; 160:149-162. [PMID: 32745763 DOI: 10.1016/j.freeradbiomed.2020.07.017] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 07/02/2020] [Accepted: 07/13/2020] [Indexed: 12/22/2022]
Abstract
The role of glutathione in autism spectrum disorder (ASD) is emerging as a major topic, due to its role in the maintenance of the intracellular redox balance. Several studies have implicated glutathione redox imbalance as a leading factor in ASD, and both ASD and many other neurodevelopmental disorders involve low levels of reduced glutathione (GSH), high levels of oxidized glutathione (GSSG), and abnormalities in the expressions of glutathione-related enzymes in the blood or brain. Glutathione metabolism, through its impact on redox environment or redox-independent mechanisms, interferes with multiple mechanisms involved in ASD pathogenesis. Glutathione-mediated regulation of glutamate receptors [e.g., N-methyl-d-aspartate (NMDA) receptor], as well as the role of glutamate as a substrate for glutathione synthesis, may be involved in the regulation of glutamate excitotoxicity. However, the interaction between glutathione and glutamate in the pathogenesis of brain diseases may vary from synergism to antagonism. Modulation of glutathione is also associated with regulation of redox-sensitive transcription factors nuclear factor kappa B (NF-κB) and activator protein 1 (AP-1) and downstream signaling (proinflammatory cytokines and inducible enzymes), thus providing a significant impact on neuroinflammation. Mitochondrial dysfunction, as well as neuronal apoptosis, may also provide a significant link between glutathione metabolism and ASD. Furthermore, it has been recently highlighted that glutathione can affect and modulate DNA methylation and epigenetics. Review analysis including research studies meeting the required criteria for analysis showed statistically significant differences between the plasma GSH and GSSG levels as well as GSH:GSSG ratio in autistic patients compared with healthy individuals (P = 0.0145, P = 0.0150 and P = 0.0202, respectively). Therefore, the existing data provide a strong background on the role of the glutathione system in ASD pathogenesis. Future research is necessary to investigate the role of glutathione redox signaling in ASD, which could potentially also lead to promising therapeutics.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine (CONEM), Mo I Rana, Norway.
| | - Alexey A Tinkov
- IM Sechenov First Moscow State Medical University, Moscow, Russia; Yaroslavl State University, Yaroslavl, Russia; Federal Research Centre of Biological Systems, Agro-technologies of the Russian Academy of Sciences, Orenburg, Russia
| | - Božena Hosnedlová
- Department of Human Pharmacology and Toxicology, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences Brno, Brno, Czech Republic; CONEM Metallomics Nanomedicine Research Group (CMNRG), Brno, Czech Republic
| | - Rene Kizek
- Department of Human Pharmacology and Toxicology, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences Brno, Brno, Czech Republic; CONEM Metallomics Nanomedicine Research Group (CMNRG), Brno, Czech Republic; Faculty of Pharmacy with Division of Laboratory Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Olga P Ajsuvakova
- IM Sechenov First Moscow State Medical University, Moscow, Russia; Yaroslavl State University, Yaroslavl, Russia; Federal Research Centre of Biological Systems, Agro-technologies of the Russian Academy of Sciences, Orenburg, Russia
| | - Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy; CONEM Scientific Secretary, Verona, Italy
| | - Margarita G Skalnaya
- IM Sechenov First Moscow State Medical University, Moscow, Russia; Federal Research Centre of Biological Systems, Agro-technologies of the Russian Academy of Sciences, Orenburg, Russia
| | | | - Maryam Dadar
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Afaf El-Ansary
- Medicinal Chemistry Department, King Saud University, Riyadh, Saudi Arabia; Autism Research and Treatment Center, Riyadh, Saudi Arabia; CONEM Saudi Autism Research Group, King Saud University, Riyadh, Saudi Arabia
| | - Hanan Qasem
- Autism Research and Treatment Center, Riyadh, Saudi Arabia; CONEM Saudi Autism Research Group, King Saud University, Riyadh, Saudi Arabia
| | - James B Adams
- School for Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ, USA
| | - Jan Aaseth
- Research Department, Innlandet Hospital Trust, Brumunddal, Norway
| | - Anatoly V Skalny
- IM Sechenov First Moscow State Medical University, Moscow, Russia; Federal Research Centre of Biological Systems, Agro-technologies of the Russian Academy of Sciences, Orenburg, Russia
| |
Collapse
|
44
|
Novel cascade reaction-based fluorescent cyanine chemosensor for cysteine detection and bioimaging in living system. Talanta 2020; 219:121291. [DOI: 10.1016/j.talanta.2020.121291] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 06/09/2020] [Accepted: 06/10/2020] [Indexed: 02/07/2023]
|
45
|
Firdaus Z, Singh N, Prajapati SK, Krishnamurthy S, Singh TD. Centella asiatica prevents D-galactose-Induced cognitive deficits, oxidative stress and neurodegeneration in the adult rat brain. Drug Chem Toxicol 2020; 45:1417-1426. [DOI: 10.1080/01480545.2020.1833907] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Zeba Firdaus
- Department of Medicinal Chemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Neha Singh
- Department of Pathology, MSR Medical College, Bengaluru, India
| | - Santosh Kumar Prajapati
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, BHU, Varanasi, India
| | - Sairam Krishnamurthy
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, BHU, Varanasi, India
| | - Tryambak Deo Singh
- Department of Medicinal Chemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
46
|
Bjørklund G, Peana M, Maes M, Dadar M, Severin B. The glutathione system in Parkinson's disease and its progression. Neurosci Biobehav Rev 2020; 120:470-478. [PMID: 33068556 DOI: 10.1016/j.neubiorev.2020.10.004] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 09/25/2020] [Accepted: 10/05/2020] [Indexed: 02/07/2023]
Abstract
Redox dysfunctions and neuro-oxidative stress play a major role in the pathophysiology and progression of Parkinson's disease (PD). Glutathione (GSH) and the reduced/oxidized glutathione (GSH/GSSG) ratio are lowered in oxidative stress conditions and may lead to increased oxidative toxicity. GSH is involved not only in neuro-immune and neuro-oxidative processes, including thiol redox signaling, but also in cell proliferation and differentiation and in the regulation of cell death, including apoptotic pathways. Lowered GSH metabolism and a low GSH/GSSG ratio following oxidative stress are associated with mitochondrial dysfunctions and constitute a critical factor in the neuroinflammatory and neurodegenerative processes accompanying PD. This review provides indirect evidence that GSH redox signaling is associated with the pathophysiology of PD. Nevertheless, it has not been delineated whether GSH redox imbalances are a causative factor in PD or whether PD-associated pathways cause the GSH redox imbalances in PD. The results show that antioxidant approaches, including neuroprotective and anti-neuroinflammatory agents, which neutralize reactive oxygen species, may have therapeutic efficacy in the treatment of PD and its progression.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine (CONEM), Mo i Rana, Norway.
| | | | - Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Impact Research Center, Deakin University, Geelong, Australia
| | - Maryam Dadar
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Beatrice Severin
- Faculty of Medicine, Ovidius University of Constanta, Constanta, Romania
| |
Collapse
|
47
|
Marí M, de Gregorio E, de Dios C, Roca-Agujetas V, Cucarull B, Tutusaus A, Morales A, Colell A. Mitochondrial Glutathione: Recent Insights and Role in Disease. Antioxidants (Basel) 2020; 9:antiox9100909. [PMID: 32987701 PMCID: PMC7598719 DOI: 10.3390/antiox9100909] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 09/17/2020] [Accepted: 09/19/2020] [Indexed: 02/08/2023] Open
Abstract
Mitochondria are the main source of reactive oxygen species (ROS), most of them deriving from the mitochondrial respiratory chain. Among the numerous enzymatic and non-enzymatic antioxidant systems present in mitochondria, mitochondrial glutathione (mGSH) emerges as the main line of defense for maintaining the appropriate mitochondrial redox environment. mGSH’s ability to act directly or as a co-factor in reactions catalyzed by other mitochondrial enzymes makes its presence essential to avoid or to repair oxidative modifications that can lead to mitochondrial dysfunction and subsequently to cell death. Since mitochondrial redox disorders play a central part in many diseases, harboring optimal levels of mGSH is vitally important. In this review, we will highlight the participation of mGSH as a contributor to disease progression in pathologies as diverse as Alzheimer’s disease, alcoholic and non-alcoholic steatohepatitis, or diabetic nephropathy. Furthermore, the involvement of mitochondrial ROS in the signaling of new prescribed drugs and in other pathologies (or in other unmet medical needs, such as gender differences or coronavirus disease of 2019 (COVID-19) treatment) is still being revealed; guaranteeing that research on mGSH will be an interesting topic for years to come.
Collapse
Affiliation(s)
- Montserrat Marí
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona-Spanish Council of Scientific Research, August Pi i Sunyer Biomedical Research Institute, 08036 Barcelona, Spain; (E.d.G.); (C.d.D.); (V.R.-A.); (B.C.); (A.T.)
- Correspondence: (M.M.); (A.M.); (A.C.); Tel.: +34-93-363-8300 (M.M.)
| | - Estefanía de Gregorio
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona-Spanish Council of Scientific Research, August Pi i Sunyer Biomedical Research Institute, 08036 Barcelona, Spain; (E.d.G.); (C.d.D.); (V.R.-A.); (B.C.); (A.T.)
| | - Cristina de Dios
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona-Spanish Council of Scientific Research, August Pi i Sunyer Biomedical Research Institute, 08036 Barcelona, Spain; (E.d.G.); (C.d.D.); (V.R.-A.); (B.C.); (A.T.)
- Departament de Biomedicina, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Vicente Roca-Agujetas
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona-Spanish Council of Scientific Research, August Pi i Sunyer Biomedical Research Institute, 08036 Barcelona, Spain; (E.d.G.); (C.d.D.); (V.R.-A.); (B.C.); (A.T.)
| | - Blanca Cucarull
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona-Spanish Council of Scientific Research, August Pi i Sunyer Biomedical Research Institute, 08036 Barcelona, Spain; (E.d.G.); (C.d.D.); (V.R.-A.); (B.C.); (A.T.)
- Departament de Biomedicina, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Anna Tutusaus
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona-Spanish Council of Scientific Research, August Pi i Sunyer Biomedical Research Institute, 08036 Barcelona, Spain; (E.d.G.); (C.d.D.); (V.R.-A.); (B.C.); (A.T.)
| | - Albert Morales
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona-Spanish Council of Scientific Research, August Pi i Sunyer Biomedical Research Institute, 08036 Barcelona, Spain; (E.d.G.); (C.d.D.); (V.R.-A.); (B.C.); (A.T.)
- Barcelona Clinic Liver Cancer Group, Liver Unit, Hospital Clínic, Network Center for Biomedical Research in Hepatic and Digestive Diseases (CIBEREHD), 08036 Barcelona, Spain
- Correspondence: (M.M.); (A.M.); (A.C.); Tel.: +34-93-363-8300 (M.M.)
| | - Anna Colell
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona-Spanish Council of Scientific Research, August Pi i Sunyer Biomedical Research Institute, 08036 Barcelona, Spain; (E.d.G.); (C.d.D.); (V.R.-A.); (B.C.); (A.T.)
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), 08036 Barcelona, Spain
- Correspondence: (M.M.); (A.M.); (A.C.); Tel.: +34-93-363-8300 (M.M.)
| |
Collapse
|
48
|
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: 2.8] [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
|
49
|
Oxidative Stress and Neuroinflammation Potentiate Each Other to Promote Progression of Dopamine Neurodegeneration. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6137521. [PMID: 32714488 PMCID: PMC7354668 DOI: 10.1155/2020/6137521] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/19/2020] [Accepted: 06/22/2020] [Indexed: 01/07/2023]
Abstract
Parkinson's disease (PD) is a chronic and complex disease of the central nervous system (CNS). Progressive loss of dopamine (DA) neurons in midbrain substantia nigra is considered to be the main cause of PD. The hallmark of PD pathology is the formation of Lewy bodies and the deposition of α-synuclein (α-syn). The mechanisms responsible for the progressive feature of DA neurodegeneration are not fully illustrated. Recently, oxidative stress and neuroinflammation have received extensive attention as two important entry points in the pathogenesis of PD. The occurrence of oxidative stress and neuroinflammation is usually derived from external influences or changes in internal environment, such as the accumulation of reactive oxygen species, exposure to a toxic environment, and the transformation of systemic inflammation. However, PD never results from a single independent factor and the simultaneous participation of oxidative stress and neuroinflammation contributed to PD development. Oxidative stress and neuroinflammation could potentiate each other to promote progression of PD. In this review, we briefly summarized the conditions of oxidative stress and neuroinflammation and the crosstalk between oxidative stress and neuroinflammation on the development of PD.
Collapse
|
50
|
Forrester SJ, Preston KJ, Cooper HA, Boyer MJ, Escoto KM, Poltronetti AJ, Elliott KJ, Kuroda R, Miyao M, Sesaki H, Akiyama T, Kimura Y, Rizzo V, Scalia R, Eguchi S. Mitochondrial Fission Mediates Endothelial Inflammation. Hypertension 2020; 76:267-276. [PMID: 32389075 PMCID: PMC7289685 DOI: 10.1161/hypertensionaha.120.14686] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 03/31/2020] [Indexed: 01/04/2023]
Abstract
Endothelial inflammation and mitochondrial dysfunction have been implicated in cardiovascular diseases, yet, a unifying mechanism tying them together remains limited. Mitochondrial dysfunction is frequently associated with mitochondrial fission/fragmentation mediated by the GTPase Drp1 (dynamin-related protein 1). Nuclear factor (NF)-κB, a master regulator of inflammation, is implicated in endothelial dysfunction and resultant complications. Here, we explore a causal relationship between mitochondrial fission and NF-κB activation in endothelial inflammatory responses. In cultured endothelial cells, TNF-α (tumor necrosis factor-α) or lipopolysaccharide induces mitochondrial fragmentation. Inhibition of Drp1 activity or expression suppresses mitochondrial fission, NF-κB activation, vascular cell adhesion molecule-1 induction, and leukocyte adhesion induced by these proinflammatory factors. Moreover, attenuations of inflammatory leukocyte adhesion were observed in Drp1 heterodeficient mice as well as endothelial Drp1 silenced mice. Intriguingly, inhibition of the canonical NF-κB signaling suppresses endothelial mitochondrial fission. Mechanistically, NF-κB p65/RelA seems to mediate inflammatory mitochondrial fission in endothelial cells. In addition, the classical anti-inflammatory drug, salicylate, seems to maintain mitochondrial fission/fusion balance against TNF-α via inhibition of NF-κB. In conclusion, our results suggest a previously unknown mechanism whereby the canonical NF-κB cascade and a mitochondrial fission pathway interdependently regulate endothelial inflammation.
Collapse
Affiliation(s)
- Steven J. Forrester
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, U.S.A
| | - Kyle J. Preston
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, U.S.A
| | - Hannah A. Cooper
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, U.S.A
| | - Michael J. Boyer
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, U.S.A
| | - Kathleen M. Escoto
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, U.S.A
| | - Anthony J. Poltronetti
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, U.S.A
| | - Katherine J. Elliott
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, U.S.A
| | - Ryohei Kuroda
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, U.S.A
| | - Masashi Miyao
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, U.S.A
- Department of Forensic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiromi Sesaki
- Department of Cell Biology, Johns Hopkins School of Medicine, Baltimore, MD, U.S.A
| | - Tomoko Akiyama
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Yayoi Kimura
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Victor Rizzo
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, U.S.A
| | - Rosario Scalia
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, 3500 N. Broad Street, Philadelphia, PA19140
| | - Satoru Eguchi
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, U.S.A
| |
Collapse
|