1
|
Chen Y, Shen YQ. Role of reactive oxygen species in regulating epigenetic modifications. Cell Signal 2024; 125:111502. [PMID: 39521028 DOI: 10.1016/j.cellsig.2024.111502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 10/24/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
Reactive oxygen species (ROS) originate from diverse sources and regulate multiple signaling pathways within the cellular environment. Their generation is intricately controlled, and disruptions in their signaling or atypical levels can precipitate pathological conditions. Epigenetics, the examination of heritable alterations in gene expression independent of changes in the genetic code, has been implicated in the pathogenesis of various diseases through aberrant epigenetic modifications. The significant contribution of epigenetic modifications to disease progression underscores their potential as crucial therapeutic targets for a wide array of medical conditions. This study begins by providing an overview of ROS and epigenetics, followed by a discussion on the mechanisms of epigenetic modifications such as DNA methylation, histone modification, and RNA modification-mediated regulation. Subsequently, a detailed examination of the interaction between ROS and epigenetic modifications is presented, offering new perspectives and avenues for exploring the mechanisms underlying specific epigenetic diseases and the development of novel therapeutics.
Collapse
Affiliation(s)
- Yutong Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Ying-Qiang Shen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China.
| |
Collapse
|
2
|
Yang B, Xu Y, Yu J, Wang Q, Fan Q, Zhao X, Qiao Y, Zhang Z, Zhou Q, Yin D, He M, He H. Salidroside pretreatment alleviates ferroptosis induced by myocardial ischemia/reperfusion through mitochondrial superoxide-dependent AMPKα2 activation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155365. [PMID: 38552436 DOI: 10.1016/j.phymed.2024.155365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/28/2023] [Accepted: 01/14/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Ferroptosis, a form of regulated cell death (RCD) that relies on excessive reactive oxygen species (ROS) generation, Fe2+accumulation, abnormal lipid metabolism and is involved in various organ ischemia/reperfusion (I/R) injury, expecially in myocardium. Mitochondria are the powerhouses of eukaryotic cells and essential in regulating multiple RCD. However, the links between mitochondria and ferroptosis are still poorly understood. Salidroside (Sal), a natural phenylpropanoid glycoside isolated from Rhodiola rosea, has mult-bioactivities. However, the effects and mechanism in alleviating ferroptosis caused by myocardial I/R injury remains unclear. PURPOSE This study aimed to investigate whether pretreated with Sal could protect the myocardium against I/R damage and the underlying mechanisms. In particular, the relationship between Sal pretreatment, AMPKα2 activity, mitochondria and ROS generation was explored. STUDY DESIGN AND METHODS Firstly, A/R or I/R injury models were employed in H9c2 cells and Sprague-Dawley rats. And then the anti-ferroptotic effects and mechanism of Sal pretreatment was detected using multi-relevant indexes in H9c2 cells. Further, how does Sal pretreatment in AMPKα2 phosphorylation was explored. Finally, these results were validated by I/R injury in rats. RESULTS Similar to Ferrostatin-1 (a ferroptosis inhibitor) and MitoTEMPO, a mitochondrial free radical scavenger, Sal pretreatment effectively alleviated Fe2+ accumulation, redox disequilibrium and maintained mitochondrial energy production and function in I/R-induced myocardial injury, as demonstrated using multifunctional, enzymatic, and morphological indices. However, these effects were abolished by downregulation of AMPKα2 using an adenovirus, both in vivo and in vitro. Moreover, the results also provided a non-canonical mechanism that, under mild mitochondrial ROS generation, Sal pretreatment upregulated and phosphorylated AMPKα2, which enhanced mitochondrial complex I activity to activate innate adaptive responses and increase cellular tolerance to A/R injury. CONCLUSION Overall, our work highlighted mitochondria are of great impotance in myocardial I/R-induced ferroptosis and demonstrated that Sal pretreatment activated AMPKα2 against I/R injury, indicating that Sal could become a candidate phytochemical for the treatment of myocardial I/R injury.
Collapse
Affiliation(s)
- Bin Yang
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Ying Xu
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Jingzhi Yu
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Qihao Wang
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Qigui Fan
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Xiaoyu Zhao
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Yang Qiao
- Jiangxi Academy of Clinical Medical Sciences, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Zeyu Zhang
- Jiangxi Academy of Clinical Medical Sciences, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Qing Zhou
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Dong Yin
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Ming He
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China.
| | - Huan He
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China.
| |
Collapse
|
3
|
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
|
4
|
Reiter RJ, Sharma R, Chuffa LGDA, Simko F, Dominguez-Rodriguez A. Mitochondrial Melatonin: Beneficial Effects in Protecting against Heart Failure. Life (Basel) 2024; 14:88. [PMID: 38255703 PMCID: PMC10820220 DOI: 10.3390/life14010088] [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: 11/28/2023] [Revised: 01/01/2024] [Accepted: 01/03/2024] [Indexed: 01/24/2024] Open
Abstract
Cardiovascular disease is the cause of physical infirmity and thousands of deaths annually. Typically, during heart failure, cardiomyocyte mitochondria falter in terms of energy production and metabolic processing. Additionally, inflammation and the accumulation of non-contractile fibrous tissue contribute to cardiac malfunction. Melatonin, an endogenously produced molecule, experimentally reduces the initiation and progression of atherosclerotic lesions, which are often the basis of coronary artery disease. The current review critically analyzes published data related to the experimental use of melatonin to forestall coronary artery pathologies. Collectively, these studies document melatonin's anti-atherosclerotic actions in reducing LDL oxidation and triglyceride levels, lowering endothelial malfunction, limiting adhesion molecule formation, preventing macrophage polarization to the M1 pro-inflammatory phenotype, changing cellular metabolism, scavenging destructive reactive oxygen species, preventing the proliferation and invasion of arterial smooth muscle cells into the lesioned area, restricting the ingrowth of blood vessels from the vasa vasorum, and solidifying the plaque cap to reduce the chance of its rupture. Diabetic hyperglycemia, which aggravates atherosclerotic plaque formation, is also inhibited by melatonin supplementation in experimental animals. The potential value of non-toxic melatonin as a possible inhibitor of cardiac pathology in humans should be seriously considered by performing clinical trials using this multifunctional molecule.
Collapse
Affiliation(s)
- Russel J. Reiter
- Department of Cell Systems and Anatomy, Long School of Medicine, UT Health San Antonio, San Antonio, TX 78229, USA
| | - Ramaswamy Sharma
- Applied Biomedical Sciences, School of Osteopathic Medicine, University of the Incarnate Word, San Antonio, TX 78235, USA
| | - Luiz Gustavo de Almeida Chuffa
- Department of Structural and Functional Biology-IBB/UNESP, Institute of Biosciences of Botucatu, Universidade Estadual Paulista, Botucatu 18618-689, São Paulo, Brazil;
| | - Fedor Simko
- Institute of Pathophysiology, Faculty of Medicine, Comenius University, 81108 Bratislava, Slovakia;
| | | |
Collapse
|
5
|
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: 0] [Impact Index Per Article: 0] [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
|
6
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [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
|
7
|
Chatterjee P, Holody CD, Kirschenman R, Graton ME, Spaans F, Phillips TJ, Case CP, Bourque SL, Lemieux H, Davidge ST. Sex-Specific Effects of Prenatal Hypoxia and a Placental Antioxidant Treatment on Cardiac Mitochondrial Function in the Young Adult Offspring. Int J Mol Sci 2023; 24:13624. [PMID: 37686430 PMCID: PMC10487956 DOI: 10.3390/ijms241713624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/25/2023] [Accepted: 09/01/2023] [Indexed: 09/10/2023] Open
Abstract
Prenatal hypoxia is associated with placental oxidative stress, leading to impaired fetal growth and an increased risk of cardiovascular disease in the adult offspring; however, the mechanisms are unknown. Alterations in mitochondrial function may result in impaired cardiac function in offspring. In this study, we hypothesized that cardiac mitochondrial function is impaired in adult offspring exposed to intrauterine hypoxia, which can be prevented by placental treatment with a nanoparticle-encapsulated mitochondrial antioxidant (nMitoQ). Cardiac mitochondrial respiration was assessed in 4-month-old rat offspring exposed to prenatal hypoxia (11% O2) from gestational day (GD)15-21 receiving either saline or nMitoQ on GD 15. Prenatal hypoxia did not alter cardiac mitochondrial oxidative phosphorylation capacity in the male offspring. In females, the NADH + succinate pathway capacity decreased by prenatal hypoxia and tended to be increased by nMitoQ. Prenatal hypoxia also decreased the succinate pathway capacity in females. nMitoQ treatment increased respiratory coupling efficiency in prenatal hypoxia-exposed female offspring. In conclusion, prenatal hypoxia impaired cardiac mitochondrial function in adult female offspring only, which was improved with prenatal nMitoQ treatment. Therefore, treatment strategies targeting placental oxidative stress in prenatal hypoxia may reduce the risk of cardiovascular disease in adult offspring by improving cardiac mitochondrial function in a sex-specific manner.
Collapse
Affiliation(s)
- Paulami Chatterjee
- Department of Physiology, University of Alberta, Edmonton, AB T6G 2R3, Canada;
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T6G 2R3, Canada; (R.K.); (M.E.G.); (F.S.)
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada; (S.L.B.); (H.L.)
| | - Claudia D. Holody
- Faculty Saint-Jean, University of Alberta, Edmonton, AB T6G 2R3, Canada;
- Department of Pediatrics, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Raven Kirschenman
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T6G 2R3, Canada; (R.K.); (M.E.G.); (F.S.)
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada; (S.L.B.); (H.L.)
| | - Murilo E. Graton
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T6G 2R3, Canada; (R.K.); (M.E.G.); (F.S.)
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada; (S.L.B.); (H.L.)
| | - Floor Spaans
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T6G 2R3, Canada; (R.K.); (M.E.G.); (F.S.)
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada; (S.L.B.); (H.L.)
| | - Tom J. Phillips
- UK Dementia Research Institute, Cardiff University, Cardiff CF10 3AT, UK;
| | - C. Patrick Case
- Musculoskeletal Research Unit, University of Bristol, Bristol BS10 5NB, UK;
| | - Stephane L. Bourque
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada; (S.L.B.); (H.L.)
- Department of Pediatrics, University of Alberta, Edmonton, AB T6G 2R3, Canada
- Department of Anesthesiology & Pain Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Hélène Lemieux
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada; (S.L.B.); (H.L.)
- Faculty Saint-Jean, University of Alberta, Edmonton, AB T6G 2R3, Canada;
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Sandra T. Davidge
- Department of Physiology, University of Alberta, Edmonton, AB T6G 2R3, Canada;
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T6G 2R3, Canada; (R.K.); (M.E.G.); (F.S.)
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada; (S.L.B.); (H.L.)
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
Genders AJ, Kuang J, Saner NJ, Botella J, Bishop DJ. Ammonium chloride administration prevents training-induced improvements in mitochondrial respiratory function in the soleus muscle of male rats. Am J Physiol Cell Physiol 2023; 324:C67-C75. [PMID: 36542512 DOI: 10.1152/ajpcell.00165.2022] [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: 04/22/2022] [Revised: 11/21/2022] [Accepted: 11/21/2022] [Indexed: 11/29/2022]
Abstract
Exercise training can increase both mitochondrial content and mitochondrial respiration. Despite its popularity, high-intensity exercise can be accompanied by mild acidosis (also present in certain pathological states), which may limit exercise-induced adaptations to skeletal muscle mitochondria. The aim of this study was to determine if administration of ammonium chloride (0.05 g/kg) to Wistar rats before each individual exercise session (5 high-intensity exercise sessions/wk for 8 wk) reduced training-induced increases in mitochondrial content (measured by citrate synthase activity and protein content of electron transport system complexes) and respiration (measured in permeabilized muscle fibers). In the soleus muscle, the exercise-training-induced increase in mitochondrial respiration was reduced in rats administered ammonium chloride compared to control animals, but mitochondrial content was not altered. These effects were not present in the white gastrocnemius muscle. In conclusion, ammonium chloride administration before each exercise session over 8 wk reduced improvements in mitochondrial respiration in the soleus muscle but did not alter mitochondrial content. This suggests that mild acidosis may affect training-induced improvements in the respiration of mitochondria in some muscles.
Collapse
Affiliation(s)
- Amanda J Genders
- Institute for Health and Sport, Victoria University, Melbourne, Australia
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Jujiao Kuang
- Institute for Health and Sport, Victoria University, Melbourne, Australia
- Australian Institute for Musculoskeletal Sciences, Melbourne, Australia
| | - Nicholas J Saner
- Institute for Health and Sport, Victoria University, Melbourne, Australia
- Human Integrative Physiology, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Javier Botella
- Institute for Health and Sport, Victoria University, Melbourne, Australia
- Metabolic Research Unit, Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Geelong, Australia
| | - David J Bishop
- Institute for Health and Sport, Victoria University, Melbourne, Australia
| |
Collapse
|
11
|
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: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 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
|
12
|
Prag HA, Aksentijevic D, Dannhorn A, Giles AV, Mulvey JF, Sauchanka O, Du L, Bates G, Reinhold J, Kula-Alwar D, Xu Z, Pellerin L, Goodwin RJA, Murphy MP, Krieg T. Ischemia-Selective Cardioprotection by Malonate for Ischemia/Reperfusion Injury. Circ Res 2022; 131:528-541. [PMID: 35959683 PMCID: PMC9426742 DOI: 10.1161/circresaha.121.320717] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Inhibiting SDH (succinate dehydrogenase), with the competitive inhibitor malonate, has shown promise in ameliorating ischemia/reperfusion injury. However, key for translation to the clinic is understanding the mechanism of malonate entry into cells to enable inhibition of SDH, its mitochondrial target, as malonate itself poorly permeates cellular membranes. The possibility of malonate selectively entering the at-risk heart tissue on reperfusion, however, remains unexplored. METHODS C57BL/6J mice, C2C12 and H9c2 myoblasts, and HeLa cells were used to elucidate the mechanism of selective malonate uptake into the ischemic heart upon reperfusion. Cells were treated with malonate while varying pH or together with transport inhibitors. Mouse hearts were either perfused ex vivo (Langendorff) or subjected to in vivo left anterior descending coronary artery ligation as models of ischemia/reperfusion injury. Succinate and malonate levels were assessed by liquid chromatography-tandem mass spectrometry LC-MS/MS, in vivo by mass spectrometry imaging, and infarct size by TTC (2,3,5-triphenyl-2H-tetrazolium chloride) staining. RESULTS Malonate was robustly protective against cardiac ischemia/reperfusion injury, but only if administered at reperfusion and not when infused before ischemia. The extent of malonate uptake into the heart was proportional to the duration of ischemia. Malonate entry into cardiomyocytes in vivo and in vitro was dramatically increased at the low pH (≈6.5) associated with ischemia. This increased uptake of malonate was blocked by selective inhibition of MCT1 (monocarboxylate transporter 1). Reperfusion of the ischemic heart region with malonate led to selective SDH inhibition in the at-risk region. Acid-formulation greatly enhances the cardioprotective potency of malonate. CONCLUSIONS Cardioprotection by malonate is dependent on its entry into cardiomyocytes. This is facilitated by the local decrease in pH that occurs during ischemia, leading to its selective uptake upon reperfusion into the at-risk tissue, via MCT1. Thus, malonate's preferential uptake in reperfused tissue means it is an at-risk tissue-selective drug that protects against cardiac ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Hiran A. Prag
- Department of Medicine (H.A.P., A.V.G., J.F.M., O.S., D.K.-A., M.P.M., T.K.), University of Cambridge, United Kingdom.,MRC Mitochondrial Biology Unit (H.A.P., A.V.G., G.B., J.R., M.M.P.), University of Cambridge, United Kingdom
| | - Dunja Aksentijevic
- Centre for Biochemical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom (D.A.)
| | - Andreas Dannhorn
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, United Kingdom (A.D., R.J.A.G.)
| | - Abigail V. Giles
- Department of Medicine (H.A.P., A.V.G., J.F.M., O.S., D.K.-A., M.P.M., T.K.), University of Cambridge, United Kingdom.,MRC Mitochondrial Biology Unit (H.A.P., A.V.G., G.B., J.R., M.M.P.), University of Cambridge, United Kingdom.,Laboratory of Cardiac Energetics, National Heart, Lung and Blood Institute, Bethesda, MD (A.V.G.)
| | - John F. Mulvey
- Department of Medicine (H.A.P., A.V.G., J.F.M., O.S., D.K.-A., M.P.M., T.K.), University of Cambridge, United Kingdom
| | - Olga Sauchanka
- Department of Medicine (H.A.P., A.V.G., J.F.M., O.S., D.K.-A., M.P.M., T.K.), University of Cambridge, United Kingdom
| | - Luping Du
- Department of Physiology and Pathophysiology, Tianjin Medical University, China (L.D., Z.X.)
| | - Georgina Bates
- MRC Mitochondrial Biology Unit (H.A.P., A.V.G., G.B., J.R., M.M.P.), University of Cambridge, United Kingdom
| | - Johannes Reinhold
- MRC Mitochondrial Biology Unit (H.A.P., A.V.G., G.B., J.R., M.M.P.), University of Cambridge, United Kingdom.,Faculty of Medicine and Health Sciences, University of East Anglia, Norwich Research Park (J.R.)
| | - Duvaraka Kula-Alwar
- Department of Medicine (H.A.P., A.V.G., J.F.M., O.S., D.K.-A., M.P.M., T.K.), University of Cambridge, United Kingdom
| | - Zhelong Xu
- Department of Physiology and Pathophysiology, Tianjin Medical University, China (L.D., Z.X.)
| | - Luc Pellerin
- Département de Physiologie, Université de Lausanne, Switzerland (L.P.).,Centre de Résonance Magnétique des Systèmes Biologiques, UMR5536 CNRS, LabEx TRAIL-IBIO, Université de Bordeaux, France (L.P.).,Inserm U1313, Université et CHU de Poitiers, France (L.P.)
| | - Richard J. A. Goodwin
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, United Kingdom (A.D., R.J.A.G.).,Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, United Kingdom (R.J.A.G.)
| | - Michael P. Murphy
- Department of Medicine (H.A.P., A.V.G., J.F.M., O.S., D.K.-A., M.P.M., T.K.), University of Cambridge, United Kingdom.,MRC Mitochondrial Biology Unit (H.A.P., A.V.G., G.B., J.R., M.M.P.), University of Cambridge, United Kingdom
| | - Thomas Krieg
- Department of Medicine (H.A.P., A.V.G., J.F.M., O.S., D.K.-A., M.P.M., T.K.), University of Cambridge, United Kingdom
| |
Collapse
|
13
|
Alipour M, Hajipour-Verdom B, Javan M, Abdolmaleki P. Static and Electromagnetic Fields Differently Affect Proliferation and Cell Death Through Acid Enhancement of ROS Generation in Mesenchymal Stem Cells. Radiat Res 2022; 198:384-395. [PMID: 35867630 DOI: 10.1667/rade-21-00037.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 06/17/2022] [Indexed: 11/03/2022]
Abstract
Magnetic fields remotely influence cellular homeostasis as a physical agent through the changes in cell physicochemical reactions. Magnetic fields affect cell fate, which may provide an important and interesting challenge in stem cell behaviors. Here, we investigated the effects of the static magnetic field (SMF, 20 mT) and electromagnetic field (EMF, 20 mT-50 Hz) on reactive oxygen species (ROS) production and the acidic pH conditions as stimuli to change cell cycle progression and cell death in mesenchymal stem cells. Results show that SMF, EMF, and their simultaneous (SMF+EMF) administration increase ROS and expression of nuclear factor erythroid 2-related factor 2 (Nrf2), superoxide dismutase 2 (SOD2), and glutathione-S-transferase (GST) as an antioxidant defense system. Besides, intracellular pH (pHi) decreases in presence of either EMF or SMF+EMF, but not SMF. Decreased ROS content using ascorbic acid in these treatments leads to increased pH compared to the magnetic field treatments alone. Furthermore, each magnetic field has different effects on the cellular process of stem cells, including cell cycle, apoptosis and necrosis. Moreover, treatment by SMF enhances the cell viability after 24 h, while EMF or SMF+EMF decreases it. These observations indicate that fluctuations of ROS generation and acid enhancement during SMF and EMF treatments may reveal their beneficial and adverse effects on the molecular and cellular mechanisms involved in the growth, death, and differentiation of stem cells.
Collapse
Affiliation(s)
| | | | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, 14115-154, Iran
| | | |
Collapse
|
14
|
Magierowska K, Korbut E, Wójcik-Grzybek D, Bakalarz D, Sliwowski Z, Cieszkowski J, Szetela M, Torregrossa R, Whiteman M, Magierowski M. Mitochondria-targeted hydrogen sulfide donors versus acute oxidative gastric mucosal injury. J Control Release 2022; 348:321-334. [PMID: 35654168 DOI: 10.1016/j.jconrel.2022.05.051] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 05/23/2022] [Accepted: 05/26/2022] [Indexed: 12/14/2022]
Abstract
Hydrogen sulfide (H2S) as a gaseous molecule prevents gastrointestinal (GI)-tract against various injuries. This study aimed to evaluate for the first time the detailed molecular mechanism of mitochondria-targeting H2S-prodrugs, AP39 and RT01 in gastroprotection against ischemia/reperfusion (I/R)-induced lesions. Wistar rats exposed to I/R were pretreated i.g. with vehicle, AP39 (0.004-2 mg/kg), RT01 (0.1 mg/kg), or with AP219 (0.1 mg/kg) as structural control without ability to release H2S. AP39 was also administered with mTOR1 inhibitor, rapamycin (1 mg/kg i.g.). Gastric damage area was assessed micro-/macroscopically, gastric blood flow (GBF) by laser flowmetry, mRNA level of HIF-1α, GPx, SOD1, SOD2, annexin-A1, SOCS3, IL-1RA, IL-1β, IL-1R1, IL-1R2, TNFR2, iNOS by real-time PCR. Gastric mucosal and/or serum content of IL-1β, IL-4, IL-5, IL-10, G-CSF, M-CSF, VEGFA, GRO, RANTES, MIP-1α, MCP1, TNF-α, TIMP1, FABP3, GST-α, STAT3/5 and phosphorylation of mTOR, NF-κB, ERK, Akt was evaluated by microbeads-fluorescent assay. Mitochondrial complexes activities were measured biochemically. RNA damage was assessed as 8-OHG by ELISA. AP39 and RT01 reduced micro-/macroscopic gastric I/R-injury increasing GBF. AP39-gastroprotection was accompanied by maintained activity of mitochondrial complexes, prevented RNA oxidation and enhanced mRNA/protein expression of SOCS3, IL-1RA, annexin-A1, GST-α, HIF-1α. Rapamycin reversed AP-39-gastroprotection. AP39-gastroprotection was followed by decreased NF-κB, ERK, IL-1β and enhanced Akt and mTOR proteins phosphorylation. AP39-prevented gastric mucosal damage caused by I/R-injury, partly by mitochondrial complex activity maintenance. AP39-mediated attenuation of gastric mucosal oxidation, hypoxia and inflammation involved mTOR1 and Akt pathways activity and modulation of HIF-1α, GST-α, SOCS3, IL1RA and TIMP1 molecular interplay.
Collapse
Affiliation(s)
| | - Edyta Korbut
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland
| | | | - Dominik Bakalarz
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland; Department of Forensic Toxicology, Institute of Forensic Research, Cracow, Poland
| | - Zbigniew Sliwowski
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland
| | - Jakub Cieszkowski
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland
| | - Małgorzata Szetela
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland
| | | | | | - Marcin Magierowski
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland.
| |
Collapse
|
15
|
Liu M, Huang Q, Zhu Y, Chen L, Li Y, Gong Z, Ai K. Harnessing reactive oxygen/nitrogen species and inflammation: Nanodrugs for liver injury. Mater Today Bio 2022; 13:100215. [PMID: 35198963 PMCID: PMC8850330 DOI: 10.1016/j.mtbio.2022.100215] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 02/04/2022] [Accepted: 02/06/2022] [Indexed: 12/11/2022] Open
Abstract
Overall, 12% of the global population (800 million) suffers from liver disease, which causes 2 million deaths every year. Liver injury involving characteristic reactive oxygen/nitrogen species (RONS) and inflammation plays a key role in progression of liver disease. As a key metabolic organ of the human body, the liver is susceptible to injury from various sources, including COVID-19 infection. Owing to unique structural features and functions of the liver, most current antioxidants and anti-inflammatory drugs are limited against liver injury. However, the characteristics of the liver could be utilized in the development of nanodrugs to achieve specific enrichment in the liver and consequently targeted treatment. Nanodrugs have shown significant potential in eliminating RONS and regulating inflammation, presenting an attractive therapeutic tool for liver disease through controlling liver injury. Therefore, the main aim of the current review is to provide a comprehensive summary of the latest developments contributing to our understanding of the mechanisms underlying nanodrugs in the treatment of liver injury via harnessing RONS and inflammation. Meanwhile, the prospects of nanodrugs for liver injury therapy are systematically discussed, which provides a sound platform for novel therapeutic insights and inspiration for design of nanodrugs to treat liver disease.
Collapse
Affiliation(s)
- Min Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Qiong Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yan Zhu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Li Chen
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Yumei Li
- Department of Assisted Reproduction, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Zhicheng Gong
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Kelong Ai
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| |
Collapse
|
16
|
Wang J, Tang X, Lu Y, Zheng Y, Zeng F, Shi W, Zhou P. Lycopene Regulates Dietary Dityrosine-Induced Mitochondrial-Lipid Homeostasis by Increasing Mitochondrial Complex Activity. Mol Nutr Food Res 2021; 66:e2100724. [PMID: 34780105 DOI: 10.1002/mnfr.202100724] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/13/2021] [Indexed: 12/22/2022]
Abstract
SCOPE Dityrosine (DT), a marker of protein oxidation, is widely found in many high-protein foods. Dietary intake of DT induces myocardial oxidative stress injury and impairs energy metabolism. Lycopene is a common dietary supplement with antioxidant and mitochondrial-lipid homeostasis modulating abilities. This study aimed to examine the effects of lycopene on DT-induced disturbances in myocardial function and energy metabolism. METHODS AND RESULTS Four-week-old C57BL/6J mice received intragastric administration of either tyrosine (420 µg kg-1 BW), DT (420 µg kg-1 BW), or lycopene at high (10 mg kg-1 BW) and low (5 mg kg-1 BW) doses for 35 days. Lycopene administration effectively reduced oxidative stress, cardiac fatty acid accumulation, and cardiac hypertrophy and improved mitochondrial performance in DT-induced mice. In vitro experiments in H9c2 cells showed that DT directly inhibited the activity of the respiratory chain complex, whereas oxidative phosphorylation and β-oxidation gene expression is upregulated. Lycopene enhanced the activity of the complexes and inhibited ROS production caused by compensatory regulation. CONCLUSION Lycopene improves DT-mediated myocardial energy homeostasis disorder by promoting the activity of respiratory chain complexes I and IV and alleviates the accumulation of cardiac fatty acids and myocardial hypertrophy.
Collapse
Affiliation(s)
- Jun Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China.,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Xue Tang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China.,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Yipin Lu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Yingying Zheng
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China.,National Enineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Fanhang Zeng
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China.,Collaborative Innovation Center of Food Safety and Quality Control, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Wentao Shi
- School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Peng Zhou
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China.,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| |
Collapse
|
17
|
Irisin: A Promising Target for Ischemia-Reperfusion Injury Therapy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5391706. [PMID: 34745418 PMCID: PMC8570861 DOI: 10.1155/2021/5391706] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/08/2021] [Accepted: 10/18/2021] [Indexed: 12/01/2022]
Abstract
Ischemia-reperfusion injury (IRI) is defined as the total combined damage that occurs during a period of ischemia and following the recovery of blood flow. Oxidative stress, mitochondrial dysfunction, and an inflammatory response are factors contributing to IRI-related damage that can each result in cell death. Irisin is a polypeptide that is proteolytically cleaved from the extracellular domain of fibronectin type III domain-containing protein 5 (FNDC5). Irisin acts as a myokine that potentially mediates beneficial effects of exercise by reducing oxidative stress, improving mitochondrial fitness, and suppressing inflammation. The existing literature also suggests a possible link between irisin and IRI, involving mechanisms similar to those associated with exercise. This article will review the pathogenesis of IRI and the potential benefits and current limitations of irisin as a therapeutic strategy for IRI, while highlighting the mechanistic correlations between irisin and IRI.
Collapse
|
18
|
Chen Y, Zhang J, Zhang M, Song Y, Zhang Y, Fan S, Ren S, Fu L, Zhang N, Hui H, Shen X. Baicalein resensitizes tamoxifen-resistant breast cancer cells by reducing aerobic glycolysis and reversing mitochondrial dysfunction via inhibition of hypoxia-inducible factor-1α. Clin Transl Med 2021; 11:e577. [PMID: 34841716 PMCID: PMC8567056 DOI: 10.1002/ctm2.577] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 09/03/2021] [Accepted: 09/06/2021] [Indexed: 12/19/2022] Open
Abstract
Drug resistance is a major hurdle for the effectiveness of tamoxifen (TAM) to provide clinical benefit. Therefore, it is essential to identify a sensitizer that could be used to improve TAM efficacy in treating TAM-resistant breast cancer. Here, we investigated the ability of baicalein to reverse TAM resistance. We found that baicalein increased the efficacy of TAM in inhibiting proliferation and inducing apoptosis of TAM-resistant cells. It also enhanced the TAM-induced growth reduction of resistant cells from NOD/SCID mouse mammary fat pads, without causing obvious systemic toxicity. Analyses using the CellMiner tool and the Kaplan-Meier plotter database showed that HIF-1α expression was inversely correlated with TAM therapeutic response in NCI-60 cancer cells and breast cancer patients. HIF-1α expression was increased in TAM-resistant cells due to an increase in mRNA levels and reduced ubiquitin-mediated degradation. Baicalein reduced HIF-1α expression by promoting its interaction with PHD2 and pVHL, thus facilitating ubiquitin ligase-mediated proteasomal degradation and thereby suppressing the nuclear translocation, binding to the hypoxia-response element, and transcriptional activity of HIF-1α. As a result, baicalein downregulated aerobic glycolysis by restricting glucose uptake, lactate production, ATP generation, lactate/pyruvate ratio and expression of HIF-1α-targeted glycolytic genes, thereby enhancing the antiproliferative efficacy of TAM. Furthermore, baicalein interfered with HIF-1α inhibition of mitochondrial biosynthesis, which increased mitochondrial DNA content and mitochondrial numbers, restored the generation of reactive oxygen species in mitochondria, and thus enhanced the TAM-induced mitochondrial apoptotic pathway. The HIF-1α stabilizer dimethyloxallyl glycine prevented the baicalein-induced downregulation of glycolysis and mitochondrial biosynthesis and reduced the effects of baicalein on reversing TAM resistance. Our results indicate that baicalein is a promising candidate to help overcome TAM resistance by sensitizing resistant cells to TAM-induced growth inhibition and apoptosis. The mechanism underlying the effects of baicalein consists of inhibition of HIF-1α-mediated aerobic glycolysis and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Yan Chen
- The State Key Laboratory of Functions and Applications of Medicinal PlantsGuizhou Medical UniversityGuizhouChina
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou ProvinceSchool of Pharmaceutical SciencesGuizhou Medical UniversityGuizhouChina
- The Union Key Laboratory of Guiyang City‐Guizhou Medical UniversitySchool of Pharmaceutical SciencesGuizhou Medical UniversityGuizhouChina
| | - Jingyu Zhang
- The State Key Laboratory of Functions and Applications of Medicinal PlantsGuizhou Medical UniversityGuizhouChina
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou ProvinceSchool of Pharmaceutical SciencesGuizhou Medical UniversityGuizhouChina
- The Union Key Laboratory of Guiyang City‐Guizhou Medical UniversitySchool of Pharmaceutical SciencesGuizhou Medical UniversityGuizhouChina
| | - Minqin Zhang
- The State Key Laboratory of Functions and Applications of Medicinal PlantsGuizhou Medical UniversityGuizhouChina
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou ProvinceSchool of Pharmaceutical SciencesGuizhou Medical UniversityGuizhouChina
- The Union Key Laboratory of Guiyang City‐Guizhou Medical UniversitySchool of Pharmaceutical SciencesGuizhou Medical UniversityGuizhouChina
| | - Yuxuan Song
- The State Key Laboratory of Functions and Applications of Medicinal PlantsGuizhou Medical UniversityGuizhouChina
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou ProvinceSchool of Pharmaceutical SciencesGuizhou Medical UniversityGuizhouChina
- The Union Key Laboratory of Guiyang City‐Guizhou Medical UniversitySchool of Pharmaceutical SciencesGuizhou Medical UniversityGuizhouChina
| | - Yue Zhang
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou ProvinceSchool of Pharmaceutical SciencesGuizhou Medical UniversityGuizhouChina
- The Key Laboratory of Optimal Utilization of Natural Medicine ResourcesSchool of Pharmaceutical SciencesGuizhou Medical UniversityGuizhouChina
| | - Shuangqin Fan
- The State Key Laboratory of Functions and Applications of Medicinal PlantsGuizhou Medical UniversityGuizhouChina
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou ProvinceSchool of Pharmaceutical SciencesGuizhou Medical UniversityGuizhouChina
- The Key Laboratory of Optimal Utilization of Natural Medicine ResourcesSchool of Pharmaceutical SciencesGuizhou Medical UniversityGuizhouChina
| | - Shuang Ren
- The State Key Laboratory of Functions and Applications of Medicinal PlantsGuizhou Medical UniversityGuizhouChina
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou ProvinceSchool of Pharmaceutical SciencesGuizhou Medical UniversityGuizhouChina
- The Union Key Laboratory of Guiyang City‐Guizhou Medical UniversitySchool of Pharmaceutical SciencesGuizhou Medical UniversityGuizhouChina
| | - Lingyun Fu
- The State Key Laboratory of Functions and Applications of Medicinal PlantsGuizhou Medical UniversityGuizhouChina
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou ProvinceSchool of Pharmaceutical SciencesGuizhou Medical UniversityGuizhouChina
- The Union Key Laboratory of Guiyang City‐Guizhou Medical UniversitySchool of Pharmaceutical SciencesGuizhou Medical UniversityGuizhouChina
| | - Nenling Zhang
- The State Key Laboratory of Functions and Applications of Medicinal PlantsGuizhou Medical UniversityGuizhouChina
- The Key Laboratory of Optimal Utilization of Natural Medicine ResourcesSchool of Pharmaceutical SciencesGuizhou Medical UniversityGuizhouChina
| | - Hui Hui
- State Key Laboratory of Natural MedicinesJiangsu Key Laboratory of Carcinogenesis and InterventionChina Pharmaceutical UniversityNanjingChina
| | - Xiangchun Shen
- The State Key Laboratory of Functions and Applications of Medicinal PlantsGuizhou Medical UniversityGuizhouChina
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou ProvinceSchool of Pharmaceutical SciencesGuizhou Medical UniversityGuizhouChina
- The Union Key Laboratory of Guiyang City‐Guizhou Medical UniversitySchool of Pharmaceutical SciencesGuizhou Medical UniversityGuizhouChina
- The Key Laboratory of Optimal Utilization of Natural Medicine ResourcesSchool of Pharmaceutical SciencesGuizhou Medical UniversityGuizhouChina
| |
Collapse
|
19
|
Zhao T, Wu W, Sui L, Huang Q, Nan Y, Liu J, Ai K. Reactive oxygen species-based nanomaterials for the treatment of myocardial ischemia reperfusion injuries. Bioact Mater 2021; 7:47-72. [PMID: 34466716 PMCID: PMC8377441 DOI: 10.1016/j.bioactmat.2021.06.006] [Citation(s) in RCA: 136] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 05/09/2021] [Accepted: 06/02/2021] [Indexed: 02/06/2023] Open
Abstract
Interventional coronary reperfusion strategies are widely adopted to treat acute myocardial infarction, but morbidity and mortality of acute myocardial infarction are still high. Reperfusion injuries are inevitable due to the generation of reactive oxygen species (ROS) and apoptosis of cardiac muscle cells. However, many antioxidant and anti-inflammatory drugs are largely limited by pharmacokinetics and route of administration, such as short half-life, low stability, low bioavailability, and side effects for treatment myocardial ischemia reperfusion injury. Therefore, it is necessary to develop effective drugs and technologies to address this issue. Fortunately, nanotherapies have demonstrated great opportunities for treating myocardial ischemia reperfusion injury. Compared with traditional drugs, nanodrugs can effectively increase the therapeutic effect and reduces side effects by improving pharmacokinetic and pharmacodynamic properties due to nanodrugs’ size, shape, and material characteristics. In this review, the biology of ROS and molecular mechanisms of myocardial ischemia reperfusion injury are discussed. Furthermore, we summarized the applications of ROS-based nanoparticles, highlighting the latest achievements of nanotechnology researches for the treatment of myocardial ischemia reperfusion injury. Cardiovascular diseases are the leading cause of death worldwide. Researches of the myocardial infarction pathology and development of new treatments have very important scientific significance in the biomedical field. Many nanomaterials have shown amazing therapeutic effects to reduce myocardial damage by eliminating ROS. Nanomaterials effectively reduced myocardial damage through eliminating ROS from NOXs, M-ETC, M-Ca2+, M-mPTP, and RIRR.
Collapse
Affiliation(s)
- Tianjiao Zhao
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410087, China.,Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410087, China
| | - Wei Wu
- Department of Geriatric Surgery, Xiangya Hospital, Central South University, Changsha, 410087, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410087, China
| | - Lihua Sui
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China.,Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China
| | - Qiong Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410087, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410087, China
| | - Yayun Nan
- Geriatric Medical Center, Ningxia People's Hospital, Yinchuan, 750003, China
| | - Jianhua Liu
- Department of Radiology, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Kelong Ai
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China.,Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China
| |
Collapse
|
20
|
Bae J, Paltzer WG, Mahmoud AI. The Role of Metabolism in Heart Failure and Regeneration. Front Cardiovasc Med 2021; 8:702920. [PMID: 34336958 PMCID: PMC8322239 DOI: 10.3389/fcvm.2021.702920] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/23/2021] [Indexed: 12/25/2022] Open
Abstract
Heart failure is the leading cause of death worldwide. The inability of the adult mammalian heart to regenerate following injury results in the development of systolic heart failure. Thus, identifying novel approaches toward regenerating the adult heart has enormous therapeutic potential for adult heart failure. Mitochondrial metabolism is an essential homeostatic process for maintaining growth and survival. The emerging role of mitochondrial metabolism in controlling cell fate and function is beginning to be appreciated. Recent evidence suggests that metabolism controls biological processes including cell proliferation and differentiation, which has profound implications during development and regeneration. The regenerative potential of the mammalian heart is lost by the first week of postnatal development when cardiomyocytes exit the cell cycle and become terminally differentiated. This inability to regenerate following injury is correlated with the metabolic shift from glycolysis to fatty acid oxidation that occurs during heart maturation in the postnatal heart. Thus, understanding the mechanisms that regulate cardiac metabolism is key to unlocking metabolic interventions during development, disease, and regeneration. In this review, we will focus on the emerging role of metabolism in cardiac development and regeneration and discuss the potential of targeting metabolism for treatment of heart failure.
Collapse
Affiliation(s)
- Jiyoung Bae
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, United States
| | - Wyatt G Paltzer
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, United States
| | - Ahmed I Mahmoud
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, United States
| |
Collapse
|
21
|
Hidalgo-Gutiérrez A, González-García P, Díaz-Casado ME, Barriocanal-Casado E, López-Herrador S, Quinzii CM, López LC. Metabolic Targets of Coenzyme Q10 in Mitochondria. Antioxidants (Basel) 2021; 10:520. [PMID: 33810539 PMCID: PMC8066821 DOI: 10.3390/antiox10040520] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/14/2021] [Accepted: 03/23/2021] [Indexed: 12/11/2022] Open
Abstract
Coenzyme Q10 (CoQ10) is classically viewed as an important endogenous antioxidant and key component of the mitochondrial respiratory chain. For this second function, CoQ molecules seem to be dynamically segmented in a pool attached and engulfed by the super-complexes I + III, and a free pool available for complex II or any other mitochondrial enzyme that uses CoQ as a cofactor. This CoQ-free pool is, therefore, used by enzymes that link the mitochondrial respiratory chain to other pathways, such as the pyrimidine de novo biosynthesis, fatty acid β-oxidation and amino acid catabolism, glycine metabolism, proline, glyoxylate and arginine metabolism, and sulfide oxidation metabolism. Some of these mitochondrial pathways are also connected to metabolic pathways in other compartments of the cell and, consequently, CoQ could indirectly modulate metabolic pathways located outside the mitochondria. Thus, we review the most relevant findings in all these metabolic functions of CoQ and their relations with the pathomechanisms of some metabolic diseases, highlighting some future perspectives and potential therapeutic implications.
Collapse
Affiliation(s)
- Agustín Hidalgo-Gutiérrez
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (P.G.-G.); (M.E.D.-C.); (E.B.-C.); (S.L.-H.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
| | - Pilar González-García
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (P.G.-G.); (M.E.D.-C.); (E.B.-C.); (S.L.-H.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
| | - María Elena Díaz-Casado
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (P.G.-G.); (M.E.D.-C.); (E.B.-C.); (S.L.-H.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
| | - Eliana Barriocanal-Casado
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (P.G.-G.); (M.E.D.-C.); (E.B.-C.); (S.L.-H.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
| | - Sergio López-Herrador
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (P.G.-G.); (M.E.D.-C.); (E.B.-C.); (S.L.-H.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
| | - Catarina M. Quinzii
- Department of Neurology, Columbia University Medical Center, New York, NY 10032, USA;
| | - Luis C. López
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (P.G.-G.); (M.E.D.-C.); (E.B.-C.); (S.L.-H.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
| |
Collapse
|
22
|
Dambrova M, Zuurbier CJ, Borutaite V, Liepinsh E, Makrecka-Kuka M. Energy substrate metabolism and mitochondrial oxidative stress in cardiac ischemia/reperfusion injury. Free Radic Biol Med 2021; 165:24-37. [PMID: 33484825 DOI: 10.1016/j.freeradbiomed.2021.01.036] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/13/2021] [Accepted: 01/14/2021] [Indexed: 12/12/2022]
Abstract
The heart is the most metabolically flexible organ with respect to the use of substrates available in different states of energy metabolism. Cardiac mitochondria sense substrate availability and ensure the efficiency of oxidative phosphorylation and heart function. Mitochondria also play a critical role in cardiac ischemia/reperfusion injury, during which they are directly involved in ROS-producing pathophysiological mechanisms. This review explores the mechanisms of ROS production within the energy metabolism pathways and focuses on the impact of different substrates. We describe the main metabolites accumulating during ischemia in the glucose, fatty acid, and Krebs cycle pathways. Hyperglycemia, often present in the acute stress condition of ischemia/reperfusion, increases cytosolic ROS concentrations through the activation of NADPH oxidase 2 and increases mitochondrial ROS through the metabolic overloading and decreased binding of hexokinase II to mitochondria. Fatty acid-linked ROS production is related to the increased fatty acid flux and corresponding accumulation of long-chain acylcarnitines. Succinate that accumulates during anoxia/ischemia is suggested to be the main source of ROS, and the role of itaconate as an inhibitor of succinate dehydrogenase is emerging. We discuss the strategies to modulate and counteract the accumulation of substrates that yield ROS and the therapeutic implications of this concept.
Collapse
Affiliation(s)
- Maija Dambrova
- Latvian Institute of Organic Synthesis, Riga, Latvia; Riga Stradins University, Riga, Latvia.
| | - Coert J Zuurbier
- Amsterdam UMC, University of Amsterdam, Laboratory of Experimental Intensive Care and Anesthesiology, Department of Anesthesiology, Amsterdam Cardiovascular Sciences, Meibergdreef 9, AZ 1105, Amsterdam, the Netherlands
| | - Vilmante Borutaite
- Neuroscience Institute, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | | | | |
Collapse
|