1
|
Tregub PP, Kulikov VP, Ibrahimli I, Tregub OF, Volodkin AV, Ignatyuk MA, Kostin AA, Atiakshin DA. Molecular Mechanisms of Neuroprotection after the Intermittent Exposures of Hypercapnic Hypoxia. Int J Mol Sci 2024; 25:3665. [PMID: 38612476 PMCID: PMC11011936 DOI: 10.3390/ijms25073665] [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: 02/19/2024] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
The review introduces the stages of formation and experimental confirmation of the hypothesis regarding the mutual potentiation of neuroprotective effects of hypoxia and hypercapnia during their combined influence (hypercapnic hypoxia). The main focus is on the mechanisms and signaling pathways involved in the formation of ischemic tolerance in the brain during intermittent hypercapnic hypoxia. Importantly, the combined effect of hypoxia and hypercapnia exerts a more pronounced neuroprotective effect compared to their separate application. Some signaling systems are associated with the predominance of the hypoxic stimulus (HIF-1α, A1 receptors), while others (NF-κB, antioxidant activity, inhibition of apoptosis, maintenance of selective blood-brain barrier permeability) are mainly modulated by hypercapnia. Most of the molecular and cellular mechanisms involved in the formation of brain tolerance to ischemia are due to the contribution of both excess carbon dioxide and oxygen deficiency (ATP-dependent potassium channels, chaperones, endoplasmic reticulum stress, mitochondrial metabolism reprogramming). Overall, experimental studies indicate the dominance of hypercapnia in the neuroprotective effect of its combined action with hypoxia. Recent clinical studies have demonstrated the effectiveness of hypercapnic-hypoxic training in the treatment of childhood cerebral palsy and diabetic polyneuropathy in children. Combining hypercapnic hypoxia with pharmacological modulators of neuro/cardio/cytoprotection signaling pathways is likely to be promising for translating experimental research into clinical medicine.
Collapse
Affiliation(s)
- Pavel P. Tregub
- Department of Pathophysiology, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
- Brain Science Institute, Research Center of Neurology, 125367 Moscow, Russia
- Scientific and Educational Resource Center “Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis”, RUDN University, 117198 Moscow, Russia; (A.V.V.); (M.A.I.); (A.A.K.); (D.A.A.)
| | - Vladimir P. Kulikov
- Department of Ultrasound and Functional Diagnostics, Altay State Medical University, 656040 Barnaul, Russia;
| | - Irada Ibrahimli
- Department of Pathophysiology, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
| | | | - Artem V. Volodkin
- Scientific and Educational Resource Center “Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis”, RUDN University, 117198 Moscow, Russia; (A.V.V.); (M.A.I.); (A.A.K.); (D.A.A.)
| | - Michael A. Ignatyuk
- Scientific and Educational Resource Center “Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis”, RUDN University, 117198 Moscow, Russia; (A.V.V.); (M.A.I.); (A.A.K.); (D.A.A.)
| | - Andrey A. Kostin
- Scientific and Educational Resource Center “Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis”, RUDN University, 117198 Moscow, Russia; (A.V.V.); (M.A.I.); (A.A.K.); (D.A.A.)
| | - Dmitrii A. Atiakshin
- Scientific and Educational Resource Center “Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis”, RUDN University, 117198 Moscow, Russia; (A.V.V.); (M.A.I.); (A.A.K.); (D.A.A.)
| |
Collapse
|
2
|
Sarkar A, Pawar SV, Chopra K, Jain M. Gamut of glycolytic enzymes in vascular smooth muscle cell proliferation: Implications for vascular proliferative diseases. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167021. [PMID: 38216067 DOI: 10.1016/j.bbadis.2024.167021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 01/05/2024] [Accepted: 01/05/2024] [Indexed: 01/14/2024]
Abstract
Vascular smooth muscle cells (VSMCs) are the predominant cell type in the media of the blood vessels and are responsible for maintaining vascular tone. Emerging evidence confirms that VSMCs possess high plasticity. During vascular injury, VSMCs switch from a "contractile" phenotype to an extremely proliferative "synthetic" phenotype. The balance between both strongly affects the progression of vascular remodeling in many cardiovascular pathologies such as restenosis, atherosclerosis and aortic aneurism. Proliferating cells demand high energy requirements and to meet this necessity, alteration in cellular bioenergetics seems to be essential. Glycolysis, fatty acid metabolism, and amino acid metabolism act as a fuel for VSMC proliferation. Metabolic reprogramming of VSMCs is dynamically variable that involves multiple mechanisms and encompasses the coordination of various signaling molecules, proteins, and enzymes. Here, we systemically reviewed the metabolic changes together with the possible treatments that are still under investigation underlying VSMC plasticity which provides a promising direction for the treatment of diseases associated with VSMC proliferation. A better understanding of the interaction between metabolism with associated signaling may uncover additional targets for better therapeutic strategies in vascular disorders.
Collapse
Affiliation(s)
- Ankan Sarkar
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Sandip V Pawar
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Kanwaljit Chopra
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Manish Jain
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India.
| |
Collapse
|
3
|
Arranz-Paraíso D, Sola Y, Baeza-Moyano D, Benítez-Martínez M, Melero-Tur S, González-Lezcano RA. Mitochondria and light: An overview of the pathways triggered in skin and retina with incident infrared radiation. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2023; 238:112614. [PMID: 36469983 DOI: 10.1016/j.jphotobiol.2022.112614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 11/18/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022]
Abstract
Slightly more than half of the solar radiation that passes through the atmosphere and reaches the Earth's surface is infrared. Over the past few years, many papers have been published on the possible positive effects of receiving this part of the electromagnetic spectrum. In this article we analyse the role of mitochondria in the supposed effects of infrared light based on the published literature. It is claimed that ATP synthesis is stimulated, which has a positive effect on the skin by increasing fibroblast proliferation, anchorage and production of collagen fibres, procollagen, and various cytokines responsible for the wound healing process, such as keratinocyte growth factor. Currently there are infrared light emitting equipment whose manufacturers and the centres where this service or treatment is offered claim that they are used for skin rejuvenation among other positive effects. Based on the literature review, it is necessary to deepen the scientific study of the mechanism of absorption of infrared radiation through the skin to better understand its possible positive effects, the risks of overexposure and to improve consumer health protection.
Collapse
Affiliation(s)
- Daniel Arranz-Paraíso
- Área de conocimiento de Tecnología Farmacéutica, Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28668 Alcorcón, Madrid, Spain.
| | - Yolanda Sola
- Group of Meteorology, Department of Applied Physics, Universitat de Barcelona, Martí i Franquès 1, 08028 Barcelona, Spain.
| | - David Baeza-Moyano
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28668 Alcorcón, Madrid, Spain.
| | - Marta Benítez-Martínez
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28668 Alcorcón, Madrid, Spain.
| | - Sofía Melero-Tur
- Departamento de arquitectura y diseño, Escuela Politécnica Superior, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28668 Alcorcón, Madrid, Spain.
| | - Roberto Alonso González-Lezcano
- Departamento de arquitectura y diseño, Escuela Politécnica Superior, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28668 Alcorcón, Madrid, Spain.
| |
Collapse
|
4
|
Tian Y, Nusantara AC, Hamoh T, Mzyk A, Tian X, Perona Martinez F, Li R, Permentier HP, Schirhagl R. Functionalized Fluorescent Nanodiamonds for Simultaneous Drug Delivery and Quantum Sensing in HeLa Cells. ACS APPLIED MATERIALS & INTERFACES 2022; 14:39265-39273. [PMID: 35984747 PMCID: PMC9437893 DOI: 10.1021/acsami.2c11688] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Here, we present multifunctional fluorescent nanodiamonds (FNDs) for simultaneous drug delivery and free radical detection. For this purpose, we modified FNDs containing nitrogen vacancy (NV) centers with a diazoxide derivative. We found that our particles enter cells more easily and are able to deliver this cancer drug into HeLa cells. The particles were characterized by infrared spectroscopy, dynamic light scattering, and secondary electron microscopy. Compared to the free drug, we observe a sustained release over 72 h rather than 12 h for the free drug. Apart from releasing the drug, with these particles, we can measure the drug's effect on free radical generation directly. This has the advantage that the response is measured locally, where the drug is released. These FNDs change their optical properties based on their magnetic surrounding. More specifically, we make use of a technique called relaxometry to detect spin noise from the free radical at the nanoscale with subcellular resolution. We further compared the results from our new technique with a conventional fluorescence assay for the detection of reactive oxygen species. This provides a new method to investigate the relationship between drug release and the response by the cell via radical formation or inhibition.
Collapse
Affiliation(s)
- Yuchen Tian
- Department
of Biomedical Engineering, Groningen University,
University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AW Groningen, Netherlands
| | - Anggrek C. Nusantara
- Department
of Biomedical Engineering, Groningen University,
University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AW Groningen, Netherlands
| | - Thamir Hamoh
- Department
of Biomedical Engineering, Groningen University,
University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AW Groningen, Netherlands
| | - Aldona Mzyk
- Department
of Biomedical Engineering, Groningen University,
University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AW Groningen, Netherlands
- Institute
of Metallurgy and Materials Science Polish Academy of Sciences, 25 Reymonta Street, 30-059, Cracow, Poland
| | - Xiaobo Tian
- Department
of Analytical Biochemistry, Interfaculty Mass Spectrometry Center,
Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, Groningen 9713 AV, The Netherlands
| | - Felipe Perona Martinez
- Department
of Biomedical Engineering, Groningen University,
University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AW Groningen, Netherlands
| | - Runrun Li
- Department
of Biomedical Engineering, Groningen University,
University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AW Groningen, Netherlands
| | - Hjalmar P. Permentier
- Department
of Analytical Biochemistry, Interfaculty Mass Spectrometry Center,
Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, Groningen 9713 AV, The Netherlands
| | - Romana Schirhagl
- Department
of Biomedical Engineering, Groningen University,
University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AW Groningen, Netherlands
| |
Collapse
|
5
|
Ahmad T, Wang J, Velez AK, Suarez-Pierre A, Clement KC, Dong J, Sebestyen K, Canner JK, Murphy MP, Lawton JS. Cardioprotective mechanisms of mitochondria-targeted S-nitrosating agent and adenosine triphosphate-sensitive potassium channel opener are mutually exclusive. JTCVS OPEN 2021; 8:338-354. [PMID: 36004142 PMCID: PMC9390287 DOI: 10.1016/j.xjon.2021.07.036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 07/30/2021] [Indexed: 11/22/2022]
Abstract
Background Myocytes exposed to stress exhibit significant swelling and reduced contractility. These consequences are ameliorated by adenosine triphosphate-sensitive potassium (KATP) channel opener diazoxide (DZX) via an unknown mechanism. KATP channel openers also provide cardioprotection in multiple animal models. Nitric oxide donors are similarly cardioprotective, and their combination with KATP activation may provide synergistic benefit. We hypothesized that mitochondria-targeted S-nitrosating agent (MitoSNO) would provide synergistic cardioprotection with DZX. Methods Myocyte volume and contractility were compared following Tyrode's physiologic solution (20 minutes) and stress (hyperkalemic cardioplegia [CPG] ± DZX; n = 5-20 each; 20 minutes) with or without MitoSNO (n = 5-11 each) at the end of stress, followed by Tyrode's solution (20 minutes). Isolated mouse hearts received CPG ± DZX (n = 8-10 each) before global ischemia (90 minutes) with or without MitoSNO (n = 8 each) at the end of ischemia, followed by reperfusion (30 minutes). Left ventricular (LV) pressures were compared using a linear mixed model to assess the impact of treatment on the outcome, adjusting for baseline and balloon volume. Results Stress (CPG) was associated with reduced myocyte contractility that was prevented by DZX and MitoSNO individually; however, their combination was associated with loss of cardioprotection. Similarly, DZX and MitoSNO improved LV function after prolonged ischemia compared with CPG alone, and cardioprotection was lost with their combination. Conclusions MitoSNO and DZX provide cardioprotection that is lost with their combination, suggesting mutually exclusive mechanisms of action. The lack of a synergistic beneficial effect informs the current knowledge of the cardioprotective mechanisms of DZX and will aid planning of future clinical trials.
Collapse
Key Words
- CPG, cardioplegia
- DZX, diazoxide
- EDP, end-diastolic pressure
- KATP, adenosine triphosphate–sensitive potassium
- KHB, Krebs–Henseleit buffer
- LV, left ventricular
- LVDP, left ventricular developed pressure
- MitoSNO, mitochondrial-selective S–nitrosating agent
- NO, nitric oxide
- ROS, reactive oxygen species
- SDH, succinate dehydrogenase
- SUR, sulfonylurea
- basic science
- ion channels
- ischemia
- preconditioning
Collapse
Affiliation(s)
- Thaniyyah Ahmad
- Division of Cardiac Surgery, Department of Surgery, Johns Hopkins University, Baltimore, Md
| | - Jie Wang
- Division of Cardiac Surgery, Department of Surgery, Johns Hopkins University, Baltimore, Md
| | - Ana Karen Velez
- Division of Cardiac Surgery, Department of Surgery, Johns Hopkins University, Baltimore, Md
| | | | - Kathleen C. Clement
- Division of Cardiac Surgery, Department of Surgery, Johns Hopkins University, Baltimore, Md
| | - Jie Dong
- Division of Cardiac Surgery, Department of Surgery, Johns Hopkins University, Baltimore, Md
| | - Krisztian Sebestyen
- Johns Hopkins Center for Outcomes Research, Johns Hopkins University, Baltimore, Md
| | - Joseph K. Canner
- Johns Hopkins Center for Outcomes Research, Johns Hopkins University, Baltimore, Md
| | - Michael P. Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Jennifer S. Lawton
- Division of Cardiac Surgery, Department of Surgery, Johns Hopkins University, Baltimore, Md
| |
Collapse
|
6
|
Höppner J, Bruni C, Distler O, Robson SC, Burmester GR, Siegert E, Distler JHW. Purinergic signaling in systemic sclerosis. Rheumatology (Oxford) 2021; 61:2770-2782. [PMID: 34849624 DOI: 10.1093/rheumatology/keab859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 11/13/2022] Open
Abstract
Systemic sclerosis (SSc) is a chronic autoimmune rheumatic disease that involves numerous organs and presents major management challenges. The histopathologic hallmarks of SSc include vasculopathy, fibrosis and autoimmune phenomena involving both innate and adaptive immune systems. Purinergic signalling is a pathway that may be implicated in the pathophysiology of several of these disease manifestations. Extracellular purines are potent signalling mediators, which have been shown to be dysregulated in SSc. As examples, purines can exacerbate vasculopathy and provoke platelet dysfunction; as well as contributing to immune dysregulation. Elements of purinergic signalling further promote organ and tissue fibrosis in several disease models. Here, we provide an overview of extracellular purine metabolism in purinergic signalling and link disorders of these to the molecular pathology of SSc. We also discuss targeting the purinergic signalling and explore the translational applications for new therapeutic options in SSc.
Collapse
Affiliation(s)
- Jakob Höppner
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Cosimo Bruni
- Department of Experimental and Clinical Medicine, Division of Rheumatology, Careggi University Hospital, University of Florence, Florence, Italy.,Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Oliver Distler
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Simon C Robson
- Departments of Anesthesia and Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Gerd R Burmester
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Elise Siegert
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| | - Jörg H W Distler
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Germany
| |
Collapse
|
7
|
Hypercapnia Modulates the Activity of Adenosine A1 Receptors and mitoK +ATP-Channels in Rat Brain When Exposed to Intermittent Hypoxia. Neuromolecular Med 2021; 24:155-168. [PMID: 34115290 DOI: 10.1007/s12017-021-08672-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 06/05/2021] [Indexed: 10/21/2022]
Abstract
The mechanisms and signaling pathways of the neuroprotective effects of hypercapnia and its combination with hypoxia are not studied sufficiently. The study aims to test the hypothesis of the potentiating effect of hypercapnia on the systems of adaptation to hypoxia, directly associated with A1-adenosine receptors and mitochondrial ATP-dependent K+ -channels (mitoK+ATP-channels). We evaluated the relative number of A1-adenosine receptors and mitoK+ATP-channels in astrocytes obtained from male Wistar rats exposed to various respiratory conditions (15 times of hypoxia and/or hypercapnia). In addition, the relative number of these molecules in astrocytes was evaluated on an in vitro model of chemical hypoxia, as well as in the cerebral cortex after photothrombotic damage. This study indicates an increase in the relative number of A1-adenosine receptors in astrocytes and in cells next to the stroke region of the cerebral cortex in rats exposed to hypoxia and hypercapnic hypoxia, but not hypercapnia alone. Hypercapnia and hypoxia increase the relative number of mitoK+ATP-channels in astrocytes and in cells of the peri-infarct region of the cerebral cortex in rats. In an in vitro study, hypercapnia mitigates the effects of acute chemical hypoxia observed in astrocytes for A1-adenosine receptors and mitoK+ATP-channels. Hypercapnia, unlike hypoxia, does not affect the relative number of A1 receptors to adenosine. At the same time, both hypercapnia and hypoxia increase the relative number of mitoK+ATP-channels, which can potentiate their protective effects with combined exposure.
Collapse
|
8
|
Signaling pathways targeting mitochondrial potassium channels. Int J Biochem Cell Biol 2020; 125:105792. [PMID: 32574707 DOI: 10.1016/j.biocel.2020.105792] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 06/08/2020] [Accepted: 06/11/2020] [Indexed: 12/11/2022]
Abstract
In this review, we describe key signaling pathways regulating potassium channels present in the inner mitochondrial membrane. The signaling cascades covered here include phosphorylation, redox reactions, modulation by calcium ions and nucleotides. The following types of potassium channels have been identified in the inner mitochondrial membrane of various tissues: ATP-sensitive, Ca2+-activated, voltage-gated and two-pore domain potassium channels. The direct roles of these channels involve regulation of mitochondrial respiration, membrane potential and synthesis of reactive oxygen species (ROS). Changes in channel activity lead to diverse pro-life and pro-death responses in different cell types. Hence, characterizing the signaling pathways regulating mitochondrial potassium channels will facilitate understanding the physiological role of these proteins. Additionally, we describe in this paper certain regulatory mechanisms, which are unique to mitochondrial potassium channels.
Collapse
|
9
|
Krajnak K. Frequency-dependent changes in mitochondrial number and generation of reactive oxygen species in a rat model of vibration-induced injury. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2020; 83:20-35. [PMID: 31971087 PMCID: PMC7737659 DOI: 10.1080/15287394.2020.1718043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Regular use of vibrating hand tools results in cold-induced vasoconstriction, finger blanching, and a reduction in tactile sensitivity and manual dexterity. Depending upon the length and frequency, vibration induces regeneration, or dysfunction and apoptosis, inflammation and an increase in reactive oxygen species (ROS) levels. These changes may be associated with mitochondria, this study examined the effects of vibration on total and functional mitochondria number. Male rats were exposed to restraint or tail vibration at 62.5, 125, or 250 Hz. The frequency-dependent effects of vibration on mitochondrial number and generation of oxidative stress were examined. After 10 days of exposure at 125 Hz, ventral tail arteries (VTA) were constricted and there was an increase in mitochondrial number and intensity of ROS staining. In the skin, the influence of vibration on arterioles displayed a similar but insignificant response in VTA. There was also a reduction in the number of small nerves with exposure to vibration at 250 Hz, and a reduction in mitochondrial number in nerves in restrained and all vibrated conditions. There was a significant rise in the size of the sensory receptors with vibration at 125 Hz, and an elevation in ROS levels. Based upon these results, mitochondria number and activity are affected by vibration, especially at frequencies at or near resonance. The influence of vibration on the vascular system may either be adaptive or maladaptive. However, the effects on cutaneous nerves might be a precursor to loss of innervation and sensory function noted in workers exposed to vibration.
Collapse
Affiliation(s)
- Kristine Krajnak
- Physical Effects Research Branch, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| |
Collapse
|
10
|
Riess ML, Elorbany R, Weihrauch D, Stowe DF, Camara AK. PPARγ-Independent Side Effects of Thiazolidinediones on Mitochondrial Redox State in Rat Isolated Hearts. Cells 2020; 9:cells9010252. [PMID: 31968546 PMCID: PMC7017211 DOI: 10.3390/cells9010252] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/17/2020] [Accepted: 01/17/2020] [Indexed: 11/16/2022] Open
Abstract
The effect of anti-diabetic thiazolidinediones (TZDs) on contributing to heart failure and cardiac ischemia/reperfusion (IR) injury is controversial. In this study we investigated the effect of select TZDs on myocardial and mitochondrial function in Brown Norway rat isolated hearts. In a first set of experiments, the TZD rosiglitazone was given acutely before global myocardial IR, and pre- and post-IR function and infarct size were assessed. In a second set of experiments, different concentrations of rosiglitazone and pioglitazone were administered in the presence or absence of the specific PPARγ antagonist GW9662, and their effects on the mitochondrial redox state were measured by online NADH and FAD autofluorescence. The administration of rosiglitazone did not significantly affect myocardial function except for transiently increasing coronary flow, but it increased IR injury compared to the control hearts. Both TZDs resulted in dose-dependent, reversible increases in mitochondrial oxidation which was not attenuated by GW9662. Taken together, these data suggest that TZDs cause excessive mitochondrial uncoupling by a PPARγ-independent mechanism. Acute rosiglitazone administration before IR was associated with enhanced cardiac injury. If translated clinically, susceptible patients on PPARγ agonists may experience enhanced myocardial IR injury by mitochondrial dysfunction.
Collapse
Affiliation(s)
- Matthias L. Riess
- Anesthesiology, TVHS VA Medical Center, Nashville, TN 37212, USA
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
- Correspondence: ; Tel.: +1-(615)-936-0277; Fax: +1-(615)-343-3916
| | - Reem Elorbany
- Interdisciplinary Scientist Training Program, University of Chicago, Chicago, IL 60637, USA;
| | - Dorothee Weihrauch
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (D.W.); (D.F.S.)
| | - David F. Stowe
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (D.W.); (D.F.S.)
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Clement J. Zablocki VA Medical Center, Milwaukee, WI 53295, USA
| | - Amadou K.S. Camara
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (D.W.); (D.F.S.)
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
11
|
Watanabe Y. Cardiac Na +/Ca 2+ exchange stimulators among cardioprotective drugs. J Physiol Sci 2019; 69:837-849. [PMID: 31664641 PMCID: PMC10717683 DOI: 10.1007/s12576-019-00721-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 10/18/2019] [Indexed: 02/06/2023]
Abstract
We previously reviewed our study of the pharmacological properties of cardiac Na+/Ca2+ exchange (NCX1) inhibitors among cardioprotective drugs, such as amiodarone, bepridil, dronedarone, cibenzoline, azimilide, aprindine, and benzyl-oxyphenyl derivatives (Watanabe et al. in J Pharmacol Sci 102:7-16, 2006). Since then we have continued our studies further and found that some cardioprotective drugs are NCX1 stimulators. Cardiac Na+/Ca2+ exchange current (INCX1) was stimulated by nicorandil (a hybrid ATP-sensitive K+ channel opener), pinacidil (a non-selective ATP-sensitive K+ channel opener), flecainide (an antiarrhythmic drug), and sodium nitroprusside (SNP) (an NO donor). Sildenafil (a phosphodiesterase-5 inhibitor) further increased the pinacidil-induced augmentation of INCX1. In paper, here I review the NCX stimulants that enhance NCX function among the cardioprotective agents we examined such as nicorandil, pinacidil, SNP, sildenafil and flecainide, in addition to atrial natriuretic (ANP) and dofetilide, which were reported by other investigators.
Collapse
Affiliation(s)
- Yasuhide Watanabe
- Division of Pharmacological Science, Department of Health Science, Hamamatsu University School of Medicine, 1-20-1 Handa-yama, Higashi-ku, Hamamatsu, 431-3192, Japan.
| |
Collapse
|
12
|
Dowrick JM, Tran K, Loiselle DS, Nielsen PMF, Taberner AJ, Han J, Ward M. The slow force response to stretch: Controversy and contradictions. Acta Physiol (Oxf) 2019; 226:e13250. [PMID: 30614655 DOI: 10.1111/apha.13250] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 12/20/2018] [Accepted: 01/02/2019] [Indexed: 12/19/2022]
Abstract
When exposed to an abrupt stretch, cardiac muscle exhibits biphasic active force enhancement. The initial, instantaneous, force enhancement is well explained by the Frank-Starling mechanism. However, the cellular mechanisms associated with the second, slower phase remain contentious. This review explores hypotheses regarding this "slow force response" with the intention of clarifying some apparent contradictions in the literature. The review is partitioned into three sections. The first section considers pathways that modify the intracellular calcium handling to address the role of the sarcoplasmic reticulum in the mechanism underlying the slow force response. The second section focuses on extracellular calcium fluxes and explores the identity and contribution of the stretch-activated, non-specific, cation channels as well as signalling cascades associated with G-protein coupled receptors. The final section introduces promising candidates for the mechanosensor(s) responsible for detecting the stretch perturbation.
Collapse
Affiliation(s)
- Jarrah M. Dowrick
- Auckland Bioengineering Institute University of Auckland Auckland New Zealand
| | - Kenneth Tran
- Auckland Bioengineering Institute University of Auckland Auckland New Zealand
| | - Denis S. Loiselle
- Auckland Bioengineering Institute University of Auckland Auckland New Zealand
- Department of Physiology University of Auckland Auckland New Zealand
| | - Poul M. F. Nielsen
- Auckland Bioengineering Institute University of Auckland Auckland New Zealand
- Department of Engineering Science University of Auckland Auckland New Zealand
| | - Andrew J. Taberner
- Auckland Bioengineering Institute University of Auckland Auckland New Zealand
- Department of Engineering Science University of Auckland Auckland New Zealand
| | - June‐Chiew Han
- Auckland Bioengineering Institute University of Auckland Auckland New Zealand
| | - Marie‐Louise Ward
- Department of Physiology University of Auckland Auckland New Zealand
| |
Collapse
|
13
|
Iguchi K, Saotome M, Yamashita K, Hasan P, Sasaki M, Maekawa Y, Watanabe Y. Pinacidil, a KATP channel opener, stimulates cardiac Na +/Ca 2+ exchanger function through the NO/cGMP/PKG signaling pathway in guinea pig cardiac ventricular myocytes. Naunyn Schmiedebergs Arch Pharmacol 2019; 392:949-959. [PMID: 30919008 DOI: 10.1007/s00210-019-01642-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 03/01/2019] [Indexed: 12/21/2022]
Abstract
Pinacidil, a nonselective ATP-sensitive K+ (KATP) channel opener, has cardioprotective effects for hypertension, ischemia/reperfusion injury, and arrhythmia. This agent abolishes early afterdepolarizations, delayed afterdepolarizations (DADs), and abnormal automaticity in canine cardiac ventricular myocytes. DADs are well known to be caused by the Na+/Ca2+ exchange current (INCX). In this study, we used the whole-cell patch-clamp technique and Fura-2/AM (Ca2+-indicator) method to investigate the effect of pinacidil on INCX in isolated guinea pig cardiac ventricular myocytes. In the patch-clamp study, pinacidil enhanced INCX in a concentration-dependent manner. The half-maximal effective concentration values were 23.5 and 23.0 μM for the Ca2+ entry (outward) and Ca2+ exit (inward) components of INCX, respectively. The pinacidil-induced INCX increase was blocked by L-NAME, a nitric oxide (NO) synthase inhibitor, by ODQ, a soluble guanylate cyclase inhibitor, and by KT5823, a cyclic guanosine monophosphate (cGMP)-dependent protein kinase (PKG) inhibitor, but not by N-2-mercaptopropyonyl glycine (MPG), a reactive oxygen species (ROS) scavenger. Glibenclamide, a nonselective KATP channel inhibitor, blocked the pinacidil-induced INCX increase, while 5-HD, a selective mitochondria KATP channel inhibitor, did not. In the Fura-2/AM study pinacidil also enhanced intracellular Ca2+ concentration, which was inhibited by L-NAME, ODQ, KT5823, and glibenclamide, but not by MPG and 5-HD. Sildenafil, a phosphodiesterase 5 inhibitor, increased further the pinacidil-induced INCX increase. Sodium nitroprusside, a NO donor, also increased INCX. In conclusion, pinacidil may stimulate cardiac Na+/Ca2+ exchanger (NCX1) by opening plasma membrane KATP channels and activating the NO/cGMP/PKG signaling pathway.
Collapse
Affiliation(s)
- Keisuke Iguchi
- Department of Internal Medicine III (Cardiology), Hamamatsu University School of Medicine, Hamamatsu, 431-3192, Japan.,Division of Pharmacological Science, Department of Health Science, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192, Japan
| | - Masao Saotome
- Department of Internal Medicine III (Cardiology), Hamamatsu University School of Medicine, Hamamatsu, 431-3192, Japan
| | - Kanna Yamashita
- Division of Pharmacological Science, Department of Health Science, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192, Japan
| | - Prottoy Hasan
- Department of Internal Medicine III (Cardiology), Hamamatsu University School of Medicine, Hamamatsu, 431-3192, Japan
| | - Miyuki Sasaki
- Division of Pharmacological Science, Department of Health Science, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192, Japan
| | - Yuichiro Maekawa
- Department of Internal Medicine III (Cardiology), Hamamatsu University School of Medicine, Hamamatsu, 431-3192, Japan
| | - Yasuhide Watanabe
- Division of Pharmacological Science, Department of Health Science, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192, Japan.
| |
Collapse
|
14
|
Abstract
20-HETE, the ω-hydroxylation product of arachidonic acid catalyzed by enzymes of the cytochrome P450 (CYP) 4A and 4F gene families, is a bioactive lipid mediator with potent effects on the vasculature including stimulation of smooth muscle cell contractility, migration and proliferation as well as activation of endothelial cell dysfunction and inflammation. Clinical studies have shown elevated levels of plasma and urinary 20-HETE in human diseases and conditions such as hypertension, obesity and metabolic syndrome, myocardial infarction, stroke, and chronic kidney diseases. Studies of polymorphic associations also suggest an important role for 20-HETE in hypertension, stroke and myocardial infarction. Animal models of increased 20-HETE production are hypertensive and are more susceptible to cardiovascular injury. The current review summarizes recent findings that focus on the role of 20-HETE in the regulation of vascular and cardiac function and its contribution to the pathology of vascular and cardiac diseases.
Collapse
Affiliation(s)
- Petra Rocic
- Department of Pharmacology, New York Medical College School of Medicine, Valhalla, NY, United States
| | | |
Collapse
|
15
|
Berry BJ, Trewin AJ, Amitrano AM, Kim M, Wojtovich AP. Use the Protonmotive Force: Mitochondrial Uncoupling and Reactive Oxygen Species. J Mol Biol 2018; 430:3873-3891. [PMID: 29626541 DOI: 10.1016/j.jmb.2018.03.025] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 03/21/2018] [Accepted: 03/26/2018] [Indexed: 02/06/2023]
Abstract
Mitochondrial respiration results in an electrochemical proton gradient, or protonmotive force (pmf), across the mitochondrial inner membrane. The pmf is a form of potential energy consisting of charge (∆ψm) and chemical (∆pH) components, that together drive ATP production. In a process called uncoupling, proton leak into the mitochondrial matrix independent of ATP production dissipates the pmf and energy is lost as heat. Other events can directly dissipate the pmf independent of ATP production as well, such as chemical exposure or mechanisms involving regulated mitochondrial membrane electrolyte transport. Uncoupling has defined roles in metabolic plasticity and can be linked through signal transduction to physiologic events. In the latter case, the pmf impacts mitochondrial reactive oxygen species (ROS) production. Although capable of molecular damage, ROS also have signaling properties that depend on the timing, location, and quantity of their production. In this review, we provide a general overview of mitochondrial ROS production, mechanisms of uncoupling, and how these work in tandem to affect physiology and pathologies, including obesity, cardiovascular disease, and immunity. Overall, we highlight that isolated bioenergetic models-mitochondria and cells-only partially recapitulate the complex link between the pmf and ROS signaling that occurs in vivo.
Collapse
Affiliation(s)
- Brandon J Berry
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Box 711/604, 575 Elmwood Ave., Rochester, NY 14642, USA.
| | - Adam J Trewin
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, Box 711/604, 575 Elmwood Ave., Rochester, NY 14642, USA.
| | - Andrea M Amitrano
- Department of Pathology, University of Rochester Medical Center, Box 609, 601 Elmwood Ave., Rochester, NY 14642, USA; Department of Microbiology and Immunology, University of Rochester Medical Center, Box 609, 601 Elmwood Ave., Rochester, NY 14642, USA.
| | - Minsoo Kim
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Box 711/604, 575 Elmwood Ave., Rochester, NY 14642, USA; Department of Pathology, University of Rochester Medical Center, Box 609, 601 Elmwood Ave., Rochester, NY 14642, USA; Department of Microbiology and Immunology, University of Rochester Medical Center, Box 609, 601 Elmwood Ave., Rochester, NY 14642, USA.
| | - Andrew P Wojtovich
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Box 711/604, 575 Elmwood Ave., Rochester, NY 14642, USA; Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, Box 711/604, 575 Elmwood Ave., Rochester, NY 14642, USA.
| |
Collapse
|
16
|
singh L, Randhawa PK, Singh N, Jaggi AS. Redox signaling in remote ischemic preconditioning-induced cardioprotection: Evidences and mechanisms. Eur J Pharmacol 2017; 809:151-155. [DOI: 10.1016/j.ejphar.2017.05.033] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Revised: 05/12/2017] [Accepted: 05/15/2017] [Indexed: 12/16/2022]
|
17
|
Daiber A, Di Lisa F, Oelze M, Kröller‐Schön S, Steven S, Schulz E, Münzel T. Crosstalk of mitochondria with NADPH oxidase via reactive oxygen and nitrogen species signalling and its role for vascular function. Br J Pharmacol 2017; 174:1670-1689. [PMID: 26660451 PMCID: PMC5446573 DOI: 10.1111/bph.13403] [Citation(s) in RCA: 183] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 09/22/2015] [Accepted: 11/30/2015] [Indexed: 12/21/2022] Open
Abstract
Cardiovascular diseases are associated with and/or caused by oxidative stress. This concept has been proven by using the approach of genetic deletion of reactive species producing (pro-oxidant) enzymes as well as by the overexpression of reactive species detoxifying (antioxidant) enzymes leading to a marked reduction of reactive oxygen and nitrogen species (RONS) and in parallel to an amelioration of the severity of diseases. Likewise, the development and progression of cardiovascular diseases is aggravated by overexpression of RONS producing enzymes as well as deletion of antioxidant RONS detoxifying enzymes. Thus, the consequences of the interaction (redox crosstalk) of superoxide/hydrogen peroxide produced by mitochondria with other ROS producing enzymes such as NADPH oxidases (Nox) are of outstanding importance and will be discussed including the consequences for endothelial nitric oxide synthase (eNOS) uncoupling as well as the redox regulation of the vascular function/tone in general (soluble guanylyl cyclase, endothelin-1, prostanoid synthesis). Pathways and potential mechanisms leading to this crosstalk will be analysed in detail and highlighted by selected examples from the current literature including hypoxia, angiotensin II-induced hypertension, nitrate tolerance, aging and others. The general concept of redox-based activation of RONS sources via "kindling radicals" and enzyme-specific "redox switches" will be discussed providing evidence that mitochondria represent key players and amplifiers of the burden of oxidative stress. LINKED ARTICLES This article is part of a themed section on Redox Biology and Oxidative Stress in Health and Disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.12/issuetoc.
Collapse
Affiliation(s)
- Andreas Daiber
- Center for Cardiology, Laboratory of Molecular CardiologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
| | - Fabio Di Lisa
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly
| | - Matthias Oelze
- Center for Cardiology, Laboratory of Molecular CardiologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
| | - Swenja Kröller‐Schön
- Center for Cardiology, Laboratory of Molecular CardiologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
| | - Sebastian Steven
- Center for Cardiology, Laboratory of Molecular CardiologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
- Center of Thrombosis and HemostasisMedical Center of the Johannes Gutenberg UniversityMainzGermany
| | - Eberhard Schulz
- Center for Cardiology, Laboratory of Molecular CardiologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
| | - Thomas Münzel
- Center for Cardiology, Laboratory of Molecular CardiologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
| |
Collapse
|
18
|
Tullio F, Penna C, Cabiale K, Femminò S, Galloni M, Pagliaro P. Cardioprotective effects of calcitonin gene-related peptide in isolated rat heart and in H9c2 cells via redox signaling. Biomed Pharmacother 2017; 90:194-202. [PMID: 28364596 DOI: 10.1016/j.biopha.2017.03.043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 03/10/2017] [Accepted: 03/14/2017] [Indexed: 02/02/2023] Open
Abstract
The calcitonin-gene-related-peptide (CGRP) release is coupled to the signaling of Angeli's salt in determining vasodilator effects. However, it is unknown whether CGRP is involved in Angeli's salt cardioprotective effects and which are the mechanisms of protection. We aimed to determine whether CGRP is involved in myocardial protection induced by Angeli's salt. We also analyzed the intracellular signaling pathway activated by CGRP. Isolated rat hearts were pre-treated with Angeli's salt or Angeli's salt plus CGRP8-37, a specific CGRP-receptor antagonist, and subjected to ischemia (30-min) and reperfusion (120-min). Moreover, we studied CGRP-induced protection during oxidative stress (H2O2) and hypoxia/reoxygenation protocols in H9c2 cardiomyocytes. Cell vitality and mitochondrial membrane potential (ΔYm, MMP) were measured using MTT and JC-1 dyes. Angeli's salt reduced infarct size and ameliorated post-ischemic cardiac function via a CGRP-receptor-dependent mechanism. Pre-treatment with CGRP increased H9c2 survival upon challenging with either H2O2 (redox stress) or hypoxia/reoxygenation (H/R stress). Under these stress conditions, reduction in MMP and cell death were partly prevented by CGRP. These CGRP beneficial effects were blocked by CGRP8-37. During H/R stress, pre-treatment with either CGRP-receptor, protein kinase C (PKC) or mitochondrial KATP channel antagonists, and pre-treatment with an antioxidant (2-mercaptopropionylglycine) blocked the protection mediated by CGRP. In conclusion, CGRP is involved in the cardioprotective effects of Angeli's salt. In H9c2 cardiomyocytes, CGRP elicits PKC-dependent and mitochondrial-KATP-redox-dependent mechanisms. Hence, CGRP is an important factor in the redox-sensible cardioprotective effects of Angeli's salt.
Collapse
Affiliation(s)
- Francesca Tullio
- Department of Clinical and Biological Sciences, University of Turin, Italy
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, Italy.
| | - Karine Cabiale
- Department of Clinical and Biological Sciences, University of Turin, Italy; Department of Veterinary Science, University of Torino, Italy
| | - Saveria Femminò
- Department of Clinical and Biological Sciences, University of Turin, Italy
| | - Marco Galloni
- Department of Veterinary Science, University of Torino, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, Italy.
| |
Collapse
|
19
|
Onukwufor JO, Stevens D, Kamunde C. Bioenergetic and volume regulatory effects of mitoKATP channel modulators protect against hypoxia-reoxygenation-induced mitochondrial dysfunction. ACTA ACUST UNITED AC 2016; 219:2743-51. [PMID: 27358470 DOI: 10.1242/jeb.140186] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 06/26/2016] [Indexed: 12/19/2022]
Abstract
The mitochondrial ATP-sensitive K(+) (mitoKATP) channel plays a significant role in mitochondrial physiology and protects against ischemic reperfusion injury in mammals. Although fish frequently face oxygen fluctuations in their environment, the role of the mitoKATP channel in regulating the responses to oxygen stress is rarely investigated in this class of animals. To elucidate whether and how the mitoKATP channel protects against hypoxia-reoxygenation (H-R)-induced mitochondrial dysfunction in fish, we first determined the mitochondrial bioenergetic effects of two key modulators of the channel, diazoxide and 5-hydroxydecanoate (5-HD), using a wide range of doses. Subsequently, the effects of low and high doses of the modulators on mitochondrial bioenergetics and volume under normoxia and after H-R using buffers with and without magnesium and ATP (Mg-ATP) were tested. In the absence of Mg-ATP (mitoKATP channel open), both low and high doses of diazoxide improved mitochondrial coupling, but only the high dose of 5-HD reversed the post-H-R coupling-enhancing effect of diazoxide. In the presence of Mg-ATP (mitoKATP channel closed), diazoxide at the low dose improved coupling post-H-R, and this effect was abolished by 5-HD at the low dose. Interestingly, both low and high doses of diazoxide reversed H-R-induced swelling under mitoKATP channel open conditions, but this effect was not sensitive to 5-HD. Under mitoKATP channel closed conditions, diazoxide at the low dose protected the mitochondria from H-R-induced swelling and 5-HD at the low dose reversed this effect. In contrast, diazoxide at the high dose failed to reduce the swelling caused by H-R, and the addition of the high dose of 5-HD enhanced mitochondrial swelling. Overall, our study showed that in the presence of Mg-ATP, both opening of mitoKATP channels and bioenergetic effects of diazoxide were protective against H-R in fish mitochondria, while in the absence of Mg-ATP only the bioenergetic effect of diazoxide was protective.
Collapse
Affiliation(s)
- John O Onukwufor
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, Prince Edward Island, Canada C1A 4P3
| | - Don Stevens
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, Prince Edward Island, Canada C1A 4P3
| | - Collins Kamunde
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, Prince Edward Island, Canada C1A 4P3
| |
Collapse
|
20
|
Arsyad A, Dobson GP. Adenosine relaxation in isolated rat aortic rings and possible roles of smooth muscle Kv channels, KATP channels and A2a receptors. BMC Pharmacol Toxicol 2016; 17:23. [PMID: 27211886 PMCID: PMC4876563 DOI: 10.1186/s40360-016-0067-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 04/29/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND An area of ongoing controversy is the role adenosine to regulate vascular tone in conduit vessels that regulate compliance, and the role of nitric oxide (NO), potassium channels and receptor subtypes involved. The aim of our study was to investigate adenosine relaxation in rat thoracic aortic rings, and the effect of inhibitors of NO, prostanoids, Kv, KATP channels, and A2a and A2b receptors. METHODS Aortic rings were freshly harvested from adult male Sprague Dawley rats and equilibrated in an organ bath containing oxygenated, modified Krebs-Henseleit solution, 11 mM glucose, pH 7.4, 37 °C. Isolated rings were pre-contracted sub-maximally with 0.3 μM norepinephrine (NE), and the effect of increasing concentrations of adenosine (1 to 1000 μM) were examined. The drugs L-NAME, indomethacin, 4-aminopyridine (4-AP), glibenclamide, 5-hydroxydecanoate, ouabain, 8-(3-chlorostyryl) caffeine and PSB-0788 were examined in intact and denuded rings. Rings were tested for viability after each experiment. RESULTS Adenosine induced a dose-dependent, triphasic relaxation response, and the mechanical removal of the endothelium significantly deceased adenosine relaxation above 10 μM. Interestingly, endothelial removal significantly decreased the responsiveness (defined as % relaxation per μM adenosine) by two-thirds between 10 and 100 μM, but not in the lower (1-10 μM) or higher (>100 μM) ranges. In intact rings, L-NAME significantly reduced relaxation, but not indomethacin. Antagonists of voltage-dependent Kv (4-AP), sarcolemma KATP (glibenclamide) and mitochondrial KATP channels (5-HD) led to significant reductions in relaxation in both intact and denuded rings, with ouabain having little or no effect. Adenosine-induced relaxation appeared to involve the A2a receptor, but not the A2b subtype. CONCLUSIONS It was concluded that adenosine relaxation in NE-precontracted rat aortic rings was triphasic and endothelium-dependent above 10 μM, and relaxation involved endothelial nitric oxide (not prostanoids) and a complex interplay between smooth muscle A2a subtype and voltage-dependent Kv, SarcKATP and MitoKATP channels. The possible in vivo significance of the regulation of arterial compliance to left ventricular function coupling is discussed.
Collapse
Affiliation(s)
- Aryadi Arsyad
- Physiology Department, Medical Faculty, Hasanuddin University, Jl. Perintis Kemerdekaan, Km. 10, Tamalanrea, Makassar, 90213, Indonesia
| | - Geoffrey P Dobson
- Heart, Trauma and Sepsis Research Laboratory, Australian Institute of Tropical Health and Medicine, College of Medicine and Dentistry, James Cook University, 1 James Cook Drive, Queensland, 4811, Australia.
| |
Collapse
|
21
|
Yang Y, Chen X, Min H, Song S, Zhang J, Fan S, Yi L, Wang H, Gu X, Ma Z, Gao Q. Persistent mitoKATP Activation Is Involved in the Isoflurane-induced Cytotoxicity. Mol Neurobiol 2016; 54:1101-1110. [DOI: 10.1007/s12035-016-9710-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 01/11/2016] [Indexed: 01/27/2023]
|
22
|
Petruş A, Duicu OM, Sturza A, Noveanu L, Kiss L, Dănilă M, Baczkó I, Muntean DM, Jost N. Modulation of mitochondrial respiratory function and ROS production by novel benzopyran analogues. Can J Physiol Pharmacol 2015; 93:811-8. [DOI: 10.1139/cjpp-2015-0041] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A substantial body of evidence indicates that pharmacological activation of mitochondrial ATP-sensitive potassium channels (mKATP) in the heart is protective in conditions associated with ischemia/reperfusion injury. Several mechanisms have been postulated to be responsible for cardioprotection, including the modulation of mitochondrial respiratory function. The aim of the present study was to characterize the dose-dependent effects of novel synthetic benzopyran analogues, derived from a BMS-191095, a selective mKATP opener, on mitochondrial respiration and reactive oxygen species (ROS) production in isolated rat heart mitochondria. Mitochondrial respiratory function was assessed by high-resolution respirometry, and H2O2 production was measured by the Amplex Red fluorescence assay. Four compounds, namely KL-1487, KL-1492, KL-1495, and KL-1507, applied in increasing concentrations (50, 75, 100, and 150 μmol/L, respectively) were investigated. When added in the last two concentrations, all compounds significantly increased State 2 and 4 respiratory rates, an effect that was not abolished by 5-hydroxydecanoate (5-HD, 100 μmol/L), the classic mKATP inhibitor. The highest concentration also elicited an important decrease of the oxidative phosphorylation in a K+ independent manner. Both concentrations of 100 and 150 μmol/L for KL-1487, KL-1492, and KL-1495, and the concentration of 150 μmol/L for KL-1507, respectively, mitigated the mitochondrial H2O2 release. In isolated rat heart mitochondria, the novel benzopyran analogues act as protonophoric uncouplers of oxidative phosphorylation and decrease the generation of reactive oxygen species in a dose-dependent manner.
Collapse
Affiliation(s)
- Alexandra Petruş
- Department of Pathophysiology, “Victor Babeş” University of Medicine and Pharmacy of Timişoara, 14, Tudor Vladimirescu st. 300173 Timisoara, Romania
| | - Oana M. Duicu
- Department of Pathophysiology, Center for Translational Research and Systems Medicine, “Victor Babeş” University of Medicine and Pharmacy of Timişoara, Romania
| | - Adrian Sturza
- Department of Pathophysiology, Center for Translational Research and Systems Medicine, “Victor Babeş” University of Medicine and Pharmacy of Timişoara, Romania
| | - Lavinia Noveanu
- Department of Pathophysiology, Center for Translational Research and Systems Medicine, “Victor Babeş” University of Medicine and Pharmacy of Timişoara, Romania
| | - Loránd Kiss
- Institute of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Szeged, Szeged, Hungary
| | - Maria Dănilă
- Department of Pathophysiology, Center for Translational Research and Systems Medicine, “Victor Babeş” University of Medicine and Pharmacy of Timişoara, Romania
| | - István Baczkó
- Department of Pathophysiology, “Victor Babeş” University of Medicine and Pharmacy of Timişoara, 14, Tudor Vladimirescu st. 300173 Timisoara, Romania
| | - Danina M. Muntean
- Department of Pathophysiology, Center for Translational Research and Systems Medicine, “Victor Babeş” University of Medicine and Pharmacy of Timişoara, Romania
| | - Norbert Jost
- Department of Pathophysiology, Center for Translational Research and Systems Medicine, “Victor Babeş” University of Medicine and Pharmacy of Timişoara, Romania
| |
Collapse
|
23
|
Marongiu E, Crisafulli A. Cardioprotection acquired through exercise: the role of ischemic preconditioning. Curr Cardiol Rev 2014; 10:336-48. [PMID: 24720421 PMCID: PMC4101198 DOI: 10.2174/1573403x10666140404110229] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 12/16/2013] [Accepted: 03/28/2014] [Indexed: 02/07/2023] Open
Abstract
A great bulk of evidence supports the concept that regular exercise training can reduce the incidence of coronary events and increase survival chances after myocardial infarction. These exercise-induced beneficial effects on the myocardium are reached by means of the reduction of several risk factors relating to cardiovascular disease, such as high cholesterol, hypertension, obesity etc. Furthermore, it has been demonstrated that exercise can reproduce the "ischemic preconditioning" (IP), which refers to the capacity of short periods of ischemia to render the myocardium more resistant to subsequent ischemic insult and to limit infarct size during prolonged ischemia. However, IP is a complex phenomenon which, along with infarct size reduction, can also provide protection against arrhythmia and myocardial stunning due to ischemia-reperfusion. Several clues demonstrate that preconditioning may be directly induced by exercise, thus inducing a protective phenotype at the heart level without the necessity of causing ischemia. Exercise appears to act as a physiological stress that induces beneficial myocardial adaptive responses at cellular level. The purpose of the present paper is to review the latest data on the role played by exercise in triggering myocardial preconditioning.
Collapse
|
24
|
Abstract
The field of mitochondrial ion channels has recently seen substantial progress, including the molecular identification of some of the channels. An integrative approach using genetics, electrophysiology, pharmacology, and cell biology to clarify the roles of these channels has thus become possible. It is by now clear that many of these channels are important for energy supply by the mitochondria and have a major impact on the fate of the entire cell as well. The purpose of this review is to provide an up-to-date overview of the electrophysiological properties, molecular identity, and pathophysiological functions of the mitochondrial ion channels studied so far and to highlight possible therapeutic perspectives based on current information.
Collapse
|
25
|
Shin BS, Kim HG, Choi OH. Mitochondrial Channel Opener Diazoxide Attenuates Hypoxia-Induced sFlt-1 Release in Human Choriocarcinoma Cells. J Menopausal Med 2014; 20:21-31. [PMID: 25371888 PMCID: PMC4217563 DOI: 10.6118/jmm.2014.20.1.21] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 03/07/2014] [Accepted: 03/07/2014] [Indexed: 11/16/2022] Open
Abstract
Objectives To examine the effect of diazoxide on hypoxia-induced soluble fms-like tyrosin kinase-1 (sFlt-1) release in JEG-3 choriocarcinoma cells. Methods Cells were cultured under normoxia (20% O2) or hypoxia (1% O2), and expression of sFlt-1 mRNA and protein release was determined by quantitative real-time reverse-transcriptase polymerase chain reaction (qRT-PCR) assays and enzyme-linked immunosorbent assay (ELISA). Results Tumor necrosis factor-alpha (TNF-α) as well as hypoxia stimulated sFlt-1 release and diazoxide inhibited both of them. The selective inhibitor of mitochondrial adenosine triphosphat (ATP)-sensitive K+ channel opener (KATP) 5-hydroxydecanoate (5-HD) completely reversed the diazoxide-induced inhibition of hypoxia-stimulated sFlt-1 release. qRT-PCR and Western blot analyses showed that diazoxide up-regulated the heme oxygenase-1 (HO-1) expression. In addition, the HO-1 inducer cobalt protoporphyrin (CoPP) and the metabolic product of HO-1 bilirubin mimicked diazoxide to inhibit sFlt-1 release and reactive oxygen species (ROS) production under hypoxia, whereas the HO-1 inhibitor zinc protoporphyrin IX (ZnPP IX) antagonized the effect of diazoxide. In cells transfected with the HO-1 siRNA, diazoxide did not exert any effect on sFlt-1 release and ROS production under hypoxia. Conclusion These results, taken together, strongly suggest that up-regulation of the HO-1 expression is the crucial mechanism responsible for the diazoxide-induced inhibition of the sFlt-1 release and ROS production under hypoxia.
Collapse
Affiliation(s)
- Byeong Seop Shin
- Department of Obstetrics and Gynecology, Pusan National University School of Medicine, Busan, Korea
| | - Hwi Gon Kim
- Department of Obstetrics and Gynecology, Pusan National University School of Medicine, Busan, Korea
| | - Ook Hwan Choi
- Department of Obstetrics and Gynecology, Pusan National University School of Medicine, Busan, Korea
| |
Collapse
|
26
|
Dymkowska D, Drabarek B, Jakubczyk J, Wojciechowska S, Zabłocki K. Potassium channel openers prevent palmitate-induced insulin resistance in C2C12 myotubes. Arch Biochem Biophys 2013; 541:47-52. [PMID: 24262853 DOI: 10.1016/j.abb.2013.11.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 10/28/2013] [Accepted: 11/08/2013] [Indexed: 10/26/2022]
Abstract
Insulin resistance (IR) of muscle cells is an early symptom of type 2 diabetes. It often results from excessive lipid accumulation in muscle fibers which under in vitro experimental conditions may be induced by incubation of muscle cells with palmitate. IR is manifested as a reduced response of cells to insulin expressed by lowered Akt kinase phosphorylation and decreased insulin-dependent glucose uptake. Stimulation of mitochondrial oxidative metabolism by mild dissipation of the mitochondrial potential is thought to increase fatty acid utilization and thereby prevent insulin resistance. Here it is shown that nicorandil and NS1619, which are openers of two different mitochondrial potassium channels, protect C2C12 myotubes from palmitate-induced insulin resistance. Preincubation of myotubes with 5-hydroxydecanoate abolishes the protective effect of nicorandil. The efficient concentrations of both openers are far below those commonly applied for cytoprotection. This is probably why their effects on the mitochondrial energy metabolism are small. These data suggest that opening of mitochondrial potassium channels could be a promising approach in prevention and therapy of insulin resistance related to dyslipidemia and obesity.
Collapse
Affiliation(s)
| | - Beata Drabarek
- Nencki Institute of Experimental Biology, Warsaw, Poland
| | | | | | | |
Collapse
|
27
|
Krenz M, Baines C, Kalogeris T, Korthuis R. Cell Survival Programs and Ischemia/Reperfusion: Hormesis, Preconditioning, and Cardioprotection. ACTA ACUST UNITED AC 2013. [DOI: 10.4199/c00090ed1v01y201309isp044] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
28
|
Zeng Z, Huang HF, He F, Wu LX, Lin J, Chen MQ. Diazoxide attenuates ischemia/reperfusion injury via upregulation of heme oxygenase-1 after liver transplantation in rats. World J Gastroenterol 2012; 18:1765-72. [PMID: 22553400 PMCID: PMC3332289 DOI: 10.3748/wjg.v18.i15.1765] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Revised: 01/17/2012] [Accepted: 04/09/2012] [Indexed: 02/06/2023] Open
Abstract
AIM: To evaluate the effects of diazoxide on ischemia/reperfusion (I/R)-injured hepatocytes and further elucidate its underlying mechanisms.
METHODS: Male Sprague-Dawley rats were randomized (8 for donor and recipient per group) into five groups: I/R group (4 h of liver cold ischemia followed by 6 h of reperfusion); heme oxygenase-1 (HO-1) small interfering RNA (siRNA) group (injection of siRNA via donor portal vein 48 h prior to harvest); diazoxide (DZ) group (injection of DZ via donor portal vein 10 min prior to harvest); HO-1 siRNA + DZ group; and siRNA control group. Blood and liver samples were collected at 6 h after reperfusion. The mRNA expressions and protein levels of HO-1 were determined by reverse transcription polymerase chain reaction and Western blotting, and tissue morphology was examined by light and transmission electron microscopy. Serum transaminases level and cytokines concentration were also measured.
RESULTS: We observed that a significant reduction of HO-1 mRNA and protein levels in HO-1 siRNA and HO-1 siRNA + DZ group when compared with I/R group, while the increases were prominent in the DZ group. Light and transmission electron microscopy indicated severe disruption of tissue with lobular distortion and mitochondrial cristae damage in the HO-1 siRNA and HO-1 siRNA + DZ groups compared with DZ group. Serum alanine aminotransferase, aspartate transaminase, tumor necrosis factor-α and interleukin-6 levels increased in the HO-1 siRNA and HO-1 siRNA + DZ groups, and decreased in the DZ group.
CONCLUSION: The protective effect of DZ may be induced by upregulation of HO-1. By inhibiting expression of HO-1, this protection pretreated with DZ was abolished.
Collapse
|
29
|
Szabò I, Leanza L, Gulbins E, Zoratti M. Physiology of potassium channels in the inner membrane of mitochondria. Pflugers Arch 2011; 463:231-46. [PMID: 22089812 DOI: 10.1007/s00424-011-1058-7] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2011] [Accepted: 10/30/2011] [Indexed: 02/06/2023]
Abstract
The inner membrane of the ATP-producing organelles of endosymbiotic origin, mitochondria, has long been considered to be poorly permeable to cations and anions, since the strict control of inner mitochondrial membrane permeability is crucial for efficient ATP synthesis. Over the past 30 years, however, it has become clear that various ion channels--along with antiporters and uniporters--are present in the mitochondrial inner membrane, although at rather low abundance. These channels are important for energy supply, and some are a decisive factor in determining whether a cell lives or dies. Their electrophysiological and pharmacological characterisations have contributed importantly to the ongoing elucidation of their pathophysiological roles. This review gives an overview of recent advances in our understanding of the functions of the mitochondrial potassium channels identified so far. Open issues concerning the possible molecular entities giving rise to the observed activities and channel protein targeting to mitochondria are also discussed.
Collapse
Affiliation(s)
- Ildikò Szabò
- Department of Biology, University of Padova, Padova, Italy.
| | | | | | | |
Collapse
|
30
|
Markou T, Makridou Z, Galatou E, Lazou A. Multiple signalling pathways underlie the protective effect of levosimendan in cardiac myocytes. Eur J Pharmacol 2011; 667:298-305. [PMID: 21664904 DOI: 10.1016/j.ejphar.2011.05.078] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2010] [Revised: 04/18/2011] [Accepted: 05/23/2011] [Indexed: 11/26/2022]
Abstract
Levosimendan is a cardiovascular drug for the treatment of acute and decompensated heart failure. The current weight of evidence on the cardioprotective effects of levosimendan originates from whole heart models and there is no information on the mechanism whereby signalling pathways are activated. In the present study, we investigated the effect of levosimendan on ischaemia/reperfusion injury and the underlying mechanism in cardiac myocytes. Pretreatment with levosimendan reversed the effects of ischaemia and ischaemia/reperfusion on cell viability and enhanced phosphorylation of Akt, p38-mitogen activated protein kinase (MAPK) and extracellular signal-regulated kinases 1/2 (ERK1/2). Inhibitors of these kinases and the blocker of the mitochondrial K(ATP) channels, 5-hydroxydecanoate, completely abolished the protection afforded by levosimendan. Levosimendan stimulated the phosphorylation of Akt, ERK1/2 and p38-MAPK with different kinetics and the activation of these pathways was dependent on the opening of the mitochondrial K(ATP) channels and the production of oxygen free radicals. The levosimendan-induced phosphorylation of ERK1/2 and Akt was reduced by inhibitors of epidermal growth factor receptor and Src. On the other hand, inhibition of the protein kinase A (PKA) pathway reduced phosphorylation of p38-MAPK. Furthermore, p38-MAPK was activated when a phosphodiesterase inhibitor or a selective PKA activator was used. Overall, our results suggest that levosimendan regulates the wiring of the natural salvaging pathways to execute the prosurvival signals. This network includes Akt, ERK1/2 and p38-MAPK. Opening of mitochondrial K(ATP) channels and the subsequent production of oxygen free radicals, the epidermal growth factor receptor/Src, and the cAMP/PKA pathways seem to mediate this response.
Collapse
Affiliation(s)
- Thomais Markou
- Laboratory of Animal Physiology, Department of Zoology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | | | | | | |
Collapse
|
31
|
Parga JA, Rodríguez-Pallares J, Joglar B, Diaz-Ruiz C, Guerra MJ, Labandeira-Garcia JL. Effect of inhibitors of NADPH oxidase complex and mitochondrial ATP-sensitive potassium channels on generation of dopaminergic neurons from neurospheres of mesencephalic precursors. Dev Dyn 2011; 239:3247-59. [PMID: 21046630 DOI: 10.1002/dvdy.22474] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Reactive oxygen species signaling has been suggested to regulate stem cell development. In the present study, we treated neurospheres of rat mesencephalic precursors with inhibitors of the NADPH oxidase complex and mitochondrial ATP-sensitive potassium (mitoKATP) channel blockers during the proliferation and/or the differentiation periods to study the effects on generation of dopaminergic neurons. Treatment with low doses (100 or 250 μM) of the NADPH inhibitor apocynin during the proliferation period increased the generation of dopaminergic neurons. However, higher doses (1 mM) were necessary during the differentiation period to induce the same effect. Treatment with general (glibenclamide) or mitochondrial (5-hydroxydecanoate) KATP channel blockers during the proliferation and differentiation periods increased the number of dopaminergic neurons. Furthermore, neither increased proliferation rate nor apoptosis had a major role in the observed increase in generation of dopaminergic neurons, which suggests that the redox state is able to regulate differentiation of precursors into dopaminergic neurons.
Collapse
Affiliation(s)
- J A Parga
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, University of Santiago de Compostela, Santiago de Compostela, Spain
| | | | | | | | | | | |
Collapse
|
32
|
Lavi R, Shainberg A, Shneyvays V, Hochauser E, Isaac A, Zinman T, Friedmann H, Lubart R. Detailed analysis of reactive oxygen species induced by visible light in various cell types. Lasers Surg Med 2010; 42:473-80. [PMID: 20662023 DOI: 10.1002/lsm.20919] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND AND OBJECTIVE Light in the visible and near infrared region stimulates various cellular processes, and thus has been used for therapeutic purposes. One of the proposed mechanisms is based on cellular production of reactive oxygen species (ROS) in response to illumination. In the present study, we followed visible light (VL)-induced hydroxyl radicals in various cell types and cellular sites using the electron paramagnetic resonance (EPR) spin-trapping technique. MATERIALS AND METHODS Fibroblasts, sperm cells, cardiomyocytes, and skeletal muscle cells were irradiated with broadband (400-800 nm) VL. To detect ROS, the EPR spin-trapping technique coupled with the spin-traps 5,5-dimethyl pyrroline-N-oxide (DMPO) or 5-(diethoxyphosphoryl)-5-methyl-1-pyrroline-N-oxide (DEPMPO) were used. To investigate the cellular sites of ROS formation, the cell-permeable molecule, isopropanol, or the nonpermeable proteins, bovine serum albumin (BSA) and superoxide dismutase (SOD), were introduced to the cells before irradiation. ROS production in mitochondria was measured using the fluorescent probe, MitoTracker Red (MTR). RESULTS AND CONCLUSIONS The concentration of .OH increased both with illumination time and with cell concentration, and decreased when N(2) was bubbled into the cell culture, suggesting that VL initiates a photochemical reaction via endogenous photosensitizers. VL was found to stimulate ROS generation both in membrane and cytoplasm. In addition, fluorescent measurments confirmed the mitochondria to be target for light-cell interaction. The findings support the hypothesis that ROS are generated in various cellular sites following light illumination.
Collapse
Affiliation(s)
- Ronit Lavi
- Department of Chemistry, Bar-Ilan University, Ramat-Gan 52900, Israel.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Giorgi C, Agnoletto C, Baldini C, Bononi A, Bonora M, Marchi S, Missiroli S, Patergnani S, Poletti F, Rimessi A, Zavan B, Pinton P. Redox control of protein kinase C: cell- and disease-specific aspects. Antioxid Redox Signal 2010; 13:1051-85. [PMID: 20136499 DOI: 10.1089/ars.2009.2825] [Citation(s) in RCA: 286] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Hormones, growth factors, electrical stimulation, and cell-cell interactions regulate numerous cellular processes by altering the levels of second messengers, thus influencing biochemical reactions inside the cells. The Protein Kinase C family (PKCs) is a group of serine/threonine kinases that are dependent on calcium (Ca(2+)), diacylglycerol, and phospholipids. Signaling pathways that induce variations on the levels of PKC activators have been implicated in the regulation of diverse cellular functions and, in turn, PKCs are key regulators of a plethora of cellular processes, including proliferation, differentiation, and tumorigenesis. Importantly, PKCs contain regions, both in the N-terminal regulatory domain and in the C-terminal catalytic domain, that are susceptible to redox modifications. In several pathophysiological conditions when the balance between oxidants, antioxidants, and alkylants is compromised, cells undergo redox stress. PKCs are cell-signaling proteins that are particularly sensitive to redox stress because modification of their redox-sensitive regions interferes with their activity and, thus, with their biological effects. In this review, we summarize the involvement of PKCs in health and disease and the importance of redox signaling in the regulation of this family of kinases.
Collapse
Affiliation(s)
- Carlotta Giorgi
- Department of Experimental and Diagnostic Medicine, Section of General Pathology, Interdisciplinary Center for the Study of Inflammation (ICSI), BioPharmaNet, University of Ferrara, Ferrara, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Chai Y, Lin YF. Stimulation of neuronal KATP channels by cGMP-dependent protein kinase: involvement of ROS and 5-hydroxydecanoate-sensitive factors in signal transduction. Am J Physiol Cell Physiol 2010; 298:C875-92. [PMID: 20053925 PMCID: PMC2853218 DOI: 10.1152/ajpcell.00196.2009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2009] [Accepted: 12/31/2009] [Indexed: 11/22/2022]
Abstract
The ATP-sensitive potassium (K(ATP)) channel couples intracellular metabolic state to membrane excitability. Recently, we demonstrated that neuronal K(ATP) channels are functionally enhanced by activation of a nitric oxide (NO)/cGMP/cGMP-dependent protein kinase (PKG) signaling cascade. In this study, we further investigated the intracellular mechanism underlying PKG stimulation of neuronal K(ATP) channels. By performing single-channel recordings in transfected HEK293 and neuroblastoma SH-SY5Y cells, we found that the increase of Kir6.2/SUR1 (i.e., the neuronal-type K(ATP)) channel currents by PKG activation in cell-attached patches was diminished by 5-hydroxydecanoate (5-HD), an inhibitor of the putative mitochondrial K(ATP) channel; N-(2-mercaptopropionyl)glycine, a reactive oxygen species (ROS) scavenger, and catalase, a hydrogen peroxide (H(2)O(2))-decomposing enzyme. These reagents also ablated NO-induced K(ATP) channel stimulation and prevented the shifts in the single-channel open- and closed-time distributions resulting from PKG activation and NO induction. Bath application of H(2)O(2) reproduced PKG stimulation of Kir6.2/SUR1 but did not activate tetrameric Kir6.2LRKR368/369/370/371AAAA channels. Moreover, neither the PKG activator nor exogenous H(2)O(2) was able to enhance the function of K(ATP) channels in the presence of Ca(2+) chelators and calmodulin antagonists, whereas the stimulatory effect of H(2)O(2) was unaffected by 5-HD. Altogether, in this report we provide novel evidence that activation of PKG stimulates neuronal K(ATP) channels by modulating intrinsic channel gating via a 5-HD-sensitive factor(s)/ROS/Ca(2+)/calmodulin signaling pathway that requires the presence of the SUR1 subunit. This signaling pathway may contribute to neuroprotection against ischemic injury and regulation of neuronal excitability and neurotransmitter release by modulating the function of neuronal K(ATP) channels.
Collapse
Affiliation(s)
- Yongping Chai
- Dept. of Physiology and Membrane Biology, Univ. of California, Davis, One Shields Ave., Davis, CA 95616, USA
| | | |
Collapse
|
35
|
An investigation of the occurrence and properties of the mitochondrial intermediate-conductance Ca2+-activated K+ channel mtKCa3.1. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2009; 1797:1260-7. [PMID: 20036632 DOI: 10.1016/j.bbabio.2009.12.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2009] [Revised: 12/15/2009] [Accepted: 12/19/2009] [Indexed: 12/24/2022]
Abstract
The mitochondrial intermediate-conductance Ca2+-activated K+ channel mtKCa3.1 has recently been discovered in the HCT116 colon tumor-derived cell line, which expresses relatively high levels of this protein also in the plasma membrane. Electrophysiological recordings revealed that the channel can exhibit different conductance states and kinetic modes, which we tentatively ascribe to post-translational modifications. To verify whether the localization of this channel in mitochondria might be a peculiarity of these cells or a more widespread feature we have checked for the presence of mtKCa3.1 in a few other cell lines using biochemical and electrophysiological approaches. It turned out to be present at least in some of the cells investigated. Functional assays explored the possibility that mtKCa3.1 might be involved in cell proliferation or play a role similar to that of the Shaker-type KV1.3 channel in lymphocytes, which interacts with outer mitochondrial membrane-inserted Bax thereby promoting apoptosis (Szabò, I. et al., Proc. Natl. Acad Sci. USA 105 (2008) 14861-14866). A specific KCa3.1 inhibitor however did not have any detectable effect on cell proliferation or death.
Collapse
|
36
|
Matejíková J, Ravingerová T, Pancza D, Čarnická S, Kolář F. Mitochondrial KATP opening confers protection against lethal myocardial injury and ischaemia-induced arrhythmias in the rat heart via PI3K/Akt-dependent and -independent mechanismsThis article is one of a selection of papers published in a special issue on Advances in Cardiovascular Research. Can J Physiol Pharmacol 2009; 87:1055-62. [DOI: 10.1139/y09-100] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Opening of mitochondrial KATP channels (mitoKATP) has been reported to underlie protection against ischaemia–reperfusion injury induced by ischaemic preconditioning (I-PC); however, the molecular mechanisms of its antiarrhythmic effect have not been fully elucidated. We explored the involvement of phosphatidylinositol 3-kinase (PI3K)/Akt in the PC-like effect of mitoKATP opener diazoxide with particular regard to its role in protection against ischaemia-induced arrhythmias. Langendorff-perfused rat hearts were subjected to 30 min LAD occlusion with or without a prior 15 min of perfusion with diazoxide (50 µmol/L) given either alone (D-PC) or in combination with the PI3K/Akt inhibitor wortmannin (100 nmol/L). In an additional protocol, ischaemia was followed by 2 h reperfusion for infarct size (IS) determination (tetrazolium staining). The total number of premature ventricular complexes over the whole period of ischaemia, episodes of ventricular tachycardia and its duration were significantly lower in the D-PC group than in the non-preconditioned controls (158 ± 20, 2 ± 0.6 and 4.6 ± 1.8 s vs. 551 ± 61, 11 ± 2 and 42 ± 8 s, respectively; p < 0.05), concomitant with a 62% reduction in the size of infarction. Wortmannin modified neither arrhythmogenesis nor IS in the non-preconditioned hearts. Bracketing of diazoxide with wortmannin did not reverse the antiarrhythmic protection, whereas the IS-limiting effect was blunted. The results indicate that in contrast with the positive role of PI3K/Akt in protection against lethal myocardial injury, its activity is not involved in suppression of ischaemia-induced arrhythmias conferred by mitoKATP opening in the rat heart.
Collapse
Affiliation(s)
- Jana Matejíková
- Institute for Heart Research, Slovak Academy of Sciences, Centre of Excellence for Cardiovascular Research, Bratislava, Slovak Republic
- Institute of Physiology, Academy of Sciences of the Czech Republic and Centre for Cardiovascular Research, Prague, Czech Republic
| | - Táňa Ravingerová
- Institute for Heart Research, Slovak Academy of Sciences, Centre of Excellence for Cardiovascular Research, Bratislava, Slovak Republic
- Institute of Physiology, Academy of Sciences of the Czech Republic and Centre for Cardiovascular Research, Prague, Czech Republic
| | - Dezider Pancza
- Institute for Heart Research, Slovak Academy of Sciences, Centre of Excellence for Cardiovascular Research, Bratislava, Slovak Republic
- Institute of Physiology, Academy of Sciences of the Czech Republic and Centre for Cardiovascular Research, Prague, Czech Republic
| | - Slávka Čarnická
- Institute for Heart Research, Slovak Academy of Sciences, Centre of Excellence for Cardiovascular Research, Bratislava, Slovak Republic
- Institute of Physiology, Academy of Sciences of the Czech Republic and Centre for Cardiovascular Research, Prague, Czech Republic
| | - František Kolář
- Institute for Heart Research, Slovak Academy of Sciences, Centre of Excellence for Cardiovascular Research, Bratislava, Slovak Republic
- Institute of Physiology, Academy of Sciences of the Czech Republic and Centre for Cardiovascular Research, Prague, Czech Republic
| |
Collapse
|
37
|
Nilakantan V, Liang H, Mortensen J, Taylor E, Johnson CP. Variable effects of the mitoK(ATP) channel modulators diazoxide and 5-HD in ATP-depleted renal epithelial cells. Mol Cell Biochem 2009; 335:211-22. [PMID: 19784759 DOI: 10.1007/s11010-009-0271-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2009] [Accepted: 09/16/2009] [Indexed: 11/26/2022]
Abstract
The role of mitochondrial K(ATP) (mitoK(ATP)) channels in renal ischemia-reperfusion injury is controversial with studies showing both protective and deleterious effects. In this study, we compared the effects of the putative mitoK(ATP) opener, diazoxide, and the mitoK(ATP) blocker, 5-hydroxydecanoate (5-HD) on cytotoxicity and apoptosis in tubular epithelial cells derived from rat (NRK-52E) and pig (LLC-PK1) following in vitro ischemic injury. Following ATP depletion-recovery, there was a significant increase in cytotoxicity in both NRK cells and LLC-PK1 cells although NRK cells were more sensitive to the injury. Diazoxide treatment attenuated cytotoxicity in both cell types and 5-HD treatment-increased cytotoxicity in the sensitive NRK cells in a superoxide-dependant manner. The protective effect of diazoxide was also reversed in the presence of 5-HD in ATP-depleted NRK cells. The ATP depletion-mediated increase in superoxide was enhanced by both diazoxide and 5-HD with the effect being more pronounced in the cells undergoing 5-HD treatment. Further, ATP depletion-induced activation of caspase-3 was decreased by diazoxide in NRK cells. In order to determine the signaling pathways involved in apoptosis, we examined the activation of Erk and JNK in ATP-depleted NRK cells. Diazoxide-activated Erk in ATP-depleted cells, but did not have any effect on JNK activation. In contrast, 5-HD did not impact Erk levels but increased JNK activation even under controlled conditions. Further, the use of a JNK inhibitor with 5-HD reversed the deleterious effects of 5-HD. This study demonstrates that in cells that are sensitive to ATP depletion-recovery, mitoK(ATP) channels protect against ATP depletion-mediated cytotoxicity and apoptosis through Erk- and JNK-dependant mechanisms.
Collapse
Affiliation(s)
- Vani Nilakantan
- Division of Transplant Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | | | | | | | | |
Collapse
|
38
|
Early Anesthetic Preconditioning in Mixed Cortical Neuronal-Glial Cell Cultures Subjected to Oxygen-Glucose Deprivation: The Role of Adenosine Triphosphate Dependent Potassium Channels and Reactive Oxygen Species in Sevoflurane-Induced Neuroprotection. Anesth Analg 2009; 108:955-63. [DOI: 10.1213/ane.0b013e318193fee7] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
39
|
Downey JM, Krieg T, Cohen MV. Mapping preconditioning's signaling pathways: an engineering approach. Ann N Y Acad Sci 2008; 1123:187-96. [PMID: 18375591 DOI: 10.1196/annals.1420.022] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Preconditioning the heart by exposure to brief cycles of ischemia-reperfusion causes it to become very resistant to ischemia-induced infarction. This protection has been shown to depend on a large number of signal transduction components whose arrangements within the cardiomyocyte are unknown. To aid the translation of this phenomenon to the clinical setting, we have attempted to map the signal transduction pathways responsible for this protection. To resolve the signaling order we have injected a signal at an intermediate point in the system transduction pathway and monitored it at a downstream site. System analysis reveals both parallel and series signaling arrangements. Separate trigger and mediator phases could be identified. The trigger phase is now well mapped. During the preconditioning ischemia, autacoids--including adenosine, opioids, and bradykinin--are released from the heart. These substances occupy their respective Gi-coupled receptors. Opioid and bradykinin receptors activate phosphatidylinositol 3-kinase (PI3-kinase) which, through phosphoinositide-dependent protein kinase, causes activation of Akt. Opioid couples through transactivation of the epidermal growth factor receptor, while bradykinin's coupling to PI3-kinase is unknown. PI3-kinase causes extracellular signal regulated kinase (ERK)-dependent activation of endothelial nitric oxide synthase. The resulting nitric oxide activates soluble guanylyl cyclase resulting in cyclic C-GMP-dependent protein kinase (PKG) activation through production of cyclic guanosine monophosphate. PKG initiates opening of ATP-sensitive potassium channels on the inner membrane of the mitochondria. Potassium entry into mitochondria causes the generation of free radicals during reperfusion when oxygen is reintroduced. Through redox signaling, these radicals activate protein kinase C (PKC) and put the heart into the protected phenotype that persists for one to two hours. Although adenosine receptors activate PI3-kinase, they also have a second direct coupling to PKC and thus bypass the mitochondrial pathway. The mediator phase occurs during the first minutes of reperfusion following the lethal ischemic insult and is still poorly defined. Briefly, PKC somehow potentiates adenosine's ability to activate signaling from low-affinity A(2b) adenosine receptors. These receptors couple to the survival kinases, Akt and ERK, believed to inhibit the formation of deadly mitochondrial permeability transition pores through the phosphorylation of glycogen synthase kinase-3beta. The proposed signaling maps reveal many points at which drugs can trigger the protected phenotype.
Collapse
Affiliation(s)
- James M Downey
- Department of Physiology and Medicine, University of South Alabama, Mobile, AL 36688, USA.
| | | | | |
Collapse
|
40
|
Dost T, Cohen MV, Downey JM. Redox signaling triggers protection during the reperfusion rather than the ischemic phase of preconditioning. Basic Res Cardiol 2008; 103:378-84. [PMID: 18347834 DOI: 10.1007/s00395-008-0718-z] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2007] [Accepted: 01/11/2008] [Indexed: 10/22/2022]
Abstract
In ischemic preconditioning (IPC) brief ischemia/reperfusion renders the heart resistant to infarction from any subsequent ischemic insult. Protection results from binding of surface receptors by ligands released during the preconditioning ischemia. The downstream pathway involves redox signaling as IPC will not protect in the presence of a free radical scavenger. To determine when in the IPC protocol the redox signaling occurs, seven groups of isolated rabbit hearts were studied. All hearts underwent 30 min of coronary branch occlusion and 2 h of reperfusion. IPC groups were subjected to 5 min of regional ischemia followed by 10 min of reperfusion prior to the 30-min coronary occlusion. The Control group had only the 30-min occlusion and 2-h reperfusion. In the second group IPC preceded the index coronary occlusion. The third group was also preconditioned, but the free radical scavenger N-2-mercaptopropionyl glycine (MPG 300 microM) was infused during the 10-min reperfusion and therefore was present in the myocardium in the distribution of the snared coronary artery during the entire reperfusion phase and also during the subsequent 30-min ischemia. In another preconditioned group MPG was added to the perfusate before the preconditioning ischemia and therefore was present in the tissue only during the preconditioning ischemia and then was washed out during reperfusion. In the fifth group MPG was added to the perfusate for only the last 5 min of the preconditioning reperfusion and therefore was present in the tissue during the last minutes of the reperfusion phase and the 30 min of ischemia. In an additional group of IPC hearts MPG was infused for only the initial 5 min of the preconditioning reperfusion and then allowed to wash out so that the scavenger was present for only the first half of the reperfusion phase. Infarct and risk zone sizes were measured by triphenyltetrazolium staining and fluorescent microspheres, resp. IPC reduced infarct size from 31.3 +/- 2.7% of the ischemic zone in control hearts to only 8.4 +/- 1.9%. MPG completely blocked IPC's protection in the third (39.4 +/- 2.8%) and sixth (36.1 +/- 7.7%) groups but did not affect its protection in groups 4 (8.1 +/- 1.5%) or 5 (7.8 +/- 1.1%). When deoxygenated buffer was used during IPC's reperfusion phase in the seventh group of hearts, protection was lost and infarct size was increased over that seen in control hearts (74.5 +/- 9.0%). Hence redox signaling occurs during the reperfusion phase of IPC, and the critical component in that reperfusion phase appears to be molecular oxygen.
Collapse
Affiliation(s)
- Turhan Dost
- Dept. of Physiology, MSB 3074, University of South Alabama, College of Medicine, Mobile, AL, 36688, USA
| | | | | |
Collapse
|
41
|
Liu Y, Yang XM, Iliodromitis EK, Kremastinos DT, Dost T, Cohen MV, Downey JM. Redox signaling at reperfusion is required for protection from ischemic preconditioning but not from a direct PKC activator. Basic Res Cardiol 2008; 103:54-9. [PMID: 17999029 PMCID: PMC2660167 DOI: 10.1007/s00395-007-0683-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2007] [Accepted: 10/01/2007] [Indexed: 10/22/2022]
Abstract
Redox signaling prior to a lethal ischemic insult is an important step in triggering the protected state in ischemic preconditioning. When the preconditioned heart is reperfused a second sequence of signal transduction events, the mediator pathway, occurs which is believed to inhibit mitochondrial permeability transition pore formation that normally destroys mitochondria in much of the reperfused tissue. Prominent among the mediator pathway's events is activation of phosphatidylinositol 3-kinase and extracellular signal-regulated kinase. Recently it was found that both activation of PKC and generation of reactive oxygen species (ROS) at the time of reperfusion are required for protection in preconditioned hearts. To establish their relative order we tested whether ROS formation at reperfusion is required in hearts protected by direct activation of PKC at reperfusion. Isolated rabbit hearts were exposed to 30 min of regional ischemia and 2 h of reperfusion. Preconditioned hearts received 5 min of global ischemia and 10 min of reperfusion prior to the index ischemia. Another group of preconditioned hearts was exposed to 300 microM of the ROS scavenger N-(2-mercaptopropionyl) glycine (MPG) for 20 min starting 5 min prior to reperfusion. Infarct size was measured by triphenyltetrazolium staining. Preconditioning reduced infarct size from 36% +/- 2% of the ischemic zone in control hearts to only 18 +/- 2%. MPG during early reperfusion completely blocked preconditioning's protection (33 +/- 3% infarction). MPG given in the same dose and schedule to non-preconditioned hearts had no effect on infarct size. In the last group phorbol 12-myristate 13-acetate (PMA) (0.05 nM) was given to non-preconditioned hearts from 1 min before to 5 min after reperfusion in addition to MPG administered as in the other groups. MPG did not block protection from an infusion of PMA as infarct size was only 9 +/- 2% of the risk zone. We conclude that while redox signaling during the first few minutes of reperfusion is an essential component of preconditioning's protective mechanism, this step occurs upstream of PKC activation.
Collapse
Affiliation(s)
- Yanping Liu
- Department of Physiology, University of South Alabama College of Medicine, Mobile, Alabama, USA
| | - Xi-Ming Yang
- Department of Physiology, University of South Alabama College of Medicine, Mobile, Alabama, USA
| | - Efstathios K. Iliodromitis
- Second University Department of Cardiology, Attikon General Hospital, Medical School, University of Athens, Athens, Greece
| | - Dimitrios Th. Kremastinos
- Second University Department of Cardiology, Attikon General Hospital, Medical School, University of Athens, Athens, Greece
| | - Turhan Dost
- Department of Physiology, University of South Alabama College of Medicine, Mobile, Alabama, USA
| | - Michael V. Cohen
- Department of Physiology, University of South Alabama College of Medicine, Mobile, Alabama, USA
- Department of Medicine, University of South Alabama College of Medicine, Mobile, Alabama, USA
| | - James M. Downey
- Department of Physiology, University of South Alabama College of Medicine, Mobile, Alabama, USA
| |
Collapse
|
42
|
Abstract
Ischemic preconditioning renders the heart resistant to infarction from ischemia/reperfusion. Over the past two decades a great deal has been learned about preconditioning's mechanism. Adenosine, bradykinin, and opioids act in parallel to trigger the preconditioned state and do so by activating PKC. While adenosine couples directly to PKC through the phospholipases, bradykinin and opioids do so through a complex pathway that includes in order: phosphatidylinositol 3-kinase (PI3-kinase), Akt, nitric oxide synthase, guanylyl cyclase, PKG, opening of mitochondrial K(ATP) channels, and activation of PKC by redox signaling. There are even differences between the opioid and bradykinin coupling as the former activates PI3-kinase through transactivation of the epidermal growth factor receptor while the latter has an unknown coupling mechanism. Protection stems from inhibition of formation of mitochondrial permeability transition pores early in reperfusion through activation of the survival kinases, Akt and ERK. These kinases are activated as a result of PKC somehow promoting signaling from adenosine A(2) receptors early in reperfusion. The survival kinases are thought to inhibit pore formation by phosphorylating GSK-3beta. The reperfused heart requires the support of the protective signals for only about an hour after which the ischemic injury is repaired and the signals are no longer needed.
Collapse
Affiliation(s)
- James M Downey
- Department of Physiology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA
| | | | | |
Collapse
|
43
|
Gaitanaki C, Kalpachidou T, Aggeli IKS, Papazafiri P, Beis I. CoCl2 induces protective events via the p38-MAPK signalling pathway and ANP in the perfused amphibian heart. ACTA ACUST UNITED AC 2007; 210:2267-77. [PMID: 17575032 DOI: 10.1242/jeb.003178] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Mitogen-activated protein kinases (MAPKs) constitute one of the most important intracellular signalling pathways. In particular, the p38-MAPK subfamily is known to be activated under various stressful conditions, such as mechanical or oxidative stress. Furthermore, cobalt chloride (CoCl2) has been shown to mimic hypoxic responses in various cell lines and cause overproduction of reactive oxygen species (ROS). In the current study, we investigated the effect of CoCl2 on p38-MAPK signalling pathway in the perfused Rana ridibunda heart. Immunoblot analysis of the phosphorylated, and thus activated, form of p38-MAPK revealed that maximum phosphorylation was attained at 500 micromol l(-1) CoCl2. A similar profile was observed for MAPKAPK2 and Hsp27 phosphorylation (direct and indirect p38-MAPK substrates, respectively). Time course analysis of p38-MAPK phosphorylation pattern showed that the kinase reached its peak within 15 min of treatment with 500 micromol l(-1) CoCl2. Similar results were obtained for Hsp27 phosphorylation. In the presence of the antioxidants Trolox or Lipoic acid, p38-MAPK CoCl2-induced phosphorylation was attenuated. Analogous results were obtained for Hsp27 and MAPKAPK2. In parallel, mRNA levels of the ANP gene, a hormone whose transcriptional regulation has previously been shown to be regulated by p38-MAPK, were examined (semi-quantitative ratiometric RT-PCR). CoCl2 treatment significantly increased ANP mRNA levels, whereas, in the presence of antioxidants, the transcript levels returned to basal values. All the above data indicate that CoCl2 stimulates compensatory mechanisms involving the p38-MAPK signalling cascade along with ANP.
Collapse
Affiliation(s)
- Catherine Gaitanaki
- Department of Animal and Human Physiology, School of Biology, University of Athens, Panepistimioupolis, 157 84 Athens, Greece
| | | | | | | | | |
Collapse
|
44
|
Minguet G, Joris J, Lamy M. Preconditioning and protection against ischaemia-reperfusion in non-cardiac organs: a place for volatile anaesthetics? Eur J Anaesthesiol 2007; 24:733-45. [PMID: 17555610 DOI: 10.1017/s0265021507000531] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
There is an increasing body of evidence that volatile anaesthetics protect myocardium against ischaemic insult by a mechanism termed 'anaesthetic preconditioning'. Anaesthetic preconditioning and ischaemic preconditioning share several common mechanisms of action. Since ischaemic preconditioning has been demonstrated in organs other than the heart, anaesthetic preconditioning might also apply in these organs and have significant clinical applications in surgical procedures carrying a high risk of ischaemia-reperfusion injury. After a brief review on myocardial preconditioning, experimental and clinical data on preconditioning in non-cardiac tissues will be presented. Potential benefits of anaesthetic preconditioning during non-cardiac surgery will be addressed.
Collapse
Affiliation(s)
- G Minguet
- University of Liège, Department of Anaesthesia and Intensive Care Medicine, Belgium.
| | | | | |
Collapse
|
45
|
Sheh YL, Hsu C, Chan SHH, Chan JYH. NADPH oxidase- and mitochondrion-derived superoxide at rostral ventrolateral medulla in endotoxin-induced cardiovascular depression. Free Radic Biol Med 2007; 42:1610-23. [PMID: 17448908 DOI: 10.1016/j.freeradbiomed.2007.02.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2006] [Revised: 01/28/2007] [Accepted: 02/20/2007] [Indexed: 11/16/2022]
Abstract
We evaluated the contribution of superoxide anion (O2*-) generated by NADPH oxidase or mitochondria in the rostral ventrolateral medulla (RVLM), where sympathetic premotor neurons for arterial pressure maintenance are located, on cardiovascular depression induced by inducible nitric oxide synthase-derived NO after Escherichia coli lipopolysaccharide (LPS) treatment. In Sprague-Dawley rats maintained under propofol anesthesia, microinjection of LPS bilaterally into the RVLM induced progressive hypotension, bradycardia, and reduction in sympathetic vasomotor outflow over our 240-min observation period. This was accompanied by an increase in O2*- production (60-240 min) in the RVLM, alongside phosphorylation of p47(phox) or p67(phox), upregulation of gp91(phox) or p47(phox) protein, and increase in Rac-1 or NADPH oxidase activity (60-120 min), and a depression of mitochondrial respiratory enzyme activity (120-240 min). Whereas inhibition of NADPH oxidase or knockdown of the gp91(phox) or p47(phox) gene blunted the early phase (60-150 min), coenzyme Q10 or mitochondrial K(ATP) channel inhibitor antagonized the delayed phase (120-240 min) of LPS-induced increase in O2*- production in RVLM and cardiovascular depression. We conclude that, whereas NADPH oxidase-derived O2*- in RVLM participates predominantly in the early phase, O2*- generated by depression in mitochondrial respiratory enzyme activity or opening of mitoK(ATP) channels mediates the delayed phase of LPS-induced cardiovascular depression.
Collapse
Affiliation(s)
- Yen-Ling Sheh
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan, Republic of China
| | | | | | | |
Collapse
|
46
|
Cohen MV, Philipp S, Krieg T, Cui L, Kuno A, Solodushko V, Downey JM. Preconditioning-mimetics bradykinin and DADLE activate PI3-kinase through divergent pathways. J Mol Cell Cardiol 2007; 42:842-51. [PMID: 17292392 PMCID: PMC1950851 DOI: 10.1016/j.yjmcc.2007.01.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2006] [Revised: 01/03/2007] [Accepted: 01/04/2007] [Indexed: 11/29/2022]
Abstract
We previously reported that pharmacological preconditioning of rabbit hearts with acetylcholine involves activation of phosphatidylinositol 3-kinase (PI3-K) through transactivation of the epidermal growth factor receptor (EGFR). Transactivation is thought to be initiated by cleavage of membrane-bound pro-heparin-binding EGF-like growth factor (HB-EGF) by a membrane metalloproteinase thus releasing HB-EGF which binds to the EGFR. This pathway leads to redox signaling with the generation of reactive oxygen species (ROS) by mitochondria. We tested whether preconditioning's physiological triggers, bradykinin and opioid, also signal through the EGFR. Both bradykinin and the synthetic delta-opioid agonist DADLE increased ROS production in isolated cardiomyocytes by approximately 50%. DADLE's effect was abrogated by either metalloproteinase inhibitor III (MPI) or the diphtheria toxin mutant CRM-197 which blocks heparin-binding EGF shedding indicating that DADLE signals through EGFR transactivation. MPI also blocked DADLE's infarct-sparing effect in whole hearts. Additionally, blocking Src kinase (a component of the EGFR's signaling complex) with PP2 or PI3-K with wortmannin blocked DADLE's effect on cardiomyocyte ROS production and PP2 blocked DADLE's salvage of ischemic myocardium. Finally, DADLE increased phosphorylation of Akt and extracellular signal-regulated protein kinases (ERK) 1/2 in left ventricular myocardium, and this increase was blocked by the EGFR antagonist AG1478. On the other hand, neither MPI nor CRM-197 prevented bradykinin from increasing ROS production, and MPI did not affect bradykinin's infarct-sparing effect in intact hearts. Conversely, both PP2 and wortmannin blocked bradykinin's effect on ROS generation and also aborted bradykinin's cardioprotective effect in intact hearts. While bradykinin also increased phosphorylation of Akt and ERK in myocardium, that increase was not affected by AG1478. Hence bradykinin, unlike acetylcholine or opioid, does not transactivate EGFR, although all 3 agonists do signal through Src and PI3-K.
Collapse
Affiliation(s)
- Michael V Cohen
- Department of Physiology, University of South Alabama, College of Medicine, Mobile, AL 36688, USA.
| | | | | | | | | | | | | |
Collapse
|
47
|
Pasdois P, Beauvoit B, Costa ADT, Vinassa B, Tariosse L, Bonoron-Adèle S, Garlid KD, Dos Santos P. Sarcoplasmic ATP-sensitive potassium channel blocker HMR1098 protects the ischemic heart: implication of calcium, complex I, reactive oxygen species and mitochondrial ATP-sensitive potassium channel. J Mol Cell Cardiol 2006; 42:631-42. [PMID: 17306295 DOI: 10.1016/j.yjmcc.2006.12.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2006] [Revised: 11/27/2006] [Accepted: 12/19/2006] [Indexed: 11/30/2022]
Abstract
The aim of this study was to investigate the effects of HMR1098, a selective blocker of sarcolemmal ATP-sensitive potassium channel (sarcK(ATP)), in Langendorff-perfused rat hearts submitted to ischemia and reperfusion. The recovery of heart hemodynamic and mitochondrial function, studied on skinned fibers, was analyzed after 30-min global ischemia followed by 20-min reperfusion. Infarct size was quantified on a regional ischemia model after 2-h reperfusion. We report that the perfusion of 10 microM HMR1098 before ischemia, delays the onset of ischemic contracture, improves recovery of cardiac function upon reperfusion, preserves the mitochondrial architecture, and finally decreases infarct size. This HMR1098-induced cardioprotection is prevented by 1 mM 2-mercaptopropionylglycine, an antioxidant, and by 100 nM nifedipine, an L-type calcium channel blocker. Concomitantly, it is shown that HMR1098 perfusion induces (i) a transient and specific inhibition of the respiratory chain complex I and, (ii) an increase in the averaged intracellular calcium concentration probed by the in situ measurement of indo-1 fluorescence. Finally, all the beneficial effects of HMR1098 were strongly inhibited by 5-hydroxydecanoate and abolished by glibenclamide, two mitoK(ATP) blockers. This study demonstrates that the HMR1098-induced cardioprotection occurs indirectly through extracellular calcium influx, respiratory chain complex inhibition, reactive oxygen species production and mitoK(ATP) opening. Taken together, these data suggest that a functional interaction between sarcK(ATP) and mitoK(ATP) exists in isolated rat heart ischemia model, which is mediated by extracellular calcium influx.
Collapse
|
48
|
Das A, Smolenski A, Lohmann SM, Kukreja RC. Cyclic GMP-dependent protein kinase Ialpha attenuates necrosis and apoptosis following ischemia/reoxygenation in adult cardiomyocyte. J Biol Chem 2006; 281:38644-52. [PMID: 17038326 DOI: 10.1074/jbc.m606142200] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cyclic GMP-dependent protein kinases protein kinase G (PKG) Ialpha and PKGIbeta are major mediators of cGMP signaling in the cardiovascular system. PKGIalpha is present in the heart, although its role in protection against ischemia/reperfusion injury is not known. We investigated the direct effect of PKGIalpha against necrosis and apoptosis following simulated ischemia (SI) and reoxygenation (RO) in cardiomyocytes. Adult rat cardiomyocytes were infected with adenoviral vectors containing hPKGIalpha or catalytically inactive mutant hPKGIalphaK390A. After 24 h, the cells were subjected to 90 min of SI and 2 h RO for necrosis (trypan blue exclusion and lactate dehydrogenase release) or 18 h RO for apoptosis studies. To evaluate the role of K(ATP) channels, subgroups of cells were treated with 5-hydroxydecanoate (100 microm), HMR1098 (30 microm), or glibenclamide (50 microm), the respective blockers of mitochondrial, sarcolemmal, or both types of K(ATP) channels prior to SI. The necrosis observed in 33.7 +/- 1.6% of total myocytes in the SI-RO control group was reduced to 18.6 +/- 0.8% by PKGIalpha (mean +/- S.E., n = 7, p < 0.001). The apoptosis observed in 17.9 +/- 1.3% of total myocytes in the SI-RO control group was reduced to 6.0 +/- 0.6% by PKGIalpha (mean +/- S.E., n = 7, p < 0.001). In addition, PKGIalpha inhibited the activation of caspase-3 after SI-RO in myocytes. Myocytes infected with the inactive PKGIalphaK390A mutant showed no protection. PKGIalpha enhanced phosphorylation of Akt, ERK1/2, and JNK, increased Bcl-2, inducible nitric-oxide synthase, endothelial nitric-oxide synthase, and decreased Bax expression. 5-Hydroxydecanoate and glibenclamide abolished PKGIalpha-mediated protection against necrosis and apoptosis. However, HMR1098, had no effect. A scavenger of reactive oxygen species, as well as inhibitors of phosphatidylinositol 3-kinase, ERK, JNK1, and NOS, also blocked PKGIalpha-mediated protection against necrosis and apoptosis. These results show that opening of mitochondrial K(ATP) channels and generation of reactive oxygen species, in association with phosphorylation of Akt, ERK, and JNK, and increased expression of NOS and Bcl-2, play an essential role in the protective effect of PKGIalpha.
Collapse
Affiliation(s)
- Anindita Das
- Department of Internal Medicine, Division of Cardiology, Virginia Commonwealth University Medical Center, Richmond, VA 23298, USA
| | | | | | | |
Collapse
|
49
|
Wang JS, Lee T, Chow SE. Role of exercise intensities in oxidized low-density lipoprotein-mediated redox status of monocyte in men. J Appl Physiol (1985) 2006; 101:740-4. [PMID: 16728523 DOI: 10.1152/japplphysiol.00144.2006] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Exercise significantly influences the progression of atherosclerosis. Oxidized LDL (ox-LDL), as a stimulator of oxidative stress, facilitates monocyte-related atherogenesis. This study investigates how exercise intensity impacts ox-LDL-mediated redox status of monocytes. Twenty-five sedentary healthy men exercised mildly, moderately, and heavily (i.e., 40, 60, and 80% maximal oxygen consumption, respectively) on a bicycle ergometer. Reactive oxygen species (ROS) production, cytosolic and mitochondrial superoxide dismutase (c-SOD and m-SOD, respectively) activities, and total and reduced-form γ-glutamylcysteinyl glycine (t-GSH and r-GSH, respectively) contents in monocytes mediated by ox-LDL were measured. This experiment obtained the following findings: 1) ox-LDL increased monocyte ROS production and was accompanied by decreased c-SOD and m-SOD activities, as well as t-GSH and r-GSH contents, whereas treating monocytes with diphenyleneiodonium (DPI) (a NADPH oxidase inhibitor) or rotenone/2-thenoyltrifluoroacetone (TTFA) (mitochondrial complex I/II inhibitors) hindered ox-LDL-induced monocyte ROS production; 2) production of ROS and reduction of m-SOD activity and r-GSH content in monocyte by ox-LDL were enhanced by heavy exercise and depressed by mild and moderate exercise; and 3) heavy exercise augmented the inhibition of ox-LDL-induced monocyte ROS production by DPI and rotenone/TTFA, whereas these DPI- and rotenone/TTFA-mediated monocyte ROS productions were unchanged in response to mild and moderate exercise. We conclude that heavy exercise increases ox-LDL-induced monocyte ROS production, possibly by decreasing m-SOD activity and r-GSH content in monocytes. However, mild and moderate exercise likely protects individuals against suppression of anti-oxidative capacity of monocyte by ox-LDL.
Collapse
Affiliation(s)
- Jong-Shyan Wang
- Institute of Coaching Science, National College of Physical Education & Sports, Tao-Yuan, Taiwan.
| | | | | |
Collapse
|
50
|
Kuno A, Critz SD, Cohen MV, Downey JM. Nicorandil opens mitochondrial K(ATP) channels not only directly but also through a NO-PKG-dependent pathway. Basic Res Cardiol 2006; 102:73-9. [PMID: 16900442 DOI: 10.1007/s00395-006-0612-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2006] [Revised: 07/04/2006] [Accepted: 07/10/2006] [Indexed: 10/24/2022]
Abstract
Nicorandil, a hybrid of nitrate generator and potassium channel opener, protects ischemic myocardium by opening mitochondrial ATP sensitive potassium (mitoK(ATP)) channels. We recently found that nitric oxide (NO) opened K(ATP) channels in rabbit hearts by a protein kinase G (PKG) mechanism. This study examined whether the NO-donor property of nicorandil also contributes to opening of mitoK(ATP) channels through PKG. MitoK(ATP) channel opening was monitored in adult rabbit cardiomyocytes by measuring reactive oxygen species (ROS) production, an established marker of channel opening. Nicorandil increased ROS production in a dose-dependent manner. The selective mitoK(ATP) channel inhibitor 5-hydroxydecanoate (200 microM) completely blocked ROS production by nicorandil at all doses. The PKG inhibitor 8-bromoguanosine-3',5'-cyclic monophosphorothioate, Rpisomer (Rp-8-Br-cGMPs, 50 microM) shifted the dose-ROS production curve to the right with an increase of the EC(50) from 2.4 x 10(-5) M to 6.9 x 10(-5) M. Rp- 8-Br-cGMPs did not affect the increase in ROS production by the selective mitoK(ATP) channel opener diazoxide while it completely blocked increased ROS production from the NO donor S-nitroso-N-acetylpenicillamine (1 microM). Furthermore ODQ, an antagonist of soluble guanylyl cyclase, blocked nicorandil's ability to increase ROS generation. These results indicate that nicorandil, in addition to its direct effect on the channels, opens mitoK(ATP) channels indirectly via a NO-PKG signaling pathway.
Collapse
Affiliation(s)
- Atsushi Kuno
- Dept. of Physiology, MSB 3070, College of Medicine University of South Alabama, Mobile, AL 36688, USA
| | | | | | | |
Collapse
|