1
|
Lei L, Yuan J, Dai Z, Xiang S, Tu Q, Cui X, Zhai S, Chen X, He Z, Fang B, Xu Z, Yu H, Tang L, Zhang C. Targeting the Labile Iron Pool with Engineered DFO Nanosheets to Inhibit Ferroptosis for Parkinson's Disease Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2409329. [PMID: 39221531 DOI: 10.1002/adma.202409329] [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: 06/29/2024] [Revised: 07/31/2024] [Indexed: 09/04/2024]
Abstract
Ferroptosis in neurons is considered one of the key factors that induces Parkinson's disease (PD), which is caused by excessive iron accumulation in the intracellular labile iron pool (LIP). The iron ions released from the LIP lead to the aberrant generation of reactive oxygen species (ROS) to trigger ferroptosis and exacerbate PD progression. Herein, a pioneering design of multifunctional nanoregulator deferoxamine (DFO)-integrated nanosheets (BDPR NSs) is presented that target the LIP to restrict ferroptosis and protect against PD. The BDPR NSs are constructed by incorporating a brain-targeting peptide and DFO into polydopamine-modified black phosphorus nanosheets. These BDPR NSs can sequester free iron ions, thereby ameliorating LIP overload and regulating iron metabolism. Furthermore, the BDPR NSs can decrease lipid peroxidation generation by mitigating ROS accumulation. More importantly, BDPR NSs can specifically accumulate in the mitochondria to suppress ROS generation and decrease mitochondrial iron accumulation. In vivo experiments demonstrated that the BDPR NSs highly efficiently mitigated dopaminergic neuronloss and its associated behavioral disorders by modulating the LIP and inhibiting ferroptosis. Thus, the BDPR-based nanovectors holds promise as a potential avenue for advancing PD therapy.
Collapse
Affiliation(s)
- Li Lei
- Department of Chemistry, Engineering Research Center for Molecular Medicine, College of Basic Medical Science, Guizhou Medical University, Guiyang, 550025, China
- Department of Neurosurgery, the Affiliated Hospital of Guizhou Medical University, Guiyang, 550025, China
| | - Jiali Yuan
- Department of Biology, Engineering Research Center for Molecular Medicine, College of Basic Medical Science, Guizhou Medical University, Guiyang, 550025, China
| | - Zhijun Dai
- Department of Biology, Engineering Research Center for Molecular Medicine, College of Basic Medical Science, Guizhou Medical University, Guiyang, 550025, China
| | - Song Xiang
- Department of Biology, Engineering Research Center for Molecular Medicine, College of Basic Medical Science, Guizhou Medical University, Guiyang, 550025, China
| | - Qiuxia Tu
- Department of Biology, Engineering Research Center for Molecular Medicine, College of Basic Medical Science, Guizhou Medical University, Guiyang, 550025, China
| | - Xing Cui
- Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, College of Pharmacy, Guizhou Medical University, Guiyang, 550025, China
| | - Suzhen Zhai
- Department of Biology, Engineering Research Center for Molecular Medicine, College of Basic Medical Science, Guizhou Medical University, Guiyang, 550025, China
| | - Xiaozhong Chen
- The Jinyang Hospital Affiliated to Guizhou Medical University: The Second People's Hospital of Guiyang, Guiyang, 550025, China
| | - Zhixu He
- Department of Biology, Engineering Research Center for Molecular Medicine, College of Basic Medical Science, Guizhou Medical University, Guiyang, 550025, China
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang, 550004, China
| | - Boyan Fang
- Parkinson Medical Center, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, 100144, China
| | - Zhiai Xu
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China
| | - Haijun Yu
- Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Lei Tang
- Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, College of Pharmacy, Guizhou Medical University, Guiyang, 550025, China
| | - Chunlin Zhang
- Department of Biology, Engineering Research Center for Molecular Medicine, College of Basic Medical Science, Guizhou Medical University, Guiyang, 550025, China
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang, 550004, China
| |
Collapse
|
2
|
Hu J, Nieminen AL, Zhong Z, Lemasters JJ. Role of Mitochondrial Iron Uptake in Acetaminophen Hepatotoxicity. LIVERS 2024; 4:333-351. [PMID: 39554796 PMCID: PMC11567147 DOI: 10.3390/livers4030024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2024] Open
Abstract
Overdose of acetaminophen (APAP) produces fulminant hepatic necrosis. The underlying mechanism of APAP hepatotoxicity involves mitochondrial dysfunction, including mitochondrial oxidant stress and the onset of mitochondrial permeability transition (MPT). Reactive oxygen species (ROS) play an important role in APAP-induced hepatotoxicity, and iron is a critical catalyst for ROS formation. This review summarizes the role of mitochondrial ROS formation in APAP hepatotoxicity and further focuses on the role of iron. Normally, hepatocytes take up Fe3+-transferrin bound to transferrin receptors via endocytosis. Concentrated into lysosomes, the controlled release of iron is required for the mitochondrial biosynthesis of heme and non-heme iron-sulfur clusters. After APAP overdose, the toxic metabolite, NAPQI, damages lysosomes, causing excess iron release and the mitochondrial uptake of Fe2+ by the mitochondrial calcium uniporter (MCU). NAPQI also inhibits mitochondrial respiration to promote ROS formation, including H2O2, with which Fe2+ reacts to form highly reactive •OH through the Fenton reaction. •OH, in turn, causes lipid peroxidation, the formation of toxic aldehydes, induction of the MPT, and ultimately, cell death. Fe2+ also facilitates protein nitration. Targeting pathways of mitochondrial iron movement and consequent iron-dependent mitochondrial ROS formation is a promising strategy to intervene against APAP hepatotoxicity in a clinical setting.
Collapse
Affiliation(s)
- Jiangting Hu
- Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Anna-Liisa Nieminen
- Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Zhi Zhong
- Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - John J Lemasters
- Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
3
|
Gao L, Li YJ, Zhao JM, Liao YX, Qin MC, Li JJ, Shi H, Wong NK, Lyu ZP, Shen JG. Mechanism of Reactive Oxygen/Nitrogen Species in Liver Ischemia-Reperfusion Injury and Preventive Effect of Chinese Medicine. Chin J Integr Med 2024:10.1007/s11655-024-3810-9. [PMID: 38941044 DOI: 10.1007/s11655-024-3810-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2024] [Indexed: 06/29/2024]
Abstract
Liver ischemia-reperfusion injury (LIRI) is a pathological process involving multiple injury factors and cell types, with different stages. Currently, protective drugs targeting a single condition are limited in efficacy, and interventions on immune cells will also be accompanied by a series of side effects. In the current bottleneck research stage, the multi-target and obvious clinical efficacy of Chinese medicine (CM) is expected to become a breakthrough point in the research and development of new drugs. In this review, we summarize the roles of reactive oxygen species (ROS) and reactive nitrogen species (RNS) in various stages of hepatic ischemia-reperfusion and on various types of cells. Combined with the current research progress in reducing ROS/RNS with CM, new therapies and mechanisms for the treatment of hepatic ischemia-reperfusion are discussed.
Collapse
Affiliation(s)
- Lei Gao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Yun-Jia Li
- The First Affiliated Hospital/the First Clinical Medicine School of Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Jia-Min Zhao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Yu-Xin Liao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Meng-Chen Qin
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Jun-Jie Li
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Hao Shi
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Nai-Kei Wong
- State Key Discipline of Infectious Diseases, Shenzhen Third People's Hospital, the Second Affiliated Hospital, Shenzhen University, Shenzhen, Guangdong Province, 518112, China
| | - Zhi-Ping Lyu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Jian-Gang Shen
- School of Chinese Medicine, the University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
4
|
Zhou Y, Qiu T, Wang T, Yu B, Xia K, Guo J, Liu Y, Ma X, Zhang L, Zou J, Chen Z, Zhou J. Research progress on the role of mitochondria in the process of hepatic ischemia-reperfusion injury. Gastroenterol Rep (Oxf) 2024; 12:goae066. [PMID: 38912038 PMCID: PMC11193119 DOI: 10.1093/gastro/goae066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 11/08/2023] [Accepted: 02/26/2024] [Indexed: 06/25/2024] Open
Abstract
During liver ischemia-reperfusion injury, existing mechanisms involved oxidative stress, calcium overload, and the activation of inflammatory responses involve mitochondrial injury. Mitochondrial autophagy, a process that maintains the normal physiological activity of mitochondria, promotes cellular metabolism, improves cellular function, and facilitates organelle renewal. Mitochondrial autophagy is involved in oxidative stress and apoptosis, of which the PINK1-Parkin pathway is a major regulatory pathway, and the deletion of PINK1 and Parkin increases mitochondrial damage, reactive oxygen species production, and inflammatory response, playing an important role in mitochondrial quality regulation. In addition, proper mitochondrial permeability translational cycle regulation can help maintain mitochondrial stability and mitigate hepatocyte death during ischemia-reperfusion injury. This mechanism is also closely related to oxidative stress, calcium overload, and the aforementioned autophagy pathway, all of which leads to the augmentation of the mitochondrial membrane permeability transition pore opening and cause apoptosis. Moreover, the release of mitochondrial DNA (mtDNA) due to oxidative stress further aggravates mitochondrial function impairment. Mitochondrial fission and fusion are non-negligible processes required to maintain the dynamic renewal of mitochondria and are essential to the dynamic stability of these organelles. The Bcl-2 protein family also plays an important regulatory role in the mitochondrial apoptosis signaling pathway. A series of complex mechanisms work together to cause hepatic ischemia-reperfusion injury (HIRI). This article reviews the role of mitochondria in HIRI, hoping to provide new therapeutic clues for alleviating HIRI in clinical practice.
Collapse
Affiliation(s)
- Yujie Zhou
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Tao Qiu
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Tianyu Wang
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Bo Yu
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Kang Xia
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Jiayu Guo
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Yiting Liu
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Xiaoxiong Ma
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Long Zhang
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Jilin Zou
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Zhongbao Chen
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Jiangqiao Zhou
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| |
Collapse
|
5
|
Yang B, Xu Y, Yu J, Wang Q, Fan Q, Zhao X, Qiao Y, Zhang Z, Zhou Q, Yin D, He M, He H. Salidroside pretreatment alleviates ferroptosis induced by myocardial ischemia/reperfusion through mitochondrial superoxide-dependent AMPKα2 activation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155365. [PMID: 38552436 DOI: 10.1016/j.phymed.2024.155365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/28/2023] [Accepted: 01/14/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Ferroptosis, a form of regulated cell death (RCD) that relies on excessive reactive oxygen species (ROS) generation, Fe2+accumulation, abnormal lipid metabolism and is involved in various organ ischemia/reperfusion (I/R) injury, expecially in myocardium. Mitochondria are the powerhouses of eukaryotic cells and essential in regulating multiple RCD. However, the links between mitochondria and ferroptosis are still poorly understood. Salidroside (Sal), a natural phenylpropanoid glycoside isolated from Rhodiola rosea, has mult-bioactivities. However, the effects and mechanism in alleviating ferroptosis caused by myocardial I/R injury remains unclear. PURPOSE This study aimed to investigate whether pretreated with Sal could protect the myocardium against I/R damage and the underlying mechanisms. In particular, the relationship between Sal pretreatment, AMPKα2 activity, mitochondria and ROS generation was explored. STUDY DESIGN AND METHODS Firstly, A/R or I/R injury models were employed in H9c2 cells and Sprague-Dawley rats. And then the anti-ferroptotic effects and mechanism of Sal pretreatment was detected using multi-relevant indexes in H9c2 cells. Further, how does Sal pretreatment in AMPKα2 phosphorylation was explored. Finally, these results were validated by I/R injury in rats. RESULTS Similar to Ferrostatin-1 (a ferroptosis inhibitor) and MitoTEMPO, a mitochondrial free radical scavenger, Sal pretreatment effectively alleviated Fe2+ accumulation, redox disequilibrium and maintained mitochondrial energy production and function in I/R-induced myocardial injury, as demonstrated using multifunctional, enzymatic, and morphological indices. However, these effects were abolished by downregulation of AMPKα2 using an adenovirus, both in vivo and in vitro. Moreover, the results also provided a non-canonical mechanism that, under mild mitochondrial ROS generation, Sal pretreatment upregulated and phosphorylated AMPKα2, which enhanced mitochondrial complex I activity to activate innate adaptive responses and increase cellular tolerance to A/R injury. CONCLUSION Overall, our work highlighted mitochondria are of great impotance in myocardial I/R-induced ferroptosis and demonstrated that Sal pretreatment activated AMPKα2 against I/R injury, indicating that Sal could become a candidate phytochemical for the treatment of myocardial I/R injury.
Collapse
Affiliation(s)
- Bin Yang
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Ying Xu
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Jingzhi Yu
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Qihao Wang
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Qigui Fan
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Xiaoyu Zhao
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Yang Qiao
- Jiangxi Academy of Clinical Medical Sciences, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Zeyu Zhang
- Jiangxi Academy of Clinical Medical Sciences, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Qing Zhou
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Dong Yin
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Ming He
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China.
| | - Huan He
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China.
| |
Collapse
|
6
|
Hu J, Nieminen AL, Weemhoff JL, Jaeschke H, Murphy LG, Dent JA, Lemasters JJ. The mitochondrial calcium uniporter mediates mitochondrial Fe 2+ uptake and hepatotoxicity after acetaminophen. Toxicol Appl Pharmacol 2023; 479:116722. [PMID: 37848124 PMCID: PMC10872750 DOI: 10.1016/j.taap.2023.116722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/23/2023] [Accepted: 10/11/2023] [Indexed: 10/19/2023]
Abstract
Acetaminophen (APAP) overdose disrupts hepatocellular lysosomes, which release ferrous iron (Fe2+) that translocates into mitochondria putatively via the mitochondrial calcium uniporter (MCU) to induce oxidative/nitrative stress, the mitochondrial permeability transition (MPT), and hepatotoxicity. To investigate how MCU deficiency affects mitochondrial Fe2+ uptake and hepatotoxicity after APAP overdose, global MCU knockout (KO), hepatocyte specific (hs) MCU KO, and wildtype (WT) mice were treated with an overdose of APAP both in vivo and in vitro. Compared to strain-specific WT mice, serum ALT decreased by 88 and 56%, respectively, in global and hsMCU KO mice at 24 h after APAP (300 mg/kg). Hepatic necrosis also decreased by 84 and 56%. By contrast, when MCU was knocked out in Kupffer cells, ALT release and necrosis were unchanged after overdose APAP. Intravital multiphoton microscopy confirmed loss of viability and mitochondrial depolarization in pericentral hepatocytes of WT mice, which was decreased in MCU KO mice. CYP2E1 expression, hepatic APAP-protein adduct formation, and JNK activation revealed that APAP metabolism was equivalent between WT and MCU KO mice. In cultured hepatocytes after APAP, loss of cell viability decreased in hsMCU KO compared to WT hepatocytes. Using fructose plus glycine to prevent cell killing, mitochondrial Fe2+ increased progressively after APAP, as revealed with mitoferrofluor (MFF), a mitochondrial Fe2+ indicator. By contrast in hsMCU KO hepatocytes, mitochondrial Fe2+ uptake after APAP was suppressed. Rhod-2 measurements showed that Ca2+ did not increase in mitochondria after APAP in either WT or KO hepatocytes. In conclusion, MCU mediates uptake of Fe2+ into mitochondria after APAP and plays a central role in mitochondrial depolarization and cell death during APAP-induced hepatotoxicity.
Collapse
Affiliation(s)
- Jiangting Hu
- Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, SC, United States of America; Departments of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States of America
| | - Anna-Liisa Nieminen
- Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, SC, United States of America; Departments of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States of America; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States of America
| | - James L Weemhoff
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States of America
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States of America
| | - Laura G Murphy
- Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, SC, United States of America; Departments of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States of America
| | - Judith A Dent
- Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, SC, United States of America; Departments of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States of America
| | - John J Lemasters
- Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, SC, United States of America; Departments of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States of America; Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC, United States of America; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States of America.
| |
Collapse
|
7
|
Tao H, Dar HY, Tian C, Banerjee S, Glazer ES, Srinivasan S, Zhu L, Pacifici R, He P. Differences in hepatocellular iron metabolism underlie sexual dimorphism in hepatocyte ferroptosis. Redox Biol 2023; 67:102892. [PMID: 37741044 PMCID: PMC10519854 DOI: 10.1016/j.redox.2023.102892] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 09/15/2023] [Indexed: 09/25/2023] Open
Abstract
Males show higher incidence and severity than females in hepatic injury and many liver diseases, but the mechanisms are not well understood. Ferroptosis, an iron-mediated lipid peroxidation-dependent death, plays an important role in the pathogenesis of liver diseases. We determined whether hepatocyte ferroptosis displays gender difference, accounting for sexual dimorphism in liver diseases. Compared to female hepatocytes, male hepatocytes were much more vulnerable to ferroptosis by iron and pharmacological inducers including RSL3 and iFSP1. Male but not female hepatocytes exhibited significant increases in mitochondrial Fe2+ and mitochondrial ROS (mtROS) contents. Female hepatocytes showed a lower expression of iron importer transferrin receptor 1 (TfR1) and mitochondrial iron importer mitoferrin 1 (Mfrn1), but a higher expression of iron storage protein ferritin heavy chain 1 (FTH1). It is well known that TfR1 expression is positively correlated with ferroptosis. Herein, we showed that silencing FTH1 enhanced while knockdown of Mfrn1 decreased ferroptosis in HepG2 cells. Removing female hormones by ovariectomy (OVX) did not dampen but rather enhanced hepatocyte resistance to ferroptosis. Mechanistically, OVX potentiated the decrease in TfR1 and increase in FTH1 expression. OVX also increased FSP1 expression in ERK-dependent manner. Elevation in FSP1 suppressed mitochondrial Fe2+ accumulation and mtROS production, constituting a novel mechanism of FSP1-mediated inhibition of ferroptosis. In conclusion, differences in hepatocellular iron handling between male and female account, at least in part, for sexual dimorphism in induced ferroptosis of the hepatocytes.
Collapse
Affiliation(s)
- Hui Tao
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Hamid Y Dar
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Cheng Tian
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Somesh Banerjee
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Evan S Glazer
- Departments of Surgery and Cancer Center, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Shanthi Srinivasan
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA; Atlanta Veterans Administration Medical Center, Decatur, GA, USA
| | - Liqin Zhu
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Roberto Pacifici
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Peijian He
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, 615 Michael Street, Atlanta, GA, 30322, USA.
| |
Collapse
|
8
|
Tao H, Dar HY, Tian C, Banerjee S, Glazer ES, Srinivasan S, Zhu L, Pacifici R, He P. Differences in Hepatocellular Iron Metabolism Underlie Sexual Dimorphism in Hepatocyte Ferroptosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.24.546395. [PMID: 37425728 PMCID: PMC10327041 DOI: 10.1101/2023.06.24.546395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Males show higher incidence and severity than females in hepatic injury and many liver diseases, but the mechanisms are not well understood. Ferroptosis, an iron-mediated lipid peroxidation-dependent death, plays an important role in the pathogenesis of liver diseases. We determined whether hepatocyte ferroptosis displays gender difference, accounting for sexual dimorphism in liver diseases. Compared to female hepatocytes, male hepatocytes were much more vulnerable to ferroptosis by iron and pharmacological inducers including RSL3 and iFSP1. Male but not female hepatocytes exhibited significant increases in mitochondrial Fe 2+ and mitochondrial ROS (mtROS) contents. Female hepatocytes showed a lower expression of iron importer transferrin receptor 1 (TfR1) and mitochondrial iron importer mitoferrin 1 (Mfrn1), but a higher expression of iron storage protein ferritin heavy chain 1 (FTH1). It is well known that TfR1 expression is positively correlated with ferroptosis. Herein, we showed that silencing FTH1 enhanced while knockdown of Mfrn1 decreased ferroptosis in HepG2 cells. Removing female hormones by ovariectomy (OVX) did not dampen but rather enhanced hepatocyte resistance to ferroptosis. Mechanistically, OVX potentiated the decrease in TfR1 and increase in FTH1 expression. OVX also increased FSP1 expression in ERK-dependent manner. Elevation in FSP1 suppressed mitochondrial Fe 2+ accumulation and mtROS production, constituting a novel mechanism of FSP1-mediated inhibition of ferroptosis. In conclusion, differences in hepatocellular iron handling between male and female account, at least in part, for sexual dimorphism in induced ferroptosis of the hepatocytes.
Collapse
|
9
|
Fedotcheva T, Shimanovsky N, Fedotcheva N. Specific Features of Mitochondrial Dysfunction under Conditions of Ferroptosis Induced by t-Butylhydroperoxide and Iron: Protective Role of the Inhibitors of Lipid Peroxidation and Mitochondrial Permeability Transition Pore Opening. MEMBRANES 2023; 13:372. [PMID: 37103799 PMCID: PMC10145271 DOI: 10.3390/membranes13040372] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/18/2023] [Accepted: 03/21/2023] [Indexed: 06/19/2023]
Abstract
Recent studies have indicated the critical importance of mitochondria in the induction and progression of ferroptosis. There is evidence indicating that tert-butyl hydroperoxide (TBH), a lipid-soluble organic peroxide, is capable of inducing ferroptosis-type cell death. We investigated the effect of TBH on the induction of nonspecific membrane permeability measured by mitochondrial swelling and on oxidative phosphorylation and NADH oxidation assessed by NADH fluo rescence. TBH and iron, as well as their combinations, induced, with a respective decrease in the lag phase, the swelling of mitochondria, inhibited oxidative phosphorylation and stimulated NADH oxidation. The lipid radical scavenger butylhydroxytoluene (BHT), the inhibitor of mitochondrial phospholipase iPLA2γ bromoenol lactone (BEL), and the inhibitor of the mitochondrial permeability transition pore (MPTP) opening cyclosporine A (CsA) were equally effective in protecting these mitochondrial functions. The radical-trapping antioxidant ferrostatin-1, a known indicator of ferroptotic alteration, restricted the swelling but was less effective than BHT. ADP and oligomycin significantly decelerated iron- and TBH-induced swelling, confirming the involvement of MPTP opening in mitochondrial dysfunction. Thus, our data showed the participation of phospholipase activation, lipid peroxidation, and the MPTP opening in the mitochondria-dependent ferroptosis. Presumably, their involvement took place at different stages of membrane damage initiated by ferroptotic stimuli.
Collapse
Affiliation(s)
- Tatiana Fedotcheva
- Science Research Laboratory of Molecular Pharmacology, Medical Biological Faculty, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Ostrovityanova St. 1, Moscow 117997, Russia
| | - Nikolai Shimanovsky
- Science Research Laboratory of Molecular Pharmacology, Medical Biological Faculty, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Ostrovityanova St. 1, Moscow 117997, Russia
| | - Nadezhda Fedotcheva
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya str., 3, Pushchino 142290, Russia
| |
Collapse
|
10
|
Liu X, Li W, Wang M, Liu N, Yang Q, He Y, Hu D, Zhu R, Yin L. Inflammatory Cell-Inspired Cascade Nanozyme Induces Intracellular Radical Storm for Enhanced Anticancer Therapy. SMALL METHODS 2023; 7:e2201641. [PMID: 36610035 DOI: 10.1002/smtd.202201641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Indexed: 06/17/2023]
Abstract
Manipulating intracellular levels of reactive oxygen and nitrogen species (RONS) is of great potential for cancer treatment. Inspired by the natural mechanism of a radical storm in inflammatory cells via activated and regulatable biocatalysis, the authors herein report a self-powered nanozyme that can enable RONS production in tumor cells via cascade reactions. The nanozymes are constructed via glucose oxidase (GOx)-templated inverse microemulsion polymerization from acrylamide, arginine-acrylamide, ferrocene-acrylate, and N,N'-bis(acryloyl)cystamine, followed by surface coating with hyaluronic acid. After targeted delivery into cancer cells, the nanozymes are dissociated by intracellular glutathione to release GOx, which decomposed glucose to generate gluconic acid and H2 O2 . Under such acidified conditions, H2 O2 efficiently oxidized pendant arginine residues to produce nitric oxide , transformed into a highly toxic hydroxyl radical and superoxide anion via ferrocene-mediated Fenton reaction and Haber-Weiss cycle, and simultaneously generated peroxynitrite anion via reaction between NO and ·O2 - , thus provoking the RONS radical storm to effectively eradicate A549 tumor cells both in vitro and in vivo. This nature-inspired enzyme-chemical dynamic therapy may provide a promising modality for anti-cancer treatment.
Collapse
Affiliation(s)
- Xun Liu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, P. R. China
- Department of Thoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, P. R. China
| | - Wei Li
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, P. R. China
| | - Mengru Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, P. R. China
| | - Ningyu Liu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, P. R. China
| | - Qiang Yang
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, P. R. China
| | - Yunjie He
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, P. R. China
| | - Duanmin Hu
- Department of Gastroenterology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, P. R. China
| | - Rongying Zhu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, P. R. China
| | - Lichen Yin
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, P. R. China
| |
Collapse
|
11
|
Mantelou AG, Barbouti A, Goussia A, Zacharioudaki A, Papoudou-Bai A, Vlachou C, Kokkoris S, Papalois A, Galaris D, Glantzounis GK. Combined administration of membrane-permeable and impermeable iron-chelating drugs attenuates ischemia/reperfusion-induced hepatic injury. Free Radic Biol Med 2022; 193:227-237. [PMID: 36243210 DOI: 10.1016/j.freeradbiomed.2022.10.266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 10/07/2022] [Accepted: 10/07/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND The underlying pathophysiological mechanisms of hepatic ischemia-reperfusion (I/R) injury have not been completely elucidated. However, it is well known that oxidative stress, caused by a burst of reactive oxygen species (ROS) production during the reperfusion phase, plays a crucial role. A growing body of evidence indicates that the intracellular availability of free iron represents a requirement for ROS-induced adverse effects, as iron catalyzes the generation of highly reactive free radicals. The aim of this study was to examine whether a combination of iron chelators with varying lipophilicity could offer enhanced protection against I/R by diminishing the conversion of weak oxidants, like H2O2, to extremely reactive ones such as hydroxyl radicals (HO.). METHODS HepG2 cells (hepatocellular carcinoma cell line) were exposed to oxidative stress conditions after pre-treatment with the iron chelators desferrioxamine (DFO) and deferiprone (DFP) alone or in combination. Labile iron pool was estimated using the calcein-acetoxymethyl ester (calcein-AM) method and DNA damage with the comet assay. We subsequently used a rabbit model (male New Zealand white rabbits) of hepatic I/R-induced injury to investigate, by measuring biochemical (ALT, ALT, ALP, γGT) and histological parameters, whether this may be true for in vivo conditions. RESULTS The combination of a membrane-permeable iron chelator (DFP) with a strong membrane-impermeable one (DFO) raises the level of protection in both hepatic cell lines exposed to oxidative stress conditions and hepatic I/R rabbit model. CONCLUSIONS Our results show that combinations of iron chelators with selected lipophilicity and iron-binding properties may represent a valuable strategy to protect against tissue damage during reperfusion after a period of ischemia.
Collapse
Affiliation(s)
- Athina G Mantelou
- HPB Unit, Department of Surgery, University Hospital of Ioannina and Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, 45110, Greece
| | - Alexandra Barbouti
- Department of Anatomy-Histology-Embryology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, 45110, Greece
| | - Anna Goussia
- Department of Pathology, University Hospital of Ioannina and Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, 45110, Greece
| | | | - Alexandra Papoudou-Bai
- Department of Pathology, University Hospital of Ioannina and Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, 45110, Greece
| | - Chara Vlachou
- Laboratory of Biological Chemistry, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, 45110, Greece
| | - Stelios Kokkoris
- First Department of Critical Care, School of Medicine, National and Kapodistrian University of Athens, Athens, 10676, Greece
| | - Apostolos Papalois
- Experimental, Educational and Research Center ELPEN, Athens, 19009, Greece; European University of Cyprus, School of Medicine, Nicosia, 2404, Cyprus
| | - Dimitrios Galaris
- Laboratory of Biological Chemistry, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, 45110, Greece
| | - Georgios K Glantzounis
- HPB Unit, Department of Surgery, University Hospital of Ioannina and Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, 45110, Greece.
| |
Collapse
|
12
|
Kholmukhamedov A, Li L, Lindsey CC, Hu J, Nieminen AL, Takemoto K, Beeson GC, Beneker CM, McInnes C, Beeson CC, Lemasters JJ. A new fluorescent sensor mitoferrofluor indicates the presence of chelatable iron in polarized and depolarized mitochondria. J Biol Chem 2022; 298:102336. [PMID: 35931111 PMCID: PMC9460511 DOI: 10.1016/j.jbc.2022.102336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 11/28/2022] Open
Abstract
Mitochondrial chelatable iron contributes to the severity of several injury processes, including ischemia/reperfusion, oxidative stress, and drug toxicity. However, methods to measure this species in living cells are lacking. To measure mitochondrial chelatable iron in living cells, here we synthesized a new fluorescent indicator, mitoferrofluor (MFF). We designed cationic MFF to accumulate electrophoretically in polarized mitochondria, where a reactive group then forms covalent adducts with mitochondrial proteins to retain MFF even after subsequent depolarization. We also show in cell-free medium that Fe2+ (and Cu2+), but not Fe3+, Ca2+, or other biologically relevant divalent cations, strongly quenched MFF fluorescence. Using confocal microscopy, we demonstrate in hepatocytes that red MFF fluorescence colocalized with the green fluorescence of the mitochondrial membrane potential (ΔΨm) indicator, rhodamine 123 (Rh123), indicating selective accumulation into the mitochondria. Unlike Rh123, mitochondria retained MFF after ΔΨm collapse. Furthermore, intracellular delivery of iron with membrane-permeant Fe3+/8-hydroxyquinoline (FeHQ) quenched MFF fluorescence by ∼80% in hepatocytes and other cell lines, which was substantially restored by the membrane-permeant transition metal chelator pyridoxal isonicotinoyl hydrazone. We also show FeHQ quenched the fluorescence of cytosolically coloaded calcein, another Fe2+ indicator, confirming that Fe3+ in FeHQ undergoes intracellular reduction to Fe2+. Finally, MFF fluorescence did not change after addition of the calcium mobilizer thapsigargin, which shows MFF is insensitive to physiologically relevant increases of mitochondrial Ca2+. In conclusion, the new sensor reagent MFF fluorescence is an indicator of mitochondrial chelatable Fe2+ in normal hepatocytes with polarized mitochondria as well as in cells undergoing loss of ΔΨm.
Collapse
Affiliation(s)
- Andaleb Kholmukhamedov
- Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, South Carolina, USA,Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Li Li
- Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, South Carolina, USA,Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Christopher C. Lindsey
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Jiangting Hu
- Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, South Carolina, USA,Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Anna-Liisa Nieminen
- Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, South Carolina, USA,Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina, USA,Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Kenji Takemoto
- Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, South Carolina, USA,Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Gyda C. Beeson
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Chad M. Beneker
- Department of Drug Discovery & Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, South Carolina, USA
| | - Campbell McInnes
- Department of Drug Discovery & Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, South Carolina, USA
| | - Craig C. Beeson
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
| | - John J. Lemasters
- Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, South Carolina, USA,Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina, USA,Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA,Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, USA,For correspondence: John J. Lemasters
| |
Collapse
|
13
|
Ritiu SA, Rogobete AF, Sandesc D, Bedreag OH, Papurica M, Popovici SE, Toma D, Ivascu RI, Velovan R, Garofil DN, Corneci D, Bratu LM, Pahontu EM, Pistol A. The Impact of General Anesthesia on Redox Stability and Epigenetic Inflammation Pathways: Crosstalk on Perioperative Antioxidant Therapy. Cells 2022; 11:1880. [PMID: 35741011 PMCID: PMC9221536 DOI: 10.3390/cells11121880] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 05/31/2022] [Accepted: 06/07/2022] [Indexed: 02/07/2023] Open
Abstract
Worldwide, the prevalence of surgery under general anesthesia has significantly increased, both because of modern anesthetic and pain-control techniques and because of better diagnosis and the increased complexity of surgical techniques. Apart from developing new concepts in the surgical field, researchers and clinicians are now working on minimizing the impact of surgical trauma and offering minimal invasive procedures due to the recent discoveries in the field of cellular and molecular mechanisms that have revealed a systemic inflammatory and pro-oxidative impact not only in the perioperative period but also in the long term, contributing to more difficult recovery, increased morbidity and mortality, and a negative financial impact. Detailed molecular and cellular analysis has shown an overproduction of inflammatory and pro-oxidative species, responsible for augmenting the systemic inflammatory status and making postoperative recovery more difficult. Moreover, there are a series of changes in certain epigenetic structures, the most important being the microRNAs. This review describes the most important molecular and cellular mechanisms that impact the surgical patient undergoing general anesthesia, and it presents a series of antioxidant therapies that can reduce systemic inflammation.
Collapse
Affiliation(s)
- Stelian Adrian Ritiu
- Clinic of Anaesthesia and Intensive Care, Emergency County Hospital “Pius Brînzeu”, 300723 Timișoara, Romania; (S.A.R.); (D.S.); (O.H.B.); (M.P.); (S.E.P.); (D.T.); (R.V.)
- Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania;
| | - Alexandru Florin Rogobete
- Clinic of Anaesthesia and Intensive Care, Emergency County Hospital “Pius Brînzeu”, 300723 Timișoara, Romania; (S.A.R.); (D.S.); (O.H.B.); (M.P.); (S.E.P.); (D.T.); (R.V.)
- Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania;
- Anaesthesia and Intensive Care Research Center (CCATITM), “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania
| | - Dorel Sandesc
- Clinic of Anaesthesia and Intensive Care, Emergency County Hospital “Pius Brînzeu”, 300723 Timișoara, Romania; (S.A.R.); (D.S.); (O.H.B.); (M.P.); (S.E.P.); (D.T.); (R.V.)
- Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania;
- Anaesthesia and Intensive Care Research Center (CCATITM), “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania
| | - Ovidiu Horea Bedreag
- Clinic of Anaesthesia and Intensive Care, Emergency County Hospital “Pius Brînzeu”, 300723 Timișoara, Romania; (S.A.R.); (D.S.); (O.H.B.); (M.P.); (S.E.P.); (D.T.); (R.V.)
- Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania;
- Anaesthesia and Intensive Care Research Center (CCATITM), “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania
| | - Marius Papurica
- Clinic of Anaesthesia and Intensive Care, Emergency County Hospital “Pius Brînzeu”, 300723 Timișoara, Romania; (S.A.R.); (D.S.); (O.H.B.); (M.P.); (S.E.P.); (D.T.); (R.V.)
- Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania;
- Anaesthesia and Intensive Care Research Center (CCATITM), “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania
| | - Sonia Elena Popovici
- Clinic of Anaesthesia and Intensive Care, Emergency County Hospital “Pius Brînzeu”, 300723 Timișoara, Romania; (S.A.R.); (D.S.); (O.H.B.); (M.P.); (S.E.P.); (D.T.); (R.V.)
- Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania;
| | - Daiana Toma
- Clinic of Anaesthesia and Intensive Care, Emergency County Hospital “Pius Brînzeu”, 300723 Timișoara, Romania; (S.A.R.); (D.S.); (O.H.B.); (M.P.); (S.E.P.); (D.T.); (R.V.)
- Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania;
| | - Robert Iulian Ivascu
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (R.I.I.); (D.C.); (A.P.)
- Clinic of Anaesthesia and Intensive Care, Central Military Emergency Hospital “Dr. Carol Davila”, 010242 Bucharest, Romania
| | - Raluca Velovan
- Clinic of Anaesthesia and Intensive Care, Emergency County Hospital “Pius Brînzeu”, 300723 Timișoara, Romania; (S.A.R.); (D.S.); (O.H.B.); (M.P.); (S.E.P.); (D.T.); (R.V.)
- Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania;
| | - Dragos Nicolae Garofil
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (R.I.I.); (D.C.); (A.P.)
| | - Dan Corneci
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (R.I.I.); (D.C.); (A.P.)
- Clinic of Anaesthesia and Intensive Care, Central Military Emergency Hospital “Dr. Carol Davila”, 010242 Bucharest, Romania
| | - Lavinia Melania Bratu
- Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania;
| | - Elena Mihaela Pahontu
- Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Adriana Pistol
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (R.I.I.); (D.C.); (A.P.)
| |
Collapse
|
14
|
Zhang S, Rao S, Yang M, Ma C, Hong F, Yang S. Role of Mitochondrial Pathways in Cell Apoptosis during He-Patic Ischemia/Reperfusion Injury. Int J Mol Sci 2022; 23:ijms23042357. [PMID: 35216473 PMCID: PMC8877300 DOI: 10.3390/ijms23042357] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/13/2022] [Accepted: 02/17/2022] [Indexed: 12/15/2022] Open
Abstract
Hepatic ischemia-reperfusion injury is a major cause of post-operative hepatic dysfunction and liver failure after transplantation. Mitochondrial pathways can be either beneficial or detrimental to hepatic cell apoptosis during hepatic ischemia/reperfusion injury, depending on multiple factors. Hepatic ischemia/reperfusion injury may be induced by opened mitochondrial permeability transition pore, released apoptosis-related proteins, up-regulated B-cell lymphoma-2 gene family proteins, unbalanced mitochondrial dynamics, and endoplasmic reticulum stress, which are integral parts of mitochondrial pathways. In this review, we discuss the role of mitochondrial pathways in apoptosis that account for the most deleterious effect of hepatic ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Sen Zhang
- Experimental Center of Pathogen Biology, College of Medicine, Nanchang University, Nanchang 330006, China; (S.Z.); (S.R.); (C.M.)
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China
| | - Sijing Rao
- Experimental Center of Pathogen Biology, College of Medicine, Nanchang University, Nanchang 330006, China; (S.Z.); (S.R.); (C.M.)
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China
| | - Meiwen Yang
- Department of Surgery, Fuzhou Medical College, Nanchang University, Fuzhou 344099, China;
| | - Chen Ma
- Experimental Center of Pathogen Biology, College of Medicine, Nanchang University, Nanchang 330006, China; (S.Z.); (S.R.); (C.M.)
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China
| | - Fengfang Hong
- Experimental Center of Pathogen Biology, College of Medicine, Nanchang University, Nanchang 330006, China; (S.Z.); (S.R.); (C.M.)
- Correspondence: (F.H.); or (S.Y.)
| | - Shulong Yang
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China
- Department of Physiology, Fuzhou Medical College, Nanchang University, Fuzhou 344099, China
- Correspondence: (F.H.); or (S.Y.)
| |
Collapse
|
15
|
Fedotcheva TA, Fedotcheva NI. Protectors of the Mitochondrial Permeability Transition Pore Activated by Iron and Doxorubicin. Curr Cancer Drug Targets 2021; 21:514-525. [PMID: 33475063 DOI: 10.2174/1568009621999210120192558] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 12/03/2020] [Accepted: 12/03/2020] [Indexed: 11/22/2022]
Abstract
AIM The study is aimed at examining of action of iron, DOX, and their complex on the Mitochondrial Permeability Transition Pore (MPTP) opening and detecting of possible protectors of MPTP in the conditions close to mitochondria-dependent ferroptosis. BACKGROUND The Toxicity of Doxorubicin (DOX) is mainly associated with free iron accumulation and mitochondrial dysfunction. DOX can provoke ferroptosis, iron-dependent cell death driven by membrane damage. The Mitochondrial Permeability Transition Pore (MPTP) is considered as a common pathway leading to the development of apoptosis, necrosis, and, possibly, ferroptosis. The influence of DOX on the Ca2+ -induced MPTP opening in the presence of iron has not yet been studied. OBJECTIVE The study was conducted on isolated liver and heart mitochondria. MPTP and succinate- ubiquinone oxidoreductase were studied as targets of DOX in mitochondria-dependent ferroptosis. The iron chelator deferoxamine (DFO), the lipid radical scavenger butyl-hydroxytoluene (BHT), and rutenium red (Rr), as a possible inhibitor of ferrous ions uptake in mitochondria, were tested as MPTP protectors. The role of medium alkalization was also examined. METHODS Changes of threshold calcium concentrations required for MPTP opening were measured by a Ca2+ selective electrode, mitochondrial membrane potential was registered by tetraphenylphosphonium (TPP+)-selective electrode, and mitochondrial swelling was recorded as a decrease in absorbance at 540 nm. The activity of Succinate Dehydrogenase (SDH) was determined by the reduction of the electron acceptor DCPIP. CONCLUSION MPTP and the respiratory complex II are identified as the main targets of the iron-dependent action of DOX on the isolated mitochondria. All MPTP protectors tested abolished or weakened the effect of iron and a complex of iron with DOX on Ca2+ -induced MPTP opening, acting in different stages of MPTP activation. These data open new approaches to the modulation of the toxic influence of DOX on mitochondria with the aim to reduce their dysfunction.
Collapse
Affiliation(s)
- Tatiana A Fedotcheva
- Science Research Laboratory of Pharmacology, Faculty of Medical Biology, N. I. Pirogov Russian National Medical Research University, Ministry of Health of the Russian Federation, Moscow, Russian Federation
| | - Nadezhda I Fedotcheva
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow region, 142290, Russian Federation
| |
Collapse
|
16
|
Fedotcheva N, Olenin A, Beloborodova N. Influence of Microbial Metabolites on the Nonspecific Permeability of Mitochondrial Membranes under Conditions of Acidosis and Loading with Calcium and Iron Ions. Biomedicines 2021; 9:biomedicines9050558. [PMID: 34067718 PMCID: PMC8156683 DOI: 10.3390/biomedicines9050558] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 12/26/2022] Open
Abstract
Mitochondrial dysfunction is currently considered one of the main causes of multiple organ failure in chronic inflammation and sepsis. The participation of microbial metabolites in disorders of bioenergetic processes in mitochondria has been revealed, but their influence on the mitochondrial membrane permeability has not yet been studied. We tested the influence of various groups of microbial metabolites, including indolic and phenolic acids, trimethylamine-N-oxide (TMAO) and acetyl phosphate (AcP), on the nonspecific permeability of mitochondrial membranes under conditions of acidosis, imbalance of calcium ions and excess free iron, which are inherent in sepsis. Changes in the parameters of the calcium-induced opening of the mitochondrial permeability transition pore (MPTP) and iron-activated swelling of rat liver mitochondria were evaluated. The most active metabolites were indole-3-carboxylic acid (ICA) and benzoic acid (BA), which activated MPTP opening and swelling under all conditions. AcP showed the opposite effect on the induction of MPTP opening, increasing the threshold concentration of calcium by 1.5 times, while TMAO activated swelling only under acidification. All the redox-dependent effects of metabolites were suppressed by the lipid radical scavenger butyl-hydroxytoluene (BHT), which indicates the participation of these microbial metabolites in the activation of membrane lipid peroxidation. Thus, microbial metabolites can directly affect the nonspecific permeability of mitochondrial membranes, if conditions of acidosis, an imbalance of calcium ions and an excess of free iron are created in the pathological state.
Collapse
Affiliation(s)
- Nadezhda Fedotcheva
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya Street 3, 142290 Pushchino, Russia
- Correspondence:
| | - Andrei Olenin
- V.I. Vernadsky Institute of Geochemistry and Analytical Chemistry, Russian Academy of Sciences, 19 Kosygin Street, 119991 Moscow, Russia;
| | - Natalia Beloborodova
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 25-2 Petrovka Street, 107031 Moscow, Russia;
| |
Collapse
|
17
|
Xue Y, Liu H, Yang XX, Pang L, Liu J, Ng KTP, Yeung OWH, Lam YF, Zhang WY, Lo CM, Man K. Inhibition of Carnitine Palmitoyltransferase 1A Aggravates Fatty Liver Graft Injury via Promoting Mitochondrial Permeability Transition. Transplantation 2021; 105:550-560. [PMID: 32890136 DOI: 10.1097/tp.0000000000003437] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND Hepatic steatosis is a major risk factor for graft failure due to increased susceptibility of fatty liver to ischemia-reperfusion injury (IRI) during transplantation. Here, we aimed to investigate the role of carnitine palmitoyltransferase 1A (CPT1A) in fatty liver graft injury and to explore the underlying mechanism and therapeutic potential on attenuating hepatic IRI. METHODS Intragraft CPT1A expression profile and the association with fatty graft injury were investigated in human and rat liver transplantation samples. The underlying mechanism and therapeutic potential of CPT1A activator against IRI were also explored in mouse hepatic ischemia-reperfusion plus major hepatectomy model and in in vitro. RESULTS CPT1A expression was significantly reduced (P = 0.0019; n = 96) in human fatty liver graft compared with normal one at early phase after transplantation. Low expression of CPT1A was significantly associated with high serum alanine aminotransferase (P = 0.0144) and aspartate aminotransferase (P = 0.0060) levels. The inhibited CPT1A and poor liver function were consistently observed in rat and mouse models with fatty livers. Furthermore, inhibition of CPT1A significantly promoted the translocation of chloride intracellular channel 1 to form chloride ion channel. The dysregulation of chloride ion channel activity subsequently triggered mitochondrial permeability transition (MPT) pore opening, exacerbated cellular oxidative stress, and energy depletion. Importantly, our intravital confocal imaging showed that CPT1A activation attenuated hepatic injury through preventing MPT after reperfusion in fatty mice. CONCLUSIONS CPT1A inhibition triggered MPT contributed to severe IRI in fatty liver graft. CPT1A restoration may offer therapeutic potential on attenuating hepatic IRI.
Collapse
Affiliation(s)
- Yan Xue
- Department of Surgery, HKU-SZH &LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
van Meteren N, Lagadic-Gossmann D, Podechard N, Gobart D, Gallais I, Chevanne M, Collin A, Burel A, Dupont A, Rault L, Chevance S, Gauffre F, Le Ferrec E, Sergent O. Extracellular vesicles released by polycyclic aromatic hydrocarbons-treated hepatocytes trigger oxidative stress in recipient hepatocytes by delivering iron. Free Radic Biol Med 2020; 160:246-262. [PMID: 32791186 DOI: 10.1016/j.freeradbiomed.2020.08.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 07/14/2020] [Accepted: 08/01/2020] [Indexed: 12/11/2022]
Abstract
A growing body of evidences indicate the major role of extracellular vesicles (EVs) as players of cell communication in the pathogenesis of liver diseases. EVs are membrane-enclosed vesicles released by cells into the extracellular environment. Oxidative stress is also a key component of liver disease pathogenesis, but no role for hepatocyte-derived EVs has yet been described in the development of this process. Recently, some polycyclic aromatic hydrocarbons (PAHs), widespread environmental contaminants, were demonstrated to induce EV release from hepatocytes. They are also well-known to trigger oxidative stress leading to cell death. Therefore, the aim of this work was to investigate the involvement of EVs derived from PAHs-treated hepatocytes (PAH-EVs) in possible oxidative damages of healthy recipient hepatocytes, using both WIF-B9 and primary rat hepatocytes. We first showed that the release of EVs from PAHs -treated hepatocytes depended on oxidative stress. PAH-EVs were enriched in proteins related to oxidative stress such as NADPH oxidase and ferritin. They were also demonstrated to contain more iron. PAH-EVs could then induce oxidative stress in recipient hepatocytes, thereby leading to apoptosis. Mitochondria and lysosomes of recipient hepatocytes exhibited significant structural alterations. All those damages were dependent on internalization of EVs that reached lysosomes with their cargoes. Lysosomes thus appeared as critical organelles for EVs to induce apoptosis. In addition, pro-oxidant components of PAH-EVs, e.g. NADPH oxidase and iron, were revealed to be necessary for this cell death.
Collapse
Affiliation(s)
- Nettie van Meteren
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Dominique Lagadic-Gossmann
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Normand Podechard
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Dimitri Gobart
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Isabelle Gallais
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Martine Chevanne
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Aurore Collin
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Agnès Burel
- Univ Rennes, Biosit, UMS 3480, US_S 018, F-35000, Rennes, France
| | - Aurélien Dupont
- Univ Rennes, Biosit, UMS 3480, US_S 018, F-35000, Rennes, France
| | | | - Soizic Chevance
- Univ Rennes, CNRS, ISCR (Institut des Sciences Chimiques de Rennes) - UMR 6226, F-35000, Rennes, France
| | - Fabienne Gauffre
- Univ Rennes, CNRS, ISCR (Institut des Sciences Chimiques de Rennes) - UMR 6226, F-35000, Rennes, France
| | - Eric Le Ferrec
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Odile Sergent
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé Environnement et Travail), UMR_S 1085, F-35000, Rennes, France.
| |
Collapse
|
19
|
Li L, Tong A, Zhang Q, Wei Y, Wei X. The molecular mechanisms of MLKL-dependent and MLKL-independent necrosis. J Mol Cell Biol 2020; 13:3-14. [PMID: 33064829 PMCID: PMC8035999 DOI: 10.1093/jmcb/mjaa055] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 07/27/2020] [Accepted: 08/14/2020] [Indexed: 02/05/2023] Open
Abstract
Necrosis, a type of unwanted and passive cell demise, usually occurs under the excessive external stress and is considered to be unregulated. However, under some special conditions such as caspase inhibition, necrosis is regulable in a well-orchestrated way. The term 'regulated necrosis' has been proposed to describe such programed necrosis. Recently, several forms of necrosis, including necroptosis, pyroptosis, ferroptosis, parthanatos, oxytosis, NETosis, and Na+/K+-ATPase-mediated necrosis, have been identified, and some crucial regulators governing regulated necrosis have also been discovered. Mixed lineage kinase domain-like pseudokinase (MLKL), a core regulator in necroptosis, acts as an executioner in response to ligands of death receptor family. Its activation requires the receptor-interacting protein kinases, RIP1 and RIP3. However, MLKL is only involved in necroptosis, i.e. MLKL is dispensable for necrosis. Therefore, this review is aimed at summarizing the molecular mechanisms of MLKL-dependent and MLKL-independent necrosis.
Collapse
Affiliation(s)
- Lu Li
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - An Tong
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiangsheng Zhang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
20
|
Jung SH, Lee W, Park SH, Lee KY, Choi YJ, Choi S, Kang D, Kim S, Chang TS, Hong SS, Lee BH. Diclofenac impairs autophagic flux via oxidative stress and lysosomal dysfunction: Implications for hepatotoxicity. Redox Biol 2020; 37:101751. [PMID: 33080439 PMCID: PMC7575798 DOI: 10.1016/j.redox.2020.101751] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/24/2020] [Accepted: 10/02/2020] [Indexed: 12/17/2022] Open
Abstract
Treatment with nonsteroidal anti-inflammatory drugs (NSAIDs) is associated with various side effects, including cardiovascular and hepatic disorders. Studies suggest that mitochondrial damage and oxidative stress are important mediators of toxicity, yet the underlying mechanisms are poorly understood. In this study, we identified that some NSAIDs, including diclofenac, inhibit autophagic flux in hepatocytes. Further detailed studies demonstrated that diclofenac induced a reactive oxygen species (ROS)-dependent increase in lysosomal pH, attenuated cathepsin activity and blocked autophagosome-lysosome fusion. The reactivation of lysosomal function by treatment with clioquinol or transfection with the transcription factor EB restored lysosomal pH and thus autophagic flux. The production of mitochondrial ROS is critical for this process since scavenging ROS reversed lysosomal dysfunction and activated autophagic flux. The compromised lysosomal activity induced by diclofenac also inhibited the fusion with and degradation of mitochondria by mitophagy. Diclofenac-induced cell death and hepatotoxicity were effectively protected by rapamycin. Thus, we demonstrated that diclofenac induces the intracellular ROS production and lysosomal dysfunction that lead to the suppression of autophagy. Impaired autophagy fails to maintain mitochondrial integrity and aggravates the cellular ROS burden, which leads to diclofenac-induced hepatotoxicity.
Collapse
Affiliation(s)
- Seung-Hwan Jung
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea
| | - Wonseok Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea
| | - Seung-Hyun Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea
| | - Kang-Yo Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea
| | - You-Jin Choi
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea
| | - Soohee Choi
- Department of Life Science, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, Republic of Korea
| | - Dongmin Kang
- Department of Life Science, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, Republic of Korea
| | - Sinri Kim
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, Republic of Korea
| | - Tong-Shin Chang
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea
| | - Soon-Sun Hong
- Department of Biomedical Sciences, College of Medicine, Inha University, Sinheung-dong, Jung-gu, Incheon, 400-712, Republic of Korea
| | - Byung-Hoon Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea.
| |
Collapse
|
21
|
Smith MJ, Fowler M, Naftalin RJ, Siow RCM. UVA irradiation increases ferrous iron release from human skin fibroblast and endothelial cell ferritin: Consequences for cell senescence and aging. Free Radic Biol Med 2020; 155:49-57. [PMID: 32387586 DOI: 10.1016/j.freeradbiomed.2020.04.024] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 04/26/2020] [Indexed: 12/13/2022]
Abstract
UVA irradiation of human dermal fibroblasts and endothelial cells induces an immediate transient increase in cytosolic Fe(II), as monitored by the fluorescence Fe(II) reporters, FeRhonox1 in cytosol and MitoFerroGreen in mitochondria. Both superoxide dismutase (SOD) inhibition by tetrathiomolybdate (ATM) and catalase inhibition by 3-amino-1, 2, 4-triazole (ATZ) increase and prolong the cytosolic Fe(II) signal after UVA irradiation. SOD inhibition with ATM also increases mitochondrial Fe(II). Thus, mitochondria do not source the UV-dependent increase in cytosolic Fe(II), but instead reflect and amplify raised cytosolic labile Fe(II) concentration. Hence control of cytosolic ferritin iron release is key to preventing UVA-induced inflammation. UVA irradiation also increases dermal endothelial cell H2O2, as monitored by the adenovirus vector Hyper-DAAO-NES(HyPer). These UVA-dependent changes in intracellular Fe(II) and H2O2 are mirrored by increases in cell superoxide, monitored with the luminescence probe L-012. UV-dependent increases in cytosolic Fe(II), H2O2 and L-012 chemiluminescence are prevented by ZnCl2 (10 μM), an effective inhibitor of Fe(II) transport via ferritin's 3-fold channels. Quercetin (10 μM), a potent membrane permeable Fe(II) chelator, abolishes the cytosolic UVA-dependent FeRhonox1, Fe(II) and HyPer, H2O2 and increase in MitoFerroGreen Fe(II) signals. The time course of the quercetin-dependent decrease in endothelial H2O2 correlates with the decrease in FeRhox1 signal and both signals are fully suppressed by preloading cells with ZnCl2. These results confirm that antioxidant enzyme activity is the key factor in controlling intracellular iron levels, and hence maintenance of cell antioxidant capacity is vitally important in prevention of skin aging and inflammation initiated by labile iron and UVA.
Collapse
Affiliation(s)
- Matthew J Smith
- King's BHF Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, Faculty of Life Sciences & Medicine, 150 Stamford Street, London, SE1 9NH, UK
| | - Mark Fowler
- Unilever Colworth Science Park, Bedfordshire, UK
| | - Richard J Naftalin
- King's BHF Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, Faculty of Life Sciences & Medicine, 150 Stamford Street, London, SE1 9NH, UK.
| | - Richard C M Siow
- King's BHF Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, Faculty of Life Sciences & Medicine, 150 Stamford Street, London, SE1 9NH, UK
| |
Collapse
|
22
|
Abstract
Iron deficiency or overload poses an increasingly complex issue in cardiovascular disease, especially heart failure. The potential benefits and side effects of iron supplementation are still a matter of concern, even though current guidelines suggest therapeutic management of iron deficiency. In this review, we sought to examine the iron metabolism and to identify the rationale behind iron supplementation and iron chelation. Cardiovascular disease is increasingly linked with iron dysmetabolism, with an increased proportion of heart failure patients being affected by decreased plasma iron levels and in turn, by the decreased quality of life. Multiple studies have concluded on a benefit of iron administration, even if just for symptomatic relief. However, new studies field evidence for negative effects of dysregulated non-bound iron and its reactive oxygen species production, with concern to heart diseases. The molecular targets of iron usage, such as the mitochondria, are prone to deleterious effects of the polyvalent metal, added by the scarcely described processes of iron elimination. Iron supplementation and iron chelation show promise of therapeutic benefit in heart failure, with the extent and mechanisms of both prospects not being entirely understood. It may be that a state of decreased systemic and increased mitochondrial iron levels proves to be a useful frame for future advancements in understanding the interconnection of heart failure and iron metabolism.
Collapse
|
23
|
Hu J, Lemasters JJ. Suppression of iron mobilization from lysosomes to mitochondria attenuates liver injury after acetaminophen overdose in vivo in mice: Protection by minocycline. Toxicol Appl Pharmacol 2020; 392:114930. [PMID: 32109512 DOI: 10.1016/j.taap.2020.114930] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 02/19/2020] [Accepted: 02/24/2020] [Indexed: 12/20/2022]
Abstract
Acetaminophen (APAP) overdose causes hepatotoxicity involving mitochondrial dysfunction. Previous studies showed that translocation of Fe2+ from lysosomes into mitochondria by the mitochondrial Ca2+ uniporter (MCU) promotes the mitochondrial permeability transition (MPT) after APAP. Here, our Aim was to assess protection by iron chelation and MCU inhibition against APAP hepatotoxicity in mice. C57BL/6 mice and hepatocytes were administered toxic doses of APAP with and without starch-desferal (an iron chelator), minocycline (MCU inhibitor), or N-acetylcysteine (NAC). In mice, starch-desferal and minocycline pretreatment decreased ALT and liver necrosis after APAP by >60%. At 24 h after APAP, loss of fluorescence of mitochondrial rhodamine 123 occurred in pericentral hepatocytes often accompanied by propidium iodide labeling, indicating mitochondrial depolarization and cell death. Starch-desferal and minocycline pretreatment decreased mitochondrial depolarization and cell death by more than half. In cultured hepatocytes, cell killing at 10 h after APAP decreased from 83% to 49%, 35% and 27%, respectively, by 1 h posttreatment with minocycline, NAC, and minocycline plus NAC. With 4 h posttreatment in vivo, minocycline and minocycline plus NAC decreased ALT and necrosis by ~20% and ~50%, respectively, but NAC alone was not effective. In conclusion, minocycline and starch-desferal decrease mitochondrial dysfunction and severe liver injury after APAP overdose, suggesting that the MPT is likely triggered by iron uptake into mitochondria through MCU. In vivo, minocycline and minocycline plus NAC posttreatment after APAP protect at later time points than NAC alone, indicating that minocycline has a longer window of efficacy than NAC.
Collapse
Affiliation(s)
- Jiangting Hu
- Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, SC 29425, United States of America; Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, United States of America
| | - John J Lemasters
- Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, SC 29425, United States of America; Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, United States of America; Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, United States of America.
| |
Collapse
|
24
|
Lv H, Shang P. The significance, trafficking and determination of labile iron in cytosol, mitochondria and lysosomes. Metallomics 2019; 10:899-916. [PMID: 29923582 DOI: 10.1039/c8mt00048d] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The labile iron pool (LIP) is a pool of chelatable and redox-active iron, not only essential for a wide variety of metabolic process, but also as a catalyst in the Fenton reaction, causing the release of hazardous reactive oxygen species (ROS) with potential for inducing oxidative stress and cell damage. The cellular LIP represents the entirety of every heterogenous sub-pool of iron, not only present in the cytosol, but also in mitochondria, lysosomes and the nucleus, which have all been detected and characterized by various fluorescent methods. Accumulated evidence indicates that alterations in the intracellular LIP can substantially contribute to a variety of injurious processes and initiate pathological development. Herein, we present our understanding of the role of the cellular LIP. To fully review the LIP, firstly, the significance of cellular labile iron in different subcellular compartments is presented. And then, the trafficking processes of cellular labile iron between/in cytosol, mitochondria and lysosomes are discussed in detail. Then, the recent progress in uncovering and assessing the cellular LIP by fluorescent methods have been noted. Overall, this summary may help to comprehensively envision the important physiological and pathological roles of the LIP and shed light on profiling the LIP in a real-time and nondestructive manner with fluorescent methods. Undoubtedly, with the advent and development of iron biology, a better understanding of iron, especially the LIP, may also enhance treatments for iron-related diseases.
Collapse
Affiliation(s)
- Huanhuan Lv
- School of Life Sciences, Northwestern Polytechnical University, Youyi Xilu 127, 7100072, Xi'an, Shaanxi, China
| | | |
Collapse
|
25
|
Creed S, McKenzie M. Measurement of Mitochondrial Membrane Potential with the Fluorescent Dye Tetramethylrhodamine Methyl Ester (TMRM). Methods Mol Biol 2019; 1928:69-76. [PMID: 30725451 DOI: 10.1007/978-1-4939-9027-6_5] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The mitochondrial membrane potential (Δψm) drives the generation of ATP by mitochondria. Interestingly, Δψm is higher in many cancer cells comparted to healthy noncancerous cell types, providing a unique metabolic marker. This feature has also been exploited for therapeutic use by utilizing drugs that specifically accumulate in the mitochondria of cancer cells with high Δψm. As such, the assessment of Δψm can provide very useful information as to the metabolic state of a cancer cell, as well as its potential for malignancy. In addition, the measurement of Δψm can also be used to test the ability of novel anticancer therapies to disrupt mitochondrial metabolism and cause cell death.Here, we outline two methods for assessing Δψm in cancer cells using confocal microscopy and the potentiometric fluorescent dye tetramethylrhodamine methyl ester (TMRM). In the first protocol, we describe a technique to quantitatively measure Δψm, which can be used to compare Δψm between different cell types. In the second protocol, we describe a technique for assessing changes to Δψm over time, which can be used to determine the effectiveness of different therapeutic compounds or drugs in modulating mitochondrial function.
Collapse
Affiliation(s)
- Sarah Creed
- Monash Micro Imaging, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Matthew McKenzie
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia. .,Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia. .,School of Life and Environmental Sciences, Deakin University, Waurn Ponds, VIC, Australia.
| |
Collapse
|
26
|
Li S, Zhao Y, Wang Z, Wang J, Liu C, Sun D. Transplantation of Amniotic Fluid-Derived Stem Cells Preconditioned with Glial Cell Line-Derived Neurotrophic Factor Gene Alleviates Renal Fibrosis. Cell Transplant 2018; 28:65-78. [PMID: 30497277 PMCID: PMC6322139 DOI: 10.1177/0963689718815850] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Amniotic fluid-derived stem cells (AFSCs), which exhibit both embryonic and mesenchymal stem cell characteristics, have been shown to mitigate the degree of renal interstitial fibrosis. The aim of the present study was to determine whether transplantation of glial cell line-derived neurotrophic factor (GDNF)–modified AFSCs is more useful than transplantation of unmodified AFSCs for the treatment of renal interstitial fibrosis. Mice were randomly assigned to a sham-operation group (sham), a unilateral ureteral obstruction (UUO)-saline solution group (UUO), an AFSC transplantation group (AFSC) and a GDNF-modified AFSC transplantation group (GDNF-AFSC) and sacrificed at days 3 and 7 post-surgery (six in each group). We showed that GDNF-AFSCs noticeably suppressed oxidative stress and inflammation; additionally, GDNF-AFSCs positively regulated peritubular capillaries (PTCs), vascular endothelial growth factor (VEGF), hypoxia inducible factor-1α (HIF-1α), and transforming growth factor-β1 (TGF-β1) protein levels. Transmission electron microscopy (TEM) revealed that mitochondrial injury induced by the UUO model was significantly ameliorated after the mice were treated with GDNF-AFSCs. Therefore, we determined that GDNF gene promotes the abilities of AFSCs to inhibit inflammatory and oxidative stress effects, repair renal microvessels, relieve tissue hypoxia and mitochondrial damage, and, ultimately, alleviate renal interstitial fibrosis.
Collapse
Affiliation(s)
- Shulin Li
- 1 Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yuan Zhao
- 1 Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Zhuojun Wang
- 1 Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Jia Wang
- 1 Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Caixia Liu
- 1 Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Dong Sun
- 1 Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,2 Department of Internal Medicine and Diagnostics, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
27
|
Bogacz M, Krauth-Siegel RL. Tryparedoxin peroxidase-deficiency commits trypanosomes to ferroptosis-type cell death. eLife 2018; 7:37503. [PMID: 30047863 PMCID: PMC6117152 DOI: 10.7554/elife.37503] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 07/24/2018] [Indexed: 01/19/2023] Open
Abstract
Tryparedoxin peroxidases, distant relatives of glutathione peroxidase 4 in higher eukaryotes, are responsible for the detoxification of lipid-derived hydroperoxides in African trypanosomes. The lethal phenotype of procyclic Trypanosoma brucei that lack the enzymes fulfils all criteria defining a form of regulated cell death termed ferroptosis. Viability of the parasites is preserved by α-tocopherol, ferrostatin-1, liproxstatin-1 and deferoxamine. Without protecting agent, the cells display, primarily mitochondrial, lipid peroxidation, loss of the mitochondrial membrane potential and ATP depletion. Sensors for mitochondrial oxidants and chelatable iron as well as overexpression of a mitochondrial iron-superoxide dismutase attenuate the cell death. Electron microscopy revealed mitochondrial matrix condensation and enlarged cristae. The peroxidase-deficient parasites are subject to lethal iron-induced lipid peroxidation that probably originates at the inner mitochondrial membrane. Taken together, ferroptosis is an ancient cell death program that can occur at individual subcellular membranes and is counterbalanced by evolutionary distant thiol peroxidases. Plants, animals and fungi all belong to a group of organisms known as eukaryotes. Their cells host a variety of compartments, with each having a specific role. For example, mitochondria are tasked with providing the energy that powers most of the processes that keep the cell alive. Membranes delimit these compartments, as well as the cells themselves. Iron is an element needed for chemical reactions that are essential for the cell to survive. Yet, the byproducts of these reactions can damage – ‘oxidize’ – the lipid molecules that form the cell’s membranes, including the one around mitochondria. Unless enzymes known as peroxidases come to repair the oxidized lipids, the cell dies in a process called ferroptosis. Scientists know that this death mechanism is programmed into the cells of humans and other complex eukaryotes. However, Bogacz and Krauth-Siegel wanted to know if ferroptosis also exists in creatures that appeared early in the evolution of eukaryotes, such as the trypanosome Trypanosoma brucei. This single-cell parasite causes sleeping sickness in humans and a disease called nagana in horses and cattle. Before it infects a mammal, T. brucei goes through an ‘insect stage’ where it lives in the tsetse fly; there, it relies on its mitochondrion to produce energy. Bogacz and Krauth-Siegel now show that if the parasites in the insect stage do not have a specific type of peroxidases, they die within a few hours. In particular, problems in the membranes of the mitochondrion stop the compartment from working properly. These peroxidases-free trypanosomes fare better if they are exposed to molecules that prevent iron from taking part in the reactions that can harm lipids. They also survive more if they are forced to create large amounts of an enzyme that relies on iron to protect the mitochondrion against oxidation. Finally, using drugs that prevent ferroptosis in human cells completely rescues these trypanosomes. Taken together, the results suggest that ferroptosis is an ancient cell death program which exists in T. brucei; and that, in the insect stage of the parasite's life cycle, this process first damages the mitochondrion. This last finding could be particularly relevant because the role of mitochondria in ferroptosis in mammals is highly debated. Yet, most of the research is done in cells that do not rely on this cellular compartment to get their energy. During their life cycle, trypanosomes are either dependent on their mitochondria, or they can find their energy through other sources: this could make them a good organism in which to dissect the precise mechanisms of ferroptosis.
Collapse
Affiliation(s)
- Marta Bogacz
- Biochemie-Zentrum der Universität Heidelberg, Heidelberg, Germany
| | | |
Collapse
|
28
|
Fang S, Yu X, Ding H, Han J, Feng J. Effects of intracellular iron overload on cell death and identification of potent cell death inhibitors. Biochem Biophys Res Commun 2018; 503:297-303. [PMID: 29890135 DOI: 10.1016/j.bbrc.2018.06.019] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 06/07/2018] [Indexed: 02/07/2023]
Abstract
Iron overload causes many diseases, while the underlying etiologies of these diseases are unclear. Cell death processes including apoptosis, necroptosis, cyclophilin D-(CypD)-dependent necrosis and a recently described additional form of regulated cell death called ferroptosis, are dependent on iron or iron-dependent reactive oxygen species (ROS). However, whether the accumulation of intracellular iron itself induces ferroptosis or other forms of cell death is largely elusive. In present study, we study the role of intracellular iron overload itself-induced cell death mechanisms by using ferric ammonium citrate (FAC) and a membrane-permeable Ferric 8-hydroxyquinoline complex (Fe-8HQ) respectively. We show that FAC-induced intracellular iron overload causes ferroptosis. We also identify 3-phosphoinositide-dependent kinase 1 (PDK1) inhibitor GSK2334470 as a potent ferroptosis inhibitor. Whereas, Fe-8HQ-induced intracellular iron overload causes unregulated necrosis, but partially activates PARP-1 dependent parthanatos. Interestingly, we identify many phenolic compounds as potent inhibitors of Fe-8HQ-induced cell death. In conclusion, intracellular iron overload-induced cell death form might be dependent on the intracellular iron accumulation rate, newly identified cell death inhibitors in our study that target ferroptosis and unregulated oxidative cell death represent potential therapeutic strategies against iron overload related diseases.
Collapse
Affiliation(s)
- Shenglin Fang
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Xiaonan Yu
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Haoxuan Ding
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Jianan Han
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Jie Feng
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, China.
| |
Collapse
|
29
|
Brini M, Leanza L, Szabo I. Lipid-Mediated Modulation of Intracellular Ion Channels and Redox State: Physiopathological Implications. Antioxid Redox Signal 2018; 28:949-972. [PMID: 28679281 DOI: 10.1089/ars.2017.7215] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Significance: Ion channels play an important role in the regulation of organelle function within the cell, as proven by increasing evidence pointing to a link between altered function of intracellular ion channels and different pathologies ranging from cancer to neurodegenerative diseases, ischemic damage, and lysosomal storage diseases. Recent Advances: A link between these pathologies and redox state as well as lipid homeostasis and ion channel function is in the focus of current research. Critical Issues: Ion channels are target of modulation by lipids and lipid messengers, although in most cases the mechanistic details have not been clarified yet. Ion channel function importantly impacts production of reactive oxygen species (ROS), especially in the case of mitochondria and lysosomes. ROS, in turn, may modulate the function of intracellular channels triggering thereby a feedback control under physiological conditions. If produced in excess, ROS can be harmful to lipids and may produce oxidized forms of these membrane constituents that ultimately affect ion channel function by triggering a "circulus vitiosus." Future Directions: The present review summarizes our current knowledge about the contribution of intracellular channels to oxidative stress and gives examples of how these channels are modulated by lipids and how this modulation may affect ROS production in ROS-related diseases. Future studies need to address the importance of the regulation of intracellular ion channels and related oxidative stress by lipids in various physiological and pathological contexts. Antioxid. Redox Signal. 28, 949-972.
Collapse
Affiliation(s)
- Marisa Brini
- Department of Biology, University of Padova, Padova, Italy
| | - Luigi Leanza
- Department of Biology, University of Padova, Padova, Italy
| | - Ildiko Szabo
- Department of Biology, University of Padova, Padova, Italy.,CNR Institute of Neuroscience, Padova, Italy
| |
Collapse
|
30
|
DeHart DN, Fang D, Heslop K, Li L, Lemasters JJ, Maldonado EN. Opening of voltage dependent anion channels promotes reactive oxygen species generation, mitochondrial dysfunction and cell death in cancer cells. Biochem Pharmacol 2017; 148:155-162. [PMID: 29289511 DOI: 10.1016/j.bcp.2017.12.022] [Citation(s) in RCA: 140] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 12/26/2017] [Indexed: 12/25/2022]
Abstract
Enhancement of aerobic glycolysis and suppression of mitochondrial metabolism characterize the pro-proliferative Warburg phenotype of cancer cells. High free tubulin in cancer cells closes voltage dependent anion channels (VDAC) to decrease mitochondrial membrane potential (ΔΨ), an effect antagonized by erastin, the canonical promotor of ferroptosis. Previously, we identified six compounds (X1-X6) that also block tubulin-dependent mitochondrial depolarization. Here, we hypothesized that VDAC opening after erastin and X1-X6 increases mitochondrial metabolism and reactive oxygen species (ROS) formation, leading to ROS-dependent mitochondrial dysfunction, bioenergetic failure and cell death. Accordingly, we characterized erastin and the two most potent structurally unrelated lead compounds, X1 and X4, on ROS formation, mitochondrial function and cell viability. Erastin, X1 and X4 increased ΔΨ followed closely by an increase in mitochondrial ROS generation within 30-60 min. Subsequently, mitochondria began to depolarize after an hour or longer indicative of mitochondrial dysfunction. N-acetylcysteine (NAC, glutathione precursor and ROS scavenger) and MitoQ (mitochondrially targeted antioxidant) blocked increased ROS formation after X1 and prevented mitochondrial dysfunction. Erastin, X1 and X4 selectively promoted cell killing in HepG2 and Huh7 human hepatocarcinoma cells compared to primary rat hepatocytes. X1 and X4-dependent cell death was blocked by NAC. These results suggest that ferroptosis induced by erastin and our erastin-like lead compounds was caused by VDAC opening, leading to increased ΔΨ, mitochondrial ROS generation and oxidative stress-induced cell death.
Collapse
Affiliation(s)
- David N DeHart
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - Diana Fang
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - Kareem Heslop
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - Li Li
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - John J Lemasters
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States; Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC, United States; Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, SC, United States; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States; Institute of Theoretical and Experimental Biophysics, Pushchino, Russia.
| | - Eduardo N Maldonado
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States; Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, SC, United States; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States.
| |
Collapse
|
31
|
Hu C, Li L. Pre-conditions for eliminating mitochondrial dysfunction and maintaining liver function after hepatic ischaemia reperfusion. J Cell Mol Med 2017; 21:1719-1731. [PMID: 28301072 PMCID: PMC5571537 DOI: 10.1111/jcmm.13129] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 01/13/2017] [Indexed: 12/16/2022] Open
Abstract
The liver, the largest organ with multiple synthesis and secretion functions in mammals, consists of hepatocytes and Kupffer, stem, endothelial, stellate and other parenchymal cells. Because of early and extensive contact with the external environment, hepatic ischaemia reperfusion (IR) may result in mitochondrial dysfunction, autophagy and apoptosis of cells and tissues under various pathological conditions. Because the liver requires a high oxygen supply to maintain normal detoxification and synthesis functions, it is extremely susceptible to ischaemia and subsequent reperfusion with blood. Consequently, hepatic IR leads to acute or chronic liver failure and significantly increases the total rate of morbidity and mortality through multiple regulatory mechanisms. An increasing number of studies indicate that mitochondrial structure and function are impaired after hepatic IR, but that the health of liver tissues or liver grafts can be effectively rescued by attenuation of mitochondrial dysfunction. In this review, we mainly focus on the subsequent therapeutic interventions related to the conservation of mitochondrial function involved in mitigating hepatic IR injury and the potential mechanisms of protection. Because mitochondria are abundant in liver tissue, clarification of the regulatory mechanisms between mitochondrial dysfunction and hepatic IR should shed light on clinical therapies for alleviating hepatic IR‐induced injury.
Collapse
Affiliation(s)
- Chenxia Hu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
32
|
Sharma N, Reja SI, Gupta N, Bhalla V, Kaur D, Arora S, Kumar M. A highly selective fluorescent probe for Fe3+ in living cells: a stress induced cell based model study. Org Biomol Chem 2017; 15:1006-1012. [DOI: 10.1039/c6ob02209j] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A rhodamine–phenanthroline conjugated fluorescent probe 4 has been designed and synthesized for selective sensing and imaging of endogenous Fe3+ ions in living cells under different stress conditions.
Collapse
Affiliation(s)
- Neetu Sharma
- Department of Chemistry
- UGC Sponsored Centre for Advanced Studies-II
- Guru Nanak Dev University
- Amritsar
- India
| | - Shahi Imam Reja
- Department of Chemistry
- UGC Sponsored Centre for Advanced Studies-II
- Guru Nanak Dev University
- Amritsar
- India
| | - Neha Gupta
- Department of Chemistry
- UGC Sponsored Centre for Advanced Studies-II
- Guru Nanak Dev University
- Amritsar
- India
| | - Vandana Bhalla
- Department of Chemistry
- UGC Sponsored Centre for Advanced Studies-II
- Guru Nanak Dev University
- Amritsar
- India
| | - Davinder Kaur
- Department of Botanical and Environmental Sciences
- Guru Nanak Dev University
- Amritsar
- India
| | - Saroj Arora
- Department of Botanical and Environmental Sciences
- Guru Nanak Dev University
- Amritsar
- India
| | - Manoj Kumar
- Department of Chemistry
- UGC Sponsored Centre for Advanced Studies-II
- Guru Nanak Dev University
- Amritsar
- India
| |
Collapse
|
33
|
The Antiapoptosis Effect of Glycyrrhizate on HepG2 Cells Induced by Hydrogen Peroxide. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:6849758. [PMID: 27891207 PMCID: PMC5116359 DOI: 10.1155/2016/6849758] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Revised: 08/25/2016] [Accepted: 09/28/2016] [Indexed: 01/26/2023]
Abstract
This study demonstrated that glycyrrhizate (GAS) could protect HEPG2 cells against damage and apoptosis induced by H2O2 (1600 μM, 4 h). Cell viability assay revealed that GAS was noncytotoxity at concentration 125 µg/mL, and GAS (5 μg/mL, 25 μg/mL, and 125 μg/mL) protected HepG2 cells against H2O2-induced cytotoxicity. H2O2 induced the HepG2 cells apoptosis, obvious morphologic changes were observed after Hochest 33258 staining, and more apoptotic cells were counted in flow cytometry assay compared to that of the natural group. Pretreatment GAS (5 μg/mL, 25 μg/mL, and 125 μg/mL) prior to H2O2 reverses the morphologic changes and reduced the apoptotic cells in HepG2 cells. GAS reduced the release of MDA, increased the activities of superoxide dismutase, and diminished the release of ALT and AST during oxidative stress in HepG2 cells. After Elisa kit detecting, GAS inhibited the caspase activity induced by H2O2, GAS decreased the level of caspase-3 and caspase-9 from mitochondria in dose-dependent manner. Western blot results showed that pretreatment GAS upregulated the expression of Bcl-2 and decreased the expression of Bax. These results reveal that GAS has the cytoprotection in HepG2 cells during ROS exposure by inhibiting the caspase activity in the mitochondria and influencing apoptogenic factors of the expression of Bax and Bcl-2.
Collapse
|
34
|
Hu J, Kholmukhamedov A, Lindsey CC, Beeson CC, Jaeschke H, Lemasters JJ. Translocation of iron from lysosomes to mitochondria during acetaminophen-induced hepatocellular injury: Protection by starch-desferal and minocycline. Free Radic Biol Med 2016; 97:418-426. [PMID: 27345134 PMCID: PMC4996678 DOI: 10.1016/j.freeradbiomed.2016.06.024] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 06/16/2016] [Accepted: 06/21/2016] [Indexed: 01/09/2023]
Abstract
Acetaminophen (APAP) overdose causes hepatotoxicity involving mitochondrial dysfunction and the mitochondrial permeability transition (MPT). Iron is a critical catalyst for ROS formation, and reactive oxygen species (ROS) play an important role in APAP-induced hepatotoxicity. Previous studies show that APAP disrupts lysosomes, which release ferrous iron (Fe(2+)) into the cytosol to trigger the MPT and cell killing. Here, our aim was to investigate whether iron released from lysosomes after APAP is then taken up into mitochondria via the mitochondrial electrogenic Ca(2+), Fe(2+) uniporter (MCFU) to cause mitochondrial dysfunction and cell death. Hepatocytes were isolated from fasted male C57BL/6 mice. Necrotic cell killing was assessed by propidium iodide fluorimetry. Mitochondrial membrane potential (ΔΨ) was visualized by confocal microscopy of rhodamine 123 (Rh123) and tetramethylrhodamine methylester (TMRM). Chelatable Fe(2+) was monitored by quenching of calcein (cytosol) and mitoferrofluor (MFF, mitochondria). ROS generation was monitored by confocal microscopy of MitoSox Red and plate reader fluorimetry of chloromethyldihydrodichlorofluorescein diacetate (cmH2DCF-DA). Administered 1h before APAP (10mM), the lysosomally targeted iron chelator, starch-desferal (1mM), and the MCFU inhibitors, Ru360 (100nM) and minocycline (4µM), decreased cell killing from 83% to 41%, 57% and 53%, respectively, after 10h. Progressive quenching of calcein and MFF began after ~4h, signifying increased cytosolic and mitochondrial chelatable Fe(2+). Mitochondria then depolarized after ~10h. Dipyridyl, a membrane-permeable iron chelator, dequenched calcein and MFF fluorescence after APAP. Starch-desferal, but not Ru360 and minocycline, suppressed cytosolic calcein quenching, whereas starch-desferal, Ru360 and minocycline all suppressed mitochondrial MFF quenching and mitochondrial depolarization. Starch-desferal, Ru360 and minocycline also each decreased ROS formation. Moreover, minocycline 1h after APAP decreased cell killing by half. In conclusion, release of Fe(2+) from lysosomes followed by uptake into mitochondria via MCFU occurs during APAP hepatotoxicity. Mitochondrial iron then catalyzes toxic hydroxyl radical formation, which triggers the MPT and cell killing. The efficacy of minocycline post-treatment shows minocycline as a possible therapeutic agent against APAP hepatotoxicity.
Collapse
Affiliation(s)
- Jiangting Hu
- Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, SC 29425, United States; Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Andaleb Kholmukhamedov
- Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, SC 29425, United States; Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Christopher C Lindsey
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Craig C Beeson
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, United States
| | - John J Lemasters
- Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, SC 29425, United States; Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, United States; Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, United States; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, United States; Institute of Theoretical & Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region 142290, Russian Federation.
| |
Collapse
|
35
|
Salvianolate Protects Hepatocytes from Oxidative Stress by Attenuating Mitochondrial Injury. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:5408705. [PMID: 27340417 PMCID: PMC4909905 DOI: 10.1155/2016/5408705] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 04/01/2016] [Accepted: 04/11/2016] [Indexed: 11/18/2022]
Abstract
Salvianolate is widely used to treat angiocardiopathy in clinic in China, but its application in liver diseases remains unclear. Our study aims to investigate the effect of Salvianolate on rat hepatic injury by protecting hepatocyte mitochondria. To evaluate the effects of Salvianolate on injured hepatocytes, alpha mouse liver 12 (AML-12) cells were induced with hydrogen peroxide (H2O2) and treated with Salvianolate. Cell viability and MitoTracker Green for mitochondria and 5,5′,6,6′-tetrachloro-1,1′,3,3′-tetraethylbenzimidazole-carbocyanide iodine (JC-1) levels and cytochrome C (Cyto-C) expressions were detected in vitro. To identify the effect of Salvianolate on protecting against mitochondria injury, male Wistar rats were injected with carbon tetrachloride (CCl4) and treated with Salvianolate (40 mg·kg−1). Serum liver function, parameters for peroxidative damage, hematoxylin and eosin (H&E) staining, and transmission electron microscope (TEM) of hepatocyte mitochondria were assayed. Our results showed that Salvianolate effectively protected hepatocytes, increased mitochondria vitality, and decreased Cyto-C expressions in vitro. Besides, Salvianolate alleviated the liver function, attenuated the indicators of peroxidation, and relieved the mitochondria injury in vivo. In conclusion, Salvianolate is effective in protecting hepatocytes from injury in vitro and in vivo, and the mechanism might be related to its protective effect on hepatocyte mitochondria against oxidative stress.
Collapse
|
36
|
Chang HC, Wu R, Shang M, Sato T, Chen C, Shapiro JS, Liu T, Thakur A, Sawicki KT, Prasad SVN, Ardehali H. Reduction in mitochondrial iron alleviates cardiac damage during injury. EMBO Mol Med 2016; 8:247-67. [PMID: 26896449 PMCID: PMC4772952 DOI: 10.15252/emmm.201505748] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 01/18/2016] [Accepted: 01/20/2016] [Indexed: 01/19/2023] Open
Abstract
Excess cellular iron increases reactive oxygen species (ROS) production and causes cellular damage. Mitochondria are the major site of iron metabolism and ROS production; however, few studies have investigated the role of mitochondrial iron in the development of cardiac disorders, such as ischemic heart disease or cardiomyopathy (CM). We observe increased mitochondrial iron in mice after ischemia/reperfusion (I/R) and in human hearts with ischemic CM, and hypothesize that decreasing mitochondrial iron protects against I/R damage and the development of CM. Reducing mitochondrial iron genetically through cardiac-specific overexpression of a mitochondrial iron export protein or pharmacologically using a mitochondria-permeable iron chelator protects mice against I/R injury. Furthermore, decreasing mitochondrial iron protects the murine hearts in a model of spontaneous CM with mitochondrial iron accumulation. Reduced mitochondrial ROS that is independent of alterations in the electron transport chain's ROS producing capacity contributes to the protective effects. Overall, our findings suggest that mitochondrial iron contributes to cardiac ischemic damage, and may be a novel therapeutic target against ischemic heart disease.
Collapse
Affiliation(s)
- Hsiang-Chun Chang
- Feinberg Cardiovascular Research Institute (FCVRI), Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Rongxue Wu
- Feinberg Cardiovascular Research Institute (FCVRI), Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Meng Shang
- Feinberg Cardiovascular Research Institute (FCVRI), Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Tatsuya Sato
- Feinberg Cardiovascular Research Institute (FCVRI), Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Chunlei Chen
- Feinberg Cardiovascular Research Institute (FCVRI), Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jason S Shapiro
- Feinberg Cardiovascular Research Institute (FCVRI), Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ting Liu
- Feinberg Cardiovascular Research Institute (FCVRI), Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Anita Thakur
- Feinberg Cardiovascular Research Institute (FCVRI), Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Konrad T Sawicki
- Feinberg Cardiovascular Research Institute (FCVRI), Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Sathyamangla V N Prasad
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Hossein Ardehali
- Feinberg Cardiovascular Research Institute (FCVRI), Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
37
|
Oxidative Stress in the Healthy and Wounded Hepatocyte: A Cellular Organelles Perspective. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:8327410. [PMID: 26788252 PMCID: PMC4691634 DOI: 10.1155/2016/8327410] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Accepted: 09/10/2015] [Indexed: 02/06/2023]
Abstract
Accurate control of the cell redox state is mandatory for maintaining the structural integrity and physiological functions. This control is achieved both by a fine-tuned balance between prooxidant and anti-oxidant molecules and by spatial and temporal confinement of the oxidative species. The diverse cellular compartments each, although structurally and functionally related, actively maintain their own redox balance, which is necessary to fulfill specialized tasks. Many fundamental cellular processes such as insulin signaling, cell proliferation and differentiation and cell migration and adhesion, rely on localized changes in the redox state of signal transducers, which is mainly mediated by hydrogen peroxide (H2O2). Therefore, oxidative stress can also occur long before direct structural damage to cellular components, by disruption of the redox circuits that regulate the cellular organelles homeostasis. The hepatocyte is a systemic hub integrating the whole body metabolic demand, iron homeostasis and detoxification processes, all of which are redox-regulated processes. Imbalance of the hepatocyte's organelles redox homeostasis underlies virtually any liver disease and is a field of intense research activity. This review recapitulates the evolving concept of oxidative stress in the diverse cellular compartments, highlighting the principle mechanisms of oxidative stress occurring in the healthy and wounded hepatocyte.
Collapse
|
38
|
Dungel P, Perlinger M, Weidinger A, Redl H, Kozlov AV. The cytoprotective effect of nitrite is based on the formation of dinitrosyl iron complexes. Free Radic Biol Med 2015; 89:300-10. [PMID: 26415027 DOI: 10.1016/j.freeradbiomed.2015.08.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 08/26/2015] [Accepted: 08/28/2015] [Indexed: 10/23/2022]
Abstract
Nitrite protects various organs from ischemia-reperfusion injury by ameliorating mitochondrial dysfunction. Here we provide evidence that this protection is due to the inhibition of iron-mediated oxidative reactions caused by the release of iron ions upon hypoxia. We show in a model of isolated rat liver mitochondria that upon hypoxia, mitochondria reduce nitrite to nitric oxide (NO) in amounts sufficient to inactivate redox-active iron ions by formation of inactive dinitrosyl iron complexes (DNIC). The scavenging of iron ions in turn prevents the oxidative modification of the outer mitochondrial membrane and the release of cytochrome c during reoxygenation. This action of nitrite protects mitochondrial function. The formation of DNIC with nitrite-derived NO could also be confirmed in an ischemia-reperfusion model in liver tissue. Our data suggest that the formation of DNIC is a key mechanism of nitrite-mediated cytoprotection.
Collapse
Affiliation(s)
- Peter Dungel
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Austrian Cluster for Tissue Regeneration, A-1200 Vienna, Austria
| | - Martin Perlinger
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Austrian Cluster for Tissue Regeneration, A-1200 Vienna, Austria
| | - Adelheid Weidinger
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Austrian Cluster for Tissue Regeneration, A-1200 Vienna, Austria
| | - Heinz Redl
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Austrian Cluster for Tissue Regeneration, A-1200 Vienna, Austria
| | - Andrey V Kozlov
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Austrian Cluster for Tissue Regeneration, A-1200 Vienna, Austria.
| |
Collapse
|
39
|
Schaffroth C, Bogacz M, Dirdjaja N, Nißen A, Krauth-Siegel RL. The cytosolic or the mitochondrial glutathione peroxidase-type tryparedoxin peroxidase is sufficient to protect procyclic Trypanosoma brucei from iron-mediated mitochondrial damage and lysis. Mol Microbiol 2015; 99:172-87. [PMID: 26374473 DOI: 10.1111/mmi.13223] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/14/2015] [Indexed: 01/15/2023]
Abstract
African trypanosomes express three virtually identical glutathione peroxidase (Px)-type enzymes that occur in the cytosol (Px I and II) and mitochondrion (Px III) and detoxify fatty acid-derived hydroperoxides. Selective deletion of the genes revealed that procyclic Trypanosoma brucei lacking either the cytosolic or mitochondrial enzyme proliferate nearly as wild-type parasites, whereas the knockout of the complete genomic locus is lethal. Flow cytometry and immunofluorescence analyses revealed that the Px I-III-deficient parasites lose their mitochondrial membrane potential, which is followed by a loss of the lysosomal signal but not the glycosomal one. Mitochondrial damage and cell lysis are prevented by Trolox, ubiquinone derivatives and the iron chelator deferoxamine, whereas starch-deferoxamine is inefficient. In glucose-rich medium, cell death is attenuated suggesting that oxidants generated by the respiratory chain contribute to the lethal phenotype. Thus, the Px-type peroxidases protect procyclic cells from an iron-mediated oxidative membrane damage that originates at the mitochondrion. This contrasts with the situation in bloodstream cells, where the lysosome is the primarily affected organelle. Strikingly, either the cytosolic or the mitochondrial form of the peroxidases is required and sufficient to protect the mitochondrion and prevent cell lysis.
Collapse
Affiliation(s)
- Corinna Schaffroth
- Biochemie-Zentrum der Universität Heidelberg (BZH), Im Neuenheimer Feld 328, 69120, Heidelberg, Germany
| | - Marta Bogacz
- Biochemie-Zentrum der Universität Heidelberg (BZH), Im Neuenheimer Feld 328, 69120, Heidelberg, Germany
| | - Natalie Dirdjaja
- Biochemie-Zentrum der Universität Heidelberg (BZH), Im Neuenheimer Feld 328, 69120, Heidelberg, Germany
| | - Amrei Nißen
- Biochemie-Zentrum der Universität Heidelberg (BZH), Im Neuenheimer Feld 328, 69120, Heidelberg, Germany
| | - R Luise Krauth-Siegel
- Biochemie-Zentrum der Universität Heidelberg (BZH), Im Neuenheimer Feld 328, 69120, Heidelberg, Germany
| |
Collapse
|
40
|
Minocycline and doxycycline, but not tetracycline, mitigate liver and kidney injury after hemorrhagic shock/resuscitation. Shock 2015; 42:256-63. [PMID: 24978888 DOI: 10.1097/shk.0000000000000213] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Despite recovery of hemodynamics by fluid resuscitation after hemorrhage, development of the systemic inflammatory response and multiple organ dysfunction syndromes can nonetheless lead to death. Minocycline and doxycycline are tetracycline derivatives that are protective in models of hypoxic, ischemic, and oxidative stress. Our aim was to determine whether minocycline and doxycycline protect liver and kidney and improve survival in a mouse model of hemorrhagic shock and resuscitation. METHODS Mice were hemorrhaged to 30 mmHg for 3 h and then resuscitated with shed blood followed by half the shed volume of lactated Ringer's solution containing tetracycline (10 mg/kg), minocycline (10 mg/kg), doxycycline (5 mg/kg), or vehicle. For pretreatment plus posttreatment, drugs were administered intraperitoneally prior to hemorrhage followed by second equal dose in Ringer's solution after blood resuscitation. Blood and tissue were harvested after 6 h. RESULTS Serum alanine aminotransferase (ALT) increased to 1,988 and 1,878 U/L after posttreatment with vehicle and tetracycline, respectively, whereas minocycline and doxycycline posttreatment decreased ALT to 857 and 863 U/L. Pretreatment plus posttreatment with minocycline and doxycycline also decreased ALT to 849 and 834 U/L. After vehicle, blood creatinine increased to 134 µM, which minocycline and doxycycline posttreatment decreased to 59 and 56 µM. Minocycline and doxycycline pretreatment plus posttreatment decreased creatinine similarly. Minocycline and doxycycline also decreased necrosis and apoptosis in liver and apoptosis in both liver and kidney, the latter assessed by TUNEL (terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling) and caspase 3 activation. Lastly after 4.5 h of hemorrhage followed by resuscitation, minocycline and doxycycline (but not tetracycline) posttreatment improved 1-week survival from 38% (vehicle) to 69% and 67%, respectively. CONCLUSION Minocycline and doxycycline were similarly protective when given before as after blood resuscitation and might therefore have clinical efficacy to mitigate liver and kidney injury after resuscitated hemorrhage.
Collapse
|
41
|
Abstract
The role of iron in the development of cancer remains unclear. We previously reported that iron reduces cell survival in a Ras/mitogen-activated protein kinase (MAPK)-dependent manner in ovarian cells; however, the underlying downstream pathway leading to reduced survival was unclear. Although levels of intracellular iron, ferritin/CD71 protein and reactive oxygen species did not correlate with iron-induced cell survival changes, we identified mitochondrial damage (via TEM) and reduced expression of outer mitochondrial membrane proteins (translocase of outer membrane: TOM20 and TOM70) in cell lines sensitive to iron. Interestingly, Ru360 (an inhibitor of the mitochondrial calcium uniporter) reversed mitochondrial changes and restored cell survival in HEY ovarian carcinoma cells treated with iron. Further, cells treated with Ru360 and iron also had reduced autophagic punctae with increased lysosomal numbers, implying cross-talk between these compartments. Mitochondrial changes were dependent on activation of the Ras/MAPK pathway since treatment with a MAPK inhibitor restored expression of TOM20/TOM70 proteins. Although glutathione antioxidant levels were reduced in HEY treated with iron, extracellular glutamate levels were unaltered. Strikingly, oxalomalate (inhibitor of aconitase, involved in glutamate production) reversed iron-induced responses in a similar manner to Ru360. Collectively, our results implicate iron in modulating cell survival in a mitochondria-dependent manner in ovarian cancer cells.
Collapse
|
42
|
Grossini E, Bellofatto K, Farruggio S, Sigaudo L, Marotta P, Raina G, De Giuli V, Mary D, Pollesello P, Minisini R, Pirisi M, Vacca G. Levosimendan inhibits peroxidation in hepatocytes by modulating apoptosis/autophagy interplay. PLoS One 2015; 10:e0124742. [PMID: 25880552 PMCID: PMC4400069 DOI: 10.1371/journal.pone.0124742] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 03/05/2015] [Indexed: 12/21/2022] Open
Abstract
Background Levosimendan protects rat liver against peroxidative injuries through mechanisms related to nitric oxide (NO) production and mitochondrial ATP-dependent K (mitoKATP) channels opening. However, whether levosimendan could modulate the cross-talk between apoptosis and autophagy in the liver is still a matter of debate. Thus, the aim of this study was to examine the role of levosimendan as a modulator of the apoptosis/autophagy interplay in liver cells subjected to peroxidation and the related involvement of NO and mitoKATP. Methods and Findings In primary rat hepatocytes that have been subjected to oxidative stress, Western blot was performed to examine endothelial and inducible NO synthase isoforms (eNOS, iNOS) activation, apoptosis/autophagy and survival signalling detection in response to levosimendan. In addition, NO release, cell viability, mitochondrial membrane potential and mitochondrial permeability transition pore opening (MPTP) were examined through specific dyes. Some of those evaluations were also performed in human hepatic stellate cells (HSC). Pre-treatment of hepatocytes with levosimendan dose-dependently counteracted the injuries caused by oxidative stress and reduced NO release by modulating eNOS/iNOS activation. In hepatocytes, while the autophagic inhibition reduced the effects of levosimendan, after the pan-caspases inhibition, cell survival and autophagy in response to levosimendan were increased. Finally, all protective effects were prevented by both mitoKATP channels inhibition and NOS blocking. In HSC, levosimendan was able to modulate the oxidative balance and inhibit autophagy without improving cell viability and apoptosis. Conclusions Levosimendan protects hepatocytes against oxidative injuries by autophagic-dependent inhibition of apoptosis and the activation of survival signalling. Such effects would involve mitoKATP channels opening and the modulation of NO release by the different NOS isoforms. In HSC, levosimendan would also play a role in cell activation and possible evolution toward fibrosis. These findings highlight the potential of levosimendan as a therapeutic agent for the treatment or prevention of liver ischemia/reperfusion injuries.
Collapse
Affiliation(s)
- Elena Grossini
- Laboratory of Physiology and Experimental Surgery, Department of Translational Medicine, University Eastern Piedmont “Amedeo Avogadro”, Via Solaroli 17, Azienda Ospedaliera Universitaria Maggiore della Carità, corso Mazzini 36, Novara, Italy
- * E-mail:
| | - Kevin Bellofatto
- Laboratory of Physiology and Experimental Surgery, Department of Translational Medicine, University Eastern Piedmont “Amedeo Avogadro”, Via Solaroli 17, Azienda Ospedaliera Universitaria Maggiore della Carità, corso Mazzini 36, Novara, Italy
| | - Serena Farruggio
- Laboratory of Physiology and Experimental Surgery, Department of Translational Medicine, University Eastern Piedmont “Amedeo Avogadro”, Via Solaroli 17, Azienda Ospedaliera Universitaria Maggiore della Carità, corso Mazzini 36, Novara, Italy
| | - Lorenzo Sigaudo
- Laboratory of Physiology and Experimental Surgery, Department of Translational Medicine, University Eastern Piedmont “Amedeo Avogadro”, Via Solaroli 17, Azienda Ospedaliera Universitaria Maggiore della Carità, corso Mazzini 36, Novara, Italy
| | - Patrizia Marotta
- Laboratory of Physiology and Experimental Surgery, Department of Translational Medicine, University Eastern Piedmont “Amedeo Avogadro”, Via Solaroli 17, Azienda Ospedaliera Universitaria Maggiore della Carità, corso Mazzini 36, Novara, Italy
| | - Giulia Raina
- Laboratory of Physiology and Experimental Surgery, Department of Translational Medicine, University Eastern Piedmont “Amedeo Avogadro”, Via Solaroli 17, Azienda Ospedaliera Universitaria Maggiore della Carità, corso Mazzini 36, Novara, Italy
| | - Veronica De Giuli
- Laboratory of Physiology and Experimental Surgery, Department of Translational Medicine, University Eastern Piedmont “Amedeo Avogadro”, Via Solaroli 17, Azienda Ospedaliera Universitaria Maggiore della Carità, corso Mazzini 36, Novara, Italy
| | - David Mary
- Laboratory of Physiology and Experimental Surgery, Department of Translational Medicine, University Eastern Piedmont “Amedeo Avogadro”, Via Solaroli 17, Azienda Ospedaliera Universitaria Maggiore della Carità, corso Mazzini 36, Novara, Italy
| | - Piero Pollesello
- Laboratory of Physiology and Experimental Surgery, Department of Translational Medicine, University Eastern Piedmont “Amedeo Avogadro”, Via Solaroli 17, Azienda Ospedaliera Universitaria Maggiore della Carità, corso Mazzini 36, Novara, Italy
| | - Rosalba Minisini
- Internal Medicine, Department of Translational Medicine, University Eastern Piedmont “Amedeo Avogadro”, Via Solaroli 17, Azienda Ospedaliera Universitaria Maggiore della Carità, corso Mazzini 36, Novara, Italy
| | - Mario Pirisi
- Internal Medicine, Department of Translational Medicine, University Eastern Piedmont “Amedeo Avogadro”, Via Solaroli 17, Azienda Ospedaliera Universitaria Maggiore della Carità, corso Mazzini 36, Novara, Italy
| | - Giovanni Vacca
- Laboratory of Physiology and Experimental Surgery, Department of Translational Medicine, University Eastern Piedmont “Amedeo Avogadro”, Via Solaroli 17, Azienda Ospedaliera Universitaria Maggiore della Carità, corso Mazzini 36, Novara, Italy
| |
Collapse
|
43
|
Haldar S, Tripathi A, Qian J, Beserra A, Suda S, McElwee M, Turner J, Hopfer U, Singh N. Prion protein promotes kidney iron uptake via its ferrireductase activity. J Biol Chem 2015; 290:5512-22. [PMID: 25572394 DOI: 10.1074/jbc.m114.607507] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Brain iron-dyshomeostasis is an important cause of neurotoxicity in prion disorders, a group of neurodegenerative conditions associated with the conversion of prion protein (PrP(C)) from its normal conformation to an aggregated, PrP-scrapie (PrP(Sc)) isoform. Alteration of iron homeostasis is believed to result from impaired function of PrP(C) in neuronal iron uptake via its ferrireductase activity. However, unequivocal evidence supporting the ferrireductase activity of PrP(C) is lacking. Kidney provides a relevant model for this evaluation because PrP(C) is expressed in the kidney, and ∼370 μg of iron are reabsorbed daily from the glomerular filtrate by kidney proximal tubule cells (PT), requiring ferrireductase activity. Here, we report that PrP(C) promotes the uptake of transferrin (Tf) and non-Tf-bound iron (NTBI) by the kidney in vivo and mainly NTBI by PT cells in vitro. Thus, uptake of (59)Fe administered by gastric gavage, intravenously, or intraperitoneally was significantly lower in PrP-knock-out (PrP(-/-)) mouse kidney relative to PrP(+/+) controls. Selective in vivo radiolabeling of plasma NTBI with (59)Fe revealed similar results. Expression of exogenous PrP(C) in immortalized PT cells showed localization on the plasma membrane and intracellular vesicles and increased transepithelial transport of (59)Fe-NTBI and to a smaller extent (59)Fe-Tf from the apical to the basolateral domain. Notably, the ferrireductase-deficient mutant of PrP (PrP(Δ51-89)) lacked this activity. Furthermore, excess NTBI and hemin caused aggregation of PrP(C) to a detergent-insoluble form, limiting iron uptake. Together, these observations suggest that PrP(C) promotes retrieval of iron from the glomerular filtrate via its ferrireductase activity and modulates kidney iron metabolism.
Collapse
Affiliation(s)
| | | | - Juan Qian
- From the Departments of Pathology and
| | | | | | | | - Jerrold Turner
- the Department of Pathology, University of Chicago, Chicago, Illinois 60637
| | - Ulrich Hopfer
- Physiology and Biophysics, Case Western Reserve University/University Hospitals Case Medical Center, Cleveland, Ohio 44106 and
| | | |
Collapse
|
44
|
Molecular responses to ischemia and reperfusion in the liver. Arch Toxicol 2015; 89:651-7. [PMID: 25566829 DOI: 10.1007/s00204-014-1437-x] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 12/09/2014] [Indexed: 01/03/2023]
Abstract
Ischemia/reperfusion (IR) injury occurs when oxygen is rapidly reintroduced into ischemic tissue, resulting in cell death and necrotic tissue damage. This is a major concern during liver transplantation procedures since there is an inevitable interruption and subsequent restoration of circulation. IR injury in liver tissue is initiated through reactive oxygen species (ROS), which are generated by hepatocytes during IR insult. Although these ROS are thought to play a protective roll since they are known to activate several pathways involved in the hypoxic response, they also trigger a localized sterile immune response that results in the recruitment of Kupffer cells and neutrophils to the site of IR insult. These immune cells generate larger quantities of ROS that trigger apoptosis and oncotic necrosis in liver tissue. In this review, we will summarize what is currently known about the response of liver tissue to IR insult at the molecular level.
Collapse
|
45
|
Grubman A, White AR, Liddell JR. Mitochondrial metals as a potential therapeutic target in neurodegeneration. Br J Pharmacol 2014; 171:2159-73. [PMID: 24206195 DOI: 10.1111/bph.12513] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2013] [Revised: 10/29/2013] [Accepted: 10/30/2013] [Indexed: 12/22/2022] Open
Abstract
Transition metals are critical for enzyme function and protein folding, but in excess can mediate neurotoxic oxidative processes. As mitochondria are particularly vulnerable to oxidative damage due to radicals generated during ATP production, mitochondrial biometal homeostasis must therefore be tightly controlled to safely harness the redox potential of metal enzyme cofactors. Dysregulation of metal functions is evident in numerous neurological disorders including Alzheimer's disease, stroke, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis and Friedrich's ataxia. This review describes the mitochondrial metal defects in these disorders and highlights novel metal-based therapeutic approaches that target mitochondrial metal homeostasis in neurological disorders.
Collapse
Affiliation(s)
- A Grubman
- Department of Pathology, University of Melbourne, Melbourne, Vic., Australia
| | | | | |
Collapse
|
46
|
Maldonado EN, Lemasters JJ. ATP/ADP ratio, the missed connection between mitochondria and the Warburg effect. Mitochondrion 2014; 19 Pt A:78-84. [PMID: 25229666 DOI: 10.1016/j.mito.2014.09.002] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 09/08/2014] [Accepted: 09/08/2014] [Indexed: 02/06/2023]
Abstract
Non-proliferating cells generate the bulk of cellular ATP by fully oxidizing respiratory substrates in mitochondria. Respiratory substrates cross the mitochondrial outer membrane through only one channel, the voltage dependent anion channel (VDAC). Once in the matrix, respiratory substrates are oxidized in the tricarboxylic acid cycle to generate mostly NADH that is further oxidized in the respiratory chain to generate a proton motive force comprised mainly of membrane potential (ΔΨ) to synthesize ATP. Mitochondrial ΔΨ then drives the release of ATP(4-) from the matrix in exchange for ADP(3-) in the cytosol via the adenine nucleotide translocator (ANT) located in the mitochondrial inner membrane. Thus, mitochondrial function in non-proliferating cells drives a high cytosolic ATP/ADP ratio, essential to inhibit glycolysis. By contrast, the bioenergetics of the Warburg phenotype of proliferating cells is characterized by enhanced aerobic glycolysis and the suppression of mitochondrial metabolism. Suppressed mitochondrial function leads to lower production of mitochondrial ATP and hence lower cytosolic ATP/ADP ratios that favor enhanced glycolysis. Thus, the cytosolic ATP/ADP ratio is a key feature that determines if cell metabolism is predominantly oxidative or glycolytic. Here, we describe two novel mechanisms to explain the suppression of mitochondrial metabolism in cancer cells: the relative closure of VDAC by free tubulin and the inactivation of ANT. Both mechanisms contribute to low ATP/ADP ratios that activate glycolysis.
Collapse
Affiliation(s)
- Eduardo N Maldonado
- Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, SC 29425, United States; Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, United States; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, United States
| | - John J Lemasters
- Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, SC 29425, United States; Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, United States; Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, United States; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, United States; Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Russia.
| |
Collapse
|
47
|
Li Q, Li C, Mahtani HK, Du J, Patel AR, Lancaster JR. Nitrosothiol formation and protection against Fenton chemistry by nitric oxide-induced dinitrosyliron complex formation from anoxia-initiated cellular chelatable iron increase. J Biol Chem 2014; 289:19917-27. [PMID: 24891512 DOI: 10.1074/jbc.m114.569764] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Dinitrosyliron complexes (DNIC) have been found in a variety of pathological settings associated with (•)NO. However, the iron source of cellular DNIC is unknown. Previous studies on this question using prolonged (•)NO exposure could be misleading due to the movement of intracellular iron among different sources. We here report that brief (•)NO exposure results in only barely detectable DNIC, but levels increase dramatically after 1-2 h of anoxia. This increase is similar quantitatively and temporally with increases in the chelatable iron, and brief (•)NO treatment prevents detection of this anoxia-induced increased chelatable iron by deferoxamine. DNIC formation is so rapid that it is limited by the availability of (•)NO and chelatable iron. We utilize this ability to selectively manipulate cellular chelatable iron levels and provide evidence for two cellular functions of endogenous DNIC formation, protection against anoxia-induced reactive oxygen chemistry from the Fenton reaction and formation by transnitrosation of protein nitrosothiols (RSNO). The levels of RSNO under these high chelatable iron levels are comparable with DNIC levels and suggest that under these conditions, both DNIC and RSNO are the most abundant cellular adducts of (•)NO.
Collapse
Affiliation(s)
- Qian Li
- From the Department of Anesthesiology and Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama 35294,
| | - Chuanyu Li
- From the Department of Anesthesiology and Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Harry K Mahtani
- From the Department of Anesthesiology and Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Jian Du
- the Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China, and
| | - Aashka R Patel
- Vestavia Hills High School, Vestavia Hills, Alabama 35216
| | - Jack R Lancaster
- From the Department of Anesthesiology and Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama 35294
| |
Collapse
|
48
|
Urrutia PJ, Mena NP, Núñez MT. The interplay between iron accumulation, mitochondrial dysfunction, and inflammation during the execution step of neurodegenerative disorders. Front Pharmacol 2014; 5:38. [PMID: 24653700 PMCID: PMC3948003 DOI: 10.3389/fphar.2014.00038] [Citation(s) in RCA: 174] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 02/19/2014] [Indexed: 12/21/2022] Open
Abstract
A growing set of observations points to mitochondrial dysfunction, iron accumulation, oxidative damage and chronic inflammation as common pathognomonic signs of a number of neurodegenerative diseases that includes Alzheimer’s disease, Huntington disease, amyotrophic lateral sclerosis, Friedrich’s ataxia and Parkinson’s disease. Particularly relevant for neurodegenerative processes is the relationship between mitochondria and iron. The mitochondrion upholds the synthesis of iron–sulfur clusters and heme, the most abundant iron-containing prosthetic groups in a large variety of proteins, so a fraction of incoming iron must go through this organelle before reaching its final destination. In turn, the mitochondrial respiratory chain is the source of reactive oxygen species (ROS) derived from leaks in the electron transport chain. The co-existence of both iron and ROS in the secluded space of the mitochondrion makes this organelle particularly prone to hydroxyl radical-mediated damage. In addition, a connection between the loss of iron homeostasis and inflammation is starting to emerge; thus, inflammatory cytokines like TNF-alpha and IL-6 induce the synthesis of the divalent metal transporter 1 and promote iron accumulation in neurons and microglia. Here, we review the recent literature on mitochondrial iron homeostasis and the role of inflammation on mitochondria dysfunction and iron accumulation on the neurodegenerative process that lead to cell death in Parkinson’s disease. We also put forward the hypothesis that mitochondrial dysfunction, iron accumulation and inflammation are part of a synergistic self-feeding cycle that ends in apoptotic cell death, once the antioxidant cellular defense systems are finally overwhelmed.
Collapse
Affiliation(s)
- Pamela J Urrutia
- Department of Biology and Research Ring on Oxidative Stress in the Nervous System, Faculty of Sciences, University of Chile Santiago, Chile
| | - Natalia P Mena
- Department of Biology and Research Ring on Oxidative Stress in the Nervous System, Faculty of Sciences, University of Chile Santiago, Chile
| | - Marco T Núñez
- Department of Biology and Research Ring on Oxidative Stress in the Nervous System, Faculty of Sciences, University of Chile Santiago, Chile
| |
Collapse
|
49
|
The role of iron and reactive oxygen species in cell death. Nat Chem Biol 2014; 10:9-17. [PMID: 24346035 DOI: 10.1038/nchembio.1416] [Citation(s) in RCA: 1587] [Impact Index Per Article: 144.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 11/19/2013] [Indexed: 02/06/2023]
Abstract
The transition metal iron is essential for life, yet potentially toxic iron-catalyzed reactive oxygen species (ROS) are unavoidable in an oxygen-rich environment. Iron and ROS are increasingly recognized as important initiators and mediators of cell death in a variety of organisms and pathological situations. Here, we review recent discoveries regarding the mechanism by which iron and ROS participate in cell death. We describe the different roles of iron in triggering cell death, targets of iron-dependent ROS that mediate cell death and a new form of iron-dependent cell death termed ferroptosis. Recent advances in understanding the role of iron and ROS in cell death offer unexpected surprises and suggest new therapeutic avenues to treat cancer, organ damage and degenerative disease.
Collapse
|
50
|
Shulga N, Pastorino JG. Mitoneet mediates TNFα-induced necroptosis promoted by exposure to fructose and ethanol. J Cell Sci 2013; 127:896-907. [PMID: 24357718 DOI: 10.1242/jcs.140764] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Fructose and ethanol are metabolized principally in the liver and are both known to contribute to the development of hepatic steatosis that can progress to hepatic steatohepatitis. The present study indentifies a synergistic interaction between fructose and ethanol in promoting hepatocyte sensitivity to TNFα-induced necroptosis. Concurrent exposure to fructose and ethanol induces the overexpression of the CDGSH iron-sulfur domain-containing protein 1 (CISD1 or mitoneet), which is localized to the outer mitochondrial membrane. The increased expression of mitoneet primes the hepatocyte for TNFα-induced cytotoxicity. Treatment with TNFα induces the translocation of a Stat3-Grim-19 complex to the mitochondria, which binds to mitoneet and promotes the rapid release of its 2Fe-2S cluster, causing an accumulation of mitochondrial iron. The dramatic increase of mitochondrial iron provokes a surge in formation of reactive oxygen species, resulting in mitochondrial injury and cell death. Additionally, mitoneet is constitutively expressed at high levels in L929 fibrosarcoma cells and is required for L929 cells to undergo TNFα-induced necroptosis in the presence of caspase inhibition, indicating the importance of mitoneet to the necroptotic form of cell death.
Collapse
Affiliation(s)
- Nataly Shulga
- Department of Molecular Biology, Rowan University School of Osteopathic Medicine, Stratford, NJ 08084, USA
| | | |
Collapse
|