1
|
Doul J, Minaříková M, Charvátová Z, Maxová H. Nitric oxide is involved in the cardioprotection of neonatal rat hearts, but not in neonatal ischemic postconditioning. Physiol Rep 2024; 12:e16147. [PMID: 39097984 PMCID: PMC11298247 DOI: 10.14814/phy2.16147] [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: 02/17/2024] [Revised: 06/29/2024] [Accepted: 07/04/2024] [Indexed: 08/06/2024] Open
Abstract
The cardioprotective effect of ischemic preconditioning (IPC) and ischemic postconditioning (IPoC) in adult hearts is mediated by nitric oxide (NO). During the early developmental period, rat hearts exhibit higher resistance to ischemia-reperfusion (I/R) injury, contain higher levels of serum nitrates, and their resistance cannot be further increased by IPC or IPoC. NOS blocker (L-NAME) lowers their high resistance. Wistar rat hearts (postnatal Days 1 and 10) were perfused according to Langendorff and exposed to 40 min of global ischemia followed by reperfusion with or without IPoC. NO and reactive oxygen species donors (DEA-NONO, SIN-1) and L-NAME were administered. Tolerance to ischemia decreased between Days 1 and 10. DEA-NONO (low concentrations) significantly increased tolerance to I/R injury on both Days 1 and 10. SIN-1 increased tolerance to I/R injury on Day 10, but not on Day 1. L-NAME significantly reduced resistance to I/R injury on Day 1, but actually increased resistance to I/R injury on Day 10. Cardioprotection by IPoC on Day 10 was not affected by either NO donors or L-NAME. It can be concluded that resistance of the neonatal heart to I/R injury is NO dependent, but unlike in adult hearts, cardioprotective interventions, such as IPoC, are most likely NO independent.
Collapse
Affiliation(s)
- Jan Doul
- Department of Pathophysiology, Second Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Marcela Minaříková
- Department of Physiology, Second Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Zuzana Charvátová
- Department of Pathophysiology, Second Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Hana Maxová
- Department of Pathophysiology, Second Faculty of MedicineCharles UniversityPragueCzech Republic
- Center for Experimental MedicineInstitute for Clinical and Experimental MedicinePragueCzech Republic
| |
Collapse
|
2
|
Vaziri Z, Saleki K, Aram C, Alijanizadeh P, Pourahmad R, Azadmehr A, Ziaei N. Empagliflozin treatment of cardiotoxicity: A comprehensive review of clinical, immunobiological, neuroimmune, and therapeutic implications. Biomed Pharmacother 2023; 168:115686. [PMID: 37839109 DOI: 10.1016/j.biopha.2023.115686] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 10/03/2023] [Accepted: 10/08/2023] [Indexed: 10/17/2023] Open
Abstract
Cancer and cardiovascular disorders are known as the two main leading causes of mortality worldwide. Cardiotoxicity is a critical and common adverse effect of cancer-related chemotherapy. Chemotherapy-induced cardiotoxicity has been associated with various cancer treatments, such as anthracyclines, immune checkpoint inhibitors, and kinase inhibitors. Different methods have been reported for the management of chemotherapy-induced cardiotoxicity. In this regard, sodium-glucose cotransporter-2 inhibitors (SGLT2i), a class of antidiabetic agents, have recently been applied to manage heart failure patients. Further, SGLT2i drugs such as EMPA exert protective cardiac and systemic effects. Moreover, it can reduce inflammation through the mediation of major inflammatory components, such as Nucleotide-binding domain-like receptor protein 3 (NLRP3) inflammasomes, Adenosine 5'-monophosphate-activated protein kinase (AMPK), and c-Jun N-terminal kinase (JNK) pathways, Signal transducer and activator of transcription (STAT), and overall decreasing transcription of proinflammatory cytokines. The clinical outcome of EMPA administration is related to improving cardiovascular risk factors, including body weight, lipid profile, blood pressure, and arterial stiffness. Intriguingly, SGLT2 suppressors can regulate microglia-driven hyperinflammation affecting neurological and cardiovascular disorders. In this review, we discuss the protective effects of EMPA in chemotherapy-induced cardiotoxicity from molecular, immunological, and neuroimmunological aspects to preclinical and clinical outcomes.
Collapse
Affiliation(s)
- Zahra Vaziri
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; USERN Office, Babol University of Medical Sciences, Babol, Iran
| | - Kiarash Saleki
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; USERN Office, Babol University of Medical Sciences, Babol, Iran; Department of e-Learning, Virtual School of Medical Education and Management, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Cena Aram
- Department of Cell & Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Parsa Alijanizadeh
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; USERN Office, Babol University of Medical Sciences, Babol, Iran
| | - Ramtin Pourahmad
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Azadmehr
- Immunology Department, Babol University of Medical Sciences, Babol, Iran
| | - Naghmeh Ziaei
- Clinical Research Development unit of Rouhani Hospital, Babol University of Medical Sciences, Babol, Iran; Department of Cardiology, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
3
|
Tang K, Zhang H, Deng J, Wang D, Liu S, Lu S, Cui Q, Chen C, Liu J, Yang Z, Li Y, Chen J, Lv J, Ma J, Huang B. Ammonia detoxification promotes CD8 + T cell memory development by urea and citrulline cycles. Nat Immunol 2023; 24:162-173. [PMID: 36471170 DOI: 10.1038/s41590-022-01365-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 10/17/2022] [Indexed: 12/12/2022]
Abstract
Amino acid metabolism is essential for cell survival, while the byproduct ammonia is toxic and can injure cellular longevity. Here we show that CD8+ memory T (TM) cells mobilize the carbamoyl phosphate (CP) metabolic pathway to clear ammonia, thus promoting memory development. CD8+ TM cells use β-hydroxybutyrylation to upregulate CP synthetase 1 and trigger the CP metabolic cascade to form arginine in the cytosol. This cytosolic arginine is then translocated into the mitochondria where it is split by arginase 2 to urea and ornithine. Cytosolic arginine is also converted to nitric oxide and citrulline by nitric oxide synthases. Thus, both the urea and citrulline cycles are employed by CD8+ T cells to clear ammonia and enable memory development. This ammonia clearance machinery might be targeted to improve T cell-based cancer immunotherapies.
Collapse
Affiliation(s)
- Ke Tang
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Cell Architecture Research Center, Huazhong University of Science and Technology, Wuhan, China
| | - Huafeng Zhang
- Department of Pathology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinghui Deng
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dianheng Wang
- Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing, China
| | - Shichuan Liu
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuya Lu
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingfa Cui
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chen Chen
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jincheng Liu
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhuoshun Yang
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yonggang Li
- Hubei Provincial Key Laboratory for Applied Toxicology, Hubei Provincial Center for Disease Control and Prevention, Wuhan, China
| | - Jie Chen
- Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing, China
| | - Jiadi Lv
- Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing, China
| | - Jingwei Ma
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Huang
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. .,Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing, China.
| |
Collapse
|
4
|
Maslov LN, Naryzhnaya NV, Sementsov AS, Derkachev IA, Gusakova SV, Sarybaev A. Role of Nitric Oxide Synthase in the Infarct-Limiting Effect of Normobaric Hypoxia. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022040202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
5
|
Chen X, Zou Z, Wang Q, Gao W, Zeng S, Ye S, Xu P, Huang M, Li K, Chen J, Zhong Z, Zhang Q, Hao B, Liu Q. Inhibition of NOS1 promotes the interferon response of melanoma cells. J Transl Med 2022; 20:205. [PMID: 35538490 PMCID: PMC9092760 DOI: 10.1186/s12967-022-03403-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 04/22/2022] [Indexed: 02/07/2023] Open
Abstract
Background NOS1 expression predicts poor prognosis in patients with melanoma. However, the molecular function of NOS1 in the type I IFN response and immune escape of melanoma is still unknown. Methods The CRISPR/Cas9 system was used to generate NOS1-knockout melanoma cells and the biological characteristics of NOS1-knockout cells were evaluated by MTT assay, clonogenic assay, EdU assay, and flow cytometric assay. The effect on tumor growth was tested in BALB/c-nu and C57BL/6 mouse models. The gene expression profiles were detected with Affymetrix microarray and RNA-seq and KEGG (Kyoto Encyclopedia of Genes and Genomes) and CLUE GO analysis was done. The clinical data and transcriptional profiles of melanoma patients from the public database TCGA (The Cancer Genome Atlas) and GEO (Gene Expression Omnibus, GSE32611) were analyzed by Qlucore Omics Explorer. Results NOS1 deletion suppressed the proliferation of melanoma A375 cells in culture, blocked cell cycling at the G0/G1 phase, and decreased the tumor growth in lung metastasis nodes in a B16 melanoma xenograft mouse model. Moreover, NOS1 knockout increased the infiltration of CD3+ immune cells in tumors. The transcriptomics analysis identified 2203 differential expression genes (DEGs) after NOS1 deletion. These DEGs indicated that NOS1 deletion downregulated mostly metabolic functions but upregulated immune response pathways. After inhibiting with NOS1 inhibitor N-PLA, melanoma cells significantly increased the response to IFN\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\upalpha $$\end{document}α by upregulation expression of IFN\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\upalpha $$\end{document}α simulation genes (ISGs), especially the components in innate immune signaling, JAK-STAT, and TOLL-LIKE pathway. Furthermore, these NOS1-regulating immune genes (NOS1-ISGs) worked as a signature to predict poor overall survival and lower response to chemotherapy in melanoma patients. Conclusion These findings provided a transcriptional evidence of NOS1 promotion on tumor growth, which is correlated with metabolic regulation and immune escape in melanoma cells. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03403-w.
Collapse
Affiliation(s)
- Xi Chen
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Science, Southern Medical University, Shatai South Road, Baiyun District, 16, Guangzhou, 510515, China
| | - Zhiwei Zou
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Science, Southern Medical University, Shatai South Road, Baiyun District, 16, Guangzhou, 510515, China
| | - Qianli Wang
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Science, Southern Medical University, Shatai South Road, Baiyun District, 16, Guangzhou, 510515, China
| | - Wenwen Gao
- First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450001, China
| | - Sisi Zeng
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Science, Southern Medical University, Shatai South Road, Baiyun District, 16, Guangzhou, 510515, China
| | - Shuangyan Ye
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Science, Southern Medical University, Shatai South Road, Baiyun District, 16, Guangzhou, 510515, China
| | - Pengfei Xu
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Science, Southern Medical University, Shatai South Road, Baiyun District, 16, Guangzhou, 510515, China
| | - Mengqiu Huang
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Science, Southern Medical University, Shatai South Road, Baiyun District, 16, Guangzhou, 510515, China
| | - Keyi Li
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Science, Southern Medical University, Shatai South Road, Baiyun District, 16, Guangzhou, 510515, China
| | - Jianping Chen
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Science, Southern Medical University, Shatai South Road, Baiyun District, 16, Guangzhou, 510515, China
| | - Zhuo Zhong
- Guangzhou Hospital of integrated Traditional and West Medicine, Guangzhou, 510800, China
| | - Qianbing Zhang
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Science, Southern Medical University, Shatai South Road, Baiyun District, 16, Guangzhou, 510515, China
| | - Bingtao Hao
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Science, Southern Medical University, Shatai South Road, Baiyun District, 16, Guangzhou, 510515, China.
| | - Qiuzhen Liu
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Science, Southern Medical University, Shatai South Road, Baiyun District, 16, Guangzhou, 510515, China. .,Pingshan District People's Hospital of Shenzhen, Shenzhen, 518118, China.
| |
Collapse
|
6
|
Zafonte RD, Wang L, Arbelaez CA, Dennison R, Teng YD. Medical Gas Therapy for Tissue, Organ, and CNS Protection: A Systematic Review of Effects, Mechanisms, and Challenges. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104136. [PMID: 35243825 PMCID: PMC9069381 DOI: 10.1002/advs.202104136] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 01/10/2022] [Indexed: 05/13/2023]
Abstract
Gaseous molecules have been increasingly explored for therapeutic development. Here, following an analytical background introduction, a systematic review of medical gas research is presented, focusing on tissue protections, mechanisms, data tangibility, and translational challenges. The pharmacological efficacies of carbon monoxide (CO) and xenon (Xe) are further examined with emphasis on intracellular messengers associated with cytoprotection and functional improvement for the CNS, heart, retina, liver, kidneys, lungs, etc. Overall, the outcome supports the hypothesis that readily deliverable "biological gas" (CO, H2 , H2 S, NO, O2 , O3 , and N2 O) or "noble gas" (He, Ar, and Xe) treatment may preserve cells against common pathologies by regulating oxidative, inflammatory, apoptotic, survival, and/or repair processes. Specifically, CO, in safe dosages, elicits neurorestoration via igniting sGC/cGMP/MAPK signaling and crosstalk between HO-CO, HIF-1α/VEGF, and NOS pathways. Xe rescues neurons through NMDA antagonism and PI3K/Akt/HIF-1α/ERK activation. Primary findings also reveal that the need to utilize cutting-edge molecular and genetic tactics to validate mechanistic targets and optimize outcome consistency remains urgent; the number of neurotherapeutic investigations is limited, without published results from large in vivo models. Lastly, the broad-spectrum, concurrent multimodal homeostatic actions of medical gases may represent a novel pharmaceutical approach to treating critical organ failure and neurotrauma.
Collapse
Affiliation(s)
- Ross D. Zafonte
- Department of Physical Medicine and RehabilitationHarvard Medical SchoolBostonMA02115USA
- Neurotrauma Recovery Research, Department of Physical Medicine and RehabilitationSpaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical SchoolBostonMA02129USA
- Spaulding Research InstituteSpaulding Rehabilitation Hospital NetworkBostonMA02129USA
| | - Lei Wang
- Department of Physical Medicine and RehabilitationHarvard Medical SchoolBostonMA02115USA
- Laboratory of SCI, Stem Cell and Recovery Neurobiology Research, Department of Physical Medicine and RehabilitationSpaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical SchoolBostonMA02129USA
| | - Christian A. Arbelaez
- Department of Physical Medicine and RehabilitationHarvard Medical SchoolBostonMA02115USA
- Laboratory of SCI, Stem Cell and Recovery Neurobiology Research, Department of Physical Medicine and RehabilitationSpaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical SchoolBostonMA02129USA
| | - Rachel Dennison
- Department of Physical Medicine and RehabilitationHarvard Medical SchoolBostonMA02115USA
- Laboratory of SCI, Stem Cell and Recovery Neurobiology Research, Department of Physical Medicine and RehabilitationSpaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical SchoolBostonMA02129USA
| | - Yang D. Teng
- Department of Physical Medicine and RehabilitationHarvard Medical SchoolBostonMA02115USA
- Neurotrauma Recovery Research, Department of Physical Medicine and RehabilitationSpaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical SchoolBostonMA02129USA
- Spaulding Research InstituteSpaulding Rehabilitation Hospital NetworkBostonMA02129USA
- Laboratory of SCI, Stem Cell and Recovery Neurobiology Research, Department of Physical Medicine and RehabilitationSpaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical SchoolBostonMA02129USA
| |
Collapse
|
7
|
Querio G, Antoniotti S, Geddo F, Tullio F, Penna C, Pagliaro P, Gallo MP. Ischemic heart disease and cardioprotection: Focus on estrogenic hormonal setting and microvascular health. Vascul Pharmacol 2021; 141:106921. [PMID: 34592428 DOI: 10.1016/j.vph.2021.106921] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/14/2021] [Accepted: 09/23/2021] [Indexed: 12/12/2022]
Abstract
Ischemic Heart Disease (IHD) is a clinical condition characterized by insufficient blood flow to the cardiac tissue, and the consequent inappropriate oxygen and nutrients supply and metabolic waste removal in the heart. In the last decade a broad scientific literature has underlined the distinct mechanism of onset and the peculiar progress of IHD between female and male patients, highlighting the estrogenic hormonal setting as a key factor of these sex-dependent divergences. In particular, estrogen-activated cardioprotective pathways exert a pivotal role for the microvascular health, and their impairment, both physiologically and pathologically driven, predispose to vascular dysfunctions. Aim of this review is to summarize the current knowledge on the estrogen receptors localization and function in the cardiovascular system, particularly focusing on sex-dependent differences in microvascular vs macrovascular dysfunction and on the experimental models that allowed the researchers to reach the current findings and sketching the leading estrogen-mediated cardioprotective mechanisms.
Collapse
Affiliation(s)
- Giulia Querio
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy
| | - Susanna Antoniotti
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy
| | - Federica Geddo
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy
| | - Francesca Tullio
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy.
| | - Maria Pia Gallo
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy.
| |
Collapse
|
8
|
Buelna-Chontal M, García-Niño WR, Silva-Palacios A, Enríquez-Cortina C, Zazueta C. Implications of Oxidative and Nitrosative Post-Translational Modifications in Therapeutic Strategies against Reperfusion Damage. Antioxidants (Basel) 2021; 10:749. [PMID: 34066806 PMCID: PMC8151040 DOI: 10.3390/antiox10050749] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/03/2021] [Accepted: 05/05/2021] [Indexed: 12/17/2022] Open
Abstract
Post-translational modifications based on redox reactions "switch on-off" the biological activity of different downstream targets, modifying a myriad of processes and providing an efficient mechanism for signaling regulation in physiological and pathological conditions. Such modifications depend on the generation of redox components, such as reactive oxygen species and nitric oxide. Therefore, as the oxidative or nitrosative milieu prevailing in the reperfused heart is determinant for protective signaling, in this review we defined the impact of redox-based post-translational modifications resulting from either oxidative/nitrosative signaling or oxidative/nitrosative stress that occurs during reperfusion damage. The role that cardioprotective conditioning strategies have had to establish that such changes occur at different subcellular levels, particularly in mitochondria, is also presented. Another section is devoted to the possible mechanism of signal delivering of modified proteins. Finally, we discuss the possible efficacy of redox-based therapeutic strategies against reperfusion damage.
Collapse
Affiliation(s)
| | | | | | | | - Cecilia Zazueta
- Department of Cardiovascular Biomedicine, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico; (M.B.-C.); (W.R.G.-N.); (A.S.-P.); (C.E.-C.)
| |
Collapse
|
9
|
Post-Translational S-Nitrosylation of Proteins in Regulating Cardiac Oxidative Stress. Antioxidants (Basel) 2020; 9:antiox9111051. [PMID: 33126514 PMCID: PMC7693965 DOI: 10.3390/antiox9111051] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/23/2020] [Accepted: 10/26/2020] [Indexed: 02/07/2023] Open
Abstract
Like other post-translational modifications (PTMs) of proteins, S-nitrosylation has been considered a key regulatory mechanism of multiple cellular functions in many physiological and disease conditions. Emerging evidence has demonstrated that S-nitrosylation plays a crucial role in regulating redox homeostasis in the stressed heart, leading to discoveries in the mechanisms underlying the pathogenesis of heart diseases and cardiac protection. In this review, we summarize recent studies in understanding the molecular and biological basis of S-nitrosylation, including the formation, spatiotemporal specificity, homeostatic regulation, and association with cellular redox status. We also outline the currently available methods that have been applied to detect S-nitrosylation. Additionally, we synopsize the up-to-date studies of S-nitrosylation in various cardiac diseases in humans and animal models, and we discuss its therapeutic potential in cardiac protection. These pieces of information would bring new insights into understanding the role of S-nitrosylation in cardiac pathogenesis and provide novel avenues for developing novel therapeutic strategies for heart diseases.
Collapse
|
10
|
Lucciola R, Vrljicak P, Gurung S, Filby C, Darzi S, Muter J, Ott S, Brosens JJ, Gargett CE. Impact of Sustained Transforming Growth Factor-β Receptor Inhibition on Chromatin Accessibility and Gene Expression in Cultured Human Endometrial MSC. Front Cell Dev Biol 2020; 8:567610. [PMID: 32984350 PMCID: PMC7490520 DOI: 10.3389/fcell.2020.567610] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 08/13/2020] [Indexed: 12/18/2022] Open
Abstract
Endometrial mesenchymal stem cells (eMSC) drive the extraordinary regenerative capacity of the human endometrium. Clinical application of eMSC for therapeutic purposes is hampered by spontaneous differentiation and cellular senescence upon large-scale expansion in vitro. A83-01, a selective transforming growth factor-β receptor (TGFβ-R) inhibitor, promotes expansion of eMSC in culture by blocking differentiation and senescence, but the underlying mechanisms are incompletely understood. In this study, we combined RNA-seq and ATAC-seq to study the impact of sustained TGFβ-R inhibition on gene expression and chromatin architecture of eMSC. Treatment of primary eMSC with A83-01 for 5 weeks resulted in differential expression of 1,463 genes. Gene ontology analysis showed enrichment of genes implicated in cell growth whereas extracellular matrix genes and genes involved in cell fate commitment were downregulated. ATAC-seq analysis demonstrated that sustained TGFβ-R inhibition results in opening and closure of 3,555 and 2,412 chromatin loci, respectively. Motif analysis revealed marked enrichment of retinoic acid receptor (RAR) binding sites, which was paralleled by the induction of RARB, encoding retinoic acid receptor beta (RARβ). Selective RARβ inhibition attenuated proliferation and clonogenicity of A83-01 treated eMSC. Taken together, our study provides new insights into the gene networks and genome-wide chromatin changes that underpin maintenance of an undifferentiated phenotype of eMSC in prolonged culture.
Collapse
Affiliation(s)
- Raffaella Lucciola
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Pavle Vrljicak
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, United Kingdom
- Tommy’s National Centre for Miscarriage Research, Warwick Medical School, University Hospitals Coventry and Warwickshire National Health Service Trust, Coventry, United Kingdom
| | - Shanti Gurung
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Caitlin Filby
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Saeedeh Darzi
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Joanne Muter
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, United Kingdom
- Tommy’s National Centre for Miscarriage Research, Warwick Medical School, University Hospitals Coventry and Warwickshire National Health Service Trust, Coventry, United Kingdom
| | - Sascha Ott
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, United Kingdom
- Tommy’s National Centre for Miscarriage Research, Warwick Medical School, University Hospitals Coventry and Warwickshire National Health Service Trust, Coventry, United Kingdom
| | - Jan J. Brosens
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, United Kingdom
- Tommy’s National Centre for Miscarriage Research, Warwick Medical School, University Hospitals Coventry and Warwickshire National Health Service Trust, Coventry, United Kingdom
| | - Caroline E. Gargett
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
11
|
Cardiovascular Therapeutic Potential of the Redox Siblings, Nitric Oxide (NO•) and Nitroxyl (HNO), in the Setting of Reactive Oxygen Species Dysregulation. Handb Exp Pharmacol 2020; 264:311-337. [PMID: 32813078 DOI: 10.1007/164_2020_389] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Reactive oxygen species (ROS) dysregulation is a hallmark of cardiovascular disease, characterised by an imbalance in the synthesis and removal of ROS. ROS such as superoxide (•O2-), hydrogen peroxide (H2O2), hydroxyl (OH•) and peroxynitrite (ONOO-) have a marked impact on cardiovascular function, contributing to the vascular impairment and cardiac dysfunction associated with diseases such as angina, hypertension, diabetes and heart failure. Central to the vascular dysfunction is a reduction in bioavailability and/or physiological effects of vasoprotective nitric oxide (NO•), leading to vasoconstriction, inflammation and vascular remodelling. In a cardiac context, increased ROS generation can also lead to modification of key proteins involved in cardiac contractility. Whilst playing a key role in the pathogenesis of cardiovascular disease, ROS dysregulation also limits the clinical efficacy of current therapies, such as nitrosovasodilators. As such, alternate therapies are sought. This review will discuss the impact of ROS dysregulation on the therapeutic utility of NO• and its redox sibling, nitroxyl (HNO). Both nitric oxide (NO) and nitroxyl (HNO) donors signal through soluble guanylyl cyclase (sGC). NO binds to the Fe(II) form of sGC and nitroxyl possibly to both sGC heme and thiol groups. In the vasculature, nitroxyl can also signal through voltage-dependent (Kv) and ATP-sensitive (KATP) K+ channels as well as calcitonin gene-related peptide (CGRP). In the heart, HNO directly targets critical thiols to increase myocardial contractility, an effect not seen with NO. The qualitative effects via elevation of cGMP are similar, i.e. lusitropic in the heart and inhibitory on vasoconstriction, inflammation, aggregation and vascular remodelling. Of pathophysiological significance is the fact the efficacy of NO donors is impaired by ROS, e.g. through chemical scavenging of NO, to generate reactive nitrogen oxide species (RNOS), whilst nitroxyl is apparently not.
Collapse
|
12
|
Gáspár R, Gömöri K, Kiss B, Szántai Á, Pálóczi J, Varga ZV, Pipis J, Váradi B, Ágg B, Csont T, Ferdinandy P, Barteková M, Görbe A. Decorin Protects Cardiac Myocytes against Simulated Ischemia/Reperfusion Injury. Molecules 2020; 25:molecules25153426. [PMID: 32731559 PMCID: PMC7436189 DOI: 10.3390/molecules25153426] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/22/2020] [Accepted: 07/23/2020] [Indexed: 01/13/2023] Open
Abstract
Search for new cardioprotective therapies is of great importance since no cardioprotective drugs are available on the market. In line with this need, several natural biomolecules have been extensively tested for their potential cardioprotective effects. Previously, we have shown that biglycan, a member of a diverse group of small leucine-rich proteoglycans, enhanced the expression of cardioprotective genes and decreased ischemia/reperfusion-induced cardiomyocyte death via a TLR-4 dependent mechanism. Therefore, in the present study we aimed to test whether decorin, a small leucine-rich proteoglycan closely related to biglycan, could exert cardiocytoprotection and to reveal possible downstream signaling pathways. Methods: Primary cardiomyocytes isolated from neonatal and adult rat hearts were treated with 0 (Vehicle), 1, 3, 10, 30 and 100 nM decorin as 20 h pretreatment and maintained throughout simulated ischemia and reperfusion (SI/R). In separate experiments, to test the mechanism of decorin-induced cardio protection, 3 nM decorin was applied in combination with inhibitors of known survival pathways, that is, the NOS inhibitor L-NAME, the PKG inhibitor KT-5823 and the TLR-4 inhibitor TAK-242, respectively. mRNA expression changes were measured after SI/R injury. Results: Cell viability of both neonatal and adult cardiomyocytes was significantly decreased due to SI/R injury. Decorin at 1, 3 and 10 nM concentrations significantly increased the survival of both neonatal and adult myocytes after SI/R. At 3nM (the most pronounced protective concentration), it had no effect on apoptotic rate of neonatal cardiac myocytes. No one of the inhibitors of survival pathways (L-NAME, KT-5823, TAK-242) influenced the cardiocytoprotective effect of decorin. MYND-type containing 19 (Zmynd19) and eukaryotic translation initiation factor 4E nuclear import factor 1 (Eif4enif1) were significantly upregulated due to the decorin treatment. In conclusion, this is the first demonstration that decorin exerts a direct cardiocytoprotective effect possibly independent of NO-cGMP-PKG and TLR-4 dependent survival signaling.
Collapse
Affiliation(s)
- Renáta Gáspár
- Metabolic Diseases and Cell Signaling (MEDICS) Research Group, Department of Biochemistry, Interdisciplinary Excellence Centre, University of Szeged, Dom ter 9, H-6720 Szeged, Hungary; (R.G.); (T.C.)
| | - Kamilla Gömöri
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, University of Szeged, Dom ter 12, H-6720 Szeged, Hungary; (K.G.); (Á.S.); (J.P.)
| | - Bernadett Kiss
- Cardiometabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvarad ter 4, H-1089 Budapest, Hungary; (B.K.); (Z.V.V.); (B.V.); (B.Á.); (P.F.)
- MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
| | - Ágnes Szántai
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, University of Szeged, Dom ter 12, H-6720 Szeged, Hungary; (K.G.); (Á.S.); (J.P.)
| | - János Pálóczi
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, University of Szeged, Dom ter 12, H-6720 Szeged, Hungary; (K.G.); (Á.S.); (J.P.)
| | - Zoltán V. Varga
- Cardiometabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvarad ter 4, H-1089 Budapest, Hungary; (B.K.); (Z.V.V.); (B.V.); (B.Á.); (P.F.)
| | - Judit Pipis
- Pharmahungary Group, Hajnoczy utca 6, H-6722 Szeged, Hungary;
| | - Barnabás Váradi
- Cardiometabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvarad ter 4, H-1089 Budapest, Hungary; (B.K.); (Z.V.V.); (B.V.); (B.Á.); (P.F.)
| | - Bence Ágg
- Cardiometabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvarad ter 4, H-1089 Budapest, Hungary; (B.K.); (Z.V.V.); (B.V.); (B.Á.); (P.F.)
- MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
- Pharmahungary Group, Hajnoczy utca 6, H-6722 Szeged, Hungary;
| | - Tamás Csont
- Metabolic Diseases and Cell Signaling (MEDICS) Research Group, Department of Biochemistry, Interdisciplinary Excellence Centre, University of Szeged, Dom ter 9, H-6720 Szeged, Hungary; (R.G.); (T.C.)
| | - Péter Ferdinandy
- Cardiometabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvarad ter 4, H-1089 Budapest, Hungary; (B.K.); (Z.V.V.); (B.V.); (B.Á.); (P.F.)
- MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
- Pharmahungary Group, Hajnoczy utca 6, H-6722 Szeged, Hungary;
| | - Monika Barteková
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, Dúbravská cesta 9, 841 04 Bratislava, Slovak
- Institute of Physiology, Comenius University in Bratislava, Sasinkova 2, 813 72 Bratislava, Slovak
- Correspondence: (M.B.); (A.G.)
| | - Anikó Görbe
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, University of Szeged, Dom ter 12, H-6720 Szeged, Hungary; (K.G.); (Á.S.); (J.P.)
- Cardiometabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvarad ter 4, H-1089 Budapest, Hungary; (B.K.); (Z.V.V.); (B.V.); (B.Á.); (P.F.)
- MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
- Pharmahungary Group, Hajnoczy utca 6, H-6722 Szeged, Hungary;
- Correspondence: (M.B.); (A.G.)
| |
Collapse
|
13
|
Boulghobra D, Coste F, Geny B, Reboul C. Exercise training protects the heart against ischemia-reperfusion injury: A central role for mitochondria? Free Radic Biol Med 2020; 152:395-410. [PMID: 32294509 DOI: 10.1016/j.freeradbiomed.2020.04.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 04/01/2020] [Accepted: 04/07/2020] [Indexed: 12/11/2022]
Abstract
Ischemic heart disease is one of the main causes of morbidity and mortality worldwide. Physical exercise is an effective lifestyle intervention to reduce the risk factors for cardiovascular disease and also to improve cardiac function and survival in patients with ischemic heart disease. Among the strategies that contribute to reduce heart damages during ischemia and reperfusion, regular physical exercise is efficient both in rodent experimental models and in humans. However, the cellular and molecular mechanisms of the cardioprotective effects of exercise remain unclear. During ischemia and reperfusion, mitochondria are crucial players in cell death, but also in cell survival. Although exercise training can influence mitochondrial function, the consequences on heart sensitivity to ischemic insults remain elusive. In this review, we describe the effects of physical activity on cardiac mitochondria and their potential key role in exercise-induced cardioprotection against ischemia-reperfusion damage. Based on recent scientific data, we discuss the role of different pathways that might help to explain why mitochondria are a key target of exercise-induced cardioprotection.
Collapse
Affiliation(s)
| | - Florence Coste
- LAPEC EA4278, Avignon Université, F-84000, Avignon, France
| | - Bernard Geny
- EA3072, «Mitochondrie, Stress Oxydant, et Protection Musculaire», Université de Strasbourg, 67000, Strasbourg, France
| | - Cyril Reboul
- LAPEC EA4278, Avignon Université, F-84000, Avignon, France.
| |
Collapse
|
14
|
Bibli SI, Papapetropoulos A, Iliodromitis EK, Daiber A, Randriamboavonjy V, Steven S, Brouckaert P, Chatzianastasiou A, Kypreos KE, Hausenloy DJ, Fleming I, Andreadou I. Nitroglycerine limits infarct size through S-nitrosation of cyclophilin D: a novel mechanism for an old drug. Cardiovasc Res 2020; 115:625-636. [PMID: 30165375 DOI: 10.1093/cvr/cvy222] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 08/23/2018] [Indexed: 12/22/2022] Open
Abstract
AIMS Nitroglycerine (NTG) given prior to an ischaemic insult exerts cardioprotective effects. However, whether administration of an acute low dose of NTG in a clinically relevant manner following an ischaemic episode limits infarct size, has not yet been explored. METHODS AND RESULTS Adult mice were subjected to acute myocardial infarction in vivo and then treated with vehicle or low-dose NTG prior to reperfusion. This treatment regimen minimized myocardial infarct size without affecting haemodynamic parameters but the protective effect was absent in mice rendered tolerant to the drug. Mechanistically, NTG was shown to nitrosate and inhibit cyclophilin D (CypD), and NTG administration failed to limit infarct size in CypD knockout mice. Additional experiments revealed lack of the NTG protective effect following genetic (knockout mice) or pharmacological inhibition (L-NAME treatment) of the endothelial nitric oxide synthase (eNOS). The protective effect of NTG was attributed to preservation of the eNOS dimer. Moreover, NTG retained its cardioprotective effects in a model of endothelial dysfunction (ApoE knockout) by preserving CypD nitrosation. Human ischaemic heart biopsies revealed reduced eNOS activity and exhibited reduced CypD nitrosation. CONCLUSION Low-dose NTG given prior to reperfusion reduces myocardial infarct size by preserving eNOS function, and the subsequent eNOS-dependent S-nitrosation of CypD, inhibiting cardiomyocyte necrosis. This novel pharmacological action of NTG warrants confirmation in clinical studies, although our data in human biopsies provide promising preliminary results.
Collapse
Affiliation(s)
- Sofia-Iris Bibli
- Laboratoty of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, Athens, Greece.,Institute for Vascular Signaling, Goethe University, Theodor Stern Kai 7, Frankfurt, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Andreas Papapetropoulos
- Laboratoty of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, Athens, Greece
| | - Efstathios K Iliodromitis
- Faculty of Medicine, Second Department of Cardiology, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Andreas Daiber
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany.,University Medical Center of Mainz, Center for Cardiology, Cardiology I, Molecular Cardiology, Mainz, Germany
| | - Voahanginirina Randriamboavonjy
- Institute for Vascular Signaling, Goethe University, Theodor Stern Kai 7, Frankfurt, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Sebastian Steven
- University Medical Center of Mainz, Center for Cardiology, Cardiology I, Molecular Cardiology, Mainz, Germany.,University Medical Center of Mainz, Center for Thrombosis and Hemostasis (CTH), Mainz, Germany
| | - Peter Brouckaert
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.,Department of Molecular Biomedical Research, VIB, Ghent, Belgium
| | - Athanasia Chatzianastasiou
- Laboratoty of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, Athens, Greece
| | - Kyriakos E Kypreos
- Department of Pharmacology, University of Patras Medical School, Patras, Greece
| | - Derek J Hausenloy
- Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore.,National Heart Research Institute Singapore, National Heart Centre, Singapore.,Yong Loo Lin School of Medicine, National University Singapore, Singapore.,The Hatter Cardiovascular Institute, University College London, London, UK.,The National Institute of Health Research University College London Hospitals Biomedical Research Centre, Research & Development, London, UK.,Department of Cardiology, Barts Heart Centre, St Bartholomew's Hospital, London, UK
| | - Ingrid Fleming
- Institute for Vascular Signaling, Goethe University, Theodor Stern Kai 7, Frankfurt, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Ioanna Andreadou
- Laboratoty of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, Athens, Greece
| |
Collapse
|
15
|
Balbi C, Costa A, Barile L, Bollini S. Message in a Bottle: Upgrading Cardiac Repair into Rejuvenation. Cells 2020; 9:cells9030724. [PMID: 32183455 PMCID: PMC7140681 DOI: 10.3390/cells9030724] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/05/2020] [Accepted: 03/12/2020] [Indexed: 12/12/2022] Open
Abstract
Ischaemic cardiac disease is associated with a loss of cardiomyocytes and an intrinsic lack of myocardial renewal. Recent work has shown that the heart retains limited cardiomyocyte proliferation, which remains inefficient when facing pathological conditions. While broadly active in the neonatal mammalian heart, this mechanism becomes quiescent soon after birth, suggesting loss of regenerative potential with maturation into adulthood. A key question is whether this temporary regenerative window can be enhanced via appropriate stimulation and further extended. Recently the search for novel therapeutic approaches for heart disease has centred on stem cell biology. The “paracrine effect” has been proposed as a promising strategy to boost endogenous reparative and regenerative mechanisms from within the cardiac tissue by exploiting the modulatory potential of soluble stem cell-secreted factors. As such, growing interest has been specifically addressed towards stem/progenitor cell-secreted extracellular vesicles (EVs), which can be easily isolated in vitro from cell-conditioned medium. This review will provide a comprehensive overview of the current paradigm on cardiac repair and regeneration, with a specific focus on the role and mechanism(s) of paracrine action of EVs from cardiac stromal progenitors as compared to exogenous stem cells in order to discuss the optimal choice for future therapy. In addition, the challenges to overcoming translational EV biology from bench to bedside for future cardiac regenerative medicine will be discussed.
Collapse
Affiliation(s)
- Carolina Balbi
- Laboratory of Cellular and Molecular Cardiology, Cardiocentro Ticino Foundation, 6900 Lugano, Switzerland;
| | - Ambra Costa
- Regenerative Medicine Laboratory, Dept. of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy;
| | - Lucio Barile
- Laboratory for Cardiovascular Theranostics, Cardiocentro Ticino Foundation, 6900 Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6900 Lugano, Switzerland
- Correspondence: (L.B.); (S.B.)
| | - Sveva Bollini
- Regenerative Medicine Laboratory, Dept. of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy;
- Correspondence: (L.B.); (S.B.)
| |
Collapse
|
16
|
Sun J, Hao W, Fillmore N, Ma H, Springer D, Yu ZX, Sadowska A, Garcia A, Chen R, Muniz-Medina V, Rosenthal K, Lin J, Kuruvilla D, Osbourn J, Karathanasis SK, Walker J, Murphy E. Human Relaxin-2 Fusion Protein Treatment Prevents and Reverses Isoproterenol-Induced Hypertrophy and Fibrosis in Mouse Heart. J Am Heart Assoc 2019; 8:e013465. [PMID: 31818212 PMCID: PMC6951077 DOI: 10.1161/jaha.119.013465] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background Heart failure is one of the leading causes of death in Western countries, and there is a need for new therapeutic approaches. Relaxin‐2 is a peptide hormone that mediates pleiotropic cardiovascular effects, including antifibrotic, angiogenic, vasodilatory, antiapoptotic, and anti‐inflammatory effects in vitro and in vivo. Methods and Results We developed RELAX10, a fusion protein composed of human relaxin‐2 hormone and the Fc of a human antibody, to test the hypothesis that extended exposure of the relaxin‐2 peptide could reduce cardiac hypertrophy and fibrosis. RELAX10 demonstrated the same specificity and similar in vitro activity as the relaxin‐2 peptide. The terminal half‐life of RELAX10 was 7 days in mouse and 3.75 days in rat after subcutaneous administration. We evaluated whether treatment with RELAX10 could prevent and reverse isoproterenol‐induced cardiac hypertrophy and fibrosis in mice. Isoproterenol administration in mice resulted in increased cardiac hypertrophy and fibrosis compared with vehicle. Coadministration with RELAX10 significantly attenuated the cardiac hypertrophy and fibrosis compared with untreated animals. Isoproterenol administration significantly increased transforming growth factor β1 (TGF‐β1)–induced fibrotic signaling, which was attenuated by RELAX10. We found that RELAX10 also significantly increased protein kinase B/endothelial NO synthase signaling and protein S‐nitrosylation. In the reversal study, RELAX10‐treated animals showed significantly reduced cardiac hypertrophy and collagen levels. Conclusions These findings support a potential role for RELAX10 in the treatment of heart failure.
Collapse
Affiliation(s)
- Junhui Sun
- Cardiac Physiology Section/Cardiovascular Branch National Heart, Lung, and Blood Institute/National Institutes of Health Bethesda MD
| | | | - Natasha Fillmore
- Cardiac Physiology Section/Cardiovascular Branch National Heart, Lung, and Blood Institute/National Institutes of Health Bethesda MD
| | - Hanley Ma
- Cardiac Physiology Section/Cardiovascular Branch National Heart, Lung, and Blood Institute/National Institutes of Health Bethesda MD
| | - Danielle Springer
- Murine Phenotyping Core National Heart, Lung, and Blood Institute/National Institutes of Health Bethesda MD
| | - Zu-Xi Yu
- Pathology Core National Heart, Lung, and Blood Institute/National Institutes of Health Bethesda MD
| | | | | | | | | | | | | | | | | | | | | | - Elizabeth Murphy
- Cardiac Physiology Section/Cardiovascular Branch National Heart, Lung, and Blood Institute/National Institutes of Health Bethesda MD
| |
Collapse
|
17
|
Giri S, Katakia YT, Chatterjee S, Gajalakshmi P. Breast cancer drugs perturb fundamental vascular functions of endothelial cells by attenuating protein S-nitrosylation. Clin Exp Pharmacol Physiol 2019; 47:7-15. [PMID: 31549415 DOI: 10.1111/1440-1681.13181] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 08/28/2019] [Accepted: 09/20/2019] [Indexed: 11/29/2022]
Abstract
Cardiovascular side effects of broadly used chemotherapeutic drugs such as Tamoxifen citrate (TC), Capecitabine (CP) and Epirubicin (EP) among cancer survivors are well established. Nitric oxide (NO) is known to protect cardiovascular tissues under conditions of stress. NO can act through cyclic guanosine monophosphate (cGMP)-dependent and -independent pathways. Particularly, the S-nitrosylation of SH-groups in a protein by NO falls under cGMP-independent effects of NO. TC, CP, and EP are hypothesized as interfering with cellular protein S-nitrosylation, which, in turn, may lead to endothelial dysfunctions. The results show that all three drugs attenuate nitrosylated proteins in endothelial cells. A significant reduction in endogenous S-nitrosylated proteins was revealed by Saville-Griess assay, immunofluorescence and western blot. Incubation with the drugs causes a reduction in endothelial migration, vasodilation and tube formation, while the addition of S-nitrosoglutathione (GSNO) has a reversal of this effect. In conclusion, results indicate the possibility of decreased cellular nitrosothiols as being one of the reasons for endothelial dysfunctions under TC, CP and EP treatment. Identification of the down-regulated S-nitrosylated proteins so as to correlate their implications on fundamental vascular functions could be an interesting phenomenon.
Collapse
Affiliation(s)
- Suvendu Giri
- Department of Biotechnology & AU-KBC Research Centre, Anna University, Chennai, India
| | - Yash Tushar Katakia
- Department of Biotechnology & AU-KBC Research Centre, Anna University, Chennai, India
| | - Suvro Chatterjee
- Department of Biotechnology & AU-KBC Research Centre, Anna University, Chennai, India
| | | |
Collapse
|
18
|
Casin KM, Fallica J, Mackowski N, Veenema RJ, Chan A, St Paul A, Zhu G, Bedja D, Biswal S, Kohr MJ. S-Nitrosoglutathione Reductase Is Essential for Protecting the Female Heart From Ischemia-Reperfusion Injury. Circ Res 2019; 123:1232-1243. [PMID: 30571462 DOI: 10.1161/circresaha.118.313956] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
RATIONALE Protein S-nitros(yl)ation (SNO) has been implicated as an essential mediator of nitric oxide-dependent cardioprotection. Compared with males, female hearts exhibit higher baseline levels of protein SNO and associated with this, reduced susceptibility to myocardial ischemia-reperfusion injury. Female hearts also exhibit enhanced S-nitrosoglutathione reductase (GSNO-R) activity, which would typically favor decreased SNO levels as GSNO-R mediates SNO catabolism. OBJECTIVE Because female hearts exhibit higher SNO levels, we hypothesized that GSNO-R is an essential component of sex-dependent cardioprotection in females. METHODS AND RESULTS Male and female wild-type mouse hearts were subjected to ex vivo ischemia-reperfusion injury with or without GSNO-R inhibition (N6022). Control female hearts exhibited enhanced functional recovery and decreased infarct size versus control males. Interestingly, GSNO-R inhibition reversed this sex disparity, significantly reducing injury in male hearts, and exacerbating injury in females. Similar results were obtained with male and female GSNO-R-/- hearts using ex vivo and in vivo models of ischemia-reperfusion injury. Assessment of SNO levels using SNO-resin assisted capture revealed an increase in total SNO levels with GSNO-R inhibition in males, whereas total SNO levels remained unchanged in females. However, we found that although GSNO-R inhibition significantly increased SNO at the cardioprotective Cys39 residue of nicotinamide adenine dinucleotide (NADH) dehydrogenase subunit 3 in males, SNO-NADH dehydrogenase subunit 3 levels were surprisingly reduced in N6022-treated female hearts. Because GSNO-R also acts as a formaldehyde dehydrogenase, we examined postischemic formaldehyde levels and found that they were nearly 2-fold higher in N6022-treated female hearts compared with nontreated hearts. Importantly, the mitochondrial aldehyde dehydrogenase 2 activator, Alda-1, rescued the phenotype in GSNO-R-/- female hearts, significantly reducing infarct size. CONCLUSIONS These striking findings point to GSNO-R as a critical sex-dependent mediator of myocardial protein SNO and formaldehyde levels and further suggest that different therapeutic strategies may be required to combat ischemic heart disease in males and females.
Collapse
Affiliation(s)
- Kevin M Casin
- From the Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (K.M.C., J.F., N.M., R.J.V., A.C., A.S.P., S.B., M.J.K.)
| | - Jonathan Fallica
- From the Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (K.M.C., J.F., N.M., R.J.V., A.C., A.S.P., S.B., M.J.K.)
| | - Nathan Mackowski
- From the Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (K.M.C., J.F., N.M., R.J.V., A.C., A.S.P., S.B., M.J.K.)
| | - Ryne J Veenema
- From the Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (K.M.C., J.F., N.M., R.J.V., A.C., A.S.P., S.B., M.J.K.)
| | - Ashley Chan
- From the Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (K.M.C., J.F., N.M., R.J.V., A.C., A.S.P., S.B., M.J.K.)
| | - Amanda St Paul
- From the Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (K.M.C., J.F., N.M., R.J.V., A.C., A.S.P., S.B., M.J.K.)
| | - Guangshuo Zhu
- Cardiology Division, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (G.Z., D.B.)
| | - Djahida Bedja
- Cardiology Division, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (G.Z., D.B.)
| | - Shyam Biswal
- From the Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (K.M.C., J.F., N.M., R.J.V., A.C., A.S.P., S.B., M.J.K.)
| | - Mark J Kohr
- From the Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (K.M.C., J.F., N.M., R.J.V., A.C., A.S.P., S.B., M.J.K.)
| |
Collapse
|
19
|
Zadek F, Spina S, Hu J, Berra L. Nitric Oxide Treatment for Lungs and Beyond. Novel Insights from Recent Literature. Am J Respir Crit Care Med 2019; 200:628-630. [PMID: 31185176 PMCID: PMC6727154 DOI: 10.1164/rccm.201901-0037rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 06/11/2019] [Indexed: 12/17/2022] Open
Affiliation(s)
- Francesco Zadek
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Stefano Spina
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Jie Hu
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Lorenzo Berra
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
20
|
Román-Anguiano NG, Correa F, Cano-Martínez A, de la Peña-Díaz A, Zazueta C. Cardioprotective effects of Prolame and SNAP are related with nitric oxide production and with diminution of caspases and calpain-1 activities in reperfused rat hearts. PeerJ 2019; 7:e7348. [PMID: 31392096 PMCID: PMC6673759 DOI: 10.7717/peerj.7348] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 06/25/2019] [Indexed: 01/26/2023] Open
Abstract
Cardiac tissue undergoes changes during ischemia-reperfusion (I-R) that compromise its normal function. Cell death is one of the consequences of such damage, as well as diminution in nitric oxide (NO) content. This signaling molecule regulates the function of the cardiovascular system through dependent and independent effects of cyclic guanosine monophosphate (cGMP). The independent cGMP pathway involves post-translational modification of proteins by S-nitrosylation. Studies in vitro have shown that NO inhibits the activity of caspases and calpains through S-nitrosylation of a cysteine located in their catalytic site, so we propose to elucidate if the regulatory mechanisms of NO are related with changes in S-nitrosylation of cell death proteins in the ischemic-reperfused myocardium. We used two compounds that increase the levels of NO by different mechanisms: Prolame, an amino-estrogenic compound with antiplatelet and anticoagulant effects that induces the increase of NO levels in vivo by activating the endothelial nitric oxide synthase (eNOS) and that has not been tested as a potential inhibitor of apoptosis. On the other hand, S-Nitroso-N-acetylpenicillamine (SNAP), a synthetic NO donor that has been shown to decrease cell death after inducing hypoxia-reoxygenation in cell cultures. Main experimental groups were Control, I-R, I-R+Prolame and I-R+SNAP. Additional groups were used to evaluate the NO action pathways. Contractile function represented as heart rate and ventricular pressure was evaluated in a Langendorff system. Infarct size was measured with 2,3,5-triphenyltetrazolium chloride stain. NO content was determined indirectly by measuring nitrite levels with the Griess reaction and cGMP content was measured by Enzyme-Linked ImmunoSorbent Assay. DNA integrity was evaluated by DNA laddering visualized on an agarose gel and by Terminal deoxynucleotidyl transferase dUTP Nick-End Labeling assay. Activities of caspase-3, caspase-8, caspase-9 and calpain-1 were evaluated spectrophotometrically and the content of caspase-3 and calpain-1 by western blot. S-nitrosylation of caspase-3 and calpain-1 was evaluated by labeling S-nitrosylated cysteines. Our results show that both Prolame and SNAP increased NO content and improved functional recovery in post-ischemic hearts. cGMP-dependent and S-nitrosylation pathways were activated in both groups, but the cGMP-independent pathway was preferentially activated by SNAP, which induced higher levels of NO than Prolame. Although SNAP effectively diminished the activity of all the proteases, a correlative link between the activity of these proteases and S-nitrosylation was not fully established.
Collapse
Affiliation(s)
| | - Francisco Correa
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiologia Ignacio Chávez, México, México
| | - Agustina Cano-Martínez
- Departamento de Fisiología, Instituto Nacional de Cardiologia Ignacio Chávez, México, México
| | - Aurora de la Peña-Díaz
- Departamento de Biología Molecular, Instituto Nacional de Cardiologia Ignacio Chávez, México, México.,Departamento de Farmacología, Universidad Nacional Autónoma de México, México, Mexico
| | - Cecilia Zazueta
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiologia Ignacio Chávez, México, México
| |
Collapse
|
21
|
Naringenin Attenuates Myocardial Ischemia-Reperfusion Injury via cGMP-PKGI α Signaling and In Vivo and In Vitro Studies. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:7670854. [PMID: 30728891 PMCID: PMC6341255 DOI: 10.1155/2019/7670854] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 10/26/2018] [Indexed: 12/27/2022]
Abstract
Endoplasmic reticulum (ER) stress and oxidative stress contribute greatly to myocardial ischemia-reperfusion (MI/R) injury. Naringenin, a flavonoid derived from the citrus genus, exerts cardioprotective effects. However, the effects of naringenin on ER stress as well as oxidative stress under MI/R condition and the detailed mechanisms remain poorly defined. This study investigated the protective effect of naringenin on MI/R-injured heart with a focus on cyclic guanosine monophosphate- (cGMP-) dependent protein kinase (PKG) signaling. Sprague-Dawley rats were treated with naringenin (50 mg/kg/d) and subjected to MI/R surgery with or without KT5823 (2 mg/kg, a selective inhibitor of PKG) cotreatment. Cellular experiment was conducted on H9c2 cardiomyoblasts subjected to simulated ischemia-reperfusion treatment. Before the treatment, the cells were incubated with naringenin (80 μmol/L). PKGIα siRNA was employed to inhibit PKG signaling. Our in vivo and in vitro data showed that naringenin effectively improved heart function while it attenuated myocardial apoptosis and infarction. Furthermore, pretreatment with naringenin suppressed MI/R-induced oxidative stress as well as ER stress as evidenced by decreased superoxide generation, myocardial MDA level, gp91phox expression, and phosphorylation of PERK, IRE1α, and EIF2α as well as reduced ATF6 and CHOP. Importantly, naringenin significantly activated myocardial cGMP-PKGIα signaling while inhibition of PKG signaling with KT5823 (in vivo) or siRNA (in vitro) not only abolished these actions but also blunted naringenin's inhibitory effects against oxidative stress and ER stress. In summary, our study demonstrates that naringenin treatment protects against MI/R injury by reducing oxidative stress and ER stress via cGMP-PKGIα signaling. Its cardioprotective effect deserves further clinical study.
Collapse
|
22
|
Bibli SI, Zhou Z, Zukunft S, Fisslthaler B, Andreadou I, Szabo C, Brouckaert P, Fleming I, Papapetropoulos A. Tyrosine phosphorylation of eNOS regulates myocardial survival after an ischaemic insult: role of PYK2. Cardiovasc Res 2018; 113:926-937. [PMID: 28444132 DOI: 10.1093/cvr/cvx058] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Accepted: 03/20/2017] [Indexed: 02/06/2023] Open
Abstract
Aims Endothelial nitric oxide (NO) synthase (eNOS) is known to play a cardioprotective protective. However, the molecular mechanisms regulating eNOS activity during ischaemia/reperfusion (I/R) injury are incompletely understood. eNOS is a substrate for several kinases that positively or negatively affect its enzymatic activity. Herein, we sought to correlate eNOS phosphorylation status with cardiomyocyte survival and we investigated the contribution of the proline-rich tyrosine kinase 2 (PYK2)/eNOS axis to the regulation of myocardial infarct size in vivo. Methods and results Exposure of H9c2 cardiomyocytes to H2O2 lead to PYK2 phosphorylation on its activator site (Y402) and eNOS phosphorylation on the inhibitor site Y656 and the activator site S1176. Both H2O2-induced eNOS phosphorylation events were abolished by PYK2 pharmacological inhibition or gene knockdown. Activity assays demonstrated that phosphorylation of the tyrosine inhibitory site exerts a dominant effect over S1176. In cardiomyocytes subjected to oxidative stress or oxygen-glucose deprivation, inhibition of PYK2 limited cell injury; this effect was prevented by inhibition of NO production. In vivo, ischaemia-reperfusion induced an early activation of PYK2, leading to eNOS phosphorylation on Y656, which, in turn, reduced NO output, as judged by the low tissue levels of its downstream effector cGMP. Moreover, pharmacological blockade of PYK2 alleviated eNOS inhibition and prevented cardiac damage following I/R injury in wild-type, but not in eNOS KO mice. Conclusion The current studies demonstrate that PYK2 is a pivotal regulator of eNOS function in myocardial infarction and identify PYK2 as a novel therapeutic target for cardioprotection.
Collapse
Affiliation(s)
- Sofia-Iris Bibli
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimioupolis, Zografou 15771, Greece.,"George P. Livanos and Marianthi Simou" Laboratories, First Department of Pulmonary and Critical Care Medicine, Evangelismos Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, Ipsilantou 45-47, Athens, 10675 Greece.,Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, Frankfurt am Main, D-60590, Germany
| | - Zongmin Zhou
- "George P. Livanos and Marianthi Simou" Laboratories, First Department of Pulmonary and Critical Care Medicine, Evangelismos Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, Ipsilantou 45-47, Athens, 10675 Greece
| | - Sven Zukunft
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, Frankfurt am Main, D-60590, Germany
| | - Beate Fisslthaler
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, Frankfurt am Main, D-60590, Germany
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimioupolis, Zografou 15771, Greece
| | - Csaba Szabo
- Department of Anesthesiology, University of Texas Medical Branch, 601 Harborside Drive, Galveston, 77555 TX, USA
| | - Peter Brouckaert
- Department of Biomedical Molecular Biology, Ghent University, Ghent B-9052, Belgium.,Laboratory for Molecular Pathology and Experimental Therapy, Inflammation Research Center, VIB, Ghent B-9052, Belgium
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, Frankfurt am Main, D-60590, Germany
| | - Andreas Papapetropoulos
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimioupolis, Zografou 15771, Greece.,"George P. Livanos and Marianthi Simou" Laboratories, First Department of Pulmonary and Critical Care Medicine, Evangelismos Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, Ipsilantou 45-47, Athens, 10675 Greece.,Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation of the Academy of Athens, Soranou Ephessiou 4, Athens 11527, Greece
| |
Collapse
|
23
|
Possible role of mitochondrial K-ATP channel and nitric oxide in protection of the neonatal rat heart. Mol Cell Biochem 2018; 450:35-42. [PMID: 29802596 PMCID: PMC6328520 DOI: 10.1007/s11010-018-3370-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 05/17/2018] [Indexed: 12/20/2022]
Abstract
Cardioprotective effect of ischemic preconditioning (IPC) and ischemic postconditioning (IPoC) in adult hearts is mediated by mitochondrial-K-ATP channels and nitric oxide (NO). During early developmental period, rat hearts exhibit higher resistance to ischemia–reperfusion (I/R) injury and their resistance cannot be further increased by IPC or IPoC. Therefore, we have speculated, whether mechanisms responsible for high resistance of neonatal heart may be similar to those of IPC and IPoC. To test this hypothesis, rat hearts isolated on days 1, 4, 7, and 10 of postnatal life were perfused according to Langendorff. Developed force (DF) of contraction was measured. Hearts were exposed to 40 min of global ischemia followed by reperfusion up to the maximum recovery of DF. IPoC was induced by 5 cycles of 10-s ischemia. Mito-K-ATP blocker (5-HD) was administered 5 min before ischemia and during first 20 min of reperfusion. Another group of hearts was isolated for biochemical analysis of 3-nitrotyrosine, and serum samples were taken to measure nitrate levels. Tolerance to ischemia did not change from day 1 to day 4 but decreased on days 7 and 10. 5-HD had no effect either on neonatal resistance to I/R injury or on cardioprotective effect of IPoC on day 10. Significant difference was found in serum nitrate levels between days 1 and 10 but not in tissue 3-nitrotyrosine content. It can be concluded that while there appears to be significant difference of NO production, mito-K-ATP and ROS probably do not play role in the high neonatal resistance to I/R injury.
Collapse
|
24
|
cGMP at the centre of attention: emerging strategies for activating the cardioprotective PKG pathway. Basic Res Cardiol 2018; 113:24. [PMID: 29766323 PMCID: PMC5954070 DOI: 10.1007/s00395-018-0679-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 04/04/2018] [Indexed: 12/22/2022]
Abstract
The nitric oxide (NO)-protein kinase G (PKG) pathway has been known for some time to be an important target for cardioprotection against ischaemia/reperfusion injury and heart failure. While many approaches for reducing infarct size in patients have failed in the past, the advent of novel drugs that modulate cGMP and its downstream targets shows very promising results in recent preclinical and clinical studies. Here, we review main aspects of the NO-PKG pathway in light of recent drug development and summarise potential cardioprotective strategies in which cGMP is the main player.
Collapse
|
25
|
Hentia C, Rizzato A, Camporesi E, Yang Z, Muntean DM, Săndesc D, Bosco G. An overview of protective strategies against ischemia/reperfusion injury: The role of hyperbaric oxygen preconditioning. Brain Behav 2018; 8:e00959. [PMID: 29761012 PMCID: PMC5943756 DOI: 10.1002/brb3.959] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 02/12/2018] [Accepted: 02/18/2018] [Indexed: 12/23/2022] Open
Abstract
INTRODUCTION Ischemia/reperfusion (I/R) injury, such as myocardial infarction, stroke, and peripheral vascular disease, has been recognized as the most frequent causes of devastating disorders and death currently. Protective effect of various preconditioning stimuli, including hyperbaric oxygen (HBO), has been proposed in the management of I/R. METHODS In this study, we searched and reviewed up-to-date published papers to explore the pathophysiology of I/R injury and to understand the mechanisms underlying the protective effect of HBO as conditioning strategy. RESULTS Animal study and clinic observation support the notion that HBO therapy and conditioning provide beneficial effect against the deleterious effects of postischemic reperfusion. Several explanations have been proposed. The first likely mechanism may be that HBO counteracts hypoxia and reduces I/R injury by improving oxygen delivery to an area with diminished blood flow. Secondly, by reducing hypoxia-ischemia, HBO reduces all the pathological events as a consequence of hypoxia, including tissue edema, increased affective area permeability, postischemia derangement of tissue metabolism, and inflammation. Thirdly, HBO may directly affect cell apoptosis, signal transduction, and gene expression in those that are sensitive to oxygen or hypoxia. HBO provides a reservoir of oxygen at cellular level not only carried by blood, but also by diffusion from the interstitial tissue where it reaches high concentration that may last for several hours, improves endothelial function and rheology, and decreases local inflammation and edema. CONCLUSION Evidence suggests the benefits of HBO when used as a preconditioning stimulus in the setting of I/R injury. Translating the beneficial effects of HBO into current practice requires, as for the "conditioning strategies", a thorough consideration of risk factors, comorbidities, and comedications that could interfere with HBO-related protection.
Collapse
Affiliation(s)
- Ciprian Hentia
- Master II level in Hyperbaric Medicine Department of Biomedical Sciences University of Padova Padova Italy.,Faculty of Medicine "Victor Babeș" University of Medicine and Pharmacy Timișoara Romania
| | - Alex Rizzato
- Master II level in Hyperbaric Medicine Department of Biomedical Sciences University of Padova Padova Italy
| | | | - Zhongjin Yang
- The Institute for Human Performance SUNY Upstate Medical University Syracuse NY USA
| | - Danina M Muntean
- Faculty of Medicine "Victor Babeș" University of Medicine and Pharmacy Timișoara Romania.,Center for Translational Research and Systems Medicine "Victor Babeș" University of Medicine and Pharmacy Timișoara Romania
| | - Dorel Săndesc
- Faculty of Medicine "Victor Babeș" University of Medicine and Pharmacy Timișoara Romania
| | - Gerardo Bosco
- Master II level in Hyperbaric Medicine Department of Biomedical Sciences University of Padova Padova Italy
| |
Collapse
|
26
|
Singh L, Virdi JK, Maslov LN, Singh N, Jaggi AS. Investigating the possible mechanisms involved in adenosine preconditioning-induced cardioprotection in rats. Cardiovasc Ther 2018; 36:e12328. [PMID: 29604187 DOI: 10.1111/1755-5922.12328] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 02/23/2018] [Accepted: 03/24/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Adenosine is a breakdown product of adenosine triphosphate and plays an important role in pharmacological preconditioning. The cardioprotective effects of adenosine preconditioning are well established. However, the possible mechanisms need to be explored. AIM This study was aimed to investigate the possible mechanisms involved in adenosine preconditioning-induced cardioprotection in rats. METHODS Rat heart was isolated and perfused on Langendorff apparatus. Global ischemia for 30 minutes followed by reperfusion for 120 minutes was employed to produce myocardial injury. Myocardial injury was assessed by measuring myocardial infarct size, release of lactate dehydrogenase (LDH) and creatine kinase (CK) in the coronary effluent and hemodynamic parameters including left ventricular developed pressure (LVDP), dp/dtmax, and dp/dtmin . Serum nitrite levels were measured as an index of nitric oxide release in blood. RESULTS Adenosine (4 mg/kg) preconditioning significantly decreased ischemia-reperfusion-induced increase in LDH, CK release, infarct size, improved LVDP, dp/dtmax and dp/dtmin, and increased serum nitrite levels. Pretreatment with L-NAME, a specific NOS inhibitor, (5 mg/kg) and montelukast, leukotriene receptor antagonist, (10 mg/kg) significantly abrogated the cardioprotective effect of adenosine preconditioning. However, seratrodast, thromboxane A2 antagonist, (15 mg/kg) had no effect on adenosine-induced cardioprotection. Sodium nitroprusside (SNP) preconditioning also produced cardioprotective effects. However, caffeine (20 mg/kg) (adenosine receptor blocker) and seratrodast (15 mg/kg) had no effect on SNP-induced cardioprotection. Administration of montelukast abrogated the cardioprotective effects of SNP preconditioning-induced cardioprotection. CONCLUSION Adenosine preconditioning may increase the release of nitric oxide, which in turn may increase the release of cysteinyl leukotrienes to confer cardioprotection.
Collapse
Affiliation(s)
- Lovedeep Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Jasleen K Virdi
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Leonid N Maslov
- Laboratory of Experimental Cardiology, Federal State Budgetary Scientific Institution, Research Institute for Cardiology, Tomsk, Russia
| | - Nirmal Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Amteshwar S Jaggi
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| |
Collapse
|
27
|
Wang SB, Venkatraman V, Crowgey EL, Liu T, Fu Z, Holewinski R, Ranek M, Kass DA, O'Rourke B, Van Eyk JE. Protein S-Nitrosylation Controls Glycogen Synthase Kinase 3β Function Independent of Its Phosphorylation State. Circ Res 2018; 122:1517-1531. [PMID: 29563102 DOI: 10.1161/circresaha.118.312789] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 03/13/2018] [Accepted: 03/19/2018] [Indexed: 01/11/2023]
Abstract
RATIONALE GSK-3β (glycogen synthase kinase 3β) is a multifunctional and constitutively active kinase known to regulate a myriad of cellular processes. The primary mechanism to regulate its function is through phosphorylation-dependent inhibition at serine-9 residue. Emerging evidence indicates that there may be alternative mechanisms that control GSK-3β for certain functions. OBJECTIVES Here, we sought to understand the role of protein S-nitrosylation (SNO) on the function of GSK-3β. SNO-dependent modulation of the localization of GSK-3β and its ability to phosphorylate downstream targets was investigated in vitro, and the network of proteins differentially impacted by phospho- or SNO-dependent GSK-3β regulation and in vivo SNO modification of key signaling kinases during the development of heart failure was also studied. METHODS AND RESULTS We found that GSK-3β undergoes site-specific SNO both in vitro, in HEK293 cells, H9C2 myoblasts, and primary neonatal rat ventricular myocytes, as well as in vivo, in hearts from an animal model of heart failure and sudden cardiac death. S-nitrosylation of GSK-3β significantly inhibits its kinase activity independent of the canonical phospho-inhibition pathway. S-nitrosylation of GSK-3β promotes its nuclear translocation and access to novel downstream phosphosubstrates which are enriched for a novel amino acid consensus sequence motif. Quantitative phosphoproteomics pathway analysis reveals that nuclear GSK-3β plays a central role in cell cycle control, RNA splicing, and DNA damage response. CONCLUSIONS The results indicate that SNO has a differential effect on the location and activity of GSK-3β in the cytoplasm versus the nucleus. SNO modification of GSK-3β occurs in vivo and could contribute to the pathobiology of heart failure and sudden cardiac death.
Collapse
Affiliation(s)
- Sheng-Bing Wang
- From the Department of Medicine (S.-B.W., V.V., T.L., R.H., M.R., D.A.K., B.O'R., J.E.V.E.)
| | - Vidya Venkatraman
- From the Department of Medicine (S.-B.W., V.V., T.L., R.H., M.R., D.A.K., B.O'R., J.E.V.E.).,Johns Hopkins University, Baltimore, MD; Department of Medicine, Advanced Clinical Biosystems Research Institute, The Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA (V.V., R.H., J.E.V.E.)
| | - Erin L Crowgey
- Department of Nemours Biomedical Research, Nemours Alfred I. duPont Hospital for Children, Wilmington, DE (E.L.C.)
| | - Ting Liu
- From the Department of Medicine (S.-B.W., V.V., T.L., R.H., M.R., D.A.K., B.O'R., J.E.V.E.)
| | | | - Ronald Holewinski
- From the Department of Medicine (S.-B.W., V.V., T.L., R.H., M.R., D.A.K., B.O'R., J.E.V.E.).,Johns Hopkins University, Baltimore, MD; Department of Medicine, Advanced Clinical Biosystems Research Institute, The Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA (V.V., R.H., J.E.V.E.)
| | - Mark Ranek
- From the Department of Medicine (S.-B.W., V.V., T.L., R.H., M.R., D.A.K., B.O'R., J.E.V.E.)
| | - David A Kass
- From the Department of Medicine (S.-B.W., V.V., T.L., R.H., M.R., D.A.K., B.O'R., J.E.V.E.)
| | - Brian O'Rourke
- From the Department of Medicine (S.-B.W., V.V., T.L., R.H., M.R., D.A.K., B.O'R., J.E.V.E.)
| | - Jennifer E Van Eyk
- From the Department of Medicine (S.-B.W., V.V., T.L., R.H., M.R., D.A.K., B.O'R., J.E.V.E.) .,Johns Hopkins University, Baltimore, MD; Department of Medicine, Advanced Clinical Biosystems Research Institute, The Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA (V.V., R.H., J.E.V.E.)
| |
Collapse
|
28
|
Shao Q, Casin KM, Mackowski N, Murphy E, Steenbergen C, Kohr MJ. Adenosine A1 receptor activation increases myocardial protein S-nitrosothiols and elicits protection from ischemia-reperfusion injury in male and female hearts. PLoS One 2017; 12:e0177315. [PMID: 28493997 PMCID: PMC5426678 DOI: 10.1371/journal.pone.0177315] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 04/25/2017] [Indexed: 11/18/2022] Open
Abstract
Nitric oxide (NO) plays an important role in cardioprotection, and recent work from our group and others has implicated protein S-nitrosylation (SNO) as a critical component of NO-mediated protection in different models, including ischemic pre- and post-conditioning and sex-dependent cardioprotection. However, studies have yet to examine whether protein SNO levels are similarly increased with pharmacologic preconditioning in male and female hearts, and whether an increase in protein SNO levels, which is protective in male hearts, is sufficient to increase baseline protection in female hearts. Therefore, we pharmacologically preconditioned male and female hearts with the adenosine A1 receptor agonist N6-cyclohexyl adenosine (CHA). CHA administration prior to ischemia significantly improved functional recovery in both male and female hearts compared to baseline in a Langendorff-perfused heart model of ischemia-reperfusion injury (% of preischemic function ± SE: male baseline: 37.5±3.4% vs. male CHA: 55.3±3.2%; female baseline: 61.4±5.7% vs. female CHA: 76.0±6.2%). In a separate set of hearts, we found that CHA increased p-Akt and p-eNOS levels. We also used SNO-resin-assisted capture with LC-MS/MS to identify SNO proteins in male and female hearts, and determined that CHA perfusion induced a modest increase in protein SNO levels in both male (11.4%) and female (12.3%) hearts compared to baseline. These findings support a potential role for protein SNO in a model of pharmacologic preconditioning, and provide evidence to suggest that a modest increase in protein SNO levels is sufficient to protect both male and female hearts from ischemic injury. In addition, a number of the SNO proteins identified with CHA treatment were also observed with other forms of cardioprotective stimuli in prior studies, further supporting a role for protein SNO in cardioprotection.
Collapse
Affiliation(s)
- Qin Shao
- Department of Cardiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Kevin M. Casin
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Nathan Mackowski
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Elizabeth Murphy
- Systems Biology Center, National Heart, Lung and Blood Institute, Bethesda, Maryland, United States of America
| | - Charles Steenbergen
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Mark J. Kohr
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
29
|
Xie F, Rong B, Wang TC, Hao L, Lin MJ, Zhong JQ. Interaction between nitric oxide signaling and gap junctions during ischemic preconditioning: Importance of S-nitrosylation vs. protein kinase G activation. Nitric Oxide 2017; 65:37-42. [PMID: 28216239 DOI: 10.1016/j.niox.2017.02.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 11/18/2016] [Accepted: 02/03/2017] [Indexed: 12/13/2022]
Abstract
Much effort has been dedicated to exploring the mechanisms of IPC, and the GJ is one of the proposed targets of IPC. Several lines of evidence have indicated that NO affects GJ permeability regulation and expression of connexin isoforms. NO-induced stimulation of the sGC-cGMP pathway and the subsequent PKG activation could lead directly to connexin phosphorylation and GJ coupling modification. Additionally, because NO-induced cardioprotection against I/R injury beyond the cGMP/PKG-dependent pathway has been reported in isolated cardiomyocytes, it has been posited that NO-mediated GJ coupling might be independent from the activation of the NO-induced cGMP/PKG pathway during IPC. S-nitrosylation by NO exerts a major influence in IPC-induced cardioprotection. It has been suggested that NO-mediated cardioprotection during IPC was not dependent on sGC/cGMP/PKG but on SNO signaling. We need more researches to prove that which signaling pathway (S-nitrosylation or protein kinase G activation) is the major one modulating GJ coupling during IPC. The aim of review article is to discuss the possible signaling pathways of NO in regulating GJ during IPC.
Collapse
Affiliation(s)
- Fei Xie
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China; Emergency Department, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Bing Rong
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China; Cadre Health Department, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Tian-Cheng Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Li Hao
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China; School of Medicine, Shandong University, Jinan, China
| | - Ming-Jie Lin
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China; School of Medicine, Shandong University, Jinan, China
| | - Jing-Quan Zhong
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| |
Collapse
|
30
|
Menazza S, Sun J, Appachi S, Chambliss KL, Kim SH, Aponte A, Khan S, Katzenellenbogen JA, Katzenellenbogen BS, Shaul PW, Murphy E. Non-nuclear estrogen receptor alpha activation in endothelium reduces cardiac ischemia-reperfusion injury in mice. J Mol Cell Cardiol 2017; 107:41-51. [PMID: 28457941 DOI: 10.1016/j.yjmcc.2017.04.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 04/24/2017] [Accepted: 04/25/2017] [Indexed: 02/07/2023]
Abstract
Steroid hormone receptors including estrogen receptors (ER) classically function as ligand-regulated transcription factors. However, estrogens also elicit cellular effects through binding to extra-nuclear ER (ERα, ERβ, and G protein-coupled ER or GPER) that are coupled to kinases. How extra-nuclear ER actions impact cardiac ischemia-reperfusion (I/R) injury is unknown. We treated ovariectomized wild-type female mice with estradiol or an estrogen-dendrimer conjugate (EDC), which selectively activates extra-nuclear ER, or vehicle interventions for two weeks. I/R injury was then evaluated in isolated Langendorff perfused hearts. Two weeks of treatment with estradiol significantly decreased infarct size and improved post-ischemic contractile function. Similarly, EDC treatment significantly decreased infarct size and increased post-ischemic functional recovery compared to vehicle-treated hearts. EDC also caused an increase in myocardial protein S-nitrosylation, consistent with previous studies showing a role for this post-translational modification in cardioprotection. In further support of a role for S-nitrosylation, inhibition of nitric oxide synthase, but not soluble guanylyl cyclase blocked the EDC mediated protection. The administration of ICI182,780, which is an agonist of G-protein coupled estrogen receptor (GPER) and an antagonist of ERα and ERβ, did not result in protection; however, ICI182,780 significantly blocked EDC-mediated cardioprotection, indicating participation of ERα and/or ERβ. In studies determining the specific ER subtype and cellular target involved, EDC decreased infarct size and improved functional recovery in mice lacking ERα in cardiomyocytes. In contrast, protection was lost in mice deficient in endothelial cell ERα. Thus, extra-nuclear ERα activation in endothelium reduces cardiac I/R injury in mice, and this likely entails increased protein S-nitrosylation. Since EDC does not stimulate uterine growth, in the clinical setting EDC-like compounds may provide myocardial protection without undesired uterotrophic and cancer-promoting effects.
Collapse
Affiliation(s)
- Sara Menazza
- Systems Biology Center, National Heart Lung and Blood Institute, NIH, Bethesda, MD, United States
| | - Junhui Sun
- Systems Biology Center, National Heart Lung and Blood Institute, NIH, Bethesda, MD, United States
| | - Swathi Appachi
- Systems Biology Center, National Heart Lung and Blood Institute, NIH, Bethesda, MD, United States
| | - Ken L Chambliss
- Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX, United States
| | - Sung Hoon Kim
- Department of Molecular and Integrative Physiology, United States
| | - Angel Aponte
- Proteomics Core, NHLBI, NIH, Bethesda, MD, United States
| | - Sohaib Khan
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | | | | | - Philip W Shaul
- Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX, United States
| | - Elizabeth Murphy
- Systems Biology Center, National Heart Lung and Blood Institute, NIH, Bethesda, MD, United States.
| |
Collapse
|
31
|
Bystrom P, Foley N, Toledo-Pereyra L, Quesnelle K. Ischemic preconditioning modulates ROS to confer protection in liver ischemia and reperfusion. EXCLI JOURNAL 2017; 16:483-496. [PMID: 28694752 PMCID: PMC5491905 DOI: 10.17179/excli2017-166] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 03/20/2017] [Indexed: 12/24/2022]
Abstract
Ischemia reperfusion (IR) injury is a significant cause of morbidity and mortality in liver transplantation. When oxygen is reintroduced to the liver graft it initiates a cascade of molecular reactions leading to the release of reactive oxygen species (ROS) and pro-inflammatory cytokines. These soluble mediators propagate a sterile immune response to cause significant tissue damage. Ischemic preconditioning (IPC) is one method that reduces hepatocellular injury by altering the immune response and inhibiting the production of ROS. Studies quantifying the effects of IPC in humans have demonstrated an improved liver enzyme panel in patients receiving grafts pretreated with IPC as compared to patients receiving the standard of care. In our review, we explore current literature in the field in order to describe the mechanism through which IPC regulates the production of ROS and improves IR injury.
Collapse
Affiliation(s)
- Phillip Bystrom
- Western Michigan University, Homer Stryker M.D. School of Medicine Department of Biomedical Sciences
| | - Nicole Foley
- Western Michigan University, Homer Stryker M.D. School of Medicine Department of Biomedical Sciences
| | - Luis Toledo-Pereyra
- Western Michigan University, Homer Stryker M.D. School of Medicine Department of Surgery
| | - Kelly Quesnelle
- Western Michigan University, Homer Stryker M.D. School of Medicine Department of Biomedical Sciences
| |
Collapse
|
32
|
The opposing roles of NO and oxidative stress in cardiovascular disease. Pharmacol Res 2016; 116:57-69. [PMID: 27988384 DOI: 10.1016/j.phrs.2016.12.017] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 11/15/2016] [Accepted: 12/13/2016] [Indexed: 02/07/2023]
Abstract
Nitric oxide (NO) plays a pivotal role in the maintenance of cardiovascular homeostasis. A reduction in the bioavailability of endogenous NO, manifest as a decrease in the production and/or impaired signaling, is associated with many cardiovascular diseases including hypertension, atherosclerosis, stroke and heart failure. There is substantial evidence that reactive oxygen species (ROS), generated predominantly from NADPH oxidases (Nox), are responsible for the reduced NO bioavailability in vascular and cardiac pathologies. ROS can compromise NO function via a direct inactivation of NO, together with a reduction in NO synthesis and oxidation of its receptor, soluble guanylyl cyclase. Whilst nitrovasodilators are administered to compensate for the ROS-mediated loss in NO bioactivity, their clinical utility is limited due to the development of tolerance and resistance and systemic hypotension. Moreover, efforts to directly scavenge ROS with antioxidants has had limited clinical efficacy. This review outlines the therapeutic utility of NO-based therapeutics in cardiovascular diseases and describes the source and impact of ROS in these pathologies, with particular focus on the interaction with NO. Future therapeutic approaches in the treatment of cardiovascular diseases are highlighted with a focus on nitroxyl (HNO) donors as an alternative to traditional NO donors and the development of novel Nox inhibitors.
Collapse
|
33
|
Bibli SI, Andreadou I, Glynos C, Chatzianastasiou A, Toumpanakis D, Zakynthinos S, Vasilakopoulos T, Iliodromitis EK, Papapetropoulos A. Exposure to cigarette smoke abrogates the beneficial effect of ischemic postconditioning. Am J Physiol Heart Circ Physiol 2016; 311:H1321-H1332. [PMID: 27694220 DOI: 10.1152/ajpheart.00925.2015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 09/06/2016] [Indexed: 02/08/2023]
Abstract
Cigarette smoking is one of the risk factors for coronary artery disease. Although conditioning decreases infarct size in hearts from healthy animals, comorbidities may render it ineffective. We investigated the effects of cigarette smoke (CS) exposure on intracellular myocardial signaling, infarct size after ischemia-reperfusion, and the potential interference with ischemic conditioning. Exposure of mice to CS increased blood pressure, caused cardiac hypertrophy, and upregulated the nitric oxide synthatse (NOS)/soluble guanylate cyclase (sGC)/cGMP pathway. To test the effect of CS exposure on the endogenous cardioprotective mechanisms, mice were subjected to regional myocardial ischemia and reperfusion with no further intervention or application of preconditioning (PreC) or postconditioning (PostC). Exposure to CS did not increase the infarction compared with the room air (RA)-exposed group. PreC was beneficial for both CS and RA vs. nonconditioned animals. PostC was effective only in RA animals, while the infarct size-limiting effect was not preserved in the CS group. Differences in oxidative stress markers, Akt, and endothelial NOS phosphorylation and cGMP levels were observed between RA and CS groups subjected to PostC. In conclusion, exposure to CS does not per se increase infarct size. The beneficial effect of ischemic PreC is preserved in mice exposed to CS, as it does not affect the cardioprotective signaling; in contrast, PostC fails to protect CS-exposed mice due to impaired activation of the Akt/eNOS/cGMP axis that occurs in parallel to enhanced oxidative stress.
Collapse
Affiliation(s)
- Sofia-Iris Bibli
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
- "George P. Livanos and Marianthi Simou Laboratories," 1st Department of Pulmonary and Critical Care Medicine, Evangelismos Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece; and
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Constantinos Glynos
- "George P. Livanos and Marianthi Simou Laboratories," 1st Department of Pulmonary and Critical Care Medicine, Evangelismos Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece; and
| | - Athanasia Chatzianastasiou
- "George P. Livanos and Marianthi Simou Laboratories," 1st Department of Pulmonary and Critical Care Medicine, Evangelismos Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece; and
| | - Dimitris Toumpanakis
- "George P. Livanos and Marianthi Simou Laboratories," 1st Department of Pulmonary and Critical Care Medicine, Evangelismos Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece; and
| | - Spyros Zakynthinos
- "George P. Livanos and Marianthi Simou Laboratories," 1st Department of Pulmonary and Critical Care Medicine, Evangelismos Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece; and
| | - Theodoros Vasilakopoulos
- "George P. Livanos and Marianthi Simou Laboratories," 1st Department of Pulmonary and Critical Care Medicine, Evangelismos Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece; and
| | - Efstathios K Iliodromitis
- Faculty of Medicine, 2nd Department of Cardiology, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Andreas Papapetropoulos
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece;
- "George P. Livanos and Marianthi Simou Laboratories," 1st Department of Pulmonary and Critical Care Medicine, Evangelismos Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece; and
| |
Collapse
|
34
|
Dezfulian C, Kenny E, Lamade A, Misse A, Krehel N, St Croix C, Kelley EE, Jackson TC, Uray T, Rackley J, Kochanek PM, Clark RSB, Bayir H. Mechanistic characterization of nitrite-mediated neuroprotection after experimental cardiac arrest. J Neurochem 2016; 139:419-431. [PMID: 27507435 DOI: 10.1111/jnc.13764] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 08/04/2016] [Accepted: 08/05/2016] [Indexed: 12/27/2022]
Abstract
Nitrite acts as an ischemic reservoir of nitric oxide (NO) and a potent S-nitrosating agent which reduced histologic brain injury after rat asphyxial cardiac arrest (ACA). The mechanism(s) of nitrite-mediated neuroprotection remain to be defined. We hypothesized that nitrite-mediated brain mitochondrial S-nitrosation accounts for neuroprotection by reducing reperfusion reactive oxygen species (ROS) generation. Nitrite (4 μmol) or placebo was infused IV after normothermic (37°C) ACA in randomized, blinded fashion with evaluation of neurologic function, survival, brain mitochondrial function, and ROS. Blood and CSF nitrite were quantified using reductive chemiluminescence and S-nitrosation by biotin switch. Direct neuroprotection was verified in vitro after 1 and 4 h neuronal oxygen glucose deprivation measuring neuronal death with inhibition studies to examine mechanism. Mitochondrial ROS generation was quantified by live neuronal imaging using mitoSOX. Nitrite significantly reduced neurologic disability after ACA. ROS generation was reduced in brain mitochondria from nitrite- versus placebo-treated rats after ACA with congruent preservation of brain ascorbate and reduction of ROS in brain sections using immuno-spin trapping. ATP generation was maintained with nitrite up to 24 h after ACA. Nitrite rapidly entered CSF and increased brain mitochondrial S-nitrosation. Nitrite reduced in vitro mitochondrial superoxide generation and improved survival of neurons after oxygen glucose deprivation. Protection was maintained with inhibition of soluble guanylate cyclase but lost with NO scavenging and ultraviolet irradiation. Nitrite therapy results in direct neuroprotection from ACA mediated by reductions in brain mitochondrial ROS in association with protein S-nitrosation. Neuroprotection is dependent on NO and S-nitrosothiol generation, not soluble guanylate cyclase.
Collapse
Affiliation(s)
- Cameron Dezfulian
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA. .,Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA. .,Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
| | - Elizabeth Kenny
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Andrew Lamade
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Amalea Misse
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Nicholas Krehel
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Claudette St Croix
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Eric E Kelley
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Travis C Jackson
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Thomas Uray
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Justin Rackley
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Patrick M Kochanek
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Robert S B Clark
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Hulya Bayir
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Department of Environmental and Occupational Health, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
35
|
Biasutto L, Azzolini M, Szabò I, Zoratti M. The mitochondrial permeability transition pore in AD 2016: An update. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1863:2515-30. [PMID: 26902508 DOI: 10.1016/j.bbamcr.2016.02.012] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 02/04/2016] [Accepted: 02/05/2016] [Indexed: 12/13/2022]
Abstract
Over the past 30years the mitochondrial permeability transition - the permeabilization of the inner mitochondrial membrane due to the opening of a wide pore - has progressed from being considered a curious artifact induced in isolated mitochondria by Ca(2+) and phosphate to a key cell-death-inducing process in several major pathologies. Its relevance is by now universally acknowledged and a pharmacology targeting the phenomenon is being developed. The molecular nature of the pore remains to this day uncertain, but progress has recently been made with the identification of the FOF1 ATP synthase as the probable proteic substrate. Researchers sharing this conviction are however divided into two camps: these believing that only the ATP synthase dimers or oligomers can form the pore, presumably in the contact region between monomers, and those who consider that the ring-forming c subunits in the FO sector actually constitute the walls of the pore. The latest development is the emergence of a new candidate: Spastic Paraplegia 7 (SPG7), a mitochondrial AAA-type membrane protease which forms a 6-stave barrel. This review summarizes recent developments of research on the pathophysiological relevance and on the molecular nature of the mitochondrial permeability transition pore. This article is part of a Special Issue entitled: Mitochondrial Channels edited by Pierre Sonveaux, Pierre Maechler and Jean-Claude Martinou.
Collapse
Affiliation(s)
- Lucia Biasutto
- CNR Neuroscience Institute, Viale G. Colombo 3, 35121 Padova, Italy; University of Padova, Department of Biomedical Sciences, Viale G. Colombo 3, 35121 Padova, Italy
| | - Michele Azzolini
- CNR Neuroscience Institute, Viale G. Colombo 3, 35121 Padova, Italy; University of Padova, Department of Biomedical Sciences, Viale G. Colombo 3, 35121 Padova, Italy
| | - Ildikò Szabò
- CNR Neuroscience Institute, Viale G. Colombo 3, 35121 Padova, Italy; University of Padova, Department of Biology, Viale G. Colombo 3, 35121 Padova, Italy
| | - Mario Zoratti
- CNR Neuroscience Institute, Viale G. Colombo 3, 35121 Padova, Italy; University of Padova, Department of Biomedical Sciences, Viale G. Colombo 3, 35121 Padova, Italy.
| |
Collapse
|
36
|
Donato M, Goyeneche MA, Garces M, Marchini T, Pérez V, Del Mauro J, Höcht C, Rodríguez M, Evelson P, Gelpi RJ. Myocardial triggers involved in activation of remote ischaemic preconditioning. Exp Physiol 2016; 101:708-16. [PMID: 27028009 DOI: 10.1113/ep085535] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 03/22/2016] [Indexed: 12/15/2022]
Abstract
NEW FINDINGS What is the central question of this study? Ischaemia-reperfusion of peripheral tissues protects the heart from subsequent myocardial ischaemia-reperfusion injury, a phenomenon referred to as remote ischaemic preconditioning (rIPC). This study evaluated the possible myocardial triggers of rIPC. What is the main finding and its importance? Remote ischaemic preconditioning reduces infarct size through a vagal pathway and a mechanism involving phosphorylation of Akt and endothelial nitric oxide synthase, opening of mitochondrial ATP-dependent K(+) channels and an increase in mitochondrial H2 O2 production. All these phenomena occur before the myocardial ischaemia; hence, they could act as 'triggers' of rIPC. It has been proposed that remote ischaemic preconditioning (rIPC) activates a parasympathetic neural pathway. However, the myocardial intracellular mechanism of rIPC remains unclear. Here, we characterized some of the intracellular signals participating as rIPC triggers. Isolated rat hearts were subjected to 30 min of global ischaemia and 120 min of reperfusion (Non-rIPC group). In a second group, before the isolation of the heart, an rIPC protocol (three cycles of hindlimb ischaemia-reperfusion) was performed. The infarct size was measured with tetrazolium staining. Expression/phosphorylation of Akt and endothelial nitric oxide synthase (eNOS) and mitochondrial H2 O2 production were evaluated at the end of the rIPC protocol, before myocardial ischaemia-reperfusion. The rIPC significantly decreased the infarct size and induced Akt and eNOS phosphorylation. The protective effect on infarct size was abolished by cervical vagal section, l-NAME (an NO synthesis inhibitor) and 5-hydroxydecanoate (a mitochondrial ATP-dependent K(+) channel blocker). Mitochondrial production of H2 O2 was increased by rIPC, whereas it was abolished by cervical vagal section, l-NAME and 5-hydroxydecanoate. We conclude that rIPC activates a parasympathetic vagal pathway and a mechanism involving the phosphorylation of Akt and eNOS, the opening of mitochondrial ATP-dependent K(+) channels and the release of H2 O2 by the mitochondria. All these phenomena occur before myocardial ischaemia and could act as triggers of rIPC.
Collapse
Affiliation(s)
- Martín Donato
- Institute of Cardiovascular Pathophysiology (INFICA), Department of Pathology, Faculty of Medicine, University of Buenos Aires, Argentina.,Institute of Biochemistry and Molecular Medicine (IBIMOL, UBA-CONICET), Faculty of Medicine, University of Buenos Aires, Argentina
| | - María A Goyeneche
- Institute of Cardiovascular Pathophysiology (INFICA), Department of Pathology, Faculty of Medicine, University of Buenos Aires, Argentina
| | - Mariana Garces
- Institute of Biochemistry and Molecular Medicine (IBIMOL, UBA-CONICET), Faculty of Pharmacy and Biochemistry, University of Buenos Aires, Argentina
| | - Timoteo Marchini
- Institute of Biochemistry and Molecular Medicine (IBIMOL, UBA-CONICET), Faculty of Pharmacy and Biochemistry, University of Buenos Aires, Argentina
| | - Virginia Pérez
- Institute of Cardiovascular Pathophysiology (INFICA), Department of Pathology, Faculty of Medicine, University of Buenos Aires, Argentina.,Institute of Biochemistry and Molecular Medicine (IBIMOL, UBA-CONICET), Faculty of Medicine, University of Buenos Aires, Argentina
| | - Julieta Del Mauro
- Department of Pharmacology, Faculty of Pharmacy and Biochemistry, University of Buenos Aires, Argentina
| | - Christian Höcht
- Department of Pharmacology, Faculty of Pharmacy and Biochemistry, University of Buenos Aires, Argentina
| | - Manuel Rodríguez
- Institute of Cardiovascular Pathophysiology (INFICA), Department of Pathology, Faculty of Medicine, University of Buenos Aires, Argentina.,Institute of Biochemistry and Molecular Medicine (IBIMOL, UBA-CONICET), Faculty of Medicine, University of Buenos Aires, Argentina
| | - Pablo Evelson
- Institute of Biochemistry and Molecular Medicine (IBIMOL, UBA-CONICET), Faculty of Pharmacy and Biochemistry, University of Buenos Aires, Argentina
| | - Ricardo J Gelpi
- Institute of Cardiovascular Pathophysiology (INFICA), Department of Pathology, Faculty of Medicine, University of Buenos Aires, Argentina.,Institute of Biochemistry and Molecular Medicine (IBIMOL, UBA-CONICET), Faculty of Medicine, University of Buenos Aires, Argentina
| |
Collapse
|
37
|
Correa F, Buelna-Chontal M, Chagoya V, García-Rivas G, Vigueras RM, Pedraza-Chaverri J, García-Niño WR, Hernández-Pando R, León-Contreras JC, Zazueta C. Inhibition of the nitric oxide/cyclic guanosine monophosphate pathway limited the cardioprotective effect of post-conditioning in hearts with apical myocardial infarction. Eur J Pharmacol 2015; 765:472-81. [PMID: 26387613 DOI: 10.1016/j.ejphar.2015.09.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 09/09/2015] [Accepted: 09/14/2015] [Indexed: 11/19/2022]
Abstract
Reperfusion damage involves opening of the mitochondrial permeability transition pore (mPTP) and loss of ATP synthesis. Several cardioprotective pathways are activated by ischemic or pharmacological post-conditioning (PC). The mechanisms that are activated by PC in no co-morbidity murine models include: activation of rescue kinases, oxidative stress reduction, glycolytic flux regulation and preservation of ATP synthesis. However, relatively scarce efforts have been made to define whether the efficacy of PC signaling is blunted by risk factors or systemic diseases associated with ischemic heart pathology. Experimental evidence has shown that the nitric oxide (NO)/cyclic guanosine monophosphate (cGMP) signaling is a main mechanism activated by PC in hearts without pathological history. In this work we evaluated the participation of the NO pathway, through downstream kinase activation and inhibition of mPTP in hearts with previous infarct. Myocardial infarction was induced with a single dose of isoproterenol (85 mg/kg i.p.) to male Wistar rats. After 24 h, the hearts were mounted into the Langendorff system and subjected to 30 min of ischemia and 60 min of reperfusion. PC consisted of 5 cycles of 30 s of reperfusion/30 s of ischemia, then the hearts were reperfused with or without inhibitors of the NO/cGMP pathway. PC activates the NO/cGMP pathway, as increased cGMP and NO levels were detected in isoproterenol-treated hearts. The cardioprotective effect of PC was abolished with both L-NAME (inhibitor of constitutive NO synthase) and ODQ (inhibitor of soluble guanylate cyclase), whereas the NO donor (DETA-NO) restored cardioprotection even in the presence of L-NAME or ODQ. We also found that mitochondrial structure and function was preserved in PC hearts. We conclude that PC exerts cardioprotection in hearts with previous infarct by maintaining mitochondrial structure and function through NO-dependent pathway.
Collapse
Affiliation(s)
- Francisco Correa
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología, I. Ch., Juan Badiano No. 1., Col. Sección XVI, México D.F. 14080, Mexico.
| | - Mabel Buelna-Chontal
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología, I. Ch., Juan Badiano No. 1., Col. Sección XVI, México D.F. 14080, Mexico
| | - Victoria Chagoya
- Departamento de Biología Celular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, D.F., Mexico
| | - Gerardo García-Rivas
- Centro de Innovación y Transferencia en Salud de la Escuela de Medicina del Tecnológico de Monterrey, Cátedra de Cardiología y Medicina Vascular, Instituto de Cardiología y Medicina Vascular del Tecnológico de Monterrey, Monterrey, N.L. 64710, Mexico
| | - Rosa María Vigueras
- Laboratorio de Histomorfología, Torre de Investigación, Instituto Nacional de Pediatría, SS, México D.F. 04530, Mexico
| | - José Pedraza-Chaverri
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, 04510 México DF, Mexico
| | - Wylly Ramsés García-Niño
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología, I. Ch., Juan Badiano No. 1., Col. Sección XVI, México D.F. 14080, Mexico
| | - Rogelio Hernández-Pando
- Sección de Patología Experimental, Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Av. Vasco De Quiroga 15, Tlalpan, México D.F., Mexico
| | - Juan Carlos León-Contreras
- Sección de Patología Experimental, Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Av. Vasco De Quiroga 15, Tlalpan, México D.F., Mexico
| | - Cecilia Zazueta
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología, I. Ch., Juan Badiano No. 1., Col. Sección XVI, México D.F. 14080, Mexico
| |
Collapse
|
38
|
Penna C, Angotti C, Pagliaro P. Protein S-nitrosylation in preconditioning and postconditioning. Exp Biol Med (Maywood) 2015; 239:647-62. [PMID: 24668550 DOI: 10.1177/1535370214522935] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The coronary artery disease is a leading cause of death and morbidity worldwide. This disease has a complex pathophysiology that includes multiple mechanisms. Among these is the oxidative/nitrosative stress. Paradoxically, oxidative/nitrosative signaling plays a major role in cardioprotection against ischemia/reperfusion injury. In this context, the gas transmitter nitric oxide may act through several mechanisms, such as guanylyl cyclase activation and via S-nitrosylation of proteins. The latter is a covalent modification of a protein cysteine thiol by a nitric oxide-group that generates an S-nitrosothiol. Here, we report data showing that nitric oxide and S-nitrosylation of proteins play a pivotal role not only in preconditioning but also in postconditioning cardioprotection.
Collapse
|
39
|
Abstract
Reperfusion is mandatory to salvage ischemic myocardium from infarction, but reperfusion per se contributes to injury and ultimate infarct size. Therefore, cardioprotection beyond that by timely reperfusion is needed to reduce infarct size and improve the prognosis of patients with acute myocardial infarction. The conditioning phenomena provide such cardioprotection, insofar as brief episodes of coronary occlusion/reperfusion preceding (ischemic preconditioning) or following (ischemic postconditioning) sustained myocardial ischemia with reperfusion reduce infarct size. Even ischemia/reperfusion in organs remote from the heart provides cardioprotection (remote ischemic conditioning). The present review characterizes the signal transduction underlying the conditioning phenomena, including their physical and chemical triggers, intracellular signal transduction, and effector mechanisms, notably in the mitochondria. Cardioprotective signal transduction appears as a highly concerted spatiotemporal program. Although the translation of ischemic postconditioning and remote ischemic conditioning protocols to patients with acute myocardial infarction has been fairly successful, the pharmacological recruitment of cardioprotective signaling has been largely disappointing to date.
Collapse
Affiliation(s)
- Gerd Heusch
- From the Institute for Pathophysiology, West German Heart and Vascular Centre, University of Essen Medical School, Essen, Germany.
| |
Collapse
|
40
|
Cohen MV, Downey JM. Signalling pathways and mechanisms of protection in pre- and postconditioning: historical perspective and lessons for the future. Br J Pharmacol 2015; 172:1913-32. [PMID: 25205071 PMCID: PMC4386972 DOI: 10.1111/bph.12903] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 08/22/2014] [Accepted: 08/29/2014] [Indexed: 12/19/2022] Open
Abstract
Ischaemic pre- and postconditioning are potent cardioprotective interventions that spare ischaemic myocardium and decrease infarct size after periods of myocardial ischaemia/reperfusion. They are dependent on complex signalling pathways involving ligands released from ischaemic myocardium, G-protein-linked receptors, membrane growth factor receptors, phospholipids, signalling kinases, NO, PKC and PKG, mitochondrial ATP-sensitive potassium channels, reactive oxygen species, TNF-α and sphingosine-1-phosphate. The final effector is probably the mitochondrial permeability transition pore and the signalling produces protection by preventing pore formation. Many investigators have worked to produce a roadmap of this signalling with the hope that it would reveal where one could intervene to therapeutically protect patients with acute myocardial infarction whose hearts are being reperfused. However, attempts to date to show efficacy of such an intervention in large clinical trials have been unsuccessful. Reasons for this inability to translate successes in the experimental laboratory to the clinical arena are evaluated in this review. It is suggested that all patients with acute coronary syndromes currently presenting to the hospital and being treated with platelet P2Y12 receptor antagonists, the current standard of care, are indeed already benefiting from protection from the conditioning pathways outlined earlier. If that proves to be the case, then future attempts to further decrease infarction will have to rely on interventions which protect by a different mechanism.
Collapse
Affiliation(s)
- Michael V Cohen
- Department of Physiology, University of South Alabama College of MedicineMobile, AL, USA
- Department of Medicine, University of South Alabama College of MedicineMobile, AL, USA
| | - James M Downey
- Department of Physiology, University of South Alabama College of MedicineMobile, AL, USA
| |
Collapse
|
41
|
Sun J, Nguyen T, Aponte AM, Menazza S, Kohr MJ, Roth DM, Patel HH, Murphy E, Steenbergen C. Ischaemic preconditioning preferentially increases protein S-nitrosylation in subsarcolemmal mitochondria. Cardiovasc Res 2015; 106:227-36. [PMID: 25694588 DOI: 10.1093/cvr/cvv044] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 02/08/2015] [Indexed: 12/16/2022] Open
Abstract
Nitric oxide (NO) and protein S-nitrosylation (SNO) have been shown to play important roles in ischaemic preconditioning (IPC)-induced acute cardioprotection. The majority of proteins that show increased SNO following IPC are localized to the mitochondria, and our recent studies suggest that caveolae transduce acute NO/SNO cardioprotective signalling in IPC hearts. Due to the close association between subsarcolemmal mitochondria (SSM) and the sarcolemma/caveolae, we tested the hypothesis that SSM, rather than the interfibrillar mitochondria (IFM), are major targets for NO/SNO signalling derived from caveolae-associated eNOS. Following either control perfusion or IPC, SSM and IFM were isolated from Langendorff perfused mouse hearts, and SNO was analysed using a modified biotin switch method with fluorescent maleimide fluors. In perfusion control hearts, the SNO content was higher in SSM compared with IFM (1.33 ± 0.19, ratio of SNO content Perf-SSM vs. Perf-IFM), and following IPC SNO content significantly increased preferentially in SSM, but not in IFM (1.72 ± 0.17 and 1.07 ± 0.04, ratio of SNO content IPC-SSM vs. Perf-IFM, and IPC-IFM vs. Perf-IFM, respectively). Consistent with these findings, eNOS, caveolin-3, and connexin-43 were detected in SSM, but not in IFM, and IPC resulted in a further significant increase in eNOS/caveolin-3 levels in SSM. Interestingly, we did not observe an IPC-induced increase in SNO or eNOS/caveolin-3 in SSM isolated from caveolin-3(-/-) mouse hearts, which could not be protected with IPC. In conclusion, these results suggest that SSM may be the preferential target of sarcolemmal signalling-derived post-translational protein modification (caveolae-derived eNOS/NO/SNO), thus providing an important role in IPC-induced cardioprotection.
Collapse
Affiliation(s)
- Junhui Sun
- Systems Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, 10 Center Drive, Bldg10/Rm8N206, Bethesda, MD 20892, USA
| | - Tiffany Nguyen
- Systems Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, 10 Center Drive, Bldg10/Rm8N206, Bethesda, MD 20892, USA
| | - Angel M Aponte
- Systems Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, 10 Center Drive, Bldg10/Rm8N206, Bethesda, MD 20892, USA Proteomics Core Facility, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sara Menazza
- Systems Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, 10 Center Drive, Bldg10/Rm8N206, Bethesda, MD 20892, USA
| | - Mark J Kohr
- Systems Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, 10 Center Drive, Bldg10/Rm8N206, Bethesda, MD 20892, USA Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD 21205, USA
| | - David M Roth
- Department of Anesthesiology, VA San Diego Healthcare System and University of California at San Diego, La Jolla, CA 92093, USA
| | - Hemal H Patel
- Department of Anesthesiology, VA San Diego Healthcare System and University of California at San Diego, La Jolla, CA 92093, USA
| | - Elizabeth Murphy
- Systems Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, 10 Center Drive, Bldg10/Rm8N206, Bethesda, MD 20892, USA
| | - Charles Steenbergen
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD 21205, USA
| |
Collapse
|
42
|
Pagliaro P, Penna C. Redox signalling and cardioprotection: translatability and mechanism. Br J Pharmacol 2015; 172:1974-95. [PMID: 25303224 DOI: 10.1111/bph.12975] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 09/24/2014] [Accepted: 09/30/2014] [Indexed: 12/13/2022] Open
Abstract
The morbidity and mortality from coronary artery disease (CAD) remain significant worldwide. The treatment for acute myocardial infarction has improved over the past decades, including early reperfusion of culprit coronary arteries. Although it is mandatory to reperfuse the ischaemic territory as soon as possible, paradoxically this leads to additional myocardial injury, namely ischaemia/reperfusion (I/R) injury, in which redox stress plays a pivotal role and for which no effective therapy is currently available. In this review, we report evidence that the redox environment plays a pivotal role not only in I/R injury but also in cardioprotection. In fact, cardioprotective strategies, such as pre- and post-conditioning, result in a robust reduction in infarct size in animals and the role of redox signalling is of paramount importance in these conditioning strategies. Nitrosative signalling and cysteine redox modifications, such as S-nitrosation/S-nitrosylation, are also emerging as very important mechanisms in conditioning cardioprotection. The reasons for the switch from protective oxidative/nitrosative signalling to deleterious oxidative/nitrosative/nitrative stress are not fully understood. The complex regulation of this switch is, at least in part, responsible for the diminished or lack of cardioprotection induced by conditioning protocols observed in ageing animals and with co-morbidities as well as in humans. Therefore, it is important to understand at a mechanistic level the reasons for these differences before proposing a safe and useful transition of ischaemic or pharmacological conditioning. Indeed, more mechanistic novel therapeutic strategies are required to protect the heart from I/R injury and to improve clinical outcomes in patients with CAD.
Collapse
Affiliation(s)
- P Pagliaro
- Department of Clinical and Biological Sciences, University of Torino, 10043, Orbassano, Turin, Italy
| | | |
Collapse
|
43
|
Role of phosphoinositide 3-kinase IA (PI3K-IA) activation in cardioprotection induced by ouabain preconditioning. J Mol Cell Cardiol 2015; 80:114-25. [PMID: 25575882 DOI: 10.1016/j.yjmcc.2014.12.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 12/06/2014] [Accepted: 12/26/2014] [Indexed: 11/23/2022]
Abstract
Acute myocardial infarction, the clinical manifestation of ischemia-reperfusion (IR) injury, is a leading cause of death worldwide. Like ischemic preconditioning (IPC) induced by brief episodes of ischemia and reperfusion, ouabain preconditioning (OPC) mediated by Na/K-ATPase signaling protects the heart against IR injury. Class I PI3K activation is required for IPC, but its role in OPC has not been investigated. While PI3K-IB is critical to IPC, studies have suggested that ouabain signaling is PI3K-IA-specific. Hence, a pharmacological approach was used to test the hypothesis that OPC and IPC rely on distinct PI3K-I isoforms. In Langendorff-perfused mouse hearts, OPC was initiated by 4 min of ouabain 10 μM and IPC was triggered by 4 cycles of 5 min ischemia and reperfusion prior to 40 min of global ischemia and 30 min of reperfusion. Without affecting PI3K-IB, ouabain doubled PI3K-IA activity and Akt phosphorylation at Ser(473). IPC and OPC significantly preserved cardiac contractile function and tissue viability as evidenced by left ventricular developed pressure and end-diastolic pressure recovery, reduced lactate dehydrogenase release, and decreased infarct size. OPC protection was blunted by the PI3K-IA inhibitor PI-103, but not by the PI3K-IB inhibitor AS-604850. In contrast, IPC-mediated protection was not affected by PI-103 but was blocked by AS-604850, suggesting that PI3K-IA activation is required for OPC while PI3K-IB activation is needed for IPC. Mechanistically, PI3K-IA activity is required for ouabain-induced Akt activation but not PKCε translocation. However, in contrast to PKCε translocation which is critical to protection, Akt activity was not required for OPC. Further studies shall reveal the identity of the downstream targets of this new PI3K IA-dependent branch of OPC. These findings may be of clinical relevance in patients at risk for myocardial infarction with underlying diseases and/or medication that could differentially affect the integrity of cardiac PI3K-IA and IB pathways.
Collapse
|
44
|
Farah C, Reboul C. NO Better Way to Protect the Heart during Ischemia-Reperfusion: To be in the Right Place at the Right Time. Front Pediatr 2015; 3:6. [PMID: 25705614 PMCID: PMC4319379 DOI: 10.3389/fped.2015.00006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 01/26/2015] [Indexed: 11/24/2022] Open
Affiliation(s)
- Charlotte Farah
- EA4278, LaPEC, Université d'Avignon , Avignon , France ; UMR-CNRS 9214, INSERM U1046, Université de Montpellier , Montpellier , France
| | - Cyril Reboul
- EA4278, LaPEC, Université d'Avignon , Avignon , France
| |
Collapse
|
45
|
Soetkamp D, Nguyen TT, Menazza S, Hirschhäuser C, Hendgen-Cotta UB, Rassaf T, Schlüter KD, Boengler K, Murphy E, Schulz R. S-nitrosation of mitochondrial connexin 43 regulates mitochondrial function. Basic Res Cardiol 2014; 109:433. [PMID: 25115184 PMCID: PMC4168224 DOI: 10.1007/s00395-014-0433-x] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 08/05/2014] [Accepted: 08/06/2014] [Indexed: 01/31/2023]
Abstract
S-nitrosation (SNO) of connexin 43 (Cx43)-formed channels modifies dye uptake in astrocytes and gap junctional communication in endothelial cells. Apart from forming channels at the plasma membrane of several cell types, Cx43 is also located at the inner membrane of myocardial subsarcolemmal mitochondria (SSM), but not in interfibrillar mitochondria (IFM). The absence or pharmacological blockade of mitochondrial Cx43 (mtCx43) reduces dye and potassium uptake. Lack of mtCx43 is associated with loss of endogenous cardioprotection by ischemic preconditioning (IPC), which is mediated by formation of reactive oxygen species (ROS). Whether or not mitochondrial Lucifer Yellow (LY), ion uptake, or ROS generation are affected by SNO of mtCx43 and whether or not cardioprotective interventions affect SNO of mtCx43 remains unknown. In SSM from rat hearts, application of NO donors (48 nmol to 1 mmol) increased LY uptake (0.5 mmol SNAP 38.4 ± 7.1 %, p < 0.05; 1 mmol GSNO 28.1 ± 7.4 %, p < 0.05) and the refilling rate of potassium (SNAP 227.9 ± 30.1 %, p < 0.05; GSNO 122.6 ± 28.1 %, p < 0.05). These effects were absent following blockade of Cx43 hemichannels by carbenoxolone as well as in IFM lacking Cx43. Unlike potassium, the sodium permeability was not affected by application of NO. Furthermore, mitochondrial ROS formation was increased following NO application compared to control SSM (0.5 mmol SNAP 22.9 ± 1.8 %, p < 0.05; 1 mmol GSNO 40.6 ± 7.1 %, p < 0.05), but decreased in NO treated IFM compared to control (0.5 mmol SNAP 14.4 ± 4 %, p < 0.05; 1 mmol GSNO 13.8 ± 4 %, p < 0.05). NO donor administration to isolated SSM increased SNO of mtCx43 by 109.2 ± 15.8 %. Nitrite application (48 nmol) to mice was also associated with elevated SNO of mtCx43 by 59.3 ± 18.2 % (p < 0.05). IPC by four cycles of 5 min of ischemia and 5 min of reperfusion increased SNO of mtCx43 by 41.6 ± 1.7 % (p < 0.05) when compared to control perfused rat hearts. These data suggest that SNO of mtCx43 increases mitochondrial permeability, especially for potassium and leads to increased ROS formation. The increased amount of SNO mtCx43 by IPC or nitrite administration may link NO and Cx43 in the signal transduction cascade of cardioprotective interventions.
Collapse
Affiliation(s)
- Daniel Soetkamp
- Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Williams D, Venardos KM, Byrne M, Joshi M, Horlock D, Lam NT, Gregorevic P, McGee SL, Kaye DM. Abnormal mitochondrial L-arginine transport contributes to the pathogenesis of heart failure and rexoygenation injury. PLoS One 2014; 9:e104643. [PMID: 25111602 PMCID: PMC4128716 DOI: 10.1371/journal.pone.0104643] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 07/11/2014] [Indexed: 11/29/2022] Open
Abstract
Background Impaired mitochondrial function is fundamental feature of heart failure (HF) and myocardial ischemia. In addition to the effects of heightened oxidative stress, altered nitric oxide (NO) metabolism, generated by a mitochondrial NO synthase, has also been proposed to impact upon mitochondrial function. However, the mechanism responsible for arginine transport into mitochondria and the effect of HF on such a process is unknown. We therefore aimed to characterize mitochondrial L-arginine transport and to investigate the hypothesis that impaired mitochondrial L-arginine transport plays a key role in the pathogenesis of heart failure and myocardial injury. Methods and Results In mitochondria isolated from failing hearts (sheep rapid pacing model and mouse Mst1 transgenic model) we demonstrated a marked reduction in L-arginine uptake (p<0.05 and p<0.01 respectively) and expression of the principal L-arginine transporter, CAT-1 (p<0.001, p<0.01) compared to controls. This was accompanied by significantly lower NO production and higher 3-nitrotyrosine levels (both p<0.05). The role of mitochondrial L-arginine transport in modulating cardiac stress responses was examined in cardiomyocytes with mitochondrial specific overexpression of CAT-1 (mtCAT1) exposed to hypoxia-reoxygenation stress. mtCAT1 cardiomyocytes had significantly improved mitochondrial membrane potential, respiration and ATP turnover together with significantly decreased reactive oxygen species production and cell death following mitochondrial stress. Conclusion These data provide new insights into the role of L-arginine transport in mitochondrial biology and cardiovascular disease. Augmentation of mitochondrial L-arginine availability may be a novel therapeutic strategy for myocardial disorders involving mitochondrial stress such as heart failure and reperfusion injury.
Collapse
Affiliation(s)
- David Williams
- Heart Failure Research Group, Baker IDI Heart & Diabetes Institute, Melbourne, Australia
| | - Kylie M. Venardos
- Heart Failure Research Group, Baker IDI Heart & Diabetes Institute, Melbourne, Australia
- Department of Medicine, Monash University, Melbourne, Australia
| | - Melissa Byrne
- Heart Failure Research Group, Baker IDI Heart & Diabetes Institute, Melbourne, Australia
| | - Mandar Joshi
- Heart Failure Research Group, Baker IDI Heart & Diabetes Institute, Melbourne, Australia
| | - Duncan Horlock
- Heart Failure Research Group, Baker IDI Heart & Diabetes Institute, Melbourne, Australia
| | - Nicholas T. Lam
- Heart Failure Research Group, Baker IDI Heart & Diabetes Institute, Melbourne, Australia
| | - Paul Gregorevic
- Muscle Research & Therapeutics Laboratory, Baker IDI Heart & Diabetes Institute, Melbourne, Australia
| | - Sean L. McGee
- Metabolic Research Unit, Deakin University, Geelong, Australia
| | - David M. Kaye
- Heart Failure Research Group, Baker IDI Heart & Diabetes Institute, Melbourne, Australia
- Department of Medicine, Monash University, Melbourne, Australia
- * E-mail:
| |
Collapse
|
47
|
Satohisa S, Zhang HH, Feng L, Yang YY, Huang L, Chen DB. Endogenous NO upon estradiol-17β stimulation and NO donor differentially regulate mitochondrial S-nitrosylation in endothelial cells. Endocrinology 2014; 155:3005-16. [PMID: 24877627 PMCID: PMC4098011 DOI: 10.1210/en.2013-2174] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Adduction of a nitric oxide (NO) moiety (NO(•)) to cysteines termed as S-nitrosylation (SNO) has emerged as a crucial mechanism for NO signaling crucial for mediating the vascular effects of estrogens. Mitochondrion is a known vascular risk factor; however, the effects of estrogens on mitochondrial SNO are incompletely understood. In this study we determined the effects of estradiol-17β (E2β) on mitochondrial protein SNO in primary human umbilical vein endothelial cells and compared the mitochondrial nitroso-proteomes in E2β- and a NO donor S-nitrosoglutathione (GSNO)-treated cells using a proteomics approach. Treatment with 10 nM E2β and 1 mM GSNO for 30 minutes significantly increased the levels of mitochondrial SNO-proteins. Subcellular localization of SNO-proteins showed mitochondria as the major cellular organelle for protein SNO in response to E2β and GSNO. E2β stimulated mitochondrial endothelial nitric oxide synthase (eNOS) phosphorylation and mitochondrial protein SNO that was enhanced by overexpression of mitochondrion or Golgi, but not membrane targeting eNOS constructs. We identified 11, 32, and 54 SNO-proteins in the mitochondria from the untreated, E2β-, and GSNO-treated human umbilical vein endothelial cells, respectively. Comparisons of the nitroso-proteomes revealed that common and different mitochondrial SNO-proteins were affected by endogenous NO on E2β stimulation and exogenous NO from donor. These SNO-proteins were associated with various mitochondrial functions, including energy and redox regulation, transport, iron homeostasis, translation, mitochondrial morphology, and apoptosis, etc. Collectively, we conclude that estrogens rapidly stimulate protein SNO in endothelial mitochondria via mitochondrial eNOS, providing a mechanism for mediating the vascular effects of estrogens.
Collapse
Affiliation(s)
- Seiro Satohisa
- Departments of Obstetrics and Gynecology (S.S., H-h.Z., L.F., D-b.C.), Biophysics and Physiology (Y-y.Y., L.H.), and Pathology (D-b.C.), University of California, Irvine, California 92697
| | | | | | | | | | | |
Collapse
|
48
|
Stub D, Duffy SJ, Kaye DM. Device-Based Therapy in the Prevention of Contrast-Induced Nephropathy. Interv Cardiol Clin 2014; 3:421-428. [PMID: 28582226 DOI: 10.1016/j.iccl.2014.03.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Contrast-induced nephropathy (CIN) is a common condition that is associated with short- and, likely, long-term adverse outcomes. Although periprocedural intravenous hydration is the simplest and most widely used technique to prevent CIN, the limited ability of this approach to mitigate the CIN risk in high-risk populations has provided an impetus to develop new preventive strategies. A range of potentially useful device-based approaches offers new preventive techniques. Well-designed and adequately powered randomized studies of these device-based therapies are urgently needed to determine the expanding role they will play in future clinical practice.
Collapse
Affiliation(s)
- Dion Stub
- Cardiovascular Medicine, Baker IDI Heart and Diabetes Institute, Alfred Hospital, Commercial Road, Melbourne, Victoria 3004, Australia
| | - Stephen J Duffy
- Cardiovascular Medicine, Baker IDI Heart and Diabetes Institute, Alfred Hospital, Commercial Road, Melbourne, Victoria 3004, Australia
| | - David M Kaye
- Cardiovascular Medicine, Baker IDI Heart and Diabetes Institute, Alfred Hospital, Commercial Road, Melbourne, Victoria 3004, Australia.
| |
Collapse
|
49
|
Simon JN, Duglan D, Casadei B, Carnicer R. Nitric oxide synthase regulation of cardiac excitation-contraction coupling in health and disease. J Mol Cell Cardiol 2014; 73:80-91. [PMID: 24631761 DOI: 10.1016/j.yjmcc.2014.03.004] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 03/04/2014] [Accepted: 03/05/2014] [Indexed: 02/07/2023]
Abstract
Significant advances in our understanding of the ability of nitric oxide synthases (NOS) to modulate cardiac function have provided key insights into the role NOS play in the regulation of excitation-contraction (EC) coupling in health and disease. Through both cGMP-dependent and cGMP-independent (e.g. S-nitrosylation) mechanisms, NOS have the ability to alter intracellular Ca(2+) handling and the myofilament response to Ca(2+), thereby impacting the systolic and diastolic performance of the myocardium. Findings from experiments using nitric oxide (NO) donors and NOS inhibition or gene deletion clearly implicate dysfunctional NOS as a critical contributor to many cardiovascular disease states. However, studies to date have only partially addressed NOS isoform-specific effects and, more importantly, how subcellular localization of NOS influences ion channels involved in myocardial EC coupling and excitability. In this review, we focus on the contribution of each NOS isoform to cardiac dysfunction and on the role of uncoupled NOS activity in common cardiac disease states, including heart failure, diabetic cardiomyopathy, ischemia/reperfusion injury and atrial fibrillation. We also review evidence that clearly indicates the importance of NO in cardioprotection. This article is part of a Special Issue entitled "Redox Signalling in the Cardiovascular System".
Collapse
Affiliation(s)
- Jillian N Simon
- Radcliffe Department of Medicine, Division of Cardiovascular Medicine, University of Oxford, Oxford, UK
| | - Drew Duglan
- Radcliffe Department of Medicine, Division of Cardiovascular Medicine, University of Oxford, Oxford, UK
| | - Barbara Casadei
- Radcliffe Department of Medicine, Division of Cardiovascular Medicine, University of Oxford, Oxford, UK
| | - Ricardo Carnicer
- Radcliffe Department of Medicine, Division of Cardiovascular Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
50
|
Tong G, Aponte AM, Kohr MJ, Steenbergen C, Murphy E, Sun J. Postconditioning leads to an increase in protein S-nitrosylation. Am J Physiol Heart Circ Physiol 2014; 306:H825-32. [PMID: 24441547 DOI: 10.1152/ajpheart.00660.2013] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Previous studies have shown a role for nitric oxide and S-nitrosylation (SNO) in postconditioning (PostC), but specific SNO proteins and sites have not been identified in the myocardium after PostC. In this study, we examined SNO signaling in PostC using a Langendorff-perfused mouse heart model. After 20 min of equilibrium perfusion and 25 min of global ischemia, PostC was applied at the beginning of reperfusion with six cycles of 10 s of reperfusion and 10 s of ischemia. The total period of reperfusion was 90 min. Compared with the ischemia-reperfusion (I/R) control, PostC significantly reduced postischemic contractile dysfunction and infarct size. PostC-induced protection was blocked by treatment with N(G)-nitro-l-arginine methyl ester (l-NAME) (10 μmol/l; a constitutive NO synthase inhibitor), but not by either ODQ (10 μmol/l, a highly selective soluble guanylyl cyclase inhibitor) or KT5823 (1 μmol/l, a specific protein kinase G inhibitor). Two biotin switch based methods, two dimensional CyDye-maleimide difference gel electrophoresis (2D CyDye-maleimide DIGE) and SNO-resin-assisted capture (SNO-RAC), were utilized to identify SNO-modified proteins and sites. Using 2D CyDye-maleimide DIGE analysis, PostC was found to cause a 25% or greater increase in SNO of a number of proteins, which was blocked by treatment with l-NAME in parallel with the loss of protection. Using SNO-RAC, we identified 77 unique proteins with SNO sites after PostC. These results suggest that NO-mediated SNO signaling is involved in PostC-induced cardioprotection and these data provide the first set of candidate SNO proteins in PostC hearts.
Collapse
Affiliation(s)
- Guang Tong
- Department of Cardiovascular Surgery, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, Guangdong Province, China
| | | | | | | | | | | |
Collapse
|