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
|
Zhang T, Li L, Mo X, Xie S, Liu S, Zhao N, Zhang H, Chen S, Zeng X, Wang S, Deng W, Tang Q. Matairesinol blunts adverse cardiac remodeling and heart failure induced by pressure overload by regulating Prdx1 and PI3K/AKT/FOXO1 signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156054. [PMID: 39306883 DOI: 10.1016/j.phymed.2024.156054] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 08/20/2024] [Accepted: 09/13/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND Pathological cardiac remodeling is a critical process leading to heart failure, characterized primarily by inflammation and apoptosis. Matairesinol (Mat), a key chemical component of Podocarpus macrophyllus resin, exhibits a wide range of pharmacological activities, including anti-hydatid, antioxidant, antitumor, and anti-inflammatory effects. PURPOSE This study aims to investigate whether Matairesinol alleviate cardiac hypertrophy and remodeling caused by pressure overload and to elucidate its mechanism of action. METHODS An in vitro pressure loading model was established using neonatal rat cardiomyocytes treated with angiotensin Ⅱ, while an in vivo model was created using C57 mice subjected to transverse aortic constriction (TAC). To activate the PI3K/Akt/FoxO1 pathway, Ys-49 was employed. Moreover, small interfering RNA (siRNA) and short hairpin RNA (shRNA) were utilized to silence Prdx1 expression both in vitro and in vivo. Various techniques, including echocardiography, wheat germ agglutinin (WGA) staining, HE staining, PSR staining, and Masson trichrome staining, were used to assess cardiac function, cardiomyocyte cross-sectional area, and fibrosis levels in rats. Apoptosis in myocardial tissue and in vitro was detected by TUNEL assay, while reactive oxygen species (ROS) content in tissues and cells was measured using DHE staining. Furthermore, the affinity of Prdx1 with Mat and PI3K was analyzed using computer-simulated molecular docking. Western blotting and RT-PCR were utilized to evaluate Prdx1 levels and proteins related to apoptosis and oxidative stress, as well as the mRNA levels of cardiac hypertrophy and fibrosis-related indicators. RESULTS Mat significantly alleviated cardiac hypertrophy and fibrosis induced by TAC, preserved cardiac function, and markedly reduced cardiomyocyte apoptosis and oxidative damage. In vitro, mat attenuated ang Ⅱ - induced hypertrophy of nrvms and activation of neonatal rat fibroblasts. Notably, activation of the PI3K/Akt/FoxO1 pathway and downregulation of Prdx1 expression were observed in TAC mice; however, these effects were reversed by Mat treatment. Furthermore, Prdx1 knockdown activated the PI3K/Akt/FoxO1 pathway, leading to exacerbation of the disease. Molecular docking indicated that Molecular docking indicated that Mat upregulated Prdx1 expression by binding to it, thereby inhibiting the PI3K/Akt/FoxO1 pathway and protecting the heart by restoring Prdx1 expression levels. CONCLUSION Matairesinol alleviates pressure overload-induced cardiac remodeling both in vivo and in vitro by upregulating Prdx1 expression and inhibiting the PI3K/Akt/FoxO1 pathway. This study highlights the therapeutic potential of Matairesinol in the treatment of cardiac hypertrophy and remodeling, providing a promising avenue for future research and clinical application.
Collapse
Key Words
- ANP, atrial natriuretic peptide
- Abbreviations: MAT, matairesinol
- BNP, B-type natriuretic peptide
- Cardiac fibrosis
- Cardiac hypertrophy
- Cardiac remodeling
- LV, left ventricular
- LVEDd, left ventricular end-diastolic dimension
- LVEF, left ventricular ejection fraction
- Matairesinol
- NRCFS, neonatal rat cardiac fibroblasts
- PRDX 1
- PRDX1, peroxiredoxin 1
- ROS, reactive oxygen species
- Sh-RNA, short-hairpin RNA
- Si-RNA, small interfering RNA
- TAC, transverse aortic contraction
- β-MHC, Β-myosin heavy chain
Collapse
Affiliation(s)
- Tong Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Lanlan Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Xiaotong Mo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Saiyang Xie
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Shiqiang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Nan Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Heng Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Si Chen
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, PR China
| | - Xiaofeng Zeng
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, PR China
| | - Shasha Wang
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, PR China
| | - Wei Deng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Qizhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China.
| |
Collapse
|
3
|
Chan JSF, Tabatabaei Dakhili SA, Lorenzana-Carrillo MA, Gopal K, Pulente SM, Greenwell AA, Yang K, Saed CT, Stenlund MJ, Ferrari SR, Mangra-Bala IA, Shafaati T, Bhat RK, Eaton F, Overduin M, Jørgensen SB, Steinberg GR, Mulvihill EE, Sutendra G, Ussher JR. Growth differentiation factor 15 alleviates diastolic dysfunction in mice with experimental diabetic cardiomyopathy. Cell Rep 2024; 43:114573. [PMID: 39093701 DOI: 10.1016/j.celrep.2024.114573] [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: 03/25/2024] [Revised: 06/19/2024] [Accepted: 07/17/2024] [Indexed: 08/04/2024] Open
Abstract
Growth differentiation factor 15 (GDF15) is a peptide with utility in obesity, as it decreases appetite and promotes weight loss. Because obesity increases the risk for type 2 diabetes (T2D) and cardiovascular disease, it is imperative to understand the cardiovascular actions of GDF15, especially since elevated GDF15 levels are an established biomarker for heart failure. As weight loss should be encouraged in the early stages of obesity-related prediabetes/T2D, where diabetic cardiomyopathy is often present, we assessed whether treatment with GDF15 influences its pathology. We observed that GDF15 treatment alleviates diastolic dysfunction in mice with T2D independent of weight loss. This cardioprotection was associated with a reduction in cardiac inflammation, which was likely mediated via indirect actions, as direct treatment of adult mouse cardiomyocytes and differentiated THP-1 human macrophages with GDF15 failed to alleviate lipopolysaccharide-induced inflammation. Therapeutic manipulation of GDF15 action may thus have utility for both obesity and diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Jordan S F Chan
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, AB T6G 2E1, Canada; Cardiovascular Research Institute, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Seyed Amirhossein Tabatabaei Dakhili
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, AB T6G 2E1, Canada; Cardiovascular Research Institute, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Maria Areli Lorenzana-Carrillo
- Cardiovascular Research Institute, University of Alberta, Edmonton, AB T6G 1C9, Canada; Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2G3, Canada
| | - Keshav Gopal
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, AB T6G 2E1, Canada; Cardiovascular Research Institute, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Serena M Pulente
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada; University of Ottawa Heart Institute, Ottawa, ON K1Y 4W7, Canada
| | - Amanda A Greenwell
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, AB T6G 2E1, Canada; Cardiovascular Research Institute, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Kunyan Yang
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, AB T6G 2E1, Canada; Cardiovascular Research Institute, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Christina T Saed
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, AB T6G 2E1, Canada; Cardiovascular Research Institute, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Magnus J Stenlund
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, AB T6G 2E1, Canada; Cardiovascular Research Institute, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Sally R Ferrari
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, AB T6G 2E1, Canada; Cardiovascular Research Institute, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Indiresh A Mangra-Bala
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, AB T6G 2E1, Canada; Cardiovascular Research Institute, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Tanin Shafaati
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, AB T6G 2E1, Canada; Cardiovascular Research Institute, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Rakesh K Bhat
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Farah Eaton
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, AB T6G 2E1, Canada; Cardiovascular Research Institute, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Michael Overduin
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | | | - Gregory R Steinberg
- Centre for Metabolism, Obesity, Diabetes Research, McMaster University, Hamilton, ON L8S 4K1, Canada; Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Erin E Mulvihill
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada; University of Ottawa Heart Institute, Ottawa, ON K1Y 4W7, Canada
| | - Gopinath Sutendra
- Cardiovascular Research Institute, University of Alberta, Edmonton, AB T6G 1C9, Canada; Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2G3, Canada
| | - John R Ussher
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, AB T6G 2E1, Canada; Cardiovascular Research Institute, University of Alberta, Edmonton, AB T6G 1C9, Canada.
| |
Collapse
|
4
|
Bil-Lula I, Kuliczkowski W, Krzywonos-Zawadzka A, Frydrychowski P, Stygar D, Hałucha K, Noszczyk-Nowak A. Mixture of Doxycycline, ML-7 and L-NAME Restores the Pro- and Antioxidant Balance during Myocardial Infarction-In Vivo Pig Model Study. Biomedicines 2024; 12:784. [PMID: 38672140 PMCID: PMC11047935 DOI: 10.3390/biomedicines12040784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
The restoration of blood flow to the ischemic myocardium inflicts ischemia/reperfusion (I/R) heart injury (IRI). The main contributors to IRI are increased oxidative stress and subsequent excessive production of ROS, increased expression of NOS and peroxinitate, activation of MMPs, and enhanced posttranslational modifications of contractile proteins, which make them more susceptible to proteolytic degradation. Since the pathophysiology of IRI is a complex issue, and thus, various therapeutic strategies are required to prevent or reduce IRI and microvascular dysfunction, in the current study we proposed an innovative multi-drug therapy using low concentrations of drugs applied intracoronary to reach microvessels in order to stabilize the pro- and antioxidant balance during a MI in an in vivo pig model. The ability of a mixture of doxycycline (1 μM), ML-7 (0.5 μM), and L-NAME (2 μM) to modulate the pro- and antioxidative balance was tested in the left ventricle tissue and blood samples. Data showed that infusion of a MIX reduced the total oxidative status (TOS), oxidative stress index (OSI), and malondialdehyde (MDA). It also increased the total antioxidant capacity, confirming its antioxidative properties. MIX administration also reduced the activity of MMP-2 and MMP-9, and then decreased the release of MLC1 and BNP-26 into plasma. This study demonstrated that intracoronary administration of low concentrations of doxycycline in combination with ML-7 and L-NAME is incredibly efficient in regulating pro- and antioxidant balance during MI.
Collapse
Affiliation(s)
- Iwona Bil-Lula
- Department of Medical Laboratory Diagnostics, Division of Clinical Chemistry and Laboratory Haematology, Wrocław Medical University, 50-556 Wrocław, Poland; (A.K.-Z.); (K.H.)
| | - Wiktor Kuliczkowski
- Institute of Heart Diseases, Wroclaw Medical University, 50-556 Wroclaw, Poland;
| | - Anna Krzywonos-Zawadzka
- Department of Medical Laboratory Diagnostics, Division of Clinical Chemistry and Laboratory Haematology, Wrocław Medical University, 50-556 Wrocław, Poland; (A.K.-Z.); (K.H.)
| | - Piotr Frydrychowski
- Department of Internal Medicine and Clinic of Diseases of Horses, Dogs and Cats, Faculty of Veterinary Medicine, Wrocław University of Environmental and Life Sciences, Grunwaldzki Square 47, 50-366 Wrocław, Poland; (P.F.); (A.N.-N.)
| | - Dominika Stygar
- Department of Physiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Jordana 19 Street, 41-808 Zabrze, Poland;
- SLU University Animal Hospital, Swedish University of Agricultural Sciences, SE-750 07 Uppsala, Sweden
| | - Kornela Hałucha
- Department of Medical Laboratory Diagnostics, Division of Clinical Chemistry and Laboratory Haematology, Wrocław Medical University, 50-556 Wrocław, Poland; (A.K.-Z.); (K.H.)
| | - Agnieszka Noszczyk-Nowak
- Department of Internal Medicine and Clinic of Diseases of Horses, Dogs and Cats, Faculty of Veterinary Medicine, Wrocław University of Environmental and Life Sciences, Grunwaldzki Square 47, 50-366 Wrocław, Poland; (P.F.); (A.N.-N.)
| |
Collapse
|
5
|
Czerwińska K, Januszewska L, Markiewicz-Górka I, Jaremków A, Martynowicz H, Pawlas K, Mazur G, Poręba R, Gać P. Selenoprotein P, peroxiredoxin-5, renalase, and total antioxidant status in patients with suspected obstructive sleep apnea. Sleep Breath 2024; 28:211-219. [PMID: 37495908 PMCID: PMC10954901 DOI: 10.1007/s11325-023-02880-7] [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: 02/08/2023] [Revised: 06/26/2023] [Accepted: 06/27/2023] [Indexed: 07/28/2023]
Abstract
PURPOSE The aim of this study was to investigate the relationship between selenoprotein P, peroxiredoxin-5, renalase, total antioxidant status (TAS), mean blood pressure (mBP), and apnea-hypopnea index (AHI). METHODS The study group consisted of 112 patients hospitalized to verify the diagnosis of obstructive sleep apnea (OSA). The inclusion criteria were consent to participate in the study and age ≥ 18 years. Patients with active proliferative disease, severe systemic diseases, or mental diseases were excluded from the study. Each patient underwent full polysomnography and had blood pressure measured. Blood samples were collected and laboratory test was performed. RESULTS Among 112 patients enrolled, there was a statistically significant negative linear correlation between blood pressure values (sBP, dBP, mBP) and selenoprotein P, renalase, and TAS levels. Similarly, there was a negative linear correlation between AHI and selenoprotein P, renalase, and TAS levels, but none between AHI and peroxiredoxin-5. Based on the obtained regression models, higher selenoprotein P, peroxiredoxin-5, and renalase levels were independently associated with higher TAS. Lower mBP values were independently associated with the use of antihypertensive drugs, higher TAS, and younger age. Male gender, higher BMI, and higher mBP were independently associated with higher AHI. CONCLUSIONS Higher concentrations of selenoprotein P, peroxiredoxin-5, and renalase were associated with higher TAS, which confirms their antioxidant properties. There was an indirect connection between tested antioxidants and blood pressure values.
Collapse
Affiliation(s)
- Karolina Czerwińska
- Division of Environmental Health and Occupational Medicine, Department of Population Health, Wroclaw Medical University, Mikulicza-Radeckiego 7, 50-368, Wroclaw, PL, Poland
| | - Lidia Januszewska
- Division of Environmental Health and Occupational Medicine, Department of Population Health, Wroclaw Medical University, Mikulicza-Radeckiego 7, 50-368, Wroclaw, PL, Poland
| | - Iwona Markiewicz-Górka
- Division of Environmental Health and Occupational Medicine, Department of Population Health, Wroclaw Medical University, Mikulicza-Radeckiego 7, 50-368, Wroclaw, PL, Poland
| | - Aleksandra Jaremków
- Division of Environmental Health and Occupational Medicine, Department of Population Health, Wroclaw Medical University, Mikulicza-Radeckiego 7, 50-368, Wroclaw, PL, Poland
| | - Helena Martynowicz
- Department of Internal and Occupational Diseases, Hypertension and Clinical Oncology, Wroclaw Medical University, Borowska 213, 50-556, Wroclaw, PL, Poland
| | - Krystyna Pawlas
- Division of Environmental Health and Occupational Medicine, Department of Population Health, Wroclaw Medical University, Mikulicza-Radeckiego 7, 50-368, Wroclaw, PL, Poland
| | - Grzegorz Mazur
- Department of Internal and Occupational Diseases, Hypertension and Clinical Oncology, Wroclaw Medical University, Borowska 213, 50-556, Wroclaw, PL, Poland
| | - Rafał Poręba
- Department of Internal and Occupational Diseases, Hypertension and Clinical Oncology, Wroclaw Medical University, Borowska 213, 50-556, Wroclaw, PL, Poland
| | - Paweł Gać
- Division of Environmental Health and Occupational Medicine, Department of Population Health, Wroclaw Medical University, Mikulicza-Radeckiego 7, 50-368, Wroclaw, PL, Poland.
| |
Collapse
|
6
|
Stachowicz A, Sadiq A, Walker B, Sundararaman N, Fert-Bober J. Treatment of human cardiac fibroblasts with the protein arginine deiminase inhibitor BB-Cl-amidine activates the Nrf2/HO-1 signaling pathway. Biomed Pharmacother 2023; 167:115443. [PMID: 37703660 DOI: 10.1016/j.biopha.2023.115443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/27/2023] [Accepted: 09/04/2023] [Indexed: 09/15/2023] Open
Abstract
BACKGROUND Cardiac fibrosis contributes to end-stage extracellular matrix remodeling and heart failure (HF). Cardiac fibroblasts (CFs) differentiate into myofibroblasts (myoFbs) to preserve the structural integrity of the heart; however, the molecular mechanisms regulating CF transdifferentiation remain poorly understood. Protein arginine deiminase (PAD), which converts arginine to citrulline, has been shown to play a role in myocardial infarction, fibrosis, and HF. This study aimed to investigate the role of PAD in CF differentiation to myoFbs and identify the citrullinated proteins that were associated with phenotypic changes in CFs. RESULTS Gene expression analysis showed that PAD1 and PAD2 isoforms, but not PAD4 isoforms, were abundant in both CFs and myoFbs, and PAD1 was significantly upregulated in myoFbs. The pan-PAD inhibitor BB-Cl-amidine (BB-Cl) downregulated the mRNA expression of PAD1 and PAD2 as well as the protein expression of the fibrosis marker COL1A1 in CFs and myoFbs. Interestingly, a proteomic approach pointed to the activation of the Nrf2/HO-1 signaling pathway upon BB-Cl treatment in CFs and myoFbs. BB-Cl administration resulted in the upregulation of HO-1 at both the gene and protein levels in CFs and myoFbs. Importantly, the protein citrullination landscape of CFs consisting of 86 novel citrullination sites associated with focal adhesion (FN1(R1054)), inflammation (TAGLN(R12)) and DNA replication (EEF2(R767)) pathways was identified. CONCLUSIONS In summary, we revealed that BB-Cl treatment resulted in increased HO-1 expression via the Nrf2 pathway, which could prevent excessive tissue damage, thereby leading to substantial clinical benefits for the treatment of cardiac fibrosis.
Collapse
Affiliation(s)
- Aneta Stachowicz
- Chair of Pharmacology, Jagiellonian University Medical College, Krakow, Poland; Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Alia Sadiq
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Brian Walker
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Niveda Sundararaman
- Advanced Clinical Biosystems Research Institute, Precision Biomarker Laboratories, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Justyna Fert-Bober
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Advanced Clinical Biosystems Research Institute, Precision Biomarker Laboratories, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
7
|
Differential plasma protein expression after ingestion of essential amino acid-based dietary supplement verses whey protein in low physical functioning older adults. GeroScience 2023:10.1007/s11357-023-00725-5. [PMID: 36720768 PMCID: PMC10400527 DOI: 10.1007/s11357-023-00725-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 01/02/2023] [Indexed: 02/02/2023] Open
Abstract
In a recent randomized, double-blind, placebo-controlled trial, we were able to demonstrate the superiority of a dietary supplement composed of essential amino acids (EAAs) over whey protein, in older adults with low physical function. In this paper, we describe the comparative plasma protein expression in the same subject groups of EAAs vs whey. The plasma proteomics data was generated using SOMA scan assay. A total of twenty proteins were found to be differentially expressed in both groups with a 1.5-fold change. Notably, five proteins showed a significantly higher fold change expression in the EAA group which included adenylate kinase isoenzyme 1, casein kinase II 2-alpha, Nascent polypeptide-associated complex subunit alpha, peroxiredoxin-1, and peroxiredoxin-6. These five proteins might have played a significant role in providing energy for the improved cardiac and muscle strength of older adults with LPF. On the other hand, fifteen proteins showed slightly lower fold change expression in the EAA group. Some of these 15 proteins regulate metabolism and were found to be associated with inflammation or other comorbidities. Gene Ontology (GO) enrichment analysis showed the association of these proteins with several biological processes. Furthermore, protein-protein interaction network analysis also showed distinct networks between upregulated and downregulated proteins. In conclusion, the important biological roles of the upregulated proteins plus better physical function of participants in the EAAs vs whey group demonstrated that EAAs have the potential to improve muscle strength and physical function in older adults. This study was registered with ClinicalTrials.gov: NCT03424265 "Nutritional interventions in heart failure."
Collapse
|
8
|
Brandt M, Dörschmann H, Khraisat S, Knopp T, Ringen J, Kalinovic S, Garlapati V, Siemer S, Molitor M, Göbel S, Stauber R, Karbach SH, Münzel T, Daiber A, Wenzel P. Telomere Shortening in Hypertensive Heart Disease Depends on Oxidative DNA Damage and Predicts Impaired Recovery of Cardiac Function in Heart Failure. Hypertension 2022; 79:2173-2184. [PMID: 35862118 DOI: 10.1161/hypertensionaha.121.18935] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Heart failure (HF) coincides with cardiomyocyte telomere shortening. Arterial hypertension is the most prominent risk factor for HF. Both HF and arterial hypertension are associated with dysregulation of the neurohormonal axis. How neurohormonal activation is linked to telomere shortening in the pathogenesis of HF is incompletely understood. METHODS Cardiomyocyte telomere length was assessed in a mouse model of hypertensive HF induced by excess neurohormonal activation (AngII [angiotensin II] infusion, high salt diet, and uninephrectomy), in AngII-stimulated cardiomyocytes and in endomyocardial biopsies from patients with HF. Superoxide production, expression of NOX2 (NADPH oxidase 2) and PRDX1 (peroxiredoxin 1) and HDAC6 (histone deacetylase 6) activity were assessed. RESULTS Telomere shortening occurred in vitro and in vivo, correlating with both left ventricular (LV) dilatation and LV systolic function impairment. Telomere shortening coincided with increased superoxide production, increased NOX2 expression, increased HDAC6 activity, loss of the telomere-specific antioxidant PRDX1, and increased oxidative DNA-damage. NOX2 knockout prevented PRDX1 depletion, DNA-damage and telomere shortening confirming this enzyme as a critical source of reactive oxygen species. Cotreatment with the NOX inhibitor apocynin ameliorated hypertensive HF and telomere shortening. Similarly, treatment with the HDAC6 inhibitor tubastatin A, which increases PRDX1 bioavailability, prevented telomere shortening in adult cardiomyocytes. To explore the clinical relevance of our findings, we examined endomyocardial biopsies from an all-comer population of patients with HF with reduced ejection fraction. Here, cardiomyocyte telomere length predicted the recovery of cardiac function. CONCLUSIONS Cardiomyocyte telomere shortening and oxidative damage in heart failure with reduced ejection fraction induced by excess neurohormonal activation depends on NOX2-derived superoxide and may help to stratify HF therapy.
Collapse
Affiliation(s)
- Moritz Brandt
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.).,Center for Thrombosis and Hemostasis' University Medical Center Mainz' Mainz' Germany (M.B., H.D., T.K., J.R., V.G., M.M., S.H.K., T.M., A.D., P.W.).,German Center for Cardiovascular Research (DZHK) - Partner site Rhine-Main (M.B., T.K., J.R., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.)
| | - Hendrik Dörschmann
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.)
| | - Sana'a Khraisat
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.)
| | - Tanja Knopp
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.).,Center for Thrombosis and Hemostasis' University Medical Center Mainz' Mainz' Germany (M.B., H.D., T.K., J.R., V.G., M.M., S.H.K., T.M., A.D., P.W.).,German Center for Cardiovascular Research (DZHK) - Partner site Rhine-Main (M.B., T.K., J.R., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.)
| | - Julia Ringen
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.).,Center for Thrombosis and Hemostasis' University Medical Center Mainz' Mainz' Germany (M.B., H.D., T.K., J.R., V.G., M.M., S.H.K., T.M., A.D., P.W.).,German Center for Cardiovascular Research (DZHK) - Partner site Rhine-Main (M.B., T.K., J.R., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.)
| | - Sanela Kalinovic
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.)
| | - Venkata Garlapati
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.).,Center for Thrombosis and Hemostasis' University Medical Center Mainz' Mainz' Germany (M.B., H.D., T.K., J.R., V.G., M.M., S.H.K., T.M., A.D., P.W.).,German Center for Cardiovascular Research (DZHK) - Partner site Rhine-Main (M.B., T.K., J.R., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.)
| | - Svenja Siemer
- Department of Otolaryngology, Head and Neck Surgery, University Medical Center Mainz' Mainz' Germany (S.S., R.S.)
| | - Michael Molitor
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.).,Center for Thrombosis and Hemostasis' University Medical Center Mainz' Mainz' Germany (M.B., H.D., T.K., J.R., V.G., M.M., S.H.K., T.M., A.D., P.W.).,German Center for Cardiovascular Research (DZHK) - Partner site Rhine-Main (M.B., T.K., J.R., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.)
| | - Sebastian Göbel
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.)
| | - Roland Stauber
- Department of Otolaryngology, Head and Neck Surgery, University Medical Center Mainz' Mainz' Germany (S.S., R.S.)
| | - Susanne Helena Karbach
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.).,Center for Thrombosis and Hemostasis' University Medical Center Mainz' Mainz' Germany (M.B., H.D., T.K., J.R., V.G., M.M., S.H.K., T.M., A.D., P.W.).,German Center for Cardiovascular Research (DZHK) - Partner site Rhine-Main (M.B., T.K., J.R., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.)
| | - Thomas Münzel
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.).,Center for Thrombosis and Hemostasis' University Medical Center Mainz' Mainz' Germany (M.B., H.D., T.K., J.R., V.G., M.M., S.H.K., T.M., A.D., P.W.).,German Center for Cardiovascular Research (DZHK) - Partner site Rhine-Main (M.B., T.K., J.R., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.)
| | - Andreas Daiber
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.).,Center for Thrombosis and Hemostasis' University Medical Center Mainz' Mainz' Germany (M.B., H.D., T.K., J.R., V.G., M.M., S.H.K., T.M., A.D., P.W.).,German Center for Cardiovascular Research (DZHK) - Partner site Rhine-Main (M.B., T.K., J.R., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.)
| | - Philip Wenzel
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.).,Center for Thrombosis and Hemostasis' University Medical Center Mainz' Mainz' Germany (M.B., H.D., T.K., J.R., V.G., M.M., S.H.K., T.M., A.D., P.W.).,German Center for Cardiovascular Research (DZHK) - Partner site Rhine-Main (M.B., T.K., J.R., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.).,Department of Biochemistry, Cardiovascular Research Institute Maastricht School for Cardiovascular Diseases (CARIM), Maastricht University, Maastricht, the Netherlands (P.W.)
| |
Collapse
|
9
|
Mitophagy: A Potential Target for Pressure Overload-Induced Cardiac Remodelling. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2849985. [PMID: 36204518 PMCID: PMC9532135 DOI: 10.1155/2022/2849985] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 07/16/2022] [Accepted: 08/11/2022] [Indexed: 11/18/2022]
Abstract
The pathological mechanisms underlying cardiac remodelling and cardiac dysfunction caused by pressure overload are poorly understood. Mitochondrial damage and functional dysfunction, including mitochondrial bioenergetic disorder, oxidative stress, and mtDNA damage, contribute to heart injury caused by pressure overload. Mitophagy, an important regulator of mitochondrial homeostasis and function, is triggered by mitochondrial damage and participates in the pathological process of cardiovascular diseases. Recent studies indicate that mitophagy plays a critical role in the pressure overload model, but evidence on the causal relationship between mitophagy abnormality and pressure overload-induced heart injury is inconclusive. This review summarises the mechanism, role, and regulation of mitophagy in the pressure overload model. It also pays special attention to active compounds that may regulate mitophagy in pressure overload, which provide clues for possible clinical applications.
Collapse
|
10
|
Koniari I, Velissaris D, Kounis NG, Koufou E, Artopoulou E, de Gregorio C, Mplani V, Paraskevas T, Tsigkas G, Hung MY, Plotas P, Lambadiari V, Ikonomidis I. Anti-Diabetic Therapy, Heart Failure and Oxidative Stress: An Update. J Clin Med 2022; 11:4660. [PMID: 36012897 PMCID: PMC9409680 DOI: 10.3390/jcm11164660] [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: 06/23/2022] [Revised: 07/31/2022] [Accepted: 08/01/2022] [Indexed: 11/17/2022] Open
Abstract
Diabetes mellitus (DM) and heart failure (HF) are two chronic disorders that affect millions worldwide. Hyperglycemia can induce excessive generation of highly reactive free radicals that promote oxidative stress and further exacerbate diabetes progression and its complications. Vascular dysfunction and damage to cellular proteins, membrane lipids and nucleic acids can stem from overproduction and/or insufficient removal of free radicals. The aim of this article is to review the literature regarding the use of antidiabetic drugs and their role in glycemic control in patients with heart failure and oxidative stress. Metformin exerts a minor benefit to these patients. Thiazolidinediones are not recommended in diabetic patients, as they increase the risk of HF. There is a lack of robust evidence on the use of meglinitides and acarbose. Insulin and dipeptidyl peptidase-4 (DPP-4) inhibitors may have a neutral cardiovascular effect on diabetic patients. The majority of current research focuses on sodium glucose cotransporter 2 (SGLT2) inhibitors and glucagon-like peptide 1 (GLP-1) receptor agonists. SGLT2 inhibitors induce positive cardiovascular effects in diabetic patients, leading to a reduction in cardiovascular mortality and HF hospitalization. GLP-1 receptor agonists may also be used in HF patients, but in the case of chronic kidney disease, SLGT2 inhibitors should be preferred.
Collapse
Affiliation(s)
- Ioanna Koniari
- Department of Cardiology, University Hospital of South Manchester NHS Foundation Trust, Manchester M23 9LT, UK
| | - Dimitrios Velissaris
- Department of Internal Medicine, University Hospital of Patras, 26500 Patras, Greece
| | - Nicholas G. Kounis
- Department of Cardiology, University Hospital of Patras, 26500 Patras, Greece
| | - Eleni Koufou
- Department of Cardiology, University Hospital of Patras, 26500 Patras, Greece
| | - Eleni Artopoulou
- Department of Internal Medicine, University Hospital of Patras, 26500 Patras, Greece
| | - Cesare de Gregorio
- Department of Clinical and Experimental Medicine, University of Messina Medical School, 98122 Messina, Italy
| | - Virginia Mplani
- Intensive Care Unit, Patras University Hospital, 26500 Patras, Greece
| | | | - Grigorios Tsigkas
- Department of Cardiology, University Hospital of Patras, 26500 Patras, Greece
| | - Ming-Yow Hung
- Division of Cardiology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei 11031, Taiwan
| | - Panagiotis Plotas
- Laboratory Primary Health Care, School of Health Rehabilitation Sciences, University of Patras, 26504 Patras, Greece
| | - Vaia Lambadiari
- Second Department of Internal Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Ignatios Ikonomidis
- Second Cardiology Department, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| |
Collapse
|
11
|
Hao W, Li M, Cai Q, Wu S, Li X, He Q, Hu Y. Roles of NRF2 in Fibrotic Diseases: From Mechanisms to Therapeutic Approaches. Front Physiol 2022; 13:889792. [PMID: 35721561 PMCID: PMC9203969 DOI: 10.3389/fphys.2022.889792] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/29/2022] [Indexed: 11/24/2022] Open
Abstract
Fibrosis is a persistent inflammatory response that causes scarring and tissue sclerosis by stimulating myofibroblasts to create significant quantities of extracellular matrix protein deposits in the tissue. Oxidative stress has also been linked to the development of fibrosis in several studies. The nuclear erythroid 2-related factor 2 (NRF2) transcription factor controls the expression of several detoxification and antioxidant genes. By binding to antioxidant response elements, NRF2 is activated by oxidative or electrophilic stress and promotes its target genes, resulting in a protective effect on cells. NRF2 is essential for cell survival under oxidative stress conditions. This review describes Kelch-like epichlorohydrin-associated protein 1 (KEAP1)/NRF2 signaling mechanisms and presents recent research advances regarding NRF2 and its involvement in primary fibrotic lesions such as pulmonary fibrosis, hepatic fibrosis, myocardial fibrosis, and renal fibrosis. The related antioxidant substances and drugs are described, along with the mechanisms by which KEAP1/NRF2 regulation positively affects the therapeutic response. Finally, the therapeutic prospects and potential value of NRF2 in fibrosis are summarized. Further studies on NRF2 may provide novel therapeutic approaches for fibrosis.
Collapse
Affiliation(s)
- Wenlong Hao
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Minghao Li
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Qingmin Cai
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Shiying Wu
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Xiangyao Li
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Quanyu He
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Yongbin Hu
- Department of Pathology, Basic Medical School, Central South University, Changsha, China
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Yongbin Hu,
| |
Collapse
|
12
|
Liu J, Li W, Deng KQ, Tian S, Liu H, Shi H, Fang Q, Liu Z, Chen Z, Tian T, Gan S, Hu F, Hu M, Cheng X, Ji YX, Zhang P, She ZG, Zhang XJ, Chen S, Cai J, Li H. The E3 Ligase TRIM16 Is a Key Suppressor of Pathological Cardiac Hypertrophy. Circ Res 2022; 130:1586-1600. [PMID: 35437018 DOI: 10.1161/circresaha.121.318866] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 04/06/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Pathological cardiac hypertrophy is one of the leading causes of heart failure with highly complicated pathogeneses. The E3 ligase TRIM16 (tripartite motif-containing protein 16) has been recognized as a pivotal regulator to control cell survival, immune response, and oxidativestress. However, the role of Trim16 in cardiac hypertrophy is unknown. METHODS We generated cardiac-specific knockout mice and adeno-associated virus serotype 9-Trim16 mice to evaluate the function of Trim16 in pathological myocardial hypertrophy. The direct effect of TRIM16 on cardiomyocyte enlargement was examined using an adenovirus system. Furthermore, we combined RNA-sequencing and interactome analysis that was followed by multiple molecular biological methodologies to identify the direct target and corresponding molecular events contributing to TRIM16 function. RESULTS We found an intimate correlation of Trim16 expression with hypertrophy-related heart failure in both human and mouse. Our functional investigations and unbiased transcriptomic analyses clearly demonstrated that Trim16 deficiency markedly exacerbated cardiomyocyte enlargement in vitro and in transverse aortic constriction-induced cardiac hypertrophy mouse model, whereas Trim16 overexpression attenuated cardiac hypertrophy and remodeling. Mechanistically, Prdx1 (peroxiredoxin 1) is an essential target of Trim16 in cardiac hypertrophy. We found that Trim16 interacts with Prdx1 and inhibits its phosphorylation, leading to a robust enhancement of its downstream Nrf2 (nuclear factor-erythroid 2-related factor 2) pathway to block cardiac hypertrophy. Trim16-blocked Prdx1 phosphorylation was largely dependent on a direct interaction between Trim16 and Src and the resultant Src ubiquitinational degradation. Notably, Prdx1 knockdown largely abolished the anti-hypertrophic effects of Trim16 overexpression. CONCLUSIONS Our findings provide the first evidence supporting Trim16 as a novel suppressor of pathological cardiac hypertrophy and indicate that targeting the Trim16-Prdx1 axis represents a promising therapeutic strategy for hypertrophy-related heart failure.
Collapse
Affiliation(s)
- Jiayi Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (J.L., W.L., T.T., Z.-G.S., H.Li.)
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li)
| | - Wei Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (J.L., W.L., T.T., Z.-G.S., H.Li.)
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li)
| | - Ke-Qiong Deng
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li)
- Department of Cardiology (K.-Q.D., Z.C.), Zhongnan Hospital of Wuhan University, China
| | - Song Tian
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li)
| | - Hui Liu
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li)
- Gannan Innovation and Translational Medicine Research Institute (H. Liu, M.H., X.C.), Gannan Medical University, Ganzhou, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education (H. Liu, M.H., X.C.), Gannan Medical University, Ganzhou, China
| | - Hongjie Shi
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li)
- School of Basic Medical Sciences, Wuhan University, China (H.S., S.G., Y.-X.J., P.Z., X.-J.Z., H. Li)
| | - Qian Fang
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li)
| | - Zhen Liu
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li)
| | - Ze Chen
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li)
- Department of Cardiology (K.-Q.D., Z.C.), Zhongnan Hospital of Wuhan University, China
| | - Tian Tian
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (J.L., W.L., T.T., Z.-G.S., H.Li.)
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li)
| | - Shanyu Gan
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li)
- School of Basic Medical Sciences, Wuhan University, China (H.S., S.G., Y.-X.J., P.Z., X.-J.Z., H. Li)
| | - Fengjiao Hu
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li)
- Medical Science Research Center (F.H., H. Li), Zhongnan Hospital of Wuhan University, China
| | - Manli Hu
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li)
- Gannan Innovation and Translational Medicine Research Institute (H. Liu, M.H., X.C.), Gannan Medical University, Ganzhou, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education (H. Liu, M.H., X.C.), Gannan Medical University, Ganzhou, China
| | - Xu Cheng
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li)
- Gannan Innovation and Translational Medicine Research Institute (H. Liu, M.H., X.C.), Gannan Medical University, Ganzhou, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education (H. Liu, M.H., X.C.), Gannan Medical University, Ganzhou, China
| | - Yan-Xiao Ji
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li)
- School of Basic Medical Sciences, Wuhan University, China (H.S., S.G., Y.-X.J., P.Z., X.-J.Z., H. Li)
| | - Peng Zhang
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li)
- School of Basic Medical Sciences, Wuhan University, China (H.S., S.G., Y.-X.J., P.Z., X.-J.Z., H. Li)
| | - Zhi-Gang She
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (J.L., W.L., T.T., Z.-G.S., H.Li.)
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li)
| | - Xiao-Jing Zhang
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li)
- School of Basic Medical Sciences, Wuhan University, China (H.S., S.G., Y.-X.J., P.Z., X.-J.Z., H. Li)
| | - Shaoze Chen
- Department of Cardiology, Huanggang Central Hospital, China (S.C.)
- Huanggang Institute of Translational Medicine, Huanggang, China (S.C.)
| | - Jingjing Cai
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, China (J.C.)
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (J.L., W.L., T.T., Z.-G.S., H.Li.)
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li)
- Medical Science Research Center (F.H., H. Li), Zhongnan Hospital of Wuhan University, China
- School of Basic Medical Sciences, Wuhan University, China (H.S., S.G., Y.-X.J., P.Z., X.-J.Z., H. Li)
| |
Collapse
|
13
|
Shi X, Dorsey A, Qiu H. New Progress in the Molecular Regulations and Therapeutic Applications in Cardiac Oxidative Damage Caused by Pressure Overload. Antioxidants (Basel) 2022; 11:antiox11050877. [PMID: 35624741 PMCID: PMC9137593 DOI: 10.3390/antiox11050877] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/25/2022] [Accepted: 04/26/2022] [Indexed: 12/11/2022] Open
Abstract
Chronic pressure overload is a key risk factor for mortality due to its subsequent development of heart failure, in which the underlying molecular mechanisms remain vastly undetermined. In this review, we updated the latest advancements for investigating the role and relevant mechanisms of oxidative stress involved in the pathogenesis of pressure-overload-induced cardiomyopathy and cardiac dysfunction, focusing on significant biological sources of reactive oxygen species (free radical) production, antioxidant defenses, and their association with the cardiac metabolic remodeling in the stressed heart. We also summarize the newly developed preclinical therapeutic approaches in animal models for pressure-overload-induced myocardial damage. This review aims to enhance the current understanding of the mechanisms of chronic hypertensive heart failure and potentially improve the development of better therapeutic strategies for the associated diseases.
Collapse
Affiliation(s)
| | | | - Hongyu Qiu
- Correspondence: ; Tel.: +1-404-413-3371; Fax: +1-404-413-9566
| |
Collapse
|
14
|
Methylene blue prevents osteoarthritis progression and relieves pain in rats via upregulation of Nrf2/PRDX1. Acta Pharmacol Sin 2022; 43:417-428. [PMID: 33833406 DOI: 10.1038/s41401-021-00646-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 02/04/2021] [Indexed: 02/06/2023]
Abstract
Oxidative stress-related cartilage degeneration, synovitis, and joint pain play vital roles in the progress of osteoarthritis (OA). Anti-oxidative stress agents not only prevent structural damage progression but also relieve OA-related pain. In this study, we investigated the therapeutic effect of methylene blue (MB), a classical and important anti-oxidant with strong neural affinity. Experimental OA was established in rats by radial transection of medial collateral ligament and medial meniscus (MCLT + MMT) of the right knee joint. The OA rats received intra-articular injection of MB (1 mg/kg) every week starting one week after surgery. We showed that MB administration exerted significant cartilage protection, synovitis inhibition as well as pain relief in OA rats. In human chondrocytes and fibroblast-like synoviocytes, MB significantly attenuated tert-butyl hydroperoxide (TBHP)-induced inflammatory response and oxidative stress. We demonstrated that these effects of MB resulted from dual targets of important antioxidant enzymes, Nrf2 and PRDX1, which also mutually reinforcing and participated in an interaction. Furthermore, we found that calcitonin gene-related peptide (CGRP), a neural inflammatory mediator, was accumulated around the vessel in synovium and subchondral bone in OA rats and in TBHP-treated primary cortical neurons; MB administration significantly inhibited CGRP expression through upregulation of Nrf2 and PRDX1. Taken together, these results suggest that MB ameliorates oxidative stress via Nrf2/PRDX1 regulation to prevent progression and relieve pain of OA.
Collapse
|
15
|
Peroxiredoxin-5 Knockdown Accelerates Pressure Overload-Induced Cardiac Hypertrophy in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5067544. [PMID: 35132351 PMCID: PMC8817848 DOI: 10.1155/2022/5067544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 12/21/2021] [Indexed: 11/29/2022]
Abstract
A recent study showed that peroxiredoxins (Prxs) play an important role in the development of pathological cardiac hypertrophy. However, the involvement of Prx5 in cardiac hypertrophy remains unclear. Therefore, this study is aimed at investigating the role and mechanisms of Prx5 in pathological cardiac hypertrophy and dysfunction. Transverse aortic constriction (TAC) surgery was performed to establish a pressure overload-induced cardiac hypertrophy model. In this study, we found that Prx5 expression was upregulated in hypertrophic hearts and cardiomyocytes. In addition, Prx5 knockdown accelerated pressure overload-induced cardiac hypertrophy and dysfunction in mice by activating oxidative stress and cardiomyocyte apoptosis. Importantly, heart deterioration caused by Prx5 knockdown was related to mitogen-activated protein kinase (MAPK) pathway activation. These findings suggest that Prx5 could be a novel target for treating cardiac hypertrophy and heart failure.
Collapse
|
16
|
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: 32] [Impact Index Per Article: 8.0] [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
|
17
|
Andreadou I, Efentakis P, Frenis K, Daiber A, Schulz R. Thiol-based redox-active proteins as cardioprotective therapeutic agents in cardiovascular diseases. Basic Res Cardiol 2021; 116:44. [PMID: 34275052 DOI: 10.1007/s00395-021-00885-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 06/17/2021] [Indexed: 12/12/2022]
Abstract
Thiol-based redox compounds, namely thioredoxins (Trxs), glutaredoxins (Grxs) and peroxiredoxins (Prxs), stand as a pivotal group of proteins involved in antioxidant processes and redox signaling. Glutaredoxins (Grxs) are considered as one of the major families of proteins involved in redox regulation by removal of S-glutathionylation and thereby reactivation of other enzymes with thiol-dependent activity. Grxs are also coupled to Trxs and Prxs recycling and thereby indirectly contribute to reactive oxygen species (ROS) detoxification. Peroxiredoxins (Prxs) are a ubiquitous family of peroxidases, which play an essential role in the detoxification of hydrogen peroxide, aliphatic and aromatic hydroperoxides, and peroxynitrite. The Trxs, Grxs and Prxs systems, which reversibly induce thiol modifications, regulate redox signaling involved in various biological events in the cardiovascular system. This review focuses on the current knowledge of the role of Trxs, Grxs and Prxs on cardiovascular pathologies and especially in cardiac hypertrophy, ischemia/reperfusion (I/R) injury and heart failure as well as in the presence of cardiovascular risk factors, such as hypertension, hyperlipidemia, hyperglycemia and metabolic syndrome. Further studies on the roles of thiol-dependent redox systems in the cardiovascular system will support the development of novel protective and therapeutic strategies against cardiovascular diseases.
Collapse
Affiliation(s)
- Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece.
| | - Panagiotis Efentakis
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Katie Frenis
- Department of Cardiology 1, Molecular Cardiology, University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Andreas Daiber
- Department of Cardiology 1, Molecular Cardiology, University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131, Mainz, Germany.,Partner Site Rhine-Main, German Center for Cardiovascular Research (DZHK), Langenbeckstr 1, 55131, Mainz, Germany
| | - Rainer Schulz
- Institute of Physiology, Justus Liebig University Giessen, Giessen, Germany.
| |
Collapse
|
18
|
Ramachandra CJA, Cong S, Chan X, Yap EP, Yu F, Hausenloy DJ. Oxidative stress in cardiac hypertrophy: From molecular mechanisms to novel therapeutic targets. Free Radic Biol Med 2021; 166:297-312. [PMID: 33675957 DOI: 10.1016/j.freeradbiomed.2021.02.040] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 02/11/2021] [Accepted: 02/26/2021] [Indexed: 02/06/2023]
Abstract
When faced with increased workload the heart undergoes remodelling, where it increases its muscle mass in an attempt to preserve normal function. This is referred to as cardiac hypertrophy and if sustained, can lead to impaired contractile function. Experimental evidence supports oxidative stress as a critical inducer of both genetic and acquired forms of cardiac hypertrophy, a finding which is reinforced by elevated levels of circulating oxidative stress markers in patients with cardiac hypertrophy. These observations formed the basis for using antioxidants as a therapeutic means to attenuate cardiac hypertrophy and improve clinical outcomes. However, the use of antioxidant therapies in the clinical setting has been associated with inconsistent results, despite antioxidants having been shown to exert protection in several animal models of cardiac hypertrophy. This has forced us to revaluate the mechanisms, both upstream and downstream of oxidative stress, where recent studies demonstrate that apart from conventional mediators of oxidative stress, metabolic disturbances, mitochondrial dysfunction and inflammation as well as dysregulated autophagy and protein homeostasis contribute to disease pathophysiology through mechanisms involving oxidative stress. Importantly, novel therapeutic targets have been identified to counteract oxidative stress and attenuate cardiac hypertrophy but more interestingly, the repurposing of drugs commonly used to treat metabolic disorders, hypertension, peripheral vascular disease, sleep disorders and arthritis have also been shown to improve cardiac function through suppression of oxidative stress. Here, we review the latest literature on these novel mechanisms and intervention strategies with the aim of better understanding the complexities of oxidative stress for more precise targeted therapeutic approaches to prevent cardiac hypertrophy.
Collapse
Affiliation(s)
- Chrishan J A Ramachandra
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore; Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore.
| | - Shuo Cong
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore; Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore; Yong Loo Lin School of Medicine, National University Singapore, Singapore
| | - Xavier Chan
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore; Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore; Faculty of Science, National University of Singapore, Singapore
| | - En Ping Yap
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore; Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore; Yong Loo Lin School of Medicine, National University Singapore, Singapore
| | - Fan Yu
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore; Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore; Yong Loo Lin School of Medicine, National University Singapore, Singapore
| | - Derek J Hausenloy
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore; Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore; Yong Loo Lin School of Medicine, National University Singapore, Singapore; The Hatter Cardiovascular Institute, University College London, London, UK; Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan
| |
Collapse
|
19
|
Palmatine Protects against Cerebral Ischemia/Reperfusion Injury by Activation of the AMPK/Nrf2 Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6660193. [PMID: 33777318 PMCID: PMC7981182 DOI: 10.1155/2021/6660193] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 02/05/2021] [Accepted: 02/21/2021] [Indexed: 02/07/2023]
Abstract
Palmatine (PAL), a natural isoquinoline alkaloid, possesses extensive biological and pharmaceutical activities, including antioxidative stress, anti-inflammatory, antitumor, neuroprotective, and gastroprotective activities. However, it is unknown whether PAL has a protective effect against ischemic stroke and cerebral ischemia/reperfusion (I/R) injury. In the present study, a transient middle cerebral artery occlusion (MCAO) mouse model was used to mimic ischemic stroke and cerebral I/R injury in mice. Our study demonstrated that PAL treatment ameliorated cerebral I/R injury by decreasing infarct volume, neurological scores, and brain water content. PAL administration attenuated oxidative stress, the inflammatory response, and neuronal apoptosis in mice after cerebral I/R injury. In addition, PAL treatment also decreases hypoxia and reperfusion- (H/R-) induced neuronal injury by reducing oxidative stress, the inflammatory response, and neuronal apoptosis. Moreover, the neuroprotective effects of PAL were associated with the activation of the AMP-activated protein kinase (AMPK)/nuclear factor E2-related factor 2 (Nrf2) pathway, and Nrf2 knockdown offsets PAL-mediated antioxidative stress and anti-inflammatory effects. Therefore, our results suggest that PAL may be a novel treatment strategy for ischemic stroke and cerebral I/R injury.
Collapse
|
20
|
1-O-Hexyl-2,3,5-Trimethylhydroquinone Ameliorates the Development of Preeclampsia through Suppression of Oxidative Stress and Endothelial Cell Apoptosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8839394. [PMID: 33542786 PMCID: PMC7840260 DOI: 10.1155/2021/8839394] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 12/26/2020] [Accepted: 01/06/2021] [Indexed: 12/14/2022]
Abstract
1-O-Hexyl-2,3,5-trimethylhydroquinone (HTHQ), a potent nuclear factor-E2-related factor 2 (Nrf2) activator, has potent antioxidant activity by scavenging reactive oxygen species (ROS). However, the role of HTHQ on the development of preeclampsia (PE) and the underlying mechanisms have barely been explored. In the present study, PE model was induced by adenovirus-mediated overexpression of soluble fms-like tyrosine kinase 1 (sFlt-1) in pregnant mice. The results showed that HTHQ treatment significantly relieved the high systolic blood pressure (SBP) and proteinuria and increased the fetal weight and fetal weight/placenta weight in preeclamptic mice. Furthermore, we found that HTHQ treatment significantly decreased soluble endoglin (sEng), endothelin-1 (ET-1), and activin A and restored vascular endothelial growth factor (VEGF) in preeclamptic mice. In addition, HTHQ treatment inhibited oxidative stress and endothelial cell apoptosis by increasing the levels of Nrf2 and its downstream haemoxygenase-1 (HO-1) protein. In line with the data in vivo, we discovered that HTHQ treatment attenuated oxidative stress and cell apoptosis in human umbilical vein endothelial cells (HUVECs) following hypoxia and reperfusion (H/R), and the HTHQ-mediated protection was lost after transfected with siNrf2. In conclusion, these results suggested that HTHQ ameliorates the development of preeclampsia through suppression of oxidative stress and endothelial cell apoptosis.
Collapse
|