1
|
Friedman J, Ghio M, Cotton-Betteridge A, Karim M, Packer J, Engelhardt D, Abdullah S, Shaheen F, Taghavi S, Duchesne J, Jackson-Weaver O. Infrarenal Aortic Occlusion Causes Endothelial Injury via Mitochondrial Reactive Oxygen Species. J Surg Res 2025; 309:139-145. [PMID: 40253934 DOI: 10.1016/j.jss.2025.03.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 02/14/2025] [Accepted: 03/22/2025] [Indexed: 04/22/2025]
Abstract
INTRODUCTION The endothelial glycocalyx is an anti-inflammatory, anti-coagulant coating of endothelial cells which participates in many physiologic processes. Damage to the glycocalyx occurs secondary to several disease states, and is especially pronounced in ischemia-reperfusion (I/R) injury. In models of trauma and hemorrhage, this damage has been shown to occur secondary to disordered succinate metabolism leading to increased mitochondrial reactive oxygen species (mitoROS). Aortic occlusion has also been associated with I/R injury to the vascular endothelium, though the precise mechanisms by which this occurs, and whether that injury includes glycocalyx shedding, are largely unknown. Consequently, very few therapeutic strategies for protection of the glycocalyx have been investigated. The aim of this study was to determine whether an experimental model of infrarenal aortic occlusion, with or without hemorrhage, causes a mitochondrial ROS-dependent glycocalyx shedding, and at what point this shedding predominantly occurs. We additionally aimed to determine whether plasma succinate was elevated following aortic occlusion. MATERIALS AND METHODS Male Sprague Dawley rats were anesthetized and laparotomy performed. A jugular catheter was placed and a baseline blood sample collected. In the hemorrhage group, 3 mL of blood was drawn from the jugular catheter. The infrarenal abdominal aorta was isolated and clamped with an atraumatic vascular clamp. Thirty minutes after clamping a second blood sample was collected, followed by unclamping. Blood samples were collected 2 and 15 min after reperfusion. Blood was centrifuged, and succinate and glycocalyx-component syndecan-1 were measured in the plasma using an ELISA. All rats in the treatment arm received identical hemorrhage, clamping, and blood sampling, but were treated with mitochondrial-targeted ROS scavenger mitoTEMPOL prior to clamping the aorta. After euthanizing the rats, calf muscle of the hemorrhage group was flash frozen, sectioned, and stained for glycocalyx with wheat germ agglutinin. RESULTS Plasma syndecan-1 levels were significantly (P < 0.05) elevated 2 mins after unclamping the aorta compared to baseline in both groups. Levels remained increased, though not statistically significantly, at 15 min after reperfusion. No significant increase in syndecan-1 was seen after 30 min of ischemia. MitoTEMPOL treatment prevented an increase in plasma syndecan-1. Correspondingly, glycocalyx staining intensity in mitoTEMPOL-treated rats was increased compared to control. Plasma succinate was significantly elevated at 15 min after reperfusion and was not affected by treatment with mitoTEMPOL. CONCLUSIONS To our knowledge this is the first study which demonstrates a successful therapeutic strategy in the treatment of endothelial glycocalyx shedding caused by aortic occlusion and hemorrhage in an animal model. Our study shows that glycocalyx shedding is attributable to mitochondrial ROS, and occurs following distal reperfusion rather than occlusion of the aorta or during hemorrhage. Consistent with findings in hemorrhagic shock, plasma succinate levels are elevated as a result of aortic occlusion and are upstream of ROS. Taken together, our findings suggest that in the setting of aortic occlusion, a therapeutic intervention could be performed prior to reperfusion which would substantially reduce injury to the vascular endothelium.
Collapse
Affiliation(s)
- Jessica Friedman
- Department of Surgery, Tulane University School of Medicine, New Orleans, Louisiana; Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Michael Ghio
- Department of Surgery, Tulane University School of Medicine, New Orleans, Louisiana
| | | | - Mardeen Karim
- Tulane University School of Medicine, New Orleans, Louisiana
| | - Jacob Packer
- Tulane University School of Medicine, New Orleans, Louisiana
| | - David Engelhardt
- Department of Surgery, Tulane University School of Medicine, New Orleans, Louisiana
| | - Sarah Abdullah
- Department of Surgery, Tulane University School of Medicine, New Orleans, Louisiana
| | - Farhana Shaheen
- Department of Surgery, Tulane University School of Medicine, New Orleans, Louisiana
| | - Sharven Taghavi
- Department of Surgery, Tulane University School of Medicine, New Orleans, Louisiana
| | - Juan Duchesne
- Department of Surgery, Tulane University School of Medicine, New Orleans, Louisiana
| | - Olan Jackson-Weaver
- Department of Surgery, Tulane University School of Medicine, New Orleans, Louisiana.
| |
Collapse
|
2
|
Seyfried TN, Lee DC, Duraj T, Ta NL, Mukherjee P, Kiebish M, Arismendi-Morillo G, Chinopoulos C. The Warburg hypothesis and the emergence of the mitochondrial metabolic theory of cancer. J Bioenerg Biomembr 2025:10.1007/s10863-025-10059-w. [PMID: 40199815 DOI: 10.1007/s10863-025-10059-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/20/2025] [Indexed: 04/10/2025]
Abstract
Otto Warburg originally proposed that cancer arose from a two-step process. The first step involved a chronic insufficiency of mitochondrial oxidative phosphorylation (OxPhos), while the second step involved a protracted compensatory energy synthesis through lactic acid fermentation. His extensive findings showed that oxygen consumption was lower while lactate production was higher in cancerous tissues than in non-cancerous tissues. Warburg considered both oxygen consumption and extracellular lactate as accurate markers for ATP production through OxPhos and glycolysis, respectively. Warburg's hypothesis was challenged from findings showing that oxygen consumption remained high in some cancer cells despite the elevated production of lactate suggesting that OxPhos was largely unimpaired. New information indicates that neither oxygen consumption nor lactate production are accurate surrogates for quantification of ATP production in cancer cells. Warburg also did not know that a significant amount of ATP could come from glutamine-driven mitochondrial substrate level phosphorylation in the glutaminolysis pathway with succinate produced as end product, thus confounding the linkage of oxygen consumption to the origin of ATP production within mitochondria. Moreover, new information shows that cytoplasmic lipid droplets and elevated aerobic lactic acid fermentation are both biomarkers for OxPhos insufficiency. Warburg's original hypothesis can now be linked to a more complete understanding of how OxPhos insufficiency underlies dysregulated cancer cell growth. These findings can also address several questionable assumptions regarding the origin of cancer thus allowing the field to advance with more effective therapeutic strategies for a less toxic metabolic management and prevention of cancer.
Collapse
Affiliation(s)
- Thomas N Seyfried
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA.
| | - Derek C Lee
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA
| | - Tomas Duraj
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA
| | - Nathan L Ta
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA
| | - Purna Mukherjee
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA
| | | | - Gabriel Arismendi-Morillo
- Facultad de Medicina, Instituto de Investigaciones Biológicas, Universidad del Zulia, Maracaibo, Venezuela
- Department of Medicine, Faculty of Health Sciences, University of Deusto, Bilbao (Bizkaia), Spain
| | - Christos Chinopoulos
- Department of Medical Biochemistry, Semmelweis University, Budapest, 1094, Hungary
| |
Collapse
|
3
|
Yin XH, Wang XY, Liu SC, Chen XX, Yan L, Li L, Le He G, Yang M, Liu ZK. SIRT5 -mediated desuccinylation of UQCRC2 attenuates osteogenic differentiation of aged BM-MSCs through impairing mitochondrial homeostasis. Cell Signal 2025; 128:111636. [PMID: 39892725 DOI: 10.1016/j.cellsig.2025.111636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/19/2025] [Accepted: 01/29/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND The osteogenic differentiation potential of bone marrow mesenchymal stem cells (BM-MSCs) is critical for bone regeneration and repair. In recent years, the role of protein succinylation modification in regulating cellular metabolism has garnered increasing attention. However, its mechanism in osteogenic differentiation remains unclear. METHODS Oxygen consumption rate (OCR) and mitochondrial ROS (mtROS) were detected to assess mitochondrial function in BM-MSCs with successive passages. Alizarin red staining and western blot experiments were used to evaluate osteogenic differentiation capacity. Succinylation modification omics and Co-IP detection were conducted to determine SIRT5-mediated desuccinylation of UQCRC2. RESULTS Bioinformatics analysis revealed that sirtuin 5 (SIRT5) expression is upregulated with multiple rounds of BM-MSCs' passages, and is associated with biological pathways such as oxidative phosphorylation (OXPHOS), cellular senescence, and inhibition of osteogenic differentiation. Experiments in vitro confirmed the up-regulation of SIRT5 and the suppression of osteogenic differentiation with the increased times of BM-MSCs' passages. Overexpression of SIRT5 enhanced OXPHOS and elevated mtROS levels, but reduced the expression of Runx2 and osteocalcin, and decreased calcified nodules, thereby inhibiting the osteogenic differentiation of BM-MSCs. SIRT5-mediated desuccinylation of ubiquinol-cytochrome C reductase core protein 2 (UQCRC2) at the site of K250 promoted UQCRC2 translocation from cytoplasm to mitochondria, which enhanced the activity of mitochondrial respiratory complex III. It further increased mtROS, accelerated cellular senescence and inhibited the osteogenic differentiation of BM-MSCs. CONCLUSION SIRT5 reduces succinylation modification of UQCRC2, promotes mitochondrial respiration and mtROS, and thus reduces the osteogenic differentiation ability of BM-MSCs cells. SIRT5 might be a potential target to prevent the suppression of osteogenic differentiation of of BM-MSCs after multiple rounds passages.
Collapse
Affiliation(s)
- Xin Hua Yin
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, China
| | - Xiao Yuan Wang
- Physical Examination Center, Xi'an International Medical Center Hospital, Xi'an, China
| | - Shi Chang Liu
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, China
| | - Xu Xu Chen
- Department of Sports Medicine, Hong Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, China
| | - Liang Yan
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, China
| | - Liang Li
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, China
| | - Gao Le He
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, China
| | - Ming Yang
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, China.
| | - Zhong Kai Liu
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, China.
| |
Collapse
|
4
|
Chen W, Luo X, Li W, Li X, Wang Y, Zhang R, Liu B, Zhu L, Liu Z, Cheng Y. Uncovering the active ingredients of Xinbao pill against chronic heart failure: A chemical profiling, pharmacokinetics and pharmacodynamics integrated study. JOURNAL OF ETHNOPHARMACOLOGY 2025; 342:119418. [PMID: 39880064 DOI: 10.1016/j.jep.2025.119418] [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: 10/24/2024] [Revised: 01/04/2025] [Accepted: 01/25/2025] [Indexed: 01/31/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Xinbao pill (XBP) is a renowned Chinese patent medicine, primarily efficacious in warming and nourishing the heart and kidneys, supplementing Qi to boost Yang, and promoting blood circulation to remove blood stasis. XBP has been utilized for the treatment of chronic heart failure (CHF) for nearly 30 years, but the lack of clarity regarding the active ingredients of XBP against CHF has hindered its clinical application and further promotion. AIM OF THE STUDY To comprehensively elucidate the efficacy-specific ingredients and potential mechanism of XBP against CHF. METHODS The efficacy, chemical profiling and pharmacokinetics of XBP was assessed in a CHF model rat. The anti-CHF mechanism of the mixture of the likely active ingredients was clarified by targeted metabolomics and western blotting analysis. RESULTS XBP alleviated CHF by enhancing cardiac function, reducing NT-pro BNP, mitigating myocardial damage and degrading extracellular collagen. Following XBP administration, ginsenosides exposed relatively abundant in sham or CHF rats. Ginsenoside Rg1 and notoginsenoside R1 showed downward trends in AUC0-t values in CHF group, accompanied by increasing trends in CL/F values. Moreover, CHF rats presented significantly elevated levels of ginsenoside Rg1, ginsenoside Rg2 and notoginsenoside R1 in heart. The mixture of ginsenoside Rg1, ginsenoside Rg2 and notoginsenoside R1 demonstrated remarkable efficacy in ameliorating CHF as XBP did. Notably, these three compounds were predominantly localized in mitochondria and exhibited significant potential to enhance mitochondrial homeostasis by inhibiting heme synthesis pathway-mediated decomposition of succinyl CoA. CONCLUSIONS Our research provides valuable insights that ginsenoside Rg1, ginsenoside Rg2 and notoginsenoside R1 may constitute the anti-CHF active ingredients of XBP for facilitating mitochondrial homeostasis by the suppression of heme synthesis to increase succinyl CoA.
Collapse
Affiliation(s)
- Weiying Chen
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; The Second Clinical Medicial College, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Xiang Luo
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Wentao Li
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Xiaocui Li
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Ying Wang
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Rong Zhang
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Bo Liu
- The Second Clinical Medicial College, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Lijun Zhu
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Zhongqiu Liu
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Yuanyuan Cheng
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| |
Collapse
|
5
|
Qin Z, Chen J, Liu F, Li B, Zhang C, Wang X, Liu L, Wang M, Wang T, Wang S, Yu F, Wang S, Yang J. Jellyfish stings-induced cardiac failure was ameliorated through AAG-mediated glycogen-driven ATP production. EXPLORATION (BEIJING, CHINA) 2025; 5:20230089. [PMID: 40040825 PMCID: PMC11875447 DOI: 10.1002/exp.20230089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 01/16/2024] [Indexed: 07/27/2024]
Abstract
Jellyfish stings have become a common injury among fishermen and divers. Severe jellyfish stings could worsen cardiac function and even cause cardiac complications, ultimately leading to cardiac failure (CF). Currently, there are no effective drugs available. Single cell sequencing revealed alpha-1 acid glycoprotein (AAG), an energy regulatory protein targeting to glycogen, was highly expressed in jellyfish stings-induced CF patients. However, the mechanism remains elusive. It is postulated that AAG could increase glycogen metabolism, protecting against jellyfish stings-induced CF. AAG deletion exacerbated CF, while exogenous and endogenous AAG ameliorated CF. AAG also rescued the decline triggered by the AAG knockout (KO). Intriguingly, AAG improved cardiac function and metabolic adaptation by glycogen-driven ATP production, shifting mitochondrial/glycolytic ATP production towards glycolysis. Sorted by single-cell RNA sequencing and spatial transcription technology, CC-chemokine receptor 5 (CCR5) and Peroxisome proliferator-activated receptor-gamma coactivator-1alpha (PGC-1α) were differentially expressed. Mechanistically, CCR5 inhibitor MVC abolished AAG's protective effect and PGC-1α overexpression. Collectively, jellyfish stings-induced CF was ameliorated through AAG-mediated glycogen-driven ATP production, promoting glycolytic/mitochondrial metabolic switches to rely energetically primarily on glycolysis, which might serve as a therapeutic target of CF.
Collapse
Affiliation(s)
- Zhen Qin
- PLA Naval Medical CenterNaval Medical University (Second Military Medical University)ShanghaiChina
| | - Jinhong Chen
- PLA Naval Medical CenterNaval Medical University (Second Military Medical University)ShanghaiChina
| | - Fang Liu
- PLA Naval Medical CenterNaval Medical University (Second Military Medical University)ShanghaiChina
| | - Bingbing Li
- PLA Naval Medical CenterNaval Medical University (Second Military Medical University)ShanghaiChina
| | - Chenchen Zhang
- PLA Naval Medical CenterNaval Medical University (Second Military Medical University)ShanghaiChina
| | - Xiuxiu Wang
- PLA Naval Medical CenterNaval Medical University (Second Military Medical University)ShanghaiChina
| | - Lin Liu
- PLA Naval Medical CenterNaval Medical University (Second Military Medical University)ShanghaiChina
- School of Health Sciences and EngineeringUniversity of Shanghai for Science and TechnologyShanghaiChina
| | - Mingke Wang
- PLA Naval Medical CenterNaval Medical University (Second Military Medical University)ShanghaiChina
| | | | - Su Wang
- Center for Translational NeuromedicineUniversity of Rochester Medical CenterRochesterNew YorkUSA
| | - Feifei Yu
- PLA Naval Medical CenterNaval Medical University (Second Military Medical University)ShanghaiChina
| | - Shifeng Wang
- PLA Naval Medical CenterNaval Medical University (Second Military Medical University)ShanghaiChina
| | - Jishun Yang
- PLA Naval Medical CenterNaval Medical University (Second Military Medical University)ShanghaiChina
| |
Collapse
|
6
|
Qiu M, Geng H, Zou C, Zhao X, Zhao C, Xie J, Wang J, Zhang N, Hu Y, Fu Y, Wang J, Hu X. Intestinal inflammation exacerbates endometritis through succinate production by gut microbiota and SUCNR1-mediated proinflammatory response. Int Immunopharmacol 2025; 146:113919. [PMID: 39736240 DOI: 10.1016/j.intimp.2024.113919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/13/2024] [Accepted: 12/18/2024] [Indexed: 01/01/2025]
Abstract
Endometritis poses higher health risks to women. Clinical practice has found that gastrointestinal dysfunction is more likely to lead to the occurrence of endometritis. However, the mechanism is unclear. This study explored the influence and mechanism of DSS-induced intestinal inflammation on endometritis. Our findings demonstrate that DSS-induced intestinal inflammation can worsen LPS-induced endometritis in mice, and this effect is dependent on the gut microbiota, as depleting the gut microbiota eliminates this protective effect. Similarly, FMT from DSS-treated mice to recipient mice exacerbates LPS-induced endometritis. In addition, treatment of DSS disrupted an imbalance of succinate-producing and succinate-consuming bacteria and increased the levels of succinate in the gut and uterine tissues. Furthermore, treatment with succinate aggravates LPS-induced endometritis by activating the succinate receptor 1 (SUCNR1), evidenced by inhibition of the activation of SUCNR1 reversed the inflammatory response in uterine tissues induced by succinate during endometritis induced by LPS. Collectively, the results suggested that dysbiosis of the gut microbiota exacerbates LPS-induced endometritis by production and migration of succinate from gut to uterine tissues via the gut-uterus axis, then activates the SUCNR1. This identifies gut-derived succinate as a novel target for treating endometritis, and it indicates that targeting the gut microbiota and its metabolism could be a potential strategy for intervention in endometritis.
Collapse
Affiliation(s)
- Min Qiu
- Department of Gynecology, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin, China; Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China
| | - Huafeng Geng
- Department of Gynecology, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin, China
| | - Chenyu Zou
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China
| | - Xiaotong Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China
| | - Caijun Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China
| | - Jiaxin Xie
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China
| | - Jinnan Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China
| | - Naisheng Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China
| | - Yubo Hu
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Erdao District, 126 Sendai Street, Changchun, Jilin Province 130033, China
| | - Yunhe Fu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China
| | - Junrong Wang
- Department of Gynecology, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin, China.
| | - Xiaoyu Hu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China.
| |
Collapse
|
7
|
Li YC, Cheng ML. Carvedilol Confers Ferroptosis Resistance in HL-1 Cells by Upregulating GPX4, FTH1, and FTL1 and Inducing Metabolic Remodeling Under Hypoxia/Reoxygenation. Antioxidants (Basel) 2024; 14:7. [PMID: 39857341 PMCID: PMC11762394 DOI: 10.3390/antiox14010007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025] Open
Abstract
Hypoxia/reoxygenation (HR) often occurs under cardiac pathological conditions, and HR-induced oxidative stress usually leads to cardiomyocyte damage. Carvedilol, a non-selective β-blocker, is used clinically to treat cardiac ischemia diseases. Moreover, Carvedilol has also been reported to have an antioxidant ability by reducing lipid peroxidation. However, the mechanism of Carvedilol to inhibit lipid peroxidation is still elusive. To explore the protective mechanism of Carvedilol to resist lipid peroxidation on cardiomyocytes, HL-1 cells were cultured under normoxia, hypoxia, and HR and treated with Carvedilol to investigate the alteration on metabolism, protein expression, and mRNA level to explain its oxidative mechanism. The study found that Carvedilol upregulated glutathione peroxidase 4 (GPX4) protein expression to resist HR-induced lipid peroxidation by metabolic remodeling under HR. Also, Carvedilol promoted ferroptosis-related genes, ferritin heavy chain 1 (FTH1) and ferritin light chain 1 (FTL1) mRNA levels, to reduce lipid peroxidation under both hypoxia and HR. In conclusion, our study explores a mechanism by which Carvedilol inhibits ferroptosis by upregulating GPX4, FTH1, and FTL1 levels to downregulate lipid peroxidation under HR. The study provides a potential strategy for using Carvedilol in clinical applications, inspiring further research and development in the area of heart diseases.
Collapse
Affiliation(s)
- Yi-Chin Li
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan;
| | - Mei-Ling Cheng
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan;
- Metabolomics Core Laboratory, Healthy Aging Research Center, Chang Gung University, Taoyuan City 33302, Taiwan
- Clinical Metabolomics Core Laboratory, Chang Gung Memorial Hospital, Taoyuan City 33305, Taiwan
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan
| |
Collapse
|
8
|
Cheng YH, Chiang CY, Wu CH, Chien CT. 2'-Hydroxycinnamaldehyde, a Natural Product from Cinnamon, Alleviates Ischemia/Reperfusion-Induced Microvascular Dysfunction and Oxidative Damage in Rats by Upregulating Cytosolic BAG3 and Nrf2/HO-1. Int J Mol Sci 2024; 25:12962. [PMID: 39684673 DOI: 10.3390/ijms252312962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/20/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024] Open
Abstract
2'-Hydroxycinnamaldehyde (HCA), a natural product isolated from the bark of Cinnamomum cassia, has anti-inflammatory and anti-tumor activities. In this study, we explored whether HCA preconditioning could protect the heart against ischemia/reperfusion (I/R)-induced oxidative injury through cytosolic Bcl-2-associated athanogene 3 (BAG3) upregulation. In vivo HCA preconditioning was performed intraperitoneally in adult male Wistar rats (50 mg/kg body weight) three times/week for 2 weeks before cardiac I/R injury. The animals were divided into sham control (sham), I/R, and HCA preconditioning plus I/R (HCA+I/R) groups. We examined left ventricular pressure cardiac hemodynamics, the microcirculation, electrocardiograms, infarct size, and oxidative stress and performed Western blots, immunohistochemistry, and cytokine array assays. HCA pretreatment, via BAG3 overexpression, inhibited H2O2-induced H9c2 cell death. Cardiac I/R injury increased ST-segment elevation, left ventricular end-diastolic pressure, infarct size, myocardial disruption, tissue edema, erythrocyte accumulation, leukocyte infiltration, reactive oxygen species, malondialdehyde, 8-isoprostane, caspase 3-mediated apoptosis, 4HNE/GPX4-mediated ferroptosis, and fibrosis but decreased the microcirculation, cytosolic BAG3, and Beclin-1/LC3 II-mediated autophagy in the I/R hearts. HCA preconditioning significantly decreased these oxidative injuries by increasing cardiac cytosolic BAG3 and Nrf2/HO-1 signaling. HCA preconditioning significantly decreased cardiac I/R-enhanced mitochondrial fission DRP1 expression. Our data suggest that HCA preconditioning can efficiently improve myocardial I/R injury-induced cardiac dysfunction, apoptosis, ferroptosis, mitochondrial fission, and autophagy inhibition through cardiac BAG3 and Nrf2/HO-1 upregulation.
Collapse
Affiliation(s)
- Yu-Hsuan Cheng
- School of Life Science, National Taiwan Normal University, Taipei 117, Taiwan
| | - Chih-Yao Chiang
- Department of Medicial Research and Division of Cardiovascular Surgery, Cardiovascular Center, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan
- Division of Cardiovascular Surgery, National Defense Medical Center, Taipei 114, Taiwan
| | - Chung-Hsin Wu
- School of Life Science, National Taiwan Normal University, Taipei 117, Taiwan
| | - Chiang-Ting Chien
- School of Life Science, National Taiwan Normal University, Taipei 117, Taiwan
| |
Collapse
|
9
|
Lee DC, Ta L, Mukherjee P, Duraj T, Domin M, Greenwood B, Karmacharya S, Narain NR, Kiebish M, Chinopoulos C, Seyfried TN. Amino Acid and Glucose Fermentation Maintain ATP Content in Mouse and Human Malignant Glioma Cells. ASN Neuro 2024; 16:2422268. [PMID: 39621724 PMCID: PMC11792161 DOI: 10.1080/17590914.2024.2422268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 10/22/2024] [Indexed: 02/06/2025] Open
Abstract
Energy is necessary for tumor cell viability and growth. Aerobic glucose-driven lactic acid fermentation is a common metabolic phenotype seen in most cancers including malignant gliomas. This metabolic phenotype is linked to abnormalities in mitochondrial structure and function. A luciferin-luciferase bioluminescence ATP assay was used to measure the influence of amino acids, glucose, and oxygen on ATP content and viability in mouse (VM-M3 and CT-2A) and human (U-87MG) glioma cells that differed in cell biology, genetic background, and species origin. Oxygen consumption was measured using the Resipher system. Extracellular lactate and succinate were measured as end products of the glycolysis and glutaminolysis pathways, respectively. The results showed that: (1) glutamine was a source of ATP content irrespective of oxygen. No other amino acid could replace glutamine in sustaining ATP content and viability; (2) ATP content persisted in the absence of glucose and under hypoxia, ruling out substantial contribution through either glycolysis or oxidative phosphorylation (OxPhos) under these conditions; (3) Mitochondrial complex IV inhibition showed that oxygen consumption was not an accurate measure for ATP production through OxPhos. The glutaminase inhibitor, 6-diazo-5-oxo-L-norleucine (DON), reduced ATP content and succinate export in cells grown in glutamine. The data suggests that mitochondrial substrate level phosphorylation in the glutamine-driven glutaminolysis pathway contributes to ATP content in these glioma cells. A new model is presented highlighting the synergistic interaction between the high-throughput glycolysis and glutaminolysis pathways that drive malignant glioma growth and maintain ATP content through the aerobic fermentation of both glucose and glutamine.
Collapse
Affiliation(s)
- Derek C. Lee
- Department of Biology, Boston College, Massachusetts, USA
| | - Linh Ta
- Department of Biology, Boston College, Massachusetts, USA
| | | | - Tomas Duraj
- Department of Biology, Boston College, Massachusetts, USA
| | - Marek Domin
- Mass Spectrometry Center, Chemistry Department, Boston College, Massachusetts, USA
| | | | | | | | | | | | | |
Collapse
|
10
|
Paulin Beske R, Meyer MAS, Emil Roelsgaard Obling L, Eifer Møller J, Kjaergaard J, Johansson PI, Hassager C. Interleukin 6 blockage alters the plasma metabolome in out-of-hospital cardiac arrest. Resuscitation 2024; 205:110425. [PMID: 39510308 DOI: 10.1016/j.resuscitation.2024.110425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/21/2024] [Accepted: 11/03/2024] [Indexed: 11/15/2024]
Abstract
BACKGROUND Comatose patients resuscitated from out-of-hospital cardiac arrest (OHCA) exhibit a systemic inflammatory response, as indicated by elevated interleukin-6 (IL-6) levels, which is associated with increased mortality. Tocilizumab, an IL-6 receptor antagonist that reduced C-reactive protein response and markers of myocardial injury in a phase II OHCA trial. AIM To describe the early effects of tocilizumab on circulating levels of metabolites in comatose patients resuscitated from OHCA. METHOD Patients from the phase-II double-blinded randomized trial (NCT: 03863015) were included in this substudy. A total of 85 comatose patients resuscitated from OHCA were randomized at the time of arrival to the hospital to either tocilizumab 8 mg/kg or placebo, of which 80 received the intervention and did not later withdraw from the study. Plasma samples before randomization and 48 h later were analyzed by a targeted metabolomics approach quantifying 60 circulating metabolites. RESULTS Of 80 enrolled patients (median age 62 years (IQR: 54-72), men 66 (83 %)), 39 were randomized to tocilizumab group and 41 to placebo. Comorbidities and cardiac arrest characteristics were overall well-balanced. At hospital arrival, levels of metabolites from the tricarboxylic acid (TCA) cycle were associated with time to return of spontaneous circulation and independently with early levels of IL-6 (all p < 0.05). The early levels of medium-chain acylcarnitines were associated with age, NT-proBNP, estimated glomerular filtration rate, and marker of neurological injury (neurofilament light chain) (all p < 0.01). At 48 h, tocilizumab increased the levels of plasma amino acids, especially threonine, glycine, and serine, by more than a factor of 1.5 (p < 0.01). Two eicosanoids 15(S)-HETE and 12(S)-HETE were 1.9 times higher (p < 0.01). CONCLUSION Blocking the IL-6 receptor with tocilizumab early after OHCA impacts circulating metabolites, particularly those within the glycine, serine, and threonine pathways, highlighting the connection between acute systemic inflammation and metabolism. Further, early levels of TCA metabolites are independently associated with early inflammatory response and early medium-chain acylcarnitine with later markers of neurological injury.
Collapse
Affiliation(s)
- Rasmus Paulin Beske
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Center for Endotheliomics, CAG, Department of Clinical Immunology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark.
| | - Martin A S Meyer
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | | | - Jacob Eifer Møller
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Cardiology, Odense University Hospital, Odense, Denmark
| | - Jesper Kjaergaard
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Pär I Johansson
- Center for Endotheliomics, CAG, Department of Clinical Immunology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Christian Hassager
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
11
|
Tian L, Liu Q, Guo H, Zang H, Li Y. Fighting ischemia-reperfusion injury: Focusing on mitochondria-derived ferroptosis. Mitochondrion 2024; 79:101974. [PMID: 39461581 DOI: 10.1016/j.mito.2024.101974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/12/2024] [Accepted: 10/12/2024] [Indexed: 10/29/2024]
Abstract
Ischemia-reperfusion injury (IRI) is a major cause of mortality and morbidity. Current treatments for IRI have limited efficacy and novel therapeutic strategies are needed. Mitochondrial dysfunction not only initiates IRI but also plays a significant role in ferroptosis pathogenesis. Recent studies have highlighted that targeting mitochondrial pathways is a promising therapeutic approach for ferroptosis-induced IRI. The association between ferroptosis and IRI has been reviewed many times, but our review provides the first comprehensive overview with a focus on recent mitochondrial research. First, we present the role of mitochondria in ferroptosis. Then, we summarize the evidence on mitochondrial manipulation of ferroptosis in IRI and review recent therapeutic strategies aimed at targeting mitochondria-related ferroptosis to mitigate IRI. We hope our review will provide new ideas for the treatment of IRI and accelerate the transition from bench to bedside.
Collapse
Affiliation(s)
- Lei Tian
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Qian Liu
- Department of Anesthesiology, Zigong First People's Hospital, Zigong Academy of Medical Sciences, Zigong, China
| | - Hong Guo
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Honggang Zang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Yulan Li
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou, China.
| |
Collapse
|
12
|
Cárdenas P, Nuñez-Allimant C, Silva K, Cid-Salinas C, León AC, Vallotton Z, Lorca RA, de Oliveira LCG, Casarini DE, Céspedes C, Prieto MC, Gonzalez AA. OXGR1-Dependent (Pro)Renin Receptor Upregulation in Collecting Ducts of the Clipped Kidney Contributes to Na + Balance in Goldblatt Hypertensive Mice. Int J Mol Sci 2024; 25:10045. [PMID: 39337535 PMCID: PMC11432382 DOI: 10.3390/ijms251810045] [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: 08/17/2024] [Revised: 09/13/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
The two-kidney, one-clip (2K1C) Goldblatt rodent model elicits a reduction in renal blood flow (RBF) in the clipped kidney (CK). The reduced RBF and oxygen bio-ability causes the accumulation of the tricarboxylic cycle intermediary, α-ketoglutarate, which activates the oxoglutarate receptor-1 (OXGR1). In the kidney, OXGR1 is abundantly expressed in intercalated cells (ICs) of the collecting duct (CD), thus contributing to sodium transport and electrolyte balance. The (pro)renin receptor (PRR), a member of the renin-angiotensin system (RAS), is a key regulator of sodium reabsorption and blood pressure (BP) that is expressed in ICs. The PRR is upregulated in 2K1C rats. Here, we tested the hypothesis that chronic reduction in RBF in the CK leads to OXGR1-dependent PRR upregulation in the CD and alters sodium balance and BP in 2K1C mice. To determine the role of OXGR1 in regulating the PRR in the CDs during renovascular hypertension, we performed 2K1C Goldblatt surgery (clip = 0.13 mm internal gap, 14 days) in two groups of male mice: (1) mice treated with Montelukast (OXGR1 antagonist; 5 mg/Kg/day); (2) OXGR1-/- knockout mice. Wild-type and sham-operated mice were used as controls. After 14 days, 2K1C mice showed increased systolic BP (SBP) (108 ± 11 vs. control 82 ± 5 mmHg, p < 0.01) and a lower natriuretic response after the saline challenge test. The CK group showed upregulation of erythropoietin, augmented α-ketoglutarate, and increased PRR expression in the renal medulla. The CK of OXGR1 knockout mice and mice subjected to the OXGR1 antagonist elicited impaired PRR upregulation, attenuated SBP, and better natriuretic responses. In 2K1C mice, the effect of reduced RBF on the OXGR1-dependent PRR upregulation in the CK may contribute to the anti-natriuretic and increased SBP responses.
Collapse
MESH Headings
- Animals
- Mice
- Kidney Tubules, Collecting/metabolism
- Receptors, Cell Surface/metabolism
- Receptors, Cell Surface/genetics
- Male
- Up-Regulation
- Sodium/metabolism
- Hypertension, Renovascular/metabolism
- Hypertension, Renovascular/genetics
- Blood Pressure
- Mice, Knockout
- Prorenin Receptor
- Kidney/metabolism
- Disease Models, Animal
- Renin-Angiotensin System
- Mice, Inbred C57BL
- Receptors, G-Protein-Coupled/metabolism
- Receptors, G-Protein-Coupled/genetics
- Receptors, Purinergic P2
Collapse
Affiliation(s)
- Pilar Cárdenas
- Institute of Chemistry, Pontificia Universidad Catoólica de Valparaióso, Valparaióso 2340000, Chile; (P.C.); (C.N.-A.); (K.S.); (C.C.-S.); (A.C.L.)
| | - Camila Nuñez-Allimant
- Institute of Chemistry, Pontificia Universidad Catoólica de Valparaióso, Valparaióso 2340000, Chile; (P.C.); (C.N.-A.); (K.S.); (C.C.-S.); (A.C.L.)
| | - Katherin Silva
- Institute of Chemistry, Pontificia Universidad Catoólica de Valparaióso, Valparaióso 2340000, Chile; (P.C.); (C.N.-A.); (K.S.); (C.C.-S.); (A.C.L.)
| | - Catalina Cid-Salinas
- Institute of Chemistry, Pontificia Universidad Catoólica de Valparaióso, Valparaióso 2340000, Chile; (P.C.); (C.N.-A.); (K.S.); (C.C.-S.); (A.C.L.)
| | - Allison C. León
- Institute of Chemistry, Pontificia Universidad Catoólica de Valparaióso, Valparaióso 2340000, Chile; (P.C.); (C.N.-A.); (K.S.); (C.C.-S.); (A.C.L.)
| | - Zoe Vallotton
- Department of Physiology, Tulane Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA 70112, USA; (Z.V.); (M.C.P.)
| | - Ramón A. Lorca
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Lilian Caroline Gonçalves de Oliveira
- Departamento de Medicina, Disciplina de Nefrologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-062, Brazil; (L.C.G.d.O.); (D.E.C.)
| | - Dulce E Casarini
- Departamento de Medicina, Disciplina de Nefrologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-062, Brazil; (L.C.G.d.O.); (D.E.C.)
| | - Carlos Céspedes
- Faculty of Medicine and Science, Universidad San Sebastián, Santiago 7510602, Chile;
| | - Minolfa C. Prieto
- Department of Physiology, Tulane Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA 70112, USA; (Z.V.); (M.C.P.)
| | - Alexis A. Gonzalez
- Institute of Chemistry, Pontificia Universidad Catoólica de Valparaióso, Valparaióso 2340000, Chile; (P.C.); (C.N.-A.); (K.S.); (C.C.-S.); (A.C.L.)
| |
Collapse
|
13
|
Jeon KI, Kumar A, Brookes PS, Nehrke K, Huxlin KR. Manipulating mitochondrial pyruvate carrier function causes metabolic remodeling in corneal myofibroblasts that ameliorates fibrosis. Redox Biol 2024; 75:103235. [PMID: 38889622 PMCID: PMC11231598 DOI: 10.1016/j.redox.2024.103235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 05/28/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024] Open
Abstract
Myofibroblasts are key cellular effectors of corneal wound healing from trauma, surgery, or infection. However, their persistent deposition of disorganized extracellular matrix can also cause corneal fibrosis and visual impairment. Recent work showed that the PPARγ agonist Troglitazone can mitigate established corneal fibrosis, and parallel in vitro data suggested this occurred through inhibition of the mitochondrial pyruvate carrier (MPC) rather than PPARγ. In addition to oxidative phosphorylation (Ox-Phos), pyruvate and other mitochondrial metabolites provide carbon for the synthesis of biological macromolecules. However, it is currently unclear how these roles selectively impact fibrosis. Here, we performed bioenergetic, metabolomic, and epigenetic analyses of corneal fibroblasts treated with TGF-β1 to stimulate myofibroblast trans-differentiation, with further addition of Troglitazone or the MPC inhibitor UK5099, to identify MPC-dependencies that may facilitate remodeling and loss of the myofibroblast phenotype. Our results show that a shift in energy metabolism is associated with, but not sufficient to drive cellular remodeling. Metabolites whose abundances were sensitive to MPC inhibition suggest that sustained carbon influx into the Krebs' cycle is prioritized over proline synthesis to fuel collagen deposition. Furthermore, increased abundance of acetyl-CoA and increased histone H3 acetylation suggest that epigenetic mechanisms downstream of metabolic remodeling may reinforce cellular phenotypes. Overall, our results highlight a novel molecular target and metabolic vulnerability that affects myofibroblast persistence in the context of corneal wounding.
Collapse
Affiliation(s)
- Kye-Im Jeon
- Dept. Ophthalmology, Flaum Eye Institute and Center for Visual Science, University of Rochester, Rochester, NY, USA
| | - Ankita Kumar
- Dept. Ophthalmology, Flaum Eye Institute and Center for Visual Science, University of Rochester, Rochester, NY, USA
| | - Paul S Brookes
- Dept. Anesthesiology and Perioperative Medicine, University of Rochester, Rochester, NY, USA
| | - Keith Nehrke
- Dept. Medicine-Nephrology Division, University of Rochester, Rochester, NY, USA
| | - Krystel R Huxlin
- Dept. Ophthalmology, Flaum Eye Institute and Center for Visual Science, University of Rochester, Rochester, NY, USA.
| |
Collapse
|
14
|
Condori LDM, Vivas CV, Barreto YB, Gomes LF, Alencar AM, Bloise AC. Effects of Hypoxia and Reoxygenation on Metabolic Profiles of Cardiomyocytes. Cell Biochem Biophys 2024; 82:969-985. [PMID: 38498099 DOI: 10.1007/s12013-024-01249-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/05/2024] [Indexed: 03/20/2024]
Abstract
In vitro cellular models provide valuable insights into the adaptive biochemical mechanisms triggered by cells to cope with the stress situation induced by hypoxia and reoxygenation cycles. The first biological data generated in studies based on this micrometric life-scale has the potential to provide us a global overview about the main biochemical phenomena presented in some reported preconditioning therapies in life-scale of higher dimensions. Thus, in this study, a cell incubator was designed and manufactured to produce a cellular model of heart hypoxia followed by reoxygenation (HfR) through consecutive repetitions of hypoxia-normoxia gas exchange. Samples of cellular extracts and culture media were obtained from non-proliferative cardiomyocytes (CMs) cultivated under challenging HfR (stressed CMs) and regular cultivation (unstressed CMs) in rounds of four days for each case. Metabolomic based on proton magnetic resonance spectroscopy (1H-MRS) was used as an analytical approach to identify and quantify the metabolomes of these samples, the endo- and exo-metabolome. Despite the stressed CMs presented over 90% higher cellular death rate compared to the unstressed CMs, the metabolic profiles indicates that the surviving cells up-regulate their amino acid metabolism either by active protein degradation or by the consumption of culture media components to increase coenzyme A-dependent metabolic pathways. This cell auto-regulation mechanism could be well characterized in the first two days when the difference smears off under once the metabolomes become similar. The metabolic adaptations of stressed CMs identified the relevance of the cyclic oxidation/reduction reactions of nicotinamide adenine dinucleotide phosphate molecules, NADP+/NADPH, and the increased tricarboxylic acid cycle activity in an environment overloaded with such a powerful antioxidant agent to survive an extreme HfR challenge. Thus, the combination of cellular models based on CMs, investigative methods, such as metabolomic and 1H-MRS, and the instrumental development of hypoxia incubator shown in this work were able to provide the first biochemical evidences behind therapies of gaseous exchanges paving the way to future assays.
Collapse
Affiliation(s)
| | | | - Yan Borges Barreto
- Universidade de Sao Paulo, Instituto de Fisica, Rua do Matao 1371, Sao Paulo, Brazil
| | - Ligia Ferreira Gomes
- Universidade de Sao Paulo, Instituto de Fisica, Rua do Matao 1371, Sao Paulo, Brazil.
| | | | - Antonio Carlos Bloise
- Universidade de Sao Paulo, Instituto de Fisica, Rua do Matao 1371, Sao Paulo, Brazil.
| |
Collapse
|
15
|
Chinopoulos C. Complex I activity in hypoxia: implications for oncometabolism. Biochem Soc Trans 2024; 52:529-538. [PMID: 38526218 PMCID: PMC11088919 DOI: 10.1042/bst20230189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 03/06/2024] [Accepted: 03/14/2024] [Indexed: 03/26/2024]
Abstract
Certain cancer cells within solid tumors experience hypoxia, rendering them incapable of oxidative phosphorylation (OXPHOS). Despite this oxygen deficiency, these cells exhibit biochemical pathway activity that relies on NAD+. This mini-review scrutinizes the persistent, residual Complex I activity that oxidizes NADH in the absence of oxygen as the electron acceptor. The resulting NAD+ assumes a pivotal role in fueling the α-ketoglutarate dehydrogenase complex, a critical component in the oxidative decarboxylation branch of glutaminolysis - a hallmark oncometabolic pathway. The proposition is that through glutamine catabolism, high-energy phosphate intermediates are produced via substrate-level phosphorylation in the mitochondrial matrix substantiated by succinyl-CoA ligase, partially compensating for an OXPHOS deficiency. These insights provide a rationale for exploring Complex I inhibitors in cancer treatment, even when OXPHOS functionality is already compromised.
Collapse
|
16
|
Lian J, Liu W, Hu Q, Zhang X. Succinylation modification: a potential therapeutic target in stroke. Neural Regen Res 2024; 19:781-787. [PMID: 37843212 PMCID: PMC10664134 DOI: 10.4103/1673-5374.382229] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 04/27/2023] [Accepted: 06/26/2023] [Indexed: 10/17/2023] Open
Abstract
Stroke is a leading cause of mortality and disability worldwide. Ischemic cell death triggered by the compromised supply of blood oxygen and glucose is one of the major pathophysiology of stroke-induced brain injury. Impaired mitochondrial energy metabolism is observed minutes after stroke and is closely associated with the progression of neuropathology. Recently, a new type of post-translational modification, known as lysine succinylation, has been recognized to play a significant role in mitochondrial energy metabolism after ischemia. However, the role of succinylation modification in cell metabolism after stroke and its regulation are not well understood. We aimed to review the effects of succinylation on energy metabolism, reactive oxygen species generation, and neuroinflammation, as well as Sirtuin 5 mediated desuccinylation after stroke. We also highlight the potential of targeting succinylation/desuccinylation as a promising strategy for the treatment of stroke. The succinylation level is dynamically regulated by the nonenzymatic or enzymatic transfer of a succinyl group to a protein on lysine residues and the removal of succinyl catalyzed by desuccinylases. Mounting evidence has suggested that succinylation can regulate the metabolic pathway through modulating the activity or stability of metabolic enzymes. Sirtuins, especially Sirtuin 5, are characterized for their desuccinylation activity and have been recognized as a critical regulator of metabolism through desuccinylating numerous metabolic enzymes. Imbalance between succinylation and desuccinylation has been implicated in the pathophysiology of stroke. Pharmacological agents that enhance the activity of Sirtuin 5 have been employed to promote desuccinylation and improve mitochondrial metabolism, and neuroprotective effects of these agents have been observed in experimental stroke studies. However, their therapeutic efficacy in stroke patients should be validated.
Collapse
Affiliation(s)
- Jie Lian
- Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenwu Liu
- Department of Diving and Hyperbaric Medicine, Naval Medical Center, Naval Medical University, Shanghai, China
| | - Qin Hu
- Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaohua Zhang
- Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
17
|
Tabata Fukushima C, Dancil IS, Clary H, Shah N, Nadtochiy SM, Brookes PS. Reactive oxygen species generation by reverse electron transfer at mitochondrial complex I under simulated early reperfusion conditions. Redox Biol 2024; 70:103047. [PMID: 38295577 PMCID: PMC10844975 DOI: 10.1016/j.redox.2024.103047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/09/2024] [Accepted: 01/15/2024] [Indexed: 02/02/2024] Open
Abstract
Ischemic tissues accumulate succinate, which is rapidly oxidized upon reperfusion, driving a burst of mitochondrial reactive oxygen species (ROS) generation that triggers cell death. In isolated mitochondria with succinate as the sole metabolic substrate under non-phosphorylating conditions, 90 % of ROS generation is from reverse electron transfer (RET) at the Q site of respiratory complex I (Cx-I). Together, these observations suggest Cx-I RET is the source of pathologic ROS in reperfusion injury. However, numerous factors present in early reperfusion may impact Cx-I RET, including: (i) High [NADH]; (ii) High [lactate]; (iii) Mildly acidic pH; (iv) Defined ATP/ADP ratios; (v) Presence of the nucleosides adenosine and inosine; and (vi) Defined free [Ca2+]. Herein, experiments with mouse cardiac mitochondria revealed that under simulated early reperfusion conditions including these factors, total mitochondrial ROS generation was only 56 ± 17 % of that seen with succinate alone (mean ± 95 % confidence intervals). Of this ROS, only 52 ± 20 % was assignable to Cx-I RET. A further 14 ± 7 % could be assigned to complex III, with the remainder (34 ± 11 %) likely originating from other ROS sources upstream of the Cx-I Q site. Together, these data suggest the relative contribution of Cx-I RET ROS to reperfusion injury may be overestimated, and other ROS sources may contribute a significant fraction of ROS in early reperfusion.
Collapse
Affiliation(s)
- Caio Tabata Fukushima
- Departments of Anesthesiology, University of Rochester Medical Center, USA; Departments of Biochemistry, University of Rochester Medical Center, USA; Pharmacology and Physiology, University of Rochester Medical Center, USA
| | - Ian-Shika Dancil
- Departments of Anesthesiology, University of Rochester Medical Center, USA
| | - Hannah Clary
- Departments of Biochemistry, University of Rochester Medical Center, USA
| | - Nidhi Shah
- Pharmacology and Physiology, University of Rochester Medical Center, USA
| | - Sergiy M Nadtochiy
- Departments of Anesthesiology, University of Rochester Medical Center, USA
| | - Paul S Brookes
- Departments of Anesthesiology, University of Rochester Medical Center, USA; Pharmacology and Physiology, University of Rochester Medical Center, USA.
| |
Collapse
|
18
|
Kim MJ, Oh CJ, Hong CW, Jeon JH. Comprehensive overview of the role of mitochondrial dysfunction in the pathogenesis of acute kidney ischemia-reperfusion injury: a narrative review. JOURNAL OF YEUNGNAM MEDICAL SCIENCE 2024; 41:61-73. [PMID: 38351610 DOI: 10.12701/jyms.2023.01347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 01/10/2024] [Indexed: 05/08/2024]
Abstract
Acute kidney ischemia-reperfusion (IR) injury is a life-threatening condition that predisposes individuals to chronic kidney disease. Since the kidney is one of the most energy-demanding organs in the human body and mitochondria are the powerhouse of cells, mitochondrial dysfunction plays a central role in the pathogenesis of IR-induced acute kidney injury. Mitochondrial dysfunction causes a reduction in adenosine triphosphate production, loss of mitochondrial dynamics (represented by persistent fragmentation), and impaired mitophagy. Furthermore, the pathological accumulation of succinate resulting from fumarate reduction under oxygen deprivation (ischemia) in the reverse flux of the Krebs cycle can eventually lead to a burst of reactive oxygen species driven by reverse electron transfer during the reperfusion phase. Accumulating evidence indicates that improving mitochondrial function, biogenesis, and dynamics, and normalizing metabolic reprogramming within the mitochondria have the potential to preserve kidney function during IR injury and prevent progression to chronic kidney disease. In this review, we summarize recent advances in understanding the detrimental role of metabolic reprogramming and mitochondrial dysfunction in IR injury and explore potential therapeutic strategies for treating kidney IR injury.
Collapse
Affiliation(s)
- Min-Ji Kim
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Korea
| | - Chang Joo Oh
- Research Institute of Aging and Metabolism, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Chang-Won Hong
- Department of Physiology, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Jae-Han Jeon
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Korea
| |
Collapse
|
19
|
Tu C, Caudal A, Liu Y, Gorgodze N, Zhang H, Lam CK, Dai Y, Zhang A, Wnorowski A, Wu MA, Yang H, Abilez OJ, Lyu X, Narayan SM, Mestroni L, Taylor MRG, Recchia FA, Wu JC. Tachycardia-induced metabolic rewiring as a driver of contractile dysfunction. Nat Biomed Eng 2024; 8:479-494. [PMID: 38012305 PMCID: PMC11088531 DOI: 10.1038/s41551-023-01134-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 10/15/2023] [Indexed: 11/29/2023]
Abstract
Prolonged tachycardia-a risk factor for cardiovascular morbidity and mortality-can induce cardiomyopathy in the absence of structural disease in the heart. Here, by leveraging human patient data, a canine model of tachycardia and engineered heart tissue generated from human induced pluripotent stem cells, we show that metabolic rewiring during tachycardia drives contractile dysfunction by promoting tissue hypoxia, elevated glucose utilization and the suppression of oxidative phosphorylation. Mechanistically, a metabolic shift towards anaerobic glycolysis disrupts the redox balance of nicotinamide adenine dinucleotide (NAD), resulting in increased global protein acetylation (and in particular the acetylation of sarcoplasmic/endoplasmic reticulum Ca2+-ATPase), a molecular signature of heart failure. Restoration of NAD redox by NAD+ supplementation reduced sarcoplasmic/endoplasmic reticulum Ca2+-ATPase acetylation and accelerated the functional recovery of the engineered heart tissue after tachycardia. Understanding how metabolic rewiring drives tachycardia-induced cardiomyopathy opens up opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Chengyi Tu
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Arianne Caudal
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Yu Liu
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Nikoloz Gorgodze
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Hao Zhang
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Chi Keung Lam
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Yuqin Dai
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Angela Zhang
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Greenstone Biosciences, Palo Alto, CA, USA
| | - Alexa Wnorowski
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Matthew A Wu
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Greenstone Biosciences, Palo Alto, CA, USA
| | - Huaxiao Yang
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Oscar J Abilez
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Xuchao Lyu
- Department of Pathology, Stanford University, Stanford, CA, USA
| | | | - Luisa Mestroni
- Human Medical Genetics and Genomics, University of Colorado, Aurora, CO, USA
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado, Aurora, CO, USA
| | - Matthew R G Taylor
- Human Medical Genetics and Genomics, University of Colorado, Aurora, CO, USA
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado, Aurora, CO, USA
| | - Fabio A Recchia
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
- Scuola Superiore Sant'Anna, Pisa, Italy
- Institute of Clinical Physiology of the National Research Council, Pisa, Italy
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA.
- Department of Medicine, Stanford University, Stanford, CA, USA.
- Department of Radiology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
20
|
Wang X, Li M, Wang F, Mao G, Wu J, Han R, Sheng R, Qin Z, Ni H. TIGAR reduces neuronal ferroptosis by inhibiting succinate dehydrogenase activity in cerebral ischemia. Free Radic Biol Med 2024; 216:89-105. [PMID: 38494143 DOI: 10.1016/j.freeradbiomed.2024.03.011] [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: 10/21/2023] [Revised: 01/12/2024] [Accepted: 03/14/2024] [Indexed: 03/19/2024]
Abstract
Ischemia Stroke (IS) is an acute neurological condition with high morbidity, disability, and mortality due to a severe reduction in local cerebral blood flow to the brain and blockage of oxygen and glucose supply. Oxidative stress induced by IS predisposes neurons to ferroptosis. TP53-induced glycolysis and apoptosis regulator (TIGAR) inhibits the intracellular glycolytic pathway to increase pentose phosphate pathway (PPP) flux, promotes NADPH production and thus generates reduced glutathione (GSH) to scavenge reactive oxygen species (ROS), and thus shows strong antioxidant effects to ameliorate cerebral ischemia/reperfusion injury. However, in the current study, prolonged ischemia impaired the PPP, and TIGAR was unable to produce NADPH but was still able to reduce neuronal ferroptosis and attenuate ischemic brain injury. Ferroptosis is a form of cell death caused by free radical-driven lipid peroxidation, and the vast majority of ROS leading to oxidative stress are generated by mitochondrial succinate dehydrogenase (SDH) driving reverse electron transfer (RET) via the mitochondrial electron transport chain. Overexpression of TIGAR significantly inhibited hypoxia-induced enhancement of SDH activity, and TIGAR deficiency further enhanced SDH activity. We also found that the inhibitory effect of TIGAR on SDH activity was related to its mitochondrial translocation under hypoxic conditions. TIGAR may inhibit SDH activity by mediating post-translational modifications (acetylation and succinylation) of SDH A through interaction with SDH A. SDH activity inhibition reduces neuronal ferroptosis by decreasing ROS production, eliminating MitoROS levels and attenuating lipid peroxide accumulation. Notably, TIGAR-mediated inhibition of SDH activity and ferroptosis was not dependent on the PPP-NADPH-GPX4 pathways. In conclusion, mitochondrial translocation of TIGAR in prolonged ischemia is an important pathway to reduce neuronal ferroptosis and provide sustainable antioxidant defense for the brain under prolonged ischemia, further complementing the mechanism of TIGAR resistance to oxidative stress induced by IS.
Collapse
Affiliation(s)
- Xinxin Wang
- Department of Brain Research, Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Mei Li
- Department of Brain Research, Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Fan Wang
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Guanghui Mao
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Junchao Wu
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Rong Han
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Rui Sheng
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Zhenghong Qin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China; Institute of Heath Technology, Global Institute of Software Technology, Qingshan Road, Suzhou Science & Technology Tower, Hi-Tech Area, Suzhou, 215163, China.
| | - Hong Ni
- Department of Brain Research, Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, China.
| |
Collapse
|
21
|
Beutner G, Burris JR, Collins MP, Kulkarni CA, Nadtochiy SM, de Mesy Bentley KL, Cohen ED, Brookes PS, Porter GA. Coordinated metabolic responses to cyclophilin D deletion in the developing heart. iScience 2024; 27:109157. [PMID: 38414851 PMCID: PMC10897919 DOI: 10.1016/j.isci.2024.109157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 12/02/2023] [Accepted: 02/03/2024] [Indexed: 02/29/2024] Open
Abstract
In the embryonic heart, the activation of the mitochondrial electron transport chain (ETC) coincides with the closure of the cyclophilin D (CypD) regulated mitochondrial permeability transition pore (mPTP). However, it remains to be established whether the absence of CypD has a regulatory effect on mitochondria during cardiac development. Using a variety of assays to analyze cardiac tissue from wildtype and CypD knockout mice from embryonic day (E)9.5 to adult, we found that mitochondrial structure, function, and metabolism show distinct transitions. Deletion of CypD altered the timing of these transitions as the mPTP was closed at all ages, leading to coupled ETC activity in the early embryo, decreased citrate synthase activity, and an altered metabolome particularly after birth. Our results suggest that manipulating CypD activity may control myocyte proliferation and differentiation and could be a tool to increase ATP production and cardiac function in immature hearts.
Collapse
Affiliation(s)
- Gisela Beutner
- Department of Pediatrics, Division of Cardiology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Jonathan Ryan Burris
- Department of Pediatrics, Division of Cardiology, University of Rochester Medical Center, Rochester, NY 14642, USA
- Department of Pediatrics, Division of Neonatology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Michael P. Collins
- Department of Pediatrics, Division of Cardiology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Chaitanya A. Kulkarni
- Department of Anesthesiology & Perioperative Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Sergiy M. Nadtochiy
- Department of Anesthesiology & Perioperative Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Karen L. de Mesy Bentley
- Department of Pathology & Laboratory Medicine and the Electron Microscope Resource, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Ethan D. Cohen
- Department of Pediatrics, Division of Cardiology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Paul S. Brookes
- Department of Anesthesiology & Perioperative Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - George A. Porter
- Department of Pediatrics, Division of Cardiology, University of Rochester Medical Center, Rochester, NY 14642, USA
- Departments of Medicine (Aab Cardiovascular Research Institute) and Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
22
|
Irerhievwie O, Ichipi-Ifukor PC, Asagba SO. Hepatocellular degeneration in mice co-exposed to in-utero aluminium and cadmium: Implication of a disordered antioxidant and energy homeostatic response in the liver. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2024; 106:104375. [PMID: 38262495 DOI: 10.1016/j.etap.2024.104375] [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: 11/05/2023] [Revised: 12/08/2023] [Accepted: 01/17/2024] [Indexed: 01/25/2024]
Abstract
The current study comprised four groups of pregnant animals viz; Control (CTR) received 10 ml/kg of normal saline, Al:10 mg/kg of AlCl3, Cd: 1.5 mg/Kg of CdCl2 Al+Cd; 10 mg/kg of AlCl3 and 1.5 mg/Kg of CdCl2. Treatment was done from pregnancy days (PNT) 7-20. After delivery, male animals were weaned on PSD 21 and sacrificed on PSD 78. From the study significant increases on serum liver enzymes in the group exposed to Cd and that exposed to Al+Cd were observed. The study further showed altered serum and hepatic antioxidant balance for the Cd, Al and Al+Cd groups compared to control. Similarly, lactate dehydrogenase (LDH) and succinate dehydrogenase (SDH) activities in the liver were elevated in Cd and Al+Cd groups while an altered liver histological feature in treated groups were also observed. it was concluded that in utero co-exposure to Al and Cd had the ability to alter hepatic functional indices.
Collapse
|
23
|
Nakano H, Nakano A. The role of metabolism in cardiac development. Curr Top Dev Biol 2024; 156:201-243. [PMID: 38556424 DOI: 10.1016/bs.ctdb.2024.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
Metabolism is the fundamental process that sustains life. The heart, in particular, is an organ of high energy demand, and its energy substrates have been studied for more than a century. In recent years, there has been a growing interest in understanding the role of metabolism in the early differentiation of pluripotent stem cells and in cancer research. Studies have revealed that metabolic intermediates from glycolysis and the tricarboxylic acid cycle act as co-factors for intracellular signal transduction, playing crucial roles in regulating cell behaviors. Mitochondria, as the central hub of metabolism, are also under intensive investigation regarding the regulation of their dynamics. The metabolic environment of the fetus is intricately linked to the maternal metabolic status, and the impact of the mother's nutrition and metabolic health on fetal development is significant. For instance, it is well known that maternal diabetes increases the risk of cardiac and nervous system malformations in the fetus. Another notable example is the decrease in the risk of neural tube defects when pregnant women are supplemented with folic acid. These examples highlight the profound influence of the maternal metabolic environment on the fetal organ development program. Therefore, gaining insights into the metabolic environment within developing fetal organs is critical for deepening our understanding of normal organ development. This review aims to summarize recent findings that build upon the historical recognition of the environmental and metabolic factors involved in the developing embryo.
Collapse
Affiliation(s)
- Haruko Nakano
- Department of Molecular, Cell, and Developmental Biology, UCLA, Los Angeles, CA, United States
| | - Atsushi Nakano
- Department of Molecular, Cell, and Developmental Biology, UCLA, Los Angeles, CA, United States; Cardiology Division, Department of Medicine, UCLA, Los Angeles, CA, United States; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, CA, United States; Molecular Biology Institute, UCLA, Los Angeles, CA, United States; Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan.
| |
Collapse
|
24
|
Ashok S, Raji SR, Manjunatha S, Srinivas G. Impairment of substrate-mediated mitochondrial respiration in cardiac cells by chloroquine. Mol Cell Biochem 2024; 479:373-382. [PMID: 37074504 PMCID: PMC10113731 DOI: 10.1007/s11010-023-04740-0] [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: 02/10/2023] [Accepted: 04/09/2023] [Indexed: 04/20/2023]
Abstract
Chloroquine (CQ) has a long clinical history as an anti-malarial agent and also being used for the treatment of other infections and autoimmune diseases. Recently, this lysosomotropic agent and its derivatives are also been tested as adjuncts alongside conventional anti-cancer treatments in combinatorial therapies. However, their reported cardiotoxicity tends to raise concern over their indiscriminate use. Even though the influence of CQ and its derivatives on cardiac mitochondria is extensively studied in disease models, their impact on cardiac mitochondrial respiration under physiological conditions remains inconclusive. In this study, we aimed to evaluate the impact of CQ on cardiac mitochondrial respiration using both in-vitro and in-vivo model systems. Using high-resolution respirometry in isolated cardiac mitochondria from male C57BL/6 mice treated with intraperitoneal injection of 10 mg/kg/day of CQ for 14 days, CQ was found to impair substrate-mediated mitochondrial respiration in cardiac tissue. In an in-vitro model of H9C2 cardiomyoblasts, incubation with 50 µM of CQ for 24 h disrupted mitochondrial membrane potential, produced mitochondrial fragmentation, decreased mitochondrial respiration and induced superoxide generation. Altogether, our study results indicate that CQ has a deleterious impact on cardiac mitochondrial bioenergetics which in turn suggests that CQ treatment could be an added burden, especially in patients affected with diseases with underlying cardiac complications. As CQ is an inhibitor of the lysosomal pathway, the observed effect could be an outcome of the accumulation of dysfunctional mitochondria due to autophagy inhibition.
Collapse
Affiliation(s)
- Sivasailam Ashok
- Department of Biochemistry, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala, 695011, India
| | - Sasikala Rajendran Raji
- Department of Biochemistry, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala, 695011, India
| | - Shankarappa Manjunatha
- Dr B C Roy Multispeciality Medical Research Centre, Indian Institute of Technology, Kharagpur, Kharagpur, 721302, India.
| | - Gopala Srinivas
- Department of Biochemistry, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala, 695011, India.
| |
Collapse
|
25
|
Ravasz D, Bui D, Nazarian S, Pallag G, Karnok N, Roberts J, Marzullo BP, Tennant DA, Greenwood B, Kitayev A, Hill C, Komlódi T, Doerrier C, Cunatova K, Fernandez-Vizarra E, Gnaiger E, Kiebish MA, Raska A, Kolev K, Czumbel B, Narain NR, Seyfried TN, Chinopoulos C. Residual Complex I activity and amphidirectional Complex II operation support glutamate catabolism through mtSLP in anoxia. Sci Rep 2024; 14:1729. [PMID: 38242919 PMCID: PMC10798963 DOI: 10.1038/s41598-024-51365-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 01/04/2024] [Indexed: 01/21/2024] Open
Abstract
Anoxia halts oxidative phosphorylation (OXPHOS) causing an accumulation of reduced compounds in the mitochondrial matrix which impedes dehydrogenases. By simultaneously measuring oxygen concentration, NADH autofluorescence, mitochondrial membrane potential and ubiquinone reduction extent in isolated mitochondria in real-time, we demonstrate that Complex I utilized endogenous quinones to oxidize NADH under acute anoxia. 13C metabolic tracing or untargeted analysis of metabolites extracted during anoxia in the presence or absence of site-specific inhibitors of the electron transfer system showed that NAD+ regenerated by Complex I is reduced by the 2-oxoglutarate dehydrogenase Complex yielding succinyl-CoA supporting mitochondrial substrate-level phosphorylation (mtSLP), releasing succinate. Complex II operated amphidirectionally during the anoxic event, providing quinones to Complex I and reducing fumarate to succinate. Our results highlight the importance of quinone provision to Complex I oxidizing NADH maintaining glutamate catabolism and mtSLP in the absence of OXPHOS.
Collapse
Affiliation(s)
- Dora Ravasz
- Department of Biochemistry, Semmelweis University, Budapest, 1094, Hungary
| | - David Bui
- Department of Biochemistry, Semmelweis University, Budapest, 1094, Hungary
| | - Sara Nazarian
- Department of Biochemistry, Semmelweis University, Budapest, 1094, Hungary
| | - Gergely Pallag
- Department of Biochemistry, Semmelweis University, Budapest, 1094, Hungary
| | - Noemi Karnok
- Department of Biochemistry, Semmelweis University, Budapest, 1094, Hungary
| | - Jennie Roberts
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Bryan P Marzullo
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Daniel A Tennant
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | | | | | | | - Timea Komlódi
- Department of Biochemistry, Semmelweis University, Budapest, 1094, Hungary
- Oroboros Instruments, Innsbruck, Austria
| | | | - Kristyna Cunatova
- Department of Biomedical Sciences, University of Padova, 35131, Padova, Italy
| | | | | | | | - Alexandra Raska
- Department of Biochemistry, Semmelweis University, Budapest, 1094, Hungary
| | - Krasimir Kolev
- Department of Biochemistry, Semmelweis University, Budapest, 1094, Hungary
| | - Bence Czumbel
- Department of Biochemistry, Semmelweis University, Budapest, 1094, Hungary
| | | | - Thomas N Seyfried
- Biology Department, Boston College, Chestnut Hill, Boston, MA, 02467, USA
| | | |
Collapse
|
26
|
Szabo E, Nagy B, Czajlik A, Komlodi T, Ozohanics O, Tretter L, Ambrus A. Mitochondrial Alpha-Keto Acid Dehydrogenase Complexes: Recent Developments on Structure and Function in Health and Disease. Subcell Biochem 2024; 104:295-381. [PMID: 38963492 DOI: 10.1007/978-3-031-58843-3_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
The present work delves into the enigmatic world of mitochondrial alpha-keto acid dehydrogenase complexes discussing their metabolic significance, enzymatic operation, moonlighting activities, and pathological relevance with links to underlying structural features. This ubiquitous family of related but diverse multienzyme complexes is involved in carbohydrate metabolism (pyruvate dehydrogenase complex), the citric acid cycle (α-ketoglutarate dehydrogenase complex), and amino acid catabolism (branched-chain α-keto acid dehydrogenase complex, α-ketoadipate dehydrogenase complex); the complexes all function at strategic points and also participate in regulation in these metabolic pathways. These systems are among the largest multienzyme complexes with at times more than 100 protein chains and weights ranging up to ~10 million Daltons. Our chapter offers a wealth of up-to-date information on these multienzyme complexes for a comprehensive understanding of their significance in health and disease.
Collapse
Affiliation(s)
- Eszter Szabo
- Department of Biochemistry, Semmelweis University, Budapest, Hungary
| | - Balint Nagy
- Department of Biochemistry, Semmelweis University, Budapest, Hungary
| | - Andras Czajlik
- Department of Biochemistry, Semmelweis University, Budapest, Hungary
| | - Timea Komlodi
- Department of Biochemistry, Semmelweis University, Budapest, Hungary
| | - Oliver Ozohanics
- Department of Biochemistry, Semmelweis University, Budapest, Hungary
| | - Laszlo Tretter
- Department of Biochemistry, Semmelweis University, Budapest, Hungary
| | - Attila Ambrus
- Department of Biochemistry, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
27
|
Edosuyi O, Igbe I, Oyekan A. Fumarate and its downstream signalling pathways in the cardiorenal system: Recent insights and novel expositions in the etiology of hypertension. Eur J Pharmacol 2023; 961:176186. [PMID: 37944846 PMCID: PMC10843741 DOI: 10.1016/j.ejphar.2023.176186] [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: 08/03/2023] [Revised: 11/02/2023] [Accepted: 11/03/2023] [Indexed: 11/12/2023]
Abstract
Hypertension, a risk factor for cardiorenal disease has a huge global health impact. Hence, there is a continuous search for new therapeutic targets and putative antihypertensive ligands. This search has transcended into the realm of mitochondrial metabolism which has been reported to underline the etiology of certain diseases, including hypertension. Recently, genetic alterations in the tricarboxylic acid (TCA) cycle enzyme, fumarase, which converts fumarate to malate, reportedly worsened salt-sensitive hypertension. These novel expositions shifted focus into the activity of TCA in the pathogenesis of hypertension. There is now evidence to show that a mechanistic link exists between blood pressure regulation and intermediaries in the TCA cycle involving fumarate metabolism. Fumarate has been reported to mediate the actions of endogenous ligands such as nitric oxide (NO), and hypoxia inducible factor (HIF)-1α. Similarly, there has been upregulation of protective genes such as nuclear erythroid factor 2 (Nrf2) and reduction in the expression of certain markers like kidney injury molecule 1 (KIM-1). There are reports of interactions with endogenous enzymes such as catalase (CAT) and renin via the activation of GPR91. Fumarate has also been shown to modulate the actions of renal ion channels and by extension, natriuresis. These actions of fumarate have conferred a reno- and cardio-protective effect in hypertension. This review evaluates the role of the TCA cycle, its mechanistic links, and significant contribution to blood pressure regulation with a view to understanding the possibility of a new pathological axis which may be involved in the pathogenesis of hypertension.
Collapse
Affiliation(s)
- Osaze Edosuyi
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, University of Benin, PMB 1154, Benin City, Nigeria; Center for Cardiovascular Diseases, Gray Hall Suites, Rm 256, College of Pharmacy & Health Sciences, Texas Southern University, 3100, Cleburne Street, Houston, TX, USA.
| | - Ighodaro Igbe
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, University of Benin, PMB 1154, Benin City, Nigeria
| | - Adebayo Oyekan
- Center for Cardiovascular Diseases, Gray Hall Suites, Rm 256, College of Pharmacy & Health Sciences, Texas Southern University, 3100, Cleburne Street, Houston, TX, USA
| |
Collapse
|
28
|
Szibor M, Mühlon M, Doenst T, Pohjoismäki JLO. Spatial adjustment of bioenergetics, a possible determinant of contractile adaptation and development of contractile failure. FRONTIERS IN MOLECULAR MEDICINE 2023; 3:1305960. [PMID: 39086691 PMCID: PMC11285667 DOI: 10.3389/fmmed.2023.1305960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 11/23/2023] [Indexed: 08/02/2024]
Abstract
Cardiomyocytes depend on mitochondrial oxidative phosphorylation (OXPHOS) for energy metabolism, which is facilitated by the mitochondrial electron transfer system (ETS). In a series of thermogenic redox reactions, electrons are shuttled through the ETS to oxygen as the final electron acceptor. This electron transfer is coupled to proton translocation across the inner mitochondrial membrane, which itself is the main driving force for ATP production. Oxygen availability is thus a prerequisite for ATP production and consequently contractility. Notably, cardiomyocytes are exceptionally large cells and densely packed with contractile structures, which constrains intracellular oxygen distribution. Moreover, oxygen must pass through layers of actively respiring mitochondria to reach the ones located in the innermost contractile compartment. Indeed, uneven oxygen distribution was observed in cardiomyocytes, suggesting that local ATP supply may also vary according to oxygen availability. Here, we discuss how spatial adjustment of bioenergetics to intracellular oxygen fluctuations may underlie cardiac contractile adaptation and how this adaptation may pose a risk for the development of contractile failure.
Collapse
Affiliation(s)
- Marten Szibor
- Department of Cardiothoracic Surgery, Center for Sepsis Control and Care (CSCC), Jena University Hospital, Friedrich-Schiller University of Jena, Jena, Germany
- BioMediTech and Tampere University Hospital, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Marie Mühlon
- Department of Cardiothoracic Surgery, Center for Sepsis Control and Care (CSCC), Jena University Hospital, Friedrich-Schiller University of Jena, Jena, Germany
| | - Torsten Doenst
- Department of Cardiothoracic Surgery, Center for Sepsis Control and Care (CSCC), Jena University Hospital, Friedrich-Schiller University of Jena, Jena, Germany
| | - Jaakko L. O. Pohjoismäki
- Department of Environmental and Biological Sciences, University of Eastern Finland, Joensuu, Finland
| |
Collapse
|
29
|
Fukushima CT, Dancil IS, Clary H, Shah N, Nadtochiy SM, Brookes PS. Reactive Oxygen Species Generation by Reverse Electron Transfer at Mitochondrial Complex I Under Simulated Early Reperfusion Conditions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.21.568136. [PMID: 38045326 PMCID: PMC10690194 DOI: 10.1101/2023.11.21.568136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Ischemic tissues accumulate succinate, which is rapidly oxidized upon reperfusion, driving a burst of mitochondrial reactive oxygen species (ROS) generation that triggers cell death. In isolated mitochondria with succinate as the sole metabolic substrate under non-phosphorylating conditions, 90% of ROS generation is from reverse electron transfer (RET) at the Q site of respiratory complex I (Cx-I). Together, these observations suggest Cx-I RET is the source of pathologic ROS in reperfusion injury. However, numerous factors present in early reperfusion may impact Cx-I RET, including: (i) High [NADH]; (ii) High [lactate]; (iii) Mildly acidic pH; (iv) Defined ATP/ADP ratios; (v) Presence of the nucleosides adenosine and inosine; and (vi) Defined free [Ca2+]. Herein, experiments with mouse cardiac mitochondria revealed that under simulated early reperfusion conditions including these factors, overall mitochondrial ROS generation was only 56% of that seen with succinate alone, and only 52% of this ROS was assignable to Cx-I RET. The residual non-RET ROS could be partially assigned to complex III (Cx-III) with the remainder likely originating from other ROS sources upstream of the Cx-I Q site. Together, these data suggest the relative contribution of Cx-I RET ROS to reperfusion injury may be overestimated, and other ROS sources may contribute a significant fraction of ROS in early reperfusion.
Collapse
Affiliation(s)
- Caio Tabata Fukushima
- Department of Anesthesiology, University of Rochester Medical Center
- Department of Biochemistry, University of Rochester Medical Center
- Department of Pharmacology and Physiology, University of Rochester Medical Center
| | - Ian-Shika Dancil
- Department of Anesthesiology, University of Rochester Medical Center
| | - Hannah Clary
- Department of Biochemistry, University of Rochester Medical Center
| | - Nidhi Shah
- Department of Pharmacology and Physiology, University of Rochester Medical Center
| | | | - Paul S. Brookes
- Department of Anesthesiology, University of Rochester Medical Center
- Department of Pharmacology and Physiology, University of Rochester Medical Center
| |
Collapse
|
30
|
Heusch G, Andreadou I, Bell R, Bertero E, Botker HE, Davidson SM, Downey J, Eaton P, Ferdinandy P, Gersh BJ, Giacca M, Hausenloy DJ, Ibanez B, Krieg T, Maack C, Schulz R, Sellke F, Shah AM, Thiele H, Yellon DM, Di Lisa F. Health position paper and redox perspectives on reactive oxygen species as signals and targets of cardioprotection. Redox Biol 2023; 67:102894. [PMID: 37839355 PMCID: PMC10590874 DOI: 10.1016/j.redox.2023.102894] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/04/2023] [Accepted: 09/15/2023] [Indexed: 10/17/2023] Open
Abstract
The present review summarizes the beneficial and detrimental roles of reactive oxygen species in myocardial ischemia/reperfusion injury and cardioprotection. In the first part, the continued need for cardioprotection beyond that by rapid reperfusion of acute myocardial infarction is emphasized. Then, pathomechanisms of myocardial ischemia/reperfusion to the myocardium and the coronary circulation and the different modes of cell death in myocardial infarction are characterized. Different mechanical and pharmacological interventions to protect the ischemic/reperfused myocardium in elective percutaneous coronary interventions and coronary artery bypass grafting, in acute myocardial infarction and in cardiotoxicity from cancer therapy are detailed. The second part keeps the focus on ROS providing a comprehensive overview of molecular and cellular mechanisms involved in ischemia/reperfusion injury. Starting from mitochondria as the main sources and targets of ROS in ischemic/reperfused myocardium, a complex network of cellular and extracellular processes is discussed, including relationships with Ca2+ homeostasis, thiol group redox balance, hydrogen sulfide modulation, cross-talk with NAPDH oxidases, exosomes, cytokines and growth factors. While mechanistic insights are needed to improve our current therapeutic approaches, advancements in knowledge of ROS-mediated processes indicate that detrimental facets of oxidative stress are opposed by ROS requirement for physiological and protective reactions. This inevitable contrast is likely to underlie unsuccessful clinical trials and limits the development of novel cardioprotective interventions simply based upon ROS removal.
Collapse
Affiliation(s)
- Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Duisburg-Essen, Essen, Germany.
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Robert Bell
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - Edoardo Bertero
- Chair of Cardiovascular Disease, Department of Internal Medicine and Specialties, University of Genova, Genova, Italy
| | - Hans-Erik Botker
- Department of Cardiology, Institute for Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - James Downey
- Department of Physiology, University of South Alabama, Mobile, AL, USA
| | - Philip Eaton
- William Harvey Research Institute, Queen Mary University of London, Heart Centre, Charterhouse Square, London, United Kingdom
| | - Peter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Bernard J Gersh
- Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Mauro Giacca
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College, London, United Kingdom
| | - Derek J Hausenloy
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom; Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, National Heart Research Institute Singapore, National Heart Centre, Yong Loo Lin School of Medicine, National University Singapore, Singapore
| | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), IIS-Fundación Jiménez Díaz University Hospital, and CIBERCV, Madrid, Spain
| | - Thomas Krieg
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center, University Clinic Würzburg, Würzburg, Germany
| | - Rainer Schulz
- Institute for Physiology, Justus-Liebig -Universität, Giessen, Germany
| | - Frank Sellke
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI, USA
| | - Ajay M Shah
- King's College London British Heart Foundation Centre of Excellence, London, United Kingdom
| | - Holger Thiele
- Heart Center Leipzig at University of Leipzig and Leipzig Heart Science, Leipzig, Germany
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - Fabio Di Lisa
- Dipartimento di Scienze Biomediche, Università degli studi di Padova, Padova, Italy.
| |
Collapse
|
31
|
Okoye CN, Koren SA, Wojtovich AP. Mitochondrial complex I ROS production and redox signaling in hypoxia. Redox Biol 2023; 67:102926. [PMID: 37871533 PMCID: PMC10598411 DOI: 10.1016/j.redox.2023.102926] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/29/2023] [Accepted: 10/06/2023] [Indexed: 10/25/2023] Open
Abstract
Mitochondria are a main source of cellular energy. Oxidative phosphorylation (OXPHOS) is the major process of aerobic respiration. Enzyme complexes of the electron transport chain (ETC) pump protons to generate a protonmotive force (Δp) that drives OXPHOS. Complex I is an electron entry point into the ETC. Complex I oxidizes nicotinamide adenine dinucleotide (NADH) and transfers electrons to ubiquinone in a reaction coupled with proton pumping. Complex I also produces reactive oxygen species (ROS) under various conditions. The enzymatic activities of complex I can be regulated by metabolic conditions and serves as a regulatory node of the ETC. Complex I ROS plays diverse roles in cell metabolism ranging from physiologic to pathologic conditions. Progress in our understanding indicates that ROS release from complex I serves important signaling functions. Increasing evidence suggests that complex I ROS is important in signaling a mismatch in energy production and demand. In this article, we review the role of ROS from complex I in sensing acute hypoxia.
Collapse
Affiliation(s)
- Chidozie N Okoye
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Shon A Koren
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Andrew P Wojtovich
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA; Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, 14642, USA.
| |
Collapse
|
32
|
Tokarska-Schlattner M, Zeaiter N, Cunin V, Attia S, Meunier C, Kay L, Achouri A, Hiriart-Bryant E, Couturier K, Tellier C, El Harras A, Elena-Herrmann B, Khochbin S, Le Gouellec A, Schlattner U. Multi-Method Quantification of Acetyl-Coenzyme A and Further Acyl-Coenzyme A Species in Normal and Ischemic Rat Liver. Int J Mol Sci 2023; 24:14957. [PMID: 37834405 PMCID: PMC10573920 DOI: 10.3390/ijms241914957] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 09/29/2023] [Accepted: 09/30/2023] [Indexed: 10/15/2023] Open
Abstract
Thioesters of coenzyme A (CoA) carrying different acyl chains (acyl-CoAs) are central intermediates of many metabolic pathways and donor molecules for protein lysine acylation. Acyl-CoA species largely differ in terms of cellular concentrations and physico-chemical properties, rendering their analysis challenging. Here, we compare several approaches to quantify cellular acyl-CoA concentrations in normal and ischemic rat liver, using HPLC and LC-MS/MS for multi-acyl-CoA analysis, as well as NMR, fluorimetric and spectrophotometric techniques for the quantification of acetyl-CoAs. In particular, we describe a simple LC-MS/MS protocol that is suitable for the relative quantification of short and medium-chain acyl-CoA species. We show that ischemia induces specific changes in the short-chain acyl-CoA relative concentrations, while mild ischemia (1-2 min), although reducing succinyl-CoA, has little effects on acetyl-CoA, and even increases some acyl-CoA species upstream of the tricarboxylic acid cycle. In contrast, advanced ischemia (5-6 min) also reduces acetyl-CoA levels. Our approach provides the keys to accessing the acyl-CoA metabolome for a more in-depth analysis of metabolism, protein acylation and epigenetics.
Collapse
Affiliation(s)
- Malgorzata Tokarska-Schlattner
- University Grenoble Alpes, Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), 38058 Grenoble, France; (N.Z.); (S.A.); (L.K.); (A.A.); (E.H.-B.); (K.C.); (C.T.)
| | - Nour Zeaiter
- University Grenoble Alpes, Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), 38058 Grenoble, France; (N.Z.); (S.A.); (L.K.); (A.A.); (E.H.-B.); (K.C.); (C.T.)
| | - Valérie Cunin
- University Grenoble Alpes, CNRS UMR 5525, Laboratory TIMC—Translational Microbiology, Evolution, Engineering (TREE), Service de Biochimie, Biologie Moléculaire et Toxicologie Environnementale, CHU Grenoble-Alpes, 38058 Grenoble, France; (V.C.); (C.M.); (A.L.G.)
| | - Stéphane Attia
- University Grenoble Alpes, Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), 38058 Grenoble, France; (N.Z.); (S.A.); (L.K.); (A.A.); (E.H.-B.); (K.C.); (C.T.)
| | - Cécile Meunier
- University Grenoble Alpes, CNRS UMR 5525, Laboratory TIMC—Translational Microbiology, Evolution, Engineering (TREE), Service de Biochimie, Biologie Moléculaire et Toxicologie Environnementale, CHU Grenoble-Alpes, 38058 Grenoble, France; (V.C.); (C.M.); (A.L.G.)
| | - Laurence Kay
- University Grenoble Alpes, Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), 38058 Grenoble, France; (N.Z.); (S.A.); (L.K.); (A.A.); (E.H.-B.); (K.C.); (C.T.)
| | - Amel Achouri
- University Grenoble Alpes, Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), 38058 Grenoble, France; (N.Z.); (S.A.); (L.K.); (A.A.); (E.H.-B.); (K.C.); (C.T.)
| | - Edwige Hiriart-Bryant
- University Grenoble Alpes, Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), 38058 Grenoble, France; (N.Z.); (S.A.); (L.K.); (A.A.); (E.H.-B.); (K.C.); (C.T.)
| | - Karine Couturier
- University Grenoble Alpes, Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), 38058 Grenoble, France; (N.Z.); (S.A.); (L.K.); (A.A.); (E.H.-B.); (K.C.); (C.T.)
| | - Cindy Tellier
- University Grenoble Alpes, Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), 38058 Grenoble, France; (N.Z.); (S.A.); (L.K.); (A.A.); (E.H.-B.); (K.C.); (C.T.)
| | - Abderrafek El Harras
- University Grenoble Alpes, Inserm U1209 and CNRS UMR5309, Institute for Advanced Biosciences (IAB), 38058 Grenoble, France; (A.E.H.); (B.E.-H.); (S.K.)
| | - Bénédicte Elena-Herrmann
- University Grenoble Alpes, Inserm U1209 and CNRS UMR5309, Institute for Advanced Biosciences (IAB), 38058 Grenoble, France; (A.E.H.); (B.E.-H.); (S.K.)
| | - Saadi Khochbin
- University Grenoble Alpes, Inserm U1209 and CNRS UMR5309, Institute for Advanced Biosciences (IAB), 38058 Grenoble, France; (A.E.H.); (B.E.-H.); (S.K.)
| | - Audrey Le Gouellec
- University Grenoble Alpes, CNRS UMR 5525, Laboratory TIMC—Translational Microbiology, Evolution, Engineering (TREE), Service de Biochimie, Biologie Moléculaire et Toxicologie Environnementale, CHU Grenoble-Alpes, 38058 Grenoble, France; (V.C.); (C.M.); (A.L.G.)
| | - Uwe Schlattner
- University Grenoble Alpes, Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), 38058 Grenoble, France; (N.Z.); (S.A.); (L.K.); (A.A.); (E.H.-B.); (K.C.); (C.T.)
- Institut Universitaire de France (IUF), 75231 Paris, France
| |
Collapse
|
33
|
HENG X, WANG Z, YANG L, LI L, HUANG S. Dangua Fang regulating tricarboxylic acid cycle and respiratory chain and its mechanism in diabetic rats. J TRADIT CHIN MED 2023; 43:1150-1159. [PMID: 37946477 PMCID: PMC10623262 DOI: 10.19852/j.cnki.jtcm.20230904.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 09/21/2022] [Indexed: 11/12/2023]
Abstract
OBJECTIVE To investigate the influence and possible targets of Dangua Fang on tricarboxylic acid (TCA) cycle and respiratory chain to enrich the prescription's mechanism of effective intervention on glycolipid metabolic diseases such as type 2 diabetes. METHODS After interventional rats were fed with high glucose and high fat diet ad libitum for 4 weeks, intraperitoneally injected streptozotocin to induce diabetic model. According to blood glucose level,28 diabetic rats were selected and continued to be fed with high glucose and high fat diet, were stratified by body weight, and divided randomly by blood glucose into Model group (was given sterile water by gastric perfusion and injected aquae pro injection intraperitoneally), Dangua group [Dangua liquor 20.5 g·kg-1·d-1 by perfusion and aquae pro injection intraperitoneally], Inhibitor group [sterile water by perfusion and nicotinamide phosphoribosyl transferase (Nampt) specific blocker GEN-617 1.25 mg/kg intraperitoneally], DanInhit group (Dangua liquor and GEN-617 synchronously). Control group were continuously fed with ordinary diet. The intervention was last for 10 weeks. Body weight (BW), liver index (LI), glycosylated hemoglobin (HbA1c), TC, TG, free fatty acids (FFA), creatinine (Cr), and A-ketoglutarate (α-KG), Iso-citric acid (ICA), oxaloacetic acid (OAA) were tested. The cytochrome C oxidase (COX) and Succinate dehydrogenase (SDH) were evaluated by Colorimetry; Nampt protein, Adenosine triphosphate (ATP) synthase (ATPs), Nicotinamide adenine dinucleotide (NAD+)and its reduced (NADH) in liver were measured by enzyme linked immunosorbent assay. The expressions of Nampt and mitochondrialnadhdehydrogenase-1 (mt-ND1) gene in liver was assessed by real-time polymerase chain reaction. Hepatic tissue staining was also completed. RESULTS The levels of BW, ICA, α-KG and Nampt-mRNA in the Model group are lower than that in the Normal group (P < 0.05), conversely, liver weight, LI, TC, HbA1c, SDH and ATPs, mt-ND1-mRNA, and Nampt protein in the Model group are higher (P < 0.01, P < 0.05). Compared with Model group, the levels of ICA, Nampt-mRNA and Nampt in Dangua group are significantly increased, and FFA obviously raised (P < 0.01 and P < 0.05); liver weight, BW, SDH are obviously lower, and HbA1c decreased significantly (P < 0.01, P < 0.05). TG, FFA and Nampt protein increased in the DanInhit group, TC, TG, BW obviously increased in the Inhibitor group, but SDH is decreased in both the two groups (P < 0.05, P < 0.01). Compared with Dangua group, DanInhib group has the lower levels of ICA, mt-ND1-mRNA, Nampt-mRNA, and the higher level of BW, LI and HbA1c. In the Inhibitor group, ICA and Nampt protein decreased, BW and LI, HbA1c and TG increased (P < 0.01 or P < 0.05). Tissue staining display that, in the model group there is obvious pathologic changes ie: fibrosis, steatosis and inflammatory cell infiltration. Lesions in the Dangua group are mild, and those of Inhibitor group are more obvious than the Model group, and DanInhit group is intermediately affected compared to Dangua group and Inhibitor group. CONCLUSION Dangua Fang increases the metabolic flux of TCA cycle and optimizes respiratory chain function by up-regulating Nampt expression.
Collapse
Affiliation(s)
- Xianpei HENG
- 1 Department of Endocrinology, People's Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou 350004, China
| | - Zhita WANG
- 1 Department of Endocrinology, People's Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou 350004, China
| | - Liuqing YANG
- 1 Department of Endocrinology, People's Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou 350004, China
| | - Liang LI
- 1 Department of Endocrinology, People's Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou 350004, China
| | - Suping HUANG
- 2 Academy of Integrative Medicine Fujian, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| |
Collapse
|
34
|
Oh CJ, Kim MJ, Lee JM, Kim DH, Kim IY, Park S, Kim Y, Lee KB, Lee SH, Lim CW, Kim M, Lee JY, Pagire HS, Pagire SH, Bae MA, Chanda D, Thoudam T, Khang AR, Harris RA, Ahn JH, Jeon JH, Lee IK. Inhibition of pyruvate dehydrogenase kinase 4 ameliorates kidney ischemia-reperfusion injury by reducing succinate accumulation during ischemia and preserving mitochondrial function during reperfusion. Kidney Int 2023; 104:724-739. [PMID: 37399974 DOI: 10.1016/j.kint.2023.06.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 05/26/2023] [Accepted: 06/02/2023] [Indexed: 07/05/2023]
Abstract
Ischemia-reperfusion (IR) injury, a leading cause of acute kidney injury (AKI), is still without effective therapies. Succinate accumulation during ischemia followed by its oxidation during reperfusion leads to excessive reactive oxygen species (ROS) and severe kidney damage. Consequently, the targeting of succinate accumulation may represent a rational approach to the prevention of IR-induced kidney injury. Since ROS are generated primarily in mitochondria, which are abundant in the proximal tubule of the kidney, we explored the role of pyruvate dehydrogenase kinase 4 (PDK4), a mitochondrial enzyme, in IR-induced kidney injury using proximal tubule cell-specific Pdk4 knockout (Pdk4ptKO) mice. Knockout or pharmacological inhibition of PDK4 ameliorated IR-induced kidney damage. Succinate accumulation during ischemia, which is responsible for mitochondrial ROS production during reperfusion, was reduced by PDK4 inhibition. PDK4 deficiency established conditions prior to ischemia resulting in less succinate accumulation, possibly because of a reduction in electron flow reversal in complex II, which provides electrons for the reduction of fumarate to succinate by succinate dehydrogenase during ischemia. The administration of dimethyl succinate, a cell-permeable form of succinate, attenuated the beneficial effects of PDK4 deficiency, suggesting that the kidney-protective effect is succinate-dependent. Finally, genetic or pharmacological inhibition of PDK4 prevented IR-induced mitochondrial damage in mice and normalized mitochondrial function in an in vitro model of IR injury. Thus, inhibition of PDK4 represents a novel means of preventing IR-induced kidney injury, and involves the inhibition of ROS-induced kidney toxicity through reduction in succinate accumulation and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Chang Joo Oh
- Research Institute of Aging and Metabolism, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Min-Ji Kim
- Department of Internal Medicine, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Ji-Min Lee
- Cell & Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Dong Hun Kim
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu, Republic of Korea
| | - Il-Young Kim
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon, Republic of Korea; Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Sanghee Park
- Department of Exercise Rehabilitation, Gachon University, Incheon, Republic of Korea
| | - Yeongmin Kim
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea
| | - Kyung-Bok Lee
- Center for Research Equipment (104-Dong), Korea Basic Science Institute, Ochang, Cheongju, Chungbuk, Republic of Korea
| | - Sang-Hee Lee
- Center for Research Equipment (104-Dong), Korea Basic Science Institute, Ochang, Cheongju, Chungbuk, Republic of Korea
| | - Chae Won Lim
- Department of Medicine, Graduate School, Daegu Catholic University, Gyeongsan, Gyeongbuk, Republic of Korea
| | - Myeongjin Kim
- Department of Medicine, Graduate School, Daegu Catholic University, Gyeongsan, Gyeongbuk, Republic of Korea
| | - Jung-Yi Lee
- Research Institute of Aging and Metabolism, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Haushabhau S Pagire
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Suvarna H Pagire
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Myung Ae Bae
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Dipanjan Chanda
- Research Institute of Aging and Metabolism, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Themis Thoudam
- Research Institute of Aging and Metabolism, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Ah Reum Khang
- Department of Internal Medicine, Pusan National University Yangsan Hospital, Pusan National University College of Medicine, Yangsan, Republic of Korea
| | - Robert A Harris
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Jin Hee Ahn
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea.
| | - Jae-Han Jeon
- Research Institute of Aging and Metabolism, Kyungpook National University School of Medicine, Daegu, Republic of Korea; Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea.
| | - In-Kyu Lee
- Research Institute of Aging and Metabolism, Kyungpook National University School of Medicine, Daegu, Republic of Korea; Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea.
| |
Collapse
|
35
|
Prag HA, Murphy MP, Krieg T. Preventing mitochondrial reverse electron transport as a strategy for cardioprotection. Basic Res Cardiol 2023; 118:34. [PMID: 37639068 PMCID: PMC10462584 DOI: 10.1007/s00395-023-01002-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/04/2023] [Accepted: 08/06/2023] [Indexed: 08/29/2023]
Abstract
In the context of myocardial infarction, the burst of superoxide generated by reverse electron transport (RET) at complex I in mitochondria is a crucial trigger for damage during ischaemia/reperfusion (I/R) injury. Here we outline the necessary conditions for superoxide production by RET at complex I and how it can occur during reperfusion. In addition, we explore various pathways that are implicated in generating the conditions for RET to occur and suggest potential therapeutic strategies to target RET, aiming to achieve cardioprotection.
Collapse
Affiliation(s)
- Hiran A Prag
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK.
| | - Michael P Murphy
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK.
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, CB2 0XY, UK.
| | - Thomas Krieg
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK.
| |
Collapse
|
36
|
Sánchez-Pérez P, Mata A, Torp MK, López-Bernardo E, Heiestad CM, Aronsen JM, Molina-Iracheta A, Jiménez-Borreguero LJ, García-Roves P, Costa ASH, Frezza C, Murphy MP, Stenslokken KO, Cadenas S. Energy substrate metabolism, mitochondrial structure and oxidative stress after cardiac ischemia-reperfusion in mice lacking UCP3. Free Radic Biol Med 2023; 205:244-261. [PMID: 37295539 DOI: 10.1016/j.freeradbiomed.2023.05.014] [Citation(s) in RCA: 3] [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: 03/19/2023] [Revised: 04/22/2023] [Accepted: 05/15/2023] [Indexed: 06/12/2023]
Abstract
Myocardial ischemia-reperfusion (IR) injury may result in cardiomyocyte dysfunction. Mitochondria play a critical role in cardiomyocyte recovery after IR injury. The mitochondrial uncoupling protein 3 (UCP3) has been proposed to reduce mitochondrial reactive oxygen species (ROS) production and to facilitate fatty acid oxidation. As both mechanisms might be protective following IR injury, we investigated functional, mitochondrial structural, and metabolic cardiac remodeling in wild-type mice and in mice lacking UCP3 (UCP3-KO) after IR. Results showed that infarct size in isolated perfused hearts subjected to IR ex vivo was larger in adult and old UCP3-KO mice than in equivalent wild-type mice, and was accompanied by higher levels of creatine kinase in the effluent and by more pronounced mitochondrial structural changes. The greater myocardial damage in UCP3-KO hearts was confirmed in vivo after coronary artery occlusion followed by reperfusion. S1QEL, a suppressor of superoxide generation from site IQ in complex I, limited infarct size in UCP3-KO hearts, pointing to exacerbated superoxide production as a possible cause of the damage. Metabolomics analysis of isolated perfused hearts confirmed the reported accumulation of succinate, xanthine and hypoxanthine during ischemia, and a shift to anaerobic glucose utilization, which all recovered upon reoxygenation. The metabolic response to ischemia and IR was similar in UCP3-KO and wild-type hearts, being lipid and energy metabolism the most affected pathways. Fatty acid oxidation and complex I (but not complex II) activity were equally impaired after IR. Overall, our results indicate that UCP3 deficiency promotes enhanced superoxide generation and mitochondrial structural changes that increase the vulnerability of the myocardium to IR injury.
Collapse
Affiliation(s)
- Patricia Sánchez-Pérez
- Centro de Biología Molecular "Severo Ochoa" (CSIC/UAM), 28049, Madrid, Spain; Instituto de Investigación Sanitaria Princesa (IIS-IP), 28006, Madrid, Spain
| | - Ana Mata
- Centro de Biología Molecular "Severo Ochoa" (CSIC/UAM), 28049, Madrid, Spain; Instituto de Investigación Sanitaria Princesa (IIS-IP), 28006, Madrid, Spain
| | - May-Kristin Torp
- Centro de Biología Molecular "Severo Ochoa" (CSIC/UAM), 28049, Madrid, Spain; Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, PB1110, N-0317, Oslo, Norway
| | - Elia López-Bernardo
- Centro de Biología Molecular "Severo Ochoa" (CSIC/UAM), 28049, Madrid, Spain; Instituto de Investigación Sanitaria Princesa (IIS-IP), 28006, Madrid, Spain
| | - Christina M Heiestad
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, PB1110, N-0317, Oslo, Norway
| | - Jan Magnus Aronsen
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, PB1110, N-0317, Oslo, Norway; Bjørknes College, 0456, Oslo, Norway
| | | | - Luis J Jiménez-Borreguero
- Instituto de Investigación Sanitaria Princesa (IIS-IP), 28006, Madrid, Spain; Servicio de Cardiología, Hospital Universitario de La Princesa, 28006, Madrid, Spain; Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Pablo García-Roves
- Department of Physiological Sciences, Universitat de Barcelona, 08907, Barcelona, Spain; Nutrition, Metabolism and Gene Therapy Group, Diabetes and Metabolism Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Ana S H Costa
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Center, Cambridge Biomedical Campus, Cambridge, CB2 0XZ, UK
| | - Christian Frezza
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Center, Cambridge Biomedical Campus, Cambridge, CB2 0XZ, UK
| | - Michael P Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge, Wellcome Trust/MRC Building, Cambridge, CB2 0XY, UK
| | - Kåre-Olav Stenslokken
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, PB1110, N-0317, Oslo, Norway
| | - Susana Cadenas
- Centro de Biología Molecular "Severo Ochoa" (CSIC/UAM), 28049, Madrid, Spain; Instituto de Investigación Sanitaria Princesa (IIS-IP), 28006, Madrid, Spain.
| |
Collapse
|
37
|
Huwyler F, Eden J, Binz J, Cunningham L, Sousa Da Silva RX, Clavien P, Dutkowski P, Tibbitt MW, Hefti M. A Spectrofluorometric Method for Real-Time Graft Assessment and Patient Monitoring. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301537. [PMID: 37265001 PMCID: PMC10427358 DOI: 10.1002/advs.202301537] [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: 03/09/2023] [Revised: 05/03/2023] [Indexed: 06/03/2023]
Abstract
Biomarkers are powerful clinical diagnostics and predictors of patient outcome. However, robust measurements often require time and expensive laboratory equipment, which is insufficient to track rapid changes and limits direct use in the operating room. Here, this study presents a portable spectrophotometric device for continuous real-time measurements of fluorescent and non-fluorescent biomarkers at the point of care. This study measures the mitochondrial damage biomarker flavin mononucleotide (FMN) in 26 extended criteria human liver grafts undergoing hypothermic oxygenated perfusion to guide clinical graft assessment. Real-time data identified seven organs unsuitable for transplant that are discarded. The remaining grafts are transplanted and FMN values correlated with post-transplant indicators of liver function and patient recovery. Further, this study shows how this device can be used to monitor dialysis patients by measuring creatinine in real-time. Our approach provides a simple method to monitor biomarkers directly within biological fluids to improve organ assessment, patient care, and biomarker discovery.
Collapse
Affiliation(s)
- Florian Huwyler
- Macromolecular Engineering Lab, Department of Mechanical and Process EngineeringETH ZurichZurich8092Switzerland
- Department of Surgery and Transplantation, Swiss Hepato‐Pancreato‐Biliary (HPB) and Transplant CenterUniversity Hospital ZurichZurich8091Switzerland
- Wyss Zurich Translational CenterETH Zurich and University of ZurichZurich8092Switzerland
| | - Janina Eden
- Department of Surgery and Transplantation, Swiss Hepato‐Pancreato‐Biliary (HPB) and Transplant CenterUniversity Hospital ZurichZurich8091Switzerland
| | - Jonas Binz
- Macromolecular Engineering Lab, Department of Mechanical and Process EngineeringETH ZurichZurich8092Switzerland
| | - Leslie Cunningham
- Macromolecular Engineering Lab, Department of Mechanical and Process EngineeringETH ZurichZurich8092Switzerland
- Department of Surgery and Transplantation, Swiss Hepato‐Pancreato‐Biliary (HPB) and Transplant CenterUniversity Hospital ZurichZurich8091Switzerland
- Wyss Zurich Translational CenterETH Zurich and University of ZurichZurich8092Switzerland
| | - Richard X. Sousa Da Silva
- Department of Surgery and Transplantation, Swiss Hepato‐Pancreato‐Biliary (HPB) and Transplant CenterUniversity Hospital ZurichZurich8091Switzerland
- Wyss Zurich Translational CenterETH Zurich and University of ZurichZurich8092Switzerland
| | - Pierre‐Alain Clavien
- Department of Surgery and Transplantation, Swiss Hepato‐Pancreato‐Biliary (HPB) and Transplant CenterUniversity Hospital ZurichZurich8091Switzerland
- Wyss Zurich Translational CenterETH Zurich and University of ZurichZurich8092Switzerland
| | - Philipp Dutkowski
- Department of Surgery and Transplantation, Swiss Hepato‐Pancreato‐Biliary (HPB) and Transplant CenterUniversity Hospital ZurichZurich8091Switzerland
| | - Mark W. Tibbitt
- Macromolecular Engineering Lab, Department of Mechanical and Process EngineeringETH ZurichZurich8092Switzerland
- Wyss Zurich Translational CenterETH Zurich and University of ZurichZurich8092Switzerland
| | - Max Hefti
- Wyss Zurich Translational CenterETH Zurich and University of ZurichZurich8092Switzerland
| |
Collapse
|
38
|
Goetzman E, Gong Z, Zhang B, Muzumdar R. Complex II Biology in Aging, Health, and Disease. Antioxidants (Basel) 2023; 12:1477. [PMID: 37508015 PMCID: PMC10376733 DOI: 10.3390/antiox12071477] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/11/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Aging is associated with a decline in mitochondrial function which may contribute to age-related diseases such as neurodegeneration, cancer, and cardiovascular diseases. Recently, mitochondrial Complex II has emerged as an important player in the aging process. Mitochondrial Complex II converts succinate to fumarate and plays an essential role in both the tricarboxylic acid (TCA) cycle and the electron transport chain (ETC). The dysfunction of Complex II not only limits mitochondrial energy production; it may also promote oxidative stress, contributing, over time, to cellular damage, aging, and disease. Intriguingly, succinate, the substrate for Complex II which accumulates during mitochondrial dysfunction, has been shown to have widespread effects as a signaling molecule. Here, we review recent advances related to understanding the function of Complex II, succinate signaling, and their combined roles in aging and aging-related diseases.
Collapse
Affiliation(s)
- Eric Goetzman
- Division of Genetic and Genomic Medicine, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Zhenwei Gong
- Division of Endocrinology, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Bob Zhang
- Division of Genetic and Genomic Medicine, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Radhika Muzumdar
- Division of Endocrinology, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA 15260, USA
| |
Collapse
|
39
|
Wu KK. Extracellular Succinate: A Physiological Messenger and a Pathological Trigger. Int J Mol Sci 2023; 24:11165. [PMID: 37446354 DOI: 10.3390/ijms241311165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/01/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
When tissues are under physiological stresses, such as vigorous exercise and cold exposure, skeletal muscle cells secrete succinate into the extracellular space for adaptation and survival. By contrast, environmental toxins and injurious agents induce cellular secretion of succinate to damage tissues, trigger inflammation, and induce tissue fibrosis. Extracellular succinate induces cellular changes and tissue adaptation or damage by ligating cell surface succinate receptor-1 (SUCNR-1) and activating downstream signaling pathways and transcriptional programs. Since SUCNR-1 mediates not only pathological processes but also physiological functions, targeting it for drug development is hampered by incomplete knowledge about the characteristics of its physiological vs. pathological actions. This review summarizes the current status of extracellular succinate in health and disease and discusses the underlying mechanisms and therapeutic implications.
Collapse
Affiliation(s)
- Kenneth K Wu
- Institute of Cellular and System Medicine, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County 35053, Taiwan
- Institute of Biotechnology, College of Life Science, National Tsing-Hua University, Hsinchu 30013, Taiwan
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan
| |
Collapse
|
40
|
Abdullah S, Ghio M, Cotton-Betteridge A, Vinjamuri A, Drury R, Packer J, Aras O, Friedman J, Karim M, Engelhardt D, Kosowski E, Duong K, Shaheen F, McGrew PR, Harris CT, Reily R, Sammarco M, Chandra PK, Pociask D, Kolls J, Katakam PV, Smith A, Taghavi S, Duchesne J, Jackson-Weaver O. Succinate metabolism and membrane reorganization drives the endotheliopathy and coagulopathy of traumatic hemorrhage. SCIENCE ADVANCES 2023; 9:eadf6600. [PMID: 37315138 PMCID: PMC10266735 DOI: 10.1126/sciadv.adf6600] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 05/10/2023] [Indexed: 06/16/2023]
Abstract
Acute hemorrhage commonly leads to coagulopathy and organ dysfunction or failure. Recent evidence suggests that damage to the endothelial glycocalyx contributes to these adverse outcomes. The physiological events mediating acute glycocalyx shedding are undefined, however. Here, we show that succinate accumulation within endothelial cells drives glycocalyx degradation through a membrane reorganization-mediated mechanism. We investigated this mechanism in a cultured endothelial cell hypoxia-reoxygenation model, in a rat model of hemorrhage, and in trauma patient plasma samples. We found that succinate metabolism by succinate dehydrogenase mediates glycocalyx damage through lipid oxidation and phospholipase A2-mediated membrane reorganization, promoting the interaction of matrix metalloproteinase 24 (MMP24) and MMP25 with glycocalyx constituents. In a rat hemorrhage model, inhibiting succinate metabolism or membrane reorganization prevented glycocalyx damage and coagulopathy. In patients with trauma, succinate levels were associated with glycocalyx damage and the development of coagulopathy, and the interaction of MMP24 and syndecan-1 was elevated compared to healthy controls.
Collapse
Affiliation(s)
- Sarah Abdullah
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Michael Ghio
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA, USA
| | | | | | - Robert Drury
- Tulane University School of Medicine, New Orleans, LA, USA
| | - Jacob Packer
- Tulane University School of Medicine, New Orleans, LA, USA
| | - Oguz Aras
- Tulane University School of Medicine, New Orleans, LA, USA
| | - Jessica Friedman
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Mardeen Karim
- Tulane University School of Medicine, New Orleans, LA, USA
| | | | | | - Kelby Duong
- Tulane University School of Medicine, New Orleans, LA, USA
| | - Farhana Shaheen
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Patrick R. McGrew
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA, USA
- University Medical Center, New Orleans, LA, USA
| | - Charles T. Harris
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA, USA
- University Medical Center, New Orleans, LA, USA
| | - Robert Reily
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA, USA
- University Medical Center, New Orleans, LA, USA
| | - Mimi Sammarco
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Partha K. Chandra
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Derek Pociask
- Tulane University School of Medicine, Center for Translational Research in Infection and Inflammation, New Orleans, LA, USA
| | - Jay Kolls
- Tulane University School of Medicine, Center for Translational Research in Infection and Inflammation, New Orleans, LA, USA
| | - Prasad V. Katakam
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Alison Smith
- Louisiana State University Health Sciences Center, New Orleans, LA, USA
- University Medical Center, New Orleans, LA, USA
| | - Sharven Taghavi
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA, USA
- University Medical Center, New Orleans, LA, USA
| | - Juan Duchesne
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA, USA
- University Medical Center, New Orleans, LA, USA
| | - Olan Jackson-Weaver
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA, USA
| |
Collapse
|
41
|
Husková Z, Kikerlová S, Miklovič M, Kala P, Papoušek F, Neckář J. Inappropriate activation of the renin-angiotensin system improves cardiac tolerance to ischemia/reperfusion injury in rats with late angiotensin II-dependent hypertension. Front Physiol 2023; 14:1151308. [PMID: 37389123 PMCID: PMC10301744 DOI: 10.3389/fphys.2023.1151308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 06/05/2023] [Indexed: 07/01/2023] Open
Abstract
The aim of the study was to clarify the role of the interplay between hypertension and the renin-angiotensin system (RAS) in the pathophysiology of myocardial ischemia/reperfusion (I/R) injury. We hypothesized that in the late phase of hypertension with already developed signs of end-organ damage, inappropriate RAS activation could impair cardiac tolerance to I/R injury. Experiments were performed in male Cyp1a1-Ren-2 transgenic rats with inducible hypertension. The early phase of ANG II-dependent hypertension was induced by 5 days and the late phase by the 13 days dietary indole-3-carbinol (I3C) administration. Noninduced rats served as controls. Echocardiography and pressure-volume analysis were performed, angiotensins' levels were measured and cardiac tolerance to ischemia/reperfusion injury was studied. The infarct size was significantly reduced (by 50%) in 13 days I3C-induced hypertensive rats with marked cardiac hypertrophy, this reduction was abolished by losartan treatment. In the late phase of hypertension there are indications of a failing heart, mainly in reduced preload recruitable stroke work (PRSW), but only nonsignificant trends in worsening of some other parameters, showing that the myocardium is in a compensated phase. The influence of the RAS depends on the balance between the vasoconstrictive and the opposed vasodilatory axis. In the initial stage of hypertension, the vasodilatory axis of the RAS prevails, and with the development of hypertension the vasoconstrictive axis of the RAS becomes stronger. We observed a clear effect of AT1 receptor blockade on maximum pressure in left ventricle, cardiac hypertrophy and ANG II levels. In conclusion, we confirmed improved cardiac tolerance to I/R injury in hypertensive hypertrophied rats and showed that, in the late phase of hypertension, the myocardium is in a compensated phase.
Collapse
Affiliation(s)
- Zuzana Husková
- Center of Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - Soňa Kikerlová
- Center of Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - Matúš Miklovič
- Center of Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czechia
- Department of Pathophysiology, 2nd Faculty of Medicine, Charles University, Prague, Czechia
| | - Petr Kala
- Center of Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czechia
- Department of Cardiology, 2nd Medical Faculty, Charles University and University Hospital Motol, Prague, Czechia
| | - František Papoušek
- Laboratory of Developmental Cardiology, Institute of Physiology, Academy of Sciences of the Czech Republic (ASCR), Prague, Czechia
| | - Jan Neckář
- Center of Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czechia
- Laboratory of Developmental Cardiology, Institute of Physiology, Academy of Sciences of the Czech Republic (ASCR), Prague, Czechia
| |
Collapse
|
42
|
Wang Q, Zuurbier CJ, Huhn R, Torregroza C, Hollmann MW, Preckel B, van den Brom CE, Weber NC. Pharmacological Cardioprotection against Ischemia Reperfusion Injury-The Search for a Clinical Effective Therapy. Cells 2023; 12:1432. [PMID: 37408266 DOI: 10.3390/cells12101432] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/10/2023] [Accepted: 05/17/2023] [Indexed: 07/07/2023] Open
Abstract
Pharmacological conditioning aims to protect the heart from myocardial ischemia-reperfusion injury (IRI). Despite extensive research in this area, today, a significant gap remains between experimental findings and clinical practice. This review provides an update on recent developments in pharmacological conditioning in the experimental setting and summarizes the clinical evidence of these cardioprotective strategies in the perioperative setting. We start describing the crucial cellular processes during ischemia and reperfusion that drive acute IRI through changes in critical compounds (∆GATP, Na+, Ca2+, pH, glycogen, succinate, glucose-6-phosphate, mitoHKII, acylcarnitines, BH4, and NAD+). These compounds all precipitate common end-effector mechanisms of IRI, such as reactive oxygen species (ROS) generation, Ca2+ overload, and mitochondrial permeability transition pore opening (mPTP). We further discuss novel promising interventions targeting these processes, with emphasis on cardiomyocytes and the endothelium. The limited translatability from basic research to clinical practice is likely due to the lack of comorbidities, comedications, and peri-operative treatments in preclinical animal models, employing only monotherapy/monointervention, and the use of no-flow (always in preclinical models) versus low-flow ischemia (often in humans). Future research should focus on improved matching between preclinical models and clinical reality, and on aligning multitarget therapy with optimized dosing and timing towards the human condition.
Collapse
Affiliation(s)
- Qian Wang
- Department of Anesthesiology-L.E.I.C.A., Amsterdam University Medical Centers, Location AMC, Cardiovascular Science, Meibergdreef 11, 1105 AZ Amsterdam, The Netherlands
| | - Coert J Zuurbier
- Department of Anesthesiology-L.E.I.C.A., Amsterdam University Medical Centers, Location AMC, Cardiovascular Science, Meibergdreef 11, 1105 AZ Amsterdam, The Netherlands
| | - Ragnar Huhn
- Department of Anesthesiology, Kerckhoff-Clinic-Center for Heart, Lung, Vascular and Rheumatic Disease, Justus-Liebig-University Giessen, Benekestr. 2-8, 61231 Bad Nauheim, Germany
| | - Carolin Torregroza
- Department of Anesthesiology, Kerckhoff-Clinic-Center for Heart, Lung, Vascular and Rheumatic Disease, Justus-Liebig-University Giessen, Benekestr. 2-8, 61231 Bad Nauheim, Germany
| | - Markus W Hollmann
- Department of Anesthesiology-L.E.I.C.A., Amsterdam University Medical Centers, Location AMC, Cardiovascular Science, Meibergdreef 11, 1105 AZ Amsterdam, The Netherlands
| | - Benedikt Preckel
- Department of Anesthesiology-L.E.I.C.A., Amsterdam University Medical Centers, Location AMC, Cardiovascular Science, Meibergdreef 11, 1105 AZ Amsterdam, The Netherlands
| | - Charissa E van den Brom
- Department of Anesthesiology-L.E.I.C.A., Amsterdam University Medical Centers, Location AMC, Cardiovascular Science, Meibergdreef 11, 1105 AZ Amsterdam, The Netherlands
| | - Nina C Weber
- Department of Anesthesiology-L.E.I.C.A., Amsterdam University Medical Centers, Location AMC, Cardiovascular Science, Meibergdreef 11, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
43
|
Markevich NI, Markevich LN. Computational Modeling Analysis of Kinetics of Fumarate Reductase Activity and ROS Production during Reverse Electron Transfer in Mitochondrial Respiratory Complex II. Int J Mol Sci 2023; 24:ijms24098291. [PMID: 37175997 PMCID: PMC10179487 DOI: 10.3390/ijms24098291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 04/23/2023] [Accepted: 04/28/2023] [Indexed: 05/15/2023] Open
Abstract
Reverse electron transfer in mitochondrial complex II (CII) plays an important role in hypoxia/anoxia, in particular, in ischemia, when the blood supply to an organ is disrupted and oxygen is not available. A computational model of CII was developed in this work to facilitate the quantitative analysis of the kinetics of quinol-fumarate reduction as well as ROS production during reverse electron transfer in CII. The model consists of 20 ordinary differential equations and 7 moiety conservation equations. The parameter values were determined at which the kinetics of electron transfer in CII in both forward and reverse directions would be explained simultaneously. The possibility of the existence of the "tunnel diode" behavior in the reverse electron transfer in CII, where the driving force is QH2, was tested. It was found that any high concentrations of QH2 and fumarate are insufficient for the appearance of a tunnel effect. The results of computer modeling show that the maximum rate of succinate production cannot provide a high concentration of succinate in ischemia. Furthermore, computational modeling results predict a very low rate of ROS production, about 50 pmol/min/mg mitochondrial protein, which is considerably less than 1000 pmol/min/mg protein observed in CII in forward direction.
Collapse
Affiliation(s)
- Nikolay I Markevich
- Institute of Theoretical and Experimental Biophysics of RAS, Pushchino, Moscow 142290, Russia
| | - Lubov N Markevich
- Institute of Cell Biophysics of RAS, Pushchino, Moscow 142290, Russia
| |
Collapse
|
44
|
Consegal M, Barba I, García Del Blanco B, Otaegui I, Rodríguez-Palomares JF, Martí G, Serra B, Bellera N, Ojeda-Ramos M, Valente F, Carmona MÁ, Miró-Casas E, Sambola A, Lidón RM, Bañeras J, Barrabés JA, Rodríguez C, Benito B, Ruiz-Meana M, Inserte J, Ferreira-González I, Rodríguez-Sinovas A. Spontaneous reperfusion enhances succinate concentration in peripheral blood from stemi patients but its levels does not correlate with myocardial infarct size or area at risk. Sci Rep 2023; 13:6907. [PMID: 37106099 PMCID: PMC10140265 DOI: 10.1038/s41598-023-34196-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 04/25/2023] [Indexed: 04/29/2023] Open
Abstract
Succinate is enhanced during initial reperfusion in blood from the coronary sinus in ST-segment elevation myocardial infarction (STEMI) patients and in pigs submitted to transient coronary occlusion. Succinate levels might have a prognostic value, as they may correlate with edema volume or myocardial infarct size. However, blood from the coronary sinus is not routinely obtained in the CathLab. As succinate might be also increased in peripheral blood, we aimed to investigate whether peripheral plasma concentrations of succinate and other metabolites obtained during coronary revascularization correlate with edema volume or infarct size in STEMI patients. Plasma samples were obtained from peripheral blood within the first 10 min of revascularization in 102 STEMI patients included in the COMBAT-MI trial (initial TIMI 1) and from 9 additional patients with restituted coronary blood flow (TIMI 2). Metabolite concentrations were analyzed by 1H-NMR. Succinate concentration averaged 0.069 ± 0.0073 mmol/L in patients with TIMI flow ≤ 1 and was significantly increased in those with TIMI 2 at admission (0.141 ± 0.058 mmol/L, p < 0.05). However, regression analysis did not detect any significant correlation between most metabolite concentrations and infarct size, extent of edema or other cardiac magnetic resonance (CMR) variables. In conclusion, spontaneous reperfusion in TIMI 2 patients associates with enhanced succinate levels in peripheral blood, suggesting that succinate release increases overtime following reperfusion. However, early plasma levels of succinate and other metabolites obtained from peripheral blood does not correlate with the degree of irreversible injury or area at risk in STEMI patients, and cannot be considered as predictors of CMR variables.Trial registration: Registered at www.clinicaltrials.gov (NCT02404376) on 31/03/2015. EudraCT number: 2015-001000-58.
Collapse
Affiliation(s)
- Marta Consegal
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Ignasi Barba
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
- Faculty of Medicine, University of Vic - Central University of Catalonia (UVicUCC), Can Baumann. Ctra. de Roda, 70, 08500, Vic, Spain
| | - Bruno García Del Blanco
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Imanol Otaegui
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - José F Rodríguez-Palomares
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Gerard Martí
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Bernat Serra
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Neus Bellera
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Manuel Ojeda-Ramos
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Filipa Valente
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Maria Ángeles Carmona
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Elisabet Miró-Casas
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Antonia Sambola
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Rosa María Lidón
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Jordi Bañeras
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - José Antonio Barrabés
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Cristina Rodríguez
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
- Institut de Recerca Hospital de la Santa Creu i Sant Pau (IRHSCSP), Barcelona, Spain
| | - Begoña Benito
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Marisol Ruiz-Meana
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Inserte
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Ignacio Ferreira-González
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain.
- Departament de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain.
- Centro de Investigación Biomédica en Red (CIBER) de Epidemiología y Salud Pública, CIBERESP, Instituto de Salud Carlos III, Madrid, Spain.
| | - Antonio Rodríguez-Sinovas
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain.
- Departament de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain.
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
45
|
Nanadikar MS, Vergel Leon AM, Guo J, van Belle GJ, Jatho A, Philip ES, Brandner AF, Böckmann RA, Shi R, Zieseniss A, Siemssen CM, Dettmer K, Brodesser S, Schmidtendorf M, Lee J, Wu H, Furdui CM, Brandenburg S, Burgoyne JR, Bogeski I, Riemer J, Chowdhury A, Rehling P, Bruegmann T, Belousov VV, Katschinski DM. IDH3γ functions as a redox switch regulating mitochondrial energy metabolism and contractility in the heart. Nat Commun 2023; 14:2123. [PMID: 37055412 PMCID: PMC10102218 DOI: 10.1038/s41467-023-37744-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 03/29/2023] [Indexed: 04/15/2023] Open
Abstract
Redox signaling and cardiac function are tightly linked. However, it is largely unknown which protein targets are affected by hydrogen peroxide (H2O2) in cardiomyocytes that underly impaired inotropic effects during oxidative stress. Here, we combine a chemogenetic mouse model (HyPer-DAO mice) and a redox-proteomics approach to identify redox sensitive proteins. Using the HyPer-DAO mice, we demonstrate that increased endogenous production of H2O2 in cardiomyocytes leads to a reversible impairment of cardiac contractility in vivo. Notably, we identify the γ-subunit of the TCA cycle enzyme isocitrate dehydrogenase (IDH)3 as a redox switch, linking its modification to altered mitochondrial metabolism. Using microsecond molecular dynamics simulations and experiments using cysteine-gene-edited cells reveal that IDH3γ Cys148 and 284 are critically involved in the H2O2-dependent regulation of IDH3 activity. Our findings provide an unexpected mechanism by which mitochondrial metabolism can be modulated through redox signaling processes.
Collapse
Affiliation(s)
- Maithily S Nanadikar
- Institute of Cardiovascular Physiology, University Medical Center Göttingen, Georg-August, University Göttingen, 37073, Göttingen, Germany
| | - Ana M Vergel Leon
- Institute of Cardiovascular Physiology, University Medical Center Göttingen, Georg-August, University Göttingen, 37073, Göttingen, Germany
| | - Jia Guo
- Institute of Cardiovascular Physiology, University Medical Center Göttingen, Georg-August, University Göttingen, 37073, Göttingen, Germany
| | - Gijsbert J van Belle
- Institute of Cardiovascular Physiology, University Medical Center Göttingen, Georg-August, University Göttingen, 37073, Göttingen, Germany
| | - Aline Jatho
- Institute of Cardiovascular Physiology, University Medical Center Göttingen, Georg-August, University Göttingen, 37073, Göttingen, Germany
| | - Elvina S Philip
- Institute of Cardiovascular Physiology, University Medical Center Göttingen, Georg-August, University Göttingen, 37073, Göttingen, Germany
| | - Astrid F Brandner
- Computational Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058, Erlangen, Germany
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK
| | - Rainer A Böckmann
- Computational Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058, Erlangen, Germany
- Erlangen National High-Performance Computing Center (NHR@FAU), Erlangen, Germany
| | - Runzhu Shi
- Institute of Cardiovascular Physiology, University Medical Center Göttingen, Georg-August, University Göttingen, 37073, Göttingen, Germany
| | - Anke Zieseniss
- Institute of Cardiovascular Physiology, University Medical Center Göttingen, Georg-August, University Göttingen, 37073, Göttingen, Germany
| | - Carla M Siemssen
- Institute of Cardiovascular Physiology, University Medical Center Göttingen, Georg-August, University Göttingen, 37073, Göttingen, Germany
| | - Katja Dettmer
- Institute of Functional Genomics, University of Regensburg, 93053, Regensburg, Germany
| | - Susanne Brodesser
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), 50931, Cologne, Germany
| | - Marlen Schmidtendorf
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), 50931, Cologne, Germany
| | - Jingyun Lee
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Hanzhi Wu
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Cristina M Furdui
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Sören Brandenburg
- Clinic of Cardiology & Pneumology, University Medical Center Göttingen, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Joseph R Burgoyne
- King's College London, School of Cardiovascular Medicine & Sciences, The British Heart Foundation Centre of Excellence, SE1 7EH, London, UK
| | - Ivan Bogeski
- Institute of Cardiovascular Physiology, University Medical Center Göttingen, Georg-August, University Göttingen, 37073, Göttingen, Germany
| | - Jan Riemer
- Institute for Biochemistry, Redox Metabolism and CECAD, University of Cologne, 50674, Cologne, Germany
| | - Arpita Chowdhury
- Institute of Cellular Biochemistry, University Medical Center Göttingen, 37073, Göttingen, Germany
| | - Peter Rehling
- Institute of Cellular Biochemistry, University Medical Center Göttingen, 37073, Göttingen, Germany
- Cluster of Excellence, Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells (MBExC), University of Göttingen, Göttingen, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Translational Neuroinflammation and Automated Microscopy, Göttingen, Germany
- Max Planck Institute for Multidisciplinary Sciences, 37077, Göttingen, Germany
| | - Tobias Bruegmann
- Institute of Cardiovascular Physiology, University Medical Center Göttingen, Georg-August, University Göttingen, 37073, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
- Cluster of Excellence, Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells (MBExC), University of Göttingen, Göttingen, Germany
| | - Vsevolod V Belousov
- Institute of Cardiovascular Physiology, University Medical Center Göttingen, Georg-August, University Göttingen, 37073, Göttingen, Germany
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Agency, 117997, Moscow, Russia
| | - Dörthe M Katschinski
- Institute of Cardiovascular Physiology, University Medical Center Göttingen, Georg-August, University Göttingen, 37073, Göttingen, Germany.
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany.
| |
Collapse
|
46
|
Johansen A, Thiede B, Anonsen JH, Nilsson GE. Surviving without oxygen involves major tissue specific changes in the proteome of crucian carp ( Carassius carassius). PeerJ 2023; 11:e14890. [PMID: 36915662 PMCID: PMC10007964 DOI: 10.7717/peerj.14890] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 01/23/2023] [Indexed: 03/10/2023] Open
Abstract
The crucian carp (Carassius carassius) can survive complete oxygen depletion (anoxia) for several months at low temperatures, making it an excellent model for studying molecular adaptations to anoxia. Still, little is known about how its global proteome responds to anoxia and reoxygenation. By applying mass spectrometry-based proteome analyses on brain, heart and liver tissue from crucian carp exposed to normoxia, five days anoxia, and reoxygenation, we found major changes in particularly cardiac and hepatic protein levels in response to anoxia and reoxygenation. These included tissue-specific differences in mitochondrial proteins involved in aerobic respiration and mitochondrial membrane integrity. Enzymes in the electron transport system (ETS) decreased in heart and increased massively in liver during anoxia and reoxygenation but did not change in the brain. Importantly, the data support a special role for the liver in succinate handling upon reoxygenation, as suggested by a drastic increase of components of the ETS and uncoupling protein 2, which could allow for succinate metabolism without excessive formation of reactive oxygen species (ROS). Also during reoxygenation, the levels of proteins involved in the cristae junction organization of the mitochondria changed in the heart, possibly functioning to suppress ROS formation. Furthermore, proteins involved in immune (complement) system activation changed in the anoxic heart compared to normoxic controls. The results emphasize that responses to anoxia are highly tissue-specific and related to organ function.
Collapse
Affiliation(s)
| | - Bernd Thiede
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Jan Haug Anonsen
- Department of Biosciences, University of Oslo, Oslo, Norway
- Climate & Environment Department, NORCE, Norwegian Research Centre AS, Stavanger, Norway
| | | |
Collapse
|
47
|
Mitochondrial Dysfunction: The Hidden Player in the Pathogenesis of Atherosclerosis? Int J Mol Sci 2023; 24:ijms24021086. [PMID: 36674602 PMCID: PMC9861427 DOI: 10.3390/ijms24021086] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/29/2022] [Accepted: 01/04/2023] [Indexed: 01/09/2023] Open
Abstract
Atherosclerosis is a multifactorial inflammatory pathology that involves metabolic processes. Improvements in therapy have drastically reduced the prognosis of cardiovascular disease. Nevertheless, a significant residual risk is still relevant, and is related to unmet therapeutic targets. Endothelial dysfunction and lipid infiltration are the primary causes of atherosclerotic plaque progression. In this contest, mitochondrial dysfunction can affect arterial wall cells, in particular macrophages, smooth muscle cells, lymphocytes, and endothelial cells, causing an increase in reactive oxygen species (ROS), leading to oxidative stress, chronic inflammation, and intracellular lipid deposition. The detection and characterization of mitochondrial DNA (mtDNA) is crucial for assessing mitochondrial defects and should be considered the goal for new future therapeutic interventions. In this review, we will focus on a new idea, based on the analysis of data from many research groups, namely the link between mitochondrial impairment and endothelial dysfunction and, in particular, its effect on atherosclerosis and aging. Therefore, we discuss known and novel mitochondria-targeting therapies in the contest of atherosclerosis.
Collapse
|
48
|
Milliken AS, Ciesla JH, Nadtochiy SM, Brookes PS. Distinct effects of intracellular vs. extracellular acidic pH on the cardiac metabolome during ischemia and reperfusion. J Mol Cell Cardiol 2023; 174:101-114. [PMID: 36481511 PMCID: PMC9868090 DOI: 10.1016/j.yjmcc.2022.11.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/22/2022] [Accepted: 11/25/2022] [Indexed: 12/10/2022]
Abstract
Tissue ischemia results in intracellular pH (pHIN) acidification, and while metabolism is a known driver of acidic pHIN, less is known about how acidic pHIN regulates metabolism. Furthermore, acidic extracellular (pHEX) during early reperfusion confers cardioprotection, but how this impacts metabolism is unclear. Herein we employed LCMS based targeted metabolomics to analyze perfused mouse hearts exposed to: (i) control perfusion, (ii) hypoxia, (iii) ischemia, (iv) enforced acidic pHIN, (v) control reperfusion, and (vi) acidic pHEX (6.8) reperfusion. Surprisingly little overlap was seen between metabolic changes induced by hypoxia, ischemia, and acidic pHIN. Acidic pHIN elevated metabolites in the top half of glycolysis, and enhanced glutathione redox state. Meanwhile, acidic pHEX reperfusion induced substantial metabolic changes in addition to those seen in control reperfusion. This included elevated metabolites in the top half of glycolysis, prevention of purine nucleotide loss, and an enhancement in glutathione redox state. These data led to hypotheses regarding potential roles for methylglyoxal inhibiting the mitochondrial permeability transition pore, and for acidic inhibition of ecto-5'-nucleotidase, as potential mediators of cardioprotection by acidic pHEX reperfusion. However, neither hypothesis was supported by subsequent experiments. In contrast, analysis of cardiac effluents revealed complex effects of pHEX on metabolite transport, suggesting that mildly acidic pHEX may enhance succinate release during reperfusion. Overall, each intervention had distinct and overlapping metabolic effects, suggesting acidic pH is an independent metabolic regulator regardless which side of the cell membrane it is imposed.
Collapse
Affiliation(s)
- Alexander S Milliken
- Department of Pharmacology and Physiology, University of Rochester Medical Center, USA
| | - Jessica H Ciesla
- Department of Biochemistry, University of Rochester Medical Center, USA
| | - Sergiy M Nadtochiy
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, USA
| | - Paul S Brookes
- Department of Pharmacology and Physiology, University of Rochester Medical Center, USA; Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, USA.
| |
Collapse
|
49
|
Abstract
Succinate is a circulating metabolite, and the relationship between abnormal changes in the physiological concentration of succinate and inflammatory diseases caused by the overreaction of certain immune cells has become a research focus. Recent investigations have shown that succinate produced by the gut microbiota has the potential to regulate host homeostasis and treat diseases such as inflammation. Gut microbes are important for maintaining intestinal homeostasis. Microbial metabolites serve as nutrients in energy metabolism, and act as signal molecules that stimulate host cell and organ function and affect the structural balance between symbiotic gut microorganisms. This review focuses on succinate as a metabolite of both host cells and gut microbes and its involvement in regulating the gut - immune tissue axis by activating intestinal mucosal cells, including macrophages, dendritic cells, and intestinal epithelial cells. We also examined its role as the mediator of microbiota - host crosstalk and its potential function in regulating intestinal microbiota homeostasis. This review explores feasible ways to moderate succinate levels and provides new insights into succinate as a potential target for microbial therapeutics for humans.
Collapse
Affiliation(s)
- Yi-Han Wei
- College of Animal Science, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangzhou, China
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Xi Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jiang-Chao Zhao
- Department of Animal Science, Division of Agriculture, University of Arkansas, Fayetteville, AR, USA
| | - Xiu-Qi Wang
- College of Animal Science, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangzhou, China
| | - Chun-Qi Gao
- College of Animal Science, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangzhou, China
| |
Collapse
|
50
|
Kim JS, Chapman WC, Lin Y. Mitochondrial Autophagy in Ischemic Aged Livers. Cells 2022; 11:cells11244083. [PMID: 36552847 PMCID: PMC9816943 DOI: 10.3390/cells11244083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/09/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Mitochondrial autophagy (mitophagy) is a central catabolic event for mitochondrial quality control. Defective or insufficient mitophagy, thus, can result in mitochondrial dysfunction, and ultimately cell death. There is a strong causal relationship between ischemia/reperfusion (I/R) injury and mitochondrial dysfunction following liver resection and transplantation. Compared to young patients, elderly patients poorly tolerate I/R injury. Accumulation of abnormal mitochondria after I/R is more prominent in aged livers than in young counterparts. This review highlights how altered autophagy is mechanistically involved in age-dependent hypersensitivity to reperfusion injury.
Collapse
Affiliation(s)
- Jae-Sung Kim
- Department of Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA; (W.C.C.); (Y.L.)
- Department of Cell Biology & Physiology, Washington University in St. Louis, St. Louis, MO 63110, USA
- Correspondence:
| | - William C. Chapman
- Department of Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA; (W.C.C.); (Y.L.)
| | - Yiing Lin
- Department of Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA; (W.C.C.); (Y.L.)
| |
Collapse
|