1
|
Zhang H, Xu T, Mei X, Zhao Q, Yang Q, Zeng X, Ma Z, Zhou H, Zeng Q, Xu D, Ren H. PINK1 modulates Prdx2 to reduce lipotoxicity-induced apoptosis and attenuate cardiac dysfunction in heart failure mice with a preserved ejection fraction. Clin Transl Med 2025; 15:e70166. [PMID: 39763059 PMCID: PMC11705485 DOI: 10.1002/ctm2.70166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 12/12/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
INTRODUCTION Heart failure with preserved ejection fraction (HFpEF) is a complex condition characterized by metabolic dysfunction and myocardial lipotoxicity. The roles of PTEN-induced kinase 1 (PINK1) and peroxiredoxin-2 (Prdx2) in HFpEF pathogenesis remain unclear. OBJECTIVE This study aimed to investigate the interaction between PINK1 and Prdx2 to mitigate cardiac diastolic dysfunction in HFpEF. METHODS In vivo, PINK1-knockout mice and cardiac-specific PINK1-overexpressing transgenic mice were used to establish an HFpEF mouse model via a high-fat diet and L-NAME. Myocardial lipotoxicity was induced by palmitic acid in vitro. Immunoprecipitation, western blotting and immunofluorescence analysis were performed to elucidate the molecular mechanisms involved. RESULTS We determined that PINK1 and Prdx2 were downregulated in the HFpEF mouse model. In vivo, PINK1 ablation exacerbated the reduction in Prdx2 expression, worsening cardiac dysfunction in HFpEF mice. Conversely, PINK1 overexpression restored Prdx2 levels and decreased reactive oxygen species and apoptosis, thereby reducing fibrosis and inflammation and ameliorating cardiac diastolic dysfunction in HFpEF mice. In vitro, an interaction between the N-terminal region (amino acids 1-133) of PINK1 and Prdx2 was identified. The overexpression of PINK1 induced Prdx2 expression and effectively attenuated palmitic acid-induced apoptosis through the c-Jun amino-terminal kinase (JNK) and mitogen-activated protein kinase (p38) pathways, whereas siRNA-mediated Prdx2 knockdown abolished the protective effect of PINK1. CONCLUSION PINK1 alleviates lipotoxicity-induced myocardial apoptosis and improves diastolic dysfunction in HFpEF through Prdx2, highlighting PINK1 overexpression as a potential therapeutic strategy for HFpEF. KEY POINTS Our investigation discloses a pivotal relationship between PINK1 and Prdx2 in the context of HFpEF. Notably, PINK1, in addition to its role in mitochondrial autophagy, can increase Prdx2 expression, effectively remove ROS and attenuate cardiomyocyte apoptosis by modulating the JNK and p38 pathways, thereby alleviating myocardial lipotoxicity and improving HFpEF cardiac function. Our studies offer valuable insights, opening avenues for the development of innovative therapeutic strategies in the prevention and treatment of HFpEF.
Collapse
Affiliation(s)
- Hao Zhang
- State Key Laboratory of Organ Failure ResearchDepartment of CardiologyNanfang HospitalSouthern Medical UniversityGuangzhouChina
- Key Laboratory For Organ Failure ResearchMinistry of Education of the People's Republic of ChinaGuangzhouChina
| | - Tianyu Xu
- NHC Key Laboratory of Assisted Circulation, Department of CardiologyThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Xiyuan Mei
- State Key Laboratory of Organ Failure ResearchDepartment of CardiologyNanfang HospitalSouthern Medical UniversityGuangzhouChina
- Key Laboratory For Organ Failure ResearchMinistry of Education of the People's Republic of ChinaGuangzhouChina
| | - Qiming Zhao
- State Key Laboratory of Organ Failure ResearchDepartment of CardiologyNanfang HospitalSouthern Medical UniversityGuangzhouChina
- Key Laboratory For Organ Failure ResearchMinistry of Education of the People's Republic of ChinaGuangzhouChina
| | - Qiling Yang
- State Key Laboratory of Organ Failure ResearchDepartment of CardiologyNanfang HospitalSouthern Medical UniversityGuangzhouChina
- Key Laboratory For Organ Failure ResearchMinistry of Education of the People's Republic of ChinaGuangzhouChina
| | - Xianghui Zeng
- State Key Laboratory of Organ Failure ResearchDepartment of CardiologyNanfang HospitalSouthern Medical UniversityGuangzhouChina
- Key Laboratory For Organ Failure ResearchMinistry of Education of the People's Republic of ChinaGuangzhouChina
| | - Zhuang Ma
- State Key Laboratory of Organ Failure ResearchDepartment of CardiologyNanfang HospitalSouthern Medical UniversityGuangzhouChina
- Key Laboratory For Organ Failure ResearchMinistry of Education of the People's Republic of ChinaGuangzhouChina
| | - Haobin Zhou
- State Key Laboratory of Organ Failure ResearchDepartment of CardiologyNanfang HospitalSouthern Medical UniversityGuangzhouChina
- Key Laboratory For Organ Failure ResearchMinistry of Education of the People's Republic of ChinaGuangzhouChina
| | - Qingchun Zeng
- State Key Laboratory of Organ Failure ResearchDepartment of CardiologyNanfang HospitalSouthern Medical UniversityGuangzhouChina
- Key Laboratory For Organ Failure ResearchMinistry of Education of the People's Republic of ChinaGuangzhouChina
| | - Dingli Xu
- State Key Laboratory of Organ Failure ResearchDepartment of CardiologyNanfang HospitalSouthern Medical UniversityGuangzhouChina
- Key Laboratory For Organ Failure ResearchMinistry of Education of the People's Republic of ChinaGuangzhouChina
| | - Hao Ren
- Key Laboratory For Organ Failure ResearchMinistry of Education of the People's Republic of ChinaGuangzhouChina
- Department of RheumatologyNanfang HospitalSouthern Medical UniversityGuangzhouChina
| |
Collapse
|
2
|
Kweon HY, Song EJ, Jeong SJ, Lee S, Sonn SK, Seo S, Jin J, Kim S, Kim TK, Moon SH, Kim D, Park YM, Woo HA, Oh GT. Extracellular peroxiredoxin 5 exacerbates atherosclerosis via the TLR4/MyD88 pathway. Atherosclerosis 2025; 400:119052. [PMID: 39549462 DOI: 10.1016/j.atherosclerosis.2024.119052] [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: 05/20/2024] [Revised: 11/08/2024] [Accepted: 11/08/2024] [Indexed: 11/18/2024]
Abstract
BACKGROUNGD AND AIMS Peroxiredoxin 5 (PRDX5), an atypical 2-Cys peroxiredoxin (PRDX), is known to regulate global oxidative stresses and inflammatory responses. Inflammation and oxidative stress are pivotal factors in the development of atherosclerosis, especially in the context of vascular endothelial dysfunction. However, effects of PRDX5 on atherosclerosis remain unclear. This study aimed to elucidate the role of PRDX5 in the pathogenesis of atherosclerosis. METHODS For in vivo analysis, normal chow diet 60-week old Apolipoprotein E knockout (ApoE-/-) and Prdx5-/-; ApoE-/- mice were used for the experiments. For in vitro analysis, human umbilical vein endothelial cells (HUVECs) were stimulated with oxidized LDL (oxLDL; 50 ng/ml) for 24hrs, following serum starvation by incubation with serum-free Endothelial Cell Growth Medium-2 (EGM-2) for 1hr. RESULTS We observed elevated PRDX5 expression under atherosclerotic conditions in both humans and mice. Unexpectedly, Prdx5-/-; ApoE-/- mice exhibited reduced plaque formation, with no discernible difference in aortic hydrogen peroxide (H2O2) levels compared to ApoE-/- mice. Additionally, there was a notable decrease in macrophage accumulation and vascular inflammation in the atherosclerotic aorta of Prdx5-/-; ApoE-/-. In vitro, HUVECs stimulated with oxLDL showed upregulated PRDX5 expression in both lysate and culture medium. Moreover, PRDX5 knockdown in oxLDL-stimulated (oxLDL-siPRDX5) HUVECs significantly reduced the migration and adhesion of human monocytic cells (THP-1) to HUVECs, indicating diminished vascular immune responses. Mechanistically, both in vivo and in vitro, PRDX5 deficiency inhibited the Toll-like receptor 4 (TLR4)/Myeloid differentiation primary response 88 (MyD88) signaling pathway, resulting in reduced nuclear factor kappa B (NF-κB) and P38 phosphorylation. Furthermore, treatment with recombinant PRDX5 (rPRDX5) protein restored TLR4/MyD88 signaling in oxLDL-siPRDX5 HUVECs. CONCLUSIONS These data demonstrate that extracellular PRDX5 contributes to endothelial inflammation, promoting macrophage accumulation in the atherosclerotic aorta through activation of TLR4/MyD88/NF-κB and P38 signaling pathways, thereby exacerbating the progression of atherosclerosis.
Collapse
Affiliation(s)
- Hyae Yon Kweon
- Heart-Immune-Brain Network Research Center, Department of Life Science and College of Natural Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Eun Ju Song
- Heart-Immune-Brain Network Research Center, Department of Life Science and College of Natural Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Se-Jin Jeong
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, Saint Louis, United States
| | - SoonHo Lee
- Heart-Immune-Brain Network Research Center, Department of Life Science and College of Natural Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Seong-Keun Sonn
- Heart-Immune-Brain Network Research Center, Department of Life Science and College of Natural Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Seungwoon Seo
- Imvastech Inc., 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, Republic of Korea
| | - Jing Jin
- Heart-Immune-Brain Network Research Center, Department of Life Science and College of Natural Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Sinai Kim
- Heart-Immune-Brain Network Research Center, Department of Life Science and College of Natural Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Tae Kyeong Kim
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Dr, Farmington, CT 06032, United States
| | - Shin Hye Moon
- Heart-Immune-Brain Network Research Center, Department of Life Science and College of Natural Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Doyeon Kim
- Heart-Immune-Brain Network Research Center, Department of Life Science and College of Natural Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Young Mi Park
- Department of Molecular Medicine, Ewha Womans University School of Medicine, Seoul, 03760, Republic of Korea
| | - Hyun Ae Woo
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea; College of Pharmacy, Graduate School of Applied Science and Technology for Skin Health and Aesthetics, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Goo Taeg Oh
- Heart-Immune-Brain Network Research Center, Department of Life Science and College of Natural Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea; Imvastech Inc., 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, Republic of Korea.
| |
Collapse
|
3
|
Shao R, Chen R, Zheng Q, Yao M, Li K, Cao Y, Jiang L. Oxidative stress disrupts vascular microenvironmental homeostasis affecting the development of atherosclerosis. Cell Biol Int 2024; 48:1781-1801. [PMID: 39370593 DOI: 10.1002/cbin.12239] [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: 02/11/2024] [Revised: 05/20/2024] [Accepted: 06/10/2024] [Indexed: 10/08/2024]
Abstract
Atherosclerosis is primarily an inflammatory reaction of the cardiovascular system caused by endothelial damage, leading to progressive thickening and hardening of the vessel walls, as well as extensive necrosis and fibrosis of the surrounding tissues, the most necessary pathological process causing cardiovascular disease. When the body responds to harmful internal and external stimuli, excess oxygen free radicals are produced causing oxidative stress to occur in cells and tissues. Simultaneously, the activation of inflammatory immunological processes is followed by an elevation in oxygen free radicals, which directly initiates the release of cytokines and chemokines, resulting in a detrimental cycle of vascular homeostasis abnormalities. Oxidative stress contributes to the harm inflicted upon vascular endothelial cells and the decrease in nitric oxide levels. Nitric oxide is crucial for maintaining vascular homeostasis and is implicated in the development of atherosclerosis. This study examines the influence of oxidative stress on the formation of atherosclerosis, which is facilitated by the vascular milieu. It also provides an overview of the pertinent targets and pharmaceutical approaches for treating this condition.
Collapse
Affiliation(s)
- Ruifei Shao
- Medical School, Center for Translational Research in Clinical Medicine, Kunming University of Science and Technology, Kunming, China
| | - Rui Chen
- Yan'an Hospital Affiliated to Kunming Medical University, Kunming, China
| | - Qiang Zheng
- Medical School, Center for Translational Research in Clinical Medicine, Kunming University of Science and Technology, Kunming, China
| | - Mengyu Yao
- Medical School, Center for Translational Research in Clinical Medicine, Kunming University of Science and Technology, Kunming, China
| | - Kunlin Li
- Department of General Surgery II, The First People's Hospital of Yunnan Province, Kunming, China
| | - Yu Cao
- Yunnan Key Laboratory of Innovative Application of Traditional Chinese Medicine, Department of Cardiovascular Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Lihong Jiang
- Medical School, Center for Translational Research in Clinical Medicine, Kunming University of Science and Technology, Kunming, China
- Yunnan Key Laboratory of Innovative Application of Traditional Chinese Medicine, Department of Cardiovascular Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
4
|
Pawar HD, Patil Y, Patil A, Nakhate KT, Agrawal YO, Suchal K, Ojha S, Goyal SN. Cardioprotective effect of CB1 receptor antagonist AM251 against β receptor-stimulated myocardial infarction via modulation of NF-kB signaling pathway in diabetic mice. Heliyon 2024; 10:e35138. [PMID: 39161822 PMCID: PMC11332847 DOI: 10.1016/j.heliyon.2024.e35138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 07/23/2024] [Accepted: 07/23/2024] [Indexed: 08/21/2024] Open
Abstract
We substantiated the effect of AM251, a cannabinoid receptor-1 (CB1R) antagonist, against β-receptor stimulated myocardial infarction (MI) in streptozotocin (STZ)-induced diabetic mice via modulation- of the NF-kB signaling pathway. The different parameters were assessed such as ECG, hemodynamic, cardiac injury markers, oxidative stress parameters, pro-inflammatory cytokines, and histopathological abnormalities. Mice were fed a high-fat diet for 30 days. On day 7, to trigger diabetes, 150 mg/kg of STZ was injected intraperitoneally. On day 10, to determine whether diabetes developed, the blood level of glucose was monitored. From days 11-30, diabetic mice were injected with either CB1R agonist oleamide or antagonist AM251 or both, with concurrent administrations of β-agonist isoproterenol on days 28 and 29 to induce MI. In comparison to normal, the myocardial infarcted diabetic animals demonstrated alterations in ECG, hemodynamic profiles, and diminished enzymatic activities (CK-MB, LDH, SOD, GSH, catalase), with concurrently increased MDA levels, which indicated increased oxidative stress in the myocardium. Additionally, higher concentrations of cytokines that signal myocardial inflammation, such as IL-1β, IL-6, and TNF-α, were also noted. Furthermore, elevated myonecrosis, edema, and cell infiltration which is confirmed by histopathology of heart tissue. Treatment with AM251 significantly ameliorated myocardial redox status, reduced cytokines, and repaired enzymatic activities leading to subsequent recovery in cardiac function. AM251 effectively suppressed myonecrosis and edema. This study also showed that AM251 protects against myocardial inflammation and oxidative stress triggered by isoproterenol by blocking NF-kB signalling pathway. However, upregulation of the CB1R through oleamide showed significant cardiac toxicity. Conversely, the concurrent administration of oleamide and AM251 failed to induce cardiotoxic effects in isoproterenol-induced MI in diabetic mice which indicates downregulation of the CB1R might be associated with the cardioprotective effect.
Collapse
Affiliation(s)
- Harshal D. Pawar
- Department of Pharmacology, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, 424001, Maharashtra, India
| | - Yugandhara Patil
- Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, 425405, Maharashtra, India
| | - Ashwani Patil
- Department of Pharmacology, Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, 411018, Maharashtra, India
| | - Kartik T. Nakhate
- Department of Pharmacology, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, 424001, Maharashtra, India
| | - Yogeeta O. Agrawal
- Department of Pharmaceutics, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, 424001, Maharashtra, India
| | - Kapil Suchal
- VMI, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, P.O. Box 15551, United Arab Emirates
| | - Sameer N. Goyal
- Department of Pharmacology, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, 424001, Maharashtra, India
| |
Collapse
|
5
|
Sherratt SCR, Libby P, Dawoud H, Bhatt DL, Mason RP. Eicosapentaenoic Acid Improves Endothelial Nitric Oxide Bioavailability Via Changes in Protein Expression During Inflammation. J Am Heart Assoc 2024; 13:e034076. [PMID: 38958135 PMCID: PMC11292741 DOI: 10.1161/jaha.123.034076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 05/14/2024] [Indexed: 07/04/2024]
Abstract
BACKGROUND Endothelial cell (EC) dysfunction involves reduced nitric oxide (NO) bioavailability due to NO synthase uncoupling linked to increased oxidation and reduced cofactor availability. Loss of endothelial function and NO bioavailability are associated with inflammation, including leukocyte activation. Eicosapentaenoic acid (EPA) administered as icosapent ethyl reduced cardiovascular events in REDUCE-IT (Reduction of Cardiovascular Events With Icosapent Ethyl-Intervention Trial) in relation to on-treatment EPA blood levels. The mechanisms of cardiovascular protection for EPA remain incompletely elucidated but likely involve direct effects on the endothelium. METHODS AND RESULTS In this study, human ECs were treated with EPA and challenged with the cytokine IL-6 (interleukin-6). Proinflammatory responses in the ECs were confirmed by ELISA capture of sICAM-1 (soluble intercellular adhesion molecule-1) and TNF-α (tumor necrosis factor-α). Global protein expression was determined using liquid chromatography-mass spectrometry tandem mass tag. Release kinetics of NO and peroxynitrite were monitored using porphyrinic nanosensors. IL-6 challenge induced proinflammatory responses from the ECs as evidenced by increased release of sICAM-1 and TNF-α, which correlated with a loss of NO bioavailability. ECs pretreated with EPA modulated expression of 327 proteins by >1-fold (P<0.05), compared with IL-6 alone. EPA augmented expression of proteins involved in NO production, including heme oxygenase-1 and dimethylarginine dimethylaminohydrolase-1, and 34 proteins annotated as associated with neutrophil degranulation. EPA reversed the endothelial NO synthase uncoupling induced by IL-6 as evidenced by an increased [NO]/[peroxynitrite] release ratio (P<0.05). CONCLUSIONS These direct actions of EPA on EC functions during inflammation may contribute to its distinct cardiovascular benefits.
Collapse
Affiliation(s)
- Samuel C. R. Sherratt
- Department of Molecular, Cellular, and Biomedical SciencesUniversity of New HampshireDurhamNHUSA
- Elucida ResearchBeverlyMAUSA
- Mount Sinai Fuster Heart HospitalIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Peter Libby
- Department of Medicine, Cardiovascular Division, Brigham and Women’s HospitalHarvard Medical SchoolBostonMAUSA
| | - Hazem Dawoud
- Nanomedical Research LaboratoryOhio UniversityAthensOHUSA
| | - Deepak L. Bhatt
- Mount Sinai Fuster Heart HospitalIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - R. Preston Mason
- Elucida ResearchBeverlyMAUSA
- Department of Medicine, Cardiovascular Division, Brigham and Women’s HospitalHarvard Medical SchoolBostonMAUSA
| |
Collapse
|
6
|
Buneeva OA, Fedchenko VI, Kaloshina SA, Zavyalova MG, Zgoda VG, Medvedev AE. Proteomic profiling of renal tissue of normo- and hypertensive rats with the renalase peptide RP220 as an affinity ligand. BIOMEDITSINSKAIA KHIMIIA 2024; 70:145-155. [PMID: 38940203 DOI: 10.18097/pbmc20247003145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
Abstract
Renalase (RNLS) is a recently discovered protein that plays an important role in the regulation of blood pressure by acting inside and outside cells. Intracellular RNLS is a FAD-dependent oxidoreductase that oxidizes isomeric forms of β-NAD(P)H. Extracellular renalase lacking its N-terminal peptide and cofactor FAD exerts various protective effects via non-catalytic mechanisms. Certain experimental evidence exists in the literature that the RP220 peptide (a 20-mer peptide corresponding to the amino acid sequence RNLS 220-239) reproduces a number of non-catalytic effects of this protein, acting on receptor proteins of the plasma membrane. The possibility of interaction of this peptide with intracellular proteins has not been studied. Taking into consideration the known role of RNLS as a possible antihypertensive factor, the aim of this study was to perform proteomic profiling of the kidneys of normotensive and hypertensive rats using RP220 as an affinity ligand. Proteomic (semi-quantitative) identification revealed changes in the relative content of about 200 individual proteins in the kidneys of hypertensive rats bound to the affinity sorbent as compared to the kidneys of normotensive animals. Increased binding of SHR renal proteins to RP220 over the normotensive control was found for proteins involved in the development of cardiovascular pathology. Decreased binding of the kidney proteins from hypertensive animals to RP220 was noted for components of the ubiquitin-proteasome system, ribosomes, and cytoskeleton.
Collapse
Affiliation(s)
- O A Buneeva
- Institute of Biomedical Chemistry, Moscow, Russia
| | | | | | | | - V G Zgoda
- Institute of Biomedical Chemistry, Moscow, Russia
| | - A E Medvedev
- Institute of Biomedical Chemistry, Moscow, Russia
| |
Collapse
|
7
|
Balasubramanian P, Vijayarangam V, Deviparasakthi MKG, Palaniyandi T, Ravi M, Natarajan S, Viswanathan S, Baskar G, Wahab MRA, Surendran H. Implications and progression of peroxiredoxin 2 (PRDX2) in various human diseases. Pathol Res Pract 2024; 254:155080. [PMID: 38219498 DOI: 10.1016/j.prp.2023.155080] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/24/2023] [Accepted: 12/30/2023] [Indexed: 01/16/2024]
Abstract
Peroxiredoxin 2 (PRDX2), a characteristic 2-Cys enzyme is one of the foremost effective scavenger proteins against reactive oxygen species (ROS) and hydrogen peroxide (H2O2) defending cells against oxidative stress. Dysregulation of this antioxidant raises the quantity of ROS and oxidative stress implicated in several diseases. PRDX2 lowers the generation of ROS that takes part in controlling several signalling pathways occurring in neurons, protecting them from stress caused by oxidation and an inflammatory harm. Depending on the aetiological variables, the kind of cancer, and the stage of tumour development, PRDX2 may behave either as an onco-suppressor or a promoter. However, overexpression of PRDX2 may be linked to the development of numerous cancers, including those of the colon, cervix, breast, and prostate. PRDX2 also plays a beneficial effect in inflammatory diseases. PRDX2 being a thiol-specific peroxidase, is known to control proinflammatory reactions. The spilling of PRDX2, on the other hand, accelerates cognitive impairment following a stroke by triggering an inflammatory reflex. PRDX2 expression patterns in vascular cells tend to be crucial to its involvement in cardiovascular diseases. In vascular smooth muscle cells, if the protein tyrosine phosphatase is restricted, PRDX2 could avoid the neointimal thickening which relies on platelet derived growth factor (PDGF), a vital component of vascular remodelling. A proper PRDX2 balance is therefore crucial. The imbalance causes a number of illnesses, including cancers, inflammatory diseases, cardiovascular ailments, and neurological and neurodegenerative problems which are discussed in this review.
Collapse
Affiliation(s)
| | - Varshini Vijayarangam
- Department of Biotechnology, Dr. M.G.R. Educational and Research Institute, Chennai, India
| | | | - Thirunavukkarasu Palaniyandi
- Department of Biotechnology, Dr. M.G.R. Educational and Research Institute, Chennai, India; Department of Anatomy, Biomedical Research Unit and Laboratory Animal Centre, Saveetha Dental College and Hospital, SIMATS, Saveetha University, Chennai, India.
| | - Maddaly Ravi
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | - Sudhakar Natarajan
- Department of Tuberculosis, ICMR - National Institute for Research in Tuberculosis (NIRT), Chennai, India
| | - Sandhiya Viswanathan
- Department of Biotechnology, Dr. M.G.R. Educational and Research Institute, Chennai, India
| | - Gomathy Baskar
- Department of Biotechnology, Dr. M.G.R. Educational and Research Institute, Chennai, India
| | | | - Hemapreethi Surendran
- Department of Biotechnology, Dr. M.G.R. Educational and Research Institute, Chennai, India
| |
Collapse
|
8
|
Richman TR, Ermer JA, Baker J, Siira SJ, Kile BT, Linden MD, Rackham O, Filipovska A. Mitochondrial gene expression is required for platelet function and blood clotting. Cell Rep 2023; 42:113312. [PMID: 37889747 DOI: 10.1016/j.celrep.2023.113312] [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: 06/15/2022] [Revised: 07/20/2023] [Accepted: 10/05/2023] [Indexed: 10/29/2023] Open
Abstract
Platelets are anucleate blood cells that contain mitochondria and regulate blood clotting in response to injury. Mitochondria contain their own gene expression machinery that relies on nuclear-encoded factors for the biogenesis of the oxidative phosphorylation system to produce energy required for thrombosis. The autonomy of the mitochondrial gene expression machinery from the nucleus is unclear, and platelets provide a valuable model to understand its importance in anucleate cells. Here, we conditionally delete Elac2, Ptcd1, or Mtif3 in platelets, which are essential for mitochondrial gene expression at the level of RNA processing, stability, or translation, respectively. Loss of ELAC2, PTCD1, or MTIF3 leads to increased megakaryocyte ploidy, elevated circulating levels of reticulated platelets, thrombocytopenia, and consequent extended bleeding time. Impaired mitochondrial gene expression reduces agonist-induced platelet activation. Transcriptomic and proteomic analyses show that mitochondrial gene expression is required for fibrinolysis, hemostasis, and blood coagulation in response to injury.
Collapse
Affiliation(s)
- Tara R Richman
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA 6009, Australia; ARC Centre of Excellence in Synthetic Biology, QEII Medical Centre, Nedlands, WA 6009, Australia; Centre for Medical Research, The University of Western Australia, QEII Medical Centre, Nedlands, WA 6009, Australia; Telethon Kids Institute, Northern Entrance, Perth Children's Hospital, 15 Hospital Avenue, Nedlands, WA, Australia
| | - Judith A Ermer
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA 6009, Australia; ARC Centre of Excellence in Synthetic Biology, QEII Medical Centre, Nedlands, WA 6009, Australia; Centre for Medical Research, The University of Western Australia, QEII Medical Centre, Nedlands, WA 6009, Australia
| | - Jessica Baker
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA 6009, Australia; ARC Centre of Excellence in Synthetic Biology, QEII Medical Centre, Nedlands, WA 6009, Australia; Centre for Medical Research, The University of Western Australia, QEII Medical Centre, Nedlands, WA 6009, Australia; Telethon Kids Institute, Northern Entrance, Perth Children's Hospital, 15 Hospital Avenue, Nedlands, WA, Australia
| | - Stefan J Siira
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA 6009, Australia; ARC Centre of Excellence in Synthetic Biology, QEII Medical Centre, Nedlands, WA 6009, Australia; Centre for Medical Research, The University of Western Australia, QEII Medical Centre, Nedlands, WA 6009, Australia; Telethon Kids Institute, Northern Entrance, Perth Children's Hospital, 15 Hospital Avenue, Nedlands, WA, Australia
| | - Benjamin T Kile
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - Matthew D Linden
- Pathology and Laboratory Science, The University of Western Australia, Perth, WA, Australia
| | - Oliver Rackham
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA 6009, Australia; ARC Centre of Excellence in Synthetic Biology, QEII Medical Centre, Nedlands, WA 6009, Australia; Telethon Kids Institute, Northern Entrance, Perth Children's Hospital, 15 Hospital Avenue, Nedlands, WA, Australia; Curtin Medical School, Curtin University, Bentley, WA 6102, Australia; Curtin Health Innovation Research Institute, Curtin University, Bentley, WA 6102, Australia
| | - Aleksandra Filipovska
- ARC Centre of Excellence in Synthetic Biology, QEII Medical Centre, Nedlands, WA 6009, Australia; Telethon Kids Institute, Northern Entrance, Perth Children's Hospital, 15 Hospital Avenue, Nedlands, WA, Australia.
| |
Collapse
|
9
|
Matsuo H, Kawakami K, Ohara H, Kaneko T, Mashimo T, Yamada T, Nabika T. Apolipoprotein E-depletion accelerates arterial fat deposition in the spontaneously hypertensive rat. Exp Anim 2023; 72:439-445. [PMID: 37081644 PMCID: PMC10658095 DOI: 10.1538/expanim.23-0012] [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: 01/23/2023] [Accepted: 04/09/2023] [Indexed: 04/22/2023] Open
Abstract
Hypertension and atherosclerosis are often found in one patient causing serious cardiovascular events. An animal model simultaneously expressing hypertension and atherosclerosis would be useful to study such a complex risk status. We therefore attempted to introduce a null mutation of the apolipoprotein E (ApoE) gene into the spontaneously hypertensive rat (SHR) using CRISPR/Cas9 to establish a genetic model for atherosclerosis with hypertension. We successfully established SHRApoE(-/-) having a 13-bps deletion in the 5'-end of ApoE gene. Deletion of ApoE protein was confirmed by Western blotting. Blood pressure of SHRApoE(-/-) was comparable to that of SHR. Feeding the rats with high fat high cholesterol diet (HFD) caused a significant increase in LDL cholesterol as well as in triglyceride in SHRApoE(-/-). After 8 weeks of HFD loading, superficial fat deposition was observed both in the aorta and the mesenteric arteries of SHRApoE(-/-) instead of mature atheromatous lesions found in humans. In addition, a null mutation of peroxiredoxin 2 (Prdx2) was introduced into SHRApoE(-/-) to examine the effect of increased oxidative stress on the development of atherosclerosis. SHR with the double depletion of ApoE and Prdx2 did not show mature atheroma either. Further, salt loading did not promote development of atheroma although it accelerated the development of fat deposition. These results indicated that when compared with ApoE-knockout mice, SHRApoE(-/-) was more resistant to atherosclerosis even though they have severe hypertension.
Collapse
Affiliation(s)
- Hiroyuki Matsuo
- Department of Experimental Animals, Interdisciplinary Center for Science Research, Head Office for Research and Academic Information, Shimane University, 89-1 Enya-cho, Izumo, Shimane 693-8501, Japan
- Department of Functional Pathology, Shimane University Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo, Shimane 693-8501, Japan
| | - Kohei Kawakami
- Department of Experimental Animals, Interdisciplinary Center for Science Research, Head Office for Research and Academic Information, Shimane University, 89-1 Enya-cho, Izumo, Shimane 693-8501, Japan
| | - Hiroki Ohara
- Department of Functional Pathology, Shimane University Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo, Shimane 693-8501, Japan
| | - Takehito Kaneko
- Graduate School of Science and Engineering, Iwate University, 4-3-5 Ueda, Morioka, Iwate 020-8551, Japan
| | - Tomoji Mashimo
- Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Yoshidakonoe-cho, Sakyou-ku, Kyoto 606-8501, Japan
| | - Takaya Yamada
- Department of Experimental Animals, Interdisciplinary Center for Science Research, Head Office for Research and Academic Information, Shimane University, 89-1 Enya-cho, Izumo, Shimane 693-8501, Japan
| | - Toru Nabika
- Department of Functional Pathology, Shimane University Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo, Shimane 693-8501, Japan
| |
Collapse
|
10
|
Tang Y, Liu L, Jie R, Tang Y, Zhao X, Xu M, Chen M. Negative pressure wound therapy promotes wound healing of diabetic foot ulcers by up-regulating PRDX2 in wound margin tissue. Sci Rep 2023; 13:16192. [PMID: 37758743 PMCID: PMC10533814 DOI: 10.1038/s41598-023-42634-9] [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: 07/02/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
To understand the changes in the peroxiredoxin-2 (PRDX2) expression level in the wound margin tissue (T-PRDX2) of patients with diabetic foot ulcer (DFU) before and after negative pressure wound therapy (NPWT). Additionally, the study aimed to explore the association between PRDX2 expression and the treatment outcome of DFUs to provide a new theoretical basis for revealing the mechanism of NPWT promoting the healing of DFUs. Fifty-six type 2 diabetes patients with foot ulcers undergoing NPWT (the DFU group) and 28 patients with chronic lower limb skin ulcers with normal glucose tolerance undergoing NPWT (the skin ulcer control [SUC] group) were included in the study. T-PRDX2 was detected using Western blotting, and the superoxide dismutase (SOD) activity and the malondialdehyde (MDA) and glutathione (GSH) levels were detected using a biochemical method. In addition, in vitro experiments were conducted to determine the effect of PRDX2 expression on normal human dermal fibroblast (NHDF) proliferation, migration, and apoptosis. Before NPWT, the DFU group exhibited a significantly lower T-PRDX2 expression level compared with the SUC group. After one week of NPWT, the T-PRDX2 expression level, SOD activity, and GSH content in the wound margin tissues of the DFU and SUC groups significantly increased compared with the before NPWT levels. Conversely, the inflammatory indicators (white blood cell, neutrophil percentage, C-reactive protein, and procalcitonin) and MDA content were significantly lower than the before NPWT levels. The expression changes of T-PRDX2 before and after NPWT in the DFU and SUC groups were positively correlated with the 4-week wound healing rate. In vitro experiments demonstrated that PRDX2 could alleviate the oxidative stress in NHDFs, thereby promoting their proliferation and migration, while reducing cell apoptosis. NPWT promotes DFU healing by increasing T-PRDX2, and changes in the T-PRDX2 might be associated with the therapeutic effect of NPWT.
Collapse
Affiliation(s)
- Ying Tang
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei City, Anhui Province, People's Republic of China
| | - Lei Liu
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei City, Anhui Province, People's Republic of China
| | - Ruyan Jie
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei City, Anhui Province, People's Republic of China
| | - Yizhong Tang
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, People's Republic of China
| | - Xiaotong Zhao
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei City, Anhui Province, People's Republic of China
| | - Murong Xu
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei City, Anhui Province, People's Republic of China.
| | - Mingwei Chen
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei City, Anhui Province, People's Republic of China.
| |
Collapse
|
11
|
Yan X, Xie Y, Liu H, Huang M, Yang Z, An D, Jiang G. Iron accumulation and lipid peroxidation: implication of ferroptosis in diabetic cardiomyopathy. Diabetol Metab Syndr 2023; 15:161. [PMID: 37468902 DOI: 10.1186/s13098-023-01135-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/09/2023] [Indexed: 07/21/2023] Open
Abstract
Diabetic cardiomyopathy (DC) is a serious heart disease caused by diabetes. It is unrelated to hypertension and coronary artery disease and can lead to heart insufficiency, heart failure and even death. Currently, the pathogenesis of DC is unclear, and clinical intervention is mainly symptomatic therapy and lacks effective intervention objectives. Iron overdose mediated cell death, also known as ferroptosis, is widely present in the physiological and pathological processes of diabetes and DC. Iron is a key trace element in the human body, regulating the metabolism of glucose and lipids, oxidative stress and inflammation, and other biological processes. Excessive iron accumulation can lead to the imbalance of the antioxidant system in DC and activate and aggravate pathological processes such as excessive autophagy and mitochondrial dysfunction, resulting in a chain reaction and accelerating myocardial and microvascular damage. In-depth understanding of the regulating mechanisms of iron metabolism and ferroptosis in cardiovascular vessels can help improve DC management. Therefore, in this review, we summarize the relationship between ferroptosis and the pathogenesis of DC, as well as potential intervention targets, and discuss and analyze the limitations and future development prospects of these targets.
Collapse
Affiliation(s)
- Xuehua Yan
- College of Traditional Chinese Medicine, Xinjiang Medical University, Xinjiang, China
- Xinjiang Key Laboratory of Famous Prescription and Science of Formulas, Xinjiang, China
| | - Yang Xie
- Affiliated Hospital of Traditional Chinese Medicine of Xinjiang Medical University, Xinjiang, China
| | - Hongbing Liu
- College of Traditional Chinese Medicine, Xinjiang Medical University, Xinjiang, China
| | - Meng Huang
- College of Traditional Chinese Medicine, Xinjiang Medical University, Xinjiang, China
| | - Zhen Yang
- College of Traditional Chinese Medicine, Xinjiang Medical University, Xinjiang, China
| | - Dongqing An
- College of Traditional Chinese Medicine, Xinjiang Medical University, Xinjiang, China.
- Xinjiang Key Laboratory of Famous Prescription and Science of Formulas, Xinjiang, China.
- Affiliated Hospital of Traditional Chinese Medicine of Xinjiang Medical University, Xinjiang, China.
| | - Guangjian Jiang
- College of Traditional Chinese Medicine, Xinjiang Medical University, Xinjiang, China.
| |
Collapse
|
12
|
Sherratt SCR, Libby P, Dawoud H, Bhatt DL, Malinski T, Mason RP. Eicosapentaenoic acid (EPA) reduces pulmonary endothelial dysfunction and inflammation due to changes in protein expression during exposure to particulate matter air pollution. Biomed Pharmacother 2023; 162:114629. [PMID: 37027984 DOI: 10.1016/j.biopha.2023.114629] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 04/09/2023] Open
Abstract
AIMS Inhalation of air pollution small particle matter (PM) is a leading cause of cardiovascular (CV) disease. Exposure to PMs causes endothelial cell (EC) dysfunction as evidenced by nitric oxide (NO) synthase uncoupling, vasoconstriction and inflammation. Eicosapentaenoic acid (EPA) has been shown to mitigate PM-induced adverse cardiac changes in patients receiving omega-3 fatty acid supplementation. We set out to determine the pro-inflammatory effects of multiple PMs (urban and fine) on pulmonary EC NO bioavailability and protein expression, and whether EPA restores EC function under these conditions. METHODS AND RESULTS We pretreated pulmonary ECs with EPA and then exposed them to urban or fine air pollution PMs. LC/MS-based proteomic analysis to assess relative expression levels. Expression of adhesion molecules was measured by immunochemistry. The ratio of NO to peroxynitrite (ONOO-) release, an indication of eNOS coupling, was measured using porphyrinic nanosensors following calcium stimulation. Urban/fine PMs also modulated 9/12 and 13/36 proteins, respectively, linked to platelet and neutrophil degranulation pathways and caused > 50% (p < 0.001) decrease in the stimulated NO/ONOO- release ratio. EPA treatment altered expression of proteins involved in these inflammatory pathways, including a decrease in peroxiredoxin-5 and an increase in superoxide dismutase-1. EPA also increased expression of heme oxygenase-1 (HMOX1), a cytoprotective protein, by 2.1-fold (p = 0.024). EPA reduced elevations in sICAM-1 levels by 22% (p < 0.01) and improved the NO/ONOO- release ratio by > 35% (p < 0.05). CONCLUSION These cellular changes may contribute to anti-inflammatory, cytoprotective and lipid changes associated with EPA treatment during air pollution exposure.
Collapse
Affiliation(s)
- Samuel C R Sherratt
- Department of Molecular, Cellular, and Biomedical Sciences, University of New Hampshire, Durham, NH, USA; Elucida Research LLC, Beverly, MA, USA
| | - Peter Libby
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Hazem Dawoud
- Nanomedical Research Laboratory, Ohio University, Athens, OH, USA
| | - Deepak L Bhatt
- Mount Sinai Heart, Icahn School of Medicine at Mount Sinai Health System, New York, NY, USA
| | - Tadeusz Malinski
- Nanomedical Research Laboratory, Ohio University, Athens, OH, USA.
| | - R Preston Mason
- Elucida Research LLC, Beverly, MA, USA; Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
13
|
Guo J, Tang H, Huang P, Ye X, Tang C, Shu Z, Guo J, Kang X, Shi Y, Zhou B, Liang T, Tang K. Integrative single-cell RNA and ATAC sequencing reveals that the FOXO1-PRDX2-TNF axis regulates tendinopathy. Front Immunol 2023; 14:1092778. [PMID: 37223090 PMCID: PMC10200929 DOI: 10.3389/fimmu.2023.1092778] [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: 11/08/2022] [Accepted: 04/25/2023] [Indexed: 05/25/2023] Open
Abstract
Introduction Tendinopathy, the most common form of chronic tendon disorder, leads to persistent tendon pain and loss of function. Profiling the heterogeneous cellular composition in the tendon microenvironment helps to elucidate rational molecular mechanisms of tendinopathy. Methods and results In this study, through a multi-modal analysis, a single-cell RNA- and ATAC-seq integrated tendinopathy landscape was generated for the first time. We found that a specific cell subpopulation with low PRDX2 expression exhibited a higher level of inflammation, lower proliferation and migration ability, which not only promoted tendon injury but also led to microenvironment deterioration. Mechanistically, a motif enrichment analysis of chromatin accessibility showed that FOXO1 was an upstream regulator of PRDX2 transcription, and we confirmed that functional blockade of FOXO1 activity induced PRDX2 silencing. The TNF signaling pathway was significantly activated in the PRDX2-low group, and TNF inhibition effectively restored diseased cell degradation. Discussion We revealed an essential role of diseased cells in tendinopathy and proposed the FOXO1-PRDX2-TNF axis is a potential regulatory mechanism for the treatment of tendinopathy.
Collapse
Affiliation(s)
- Junfeng Guo
- Department of Orthopedics/Sports Medicine Center, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Hong Tang
- Department of Orthopedics/Sports Medicine Center, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Pan Huang
- Department of Orthopedics/Sports Medicine Center, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Xiao Ye
- Department of Orthopedics/Sports Medicine Center, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Chuyue Tang
- Department of Orthopedics/Sports Medicine Center, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Zhao Shu
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Junfeng Guo
- Department of Stomatology, The 970th Hospital of the Joint Logistics Support Force, Yantai, China
| | - Xia Kang
- Department of Orthopedics/Sports Medicine Center, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Youxing Shi
- Department of Orthopedics/Sports Medicine Center, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Binghua Zhou
- Department of Orthopedics/Sports Medicine Center, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Taotao Liang
- Department of Orthopedics/Sports Medicine Center, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Kanglai Tang
- Department of Orthopedics/Sports Medicine Center, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
14
|
Jeong SJ, Oh GT. Unbalanced Redox With Autophagy in Cardiovascular Disease. J Lipid Atheroscler 2023; 12:132-151. [PMID: 37265853 PMCID: PMC10232220 DOI: 10.12997/jla.2023.12.2.132] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 03/27/2023] [Accepted: 04/13/2023] [Indexed: 06/03/2023] Open
Abstract
Precise redox balance is essential for the optimum health and physiological function of the human body. Furthermore, an unbalanced redox state is widely believed to be part of numerous diseases, ultimately resulting in death. In this review, we discuss the relationship between redox balance and cardiovascular disease (CVD). In various animal models, excessive oxidative stress has been associated with increased atherosclerotic plaque formation, which is linked to the inflammation status of several cell types. However, various antioxidants can defend against reactive oxidative stress, which is associated with an increased risk of CVD and mortality. The different cardiovascular effects of these antioxidants are presumably due to alterations in the multiple pathways that have been mechanistically linked to accelerated atherosclerotic plaque formation, macrophage activation, and endothelial dysfunction in animal models of CVD, as well as in in vitro cell culture systems. Autophagy is a regulated cell survival mechanism that removes dysfunctional or damaged cellular organelles and recycles the nutrients for the generation of energy. Furthermore, in response to atherogenic stress, such as the generation of reactive oxygen species, oxidized lipids, and inflammatory signaling between cells, autophagy protects against plaque formation. In this review, we characterize the broad spectrum of oxidative stress that influences CVD, summarize the role of autophagy in the content of redox balance-associated pathways in atherosclerosis, and discuss potential therapeutic approaches to target CVD by stimulating autophagy.
Collapse
Affiliation(s)
- Se-Jin Jeong
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Goo Taeg Oh
- Immune and Vascular Cell Network Research Center, National Creative Initiatives, Department of Life Sciences, Ewha Womans University, Seoul, Korea
| |
Collapse
|
15
|
Liu Z. Cardiac Microvascular Dysfunction and Cardiomyopathy in Diabetes: Is Ferroptosis a Therapeutic Target? Diabetes 2023; 72:313-315. [PMID: 36806606 PMCID: PMC10090265 DOI: 10.2337/dbi22-0036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/13/2022] [Accepted: 12/20/2023] [Indexed: 02/23/2023]
Affiliation(s)
- Zhenqi Liu
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville, VA
| |
Collapse
|
16
|
Zou J, Xu C, Zhao ZW, Yin SH, Wang G. Asprosin inhibits macrophage lipid accumulation and reduces atherosclerotic burden by up-regulating ABCA1 and ABCG1 expression via the p38/Elk-1 pathway. Lab Invest 2022; 20:337. [PMID: 35902881 PMCID: PMC9331044 DOI: 10.1186/s12967-022-03542-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 07/17/2022] [Indexed: 12/27/2022]
Abstract
Background Asprosin, a newly discovered adipokine, is a C-terminal cleavage product of profibrillin. Asprosin has been reported to participate in lipid metabolism and cardiovascular disease, but its role in atherogenesis remains elusive. Methods Asprosin was overexpressed in THP-1 macrophage-derived foam cells and apoE−/− mice using the lentiviral vector. The expression of relevant molecules was determined by qRT-PCR and/or western blot. The intracellular lipid accumulation was evaluated by high-performance liquid chromatography and Oil red O staining. HE and Oil red O staining was employed to assess plaque burden in vivo. Reverse cholesterol transport (RCT) efficiency was measured using [3H]-labeled cholesterol. Results Exposure of THP-1 macrophages to oxidized low-density lipoprotein down-regulated asprosin expression. Lentivirus-mediated overexpression of asprosin promoted cholesterol efflux and inhibited lipid accumulation in THP-1 macrophage-derived foam cells. Mechanistic analysis revealed that asprosin overexpression activated p38 and stimulated the phosphorylation of ETS-like transcription factor (Elk-1) at Ser383, leading to Elk-1 nuclear translocation and the transcriptional activation of ATP binding cassette transporters A1 (ABCA1) and ABCG1. Injection of lentiviral vector expressing asprosin diminished atherosclerotic lesion area, increased plaque stability, improved plasma lipid profiles and facilitated RCT in apoE−/− mice. Asprosin overexpression also increased the phosphorylation of p38 and Elk-1 as well as up-regulated the expression of ABCA1 and ABCG1 in the aortas. Conclusion Asprosin inhibits lipid accumulation in macrophages and decreases atherosclerotic burden in apoE−/− mice by up-regulating ABCA1 and ABCG1 expression via activation of the p38/Elk-1 signaling pathway. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03542-0.
Collapse
Affiliation(s)
- Jin Zou
- The First Affiliated Hospital, Department of Cardiology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Can Xu
- The First Affiliated Hospital, Department of Cardiology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Zhen-Wang Zhao
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Shan-Hui Yin
- The First Affiliated Hospital, Department of Neonatology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Gang Wang
- The First Affiliated Hospital, Department of Cardiology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China.
| |
Collapse
|
17
|
Role of Oxidative Stress in the Pathogenesis of Atherothrombotic Diseases. Antioxidants (Basel) 2022; 11:antiox11071408. [PMID: 35883899 PMCID: PMC9312358 DOI: 10.3390/antiox11071408] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/12/2022] [Accepted: 07/19/2022] [Indexed: 12/04/2022] Open
Abstract
Oxidative stress is generated by the imbalance between reactive oxygen species (ROS) formation and antioxidant scavenger system’s activity. Increased ROS, such as superoxide anion, hydrogen peroxide, hydroxyl radical and peroxynitrite, likely contribute to the development and complications of atherosclerotic cardiovascular diseases (ASCVD). In genetically modified mouse models of atherosclerosis, the overexpression of ROS-generating enzymes and uncontrolled ROS formation appear to be associated with accelerated atherosclerosis. Conversely, the overexpression of ROS scavenger systems reduces or stabilizes atherosclerotic lesions, depending on the genetic background of the mouse model. In humans, higher levels of circulating biomarkers derived from the oxidation of lipids (8-epi-prostaglandin F2α, and malondialdehyde), as well as proteins (oxidized low-density lipoprotein, nitrotyrosine, protein carbonyls, advanced glycation end-products), are increased in conditions of high cardiovascular risk or overt ASCVD, and some oxidation biomarkers have been reported as independent predictors of ASCVD in large observational cohorts. In animal models, antioxidant supplementation with melatonin, resveratrol, Vitamin E, stevioside, acacetin and n-polyunsaturated fatty acids reduced ROS and attenuated atherosclerotic lesions. However, in humans, evidence from large, placebo-controlled, randomized trials or prospective studies failed to show any athero-protective effect of antioxidant supplementation with different compounds in different CV settings. However, the chronic consumption of diets known to be rich in antioxidant compounds (e.g., Mediterranean and high-fish diet), has shown to reduce ASCVD over decades. Future studies are needed to fill the gap between the data and targets derived from studies in animals and their pathogenetic and therapeutic significance in human ASCVD.
Collapse
|
18
|
Wei TT, Chandy M, Nishiga M, Zhang A, Kumar KK, Thomas D, Manhas A, Rhee S, Justesen JM, Chen IY, Wo HT, Khanamiri S, Yang JY, Seidl FJ, Burns NZ, Liu C, Sayed N, Shie JJ, Yeh CF, Yang KC, Lau E, Lynch KL, Rivas M, Kobilka BK, Wu JC. Cannabinoid receptor 1 antagonist genistein attenuates marijuana-induced vascular inflammation. Cell 2022; 185:1676-1693.e23. [PMID: 35489334 PMCID: PMC9400797 DOI: 10.1016/j.cell.2022.04.005] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 02/01/2022] [Accepted: 04/04/2022] [Indexed: 12/16/2022]
Abstract
Epidemiological studies reveal that marijuana increases the risk of cardiovascular disease (CVD); however, little is known about the mechanism. Δ9-tetrahydrocannabinol (Δ9-THC), the psychoactive component of marijuana, binds to cannabinoid receptor 1 (CB1/CNR1) in the vasculature and is implicated in CVD. A UK Biobank analysis found that cannabis was an risk factor for CVD. We found that marijuana smoking activated inflammatory cytokines implicated in CVD. In silico virtual screening identified genistein, a soybean isoflavone, as a putative CB1 antagonist. Human-induced pluripotent stem cell-derived endothelial cells were used to model Δ9-THC-induced inflammation and oxidative stress via NF-κB signaling. Knockdown of the CB1 receptor with siRNA, CRISPR interference, and genistein attenuated the effects of Δ9-THC. In mice, genistein blocked Δ9-THC-induced endothelial dysfunction in wire myograph, reduced atherosclerotic plaque, and had minimal penetration of the central nervous system. Genistein is a CB1 antagonist that attenuates Δ9-THC-induced atherosclerosis.
Collapse
Affiliation(s)
- Tzu-Tang Wei
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA; Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan; Chemical Biology and Molecular Biophysics, Taiwan International Graduate Program in Chemical Biology and Molecular Biophysics (TIGP-CBMB), Academia Sinica, Taipei, Taiwan
| | - Mark Chandy
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA; Greenstone Biosciences, Palo Alto, CA 94304, USA
| | - Masataka Nishiga
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA
| | - Angela Zhang
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA
| | - Kaavya Krishna Kumar
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Dilip Thomas
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA
| | - Amit Manhas
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA
| | - Siyeon Rhee
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA; Greenstone Biosciences, Palo Alto, CA 94304, USA
| | - Johanne Marie Justesen
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA 94305, USA; Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Ian Y Chen
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA
| | - Hung-Ta Wo
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA; Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
| | - Saereh Khanamiri
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA
| | - Johnson Y Yang
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA
| | | | - Noah Z Burns
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Chun Liu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA
| | - Nazish Sayed
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA
| | - Jiun-Jie Shie
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
| | - Chih-Fan Yeh
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan; Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Kai-Chien Yang
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan; Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Edward Lau
- Department of Medicine, Division of Cardiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Kara L Lynch
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Manuel Rivas
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Brian K Kobilka
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA; Greenstone Biosciences, Palo Alto, CA 94304, USA.
| |
Collapse
|
19
|
Kim S, Lee W, Jo H, Sonn SK, Jeong SJ, Seo S, Suh J, Jin J, Kweon HY, Kim TK, Moon SH, Jeon S, Kim JW, Kim YR, Lee EW, Shin HK, Park SH, Oh GT. The antioxidant enzyme Peroxiredoxin-1 controls stroke-associated microglia against acute ischemic stroke. Redox Biol 2022; 54:102347. [PMID: 35688114 PMCID: PMC9184746 DOI: 10.1016/j.redox.2022.102347] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/17/2022] [Accepted: 05/17/2022] [Indexed: 01/04/2023] Open
Affiliation(s)
- Sinai Kim
- Heart-Immune-Brain Network Research Center, Department of Life Science, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Wonhyo Lee
- Department of Biological Sciences, Ulsan National Institute of Science & Technology (UNIST), Ulsan, South Korea
| | - Huiju Jo
- Heart-Immune-Brain Network Research Center, Department of Life Science, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Seong-Keun Sonn
- Heart-Immune-Brain Network Research Center, Department of Life Science, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Se-Jin Jeong
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Seungwoon Seo
- Heart-Immune-Brain Network Research Center, Department of Life Science, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Joowon Suh
- Heart-Immune-Brain Network Research Center, Department of Life Science, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Jing Jin
- Heart-Immune-Brain Network Research Center, Department of Life Science, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Hyae Yon Kweon
- Heart-Immune-Brain Network Research Center, Department of Life Science, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Tae Kyeong Kim
- Heart-Immune-Brain Network Research Center, Department of Life Science, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Shin Hye Moon
- Heart-Immune-Brain Network Research Center, Department of Life Science, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Sejin Jeon
- Department of Biological Sciences and Biotechnology Major in Bio-Vaccine Engineering Andong National University, Andong, South Korea
| | - Jong Woo Kim
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, South Korea; Department of Functional Genomics, University of Science and Technology (UST), Daejeon, South Korea
| | - Yu Ri Kim
- KM Convergence Research Division, Korea Institute of Oriental Medicine 1672, Yuseong-daero, Yuseong-gu, Daejeon, 34054, Republic of Korea
| | - Eun-Woo Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, South Korea; Department of Functional Genomics, University of Science and Technology (UST), Daejeon, South Korea
| | - Hwa Kyoung Shin
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam, 50612, Republic of Korea
| | - Sung Ho Park
- Department of Biological Sciences, Ulsan National Institute of Science & Technology (UNIST), Ulsan, South Korea.
| | - Goo Taeg Oh
- Heart-Immune-Brain Network Research Center, Department of Life Science, Ewha Womans University, Seoul, 03760, Republic of Korea.
| |
Collapse
|
20
|
Jang YS, Lee YS, Kim DH, Oh GT, Jeon WK, Han JS. Peroxiredoxin 2 deletion impairs hippocampal-dependent memory via exacerbating transient ischemia-induced oxidative damage. Brain Res Bull 2022; 184:99-105. [PMID: 35452748 DOI: 10.1016/j.brainresbull.2022.04.004] [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: 10/13/2021] [Revised: 04/13/2022] [Accepted: 04/16/2022] [Indexed: 11/15/2022]
Abstract
Peroxiredoxin 2 (Prx2) regulates oxidative stress response in neuronal injury. The present study examined the effects of Prx2 deletion on transient global ischemia-induced hippocampal-dependent memory impairment. First, 20-min bilateral common carotid artery occlusion (BCCAO)-reperfusion and sham-operated control procedures were conducted in 6- or 7-month-old Prx2 knockout and wild-type mice. The cognitive status of these mice was assessed using the Morris water maze task with a hidden platform and a novel object recognition task 7 days after the 20-min BCCAO. Next, to evaluate neuronal degeneration and oxidative stress in the CA1 subregion of the hippocampus critical for learning and memory, we measured immunoreactive Fluro-jade C (FJC)-positive signals and 4-hydroxy-2-trans-nonenal (4-HNE) levels, respectively. The 20-min BCCAO induced cognitive impairments and increased the intensity of FJC-positive signals and 4-HNE levels of CA1 in Prx2 knockout mice but not in wild-type mice. These results suggest that Prx2 deficiency reduces resilience to transient global ischemia.
Collapse
Affiliation(s)
- Yoon-Sun Jang
- Department of Biological Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Yo-Seob Lee
- Department of Biological Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Dong-Hee Kim
- Department of Biological Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Goo Taeg Oh
- Division of Molecular Life Sciences, Ewha W. University, Seoul 03760, Republic of Korea
| | - Won Kyung Jeon
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea; Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea.
| | - Jung-Soo Han
- Department of Biological Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| |
Collapse
|
21
|
Cysteine-Rich LIM-Only Protein 4 (CRP4) Promotes Atherogenesis in the ApoE -/- Mouse Model. Cells 2022; 11:cells11081364. [PMID: 35456043 PMCID: PMC9032522 DOI: 10.3390/cells11081364] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/31/2022] [Accepted: 04/09/2022] [Indexed: 01/27/2023] Open
Abstract
Vascular smooth muscle cells (VSMCs) can switch from their contractile state to a synthetic phenotype resulting in high migratory and proliferative capacity and driving atherosclerotic lesion formation. The cysteine-rich LIM-only protein 4 (CRP4) reportedly modulates VSM-like transcriptional signatures, which are perturbed in VSMCs undergoing phenotypic switching. Thus, we hypothesized that CRP4 contributes to adverse VSMC behaviours and thereby to atherogenesis in vivo. The atherogenic properties of CRP4 were investigated in plaque-prone apolipoprotein E (ApoE) and CRP4 double-knockout (dKO) as well as ApoE-deficient CRP4 wildtype mice. dKO mice exhibited lower plaque numbers and lesion areas as well as a reduced content of α-smooth muscle actin positive cells in the lesion area, while lesion-associated cell proliferation was elevated in vessels lacking CRP4. Reduced plaque volumes in dKO correlated with significantly less intra-plaque oxidized low-density lipoprotein (oxLDL), presumably due to upregulation of the antioxidant factor peroxiredoxin-4 (PRDX4). This study identifies CRP4 as a novel pro-atherogenic factor that facilitates plaque oxLDL deposition and identifies the invasion of atherosclerotic lesions by VSMCs as important determinants of plaque vulnerability. Thus, targeting of VSMC CRP4 should be considered in plaque-stabilizing pharmacological strategies.
Collapse
|
22
|
Ginckels P, Holvoet P. Oxidative Stress and Inflammation in Cardiovascular Diseases and Cancer: Role of Non-coding RNAs. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2022; 95:129-152. [PMID: 35370493 PMCID: PMC8961704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
High oxidative stress, Th1/Th17 immune response, M1 macrophage inflammation, and cell death are associated with cardiovascular diseases. Controlled oxidative stress, Th2/Treg anti-tumor immune response, M2 macrophage inflammation, and survival are associated with cancer. MiR-21 protects against cardiovascular diseases but may induce tumor growth by retaining the anti-inflammatory M2 macrophage and Treg phenotypes and inhibiting apoptosis. Down-regulation of let-7, miR-1, miR-9, miR-16, miR-20a, miR-22a, miR-23a, miR-24a, miR-26a, miR-29, miR-30a, miR-34a, miR-124, miR-128, miR-130a, miR-133, miR-140, miR-143-145, miR-150, miR-153, miR-181a, miR-378, and miR-383 may aid cancer cells to escape from stresses. Upregulation of miR-146 and miR-223 may reduce anti-tumor immune response together with miR-21 that also protects against apoptosis. MiR-155 and silencing of let-7e, miR-125, and miR-126 increase anti-tumor immune response. MiR expression depends on oxidative stress, cytokines, MYC, and TGF-β, and expression of silencing lncRNAs and circ-RNAs. However, one lncRNA or circ-RNA may have opposite effects by targeting several miRs. For example, PVT1 induces apoptosis by targeting miR-16a and miR-30a but inhibits apoptosis by silencing miR-17. In addition, levels of a non-coding RNA in a cell type depend not only on expression in that cell type but also on an exchange of microvesicles between cell types and tumors. Although we got more insight into the function of a growing number of individual non-coding RNAs, overall, we do not know enough how several of them interact in functional networks and how their expression changes at different stages of disease progression.
Collapse
Affiliation(s)
- Pieterjan Ginckels
- Department of Architecture, Brussels and Gent, KU Leuven, Leuven, Belgium
| | - Paul Holvoet
- Experimental Cardiology, KU Leuven, Leuven, Belgium,To whom all correspondence should be addressed: Paul Holvoet, Experimental
Cardiology, KU Leuven, Belgium; ; ORCID iD:
https://orcid.org/0000-0001-9201-0772
| |
Collapse
|
23
|
Bowdridge EC, DeVallance E, Garner KL, Griffith JA, Schafner K, Seaman M, Engels KJ, Wix K, Batchelor TP, Goldsmith WT, Hussain S, Nurkiewicz TR. Nano-titanium dioxide inhalation exposure during gestation drives redox dysregulation and vascular dysfunction across generations. Part Fibre Toxicol 2022; 19:18. [PMID: 35260159 PMCID: PMC8905816 DOI: 10.1186/s12989-022-00457-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 02/24/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Pregnancy is associated with many rapid biological adaptations that support healthy development of the growing fetus. One of which is critical to fetal health and development is the coordination between maternal liver derived substrates and vascular delivery. This crucial adaptation can be potentially derailed by inhalation of toxicants. Engineered nanomaterials (ENM) are commonly used in household and industrial products as well as in medicinal applications. As such, the potential risk of exposure remains a concern, especially during pregnancy. We have previously reported that ENM inhalation leads to upregulation in the production of oxidative species. Therefore, we aimed to determine if F0 dam maternal nano-TiO2 inhalation exposure (exclusively) resulted in altered H2O2 production capacity and changes in downstream redox pathways in the F0 dams and subsequent F1 pups. Additionally, we investigated whether this persisted into adulthood within the F1 generation and how this impacted F1 gestational outcomes and F2 fetal health and development. We hypothesized that maternal nano-TiO2 inhalation exposure during gestation in the F0 dams would result in upregulated H2O2 production in the F0 dams as well as her F1 offspring. Additionally, this toxicological insult would result in gestational vascular dysfunction in the F1 dams yielding smaller F2 generation pups. RESULTS Our results indicate upregulation of hepatic H2O2 production capacity in F0 dams, F1 offspring at 8 weeks and F1 females at gestational day 20. H2O2 production capacity was accompanied by a twofold increase in phosphorylation of the redox sensitive transcription factor NF-κB. In cell culture, naïve hepatocytes exposed to F1-nano-TiO2 plasma increased H2O2 production. Overnight exposure of these hepatocytes to F1 plasma increased H2O2 production capacity in a partially NF-κB dependent manner. Pregnant F1- nano-TiO2 females exhibited estrogen disruption (12.12 ± 3.1 pg/ml vs. 29.81 ± 8.8 pg/ml sham-control) and vascular dysfunction similar to their directly exposed mothers. F1-nano-TiO2 uterine artery H2O2 production capacity was also elevated twofold. Dysfunctional gestational outcomes in the F1-nano-TiO2 dams resulted in smaller F1 (10.22 ± 0.6 pups vs. sham-controls 12.71 ± 0.96 pups) and F2 pups (4.93 ± 0.47 g vs. 5.78 ± 0.09 g sham-control pups), and fewer F1 male pups (4.38 ± 0.3 pups vs. 6.83 ± 0.84 sham-control pups). CONCLUSION In conclusion, this manuscript provides critical evidence of redox dysregulation across generations following maternal ENM inhalation. Furthermore, dysfunctional gestational outcomes are observed in the F1-nano-TiO2 generation and impact the development of F2 offspring. In total, this data provides strong initial evidence that maternal ENM exposure has robust biological impacts that persists in at least two generations.
Collapse
Affiliation(s)
- Elizabeth C. Bowdridge
- grid.268154.c0000 0001 2156 6140Department of Physiology and Pharmacology, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, West Virginia University, Morgantown, WV 26505-9229 USA ,grid.268154.c0000 0001 2156 6140Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV USA
| | - Evan DeVallance
- grid.268154.c0000 0001 2156 6140Department of Physiology and Pharmacology, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, West Virginia University, Morgantown, WV 26505-9229 USA ,grid.268154.c0000 0001 2156 6140Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV USA
| | - Krista L. Garner
- grid.268154.c0000 0001 2156 6140Department of Physiology and Pharmacology, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, West Virginia University, Morgantown, WV 26505-9229 USA ,grid.268154.c0000 0001 2156 6140Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV USA
| | - Julie A. Griffith
- grid.268154.c0000 0001 2156 6140Department of Physiology and Pharmacology, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, West Virginia University, Morgantown, WV 26505-9229 USA ,grid.268154.c0000 0001 2156 6140Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV USA
| | - Kallie Schafner
- grid.268154.c0000 0001 2156 6140Department of Physiology and Pharmacology, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, West Virginia University, Morgantown, WV 26505-9229 USA ,grid.268154.c0000 0001 2156 6140Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV USA
| | - Madison Seaman
- grid.268154.c0000 0001 2156 6140Department of Physiology and Pharmacology, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, West Virginia University, Morgantown, WV 26505-9229 USA
| | - Kevin J. Engels
- grid.268154.c0000 0001 2156 6140Department of Physiology and Pharmacology, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, West Virginia University, Morgantown, WV 26505-9229 USA
| | - Kimberley Wix
- grid.268154.c0000 0001 2156 6140Department of Physiology and Pharmacology, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, West Virginia University, Morgantown, WV 26505-9229 USA
| | - Thomas P. Batchelor
- grid.268154.c0000 0001 2156 6140Department of Physiology and Pharmacology, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, West Virginia University, Morgantown, WV 26505-9229 USA ,grid.268154.c0000 0001 2156 6140Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV USA
| | - William T. Goldsmith
- grid.268154.c0000 0001 2156 6140Department of Physiology and Pharmacology, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, West Virginia University, Morgantown, WV 26505-9229 USA ,grid.268154.c0000 0001 2156 6140Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV USA
| | - Salik Hussain
- grid.268154.c0000 0001 2156 6140Department of Physiology and Pharmacology, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, West Virginia University, Morgantown, WV 26505-9229 USA ,grid.268154.c0000 0001 2156 6140Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV USA
| | - Timothy R. Nurkiewicz
- grid.268154.c0000 0001 2156 6140Department of Physiology and Pharmacology, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, West Virginia University, Morgantown, WV 26505-9229 USA ,grid.268154.c0000 0001 2156 6140Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV USA
| |
Collapse
|
24
|
Sharapov MG, Gudkov SV, Lankin VZ, Novoselov VI. Role of Glutathione Peroxidases and Peroxiredoxins in Free Radical-Induced Pathologies. BIOCHEMISTRY. BIOKHIMIIA 2021; 86:1418-1433. [PMID: 34906041 DOI: 10.1134/s0006297921110067] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In this review, we discuss the pathogenesis of some socially significant diseases associated with the development of oxidative stress, such as atherosclerosis, diabetes, and radiation sickness, as well as the possibilities of the therapeutic application of low-molecular-weight natural and synthetic antioxidants for the correction of free radical-induced pathologies. The main focus of this review is the role of two phylogenetically close families of hydroperoxide-reducing antioxidant enzymes peroxiredoxins and glutathione peroxidases - in counteracting oxidative stress. We also present examples of the application of exogenous recombinant antioxidant enzymes as therapeutic agents in the treatment of pathologies associated with free-radical processes and discuss the prospects of the therapeutic use of exogenous antioxidant enzymes, as well as the ways to improve their therapeutic properties.
Collapse
Affiliation(s)
- Mars G Sharapov
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia.
| | - Sergey V Gudkov
- Prokhorov Institute of General Physics, Russian Academy of Sciences, Moscow, 119991, Russia.,Institute of Biology and Biomedicine, Lobachevsky State University of Nizhni Novgorod, Nizhny Novgorod, 603022, Russia.,All-Russian Research Institute of Phytopathology, Bolshiye Vyazemy, 143050, Russia
| | - Vadim Z Lankin
- National Medical Research Center of Cardiology, Ministry of Health of Russia, Moscow, 121552, Russia
| | - Vladimir I Novoselov
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| |
Collapse
|
25
|
Kumar S, Sur S, Perez J, Demos C, Kang DW, Kim CW, Hu S, Xu K, Yang J, Jo H. Atorvastatin and blood flow regulate expression of distinctive sets of genes in mouse carotid artery endothelium. CURRENT TOPICS IN MEMBRANES 2021; 87:97-130. [PMID: 34696890 DOI: 10.1016/bs.ctm.2021.08.004] [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] [Indexed: 03/29/2023]
Abstract
Hypercholesterolemia is a well-known pro-atherogenic risk factor and statin is the most effective anti-atherogenic drug that lowers blood cholesterol levels. However, despite systemic hypercholesterolemia, atherosclerosis preferentially occurs in arterial regions exposed to disturbed blood flow (d-flow), while the stable flow (s-flow) regions are spared. Given their predominant effects on endothelial function and atherosclerosis, we tested whether (1) statin and flow regulate the same or independent sets of genes and (2) statin can rescue d-flow-regulated genes in mouse artery endothelial cells in vivo. To test the hypotheses, C57BL/6 J mice (8-week-old male, n=5 per group) were pre-treated with atorvastatin (10mg/kg/day, Orally) or vehicle for 5 days. Thereafter, partial carotid ligation (PCL) surgery to induce d-flow in the left carotid artery (LCA) was performed, and statin or vehicle treatment was continued. The contralateral right carotid artery (RCA) remained exposed to s-flow to be used as the control. Two days or 2 weeks post-PCL surgery, endothelial-enriched RNAs from the LCAs and RCAs were collected and subjected to microarray gene expression analysis. Statin treatment in the s-flow condition (RCA+statin versus RCA+vehicle) altered the expression of 667 genes at 2-day and 187 genes at 2-week timepoint, respectively (P<0.05, fold change (FC)≥±1.5). Interestingly, statin treatment in the d-flow condition (LCA+statin versus LCA+vehicle) affected a limited number of genes: 113 and 75 differentially expressed genes at 2-day and 2-week timepoint, respectively (P<0.05, FC≥±1.5). In contrast, d-flow in the vehicle groups (LCA+vehicle versus RCA+vehicle) differentially regulated 4061 genes at 2-day and 3169 genes at 2-week timepoint, respectively (P<0.05, FC≥±1.5). Moreover, statin treatment did not reduce the number of flow-sensitive genes (LCA+statin versus RCA+statin) compared to the vehicle groups: 1825 genes at 2-day and 3788 genes at 2-week, respectively, were differentially regulated (P<0.05, FC≥±1.5). These results revealed that both statin and d-flow regulate expression of hundreds or thousands of arterial endothelial genes, respectively, in vivo. Further, statin and d-flow regulate independent sets of endothelial genes. Importantly, statin treatment did not reverse d-flow-regulated genes except for a small number of genes. These results suggest that both statin and flow play important independent roles in atherosclerosis development and highlight the need to consider their therapeutic implications for both.
Collapse
Affiliation(s)
- Sandeep Kumar
- Wallace H. Coulter Department of Biomedical Engineering at Emory University and Georgia Institute of Technology, Atlanta, GA, United States
| | - Sanjoli Sur
- Wallace H. Coulter Department of Biomedical Engineering at Emory University and Georgia Institute of Technology, Atlanta, GA, United States
| | - Julian Perez
- Wallace H. Coulter Department of Biomedical Engineering at Emory University and Georgia Institute of Technology, Atlanta, GA, United States
| | - Catherine Demos
- Wallace H. Coulter Department of Biomedical Engineering at Emory University and Georgia Institute of Technology, Atlanta, GA, United States
| | - Dong-Won Kang
- Wallace H. Coulter Department of Biomedical Engineering at Emory University and Georgia Institute of Technology, Atlanta, GA, United States
| | - Chan Woo Kim
- Wallace H. Coulter Department of Biomedical Engineering at Emory University and Georgia Institute of Technology, Atlanta, GA, United States
| | - Sarah Hu
- Thrombosis Research Unit, Bristol Myers Squibb, Lawrence, NJ, United States
| | - Ke Xu
- Thrombosis Research Unit, Bristol Myers Squibb, Lawrence, NJ, United States
| | - Jing Yang
- Thrombosis Research Unit, Bristol Myers Squibb, Lawrence, NJ, United States
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering at Emory University and Georgia Institute of Technology, Atlanta, GA, United States; Division of Cardiology, Emory University, Atlanta, GA, United States.
| |
Collapse
|
26
|
Jeong SJ, Park JG, Oh GT. Peroxiredoxins as Potential Targets for Cardiovascular Disease. Antioxidants (Basel) 2021; 10:antiox10081244. [PMID: 34439492 PMCID: PMC8389283 DOI: 10.3390/antiox10081244] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 07/30/2021] [Indexed: 01/10/2023] Open
Abstract
Increased oxidative stress (OS) is considered a common etiology in the pathogenesis of cardiovascular disease (CVD). Therefore, the precise regulation of reactive oxygen species (ROS) in cardiovascular cells is essential to maintain normal physiological functions. Numerous regulators of cellular homeostasis are reportedly influenced by ROS. Hydrogen peroxide (H2O2), as an endogenous ROS in aerobic cells, is a toxic substance that can induce OS. However, many studies conducted over the past two decades have provided substantial evidence that H2O2 acts as a diffusible intracellular signaling messenger. Antioxidant enzymes, including superoxide dismutases, catalase, glutathione peroxidases, and peroxiredoxins (Prdxs), maintain the balance of ROS levels against augmentation of ROS production during the pathogenesis of CVD. Especially, Prdxs are regulatory sensors of transduced intracellular signals. The intracellular abundance of Prdxs that specifically react with H2O2 act as regulatory proteins. In this review, we focus on the role of Prdxs in the regulation of ROS-induced pathological changes in the development of CVD.
Collapse
Affiliation(s)
- Se-Jin Jeong
- Center for Cardiovascular Research, Washington University School of Medicine, St. Louis, MO 63110, USA;
| | - Jong-Gil Park
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
- Correspondence: (J.-G.P.); (G.T.O.); Tel.: +82-42-860-4122 (J.-G.P.); +82-2-3277-4128 (G.T.O.); Fax: +82-42-860-4149 (J.-G.P.); +82-2-3277-3760 (G.T.O.)
| | - Goo Taeg Oh
- Department of Life Sciences, Heart-Immune-Brain Network Research Center, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Korea
- Correspondence: (J.-G.P.); (G.T.O.); Tel.: +82-42-860-4122 (J.-G.P.); +82-2-3277-4128 (G.T.O.); Fax: +82-42-860-4149 (J.-G.P.); +82-2-3277-3760 (G.T.O.)
| |
Collapse
|
27
|
Li J, Wang C, Wang W, Liu L, Zhang Q, Zhang J, Wang B, Wang S, Hou L, Gao C, Yu X, Sun L. PRDX2 Protects Against Atherosclerosis by Regulating the Phenotype and Function of the Vascular Smooth Muscle Cell. Front Cardiovasc Med 2021; 8:624796. [PMID: 33791345 PMCID: PMC8006347 DOI: 10.3389/fcvm.2021.624796] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 02/09/2021] [Indexed: 01/05/2023] Open
Abstract
Peroxiredoxin 2 (PRDX2), an inhibitor of reactive oxygen species (ROS), is potentially involved in the progression of atherosclerosis (AS). The aim of this study was to explore the role and mechanism of PRDX2 in AS. The expression of PRDX2 was evaluated in 14 human carotid artery tissues with or without AS. The results showed that the positive reaction of PRDX2 was observed in the carotid artery vascular smooth muscle cells (CAVSMCs). To assess the mechanism by which PRDX2 may function in AS, the CAVSMCs were transfected with pEX4-PRDX2 and si-PRDX2. The catalase, hydrogen peroxide (H2O2) scavenger, was used to further confirm that PRDX2-induced inhibitory effects might be mediated through reducing ROS levels. Phenotype alteration and functional testing included transcription testing, immunostaining, and expression studies. The drug of MAPK signaling pathway inhibitors SB203580, SP600125, and PD98059 was used to evaluate the underlying mechanism. In this study, we found that the protein level of PRDX2 and the level of H2O2 were higher in the human AS carotid artery tissues than in the normal carotid artery tissues, accompanied with the activation of MAPK signaling pathway. The up-regulation of PRDX2 in the CAVSMCs significantly decreased the expression of ROS, collagen type I (COL I), collagen type III (COL III), vascular cell adhesion molecule-1 (VCAM-1), and intercellular adhesion molecule-1 (ICAM-1) and inhibited the proliferation, migration, and transformation of the CAVSMCs. The up-regulation of PRDX2 reversed the effect of the CAVSMCs treated with tumor necrosis factor-α (TNF-α). In addition, PRDX2 down-regulation promoted the protein levels of p-p38, p-JNK, and p-ERK, which was confirmed in relevant MAPK inhibitor treatment experiments. Our results suggest a protective role of PRDX2, as a scavenger of ROS, in AS progression through inhibiting the VSMC phenotype alteration and function via MAPK signaling pathway.
Collapse
Affiliation(s)
- Jing Li
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Cong Wang
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Wenjing Wang
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Lingzi Liu
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Qingqing Zhang
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Jun Zhang
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Bo Wang
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Shujing Wang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Institute of Glycobiology, Dalian Medical University, Dalian, China
| | - Li Hou
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Chuanzhou Gao
- Department of Electron Microscope, Dalian Medical University, Dalian, China
| | - Xiao Yu
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Lei Sun
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| |
Collapse
|
28
|
Abstract
Peroxiredoxin (Prx) refers to a family of thiol-dependent peroxidases that decompose hydrogen peroxide, lipid hydroperoxides, as well as peroxynitrite, and protect against oxidative and inflammatory stress. There are six mammalian Prx isozymes (Prx1-6), classified as typical 2-Cys, atypical 2-Cys, or 1-Cys Prxs based on the mechanism and the number of cysteine residues involved during catalysis. In addition to their well-established peroxide-scavenging activity, some Prxs also participate in the regulation of various cell signaling pathways. Extensive animal studies employing primarily gene knockout models provide substantial evidence supporting a critical protective role of Prxs in various disease processes involving oxidative and inflammatory stress. This review surveys recent research findings, published primarily in influential journals, on the involvement of various Prx isozymes in protecting against cardiovascular injury and related disorders, including diabetes, metabolic syndromes, and sepsis, whose pathophysiology all intimately involves oxidative stress and inflammation.
Collapse
|
29
|
Jeong SJ, Cho MJ, Ko NY, Kim S, Jung IH, Min JK, Lee SH, Park JG, Oh GT. Deficiency of peroxiredoxin 2 exacerbates angiotensin II-induced abdominal aortic aneurysm. Exp Mol Med 2020; 52:1587-1601. [PMID: 32929220 PMCID: PMC8080566 DOI: 10.1038/s12276-020-00498-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/21/2020] [Accepted: 06/29/2020] [Indexed: 01/02/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is an inflammatory vascular disease characterized by structural deterioration of the aorta caused by inflammation and oxidative stress, leading to aortic dilatation and rupture. Peroxiredoxin 2 (PRDX2), an antioxidant enzyme, has been reported as a potential negative regulator of inflammatory vascular diseases, and it has been identified as a protein that is increased in patients with ruptured AAA compared to patients with nonruptured AAA. In this study, we demonstrated that PRDX2 was a pivotal factor involved in the inhibition of AAA progression. PRDX2 levels were increased in AAA compared with those in normal aortas in both humans and mice. Ultrasound imaging revealed that the loss of PRDX2 accelerated the development of AAA in the early stages and increased AAA incidence in mice infused with angiotensin II (Ang II). Prdx2−/− mice infused with Ang II exhibited increased aortic dilatation and maximal aortic diameter without a change in blood pressure. Structural deterioration of the aortas from Prdx2−/− mice infused with Ang II was associated with increases in the degradation of elastin, oxidative stress, and intramural thrombi caused by microhemorrhages, immature neovessels, and the activation of matrix metalloproteinases compared to that observed in controls. Moreover, an increase in inflammatory responses, including the production of cell adhesion molecules and the accumulation of inflammatory cells and proinflammatory cytokines due to PRDX2 deficiency, accelerated Ang II-induced AAA progression. Our data confirm that PRDX2 plays a role as a negative regulator of the pathological process of AAA and suggest that increasing PRDX2 activity may be a novel strategy for the prevention and treatment of AAA. An enzyme with antioxidant properties may provide a biomarker and therapeutic agent to help treat abdominal aortic aneurysm (AAA). AAA involves the structural deterioration of the aorta through chronic inflammation and oxidative stress, and can trigger life-threatening artery rupture. An antioxidant enzyme called peroxiredoxin 2 (PRDX2) is increased in patients with ruptures, but whether its role in AAA is beneficial or detrimental is unclear. Goo Taeg Oh at the Ewha Womans University in Seoul, Jong-Gil Park at the Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea, and co-workers examined the effect of PRDX2 on AAA progression. PRDX2 suppressed structural damage in mice, limiting artery dilation and protein degradation. Loss of PRDX2 accelerated AAA development. Measuring levels of PRDX2 may indicate AAA severity in patients, while boosting the enzyme could repair aortic damage.
Collapse
Affiliation(s)
- Se-Jin Jeong
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Min Ji Cho
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience & Biotechnology, Daejeon, Republic of Korea.,Department of Biomolecular Science, University of Science & Technology (UST), Daejeon, Republic of Korea
| | - Na Young Ko
- Immune and Vascular Cell Network Research Center, National Creative Initiatives, Department of Life Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Sinai Kim
- Immune and Vascular Cell Network Research Center, National Creative Initiatives, Department of Life Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - In-Hyuk Jung
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Jeong-Ki Min
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience & Biotechnology, Daejeon, Republic of Korea.,Department of Biomolecular Science, University of Science & Technology (UST), Daejeon, Republic of Korea
| | - Sang Hak Lee
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.,Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Jong-Gil Park
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience & Biotechnology, Daejeon, Republic of Korea.
| | - Goo Taeg Oh
- Immune and Vascular Cell Network Research Center, National Creative Initiatives, Department of Life Sciences, Ewha Womans University, Seoul, Republic of Korea.
| |
Collapse
|
30
|
Jeon S, Kim TK, Jeong SJ, Jung IH, Kim N, Lee MN, Sonn SK, Seo S, Jin J, Kweon HY, Kim S, Shim D, Park YM, Lee SH, Kim KW, Cybulsky MI, Shim H, Roh TY, Park WY, Lee HO, Choi JH, Park SH, Oh GT. Anti-Inflammatory Actions of Soluble Ninjurin-1 Ameliorate Atherosclerosis. Circulation 2020; 142:1736-1751. [PMID: 32883094 DOI: 10.1161/circulationaha.120.046907] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Macrophages produce many inflammation-associated molecules, released by matrix metalloproteinases, such as adhesion molecules, and cytokines, as well, which play a crucial role in atherosclerosis. In this context, we investigated the relationship between Ninjurin-1 (Ninj1 [nerve injury-induced protein]), a novel matrix metalloproteinase 9 substrate, expression, and atherosclerosis progression. METHODS Ninj1 expression and atherosclerosis progression were assessed in atherosclerotic aortic tissue and serum samples from patients with coronary artery disease and healthy controls, and atheroprone apolipoprotein e-deficient (Apoe-/-) and wild-type mice, as well. Apoe-/- mice lacking systemic Ninj1 expression (Ninj1-/-Apoe-/-) were generated to assess the functional effects of Ninj1. Bone marrow transplantation was also used to generate low-density lipoprotein receptor-deficient (Ldlr-/-) mice that lack Ninj1 specifically in bone marrow-derived cells. Mice were fed a Western diet for 5 to 23 weeks, and atherosclerotic lesions were investigated. The anti-inflammatory role of Ninj1 was verified by treating macrophages and mice with the peptides Ninj11-56 (ML56) and Ninj126-37 (PN12), which mimic the soluble form of Ninj1 (sNinj1). RESULTS Our in vivo results conclusively showed a correlation between Ninj1 expression in aortic macrophages and the extent of human and mouse atherosclerotic lesions. Ninj1-deficient macrophages promoted proinflammatory gene expression by activating mitogen-activated protein kinase and inhibiting the phosphoinositide 3-kinase/Akt signaling pathway. Whole-body and bone marrow-specific Ninj1 deficiencies significantly increased monocyte recruitment and macrophage accumulation in atherosclerotic lesions through elevated macrophage-mediated inflammation. Macrophage Ninj1 was directly cleaved by matrix metalloproteinase 9 to generate a soluble form that exhibited antiatherosclerotic effects, as assessed in vitro and in vivo. Treatment with the sNinj1-mimetic peptides, ML56 and PN12, reduced proinflammatory gene expression in human and mouse classically activated macrophages, thereby attenuating monocyte transendothelial migration. Moreover, continuous administration of mPN12 alleviated atherosclerosis by inhibiting the enhanced monocyte recruitment and inflammation characteristics of this disorder in mice, regardless of the presence of Ninj1. CONCLUSIONS Ninj1 is a novel matrix metalloproteinase 9 substrate in macrophages, and sNinj1 is a secreted atheroprotective protein that regulates macrophage inflammation and monocyte recruitment in atherosclerosis. Moreover, sNinj1-mediated anti-inflammatory effects are conserved in human macrophages and likely contribute to human atherosclerosis.
Collapse
Affiliation(s)
- Sejin Jeon
- Immune and Vascular Cell Network Research Center, National Creative Initiatives, Department of Life Sciences (S.J., T.K.K., M.-N.L., S.-K.S., S.S., J.J., H.Y.K., S.K., G.T.O.), Ewha Womans University, Seoul, Korea
| | - Tae Kyeong Kim
- Immune and Vascular Cell Network Research Center, National Creative Initiatives, Department of Life Sciences (S.J., T.K.K., M.-N.L., S.-K.S., S.S., J.J., H.Y.K., S.K., G.T.O.), Ewha Womans University, Seoul, Korea
| | - Se-Jin Jeong
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO (S.-J.J., I.-H.J.)
| | - In-Hyuk Jung
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO (S.-J.J., I.-H.J.)
| | - Nayoung Kim
- Samsung Genome Institute, Samsung Medical Center, Seoul, Korea (N.K., W.-Y.P., H.-O.L.).,Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea (N.K., W.-Y.P., H.-O.L.)
| | - Mi-Ni Lee
- Immune and Vascular Cell Network Research Center, National Creative Initiatives, Department of Life Sciences (S.J., T.K.K., M.-N.L., S.-K.S., S.S., J.J., H.Y.K., S.K., G.T.O.), Ewha Womans University, Seoul, Korea
| | - Seong-Keun Sonn
- Immune and Vascular Cell Network Research Center, National Creative Initiatives, Department of Life Sciences (S.J., T.K.K., M.-N.L., S.-K.S., S.S., J.J., H.Y.K., S.K., G.T.O.), Ewha Womans University, Seoul, Korea
| | - Seungwoon Seo
- Immune and Vascular Cell Network Research Center, National Creative Initiatives, Department of Life Sciences (S.J., T.K.K., M.-N.L., S.-K.S., S.S., J.J., H.Y.K., S.K., G.T.O.), Ewha Womans University, Seoul, Korea
| | - Jing Jin
- Immune and Vascular Cell Network Research Center, National Creative Initiatives, Department of Life Sciences (S.J., T.K.K., M.-N.L., S.-K.S., S.S., J.J., H.Y.K., S.K., G.T.O.), Ewha Womans University, Seoul, Korea
| | - Hyae Yon Kweon
- Immune and Vascular Cell Network Research Center, National Creative Initiatives, Department of Life Sciences (S.J., T.K.K., M.-N.L., S.-K.S., S.S., J.J., H.Y.K., S.K., G.T.O.), Ewha Womans University, Seoul, Korea
| | - Sinai Kim
- Immune and Vascular Cell Network Research Center, National Creative Initiatives, Department of Life Sciences (S.J., T.K.K., M.-N.L., S.-K.S., S.S., J.J., H.Y.K., S.K., G.T.O.), Ewha Womans University, Seoul, Korea
| | - Dahee Shim
- Department of Life Science, College of Natural Sciences, Research Institute for Natural Sciences, Hanyang University, Seoul, Korea (D.S., J.-H.C.)
| | - Young Mi Park
- Department of Molecular Medicine, Ewha Womans University School of Medicine, Seoul, Korea (Y.M.P.)
| | - Sang-Hak Lee
- Division of Cardiology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea (S.-H.L.)
| | - Kyu-Won Kim
- College of Pharmacy, Seoul National University, Seoul, Korea (K.-W.K.)
| | - Myron I Cybulsky
- Toronto General Hospital Research Institute, University Health Network, and Department of Laboratory Medicine and Pathobiology, University of Toronto, ON, Canada (M.I.C.)
| | - Hyunbo Shim
- Departments of Bioinspired Science and Life Science (H.S.), Ewha Womans University, Seoul, Korea
| | - Tae-Young Roh
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Korea (T.-Y.R.)
| | - Woong-Yang Park
- Samsung Genome Institute, Samsung Medical Center, Seoul, Korea (N.K., W.-Y.P., H.-O.L.).,Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea (N.K., W.-Y.P., H.-O.L.)
| | - Hae-Ock Lee
- Samsung Genome Institute, Samsung Medical Center, Seoul, Korea (N.K., W.-Y.P., H.-O.L.).,Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea (N.K., W.-Y.P., H.-O.L.)
| | - Jae-Hoon Choi
- Department of Life Science, College of Natural Sciences, Research Institute for Natural Sciences, Hanyang University, Seoul, Korea (D.S., J.-H.C.)
| | - Sung Ho Park
- School of Life Sciences, Ulsan National Institute of Science & Technology (UNIST), Ulsan, Korea (S.H.P.)
| | - Goo Taeg Oh
- Immune and Vascular Cell Network Research Center, National Creative Initiatives, Department of Life Sciences (S.J., T.K.K., M.-N.L., S.-K.S., S.S., J.J., H.Y.K., S.K., G.T.O.), Ewha Womans University, Seoul, Korea
| |
Collapse
|
31
|
Comparative Study of Protective Action of Exogenous 2-Cys Peroxiredoxins (Prx1 and Prx2) Under Renal Ischemia-Reperfusion Injury. Antioxidants (Basel) 2020; 9:antiox9080680. [PMID: 32751232 PMCID: PMC7465264 DOI: 10.3390/antiox9080680] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/10/2020] [Accepted: 07/27/2020] [Indexed: 01/09/2023] Open
Abstract
The pathogenesis of ischemia-reperfusion (I/R) injuries is based on oxidative stress caused by a sharp increase in the concentration of free radicals, reactive oxygen species (ROS) and secondary products of free radical oxidation of biological macromolecules during reperfusion. Application of exogenous antioxidants lowers the level of ROS in the affected tissues, suppresses or adjusts the course of oxidative stress, thereby substantially reducing the severity of I/R injury. We believe that the use of antioxidant enzymes may be the most promising line of effort since they possess higher efficiency than low molecular weight antioxidants. Among antioxidant enzymes, of great interest are peroxiredoxins (Prx1–6) which reduce a wide range of organic and inorganic peroxide substrates. In an animal model of bilateral I/R injury of kidneys (using histological, biochemical, and molecular biological methods) it was shown that intravenous administration of recombinant typical 2-Cys peroxiredoxins (Prx1 and Prx2) effectively reduces the severity of I/R damage, contributing to the normalization of the structural and functional state of the kidneys and an almost 2-fold increase in the survival of experimental animals. The use of recombinant Prx1 or Prx2 can be an efficient approach for the prevention and treatment of renal I/R injury.
Collapse
|
32
|
Chen Y, Zhao X, Wu H. Arterial Stiffness: A Focus on Vascular Calcification and Its Link to Bone Mineralization. Arterioscler Thromb Vasc Biol 2020; 40:1078-1093. [PMID: 32237904 DOI: 10.1161/atvbaha.120.313131] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This review focuses on the association between vascular calcification and arterial stiffness, highlighting the important genetic factors, systemic and local microenvironmental signals, and underlying signaling pathways and molecular regulators of vascular calcification. Elevated oxidative stress appears to be a common procalcification factor that induces osteogenic differentiation and calcification of vascular cells in a variety of disease conditions such as atherosclerosis, diabetes mellitus, and chronic kidney disease. Thus, the role of oxidative stress and oxidative stress-regulated signals in vascular smooth muscle cells and their contributions to vascular calcification are highlighted. In relation to diabetes mellitus, the regulation of both hyperglycemia and increased protein glycosylation, by AGEs (advanced glycation end products) and O-linked β-N-acetylglucosamine modification, and its role in enhancing intracellular pathophysiological signaling that promotes osteogenic differentiation and calcification of vascular smooth muscle cells are discussed. In the context of chronic kidney disease, this review details the role of calcium and phosphate homeostasis, parathyroid hormone, and specific calcification inhibitors in regulating vascular calcification. In addition, the impact of the systemic and microenvironmental factors on respective intrinsic signaling pathways that promote osteogenic differentiation and calcification of vascular smooth muscle cells and osteoblasts are compared and contrasted, aiming to dissect the commonalities and distinctions that underlie the paradoxical vascular-bone mineralization disorders in aging and diseases.
Collapse
Affiliation(s)
- Yabing Chen
- From the Departments of Pathology (Y.C.), The University of Alabama at Birmingham.,Birmingham Veterans Affairs Medical Center, Research Department, AL (Y.C.)
| | - Xinyang Zhao
- Biochemistry (X.Z.), The University of Alabama at Birmingham
| | - Hui Wu
- Pediatric Dentistry (H.W.), The University of Alabama at Birmingham
| |
Collapse
|
33
|
Negre-Salvayre A, Guerby P, Gayral S, Laffargue M, Salvayre R. Role of reactive oxygen species in atherosclerosis: Lessons from murine genetic models. Free Radic Biol Med 2020; 149:8-22. [PMID: 31669759 DOI: 10.1016/j.freeradbiomed.2019.10.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 10/16/2019] [Accepted: 10/16/2019] [Indexed: 12/19/2022]
Abstract
Atherosclerosis is a multifactorial chronic and inflammatory disease of medium and large arteries, and the major cause of cardiovascular morbidity and mortality worldwide. The pathogenesis of atherosclerosis involves a number of risk factors and complex events including hypercholesterolemia, endothelial dysfunction, increased permeability to low density lipoproteins (LDL) and their sequestration on extracellular matrix in the intima of lesion-prone areas. These events promote LDL modifications, particularly by oxidation, which generates acute and chronic inflammatory responses implicated in atherogenesis and lesion progression. Reactive oxygen species (ROS) (which include both free radical and non-free radical oxygen intermediates), play a key-role at each step of atherogenesis, in endothelial dysfunction, LDL oxidation, and inflammatory events involved in the initiation and development of atherosclerosis lesions. Most advanced knowledge supporting the "oxidative theory of atherosclerosis" i.e. the nature and the cellular sources of ROS and antioxidant defences, as well as the mechanisms involved in the redox balance, is based on the use of genetically engineered animals, i.e. transgenic, genetically modified, or altered for systems producing or neutralizing ROS in the vessels. This review summarizes the results obtained from animals genetically manipulated for various sources of ROS or antioxidant defences in the vascular wall, and their relevance (advance or limitation), for understanding the place and role of ROS in atherosclerosis.
Collapse
Affiliation(s)
| | - Paul Guerby
- Inserm U-1048, Université de Toulouse, France; Pôle de gynécologie obstétrique, Hôpital Paule-de-Viguier, CHU de Toulouse, France
| | | | | | | |
Collapse
|
34
|
Lee YJ. Knockout Mouse Models for Peroxiredoxins. Antioxidants (Basel) 2020; 9:antiox9020182. [PMID: 32098329 PMCID: PMC7070531 DOI: 10.3390/antiox9020182] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/16/2020] [Accepted: 02/20/2020] [Indexed: 12/12/2022] Open
Abstract
Peroxiredoxins (PRDXs) are members of a highly conserved peroxidase family and maintain intracellular reactive oxygen species (ROS) homeostasis. The family members are expressed in most organisms and involved in various biological processes, such as cellular protection against ROS, inflammation, carcinogenesis, atherosclerosis, heart diseases, and metabolism. In mammals, six PRDX members have been identified and are subdivided into three subfamilies: typical 2-Cys (PRDX1, PRDX2, PRDX3, and PRDX4), atypical 2-Cys (PRDX5), and 1-Cys (PRDX6) subfamilies. Knockout mouse models of PRDXs have been developed to investigate their in vivo roles. This review presents an overview of the knockout mouse models of PRDXs with emphases on the biological and physiological changes of these model mice.
Collapse
Affiliation(s)
- Young Jae Lee
- Department of Biochemistry, College of Medicine, Gachon University, Incheon 21999, Korea
| |
Collapse
|
35
|
Khosravi M, Poursaleh A, Ghasempour G, Farhad S, Najafi M. The effects of oxidative stress on the development of atherosclerosis. Biol Chem 2020; 400:711-732. [PMID: 30864421 DOI: 10.1515/hsz-2018-0397] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 11/29/2018] [Indexed: 12/13/2022]
Abstract
Atherosclerosis is a cardiovascular disease (CVD) known widely world wide. Several hypothesizes are suggested to be involved in the narrowing of arteries during process of atherogenesis. The oxidative modification hypothesis is related to oxidative and anti-oxidative imbalance and is the most investigated. The aim of this study was to review the role of oxidative stress in atherosclerosis. Furthermore, it describes the roles of oxidative/anti-oxidative enzymes and compounds in the macromolecular and lipoprotein modifications and in triggering inflammatory events. The reactive oxygen (ROS) and reactive nitrogen species (RNS) are the most important endogenous sources produced by non-enzymatic and enzymatic [myeloperoxidase (MPO), nicotinamide adenine dinucleotide phosphate (NADH) oxidase and lipoxygenase (LO)] reactions that may be balanced with anti-oxidative compounds [glutathione (GSH), polyphenols and vitamins] and enzymes [glutathione peroxidase (Gpx), peroxiredoxins (Prdx), superoxide dismutase (SOD) and paraoxonase (PON)]. However, the oxidative and anti-oxidative imbalance causes the involvement of cellular proliferation and migration signaling pathways and macrophage polarization leads to the formation of atherogenic plaques. On the other hand, the immune occurrences and the changes in extra cellular matrix remodeling can develop atherosclerosis process.
Collapse
Affiliation(s)
- Mohsen Khosravi
- Biochemistry Department, Firoozabadi Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Adeleh Poursaleh
- Biochemistry Department, Firoozabadi Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Ghasem Ghasempour
- Biochemistry Department, Firoozabadi Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Shaikhnia Farhad
- Biochemistry Department, Firoozabadi Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Najafi
- Microbial Biotechnology Research Center, Biochemistry Department, Firoozabadi Hospital, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
36
|
Cha HN, Park S, Dan Y, Kim JR, Park SY. Peroxiredoxin2 Deficiency Aggravates Aging-Induced Insulin Resistance and Declines Muscle Strength. J Gerontol A Biol Sci Med Sci 2019; 74:147-154. [PMID: 29733327 DOI: 10.1093/gerona/gly113] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 05/03/2018] [Indexed: 12/11/2022] Open
Abstract
This study examined the role of peroxiredoxin2 (Prx2) in aging-induced insulin resistance and reduction in skeletal muscle function in young (2-month-old) and old (24-month-old) Prx2 knockout (KO) and wild-type mice. Plasma insulin levels increased with aging in Prx2 KO mice but not in wild-type mice. Insulin sensitivity in the whole-body and skeletal muscle as assessed with the hyperinsulinemic-euglycemic clamp was lower in Prx2 KO mice than in wild-type mice in the old group but was not significantly different between the two genotypes in the young group. Insulin-induced activation of intracellular signaling molecules was also suppressed in old Prx2 KO mice compared to their wild-type littermates. Oxidative stress, inflammation, and p53 expression levels in skeletal muscle were higher in Prx2 KO mice than in wild-type mice in the old group but were not different between the two genotypes in the young group. p53 expression was negatively correlated with skeletal muscle insulin sensitivity in old mice. Skeletal muscle mass was similar between the two genotypes but grip strength was reduced in old Prx2 KO mice compared to old wild-type mice. These results suggest that Prx2 plays a protective role in aging-induced insulin resistance and declines in muscle strength by suppressing oxidative stress.
Collapse
Affiliation(s)
- Hye-Na Cha
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, Korea.,Smart-Aging Convergence Research Center, College of Medicine, Yeungnam University, Daegu, Korea
| | - Soyoung Park
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, Korea.,Smart-Aging Convergence Research Center, College of Medicine, Yeungnam University, Daegu, Korea
| | - Yongwook Dan
- Weinberg College of Art and Sciences, Northwestern University, Chicago, Illinois
| | - Jae-Ryong Kim
- Smart-Aging Convergence Research Center, College of Medicine, Yeungnam University, Daegu, Korea.,Department of Biochemistry and Molecular Biology, College of Medicine, Yeungnam University, Daegu, Korea
| | - So-Young Park
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, Korea.,Smart-Aging Convergence Research Center, College of Medicine, Yeungnam University, Daegu, Korea
| |
Collapse
|
37
|
Effects of the Prdx2 depletion on blood pressure and life span in spontaneously hypertensive rats. Hypertens Res 2019; 42:610-617. [PMID: 30655626 DOI: 10.1038/s41440-019-0207-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 10/17/2018] [Accepted: 10/17/2018] [Indexed: 01/12/2023]
Abstract
Oxidative stress is involved in the pathogenesis of hypertension and hypertensive organ damage. Our previous study suggested that stroke-prone spontaneously hypertensive rats (SHRSP) exhibited greater oxidative stress than SHR and that the stroke incidence was significantly greater in SHRSP than SHR. Therefore, we hypothesized that oxidative stress was responsible for the stroke susceptibility in SHRSP. The present study constructed Prdx2 (a gene coding an antioxidative enzyme)-knockout (KO) SHR to examine whether Prdx2 knockout would make SHR more vulnerable to hypertensive organ damage, including stroke. Prdx2-KO SHR were created using CRISPR/CAS9 for genome editing. Eight-week-old male SHR and Prdx2-KO SHR were fed 1% NaCl for 2 months to induce blood pressure (BP) changes and stroke occurrence. The baseline BP was significantly greater in KO SHR, and this difference disappeared after salt loading. The life span of KO SHR was significantly reduced compared to that of SHR despite no differences in BP under salt-loading. However, no stroke was observed in KO SHR. The severity of hypertensive renal and cardiac injuries did not differ significantly between the two strains, but oxidative stress, evaluated using urinary isoprostane excretion and DHE staining, was greater in KO SHR. These results indicated that the Prdx2-depletion caused a shorter life span and modest BP increase in SHR via increased oxidative stress. The pathophysiological roles of oxidative stress in this model should be clarified in future studies.
Collapse
|
38
|
Kato R, Hayashi M, Aiuchi T, Sawada N, Obama T, Itabe H. Temporal and spatial changes of peroxiredoxin 2 levels in aortic media at very early stages of atherosclerotic lesion formation in apoE-knockout mice. Free Radic Biol Med 2019; 130:348-360. [PMID: 30395970 DOI: 10.1016/j.freeradbiomed.2018.10.458] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 10/30/2018] [Accepted: 10/31/2018] [Indexed: 01/21/2023]
Abstract
The events that trigger early onset of atherosclerotic lesion formation are poorly understood. Initially, microscopic atherosclerotic lesions appear in the aortic root in 10-week-old apoE-knockout mice that are fed normal chow. Using proteome and immunohistochemical analyses, we investigated proteins in aortic media whose expression changes in athero-prone regions at the beginning of lesion formation. Protein profiles of the root/arch and thoracic/abdominal regions of aortas in 10-week-old apoE-knockout mice were analyzed using 2D-gel electrophoresis. Proteins in 81 spots with different abundance were identified. Among them, we focused on proteins related to oxidative stress and smooth muscle cells (SMCs). The level of peroxiredoxin 2 (Prx2), a major cellular antioxidant enzyme that reduces hydrogen peroxide, was lower in aortic root/arch compared with thoracic/abdominal aorta. Immunohistochemical staining demonstrated that Prx2 expression in SMCs in the aortic root was high at 4 weeks and decreased at 10 weeks in apoE-knockout mice, while Prx2 expression in the aorta was unchanged in wild-type mice. The level of Prx2 expression correlated positively with the SMC differentiation markers, α-smooth muscle actin and transgelin, suggesting that a decline in Prx2 expression accompanies SMC dedifferentiation. Accumulated acrolein-modified proteins and the infiltration of macrophages in aortic media were observed in areas with low Prx2 expression. These results showed that Prx2 expression declines in athero-prone aortic root before lesion formation, and this reduction in Prx2 expression correlates with lipid peroxidation, SMC dedifferentiation, and macrophage recruitment.
Collapse
Affiliation(s)
- Rina Kato
- Division of Biological Chemistry, Department of Pharmaceutical Sciences, Showa University School of Pharmacy, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Masataka Hayashi
- Division of Biological Chemistry, Department of Pharmaceutical Sciences, Showa University School of Pharmacy, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Toshihiro Aiuchi
- Division of Biological Chemistry, Department of Pharmaceutical Sciences, Showa University School of Pharmacy, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Naoko Sawada
- Division of Biological Chemistry, Department of Pharmaceutical Sciences, Showa University School of Pharmacy, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Takashi Obama
- Division of Biological Chemistry, Department of Pharmaceutical Sciences, Showa University School of Pharmacy, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Hiroyuki Itabe
- Division of Biological Chemistry, Department of Pharmaceutical Sciences, Showa University School of Pharmacy, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan.
| |
Collapse
|
39
|
Mass Spectrometry Based Comparative Proteomics Using One Dimensional and Two Dimensional SDS-PAGE of Rat Atria Induced with Obstructive Sleep Apnea. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1140:541-561. [DOI: 10.1007/978-3-030-15950-4_32] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
40
|
Nrf2 in aging - Focus on the cardiovascular system. Vascul Pharmacol 2018; 112:42-53. [PMID: 30170173 DOI: 10.1016/j.vph.2018.08.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 08/09/2018] [Accepted: 08/20/2018] [Indexed: 02/07/2023]
Abstract
Aging is the most critical risk factor for the development of cardiovascular diseases and their complications. Therefore, the fine-tuning of cellular response to getting older is an essential target for prospective therapies in cardiovascular medicine. One of the most promising targets might be the transcription factor Nrf2, which drives the expression of cytoprotective and antioxidative genes. Importantly, Nrf2 expression correlates with potential lifespan in rodents. However, the effect of Nrf2 activity in vascular diseases might be ambiguous and strongly depend on the cell type. On the one hand, the Nrf2 activity may protect cells from oxidative stress and senescence, on the other hand, total lack of Nrf2 is protective against atherosclerosis development. Therefore, this review aims to discuss the current knowledge on the role played by the transcription factor Nrf2 in cardiovascular diseases and its potential effects on aging.
Collapse
|
41
|
Langford TF, Huang BK, Lim JB, Moon SJ, Sikes HD. Monitoring the action of redox-directed cancer therapeutics using a human peroxiredoxin-2-based probe. Nat Commun 2018; 9:3145. [PMID: 30087344 PMCID: PMC6081480 DOI: 10.1038/s41467-018-05557-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 06/29/2018] [Indexed: 01/05/2023] Open
Abstract
Redox cancer therapeutics target the increased reliance on intracellular antioxidant systems and enhanced susceptibility to oxidant-induced stress of some cancer cells compared to normal cells. Many of these therapeutics are thought to perturb intracellular levels of the oxidant hydrogen peroxide (H2O2), a signaling molecule that modulates a number of different processes in human cells. However, fluorescent probes for this species remain limited in their ability to detect the small perturbations induced during successful treatments. We report a fluorescent sensor based upon human peroxiredoxin-2, which acts as the natural indicator of small H2O2 fluctuations in human cells. The new probe reveals peroxide-induced oxidation in human cells below the detection limit of current probes, as well as peroxiredoxin-2 oxidation caused by two different redox cancer therapeutics in living cells. This capability will be useful in elucidating the mechanism of current redox-based therapeutics and in developing new ones.
Collapse
Affiliation(s)
- Troy F Langford
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, 02139, USA
| | - Beijing K Huang
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, 02139, USA
| | - Joseph B Lim
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, 02139, USA
| | - Sun Jin Moon
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, 02139, USA
| | - Hadley D Sikes
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, 02139, USA.
| |
Collapse
|
42
|
Park SJ, Kim JH, Lee DG, Kim JM, Lee DS. Peroxiredoxin 2 deficiency accelerates age-related ovarian failure through the reactive oxygen species-mediated JNK pathway in mice. Free Radic Biol Med 2018; 123:96-106. [PMID: 29778464 DOI: 10.1016/j.freeradbiomed.2018.05.059] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 05/09/2018] [Accepted: 05/12/2018] [Indexed: 11/21/2022]
Abstract
Reactive oxygen species (ROS) produced in biological reactions have been shown to contribute to ovarian aging. Peroxiredoxin 2 (Prx2) is an antioxidant enzyme that protects cells by scavenging ROS; however, its effect on age-related, oxidative stress-associated ovarian failure has not been reported. Here, we investigated its role in age-related ovarian dysfunction and 4-vinylcyclohexene diepoxide (VCD)-induced premature ovarian failure using Prx2-deficient mice. Compared to those in wildtype (WT) mice, serum levels of anti-Müllerian hormone, 17β-estradiol, and progesterone and numbers of follicles and corpora lutea were significantly lower in 18-month-old Prx2-/- mice. Moreover, levels of Bax, cytochrome c, cleaved caspase-3, and phosphorylated JNK proteins were higher and numbers of apoptotic (terminal deoxynucleotidyl transferase dUTP nick end labeling-positive) cells were considerably greater in 18-month-old Prx2-/- ovaries than WT ovaries. Furthermore, the effects of the ovarian toxicant VCD in significantly enhancing ROS levels and apoptosis through activation of JNK-mediated apoptotic signaling were more pronounced in Prx2-/- than WT mouse embryonic fibroblasts. Expression of the steroidogenic proteins StAR, CYP11A1, and 3β-HSD and serum levels of 17β-estradiol and progesterone were also reduced to a greater extent in Prx2-/- mice than WT mice after VCD injection. This reduced steroidogenesis was rescued by addition of the Prx mimic ebselen or JNK inhibitor SP600125. This constitutes the first report that Prx2 deficiency leads to acceleration of age-related or VCD-induced ovarian failure by activation of the ROS-induced JNK pathway. These findings suggest that Prx2 plays an important role in preventing accelerated ovarian failure by inhibiting ROS-induced JNK activation.
Collapse
Affiliation(s)
- Sun-Ji Park
- School of Life Sciences and Biotechnology, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea; Renal Division, Washington University School of Medicine, St. Louis, MO, USA
| | - Jung-Hak Kim
- School of Life Sciences and Biotechnology, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea; Division of Endocrinology, Internal Medicine, University of California, Davis, CA, USA
| | - Dong Gil Lee
- School of Life Sciences and Biotechnology, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Jin-Man Kim
- Cancer Research Institute and Department of Pathology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Dong-Seok Lee
- School of Life Sciences and Biotechnology, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea.
| |
Collapse
|
43
|
Bolduc JA, Nelson KJ, Haynes AC, Lee J, Reisz JA, Graff AH, Clodfelter JE, Parsonage D, Poole LB, Furdui CM, Lowther WT. Novel hyperoxidation resistance motifs in 2-Cys peroxiredoxins. J Biol Chem 2018; 293:11901-11912. [PMID: 29884768 PMCID: PMC6066324 DOI: 10.1074/jbc.ra117.001690] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 05/29/2018] [Indexed: 01/07/2023] Open
Abstract
2-Cys peroxiredoxins (Prxs) modulate hydrogen peroxide (H2O2)-mediated cell signaling. At high H2O2 levels, eukaryotic Prxs can be inactivated by hyperoxidation and are classified as sensitive Prxs. In contrast, prokaryotic Prxs are categorized as being resistant to hyperoxidation and lack the GGLG and C-terminal YF motifs present in the sensitive Prxs. Additional molecular determinants that account for the subtle differences in the susceptibility to hyperoxidation remain to be identified. A comparison of a new, 2.15-Å-resolution crystal structure of Prx2 in the oxidized, disulfide-bonded state with the hyperoxidized structure of Prx2 and Prx1 in complex with sulfiredoxin revealed three structural regions that rearrange during catalysis. With these regions in hand, focused sequence analyses were performed comparing sensitive and resistant Prx groups. From this combinatorial approach, we discovered two novel hyperoxidation resistance motifs, motifs A and B, which were validated using mutagenesis of sensitive human Prxs and resistant Salmonella enterica serovar Typhimurium AhpC. Introduction and removal of these motifs, respectively, resulted in drastic changes in the sensitivity to hyperoxidation with Prx1 becoming 100-fold more resistant to hyperoxidation and AhpC becoming 800-fold more sensitive to hyperoxidation. The increased sensitivity of the latter AhpC variant was also confirmed in vivo These results support the function of motifs A and B as primary drivers for tuning the sensitivity of Prxs to different levels of H2O2, thus enabling the initiation of variable signaling or antioxidant responses in cells.
Collapse
Affiliation(s)
| | | | | | - Jingyun Lee
- Wake Forest Baptist Comprehensive Cancer Center, and
| | - Julie A. Reisz
- Section on Molecular Medicine, Department of Internal Medicine
| | - Aaron H. Graff
- From the Center for Structural Biology, Department of Biochemistry
| | | | - Derek Parsonage
- From the Center for Structural Biology, Department of Biochemistry
| | - Leslie B. Poole
- From the Center for Structural Biology, Department of Biochemistry, ,Wake Forest Baptist Comprehensive Cancer Center, and ,Center for Redox Biology and Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157 and ,Center for Molecular Signaling, Wake Forest University, Winston-Salem, North Carolina 27101
| | - Cristina M. Furdui
- Section on Molecular Medicine, Department of Internal Medicine, ,Wake Forest Baptist Comprehensive Cancer Center, and ,Center for Redox Biology and Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157 and ,Center for Molecular Signaling, Wake Forest University, Winston-Salem, North Carolina 27101
| | - W. Todd Lowther
- From the Center for Structural Biology, Department of Biochemistry, ,Wake Forest Baptist Comprehensive Cancer Center, and ,Center for Redox Biology and Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157 and ,Center for Molecular Signaling, Wake Forest University, Winston-Salem, North Carolina 27101, To whom correspondence should be addressed:
Center for Structural Biology, Dept. of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27157. Tel.:
336-716-7230; Fax:
336-713-1283; E-mail:
| |
Collapse
|
44
|
Sharapov MG, Fesenko EE, Novoselov VI. The Role of Peroxiredoxins in Various Diseases Caused by Oxidative Stress and the Prospects of Using Exogenous Peroxiredoxins. Biophysics (Nagoya-shi) 2018. [DOI: 10.1134/s0006350918040164] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|
45
|
Detienne G, De Haes W, Mergan L, Edwards SL, Temmerman L, Van Bael S. Beyond ROS clearance: Peroxiredoxins in stress signaling and aging. Ageing Res Rev 2018; 44:33-48. [PMID: 29580920 DOI: 10.1016/j.arr.2018.03.005] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 03/21/2018] [Indexed: 12/12/2022]
Abstract
Antioxidants were long predicted to have lifespan-promoting effects, but in general this prediction has not been well supported. While some antioxidants do seem to have a clear effect on longevity, this may not be primarily as a result of their role in the removal of reactive oxygen species, but rather mediated by other mechanisms such as the modulation of intracellular signaling. In this review we discuss peroxiredoxins, a class of proteinaceous antioxidants with redox signaling and chaperone functions, and their involvement in regulating longevity and stress resistance. Peroxiredoxins have a clear role in the regulation of lifespan and survival of many model organisms, including the mouse, Caenorhabditis elegans and Drosophila melanogaster. Recent research on peroxiredoxins - in these models and beyond - has revealed surprising new insights regarding the interplay between peroxiredoxins and longevity signaling, which will be discussed here in detail. As redox signaling is emerging as a potentially important player in the regulation of longevity and aging, increased knowledge of these fascinating antioxidants and their mode(s) of action is paramount.
Collapse
Affiliation(s)
- Giel Detienne
- Department of Biology, KU Leuven, Naamsestraat 59, 3000 Leuven, Belgium.
| | - Wouter De Haes
- Department of Biology, KU Leuven, Naamsestraat 59, 3000 Leuven, Belgium.
| | - Lucas Mergan
- Department of Biology, KU Leuven, Naamsestraat 59, 3000 Leuven, Belgium.
| | - Samantha L Edwards
- Department of Biology, KU Leuven, Naamsestraat 59, 3000 Leuven, Belgium.
| | - Liesbet Temmerman
- Department of Biology, KU Leuven, Naamsestraat 59, 3000 Leuven, Belgium.
| | - Sven Van Bael
- Department of Biology, KU Leuven, Naamsestraat 59, 3000 Leuven, Belgium.
| |
Collapse
|
46
|
Implications of plasma thiol redox in disease. Clin Sci (Lond) 2018; 132:1257-1280. [DOI: 10.1042/cs20180157] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 05/09/2018] [Accepted: 05/22/2018] [Indexed: 12/21/2022]
Abstract
Thiol groups are crucially involved in signaling/homeostasis through oxidation, reduction, and disulphide exchange. The overall thiol pool is the resultant of several individual pools of small compounds (e.g. cysteine), peptides (e.g. glutathione), and thiol proteins (e.g. thioredoxin (Trx)), which are not in equilibrium and present specific oxidized/reduced ratios. This review addresses mechanisms and implications of circulating plasma thiol/disulphide redox pools, which are involved in several physiologic processes and explored as disease biomarkers. Thiol pools are regulated by mechanisms linked to their intrinsic reactivity against oxidants, concentration of antioxidants, thiol-disulphide exchange rates, and their dynamic release/removal from plasma. Major thiol couples determining plasma redox potential (Eh) are reduced cysteine (CyS)/cystine (the disulphide form of cysteine) (CySS), followed by GSH/disulphide-oxidized glutathione (GSSG). Hydrogen peroxide and hypohalous acids are the main plasma oxidants, while water-soluble and lipid-soluble small molecules are the main antioxidants. The thiol proteome and thiol-oxidoreductases are emerging investigative areas given their specific disease-related responses (e.g. protein disulphide isomerases (PDIs) in thrombosis). Plasma cysteine and glutathione redox couples exhibit pro-oxidant changes directly correlated with ageing/age-related diseases. We further discuss changes in thiol-disulphide redox state in specific groups of diseases: cardiovascular, cancer, and neurodegenerative. These results indicate association with the disease states, although not yet clear-cut to yield specific biomarkers. We also highlight mechanisms whereby thiol pools affect atherosclerosis pathophysiology. Overall, it is unlikely that a single measurement provides global assessment of plasma oxidative stress. Rather, assessment of individual thiol pools and thiol-proteins specific to any given condition has more solid and logical perspective to yield novel relevant information on disease risk and prognosis.
Collapse
|
47
|
Ruotsalainen AK, Lappalainen JP, Heiskanen E, Merentie M, Sihvola V, Näpänkangas J, Lottonen-Raikaslehto L, Kansanen E, Adinolfi S, Kaarniranta K, Ylä-Herttuala S, Jauhiainen M, Pirinen E, Levonen AL. Nuclear factor E2-related factor 2 deficiency impairs atherosclerotic lesion development but promotes features of plaque instability in hypercholesterolaemic mice. Cardiovasc Res 2018; 115:243-254. [DOI: 10.1093/cvr/cvy143] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 06/13/2018] [Indexed: 12/27/2022] Open
Abstract
Abstract
Aims
Oxidative stress and inflammation play an important role in the progression of atherosclerosis. Transcription factor NF-E2-related factor 2 (Nrf2) has antioxidant and anti-inflammatory effects in the vessel wall, but paradoxically, global loss of Nrf2 in apoE deficient mice alleviates atherosclerosis. In this study, we investigated the effect of global Nrf2 deficiency on early and advanced atherogenesis in alternative models of atherosclerosis, LDL receptor deficient mice (LDLR−/−), and LDLR−/− mice expressing apoB-100 only (LDLR−/− ApoB100/100) having a humanized lipoprotein profile.
Methods and results
LDLR−/− mice were fed a high-fat diet (HFD) for 6 or 12 weeks and LDLR−/−ApoB100/100 mice a regular chow diet for 6 or 12 months. Nrf2 deficiency significantly reduced early and more advanced atherosclerosis assessed by lesion size and coverage in the aorta in both models. Nrf2 deficiency in LDLR−/− mice reduced total plasma cholesterol after 6 weeks of HFD and triglycerides in LDLR−/−ApoB100/100 mice on a chow diet. Nrf2 deficiency aggravated aortic plaque maturation in aged LDLR−/−ApoB100/100 mice as it increased plaque calcification. Moreover, ∼36% of Nrf2−/−LDLR−/−ApoB100/100 females developed spontaneous myocardial infarction (MI) or sudden death at 5 to 12 months of age. Interestingly, Nrf2 deficiency increased plaque instability index, enhanced plaque inflammation and calcification, and reduced fibrous cap thickness in brachiocephalic arteries of LDLR−/−ApoB100/100 female mice at age of 12 months.
Conclusions
Absence of Nrf2 reduced atherosclerotic lesion size in both atherosclerosis models, likely via systemic effects on lipid metabolism. However, Nrf2 deficiency in aged LDLR−/−ApoB100/100 mice led to an enhanced atherosclerotic plaque instability likely via increased plaque inflammation and oxidative stress, which possibly predisposed to MI and sudden death.
Collapse
Affiliation(s)
| | - Jari P Lappalainen
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
- Department of Clinical Chemistry, University of Eastern Finland and Eastern Finland Laboratory Centre, Kuopio, Finland
| | - Emmi Heiskanen
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Mari Merentie
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Virve Sihvola
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Juha Näpänkangas
- Department of Pathology, Oulu University Hospital and University of Oulu, Oulu, Finland
| | | | - Emilia Kansanen
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Simone Adinolfi
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Kai Kaarniranta
- Department of Ophthalmology, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Seppo Ylä-Herttuala
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
- Heart Center and Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland
| | - Matti Jauhiainen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
- National Institute for Health and Welfare, Genomics and Biomarkes Unit, Helsinki, Finland; and
| | - Eija Pirinen
- Research Program for Molecular Neurology, University of Helsinki, Helsinki, Finland
| | | |
Collapse
|
48
|
Aboo Bakkar Z, Fulford J, Gates PE, Jackman SR, Jones AM, Bond B, Bowtell JL. Prolonged forearm ischemia attenuates endothelium-dependent vasodilatation and plasma nitric oxide metabolites in overweight middle-aged men. Eur J Appl Physiol 2018; 118:1565-1572. [PMID: 29785503 PMCID: PMC6060779 DOI: 10.1007/s00421-018-3886-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 05/06/2018] [Indexed: 12/01/2022]
Abstract
Purpose Repeated cycles of endothelial ischemia–reperfusion injury and the resulting respiratory burst contribute to the irreversible pathophysiology of vascular diseases, and yet, the effects of ischemia reperfusion on vascular function, oxidative stress, and nitric oxide (NO) bioavailability have not been assessed simultaneously. Therefore, this study sought to examine the effects of prolonged forearm occlusion and subsequent reperfusion on NO-dependent brachial artery endothelial function. Methods Flow-mediated dilatation was measured at baseline and 15, 30, and 45 min after 20-min forearm occlusion in 14 healthy, but physically inactive middle-aged men (53.7 ± 1.2 years, BMI: 28.1 ± 0.1 kg m−2). Venous blood samples collected from the occluded arm were analyzed for NO metabolites and markers of oxidative stress. Results FMD was significantly depressed after the prolonged occlusion compared to baseline, with a significant reduction 15-min post-occlusion (6.6 ± 0.7 to 2.9 ± 0.4%, p < 0.001); FMD remained depressed after 30 min (4.1 ± 0.6%, p = 0.001), but was not significantly different to baseline after 45-min recovery (5.4 ± 0.7%, p = 0.079). Plasma nitrate (main time effect: p = 0.015) and nitrite (main time effect: p = 0.034) concentrations were significantly reduced after prolonged occlusion. Plasma catalase activity was significantly elevated at 4- (p = 0.016) and 45-min (p = 0.001) post-occlusion, but plasma peroxiredoxin 2 and protein carbonyl content did not change. Conclusions Prolonged forearm occlusion resulted in acute impairment of endothelium-dependent vasodilatation of the brachial artery for at least 30 min after reperfusion. We demonstrate that this vascular dysfunction is associated with oxidative stress and reduced NO bioavailability following reperfusion.
Collapse
Affiliation(s)
- Zainie Aboo Bakkar
- Sport and Health Sciences, College of Life and Environmental Sciences, University of Exeter, Exeter, EX1 2LU, UK
| | | | | | - Sarah R Jackman
- Sport and Health Sciences, College of Life and Environmental Sciences, University of Exeter, Exeter, EX1 2LU, UK
| | - Andrew M Jones
- Sport and Health Sciences, College of Life and Environmental Sciences, University of Exeter, Exeter, EX1 2LU, UK
| | - Bert Bond
- Sport and Health Sciences, College of Life and Environmental Sciences, University of Exeter, Exeter, EX1 2LU, UK
| | - Joanna L Bowtell
- Sport and Health Sciences, College of Life and Environmental Sciences, University of Exeter, Exeter, EX1 2LU, UK.
| |
Collapse
|
49
|
Shukitt-Hale B, Thangthaeng N, Kelly ME, Smith DE, Miller MG. Raspberry differentially improves age-related declines in psychomotor function dependent on baseline motor ability. Food Funct 2018; 8:4752-4759. [PMID: 29168860 DOI: 10.1039/c7fo00894e] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Among older adults, falls are a leading cause of distress, pain, injury, loss of confidence, and ultimately, loss of independence and death. Previous studies in our laboratory have demonstrated that berry supplementation improves the age-related declines in balance, muscle strength, and coordination that often lead to falls, even when initiated later in life. The purpose of this study was to explore the interaction between baseline motor performance and the daily intake of raspberry required to improve/preserve motor function. Aged male F344 (17 mo) rats were tested for baseline (pre-test) balance, muscle strength, and coordination, and divided into good, average, and poor performers based on their motor composite score. Rats in each category were fed with either a control, 1%, or 2% raspberry-supplemented diet for 8 weeks and then retested (post-test). Poor performers fed with 1% or 2% raspberry had higher post-test composite scores (p < 0.05), while 2% raspberry lowered post-test composite scores in the good performers (p < 0.05), compared to control-fed rats. 1% and 2% raspberry appeared to preserve the performance of good performers and improve the performance of poor performers on plank walking (p < 0.05), while 2% raspberry improved post-test grip strength of the poor performers (p < 0.05). Additionally, rats with lower post-diet composite scores had higher levels of serum IL-1β levels (r = -0.347, p < 0.05). These findings identified poor performers as being the most likely to benefit from daily consumption of ½-2 cups of raspberry to improve/preserve motor function. Therefore, increased raspberry consumption may reduce fall risk, extend independence, and improve quality of life in the aging population.
Collapse
Affiliation(s)
- Barbara Shukitt-Hale
- USDA-ARS, Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA.
| | | | | | | | | |
Collapse
|
50
|
Abstract
Reactive oxygen species (ROS) are well known for their role in mediating both physiological and pathophysiological signal transduction. Enzymes and subcellular compartments that typically produce ROS are associated with metabolic regulation, and diseases associated with metabolic dysfunction may be influenced by changes in redox balance. In this review, we summarize the current literature surrounding ROS and their role in metabolic and inflammatory regulation, focusing on ROS signal transduction and its relationship to disease progression. In particular, we examine ROS production in compartments such as the cytoplasm, mitochondria, peroxisome, and endoplasmic reticulum and discuss how ROS influence metabolic processes such as proteasome function, autophagy, and general inflammatory signaling. We also summarize and highlight the role of ROS in the regulation metabolic/inflammatory diseases including atherosclerosis, diabetes mellitus, and stroke. In order to develop therapies that target oxidative signaling, it is vital to understand the balance ROS signaling plays in both physiology and pathophysiology, and how manipulation of this balance and the identity of the ROS may influence cellular and tissue homeostasis. An increased understanding of specific sources of ROS production and an appreciation for how ROS influence cellular metabolism may help guide us in the effort to treat cardiovascular diseases.
Collapse
Affiliation(s)
- Steven J Forrester
- From the Division of Cardiology, Department of Medicine, Emory University, Atlanta GA
| | - Daniel S Kikuchi
- From the Division of Cardiology, Department of Medicine, Emory University, Atlanta GA
| | - Marina S Hernandes
- From the Division of Cardiology, Department of Medicine, Emory University, Atlanta GA
| | - Qian Xu
- From the Division of Cardiology, Department of Medicine, Emory University, Atlanta GA
| | - Kathy K Griendling
- From the Division of Cardiology, Department of Medicine, Emory University, Atlanta GA.
| |
Collapse
|