1
|
Luo Y, Liu X, Chen Y, Tang Q, He C, Ding X, Hu J, Cai Z, Li X, Qiao H, Zou Z. Targeting PAX8 sensitizes ovarian cancer cells to ferroptosis by inhibiting glutathione synthesis. Apoptosis 2024; 29:1499-1514. [PMID: 38853202 DOI: 10.1007/s10495-024-01985-y] [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] [Accepted: 05/24/2024] [Indexed: 06/11/2024]
Abstract
Ovarian cancer is a malignant tumor originating from the ovary, characterized by its high mortality rate and propensity for recurrence. In some patients, especially those with recurrent cancer, conventional treatments such as surgical resection or standard chemotherapy yield suboptimal results. Consequently, there is an urgent need for novel anti-cancer therapeutic strategies. Ferroptosis is a distinct form of cell death separate from apoptosis. Ferroptosis inducers have demonstrated promising potential in the treatment of ovarian cancer, with evidence indicating their ability to enhance ovarian cancer cell sensitivity to cisplatin. However, resistance of cancer cells to ferroptosis still remains an inevitable challenge. Here, we analyzed genome-scale CRISPR-Cas9 loss-of function screens and identified PAX8 as a ferroptosis resistance protein in ovarian cancer. We identified PAX8 as a susceptibility gene in GPX4-dependent ovarian cancer. Depletion of PAX8 rendered GPX4-dependent ovarian cancer cells significantly more sensitive to GPX4 inhibitors. Additionally, we found that PAX8 inhibited ferroptosis in ovarian cancer cells. Combined treatment with a PAX8 inhibitor and RSL3 suppressed ovarian cancer cell growth, induced ferroptosis, and was validated in a xenograft mouse model. Further exploration of the molecular mechanisms underlying PAX8 inhibition of ferroptosis mutations revealed upregulation of glutamate-cysteine ligase catalytic subunit (GCLC) expression. GCLC mediated the ferroptosis resistance induced by PAX8 in ovarian cancer. In conclusion, our study underscores the pivotal role of PAX8 as a therapeutic target in GPX4-dependent ovarian cancer. The combination of PAX8 inhibitors such as losartan and captopril with ferroptosis inducers represents a promising new approach for ovarian cancer therapy.
Collapse
Affiliation(s)
- Yanlin Luo
- Institute of Clinical Pharmacology, School of Basic Medical Science, Zhengzhou University, Zhengzhou, 450001, China
- Department of Gynecologic Oncology, The Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), Zhengzhou, 450008, China
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Xiaoli Liu
- The Fourth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510631, China
| | - Yibing Chen
- Genetic and Prenatal Diagnosis Center, Department of Gynecology and Obstetrics, First Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052, China
| | - Qing Tang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Chengsi He
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Xinyi Ding
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Jiachun Hu
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Zheyou Cai
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Xiang Li
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Hailing Qiao
- Institute of Clinical Pharmacology, School of Basic Medical Science, Zhengzhou University, Zhengzhou, 450001, China.
| | - Zhengzhi Zou
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China.
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China.
- Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou, 510631, China.
| |
Collapse
|
2
|
Niu C, Dong M, Niu Y. Role of Glutathione in Parkinson's Disease Pathophysiology and Therapeutic Potential of Polyphenols. Phytother Res 2024. [PMID: 39290049 DOI: 10.1002/ptr.8342] [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: 04/22/2024] [Revised: 08/05/2024] [Accepted: 09/02/2024] [Indexed: 09/19/2024]
Abstract
Oxidative stress is recognized to have a central role in the initiation and progression of Parkinson's disease (PD). Within the brain, neurons are particularly sensitive to oxidation due in part to their weak intrinsic antioxidant defense. Theoretically, neurons mostly depend on neighboring astrocytes to provide antioxidant protection by supplying cysteine-containing products for glutathione (GSH) synthesis. Astrocytes and neurons possess several amino acid transport systems for GSH and its precursors. Indeed, GSH is the most abundant intrinsic antioxidant in the central nervous system. The GSH depletion and/or alterations in its metabolism in the brain contribute to the pathogenesis of PD. Noteworthy, polyphenols possess potent antioxidant activity and can augment the GSH redox system. Numerous in vitro and in vivo studies have indicated that polyphenols exhibit potent neuroprotective effects in PD. Epidemiological studies have found an association between the consumption of dietary polyphenols and a lower PD risk. In this review, we summarize current knowledge on the biosynthesis and metabolism of GSH in the brain, with an emphasis on their contribution and therapeutic potential in PD. In particular, we focus on polyphenols that can increase brain GSH levels against PD. Furthermore, some current challenges and future perspectives for polyphenol-based therapies are also discussed.
Collapse
Affiliation(s)
- Chengu Niu
- Internal Medicine Residency Program, Rochester General Hospital, Rochester, New York, USA
| | - Miaoxian Dong
- The Institute of Medicine, Qiqihar Medical University, Qiqihar, China
| | - Yingcai Niu
- The Institute of Medicine, Qiqihar Medical University, Qiqihar, China
| |
Collapse
|
3
|
Lorenc-Koci E, Górny M, Chwatko G, Kamińska K, Iciek M, Rogóż Z. The effect of phencyclidine-mediated blockade of NMDA receptors in the early postnatal period on glutathione and sulfur amino acid levels in the rat brain as a potential causative factor of schizophrenia-like behavior in adulthood. Pharmacol Rep 2024; 76:863-877. [PMID: 38904712 PMCID: PMC11294273 DOI: 10.1007/s43440-024-00607-3] [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: 03/12/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 06/22/2024]
Abstract
BACKGROUND Phencyclidine, an NMDA receptor antagonist, is frequently used to model behavioral and neurochemical changes correlated with schizophrenia in laboratory animals. The present study aimed to examine the effects of repeated administration of phencyclidine during early postnatal development on the contents of glutathione and sulfur-containing amino acids, as well as the activity of antioxidant enzymes in the brain of 12-day-old rats, and schizophrenia-like symptoms in adulthood. METHODS Male Sprague-Dawley pups were administered phencyclidine (10 mg/kg) or saline subcutaneously on the postnatal days p2, p6, p9 and p12. In 12-day-old pups, 4 h after the last dose of phencyclidine, the levels of glutathione, cysteine, methionine, and homocysteine, and the enzymatic activity of superoxide dismutase (SOD), glutathione peroxidase (GPx), and glutathione reductase (GR) were measured in the frontal cortex, hippocampus, and striatum. In 70-72-day-old rats, schizophrenia-like symptoms were assessed using behavioral tests. RESULTS Biochemical data showed that perinatal phencyclidine treatment significantly reduced glutathione and cysteine levels in all brain structures studied, methionine was diminished in the striatum, and homocysteine in both the frontal cortex and striatum. GR activity was increased in the frontal cortex while SODactivity was decreased in the hippocampus. Behaviorally, perinatal phencyclidine induced long-term deficits in social and cognitive function and a decrease in locomotor activity assessed as the time of walking. Finally, perinatal treatment with phencyclidine resulted in a significant reduction in body weight gain over time. CONCLUSION Our research provides further evidence for the usefulness of the phencyclidine-induced neurodevelopmental model of schizophrenia for studying the pathogenesis of schizophrenia.
Collapse
Affiliation(s)
- Elżbieta Lorenc-Koci
- Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, Kraków, 31-343, Poland.
| | - Magdalena Górny
- The Chair of Medical Biochemistry, Jagiellonian University Medical College, 7 Kopernika Street, Kraków, 31-034, Poland
| | - Grażyna Chwatko
- Department of Environmental Chemistry, University of Łódź, 163 Pomorska Street, Łódź, 90-236, Poland
| | - Kinga Kamińska
- Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, Kraków, 31-343, Poland
| | - Małgorzata Iciek
- The Chair of Medical Biochemistry, Jagiellonian University Medical College, 7 Kopernika Street, Kraków, 31-034, Poland
| | - Zofia Rogóż
- Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, Kraków, 31-343, Poland
| |
Collapse
|
4
|
Asantewaa G, Tuttle ET, Ward NP, Kang YP, Kim Y, Kavanagh ME, Girnius N, Chen Y, Rodriguez K, Hecht F, Zocchi M, Smorodintsev-Schiller L, Scales TQ, Taylor K, Alimohammadi F, Duncan RP, Sechrist ZR, Agostini-Vulaj D, Schafer XL, Chang H, Smith ZR, O'Connor TN, Whelan S, Selfors LM, Crowdis J, Gray GK, Bronson RT, Brenner D, Rufini A, Dirksen RT, Hezel AF, Huber AR, Munger J, Cravatt BF, Vasiliou V, Cole CL, DeNicola GM, Harris IS. Glutathione synthesis in the mouse liver supports lipid abundance through NRF2 repression. Nat Commun 2024; 15:6152. [PMID: 39034312 PMCID: PMC11271484 DOI: 10.1038/s41467-024-50454-2] [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: 01/19/2023] [Accepted: 07/12/2024] [Indexed: 07/23/2024] Open
Abstract
Cells rely on antioxidants to survive. The most abundant antioxidant is glutathione (GSH). The synthesis of GSH is non-redundantly controlled by the glutamate-cysteine ligase catalytic subunit (GCLC). GSH imbalance is implicated in many diseases, but the requirement for GSH in adult tissues is unclear. To interrogate this, we have developed a series of in vivo models to induce Gclc deletion in adult animals. We find that GSH is essential to lipid abundance in vivo. GSH levels are highest in liver tissue, which is also a hub for lipid production. While the loss of GSH does not cause liver failure, it decreases lipogenic enzyme expression, circulating triglyceride levels, and fat stores. Mechanistically, we find that GSH promotes lipid abundance by repressing NRF2, a transcription factor induced by oxidative stress. These studies identify GSH as a fulcrum in the liver's balance of redox buffering and triglyceride production.
Collapse
Affiliation(s)
- Gloria Asantewaa
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Emily T Tuttle
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Nathan P Ward
- Department of Metabolism and Physiology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Yun Pyo Kang
- Department of Metabolism and Physiology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Yumi Kim
- Department of Metabolism and Physiology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Madeline E Kavanagh
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA
- Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Nomeda Girnius
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Ying Chen
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA
| | - Katherine Rodriguez
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Fabio Hecht
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Marco Zocchi
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Leonid Smorodintsev-Schiller
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - TashJaé Q Scales
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Kira Taylor
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Fatemeh Alimohammadi
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Renae P Duncan
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Zachary R Sechrist
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Diana Agostini-Vulaj
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Xenia L Schafer
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, NY, USA
| | - Hayley Chang
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Zachary R Smith
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Thomas N O'Connor
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Sarah Whelan
- Leicester Cancer Research Centre, University of Leicester, Leicester, UK
| | - Laura M Selfors
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Jett Crowdis
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - G Kenneth Gray
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | | | - Dirk Brenner
- Experimental and Molecular Immunology, Dept. of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Odense Research Center for Anaphylaxis (ORCA), Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Alessandro Rufini
- Leicester Cancer Research Centre, University of Leicester, Leicester, UK
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milan, Italy
| | - Robert T Dirksen
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Aram F Hezel
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Aaron R Huber
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Joshua Munger
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Benjamin F Cravatt
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Vasilis Vasiliou
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA
| | - Calvin L Cole
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Gina M DeNicola
- Department of Metabolism and Physiology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Isaac S Harris
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA.
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA.
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
5
|
Kim K, Gao H, Li C, Li B. The glutathione biosynthesis is involved in metamorphosis, antioxidant function, and insecticide resistance in Tribolium castaneum. PEST MANAGEMENT SCIENCE 2024; 80:2698-2709. [PMID: 38308415 DOI: 10.1002/ps.7976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 12/06/2023] [Accepted: 01/13/2024] [Indexed: 02/04/2024]
Abstract
BACKGROUND Reduced glutathione (GSH) synthesis is vital for redox homeostasis, cell-cycle regulation and apoptosis, and immune function. The glutamate-cysteine ligase catalytic subunit (Gclc) is the first and rate-limiting enzyme in GSH synthesis, suggesting the potential use of Gclc as a pesticide target. However, the functional characterization of Gclc, especially its contribution in metamorphosis, antioxidant status and insecticide resistance, is unclear in Tribolium castaneum. RESULTS In this study, we identified and cloned Gclc from T. castaneum (TcGclc) and found that its expression began to increase significantly from the late larvae (LL) stage (3.491 ± 0.490-fold). Furthermore, RNA interference-mediated knockdown of TcGclc resulted in three types of aberration (100% total aberration rate) caused by the downregulation of genes related to the 20-hydroxyecdysone (20E) pathway. This deficiency was partially rescued by exogenous 20E treatment (53.1% ± 3.2%), but not by antioxidant. Moreover, in the TcGclc knockdown group, GSH content was decreased to 62.3%, and total antioxidant capacity, glutathione peroxidase and total superoxide dismutase activities were reduced by 14.6%, 83.6%, and 82.3%, respectively. In addition, treatment with different insecticides upregulated expression of TcGclc significantly compared with a control group during the late larval stage (P < 0.01). CONCLUSION Our results indicate that TcGclc has an extensive role in metamorphosis, antioxidant function and insecticide resistance in T. castaneum, thereby expanding our understanding of GSH functions and providing a scientific basis for pest control. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- KumChol Kim
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
- Department of Life-Science, University of Science, Pyongyang, Democratic People's Republic of Korea
| | - Han Gao
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Chengjun Li
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Bin Li
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| |
Collapse
|
6
|
Evans VJ, Wu X, Tran KK, Tabofunda SK, Ding L, Yin L, Edwards P, Zhang QY, Ding X, Van Winkle LS. Impact of aging and ergothioneine pre-treatment on naphthalene toxicity in lung. Toxicol Lett 2024; 397:89-102. [PMID: 38768835 PMCID: PMC11531314 DOI: 10.1016/j.toxlet.2024.05.014] [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: 09/25/2023] [Revised: 05/10/2024] [Accepted: 05/17/2024] [Indexed: 05/22/2024]
Abstract
Aging increases susceptibility to lung disease, but the topic is understudied, especially in relation to environmental exposures with the bulk of rodent studies using young adults. This study aims to define the pulmonary toxicity of naphthalene (NA) and the impacts of a dietary antioxidant, ergothioneine (ET), in the liver and lungs of middle-aged mice. NA causes a well-characterized pattern of conducting airway epithelial injury in the lung in young adult mice, but NA's toxicity has not been characterized in middle-aged mice, aged 1-1.5 years. ET is a dietary antioxidant that is synthesized by bacteria and fungi. The ET transporter (ETT), SLC22A4, is upregulated in tissues that experience high levels of oxidative stress. In this study, middle-aged male and female C57BL/6 J mice, maintained on an ET-free synthetic diet from conception, were gavaged with 70 mg/kg of ET for five consecutive days. On day 8, the mice were exposed to a single intraperitoneal NA dose of 50, 100, 150, or 200 mg/kg. At 24 hours post NA injection samples were collected and analyzed for ET concentration and reduced (GSH) and oxidized glutathione (GSSG) concentrations. Histopathology, morphometry, and gene expression were examined. Histopathology of mice exposed to 100 mg/kg of NA suggests reduction in toxicity in the terminal airways of both male (p ≤ 0.001) and female (p ≤ 0.05) middle-aged mice by the ET pretreatment. Our findings in this study are the first to document the toxicity of NA in middle-aged mice and show some efficacy of ET in reducing NA toxicity.
Collapse
Affiliation(s)
- Veneese Jb Evans
- Center for Health and the Environment, School of Veterinary Medicine, University of California-Davis, Davis, CA 95616-8732, USA
| | - Xiangmeng Wu
- Dept of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721-0207
| | - Kyle K Tran
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California-Davis, Davis, CA 95616-8732, USA
| | - Shanlea K Tabofunda
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California-Davis, Davis, CA 95616-8732, USA
| | - Liang Ding
- Dept of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721-0207
| | - Lei Yin
- Dept of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721-0207
| | - Patricia Edwards
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California-Davis, Davis, CA 95616-8732, USA
| | - Qing-Yu Zhang
- Dept of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721-0207
| | - Xinxin Ding
- Dept of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721-0207.
| | - Laura S Van Winkle
- Center for Health and the Environment, School of Veterinary Medicine, University of California-Davis, Davis, CA 95616-8732, USA; Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California-Davis, Davis, CA 95616-8732, USA.
| |
Collapse
|
7
|
El-Sayed HS, Saad AS, Tawfik WA, Adel A, Abdelmagid MA, Momenah MA, Azab DM, Omar SE, El-Habbaa AS, Bahshwan SMA, Alghamdi AM, El-Saadony MT, El-Tarabily KA, El-Mayet FS. The role of turmeric and black pepper oil nanoemulsion in attenuating cytokine storm triggered by duck hepatitis A virus type I (DHAV-I)-induced infection in ducklings. Poult Sci 2024; 103:103404. [PMID: 38242053 PMCID: PMC10831264 DOI: 10.1016/j.psj.2023.103404] [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: 11/03/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/21/2024] Open
Abstract
The cytokine storm induced by duck hepatitis A virus type 1 (DHAV-1) infection significantly contributes to severe, rapid deaths and economic losses in the duck industry in Egypt. This study aimed to investigate the potential inhibitory effect of a nanoemulsion containing turmeric and black pepper oil on the immune response and pathogenesis of DHAV-1 in ducklings. A total of 105 ducklings from nonvaccinated breeders were divided into 5 experimental groups, each comprising 21 birds. The negative control group (G1) remained noninfected with DHAV-1 and nontreated with nanoemulsion, while the positive control group (G2) was infected with DHAV-1 but not treated with nanoemulsion. The other 2 groups (G3, the supplemented group which was noninfected with DHAV-1), and group 4 (the prophylactic group G4) which was infected with DHAV-1, both received nanoemulsion throughout the experiment. Group 5 (G5, the therapeutic group), on the other hand, which was infected with DHAV-1 received nanoemulsion only from the onset of clinical signs. At 5 days old, the ducklings in the positive control (G2), the prophylactic (G4), and the therapeutic group (G5) were infected with DHAV-1. All the ducklings in the infected groups exhibited depression, anorexia, and opisthotonos, and their livers displayed various degrees of ecchymotic hemorrhage, liver enlargement, and microscopic pathological lesions. Notably, the positive control group (G2) experienced the most severe and pronounced effects compared to the other infected groups treated with the nanoemulsion. Meanwhile, the viral RNA loads were lower in the liver tissues of the infected ducklings treated with the nanoemulsion (G4, and G5) compared to the positive control group G2. Additionally, the nanoemulsion effectively modulated proinflammatory cytokine expression, antioxidant enzymes, liver enzymes, and lipid profile of treated ducklings. In conclusion, the turmeric and black pepper oil nanoemulsion has the potential to be a therapeutic agent for regulating and modulating the immune response, decreasing DHAV-1-induced cytokine storms, and minimizing mortality and economic losses in the duck business. More research is needed to understand how turmeric and black pepper oil nanoemulsion alleviates DHVA-1-induced cytokine storms and lowers duckling mortality.
Collapse
Affiliation(s)
- Hemat S El-Sayed
- Department of Poultry Diseases, Animal Health Research Institute, Benha-Branch, Agriculture Research Center (ARC), Benha 12618, Egypt
| | - Aalaa S Saad
- Biotechnology Department, Animal Health Research Institute, Agriculture Research Center (ARC), Giza 12618, Egypt
| | - Wesam A Tawfik
- Holding Company for Biological Products and Vaccines, Dokki, Giza 12311, Egypt; NaQaa Nanotechnology Network (NNN), Giza, Egypt
| | - Amany Adel
- Reference Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, Agriculture Research Center (ARC), Giza 12618, Egypt
| | - Marwa A Abdelmagid
- Reference Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, Agriculture Research Center (ARC), Giza 12618, Egypt
| | - Maha Abdullah Momenah
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia
| | - Dalia M Azab
- Biochemistry Department (Pharmacology), Animal Health Research Institute, Benha-Branch, Agriculture Research Center (ARC), Benha 12618, Egypt
| | - Sabry E Omar
- Department of Poultry Diseases, Animal Health Research Institute, Benha-Branch, Agriculture Research Center (ARC), Benha 12618, Egypt
| | - Ayman S El-Habbaa
- Department of Virology, Faculty of Veterinary Medicine, Benha University, Moshtohor 13736, Qalyubia, Egypt
| | - Safia M A Bahshwan
- Biological Sciences Department, College of Science and Arts, King Abdulaziz University, Rabigh 21911, Saudi Arabia
| | - Amira M Alghamdi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohamed T El-Saadony
- Department of Agricultural Microbiology, Faculty of Agriculture, Zagazig University, Zagazig 44511, Egypt
| | - Khaled A El-Tarabily
- Department of Biology, College of Science, United Arab Emirates University, Al Ain 15551, United Arab Emirates.
| | - Fouad S El-Mayet
- Department of Virology, Faculty of Veterinary Medicine, Benha University, Moshtohor 13736, Qalyubia, Egypt; Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| |
Collapse
|
8
|
Grüning NM, Ralser M. Monogenic Disorders of ROS Production and the Primary Anti-Oxidative Defense. Biomolecules 2024; 14:206. [PMID: 38397443 PMCID: PMC10887155 DOI: 10.3390/biom14020206] [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: 01/01/2024] [Revised: 01/25/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
Oxidative stress, characterized by an imbalance between the production of reactive oxygen species (ROS) and the cellular anti-oxidant defense mechanisms, plays a critical role in the pathogenesis of various human diseases. Redox metabolism, comprising a network of enzymes and genes, serves as a crucial regulator of ROS levels and maintains cellular homeostasis. This review provides an overview of the most important human genes encoding for proteins involved in ROS generation, ROS detoxification, and production of reduced nicotinamide adenine dinucleotide phosphate (NADPH), and the genetic disorders that lead to dysregulation of these vital processes. Insights gained from studies on inherited monogenic metabolic diseases provide valuable basic understanding of redox metabolism and signaling, and they also help to unravel the underlying pathomechanisms that contribute to prevalent chronic disorders like cardiovascular disease, neurodegeneration, and cancer.
Collapse
Affiliation(s)
- Nana-Maria Grüning
- Department of Biochemistry, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany;
| | - Markus Ralser
- Department of Biochemistry, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany;
- The Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
- Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| |
Collapse
|
9
|
Timson RC, Khan A, Uygur B, Saad M, Yeh HW, DelGaudio NL, Weber R, Alwaseem H, Gao J, Yang C, Birsoy K. Development of a mouse model expressing a bifunctional glutathione-synthesizing enzyme to study glutathione limitation in vivo. J Biol Chem 2024; 300:105645. [PMID: 38218225 PMCID: PMC10869265 DOI: 10.1016/j.jbc.2024.105645] [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: 10/24/2023] [Revised: 12/17/2023] [Accepted: 01/03/2024] [Indexed: 01/15/2024] Open
Abstract
Glutathione (GSH) is a highly abundant tripeptide thiol that performs diverse protective and biosynthetic functions in cells. While changes in GSH availability are associated with inborn errors of metabolism, cancer, and neurodegenerative disorders, studying the limiting role of GSH in physiology and disease has been challenging due to its tight regulation. To address this, we generated cell and mouse models that express a bifunctional glutathione-synthesizing enzyme from Streptococcus thermophilus (GshF), which possesses both glutamate-cysteine ligase and glutathione synthase activities. GshF expression allows efficient production of GSH in the cytosol and mitochondria and prevents cell death in response to GSH depletion, but not ferroptosis induction, indicating that GSH is not a limiting factor under lipid peroxidation. CRISPR screens using engineered enzymes further revealed genes required for cell proliferation under cellular and mitochondrial GSH depletion. Among these, we identified the glutamate-cysteine ligase modifier subunit, GCLM, as a requirement for cellular sensitivity to buthionine sulfoximine, a glutathione synthesis inhibitor. Finally, GshF expression in mice is embryonically lethal but sustains postnatal viability when restricted to adulthood. Overall, our work identifies a conditional mouse model to investigate the limiting role of GSH in physiology and disease.
Collapse
Affiliation(s)
- Rebecca C Timson
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, New York, USA
| | - Artem Khan
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, New York, USA
| | - Beste Uygur
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, New York, USA
| | - Marwa Saad
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, New York, USA
| | - Hsi-Wen Yeh
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, New York, USA
| | - Nicole L DelGaudio
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, New York, USA
| | - Ross Weber
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Hanan Alwaseem
- The Proteomics Resource Center, The Rockefeller University, New York, New York, USA
| | - Jing Gao
- The CRISPR & Genome Editing Center, The Rockefeller University, New York, New York, USA
| | - Chingwen Yang
- The CRISPR & Genome Editing Center, The Rockefeller University, New York, New York, USA
| | - Kıvanç Birsoy
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, New York, USA.
| |
Collapse
|
10
|
Timson RC, Khan A, Uygur B, Saad M, Yeh HW, DelGaudio N, Weber R, Alwaseem H, Gao J, Yang C, Birsoy K. A mouse model to study glutathione limitation in vivo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.08.574722. [PMID: 38260639 PMCID: PMC10802487 DOI: 10.1101/2024.01.08.574722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Glutathione (GSH) is a highly abundant tripeptide thiol that performs diverse protective and biosynthetic functions in cells. While changes in GSH availability are linked to many diseases, including cancer and neurodegenerative disorders, determining the function of GSH in physiology and disease has been challenging due to its tight regulation. To address this, we generated cell and mouse models that express a bifunctional glutathione-synthesizing enzyme from Streptococcus Thermophilus (GshF). GshF expression allows efficient production of GSH in the cytosol and mitochondria and prevents cell death in response to GSH depletion, but not ferroptosis, indicating that GSH is not a limiting factor under lipid peroxidation. CRISPR screens using engineered enzymes revealed metabolic liabilities under compartmentalized GSH depletion. Finally, GshF expression in mice is embryonically lethal but sustains postnatal viability when restricted to adulthood. Overall, our work identifies a conditional mouse model to investigate the role of GSH availability in physiology and disease.
Collapse
|
11
|
Hecht F, Zocchi M, Alimohammadi F, Harris IS. Regulation of antioxidants in cancer. Mol Cell 2024; 84:23-33. [PMID: 38029751 PMCID: PMC10843710 DOI: 10.1016/j.molcel.2023.11.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/19/2023] [Accepted: 11/01/2023] [Indexed: 12/01/2023]
Abstract
Scientists in this field often joke, "If you don't have a mechanism, say it's ROS." Seemingly connected to every biological process ever described, reactive oxygen species (ROS) have numerous pleiotropic roles in physiology and disease. In some contexts, ROS act as secondary messengers, controlling a variety of signaling cascades. In other scenarios, they initiate damage to macromolecules. Finally, in their worst form, ROS are deadly to cells and surrounding tissues. A set of molecules with detoxifying abilities, termed antioxidants, is the direct counterpart to ROS. Notably, antioxidants exist in the public domain, touted as a "cure-all" for diseases. Research has disproved many of these claims and, in some cases, shown the opposite. Of all the diseases, cancer stands out in its paradoxical relationship with antioxidants. Although the field has made numerous strides in understanding the roles of antioxidants in cancer, many questions remain.
Collapse
Affiliation(s)
- Fabio Hecht
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642, USA; Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Marco Zocchi
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642, USA; Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Fatemeh Alimohammadi
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642, USA; Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Isaac S Harris
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642, USA; Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA.
| |
Collapse
|
12
|
Bejarano E, Weinberg J, Clark M, Taylor A, Rowan S, Whitcomb EA. Redox Regulation in Age-Related Cataracts: Roles for Glutathione, Vitamin C, and the NRF2 Signaling Pathway. Nutrients 2023; 15:3375. [PMID: 37571310 PMCID: PMC10421530 DOI: 10.3390/nu15153375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/19/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
Age is the biggest risk factor for cataracts, and aberrant oxidative modifications are correlated with age-related cataracts, suggesting that proper redox regulation is important for lens clarity. The lens has very high levels of antioxidants, including ascorbate and glutathione that aid in keeping the lens clear, at least in young animals and humans. We summarize current functional and genetic data supporting the hypothesis that impaired regulation of oxidative stress leads to redox dysregulation and cataract. We will focus on the essential endogenous antioxidant glutathione and the exogenous antioxidant vitamin C/ascorbate. Additionally, gene expression in response to oxidative stress is regulated in part by the transcription factor NRF2 (nuclear factor erythroid 2-related factor 2 [NFE2L2]), thus we will summarize our data regarding cataracts in Nrf2-/- mice. In this work, we discuss the function and integration of these capacities with the objective of maintaining lens clarity.
Collapse
Affiliation(s)
- Eloy Bejarano
- JM-USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA; (E.B.); (J.W.); (M.C.); (A.T.); (S.R.)
- School of Health Sciences and Veterinary, Universidad CEU Cardenal Herrera, CEU Universities, 46113 Valencia, Spain
| | - Jasper Weinberg
- JM-USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA; (E.B.); (J.W.); (M.C.); (A.T.); (S.R.)
| | - Madison Clark
- JM-USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA; (E.B.); (J.W.); (M.C.); (A.T.); (S.R.)
| | - Allen Taylor
- JM-USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA; (E.B.); (J.W.); (M.C.); (A.T.); (S.R.)
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA 02111, USA
- Department of Ophthalmology, School of Medicine, Tufts University, Boston, MA 02111, USA
- Department of Developmental, Chemical and Molecular Biology, Tufts University, Boston, MA 02111, USA
| | - Sheldon Rowan
- JM-USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA; (E.B.); (J.W.); (M.C.); (A.T.); (S.R.)
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA 02111, USA
- Department of Ophthalmology, School of Medicine, Tufts University, Boston, MA 02111, USA
| | - Elizabeth A. Whitcomb
- JM-USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA; (E.B.); (J.W.); (M.C.); (A.T.); (S.R.)
| |
Collapse
|
13
|
Yang Z, Zou S, Zhang Y, Zhang J, Zhang P, Xiao L, Xie Y, Meng M, Feng J, Kang L, Lee MH, Fang L. ACTL6A protects gastric cancer cells against ferroptosis through induction of glutathione synthesis. Nat Commun 2023; 14:4193. [PMID: 37443154 PMCID: PMC10345109 DOI: 10.1038/s41467-023-39901-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
Gastric cancer (GC), one of the most common malignant tumors in the world, exhibits a rapid metastasis rate and causes high mortality. Diagnostic markers and potential therapeutic targets for GCs are urgently needed. Here we show that Actin-like protein 6 A (ACTL6A), encoding an SWI/SNF subunit, is highly expressed in GCs. ACTL6A is found to be critical for regulating the glutathione (GSH) metabolism pathway because it upregulates γ-glutamyl-cysteine ligase catalytic subunit (GCLC) expression, thereby reducing reactive oxygen species (ROS) levels and inhibiting ferroptosis, a regulated form of cell death driven by the accumulation of lipid-based ROS. Mechanistic studies show that ACTL6A upregulates GCLC as a cotranscription factor with Nuclear factor (erythroid-derived 2)-like 2 (NRF2) and that the hydrophobic region of ACTL6A plays an important role. Our data highlight the oncogenic role of ACTL6A in GCs and indicate that inhibition of ACTL6A or GCLC could be a potential treatment strategy for GCs.
Collapse
Affiliation(s)
- Ziqing Yang
- Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Shaomin Zou
- Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Yijing Zhang
- Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Jieping Zhang
- Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Peng Zhang
- Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Lishi Xiao
- Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Yunling Xie
- Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Manqi Meng
- Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Junyan Feng
- Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Liang Kang
- Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China.
| | - Mong-Hong Lee
- Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China.
- Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China.
| | - Lekun Fang
- Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China.
- Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China.
| |
Collapse
|
14
|
Davidson EA, Chen Y, Singh S, Orlicky DJ, Thompson B, Wang Y, Charkoftaki G, Furnary TA, Cardone RL, Kibbey RG, Shearn CT, Nebert DW, Thompson DC, Vasiliou V. Endocrine pancreas-specific Gclc gene deletion causes a severe diabetes phenotype. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.13.544855. [PMID: 37398356 PMCID: PMC10312708 DOI: 10.1101/2023.06.13.544855] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Reduced glutathione (GSH) is an abundant antioxidant that regulates intracellular redox homeostasis by scavenging reactive oxygen species (ROS). Glutamate-cysteine ligase catalytic (GCLC) subunit is the rate-limiting step in GSH biosynthesis. Using the Pax6-Cre driver mouse line, we deleted expression of the Gclc gene in all pancreatic endocrine progenitor cells. Intriguingly, Gclc knockout (KO) mice, following weaning, exhibited an age-related, progressive diabetes phenotype, manifested as strikingly increased blood glucose and decreased plasma insulin levels. This severe diabetes trait is preceded by pathologic changes in islet of weanling mice. Gclc KO weanlings showed progressive abnormalities in pancreatic morphology including: islet-specific cellular vacuolization, decreased islet-cell mass, and alterations in islet hormone expression. Islets from newly-weaned mice displayed impaired glucose-stimulated insulin secretion, decreased insulin hormone gene expression, oxidative stress, and increased markers of cellular senescence. Our results suggest that GSH biosynthesis is essential for normal development of the mouse pancreatic islet, and that protection from oxidative stress-induced cellular senescence might prevent abnormal islet-cell damage during embryogenesis.
Collapse
|
15
|
Davies BM, Katayama JK, Monsivais JE, Adams JR, Dilts ME, Eberting AL, Hansen JM. Real-time analysis of dynamic compartmentalized GSH redox shifts and H 2O 2 availability in undifferentiated and differentiated cells. Biochim Biophys Acta Gen Subj 2023; 1867:130321. [PMID: 36870547 DOI: 10.1016/j.bbagen.2023.130321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 01/26/2023] [Accepted: 01/29/2023] [Indexed: 03/06/2023]
Abstract
BACKGROUND Glutathione (GSH) is the most abundant, small biothiol antioxidant. GSH redox state (Eh) supports developmental processes, yet with disrupted GSH Eh, poor developmental outcomes may occur. The role of subcellular, compartmentalized redox environments in the context of redox regulation of differentiation is not well understood. Here, using the P19 neurogenesis model of cellular differentiation, kinetics of subcellular H2O2 availability and GSH Eh were evaluated following oxidant exposure. METHODS Stably transfected P19 cell lines expressing H2O2 availability or GSH Eh sensors, Orp1-roGFP or Grx1-roGFP, respectively, targeted to the cytosol, mitochondria, or nucleus were used. Dynamic, compartmentalized changes in H2O2 availability and GSH Eh were measured via spectrophotometric and confocal microscopy over 120 min following treatment with H2O2 (100 μM) in both differentiated and undifferentiated cells. RESULTS Generally, treated undifferentiated cells showed a greater degree and duration of both H2O2 availability and GSH Eh disruption than differentiated neurons. In treated undifferentiated cells, H2O2 availability was similar in all compartments. Interestingly, in treated undifferentiated cells, mitochondrial GSH Eh was most affected in both the initial oxidation and the rebound kinetics compared to other compartments. Pretreatment with an Nrf2 inducer prevented H2O2-induced effects in all compartments of undifferentiated cells. CONCLUSIONS Disruption of redox-sensitive developmental pathways is likely stage specific, where cells that are less differentiated and/or are actively differentiating are most affected. GENERAL SIGNIFICANCE Undifferentiated cells are more susceptible to oxidant-induced redox dysregulation but are protected by chemicals that induce Nrf2. This may preserve developmental programs and diminish the potential for poor developmental outcomes.
Collapse
Affiliation(s)
- Brandon M Davies
- Cell Biology and Physiology Department, Brigham Young University, Provo, UT 84602, USA
| | - Jenna K Katayama
- Cell Biology and Physiology Department, Brigham Young University, Provo, UT 84602, USA
| | - Joshua E Monsivais
- Cell Biology and Physiology Department, Brigham Young University, Provo, UT 84602, USA
| | - James R Adams
- Cell Biology and Physiology Department, Brigham Young University, Provo, UT 84602, USA
| | - Miriam E Dilts
- Cell Biology and Physiology Department, Brigham Young University, Provo, UT 84602, USA
| | - Arielle L Eberting
- Cell Biology and Physiology Department, Brigham Young University, Provo, UT 84602, USA
| | - Jason M Hansen
- Cell Biology and Physiology Department, Brigham Young University, Provo, UT 84602, USA.
| |
Collapse
|
16
|
Fujii J, Osaki T, Soma Y, Matsuda Y. Critical Roles of the Cysteine-Glutathione Axis in the Production of γ-Glutamyl Peptides in the Nervous System. Int J Mol Sci 2023; 24:ijms24098044. [PMID: 37175751 PMCID: PMC10179188 DOI: 10.3390/ijms24098044] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
γ-Glutamyl moiety that is attached to the cysteine (Cys) residue in glutathione (GSH) protects it from peptidase-mediated degradation. The sulfhydryl group of the Cys residue represents most of the functions of GSH, which include electron donation to peroxidases, protection of reactive sulfhydryl in proteins via glutaredoxin, and glutathione conjugation of xenobiotics, whereas Cys-derived sulfur is also a pivotal component of some redox-responsive molecules. The amount of Cys that is available tends to restrict the capacity of GSH synthesis. In in vitro systems, cystine is the major form in the extracellular milieu, and a specific cystine transporter, xCT, is essential for survival in most lines of cells and in many primary cultivated cells as well. A reduction in the supply of Cys causes GPX4 to be inhibited due to insufficient GSH synthesis, which leads to iron-dependent necrotic cell death, ferroptosis. Cells generally cannot take up GSH without the removal of γ-glutamyl moiety by γ-glutamyl transferase (GGT) on the cell surface. Meanwhile, the Cys-GSH axis is essentially common to certain types of cells; primarily, neuronal cells that contain a unique metabolic system for intercellular communication concerning γ-glutamyl peptides. After a general description of metabolic processes concerning the Cys-GSH axis, we provide an overview and discuss the significance of GSH-related compounds in the nervous system.
Collapse
Affiliation(s)
- Junichi Fujii
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Yamagata 990-9585, Japan
| | - Tsukasa Osaki
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Yamagata 990-9585, Japan
| | - Yuya Soma
- Graduate School of Nursing, Yamagata University Faculty of Medicine, Yamagata 990-9585, Japan
| | - Yumi Matsuda
- Graduate School of Nursing, Yamagata University Faculty of Medicine, Yamagata 990-9585, Japan
| |
Collapse
|
17
|
Asantewaa G, Tuttle ET, Ward NP, Kang YP, Kim Y, Kavanagh ME, Girnius N, Chen Y, Duncan R, Rodriguez K, Hecht F, Zocchi M, Smorodintsev-Schiller L, Scales TQ, Taylor K, Alimohammadi F, Sechrist ZR, Agostini-Vulaj D, Schafer XL, Chang H, Smith Z, O'Connor TN, Whelan S, Selfors LM, Crowdis J, Gray GK, Bronson RT, Brenner D, Rufini A, Dirksen RT, Hezel AF, Huber AR, Munger J, Cravatt BF, Vasiliou V, Cole CL, DeNicola GM, Harris IS. Glutathione supports lipid abundance in vivo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.10.524960. [PMID: 36798186 PMCID: PMC9934595 DOI: 10.1101/2023.02.10.524960] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Cells rely on antioxidants to survive. The most abundant antioxidant is glutathione (GSH). The synthesis of GSH is non-redundantly controlled by the glutamate-cysteine ligase catalytic subunit (GCLC). GSH imbalance is implicated in many diseases, but the requirement for GSH in adult tissues is unclear. To interrogate this, we developed a series of in vivo models to induce Gclc deletion in adult animals. We find that GSH is essential to lipid abundance in vivo. GSH levels are reported to be highest in liver tissue, which is also a hub for lipid production. While the loss of GSH did not cause liver failure, it decreased lipogenic enzyme expression, circulating triglyceride levels, and fat stores. Mechanistically, we found that GSH promotes lipid abundance by repressing NRF2, a transcription factor induced by oxidative stress. These studies identify GSH as a fulcrum in the liver's balance of redox buffering and triglyceride production.
Collapse
Affiliation(s)
- Gloria Asantewaa
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, NY, USA, 14642
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14642
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14642
| | - Emily T Tuttle
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14642
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14642
| | - Nathan P Ward
- Department of Metabolism and Physiology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA, 33612
| | - Yun Pyo Kang
- Department of Metabolism and Physiology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA, 33612
| | - Yumi Kim
- Department of Metabolism and Physiology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA, 33612
| | - Madeline E Kavanagh
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA, 92037
| | - Nomeda Girnius
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA, 02115
| | - Ying Chen
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA, 06520
| | - Renae Duncan
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14642
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14642
| | - Katherine Rodriguez
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14642
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14642
| | - Fabio Hecht
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14642
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14642
| | - Marco Zocchi
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14642
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14642
| | - Leonid Smorodintsev-Schiller
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14642
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14642
| | - TashJaé Q Scales
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14642
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14642
| | - Kira Taylor
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14642
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14642
| | - Fatemeh Alimohammadi
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14642
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY, USA, 14642
| | - Zachary R Sechrist
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14642
- Department of Surgery and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA, 14642
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA, 14642
| | - Diana Agostini-Vulaj
- Department of Surgery and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA, 14642
| | - Xenia L Schafer
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, NY, USA, 14642
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14642
| | - Hayley Chang
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14642
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14642
| | - Zachary Smith
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14642
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14642
| | - Thomas N O'Connor
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14642
- Department of Surgery and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA, 14642
| | - Sarah Whelan
- Leicester Cancer Research Centre, University of Leicester, Leicester, LE2 7LX, UK
| | - Laura M Selfors
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA, 02115
| | - Jett Crowdis
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA, 02115
| | - G Kenneth Gray
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA, 02115
| | - Roderick T Bronson
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA, 02115
| | - Dirk Brenner
- Experimental & Molecular Immunology, Department of Infection and Immunity, Luxembourg Institute of Health, 29 Rue Henri Koch, Esch-sur-Alzette, Luxembourg
- Odense Research Center for Anaphylaxis (ORCA), Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Alessandro Rufini
- Leicester Cancer Research Centre, University of Leicester, Leicester, LE2 7LX, UK
- Dipartimento di Bioscienze, Università degli Studi di Milano, Via Celoria 26, 20133, Milan, Italy
| | - Robert T Dirksen
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY, USA, 14642
| | - Aram F Hezel
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14642
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14642
| | - Aaron R Huber
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA, 14642
| | - Josh Munger
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, NY, USA, 14642
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14642
| | - Benjamin F Cravatt
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA, 92037
| | - Vasilis Vasiliou
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA, 06520
| | - Calvin L Cole
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14642
- Department of Surgery and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA, 14642
| | - Gina M DeNicola
- Department of Metabolism and Physiology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA, 33612
| | - Isaac S Harris
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14642
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14642
| |
Collapse
|
18
|
Chen Y, Holland KD, Shertzer HG, Nebert DW, Dalton TP. Fatal Epileptic Seizures in Mice Having Compromised Glutathione and Ascorbic Acid Biosynthesis. Antioxidants (Basel) 2023; 12:antiox12020448. [PMID: 36830006 PMCID: PMC9952205 DOI: 10.3390/antiox12020448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 01/31/2023] [Accepted: 02/05/2023] [Indexed: 02/12/2023] Open
Abstract
Reduced glutathione (GSH) and ascorbic acid (AA) are the two most abundant low-molecular-weight antioxidants in mammalian tissues. GclmKO knockout mice lack the gene encoding the modifier subunit of the rate-limiting enzyme in GSH biosynthesis; GclmKO mice exhibit 10-40% of normal tissue GSH levels and show no overt phenotype. GuloKO knockout mice, lacking a functional Gulo gene encoding L-gulono-γ-lactone oxidase, cannot synthesize AA and depend on dietary ascorbic acid for survival. To elucidate functional crosstalk between GSH and AA in vivo, we generated the GclmKO/GuloKO double-knockout (DKO) mouse. DKO mice exhibited spontaneous epileptic seizures, proceeding to death between postnatal day (PND)14 and PND23. Histologically, DKO mice displayed neuronal loss and glial proliferation in the neocortex and hippocampus. Epileptic seizures and brain pathology in young DKO mice could be prevented with AA supplementation in drinking water (1 g/L). Remarkably, in AA-rescued adult DKO mice, the removal of AA supplementation for 2-3 weeks resulted in similar, but more severe, neocortex and hippocampal pathology and seizures, with death occurring between 12 and 21 days later. These results provide direct evidence for an indispensable, yet underappreciated, role for the interplay between GSH and AA in normal brain function and neuronal health. We speculate that the functional crosstalk between GSH and AA plays an important role in regulating glutamatergic neurotransmission and in protecting against excitotoxicity-induced brain damage.
Collapse
Affiliation(s)
- Ying Chen
- Department of Environmental and Public Health Sciences, Center for Environmental Genetics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT 06520, USA
- Correspondence: ; Tel.: +1-203-785-4694; Fax: +1-203-724-6023
| | - Katherine D. Holland
- Division of Neurology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Howard G. Shertzer
- Department of Environmental and Public Health Sciences, Center for Environmental Genetics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Daniel W. Nebert
- Department of Environmental and Public Health Sciences, Center for Environmental Genetics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- Departments of Pediatrics and Molecular & Developmental Biology, Cincinnati Children’s Research Center, Cincinnati, OH 45229, USA
| | - Timothy P. Dalton
- Department of Environmental and Public Health Sciences, Center for Environmental Genetics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| |
Collapse
|
19
|
Ren J, Wang B, Li L, Li S, Ma Y, Su L, Liu G, Liu Y, Dai Y. Glutathione ameliorates the meiotic defects of copper exposed ovine oocytes via inhibiting the mitochondrial dysfunctions. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 251:114530. [PMID: 36630773 DOI: 10.1016/j.ecoenv.2023.114530] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 12/13/2022] [Accepted: 01/07/2023] [Indexed: 05/28/2023]
Abstract
Regardless of the essential role of copper (Cu) in the physiological regulation process of mammalian reproduction, excessive exposure to Cu triggers the meiotic defects of porcine oocytes via compromising the mitochondrial functions. However, the connections between the excessive Cu exposure and meiotic defects of ovine oocytes have not been reported. In this study, the effect of copper sulfate (CuSO4) exposure on the meiotic potentials of ovine oocytes was analyzed. Subsequently, the ameliorative effect of glutathione (GSH) supplementation on the meiotic defects of CuSO4 exposed ovine oocytes was investigated. For these purposes, the in vitro maturation (IVM) of ovine cumulus oocyte complexes (COCs) was conducted in the presence of 5, 10, 20 and 40 μg/mL of CuSO4 supplementation. Subsequently, different concentrations of GSH (2, 4 and 8 mM) were added to the IVM medium containing CuSO4 solution. After IVM, the assay, including nuclear maturation, spindle organization, chromosome alignment, cytoskeleton assembly, cortical granule (CGs) dynamics, mitochondrial function, reactive oxygen species (ROS) generation, apoptosis, epigenetic modification and fertilization capacity of ovine oocytes were performed. The results showed that excessive Cu exposure triggered the meiotic defects of ovine oocytes via promoting the mitochondrial dysfunction related oxidative stress damage. Moreover, the GSH supplementation, not only ameliorated the decreased maturation potential and fertilization defect of CuSO4 exposed oocytes, but inhibited the mitochondrial dysfunction related oxidative stress damage, ROS generation, apoptosis and altered H3K27me3 expression in the CuSO4 exposed oocytes. Combined with the gene expression pattern, the finding in the present study provided fundamental bases for the ameliorative effect of GSH supplementation on the meiotic defects of CuSO4 exposed oocytes via inhibiting the mitochondrial dysfunctions, further benefiting these potential applications of GSH supplementation in the mammalian IVM system and livestock breeding suffering from the excessive Cu exposure.
Collapse
Affiliation(s)
- Jingyu Ren
- College of Life Science, Inner Mongolia University, No. 235 West Univ. Road, Hohhot, Zip Code: 010021, Inner Mongolia, China
| | - Biao Wang
- Animal Husbandry Institute, Inner Mongolia Academy of Agricultural & Animal Husbandry Sciences, No. 22 Zhaowuda Road, Hohhot, Zip Code: 010031, Inner Mongolia, China
| | - Liping Li
- College of Life Science, Inner Mongolia University, No. 235 West Univ. Road, Hohhot, Zip Code: 010021, Inner Mongolia, China
| | - Shubin Li
- Center of Reproductive Medicine, Inner Mongolia People's Hospital, No. 20 Zhaowuda Road, Hohhot, Zip Code: 010021, Inner Mongolia, China
| | - Yuzhen Ma
- Center of Reproductive Medicine, Inner Mongolia People's Hospital, No. 20 Zhaowuda Road, Hohhot, Zip Code: 010021, Inner Mongolia, China
| | - Liya Su
- Key Laboratory of Medical Cell Biology, Clinical Medicine Research Center, Affiliated Hospital of Inner Mongolia Medical University, No. 1 Tongdao North Street, Hohhot, Zip Code: 010050, Inner Mongolia, China
| | - Gang Liu
- Key Laboratory of Medical Cell Biology, Clinical Medicine Research Center, Affiliated Hospital of Inner Mongolia Medical University, No. 1 Tongdao North Street, Hohhot, Zip Code: 010050, Inner Mongolia, China.
| | - Yongbin Liu
- College of Life Science, Inner Mongolia University, No. 235 West Univ. Road, Hohhot, Zip Code: 010021, Inner Mongolia, China.
| | - Yanfeng Dai
- College of Life Science, Inner Mongolia University, No. 235 West Univ. Road, Hohhot, Zip Code: 010021, Inner Mongolia, China.
| |
Collapse
|
20
|
Xie K, Li Y, He G, Zhao X, Chen D, Yu B, Luo Y, Mao X, Huang Z, Yu J, Luo J, Zheng P, Yan H, Li H, He J. Daidzein supplementation improved fecundity in sows via modulation of ovarian oxidative stress and inflammation. J Nutr Biochem 2022; 110:109145. [PMID: 36049671 DOI: 10.1016/j.jnutbio.2022.109145] [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: 03/18/2021] [Revised: 10/15/2021] [Accepted: 08/10/2022] [Indexed: 01/13/2023]
Abstract
Adequate ovarian hormones secretion is essential for pregnancy success. Oxidative damage and following inflammation can destroy the ovarian normal function in mammals. Daidzein (DAI) is a classical isoflavonic phytoestrogen with specific oestrogenic activity. This study aimed to explore the effects of daidzein supplementation on fertility and ovarian characteristics of sows through biochemical analysis and RNA-seq technology. Twelve multiparous Yorkshire × Landrace sows were randomly divided into CON and DAI groups. We found that DAI increased total number of embryos as well as P4 and E2 levels of serum. DAI not only elevated the activities of T-AOC and GSH-Px, but also tended to decrease the content of MDA and IL-6 in the serum. In ovary, RNA-Seq identified 237 differentially expressed genes (DEGs), and GO analysis showed that these DEGs were linked to functions associated with immune dysfunction. Moreover, STRING analysis demonstrated that most interacting nodes were TLR-4, LCP2, and CD86. Furthermore, DAI decreased the content of MDA, IL-1β, IL-6, and TNF-α, and increased the activities of T-AOC and CAT in ovarian tissue. Interestingly, a partial mantel correlation showed that T-AOC was the strongest correlation between the ovarian dataset and selected DEGs. Additionally, DAI supplementation not only increased the protein expressions of Nrf2, HO-1, and NQO1, but also decreased the protein expressions of TLR-4, p-NFκB, p-AKT, and p-IκBα. Altogether, our results indicated that DAI could ameliorate ovarian oxidative stress and inflammation in sows, which might be mediated by suppressing the TLR4/NF-κB signaling pathway and activating the Nrf2/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Kunhong Xie
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China; Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan, P. R. China
| | - Yan Li
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China; Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan, P. R. China
| | - Guoru He
- New Hope Liuhe Co., Ltd. Sichuan Province, Chengdu, Sichuan, P. R. China
| | - Xuefeng Zhao
- Shandong Animal Product Quality and Safety Center, Jinan, Shangdong, P. R. China
| | - Daiwen Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China; Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan, P. R. China
| | - Bing Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China; Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan, P. R. China
| | - Yuheng Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China; Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan, P. R. China.
| | - Xiangbing Mao
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China; Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan, P. R. China
| | - Zhiqing Huang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China; Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan, P. R. China
| | - Jie Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China; Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan, P. R. China
| | - Junqiu Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China; Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan, P. R. China
| | - Ping Zheng
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China; Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan, P. R. China
| | - Hui Yan
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China; Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan, P. R. China
| | - Hua Li
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China; Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan, P. R. China
| | - Jun He
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China; Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan, P. R. China.
| |
Collapse
|
21
|
Jiang H, Sun M, Zhao Y, Liu G, Zhong L, Xue H, Chen X, Zheng Y, Wang M. The early function of cortisol in liver during Aeromonas hydrophila infection: Dynamics of the transcriptome and accessible chromatin landscapes. Front Immunol 2022; 13:989075. [PMID: 36532002 PMCID: PMC9751032 DOI: 10.3389/fimmu.2022.989075] [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: 07/08/2022] [Accepted: 11/17/2022] [Indexed: 12/02/2022] Open
Abstract
In China, channel catfish (Ictalurus punctatus) is an important aquaculture species; however, haemorrhagic disease (Aeromonas hydrophila induced disease) in these fish has caused tremendous economic loss due to high morbidity and mass mortality in the breeding industry. The role of cortisol in bacterial diseases, particularly in the acute phase, remains unclear. In this study, liver transcriptome (RNA-seq) and chromatin accessibility (ATAC-seq) analyses were employed to investigate the early functional role of cortisol in Aeromonas hydrophila-stimulated responses. Our experiments confirmed that A. hydrophila infection can initially significantly increase serum cortisol levels at 1 h after infection. At this time point, the increased serum cortisol levels can significantly regulate A. hydrophila-regulated genes by affecting both transcriptome and chromatin accessibility. Cross-analysis of RNA-seq and ATAC-seq revealed that a certain gene group (92 target_DEGs) was regulated at an early time point by cortisol. KEGG enrichment analysis revealed that the top three pathways according to target_DEGs were cancer, glutathione metabolism, and the Notch signalling pathway. The protein-protein interaction analysis of target_DEGs revealed that they may be primarily involved in cell proliferation, CD8+ T cell function, glutathione synthesis, and activation of the NF-κB signalling pathway. This suggests that after the emergence of immune stress, the early regulation of cortisol is positive against the immune response. It is possible that in this situation, the animal is attempting to avoid dangerous situations and risks and then cope with the imbalance produced by the stressor to ultimately restore homeostasis. Our results will contribute to future research on fish and provide valuable insight regarding the mechanism of immune regulation by cortisol and the study of bacterial haemorrhagic disease in channel catfish.
Collapse
|
22
|
Genetic Disruption of the γ-Glutamylcysteine Ligase in PDAC Cells Induces Ferroptosis-Independent Cell Death In Vitro without Affecting In Vivo Tumor Growth. Cancers (Basel) 2022; 14:cancers14133154. [PMID: 35804926 PMCID: PMC9264981 DOI: 10.3390/cancers14133154] [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: 03/23/2022] [Revised: 05/29/2022] [Accepted: 06/02/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary The newly described form of iron-dependent cell death, called ferroptosis, has emerged as a powerful strategy for eradicating cancer cells. This is of particular importance for pancreatic ductal adenocarcinoma (PDAC), which has been shown to be one of the most aggressive tumors, with a five-year overall survival of less than 8%. The aim of the present study is to identify the most potent and selective target for the induction of ferroptosis in PDAC cells. The results presented here are of great importance not only for the development of novel and more effective anti-cancer therapeutics, but also anticipate potential resistant mechanisms that cancer cells might deploy. This way, ferroptosis-based therapeutics may be a step ahead of highly adaptable cancer cells. Abstract The conceptualization of a novel type of cell death, called ferroptosis, opens new avenues for the development of more efficient anti-cancer therapeutics. In this context, a full understanding of the ferroptotic pathways, the players involved, their precise role, and dispensability is prerequisite. Here, we focused on the importance of glutathione (GSH) for ferroptosis prevention in pancreatic ductal adenocarcinoma (PDAC) cells. We genetically deleted a unique, rate-limiting enzyme for GSH biosynthesis, γ-glutamylcysteine ligase (GCL), which plays a key role in tumor cell proliferation and survival. Surprisingly, although glutathione peroxidase 4 (GPx4) has been described as a guardian of ferroptosis, depletion of its substrate (GSH) led preferentially to apoptotic cell death, while classical ferroptotic markers (lipid hydroperoxides) have not been observed. Furthermore, the sensitivity of PDAC cells to the pharmacological/genetic inhibition of GPx4 revealed GSH dispensability in this context. To the best of our knowledge, this is the first time that the complete dissection of the xCT-GSH-GPx4 axis in PDAC cells has been investigated in great detail. Collectively, our results revealed the necessary role of GSH in the overall redox homeostasis of PDAC cells, as well as the dispensability of this redox-active molecule for a specific, antioxidant branch dedicated to ferroptosis prevention.
Collapse
|
23
|
Pan F, Lin X, Hao L, Wang T, Song H, Wang R. The Critical Role of Ferroptosis in Hepatocellular Carcinoma. Front Cell Dev Biol 2022; 10:882571. [PMID: 35800895 PMCID: PMC9255949 DOI: 10.3389/fcell.2022.882571] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 06/03/2022] [Indexed: 12/14/2022] Open
Abstract
Liver cancer is the sixth most frequently diagnosed cancer and the third dominant cause of cancer death worldwide. Ferroptosis is characterized as an iron-dependent form of regulated cell death, with accumulation of lipid peroxides to lethal amounts. Evidences have showed that ferroptosis is closely associated with HCC, but the mechanisms are still poorly understood. In this review, we mainly summarize the roles of several typical molecules as well as radiotherapy in regulating the ferroptosis process in HCC. Chances are that this review may help address specific issues in the treatment of HCC.
Collapse
|
24
|
Malott KF, Reshel S, Ortiz L, Luderer U. Glutathione deficiency decreases lipid droplet stores and increases reactive oxygen species in mouse oocytes†. Biol Reprod 2022; 106:1218-1231. [PMID: 35238901 PMCID: PMC9198951 DOI: 10.1093/biolre/ioac032] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 01/06/2022] [Accepted: 02/01/2022] [Indexed: 12/30/2022] Open
Abstract
Glutathione (GSH) is a tripeptide thiol antioxidant that has been shown to be important to overall reproductive health. Glutamate cysteine ligase, the rate-limiting enzyme in GSH synthesis consists of a catalytic and a modifier (GCLM) subunit. We previously showed that oxidative stress in the ovary and oocytes of Gclm-/- mice is associated with accelerated age-related decline in ovarian follicles and decreased female fertility due to preimplantation embryonic mortality. Mammalian preimplantation development is a highly regulated and energy-intensive process that primarily relies on coordination between lipid droplets (LDs) and mitochondria to maintain cellular homeostasis. In this study, we hypothesized that GSH deficiency in oocytes increases oxidative stress, leading to increased mitochondrial dysfunction and decreased LD consumption, thereby decreasing oocyte developmental competence. We observed that Gclm-/- oocytes have increased oxidative stress, primarily in the form of mitochondrial superoxide and decreased subcortical mitochondrial clusters. Further, Gclm-/- oocytes have decreased mitochondrial membrane potential (ΔΨm) compared with Gclm+/+. We surmise this is likely due to the decreased availability of LDs, as we observed a significant decrease in LD content in Gclm-/- oocytes compared with Gclm+/+. The decreased oocyte LD content is likely related to an altered serum lipidome, with Gclm-/- serum having relatively lower unsaturated fatty acids and triglycerides than that of Gclm+/+ and Gclm+/- females. Altogether these data support that decreased LDs and increased oxidative stress are primary drivers of decreased oocyte developmental competence in GSH-deficient oocytes.
Collapse
Affiliation(s)
- Kelli F Malott
- Environmental Health Sciences Graduate Program, University of California, Irvine, CA, USA
- Department of Environmental and Occupational Health, University of California, Irvine, CA, USA
- Department of Medicine, University of California, Irvine, CA, USA
| | - Samantha Reshel
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA
| | - Laura Ortiz
- Department of Medicine, University of California, Irvine, CA, USA
| | - Ulrike Luderer
- Environmental Health Sciences Graduate Program, University of California, Irvine, CA, USA
- Department of Environmental and Occupational Health, University of California, Irvine, CA, USA
- Department of Medicine, University of California, Irvine, CA, USA
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA
| |
Collapse
|
25
|
Nascè A, Gariani K, Jornayvaz FR, Szanto I. NADPH Oxidases Connecting Fatty Liver Disease, Insulin Resistance and Type 2 Diabetes: Current Knowledge and Therapeutic Outlook. Antioxidants (Basel) 2022; 11:antiox11061131. [PMID: 35740032 PMCID: PMC9219746 DOI: 10.3390/antiox11061131] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/30/2022] [Accepted: 06/03/2022] [Indexed: 12/15/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), characterized by ectopic fat accumulation in hepatocytes, is closely linked to insulin resistance and is the most frequent complication of type 2 diabetes mellitus (T2DM). One of the features connecting NAFLD, insulin resistance and T2DM is cellular oxidative stress. Oxidative stress refers to a redox imbalance due to an inequity between the capacity of production and the elimination of reactive oxygen species (ROS). One of the major cellular ROS sources is NADPH oxidase enzymes (NOX-es). In physiological conditions, NOX-es produce ROS purposefully in a timely and spatially regulated manner and are crucial regulators of various cellular events linked to metabolism, receptor signal transmission, proliferation and apoptosis. In contrast, dysregulated NOX-derived ROS production is related to the onset of diverse pathologies. This review provides a synopsis of current knowledge concerning NOX enzymes as connective elements between NAFLD, insulin resistance and T2DM and weighs their potential relevance as pharmacological targets to alleviate fatty liver disease.
Collapse
Affiliation(s)
- Alberto Nascè
- Service of Endocrinology, Diabetes, Nutrition and Patient Therapeutic Education, Geneva University Hospitals, Rue Gabrielle-Perret-Gentil 4, 1205 Geneva, Switzerland; (A.N.); (K.G.)
| | - Karim Gariani
- Service of Endocrinology, Diabetes, Nutrition and Patient Therapeutic Education, Geneva University Hospitals, Rue Gabrielle-Perret-Gentil 4, 1205 Geneva, Switzerland; (A.N.); (K.G.)
- Department of Medicine, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
- Diabetes Center of the Faculty of Medicine, University of Geneva Medical School, 1211 Geneva, Switzerland
| | - François R. Jornayvaz
- Service of Endocrinology, Diabetes, Nutrition and Patient Therapeutic Education, Geneva University Hospitals, Rue Gabrielle-Perret-Gentil 4, 1205 Geneva, Switzerland; (A.N.); (K.G.)
- Department of Medicine, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
- Diabetes Center of the Faculty of Medicine, University of Geneva Medical School, 1211 Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
- Correspondence: (F.R.J.); (I.S.)
| | - Ildiko Szanto
- Service of Endocrinology, Diabetes, Nutrition and Patient Therapeutic Education, Geneva University Hospitals, Rue Gabrielle-Perret-Gentil 4, 1205 Geneva, Switzerland; (A.N.); (K.G.)
- Department of Medicine, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
- Diabetes Center of the Faculty of Medicine, University of Geneva Medical School, 1211 Geneva, Switzerland
- Correspondence: (F.R.J.); (I.S.)
| |
Collapse
|
26
|
Liu W, Wang X, Liu Y, Fang S, Wu Z, Han C, Shi W, Bao Y. Effects of early florfenicol exposure on glutathione signaling pathway and PPAR signaling pathway in chick liver. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 237:113529. [PMID: 35487170 DOI: 10.1016/j.ecoenv.2022.113529] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 04/10/2022] [Accepted: 04/14/2022] [Indexed: 06/14/2023]
Abstract
Florfenicol (FFC) is a common antibiotic for animals. The nonstandard and excessive use of FFC can cause veterinary drug residues in animals, pollute soil and marine environment, and even threaten human health. Therefore, it is necessary to study the toxicity and side effects of FFC on animals. Our previous studies have proved that FFC can cause liver injury in chicks, but there are few in-depth studies on the mechanism of FFC causing liver injury at the level of signaling pathway in chicks. Therefore, transcriptome and proteome sequencing were performed and combined analysis was performed. Sequencing results showed that 1989 genes and 917 proteins were significantly changed in chick livers after FFC exposure. These genes and proteins are related to redox, glutathione transferase activity and lipid metabolism. There are 9 significantly different genes and 7 significantly different proteins in glutathione signaling pathway. Oxidative stress may occur in the liver of chicks through the change of activation state of glutathione signaling pathway. And there are 13 significantly different genes and 18 significantly different proteins in PPAR signaling pathway. The changes of PPAR signaling pathway may induce lipid metabolism disorder in liver. The verification results of qPCR and PRM were consistent with the sequencing results. We also detected GSH-Px, GSH, GST, TG, TC and ANDP levels in liver. These changes of biochemical indicators directly confirmed oxidative stress and lipid metabolism disorders were occurred in the livers of chicks treated by FFC. In conclusion, FFC could induce liver injury in chicks by regulating the expression levels of significantly different genes and proteins in glutathione signaling pathway and PPAR signaling pathway.
Collapse
Affiliation(s)
- Wei Liu
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Xiao Wang
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Ying Liu
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Siyuan Fang
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Zhanjun Wu
- Institute of Grain and Oil Crops of Hebei Academy of Agriculture and Forestry Sciences, Shijiazhuang 050035, China
| | - Chao Han
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Wanyu Shi
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China; Hebei Provincial Veterinary Biotechnology Innovation Center, Baoding 071001, China.
| | - Yongzhan Bao
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China; Hebei Provincial Veterinary Biotechnology Innovation Center, Baoding 071001, China.
| |
Collapse
|
27
|
Liu W, Östberg NK, Yalcinkaya M, Dou H, Endo-Umeda K, Tang Y, Hou X, Xiao T, Filder T, Abramowicz S, Yang YG, Soehnlein O, Tall AR, Wang N. Erythroid lineage Jak2V617F expression promotes atherosclerosis through erythrophagocytosis and macrophage ferroptosis. J Clin Invest 2022; 132:155724. [PMID: 35587375 PMCID: PMC9246386 DOI: 10.1172/jci155724] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 05/10/2022] [Indexed: 11/17/2022] Open
Abstract
Elevated hematocrit is associated with cardiovascular risk; however, the causality and mechanisms are unclear. The JAK2V617F (Jak2VF) mutation increases cardiovascular risk in myeloproliferative disorders and in clonal hematopoiesis. Jak2VF mice with elevated WBCs, platelets, and RBCs display accelerated atherosclerosis and macrophage erythrophagocytosis. To investigate whether selective erythroid Jak2VF expression promotes atherosclerosis, we developed hyperlipidemic erythropoietin receptor Cre mice that express Jak2VF in the erythroid lineage (VFEpoR mice). VFEpoR mice without elevated blood cell counts showed increased atherosclerotic plaque necrosis, erythrophagocytosis, and ferroptosis. Selective induction of erythrocytosis with low-dose erythropoietin further exacerbated atherosclerosis with prominent ferroptosis, lipid peroxidation, and endothelial damage. VFEpoR RBCs had reduced antioxidant defenses and increased lipid hydroperoxides. Phagocytosis of human or murine WT or JAK2VF RBCs by WT macrophages induced ferroptosis, which was prevented by the ferroptosis inhibitor liproxstatin-1. Liproxstatin-1 reversed increased atherosclerosis, lipid peroxidation, ferroptosis, and endothelial damage in VFEpoR mice and in Jak2VF chimeric mice simulating clonal hematopoiesis, but had no impact in controls. Erythroid lineage Jak2VF expression led to qualitative and quantitative defects in RBCs that exacerbated atherosclerosis. Phagocytosis of RBCs by plaque macrophages promoted ferroptosis, suggesting a therapeutic target for reducing RBC-mediated cardiovascular risk.
Collapse
Affiliation(s)
- Wenli Liu
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, United States of America
| | - Nataliya K Östberg
- Physiology and Pharmacology (FyFA), Karolinska Institute, Stockholm, Sweden
| | - Mustafa Yalcinkaya
- Division of Molecular Medicine, Department of Medicine, Columbia Unicersity, New York, United States of America
| | - Huijuan Dou
- Division of Molecular Medicine, Department of Medicine, Columbia Unicersity, New York, United States of America
| | - Kaori Endo-Umeda
- Division of Biochemistry, Department of Biomedical Sciences, Nihon University School of Medicine, Tokyo, Japan
| | - Yang Tang
- Department of Hematology, The First Hospital of Jilin University, Changchun, China
| | - Xintong Hou
- Department of Hematology, The First Hospital of Jilin University, Changchun, China
| | - Tong Xiao
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, United States of America
| | - Trevor Filder
- Division of Molecular Medicine, Department of Medicine, Columbia Unicersity, New York, United States of America
| | - Sandra Abramowicz
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, United States of America
| | - Yong-Guang Yang
- Institute of Immunology, The First Hospital of Jilin University, Changchun, China
| | - Oliver Soehnlein
- Institute of Experimental Pathology, University of Münster, Munich, Germany
| | - Alan R Tall
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, United States of America
| | - Nan Wang
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, United States of America
| |
Collapse
|
28
|
Schaupp CM, Botta D, White CC, Scoville DK, Srinouanprachanh S, Bammler TK, MacDonald J, Kavanagh TJ. Persistence of improved glucose homeostasis in Gclm null mice with age and cadmium treatment. Redox Biol 2022; 49:102213. [PMID: 34953454 PMCID: PMC8715110 DOI: 10.1016/j.redox.2021.102213] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/07/2021] [Accepted: 12/13/2021] [Indexed: 12/23/2022] Open
Abstract
Antioxidant signaling/communication is among the most important cellular defense and survival pathways, and the importance of redox signaling and homeostasis in aging has been well-documented. Intracellular levels of glutathione (GSH), a very important endogenous antioxidant, both govern and are governed by the Nrf2 pathway through expression of genes involved in its biosynthesis, including the subunits of the rate-limiting enzyme (glutamate cysteine ligase, GCL) in GSH production, GCLC and GCLM. Mice homozygous null for the Gclm gene are severely deficient in GSH compared to wild-type controls, expressing approximately 10% of normal GSH levels. To compensate for GSH deficiency, Gclm null mice have upregulated redox-regulated genes, and, surprisingly, are less susceptible to certain types of oxidative damage. Furthermore, young Gclm null mice display an interesting lean phenotype, resistance to high fat diet-induced diabetes and obesity, improved insulin and glucose tolerance, and decreased expression of genes involved in lipogenesis. However, the persistence of this phenotype has not been investigated into old age, which is important in light of studies which suggest aging attenuates antioxidant signaling, particularly in response to exogenous stimuli. In this work, we addressed whether aging compromises the favorable phenotype of increased antioxidant activity and improved glucose homeostasis observed in younger Gclm null mice. We present data showing that under basal conditions and in response to cadmium exposure (2 mg/kg, dosed once via intraperitoneal injection), the phenotype previously described in young (<6 months) Gclm null mice persists into old age (24+ months). We also provide evidence that transcriptional activation of the Nrf2, AMPK, and PPARγ pathways underlie the favorable metabolic phenotype observed previously in young Gclm null mice.
Collapse
Affiliation(s)
- Christopher M Schaupp
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, 98195, USA
| | - Dianne Botta
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, 98195, USA
| | - Collin C White
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, 98195, USA
| | - David K Scoville
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, 98195, USA
| | - Sengkeo Srinouanprachanh
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, 98195, USA
| | - Theo K Bammler
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, 98195, USA
| | - James MacDonald
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, 98195, USA
| | - Terrance J Kavanagh
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
29
|
Ramendra R, Mancini M, Ayala JM, Tung LT, Isnard S, Lin J, Routy JP, Nijnik A, Langlais D. Glutathione Metabolism Is a Regulator of the Acute Inflammatory Response of Monocytes to (1→3)-β-D-Glucan. Front Immunol 2021; 12:694152. [PMID: 34858388 PMCID: PMC8631827 DOI: 10.3389/fimmu.2021.694152] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 10/26/2021] [Indexed: 12/13/2022] Open
Abstract
(1→3)-β-D-Glucan (BDG) represents a potent pathogen-associated molecular pattern (PAMP) in triggering the host response to fungal and some bacterial infections. Monocytes play a key role in recognizing BDG and governing the acute host response to infections. However, the mechanisms regulating monocyte's acute response to BDG are poorly understood. We sought to investigate the response of monocytes to BDG at the epigenetic, transcriptomic, and molecular levels. Response of human monocytes to 1, 4, and 24 hours of BDG exposure was investigated using RNA-seq, ATAC-seq, H3K27ac and H3K4me1 ChIP-seq. We show that pathways including glutathione metabolism, pentose phosphate pathway, and citric acid cycle were upregulated at the epigenetic and transcriptomic levels in response to BDG exposure. Strikingly, unlike bacterial lipopolysaccharides, BDG induced intracellular glutathione synthesis. BDG exposure also induced NADP synthesis, increased NADPH/NADP ratio, and increased expression of genes involved in the pentose phosphate pathway in a GSH-dependent manner. By inhibiting GSH synthesis with L-buthionine sulfoximine (BSO) before BDG exposure we show that the GSH pathway promotes cell survival and regulates monocyte's effector functions including NO production, phagocytosis, and cytokine production. In summary, our work demonstrates that BDG induces glutathione synthesis and metabolism in monocytes, which is a major promoter of the acute functional response of monocytes to infections.
Collapse
Affiliation(s)
- Rayoun Ramendra
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
- McGill University Research Centre on Complex Traits, Montreal, QC, Canada
- McGill University Genome Centre, Montreal, QC, Canada
| | - Mathieu Mancini
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
- McGill University Research Centre on Complex Traits, Montreal, QC, Canada
- McGill University Genome Centre, Montreal, QC, Canada
| | - Jose-Mauricio Ayala
- McGill University Research Centre on Complex Traits, Montreal, QC, Canada
- McGill University Genome Centre, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Lin Tze Tung
- McGill University Research Centre on Complex Traits, Montreal, QC, Canada
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - Stephane Isnard
- Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC, Canada
| | - John Lin
- Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC, Canada
| | - Jean-Pierre Routy
- Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC, Canada
| | - Anastasia Nijnik
- McGill University Research Centre on Complex Traits, Montreal, QC, Canada
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - David Langlais
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
- McGill University Research Centre on Complex Traits, Montreal, QC, Canada
- McGill University Genome Centre, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| |
Collapse
|
30
|
Redox Regulation and Oxidative Stress in Mammalian Oocytes and Embryos Developed In Vivo and In Vitro. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph182111374. [PMID: 34769890 PMCID: PMC8583213 DOI: 10.3390/ijerph182111374] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/25/2021] [Accepted: 10/25/2021] [Indexed: 12/18/2022]
Abstract
Oocytes and preimplantation embryos require careful regulation of the redox environment for optimal development both in vivo and in vitro. Reactive oxygen species (ROS) are generated throughout development as a result of cellular metabolism and enzyme reactions. ROS production can result in (i) oxidative eustress, where ROS are helpful signalling molecules with beneficial physiological functions and where the redox state of the cell is maintained within homeostatic range by a closely coupled system of antioxidants and antioxidant enzymes, or (ii) oxidative distress, where excess ROS are deleterious and impair normal cellular function. in vitro culture of embryos exacerbates ROS production due to a range of issues including culture-medium composition and laboratory culture conditions. This increase in ROS can be detrimental not only to assisted reproductive success rates but can also result in epigenetic and genetic changes in the embryo, resulting in transgenerational effects. This review examines the effects of oxidative stress in the oocyte and preimplantation embryo in both the in vivo and in vitro environment, identifies mechanisms responsible for oxidative stress in the oocyte/embryo in culture and approaches to reduce these problems, and briefly examines the potential impacts on future generations.
Collapse
|
31
|
Sohn SH, Sul HJ, Kim B, Kim BJ, Kim HS, Zang DY. TRK inhibitors block NFKB and induce NRF2 in TRK fusion-positive colon cancer. J Cancer 2021; 12:6356-6362. [PMID: 34659525 PMCID: PMC8489125 DOI: 10.7150/jca.60845] [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: 03/25/2021] [Accepted: 08/19/2021] [Indexed: 11/27/2022] Open
Abstract
Tropomyosin receptor kinase (TRK) fusion is one of the oncogenic driver causes of colon cancer, and tropomyosin 3-neurotrophic receptor tyrosine kinase 1 (TPM3-NTRK1) fusion has been detected in the KM12SM cell line. In the present study, we investigated anticancer mechanisms in the KM12SM cell line using three different form of dovitinib (dovitinib (free base), dovitinib lactate (mono acid), and dovitinib dilactic acid (diacid)) and four TRK inhibitors (LOXO-101, entrectinib, regorafenib, and crizotinib). Exposure of TRK inhibitors at concentrations of 10 nM resulted in the apoptosis of KM12SM cells, whereas regorafenib had no effect. Treatment with all inhibitors except regorafenib also significantly increased the expression levels of the genes nuclear factor-erythroid 2-related factor 2 (NRF2) and glutamyl cysteine ligase catalytic subunit (GCLC) in KM12SM. These drugs significantly reduced expression of the phosphorylated proteins NFκB and COX-2 in the KM12SM cell line, and significantly attenuated KM12SM cell migration, according to a Transwell migration assay. Together, these results suggest that TRK inhibitors block products of carcinogenesis by negatively regulating the NFκB signaling pathway and positively regulating the antioxidant NRF2 signaling pathway.
Collapse
Affiliation(s)
- Sung-Hwa Sohn
- Hallym Translational Research Institute, Hallym University Sacred Heart Hospital, Anyang, 14066, Republic of Korea
| | - Hee Jung Sul
- Hallym Translational Research Institute, Hallym University Sacred Heart Hospital, Anyang, 14066, Republic of Korea
| | - Bohyun Kim
- Hallym Translational Research Institute, Hallym University Sacred Heart Hospital, Anyang, 14066, Republic of Korea
| | - Bum Jun Kim
- Department of Internal Medicine, Hallym University Medical Center, Hallym University College of Medicine, Anyang-si, Gyeonggi-do 14068, Republic of Korea
| | - Hyeong Su Kim
- Department of Internal Medicine, Hallym University Medical Center, Hallym University College of Medicine, Anyang-si, Gyeonggi-do 14068, Republic of Korea
| | - Dae Young Zang
- Hallym Translational Research Institute, Hallym University Sacred Heart Hospital, Anyang, 14066, Republic of Korea.,Department of Internal Medicine, Hallym University Medical Center, Hallym University College of Medicine, Anyang-si, Gyeonggi-do 14068, Republic of Korea
| |
Collapse
|
32
|
Lim J, Ali S, Liao LS, Nguyen ES, Ortiz L, Reshel S, Luderer U. Antioxidant supplementation partially rescues accelerated ovarian follicle loss, but not oocyte quality, of glutathione-deficient mice†. Biol Reprod 2021; 102:1065-1079. [PMID: 31950131 DOI: 10.1093/biolre/ioaa009] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 12/16/2019] [Accepted: 01/13/2020] [Indexed: 12/31/2022] Open
Abstract
The tripeptide thiol antioxidant glutathione (GSH) has multiple physiological functions. Female mice lacking the modifier subunit of glutamate cysteine ligase (GCLM), the rate-limiting enzyme in GSH synthesis, have decreased GSH concentrations, ovarian oxidative stress, preimplantation embryonic mortality, and accelerated age-related decline in ovarian follicles. We hypothesized that supplementation with thiol antioxidants, N-acetyl cysteine (NAC), or α-lipoic acid (ALA) will rescue this phenotype. Gclm-/- and Gclm+/+ females received 0 or 80 mM NAC in drinking water from postnatal day (PND) 21-30; follicle growth was induced with equine chorionic gonadotropin (eCG) on PND 27, followed by an ovulatory dose of human CG and mating with a wild type male on PND 29 and zygote harvest 20 h after hCG. N-acetyl cysteine supplementation failed to rescue the low rate of second pronucleus formation in zygotes from Gclm-/- versus Gclm+/+ females. In the second study, Gclm-/- and Gclm+/+ females received diet containing 0, 150, or 600 mg/kg ALA beginning at weaning and were mated with wild type males from 8 to 20 weeks of age. α-Lipoic acid failed to rescue the decreased offspring production of Gclm-/- females. However, 150 mg/kg diet ALA partially rescued the accelerated decline in primordial follicles, as well as the increased recruitment of follicles into the growing pool and the increased percentages of follicles with γH2AX positive oocytes or granulosa cells of Gclm-/- females. We conclude that ovarian oxidative stress is the cause of accelerated primordial follicle decline, while GSH deficiency per se may be responsible for preimplantation embryonic mortality in Gclm-/- females.
Collapse
Affiliation(s)
- Jinhwan Lim
- Department of Medicine, University of California, Irvine, California, USA
| | - Samiha Ali
- Department of Medicine, University of California, Irvine, California, USA
| | - Lisa S Liao
- Department of Medicine, University of California, Irvine, California, USA
| | - Emily S Nguyen
- Department of Medicine, University of California, Irvine, California, USA
| | - Laura Ortiz
- Department of Medicine, University of California, Irvine, California, USA
| | - Samantha Reshel
- Department of Developmental and Cell Biology, University of California, Irvine, California, USA
| | - Ulrike Luderer
- Department of Medicine, University of California, Irvine, California, USA.,Department of Developmental and Cell Biology, University of California, Irvine, California, USA.,Program in Public Health, University of California, Irvine, California, USA
| |
Collapse
|
33
|
Hitchler MJ, Domann FE. The epigenetic and morphogenetic effects of molecular oxygen and its derived reactive species in development. Free Radic Biol Med 2021; 170:70-84. [PMID: 33450377 PMCID: PMC8217084 DOI: 10.1016/j.freeradbiomed.2021.01.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 01/04/2021] [Accepted: 01/05/2021] [Indexed: 12/11/2022]
Abstract
The development of multicellular organisms involves the unpacking of a complex genetic program. Extensive characterization of discrete developmental steps has revealed the genetic program is controlled by an epigenetic state. Shifting the epigenome is a group of epigenetic enzymes that modify DNA and proteins to regulate cell type specific gene expression. While the role of these modifications in development has been established, the input(s) responsible for electing changes in the epigenetic state remains unknown. Development is also associated with dynamic changes in cellular metabolism, redox, free radical production, and oxygen availability. It has previously been postulated that these changes are causal in development by affecting gene expression. This suggests that oxygen is a morphogenic compound that impacts the removal of epigenetic marks. Likewise, metabolism and reactive oxygen species influence redox signaling through iron and glutathione to limit the availability of key epigenetic cofactors such as α-ketoglutarate, ascorbate, NAD+ and S-adenosylmethionine. Given the close relationship between these cofactors and epigenetic marks it seems likely that the two are linked. Here we describe how changing these inputs might affect the epigenetic state during development to drive gene expression. Combined, these cofactors and reactive oxygen species constitute the epigenetic landscape guiding cells along differing developmental paths.
Collapse
Affiliation(s)
- Michael J Hitchler
- Department of Radiation Oncology, Kaiser Permanente Los Angeles Medical Center, 4950 Sunset Blvd, Los Angeles, CA, 90027, USA.
| | - Frederick E Domann
- Department of Radiation Oncology, Free Radical and Radiation Biology Program, University of Iowa, Iowa City, IA, 52242, USA.
| |
Collapse
|
34
|
Abstract
Glutathione (GSH) is the most abundant non-protein thiol, and plays crucial roles in the antioxidant defense system and the maintenance of redox homeostasis in neurons. GSH depletion in the brain is a common finding in patients with neurodegenerative diseases, such as Alzheimer’s disease and Parkinson’s disease, and can cause neurodegeneration prior to disease onset. Excitatory amino acid carrier 1 (EAAC1), a sodium-dependent glutamate/cysteine transporter that is selectively present in neurons, plays a central role in the regulation of neuronal GSH production. The expression of EAAC1 is posttranslationally controlled by the glutamate transporter-associated protein 3–18 (GTRAP3-18) or miR-96-5p in neurons. The regulatory mechanism of neuronal GSH production mediated by EAAC1 may be a new target in therapeutic strategies for these neurodegenerative diseases. This review describes the regulatory mechanism of neuronal GSH production and its potential therapeutic application in the treatment of neurodegenerative diseases.
Collapse
|
35
|
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: 16.7] [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
|
36
|
Lou XY, Hou TT, Liu SY, Xu HM, Lin F, Tang X, MacLeod SL, Cleves MA, Hobbs CA. Innovative approach to identify multigenomic and environmental interactions associated with birth defects in family-based hybrid designs. Genet Epidemiol 2021; 45:171-189. [PMID: 32996630 PMCID: PMC8495752 DOI: 10.1002/gepi.22363] [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: 03/27/2020] [Revised: 09/08/2020] [Accepted: 09/11/2020] [Indexed: 11/09/2022]
Abstract
Genes, including those with transgenerational effects, work in concert with behavioral, environmental, and social factors via complex biological networks to determine human health. Understanding complex relationships between causal factors underlying human health is an essential step towards deciphering biological mechanisms. We propose a new analytical framework to investigate the interactions between maternal and offspring genetic variants or their surrogate single nucleotide polymorphisms (SNPs) and environmental factors using family-based hybrid study design. The proposed approach can analyze diverse genetic and environmental factors and accommodate samples from a variety of family units, including case/control-parental triads, and case/control-parental dyads, while minimizing potential bias introduced by population admixture. Comprehensive simulations demonstrated that our innovative approach outperformed the log-linear approach, the best available method for case-control family data. The proposed approach had greater statistical power and was capable to unbiasedly estimate the maternal and child genetic effects and the effects of environmental factors, while controlling the Type I error rate against population stratification. Using our newly developed approach, we analyzed the associations between maternal and fetal SNPs and obstructive and conotruncal heart defects, with adjustment for demographic and lifestyle factors and dietary supplements. Fourteen and 11 fetal SNPs were associated with obstructive and conotruncal heart defects, respectively. Twenty-seven and 17 maternal SNPs were associated with obstructive and conotruncal heart defects, respectively. In addition, maternal body mass index was a significant risk factor for obstructive defects. The proposed approach is a powerful tool for interrogating the etiological mechanism underlying complex traits.
Collapse
Affiliation(s)
- Xiang-Yang Lou
- Department of Biostatistics, College of Public Health and Health Professions and College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Ting-Ting Hou
- Department of Biostatistics, College of Public Health and Health Professions and College of Medicine, University of Florida, Gainesville, Florida, USA
- Institute of Bioinformatics and Institute of Crop Science, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
| | - Shou-Ye Liu
- Institute of Bioinformatics and Institute of Crop Science, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
| | - Hai-Ming Xu
- Institute of Bioinformatics and Institute of Crop Science, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
| | - Feng Lin
- Institute of Bioinformatics and Institute of Crop Science, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
| | - Xinyu Tang
- The US Food and Drug Administration, Silver Spring, Maryland, USA
| | | | - Mario A. Cleves
- Department of Pediatrics, Morsani College of Medicine, Health Informatics Institute, University of South Florida, Tampa, Florida, USA
| | - Charlotte A. Hobbs
- Rady Children’s Institute for Genomic Medicine, San Diego, California, USA
| |
Collapse
|
37
|
Bessières B, Cruz E, Alberini CM. Metabolomic profiling reveals a differential role for hippocampal glutathione reductase in infantile memory formation. eLife 2021; 10:68590. [PMID: 34825649 PMCID: PMC8626085 DOI: 10.7554/elife.68590] [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: 03/19/2021] [Accepted: 11/09/2021] [Indexed: 01/12/2023] Open
Abstract
The metabolic mechanisms underlying the formation of early-life episodic memories remain poorly characterized. Here, we assessed the metabolomic profile of the rat hippocampus at different developmental ages both at baseline and following episodic learning. We report that the hippocampal metabolome significantly changes over developmental ages and that learning regulates differential arrays of metabolites according to age. The infant hippocampus had the largest number of significant changes following learning, with downregulation of 54 metabolites. Of those, a large proportion was associated with the glutathione-mediated cellular defenses against oxidative stress. Further biochemical, molecular, and behavioral assessments revealed that infantile learning evokes a rapid and persistent increase in the activity of neuronal glutathione reductase, the enzyme that regenerates reduced glutathione from its oxidized form. Inhibition of glutathione reductase selectively impaired long-term memory formation in infant but not in juvenile and adult rats, confirming its age-specific role. Thus, metabolomic profiling revealed that the hippocampal glutathione-mediated antioxidant pathway is differentially required for the formation of infantile memory.
Collapse
Affiliation(s)
| | - Emmanuel Cruz
- Center for Neural Science, New York UniversityNew YorkUnited States
| | | |
Collapse
|
38
|
Mazloomi S, Khodadadi I, Alizadeh N, Shafiee G. Association of glutamate cystein ligase (GCL) activity Peroxiredoxin 4 (prxR4) and apelin levels in women with preeclampsia. Pregnancy Hypertens 2020; 23:163-168. [PMID: 33421846 DOI: 10.1016/j.preghy.2020.12.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 12/08/2020] [Accepted: 12/18/2020] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Preeclampsia is a common disease of pregnancy that is characterized by symptoms such as high blood pressure and proteinuria. Peroxiredoxin 4 (Prx4), is a protein with antioxidant properties which is produced in placenta and protects it from antioxidant stress and recurrent miscarriage. For regeneration of Peroxiredoxin 4 need to glutathione (GSH) and Glutamate-cysteine ligase (GCL) enzyme controls the pathway of glutathione regeneration. Apelin is a paired internal ligand with a G protein coupled receptor and is associated with angiotensin receptor (AT1) as a blood pressure regulator. This study was designed to evaluate GCL enzyme activity and Peroxiredoxin 4, glutathione and apelin levels in serum of women with preeclampsia. MATERIAL AND METHODS Thirty pregnant women with preeclampsia and 30 healthy pregnant women were enrolled in this study. All participants were diagnosed by clinical examination and confirmation by Obstetrician-Gynecologist. The GCL enzyme activity and concentration of Prx4 and apelin in serum samples were measured using ready-to-use non-competitive ELISA methods while glutathione level was determined using Ellman's reagent. RESULTS The GCL enzyme activity and Prx4 level were significantly lower in preeclampsia compared with control group (p < 0.05). In addition, marked reductions were observed in the concentrations of glutathione and apelin in preeclampsia compared to the healthy pregnant women (p < 0.05). CONCLUSION This study identified the role of the GCL and Prx4 system in preeclampsia disorder and may be one of the ways to prevent and reduce the risks of preeclampsia in high-risk women using diet control and stress reduction.
Collapse
Affiliation(s)
- Sahar Mazloomi
- Ph.D Candidate, Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Iraj Khodadadi
- Professor of Biochemistry, Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Narges Alizadeh
- Obstetricians and Gynecologist, Bijar Imam Hossain Hospital, Kurdistan University of Medical Sciences, Kurdistan, Iran
| | - Gholamreza Shafiee
- Assistant Professor of Clinical Biochemistry, Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Nutrition Health Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
39
|
Park EJ, Dusabimana T, Je J, Jeong K, Yun SP, Kim HJ, Kim H, Park SW. Honokiol Protects the Kidney from Renal Ischemia and Reperfusion Injury by Upregulating the Glutathione Biosynthetic Enzymes. Biomedicines 2020; 8:biomedicines8090352. [PMID: 32942603 PMCID: PMC7555803 DOI: 10.3390/biomedicines8090352] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 08/29/2020] [Accepted: 09/13/2020] [Indexed: 12/31/2022] Open
Abstract
Glutathione (GSH) is an endogenous antioxidant found in plants, animals, fungi, and some microorganisms that protects cells by neutralizing hydrogen peroxide. Honokiol, an active ingredient of Magnolia officinalis, is known for antioxidant, anti-inflammatory, and anti-bacterial properties. We investigated the protective mechanism of honokiol through regulating cellular GSH in renal proximal tubules against acute kidney injury (AKI). First, we measured cellular GSH levels and correlated them with the expression of GSH biosynthetic enzymes after honokiol treatment in human kidney-2 (HK-2) cells. Second, we used pharmacological inhibitors or siRNA-mediated gene silencing approach to determine the signaling pathway induced by honokiol. Third, the protective effect of honokiol via de novo GSH biosynthesis was investigated in renal ischemia-reperfusion (IR) mice. Honokiol significantly increased cellular GSH levels by upregulating the subunits of glutamate-cysteine ligase (Gcl)—Gclc and Gclm. These increases were mediated by activation of nuclear factor erythroid 2-related factor 2, via PI3K/Akt and protein kinase C signaling. Consistently, honokiol treatment reduced the plasma creatinine, tubular cell death, neutrophil infiltration and lipid peroxidation in IR mice and the effect was correlated with upregulation of Gclc and Gclm. Conclusively, honokiol may benefit to patients with AKI by increasing antioxidant GSH via transcriptional activation of the biosynthetic enzymes.
Collapse
Affiliation(s)
- Eun Jung Park
- Department of Pharmacology, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 52727, Korea; (E.J.P.); (T.D.); (J.J.); (K.J.); (S.P.Y.); (H.J.K.)
| | - Theodomir Dusabimana
- Department of Pharmacology, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 52727, Korea; (E.J.P.); (T.D.); (J.J.); (K.J.); (S.P.Y.); (H.J.K.)
- Department of Convergence Medical Sciences, Institute of Health Sciences, Gyeongsang National University Graduate School, Jinju 52727, Korea
| | - Jihyun Je
- Department of Pharmacology, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 52727, Korea; (E.J.P.); (T.D.); (J.J.); (K.J.); (S.P.Y.); (H.J.K.)
| | - Kyuho Jeong
- Department of Pharmacology, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 52727, Korea; (E.J.P.); (T.D.); (J.J.); (K.J.); (S.P.Y.); (H.J.K.)
| | - Seung Pil Yun
- Department of Pharmacology, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 52727, Korea; (E.J.P.); (T.D.); (J.J.); (K.J.); (S.P.Y.); (H.J.K.)
- Department of Convergence Medical Sciences, Institute of Health Sciences, Gyeongsang National University Graduate School, Jinju 52727, Korea
| | - Hye Jung Kim
- Department of Pharmacology, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 52727, Korea; (E.J.P.); (T.D.); (J.J.); (K.J.); (S.P.Y.); (H.J.K.)
- Department of Convergence Medical Sciences, Institute of Health Sciences, Gyeongsang National University Graduate School, Jinju 52727, Korea
| | - Hwajin Kim
- Department of Pharmacology, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 52727, Korea; (E.J.P.); (T.D.); (J.J.); (K.J.); (S.P.Y.); (H.J.K.)
- Correspondence: (H.K.); (S.W.P.); Tel.: +82-55-772-8070 (H.K.); +82-55-772-8073 (S.W.P.)
| | - Sang Won Park
- Department of Pharmacology, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 52727, Korea; (E.J.P.); (T.D.); (J.J.); (K.J.); (S.P.Y.); (H.J.K.)
- Department of Convergence Medical Sciences, Institute of Health Sciences, Gyeongsang National University Graduate School, Jinju 52727, Korea
- Correspondence: (H.K.); (S.W.P.); Tel.: +82-55-772-8070 (H.K.); +82-55-772-8073 (S.W.P.)
| |
Collapse
|
40
|
Alterations in the Antioxidant Enzyme Activities in the Neurodevelopmental Rat Model of Schizophrenia Induced by Glutathione Deficiency during Early Postnatal Life. Antioxidants (Basel) 2020; 9:antiox9060538. [PMID: 32575563 PMCID: PMC7346228 DOI: 10.3390/antiox9060538] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/10/2020] [Accepted: 06/17/2020] [Indexed: 01/24/2023] Open
Abstract
The aim of the present study was to assess the effects of l-buthionine-(S,R)-sulfoximine (BSO), a glutathione (GSH) synthesis inhibitor, and GBR 12909, a dopamine reuptake inhibitor, administered alone or in combination to Sprague-Dawley rats during early postnatal development (p5-p16), on the levels of reactive oxygen species (ROS), lipid peroxidation (LP) and the activities of antioxidant enzymes: superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx) and glutathione disulfide reductase (GR) in peripheral tissues (liver, kidney) and selected brain structures (prefrontal cortex, PFC; hippocampus, HIP; and striatum, STR) of 16-day-old rats. The studied parameters were analyzed with reference to the content of GSH and sulfur amino acids, methionine (Met) and cysteine (Cys) described in our previous study. This analysis showed that treatment with a BSO + GBR 12909 combination caused significant decreases in the lipid peroxidation levels in the PFC and HIP, in spite of there being no changes in ROS. The reduction of lipid peroxidation indicates a weakening of the oxidative power of the cells, and a shift in balance in favor of reducing processes. Such changes in cellular redox signaling in the PFC and HIP during early postnatal development may result in functional changes in adulthood.
Collapse
|
41
|
Daher B, Vučetić M, Pouysségur J. Cysteine Depletion, a Key Action to Challenge Cancer Cells to Ferroptotic Cell Death. Front Oncol 2020; 10:723. [PMID: 32457843 PMCID: PMC7221143 DOI: 10.3389/fonc.2020.00723] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 04/16/2020] [Indexed: 12/11/2022] Open
Abstract
Cancer cells are characterized as highly proliferative at the expense of enhancement of metabolic rate. Consequently, cancer cells rely on antioxidant defenses to overcome the associated increased production of reactive oxygen species (ROS). The reliance of tumor metabolism on amino acids, especially amino acid transport systems, has been extensively studied over the past decade. Although cysteine is the least abundant amino acid in the cell, evidences described it as one of the most important amino acid for cell survival and growth. Regarding its multi-functionality as a nutrient, protein folding, and major component for redox balance due to its involvement in glutathione synthesis, disruption of cysteine homeostasis appears to be promising strategy for induction of cancer cell death. Ten years ago, ferroptosis, a new form of non-apoptotic cell death, has been described as a result of cysteine insufficiency leading to a collapse of intracellular glutathione level. In the present review, we summarized the metabolic networks involving the amino acid cysteine in cancer and ferroptosis and we focused on describing the recently discovered glutathione-independent pathway, a potential player in cancer ferroptosis resistance. Then, we discuss the implication of cysteine as key player in ferroptosis as a precursor for glutathione first, but also as metabolic precursor in glutathione-independent ferroptosis axis.
Collapse
Affiliation(s)
- Boutaina Daher
- Medical Biology Department, Centre Scientifique de Monaco (CSM), Monaco, Monaco
| | - Milica Vučetić
- Medical Biology Department, Centre Scientifique de Monaco (CSM), Monaco, Monaco
| | - Jacques Pouysségur
- Medical Biology Department, Centre Scientifique de Monaco (CSM), Monaco, Monaco
- Institute for Research on Cancer and Aging (IRCAN), CNRS, INSERM, Centre A. Lacassagne, Université Côte d'Azur, Nice, France
| |
Collapse
|
42
|
Intertwined ROS and Metabolic Signaling at the Neuron-Astrocyte Interface. Neurochem Res 2020; 46:23-33. [PMID: 31989468 DOI: 10.1007/s11064-020-02965-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 01/03/2020] [Accepted: 01/16/2020] [Indexed: 12/23/2022]
Abstract
Metabolism and redox signalling share critical nodes in the nervous system. In the last years, a series of major findings have challenged the current vision on how neural reactive oxygen species (ROS) are produced and handled in the nervous system. Once regarded as deleterious by-products, ROS are now shown to be essential for a metabolic and redox crosstalk. In turn, this coupling defines neural viability and function to control behaviour or leading to neurodegeneration when compromised. Findings like a different assembly of mitochondrial respiratory supercomplexes in neurons and astrocytes stands behind a divergent production of ROS in either cell type, more prominent in astrocytes. ROS levels are however tightly controlled by an antioxidant machinery in astrocytes, assumed as more efficient than that of neurons, to regulate redox signalling. By exerting this control in ROS abundance, metabolic functions are finely tuned in both neural cells. Further, a higher engagement of mitochondrial respiration and oxidative function in neurons, underpinned by redox equivalents supplied from the pentose phosphate pathway and from glia, differs from the otherwise strong glycolytic capacity of astrocytes. Here, we recapitulate major findings on how ROS and metabolism differ between neural cells but merge to define reciprocal signalling pathways, ultimately defining neural function and fate.
Collapse
|
43
|
Bebber CM, Müller F, Prieto Clemente L, Weber J, von Karstedt S. Ferroptosis in Cancer Cell Biology. Cancers (Basel) 2020; 12:E164. [PMID: 31936571 PMCID: PMC7016816 DOI: 10.3390/cancers12010164] [Citation(s) in RCA: 206] [Impact Index Per Article: 51.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 01/07/2020] [Accepted: 01/07/2020] [Indexed: 12/12/2022] Open
Abstract
A major hallmark of cancer is successful evasion of regulated forms of cell death. Ferroptosis is a recently discovered type of regulated necrosis which, unlike apoptosis or necroptosis, is independent of caspase activity and receptor-interacting protein 1 (RIPK1) kinase activity. Instead, ferroptotic cells die following iron-dependent lipid peroxidation, a process which is antagonised by glutathione peroxidase 4 (GPX4) and ferroptosis suppressor protein 1 (FSP1). Importantly, tumour cells escaping other forms of cell death have been suggested to maintain or acquire sensitivity to ferroptosis. Therefore, therapeutic exploitation of ferroptosis in cancer has received increasing attention. Here, we systematically review current literature on ferroptosis signalling, cross-signalling to cellular metabolism in cancer and a potential role for ferroptosis in tumour suppression and tumour immunology. By summarising current findings on cell biology relevant to ferroptosis in cancer, we aim to point out new conceptual avenues for utilising ferroptosis in systemic treatment approaches for cancer.
Collapse
Affiliation(s)
- Christina M. Bebber
- Department of Translational Genomics, Medical Faculty, University of Cologne, Weyertal 155b, 50931 Cologne, Germany; (C.M.B.); (F.M.); (L.P.C.); (J.W.)
- Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), Medical Faculty, University of Cologne, Joseph-Stelzmann-Straße 26, 50931 Cologne, Germany
- Department I of Internal Medicine, University Hospital of Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| | - Fabienne Müller
- Department of Translational Genomics, Medical Faculty, University of Cologne, Weyertal 155b, 50931 Cologne, Germany; (C.M.B.); (F.M.); (L.P.C.); (J.W.)
- Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), Medical Faculty, University of Cologne, Joseph-Stelzmann-Straße 26, 50931 Cologne, Germany
| | - Laura Prieto Clemente
- Department of Translational Genomics, Medical Faculty, University of Cologne, Weyertal 155b, 50931 Cologne, Germany; (C.M.B.); (F.M.); (L.P.C.); (J.W.)
- Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), Medical Faculty, University of Cologne, Joseph-Stelzmann-Straße 26, 50931 Cologne, Germany
| | - Josephine Weber
- Department of Translational Genomics, Medical Faculty, University of Cologne, Weyertal 155b, 50931 Cologne, Germany; (C.M.B.); (F.M.); (L.P.C.); (J.W.)
- Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), Medical Faculty, University of Cologne, Joseph-Stelzmann-Straße 26, 50931 Cologne, Germany
| | - Silvia von Karstedt
- Department of Translational Genomics, Medical Faculty, University of Cologne, Weyertal 155b, 50931 Cologne, Germany; (C.M.B.); (F.M.); (L.P.C.); (J.W.)
- Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), Medical Faculty, University of Cologne, Joseph-Stelzmann-Straße 26, 50931 Cologne, Germany
| |
Collapse
|
44
|
Effects of total parenteral nutrition on drug metabolism gene expression in mice. Acta Pharm Sin B 2020; 10:153-158. [PMID: 31993312 PMCID: PMC6976970 DOI: 10.1016/j.apsb.2019.10.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 09/25/2019] [Accepted: 10/08/2019] [Indexed: 02/01/2023] Open
Abstract
Parenteral nutrition-associated liver disease (PNALD) is a liver dysfunction caused by various risk factors presented in patients receiving total parenteral nutrition (TPN). Omega-6 rich Intralipid® and omega-3 rich Omegaven® are two intravenous lipid emulsions used in TPN. TPN could affect the hepatic expression of genes in anti-oxidative stress, but it's unknown whether TPN affects genes in drug metabolism. In this study, either Intralipid®- or Omegaven®-based TPN was administered to mice and the expression of a cohort of genes involved in anti-oxidative stress or drug metabolism was analyzed, glutathione (GSH) levels were measured, and protein levels for two key drug metabolism genes were determined. Overall, the expression of most genes was downregulated by Intralipid®-based TPN (Gstp1, Gstm1, 3, 6, Nqo1, Ho-1, Mt-1, Gclc, Gclm, Cyp2d9, 2f2, 2b10, and 3a11). Omegaven® showed similar results as Intralipid® except for preserving the expression of Gstm1 and Cyp3a11, and increasing Ho-1. Total GSH levels were decreased by Intralipid®, but increased by Omegaven®. CYP3A11 protein levels were increased by Omegaven®. In conclusion, TPN reduced the expression of many genes involved in anti-oxidative stress and drug metabolism in mice. However, Omegaven® preserved expression of Cyp3a11, suggesting another beneficial effect of Omegaven® in protecting liver functions.
Collapse
Key Words
- CYP450, cytochrome p450
- Drug metabolism
- FAs, fatty acids
- GADPH, glyceraldehyde 3-phosphate dehydrogenase
- GSH, glutathione
- GSSG, GSH/glutathione disulfide
- Gclc: glutamate-cysteine ligase catalytic subunit, Gclm: glutamate-cysteine ligase modifier subunit
- Glutathione
- Gpx3, glutathione peroxidase 3
- Gstm1, glutathione S-transferase, mu 1
- Gstm3, glutathione S-transferase, mu 3
- Gstm6, glutathione S-transferase, mu 6
- Gstp1, glutathione S-transferase, pi 1
- Ho-1, heme oxygenase 1
- Liver
- Mt-1, metallothionein 1
- NQO1, NAD(P)H:quinone acceptor oxidoreductase 1
- PNALD, parenteral nutrition-associated liver disease
- Parenteral nutrition-associated liver disease
- ROS, reactive oxygen species
- TPN, total parenteral nutrition
- Total parenteral nutrition
Collapse
|
45
|
Liu P, Kerins MJ, Tian W, Neupane D, Zhang DD, Ooi A. Differential and overlapping targets of the transcriptional regulators NRF1, NRF2, and NRF3 in human cells. J Biol Chem 2019; 294:18131-18149. [PMID: 31628195 PMCID: PMC6885608 DOI: 10.1074/jbc.ra119.009591] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 10/07/2019] [Indexed: 12/12/2022] Open
Abstract
The nuclear factor (erythroid 2)-like (NRF) transcription factors are a subset of cap'n'collar transcriptional regulators. They consist of three members, NRF1, NRF2, and NRF3, that regulate the expression of genes containing antioxidant-response elements (AREs) in their promoter regions. Although all NRF members regulate ARE-containing genes, each is associated with distinct roles. A comprehensive study of differential and overlapping DNA-binding and transcriptional activities of the NRFs has not yet been conducted. Here, we performed chromatin immunoprecipitation (ChIP)-exo sequencing, an approach that combines ChIP with exonuclease treatment to pinpoint regulatory elements in DNA with high precision, in conjunction with RNA-sequencing to define the transcriptional targets of each NRF member. Our approach, done in three U2OS cell lines, identified 31 genes that were regulated by all three NRF members, 27 that were regulated similarly by all three, and four genes that were differentially regulated by at least one NRF member. We also found genes that were up- or down-regulated by only one NRF member, with 84, 84, and 22 genes that were regulated by NRF1, NRF2, and NRF3, respectively. Analysis of the ARE motifs identified in ChIP peaks revealed that NRF2 prefers binding to AREs flanked by GC-rich regions and that NRF1 prefers AT-rich flanking regions. Thus, sequence preference, likely in combination with upstream signaling events, determines NRF member activation under specific cellular contexts. Our analysis provides a comprehensive description of differential and overlapping gene regulation by the transcriptional regulators NRF1, NRF2, and NRF3.
Collapse
Affiliation(s)
- Pengfei Liu
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona 85721
| | - Michael J. Kerins
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona 85721
| | - Wang Tian
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona 85721
| | - Durga Neupane
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona 85721
| | - Donna D. Zhang
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona 85721
- University of Arizona Cancer Center, University of Arizona, Tucson, Arizona 85721
| | - Aikseng Ooi
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona 85721
- University of Arizona Cancer Center, University of Arizona, Tucson, Arizona 85721
| |
Collapse
|
46
|
Górny M, Wnuk A, Kamińska A, Kamińska K, Chwatko G, Bilska-Wilkosz A, Iciek M, Kajta M, Rogóż Z, Lorenc-Koci E. Glutathione Deficiency and Alterations in the Sulfur Amino Acid Homeostasis during Early Postnatal Development as Potential Triggering Factors for Schizophrenia-Like Behavior in Adult Rats. Molecules 2019; 24:molecules24234253. [PMID: 31766654 PMCID: PMC6930621 DOI: 10.3390/molecules24234253] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 11/15/2019] [Accepted: 11/18/2019] [Indexed: 01/09/2023] Open
Abstract
Impaired glutathione (GSH) synthesis and dopaminergic transmission are important factors in the pathophysiology of schizophrenia. Our research aimed to assess the effects of l-buthionine-(S,R)-sulfoximine (BSO), a GSH synthesis inhibitor, and GBR 12909, a dopamine reuptake inhibitor, administered alone or in combination, to Sprague–Dawley rats during early postnatal development (p5–p16), on the levels of GSH, sulfur amino acids, global DNA methylation, and schizophrenia-like behavior. GSH, methionine (Met), homocysteine (Hcy), and cysteine (Cys) contents were determined in the liver, kidney, and in the prefrontal cortex (PFC) and hippocampus (HIP) of 16-day-old rats. DNA methylation in the PFC and HIP and schizophrenia-like behavior were assessed in adulthood (p90–p93). BSO caused the tissue-dependent decreases in GSH content and alterations in Met, Hcy, and Cys levels in the peripheral tissues and in the PFC and HIP. The changes in these parameters were accompanied by alterations in the global DNA methylation in the studied brain structures. Parallel to changes in the global DNA methylation, deficits in the social behaviors and cognitive functions were observed in adulthood. Only BSO + GBR 12909-treated rats exhibited behavioral alterations resembling positive symptoms in schizophrenia patients. Our results suggest the usefulness of this neurodevelopmental model for research on the pathomechanism of schizophrenia.
Collapse
Affiliation(s)
- Magdalena Górny
- The Chair of Medical Biochemistry, Jagiellonian University Medical College, 7 Kopernika Street, 31–034 Kraków, Poland; (M.G.); (A.B.-W.); (M.I.)
| | - Agnieszka Wnuk
- Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, 31–343 Kraków, Poland; (A.W.); (K.K.); (M.K.); (Z.R.)
| | - Adrianna Kamińska
- Department of Environmental Chemistry, University of Łódź, 163 Pomorska Street, 90-236 Łódź, Poland; (A.K.); (G.C.)
| | - Kinga Kamińska
- Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, 31–343 Kraków, Poland; (A.W.); (K.K.); (M.K.); (Z.R.)
| | - Grażyna Chwatko
- Department of Environmental Chemistry, University of Łódź, 163 Pomorska Street, 90-236 Łódź, Poland; (A.K.); (G.C.)
| | - Anna Bilska-Wilkosz
- The Chair of Medical Biochemistry, Jagiellonian University Medical College, 7 Kopernika Street, 31–034 Kraków, Poland; (M.G.); (A.B.-W.); (M.I.)
| | - Małgorzata Iciek
- The Chair of Medical Biochemistry, Jagiellonian University Medical College, 7 Kopernika Street, 31–034 Kraków, Poland; (M.G.); (A.B.-W.); (M.I.)
| | - Małgorzata Kajta
- Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, 31–343 Kraków, Poland; (A.W.); (K.K.); (M.K.); (Z.R.)
| | - Zofia Rogóż
- Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, 31–343 Kraków, Poland; (A.W.); (K.K.); (M.K.); (Z.R.)
| | - Elżbieta Lorenc-Koci
- Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, 31–343 Kraków, Poland; (A.W.); (K.K.); (M.K.); (Z.R.)
- Correspondence: ; Tel.: +48-126-623-272
| |
Collapse
|
47
|
Grin MA, Tikhonov SI, Petrova AS, Pogorilyy VA, Noev АN, Tatarskiy VV, Shpakovsky DB, Milaeva ER, Kalinina EV, Chernov NN, Shtil АА, Mironov AF, Kaprin AD, Filonenko EV. New Derivatives of Bacteriopurpurin with Thiolated Au (I) Complexes: Dual Darkand Light Activated Antitumor Potency. Anticancer Agents Med Chem 2019; 20:49-58. [PMID: 31368879 DOI: 10.2174/1871520619666190801102617] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 02/15/2019] [Accepted: 05/20/2019] [Indexed: 01/10/2023]
Abstract
BACKGROUND Conventional antitumor Photosensitizers (PS) are normally low toxic in the dark whereas light activation triggers massive cell death (photodynamic therapy, PDT). OBJECTIVE To expand the therapeutic potential of PS to dual potency cytocidal agents, taking advantage of the use of bacteriopurpurin for a deeper tissue penetration of light, and suitability of the tetrapyrrolic macrocycle for chemical modifications at its periphery. METHODS Conjugation of a pro-oxidant thiolate Au (I) moiety to the bacteriopurpurin core and evaluation of cytotoxicity in cell culture and in vivo. RESULTS New water-soluble derivatives showed micromolar cytotoxicity for cultured human tumor cell lines in the dark, including the subline with an altered drug response due to p53 inactivation. Cellular PDT with the selected conjugate, thiolate Au (I)-dipropoxybacteriopurpurinimide (compound 6) with two triphenylphosphine Au fragments, triggered rapid (within minutes) cell death. Damage to the plasma membrane (necrosis) was a hallmark of cell death by compound 6 both in the dark and upon light activation. Furthermore, one single i.v. injection of compound 6 caused retardation of transplanted syngeneic tumors at the tolerable dose. Illumination of tumors that accumulated compound 6 significantly synergized with the effect of 6 in the dark. CONCLUSION Complexes of virtually non-toxic, photoactivatable bacteriopurpurin with the gold-containing organic moiety are considered the dual potency antitumor agents, tentatively applicable for intractable tumors.
Collapse
Affiliation(s)
- Mikhail A Grin
- MIREA - Russian Technological University, Moscow, Russian Federation
| | - Sergei I Tikhonov
- MIREA - Russian Technological University, Moscow, Russian Federation
| | | | | | - Аlexey N Noev
- MIREA - Russian Technological University, Moscow, Russian Federation
| | - Victor V Tatarskiy
- Blokhin National Medical Research Center of Oncology, Moscow, Russian Federation
| | - Dmitry B Shpakovsky
- Department of Chemistry, M. V. Lomonosov Moscow State University, Moscow, Russian Federation
| | - Elena R Milaeva
- Department of Chemistry, M. V. Lomonosov Moscow State University, Moscow, Russian Federation
| | | | | | - Аlexander А Shtil
- Blokhin National Medical Research Center of Oncology, Moscow, Russian Federation.,Department of Chemistry, M. V. Lomonosov Moscow State University, Moscow, Russian Federation
| | - Andrey F Mironov
- MIREA - Russian Technological University, Moscow, Russian Federation
| | - Andrey D Kaprin
- National Medical Research Center of Radiology of Ministry of Health of Russia, Moscow, Russian Federation
| | - Elena V Filonenko
- National Medical Research Center of Radiology of Ministry of Health of Russia, Moscow, Russian Federation
| |
Collapse
|
48
|
Heo G, Ko KS. Long-Term Feeding of Soy Protein Attenuates Choline Deficient-Induced Adverse Effects in Wild Type Mice and Prohibitin 1 Deficient Mice Response More Sensitively. Prev Nutr Food Sci 2019; 24:32-40. [PMID: 31008094 PMCID: PMC6456240 DOI: 10.3746/pnf.2019.24.1.32] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 01/31/2019] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease, however the exact cause of NAFLD remains unknown. Methionine, an essential amino acid, is the first limiting amino acid of soy protein, and its deficiency is suggested to cause hepatocyte damage and NAFLD. The objective of this study is to examine the changes in NAFLD susceptibility with soy protein consumption and deterioration due to prohibitin 1 (PHB1) deficiency, an important protein in hepatic mitochondrial function. In this study, liver-specific phb1 +/- mice and wild-type mice were fed a normal diet, choline-deficient diet (CDD), or soy protein diet without choline (SPD) for 16 weeks. Using hematoxylin and eosin staining, we showed that SPD attenuates symptoms of hepatocyte damage and lipid accumulation induced by CDD in mouse liver. The liver damage in mice fed the SPD was alleviated by decreasing lipogenic markers and by increasing anti-inflammatory markers. Furthermore, mRNA expression of genes involved in hepatic methionine metabolism was significantly lower in liver-specific phb1 +/- mice fed with a SPD compared with wild-type mice fed with a SPD. These data suggest a CDD can cause non-alcohol related liver damage, which can be attenuated by a SPD in wild-type mice. These phenomena were not observed in liver-specific phb1 +/- mice. It may therefore be concluded that SPD attenuates CDD-induced liver damage in wild-type mice, and that PHB1 deficiency blocks the beneficial effects of SPD against CDD-induced liver damage.
Collapse
Affiliation(s)
- Gieun Heo
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Korea
| | - Kwang Suk Ko
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
49
|
Scirè A, Cianfruglia L, Minnelli C, Bartolini D, Torquato P, Principato G, Galli F, Armeni T. Glutathione compartmentalization and its role in glutathionylation and other regulatory processes of cellular pathways. Biofactors 2019; 45:152-168. [PMID: 30561781 DOI: 10.1002/biof.1476] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/23/2018] [Accepted: 10/24/2018] [Indexed: 12/20/2022]
Abstract
Glutathione is considered the major non-protein low molecular weight modulator of redox processes and the most important thiol reducing agent of the cell. The biosynthesis of glutathione occurs in the cytosol from its constituent amino acids, but this tripeptide is also present in the most important cellular districts, such as mitochondria, nucleus, and endoplasmic reticulum, thus playing a central role in several metabolic pathways and cytoprotection mechanisms. Indeed, glutathione is involved in the modulation of various cellular processes and, not by chance, it is a ubiquitous determinant for redox signaling, xenobiotic detoxification, and regulation of cell cycle and death programs. The balance between its concentration and redox state is due to a complex series of interactions between biosynthesis, utilization, degradation, and transport. All these factors are of great importance to understand the significance of cellular redox balance and its relationship with physiological responses and pathological conditions. The purpose of this review is to give an overview on glutathione cellular compartmentalization. Information on its subcellular distribution provides a deeper understanding of glutathione-dependent processes and reflects the importance of compartmentalization in the regulation of specific cellular pathways. © 2018 BioFactors, 45(2):152-168, 2019.
Collapse
Affiliation(s)
- Andrea Scirè
- Department of Life and Environmental Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Laura Cianfruglia
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Università Politecnica delle Marche, Ancona, Italy
| | - Cristina Minnelli
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Università Politecnica delle Marche, Ancona, Italy
| | - Desirée Bartolini
- Clinical Biochemistry and Human Nutrition Labs, Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Pierangelo Torquato
- Clinical Biochemistry and Human Nutrition Labs, Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Giovanni Principato
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Università Politecnica delle Marche, Ancona, Italy
| | - Francesco Galli
- Clinical Biochemistry and Human Nutrition Labs, Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Tatiana Armeni
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
50
|
Abstract
SIGNIFICANCE Angiogenesis is the formation of new vessels that sprout from existing vessels. This process is highly complex and requires a coordinated shift of the endothelial phenotype from a quiescent cell in the vessel wall into a migrating or proliferating cell. Such change in the life of the endothelial cell is induced by a variety of factors such as hypoxia, metabolic changes, or cytokines. Recent Advances: Within the last years, it became clear that the cellular redox state and oxidation of signaling molecules or phosphatases are critical modulators in angiogenesis. CRITICAL ISSUES According to the wide variety of stimuli that induce angiogenesis, a complex signaling network is needed to support a coordinated response of the endothelial cell. Reactive oxygen species (ROS) now are second messengers that either directly oxidize a target molecule or initiate a cascade of redox sensitive steps that transmit the signal. Further Directions: For the understanding of redox signaling, it is essential to recognize and accept that ROS do not represent master regulators of angiogenetic processes. They rather modulate existing signal cascades. This review summarizes some current findings on redox signaling in angiogenesis.
Collapse
Affiliation(s)
- Katrin Schröder
- 1 Institute for Cardiovascular Physiology, Goethe-University, Frankfurt, Germany.,2 German Center for Cardiovascular Research (DZHK), Rhine-Main, Frankfurt, Germany
| |
Collapse
|