101
|
Villamor E, Moreno L, Mohammed R, Pérez-Vizcaíno F, Cogolludo A. Reactive oxygen species as mediators of oxygen signaling during fetal-to-neonatal circulatory transition. Free Radic Biol Med 2019; 142:82-96. [PMID: 30995535 DOI: 10.1016/j.freeradbiomed.2019.04.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 03/22/2019] [Accepted: 04/08/2019] [Indexed: 12/20/2022]
Abstract
Reactive oxygen species (ROS) are frequently seen as pathological agents of oxidative stress. However, ROS are not always deleterious and can also act as cell signaling molecules. Vascular oxygen sensing and signaling during fetal-to-neonatal circulatory transition is a remarkable example of the physiological regulatory actions of ROS. The fetal relative hypoxic environment induces hypoxic pulmonary vasoconstriction (HPV) and ductus arteriosus (DA) relaxation favoring the presence of high pulmonary vascular resistance and right-to-left ductal shunt. At birth, the increase in oxygen tension causes relaxation of pulmonary arteries (PAs) and normoxic DA vasoconstriction (NDAV), thus diverting blood flow to the lungs. Although the response to changes in oxygen tension is diametrically opposite, the mechanisms responsible for HPV and NDAV appear to be the result of a similar interaction between triggering and modulating factors that lead to an increase in cytosolic Ca2+ concentration and Ca2+ sensitization of the contractile apparatus. Growing evidence points to an increase in ROS (mitochondria- and/or NADPH-derived superoxide and/or H2O2), leading to inhibition of voltage-gated K+ channels, membrane depolarization, and activation of voltage-gated L-type Ca2+ channels as critical events in the signaling pathway of both HPV and NDAV. Several groups of investigators have completed this pathway adding other elements such as neutral sphingomyelinase-derived ceramide, the sarcoplasmic/endoplasmic reticulum (through ryanodine and inositol 1,4,5-trisphosphate receptors), Rho kinase-mediated Ca2+ sensitization, or transient receptor potential channels. The present review focus on the role of ROS as mediators of the homeostatic oxygen sensing system during fetal and neonatal life not only in the PAs and DA but also in systemic arteries.
Collapse
Affiliation(s)
- Eduardo Villamor
- Department of Pediatrics, Maastricht University Medical Center (MUMC+), School for Oncology and Developmental Biology (GROW), Maastricht, the Netherlands.
| | - Laura Moreno
- Department of Pharmacology, School of Medicine, Universidad Complutense de Madrid, Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Riazzudin Mohammed
- Department of Pediatrics, Maastricht University Medical Center (MUMC+), School for Oncology and Developmental Biology (GROW), Maastricht, the Netherlands
| | - Francisco Pérez-Vizcaíno
- Department of Pharmacology, School of Medicine, Universidad Complutense de Madrid, Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Angel Cogolludo
- Department of Pharmacology, School of Medicine, Universidad Complutense de Madrid, Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| |
Collapse
|
102
|
Hüttemann M, Sommer N, Weissmann N, Grossman LI. Letter by Hüttemann et al Regarding Article, "Ndufs2, a Core Subunit of Mitochondrial Complex I, Is Essential for Acute Oxygen-Sensing and Hypoxic Pulmonary Vasoconstriction". Circ Res 2019; 125:e33-e34. [PMID: 31557122 DOI: 10.1161/circresaha.119.315815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Maik Hüttemann
- From the Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI (M.H., L.I.G.)
| | - Natascha Sommer
- Excellence Cluster Cardiopulmonary System, University of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen (N.S., N.W.)
| | - Norbert Weissmann
- Excellence Cluster Cardiopulmonary System, University of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen (N.S., N.W.)
| | - Lawrence I Grossman
- From the Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI (M.H., L.I.G.)
| |
Collapse
|
103
|
Olson KR. Hydrogen sulfide, reactive sulfur species and coping with reactive oxygen species. Free Radic Biol Med 2019; 140:74-83. [PMID: 30703482 DOI: 10.1016/j.freeradbiomed.2019.01.020] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 12/19/2018] [Accepted: 01/18/2019] [Indexed: 12/31/2022]
Abstract
Life began in a ferruginous (anoxic and Fe2+ dominated) world around 3.8 billion years ago (bya). Hydrogen sulfide (H2S) and other sulfur molecules from hydrothermal vents and other fissures provided many key necessities for life's origin including catalytic platforms (primordial enzymes) that also served as primitive boundaries (cell walls), substrates for organic synthesis and a continuous source of energy in the form of reducing equivalents. Anoxigenic photosynthesis oxidizing H2S followed within a few hundred million years and laid the metabolic groundwork for oxidative photosynthesis some half-billion years later that slightly and episodically increased atmospheric oxygen around 2.3 bya. This oxidized terrestrial sulfur to sulfate which was washed to the sea where it was reduced creating vast euxinic (anoxic and sulfidic) areas. It was in this environment that eukaryotic cells appeared around 1.5 bya and where they evolved for nearly 1 billion additional years. Oxidative photosynthesis finally oxidized the oceans and around 0.6 bya oxygen levels in the atmosphere and oceans began to rise toward present day levels. This is purported to have been a life-threatening event due to the prevalence of reactive oxygen species (ROS) and thus necessitated the elaboration of chemical and enzymatic antioxidant mechanisms. However, these antioxidants initially appeared around the time of anoxigenic photosynthesis suggesting a commitment to metabolism of reactive sulfur species (RSS). This review examines these events and suggests that many of the biological attributes assigned to ROS may, in fact, be due to RSS. This is underscored by observations that ROS and RSS are chemically similar, often indistinguishable by analytical methods and the fact that the bulk of biochemical and physiological experiments are performed in unphysiologically oxic environments where ROS are artifactually favored over RSS.
Collapse
Affiliation(s)
- Kenneth R Olson
- Indiana University School of Medicine-South Bend, Raclin Carmichael Hall, 1234 Notre Dame Ave, South Bend, IN 46617, USA.
| |
Collapse
|
104
|
Paracrine Mechanisms of Redox Signalling for Postmitotic Cell and Tissue Regeneration. Trends Cell Biol 2019; 29:514-530. [DOI: 10.1016/j.tcb.2019.01.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 01/10/2019] [Accepted: 01/18/2019] [Indexed: 01/08/2023]
|
105
|
Oxygen Regulates Human Pluripotent Stem Cell Metabolic Flux. Stem Cells Int 2019; 2019:8195614. [PMID: 31236115 PMCID: PMC6545818 DOI: 10.1155/2019/8195614] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 02/27/2019] [Indexed: 02/07/2023] Open
Abstract
Metabolism has been shown to alter cell fate in human pluripotent stem cells (hPSC). However, current understanding is almost exclusively based on work performed at 20% oxygen (air), with very few studies reporting on hPSC at physiological oxygen (5%). In this study, we integrated metabolic, transcriptomic, and epigenetic data to elucidate the impact of oxygen on hPSC. Using 13C-glucose labeling, we show that 5% oxygen increased the intracellular levels of glycolytic intermediates, glycogen, and the antioxidant response in hPSC. In contrast, 20% oxygen increased metabolite flux through the TCA cycle, activity of mitochondria, and ATP production. Acetylation of H3K9 and H3K27 was elevated at 5% oxygen while H3K27 trimethylation was decreased, conforming to a more open chromatin structure. RNA-seq analysis of 5% oxygen hPSC also indicated increases in glycolysis, lysine demethylases, and glucose-derived carbon metabolism, while increased methyltransferase and cell cycle activity was indicated at 20% oxygen. Our findings show that oxygen drives metabolite flux and specifies carbon fate in hPSC and, although the mechanism remains to be elucidated, oxygen was shown to alter methyltransferase and demethylase activity and the global epigenetic landscape.
Collapse
|
106
|
Sutendra G, Michelakis ED. Oxygen Sensing in the Pulmonary Circulation: A Fluid State of Affairs. Circ Res 2019; 121:323-325. [PMID: 28775008 DOI: 10.1161/circresaha.117.311518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Gopinath Sutendra
- From the Department of Medicine, University of Alberta, Edmonton, Canada.
| | | |
Collapse
|
107
|
Mello T, Simeone I, Galli A. Mito-Nuclear Communication in Hepatocellular Carcinoma Metabolic Rewiring. Cells 2019; 8:cells8050417. [PMID: 31060333 PMCID: PMC6562577 DOI: 10.3390/cells8050417] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 04/29/2019] [Accepted: 05/01/2019] [Indexed: 12/24/2022] Open
Abstract
As the main metabolic and detoxification organ, the liver constantly adapts its activity to fulfill the energy requirements of the whole body. Despite the remarkable adaptive capacity of the liver, prolonged exposure to noxious stimuli such as alcohol, viruses and metabolic disorders results in the development of chronic liver disease that can progress to hepatocellular carcinoma (HCC), which is currently the second leading cause of cancer-related death worldwide. Metabolic rewiring is a common feature of cancers, including HCC. Altered mito-nuclear communication is emerging as a driving force in the metabolic reprogramming of cancer cells, affecting all aspects of cancer biology from neoplastic transformation to acquired drug resistance. Here, we explore relevant aspects (and discuss recent findings) of mito-nuclear crosstalk in the metabolic reprogramming of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Tommaso Mello
- Clinical Gastroenterology Unit, Department of Biomedical Clinical and Experimental Sciences "Mario Serio", University of Florence, V.le Pieraccini 6, Florence 50129, Italy.
| | - Irene Simeone
- Clinical Gastroenterology Unit, Department of Biomedical Clinical and Experimental Sciences "Mario Serio", University of Florence, V.le Pieraccini 6, Florence 50129, Italy.
- University of Siena, 53100 Siena, Italy.
| | - Andrea Galli
- Clinical Gastroenterology Unit, Department of Biomedical Clinical and Experimental Sciences "Mario Serio", University of Florence, V.le Pieraccini 6, Florence 50129, Italy.
| |
Collapse
|
108
|
Can K, Menzfeld C, Rinne L, Rehling P, Kügler S, Golubiani G, Dudek J, Müller M. Neuronal Redox-Imbalance in Rett Syndrome Affects Mitochondria as Well as Cytosol, and Is Accompanied by Intensified Mitochondrial O 2 Consumption and ROS Release. Front Physiol 2019; 10:479. [PMID: 31114506 PMCID: PMC6503037 DOI: 10.3389/fphys.2019.00479] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 04/05/2019] [Indexed: 12/31/2022] Open
Abstract
Rett syndrome (RTT), an X chromosome-linked neurodevelopmental disorder affecting almost exclusively females, is associated with various mitochondrial alterations. Mitochondria are swollen, show altered respiratory rates, and their inner membrane is leaking protons. To advance the understanding of these disturbances and clarify their link to redox impairment and oxidative stress, we assessed mitochondrial respiration in defined brain regions and cardiac tissue of male wildtype (WT) and MeCP2-deficient (Mecp2-/y ) mice. Also, we quantified for the first time neuronal redox-balance with subcellular resolution in cytosol and mitochondrial matrix. Quantitative roGFP1 redox imaging revealed more oxidized conditions in the cytosol of Mecp2-/y hippocampal neurons than in WT neurons. Furthermore, cytosol and mitochondria of Mecp2-/y neurons showed exaggerated redox-responses to hypoxia and cell-endogenous reactive oxygen species (ROS) formation. Biochemical analyzes exclude disease-related increases in mitochondrial mass in Mecp2-/y hippocampus and cortex. Protein levels of complex I core constituents were slightly lower in Mecp2-/y hippocampus and cortex than in WT; those of complex V were lower in Mecp2-/y cortex. Respiratory supercomplex-formation did not differ among genotypes. Yet, supplied with the complex II substrate succinate, mitochondria of Mecp2-/y cortex and hippocampus consumed more O2 than WT. Furthermore, mitochondria from Mecp2-/y hippocampus and cortex mediated an enhanced oxidative burden. In conclusion, we further advanced the molecular understanding of mitochondrial dysfunction in RTT. Intensified mitochondrial O2 consumption, increased mitochondrial ROS generation and disturbed redox balance in mitochondria and cytosol may represent a causal chain, which provokes dysregulated proteins, oxidative tissue damage, and contributes to neuronal network dysfunction in RTT.
Collapse
Affiliation(s)
- Karolina Can
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, Georg-August-University Göttingen, Göttingen, Germany
- Zentrum Physiologie und Pathophysiologie, Institut für Neuro- und Sinnesphysiologie, Universitätsmedizin Göttingen, Georg-August-Universität Göttingen, Göttingen, Germany
| | - Christiane Menzfeld
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, Georg-August-University Göttingen, Göttingen, Germany
- Zentrum Physiologie und Pathophysiologie, Institut für Neuro- und Sinnesphysiologie, Universitätsmedizin Göttingen, Georg-August-Universität Göttingen, Göttingen, Germany
| | - Lena Rinne
- Zentrum Physiologie und Pathophysiologie, Institut für Neuro- und Sinnesphysiologie, Universitätsmedizin Göttingen, Georg-August-Universität Göttingen, Göttingen, Germany
| | - Peter Rehling
- Zentrum Biochemie und Molekulare Zellbiologie, Institut für Zellbiochemie, Universitätsmedizin Göttingen, Georg-August-Universität Göttingen, Göttingen, Germany
| | - Sebastian Kügler
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, Georg-August-University Göttingen, Göttingen, Germany
- Klinik für Neurologie, Universitätsmedizin Göttingen, Georg-August-Universität Göttingen, Göttingen, Germany
| | - Gocha Golubiani
- Zentrum Physiologie und Pathophysiologie, Institut für Neuro- und Sinnesphysiologie, Universitätsmedizin Göttingen, Georg-August-Universität Göttingen, Göttingen, Germany
- Institute of Chemical Biology, Ilia State University, Tbilisi, Georgia
| | - Jan Dudek
- Zentrum Biochemie und Molekulare Zellbiologie, Institut für Zellbiochemie, Universitätsmedizin Göttingen, Georg-August-Universität Göttingen, Göttingen, Germany
| | - Michael Müller
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, Georg-August-University Göttingen, Göttingen, Germany
- Zentrum Physiologie und Pathophysiologie, Institut für Neuro- und Sinnesphysiologie, Universitätsmedizin Göttingen, Georg-August-Universität Göttingen, Göttingen, Germany
| |
Collapse
|
109
|
Dickmeis T, Feng Y, Mione MC, Ninov N, Santoro M, Spaink HP, Gut P. Nano-Sampling and Reporter Tools to Study Metabolic Regulation in Zebrafish. Front Cell Dev Biol 2019; 7:15. [PMID: 30873407 PMCID: PMC6401643 DOI: 10.3389/fcell.2019.00015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 01/31/2019] [Indexed: 01/09/2023] Open
Abstract
In the past years, evidence has emerged that hallmarks of human metabolic disorders can be recapitulated in zebrafish using genetic, pharmacological or dietary interventions. An advantage of modeling metabolic diseases in zebrafish compared to other "lower organisms" is the presence of a vertebrate body plan providing the possibility to study the tissue-intrinsic processes preceding the loss of metabolic homeostasis. While the small size of zebrafish is advantageous in many aspects, it also has shortcomings such as the difficulty to obtain sufficient amounts for biochemical analyses in response to metabolic challenges. A workshop at the European Zebrafish Principal Investigator meeting in Trento, Italy, was dedicated to discuss the advantages and disadvantages of zebrafish to study metabolic disorders. This perspective article by the participants highlights strategies to achieve improved tissue-resolution for read-outs using "nano-sampling" approaches for metabolomics as well as live imaging of zebrafish expressing fluorescent reporter tools that inform on cellular or subcellular metabolic processes. We provide several examples, including the use of reporter tools to study the heterogeneity of pancreatic beta-cells within their tissue environment. While limitations exist, we believe that with the advent of new technologies and more labs developing methods that can be applied to minimal amounts of tissue or single cells, zebrafish will further increase their utility to study energy metabolism.
Collapse
Affiliation(s)
- Thomas Dickmeis
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
| | - Yi Feng
- Centre for Inflammation Research, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, Scotland
| | | | - Nikolay Ninov
- DFG-Center for Regenerative Therapies Dresden, Cluster of Excellence, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden, Helmholtz Zentrum München, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | | | - Herman P. Spaink
- Institute of Biology Leiden, Leiden University, Leiden, Netherlands
| | - Philipp Gut
- Nestlé Research, EPFL Innovation Park, Lausanne, Switzerland
| |
Collapse
|
110
|
Salazar C, Elorza AA, Cofre G, Ruiz-Hincapie P, Shirihai O, Ruiz LM. The OXPHOS supercomplex assembly factor HIG2A responds to changes in energetic metabolism and cell cycle. J Cell Physiol 2019; 234:17405-17419. [PMID: 30779122 DOI: 10.1002/jcp.28362] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 01/25/2019] [Accepted: 01/30/2019] [Indexed: 01/20/2023]
Abstract
HIG2A promotes cell survival under hypoxia and mediates the assembly of complex III and complex IV into respiratory chain supercomplexes. In the present study, we show that human HIGD2A and mouse Higd2a gene expressions are regulated by hypoxia, glucose, and the cell cycle-related transcription factor E2F1. The latter was found to bind the promoter region of HIGD2A. Differential expression of the HIGD2A gene was found in C57BL/6 mice in relation to tissue and age. Besides, the silencing of HIGD2A evidenced the modulation of mitochondrial dynamics proteins namely, OPA1 as a fusion protein increases, while FIS1, a fission protein, decreases. Besides, the mitochondrial membrane potential (ΔΨm) increased. The protein HIG2A is localized in the mitochondria and nucleus. Moreover, we observed that the HIG2A protein interacts with OPA1. Changes in oxygen concentration, glucose availability, and cell cycle regulate HIGD2A expression. Alterations in HIGD2A expression are associated with changes in mitochondrial physiology.
Collapse
Affiliation(s)
- Celia Salazar
- Instituto de Ciencias Biomédicas, Facultad Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
| | - Alvaro A Elorza
- Institute of Biomedical Sciences, Faculty of Medicine and Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile.,Millennium Institute of Immunology and Immunotherapy, Santiago, Chile
| | - Glenda Cofre
- Instituto de Ciencias Biomédicas, Facultad Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
| | - Paula Ruiz-Hincapie
- School of Engineering and Technology, University of Hertfordshire, Hatfield, UK
| | - Orian Shirihai
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Lina María Ruiz
- Instituto de Ciencias Biomédicas, Facultad Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
| |
Collapse
|
111
|
Sharma A, Arambula JF, Koo S, Kumar R, Singh H, Sessler JL, Kim JS. Hypoxia-targeted drug delivery. Chem Soc Rev 2019; 48:771-813. [PMID: 30575832 PMCID: PMC6361706 DOI: 10.1039/c8cs00304a] [Citation(s) in RCA: 341] [Impact Index Per Article: 56.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Hypoxia is a state of low oxygen tension found in numerous solid tumours. It is typically associated with abnormal vasculature, which results in a reduced supply of oxygen and nutrients, as well as impaired delivery of drugs. The hypoxic nature of tumours often leads to the development of localized heterogeneous environments characterized by variable oxygen concentrations, relatively low pH, and increased levels of reactive oxygen species (ROS). The hypoxic heterogeneity promotes tumour invasiveness, metastasis, angiogenesis, and an increase in multidrug-resistant proteins. These factors decrease the therapeutic efficacy of anticancer drugs and can provide a barrier to advancing drug leads beyond the early stages of preclinical development. This review highlights various hypoxia-targeted and activated design strategies for the formulation of drugs or prodrugs and their mechanism of action for tumour diagnosis and treatment.
Collapse
Affiliation(s)
- Amit Sharma
- Department of Chemistry, Korea University, Seoul, 02841, Korea.
| | | | | | | | | | | | | |
Collapse
|
112
|
Park H, He A, Tan M, Johnson JM, Dean JM, Pietka TA, Chen Y, Zhang X, Hsu FF, Razani B, Funai K, Lodhi IJ. Peroxisome-derived lipids regulate adipose thermogenesis by mediating cold-induced mitochondrial fission. J Clin Invest 2019; 129:694-711. [PMID: 30511960 PMCID: PMC6355224 DOI: 10.1172/jci120606] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 11/20/2018] [Indexed: 12/27/2022] Open
Abstract
Peroxisomes perform essential functions in lipid metabolism, including fatty acid oxidation and plasmalogen synthesis. Here, we describe a role for peroxisomal lipid metabolism in mitochondrial dynamics in brown and beige adipocytes. Adipose tissue peroxisomal biogenesis was induced in response to cold exposure through activation of the thermogenic coregulator PRDM16. Adipose-specific knockout of the peroxisomal biogenesis factor Pex16 (Pex16-AKO) in mice impaired cold tolerance, decreased energy expenditure, and increased diet-induced obesity. Pex16 deficiency blocked cold-induced mitochondrial fission, decreased mitochondrial copy number, and caused mitochondrial dysfunction. Adipose-specific knockout of the peroxisomal β-oxidation enzyme acyl-CoA oxidase 1 (Acox1-AKO) was not sufficient to affect adiposity, thermogenesis, or mitochondrial copy number, but knockdown of the plasmalogen synthetic enzyme glyceronephosphate O-acyltransferase (GNPAT) recapitulated the effects of Pex16 inactivation on mitochondrial morphology and function. Plasmalogens are present in mitochondria and decreased with Pex16 inactivation. Dietary supplementation with plasmalogens increased mitochondrial copy number, improved mitochondrial function, and rescued thermogenesis in Pex16-AKO mice. These findings support a surprising interaction between peroxisomes and mitochondria regulating mitochondrial dynamics and thermogenesis.
Collapse
Affiliation(s)
- Hongsuk Park
- Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Anyuan He
- Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Min Tan
- Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Jordan M. Johnson
- Diabetes & Metabolism Research Center, University of Utah, Salt Lake City, Utah, USA
| | - John M. Dean
- Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | | | - Yali Chen
- Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Xiangyu Zhang
- Cardiology Division, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Fong-Fu Hsu
- Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Babak Razani
- Cardiology Division, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
- Veterans Affairs St. Louis Healthcare System, John Cochran Division, St. Louis, Missouri, USA
| | - Katsuhiko Funai
- Diabetes & Metabolism Research Center, University of Utah, Salt Lake City, Utah, USA
| | - Irfan J. Lodhi
- Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
113
|
Cogolludo A, Villamor E, Perez-Vizcaino F, Moreno L. Ceramide and Regulation of Vascular Tone. Int J Mol Sci 2019; 20:ijms20020411. [PMID: 30669371 PMCID: PMC6359388 DOI: 10.3390/ijms20020411] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 01/02/2019] [Accepted: 01/16/2019] [Indexed: 02/07/2023] Open
Abstract
In addition to playing a role as a structural component of cellular membranes, ceramide is now clearly recognized as a bioactive lipid implicated in a variety of physiological functions. This review aims to provide updated information on the role of ceramide in the regulation of vascular tone. Ceramide may induce vasodilator or vasoconstrictor effects by interacting with several signaling pathways in endothelial and smooth muscle cells. There is a clear, albeit complex, interaction between ceramide and redox signaling. In fact, reactive oxygen species (ROS) activate different ceramide generating pathways and, conversely, ceramide is known to increase ROS production. In recent years, ceramide has emerged as a novel key player in oxygen sensing in vascular cells and mediating vascular responses of crucial physiological relevance such as hypoxic pulmonary vasoconstriction (HPV) or normoxic ductus arteriosus constriction. Likewise, a growing body of evidence over the last years suggests that exaggerated production of vascular ceramide may have detrimental effects in a number of pathological processes including cardiovascular and lung diseases.
Collapse
Affiliation(s)
- Angel Cogolludo
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Ciudad Universitaria S/N, 28040 Madrid, Spain.
- Ciber Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain.
| | - Eduardo Villamor
- Department of Pediatrics, Maastricht University Medical Center (MUMC+), School for Oncology and Developmental Biology (GROW), 6202 AZ Maastricht, The Netherlands.
| | - Francisco Perez-Vizcaino
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Ciudad Universitaria S/N, 28040 Madrid, Spain.
- Ciber Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain.
| | - Laura Moreno
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Ciudad Universitaria S/N, 28040 Madrid, Spain.
- Ciber Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain.
| |
Collapse
|
114
|
Chow HM, Cheng A, Song X, Swerdel MR, Hart RP, Herrup K. ATM is activated by ATP depletion and modulates mitochondrial function through NRF1. J Cell Biol 2019; 218:909-928. [PMID: 30642892 PMCID: PMC6400560 DOI: 10.1083/jcb.201806197] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 10/27/2018] [Accepted: 12/26/2018] [Indexed: 12/19/2022] Open
Abstract
Oxidative stress, resulting from neuronal activity and depleted ATP levels, activates ATM, which phosphorylates NRF1, causing nuclear translocation and up regulation of mitochondrial gene expression. In ATM deficiency, ATP levels recover more slowly, particularly in active neurons with high energy demands. Ataxia-telangiectasia (A-T) is an autosomal recessive disease caused by mutation of the ATM gene and is characterized by loss of cerebellar Purkinje cells, neurons with high physiological activity and dynamic ATP demands. Here, we show that depletion of ATP generates reactive oxygen species that activate ATM. We find that when ATM is activated by oxidative stress, but not by DNA damage, ATM phosphorylates NRF1. This leads to NRF1 dimerization, nuclear translocation, and the up-regulation of nuclear-encoded mitochondrial genes, thus enhancing the capacity of the electron transport chain (ETC) and restoring mitochondrial function. In cells lacking ATM, cells replenish ATP poorly following surges in energy demand, and chronic ATP insufficiency endangers cell survival. We propose that in the absence of ATM, cerebellar Purkinje cells cannot respond adequately to the increase in energy demands of neuronal activity. Our findings identify ATM as a guardian of mitochondrial output, as well as genomic integrity, and suggest that alternative fuel sources may ameliorate A-T disease symptoms.
Collapse
Affiliation(s)
- Hei-Man Chow
- Division of Life Science and The State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong .,Institute for Advanced Study, Hong Kong University of Science and Technology, Hong Kong
| | - Aifang Cheng
- Division of Life Science and The State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong
| | - Xuan Song
- Division of Life Science and The State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong
| | - Mavis R Swerdel
- Department of Cell Biology and Neuroscience, Rutgers University, New Brunswick, NJ
| | - Ronald P Hart
- Department of Cell Biology and Neuroscience, Rutgers University, New Brunswick, NJ
| | - Karl Herrup
- Division of Life Science and The State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong
| |
Collapse
|
115
|
Zhu Z, Chen X, Sun J, Li Q, Lian X, Li S, Wang Y, Tian L. Inhibition of nuclear thioredoxin aggregation attenuates PM 2.5-induced NF-κB activation and pro-inflammatory responses. Free Radic Biol Med 2019; 130:206-214. [PMID: 30420332 DOI: 10.1016/j.freeradbiomed.2018.10.438] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 10/19/2018] [Accepted: 10/22/2018] [Indexed: 11/17/2022]
Abstract
Exposure to fine particulate matter (PM2.5) can induce oxidative stress and proinflammatory cytokine production, which are central for the induction of PM2.5-mediated adverse effects on public health. Nuclear factor kappa B (NF-κB) signaling is essential for inflammation. The subcellular distribution of thioredoxin (Trx) is related to the activation of NF-κB, but the mechanism involved is unclear. In the current study, we focused on the relationship between the antioxidant Trx and NF-κB in human bronchial epithelial cells (BEAS-2B) after PM2.5 exposure. We inhibited the nuclear translocation of Trx by cHCEU (4-cyclohexyl-[3-(2-chloroethyl)ureido]benzene) and subsequently increased the transcriptional activity of Nrf2 to upregulate the expression of Trx by t-BHQ. Our data suggest that PM2.5 exposure induces the activation of NF-κB and the expression of the downstream proinflammatory cytokines IL-1, IL-6, IL-8 and TNF-α in BEAS-2B cells. CHCEU alleviates inflammatory cytokines by blocking Trx nuclear translocation and inhibits the DNA binding activity of NF-κB. T-BHQ could promote the transcriptional activity of Nrf2 but failed to alleviate the production of inflammatory cytokines. Furthermore, the synergistic effect of t-BHQ and cHCEU on alleviating PM2.5-induced inflammation is more effective than the use of cHCEU alone. Our findings characterize the underlying molecular mechanisms of proinflammatory responses induced by PM2.5 and show that the nuclear translocation and accumulation of Trx in nuclei play important roles in PM2.5-induced NF-κB activation and proinflammatory responses.
Collapse
Affiliation(s)
- Zhonghui Zhu
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Xiaowei Chen
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Jingping Sun
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Qiuyue Li
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Ximeng Lian
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Siling Li
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Yan Wang
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Lin Tian
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
116
|
Bailey DM, Brugniaux JV, Filipponi T, Marley CJ, Stacey B, Soria R, Rimoldi SF, Cerny D, Rexhaj E, Pratali L, Salmòn CS, Murillo Jáuregui C, Villena M, Smirl JD, Ogoh S, Pietri S, Scherrer U, Sartori C. Exaggerated systemic oxidative-inflammatory-nitrosative stress in chronic mountain sickness is associated with cognitive decline and depression. J Physiol 2019; 597:611-629. [PMID: 30397919 PMCID: PMC6332753 DOI: 10.1113/jp276898] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 11/05/2018] [Indexed: 12/15/2022] Open
Abstract
KEY POINTS Chronic mountain sickness (CMS) is a maladaptation syndrome encountered at high altitude (HA) characterised by severe hypoxaemia that carries a higher risk of stroke and migraine and is associated with increased morbidity and mortality. We examined if exaggerated oxidative-inflammatory-nitrosative stress (OXINOS) and corresponding decrease in vascular nitric oxide bioavailability in patients with CMS (CMS+) is associated with impaired cerebrovascular function and adverse neurological outcome. Systemic OXINOS was markedly elevated in CMS+ compared to healthy HA (CMS-) and low-altitude controls. OXINOS was associated with blunted cerebral perfusion and vasoreactivity to hypercapnia, impaired cognition and, in CMS+, symptoms of depression. These findings are the first to suggest that a physiological continuum exists for hypoxaemia-induced systemic OXINOS in HA dwellers that when excessive is associated with accelerated cognitive decline and depression, helping identify those in need of more specialist neurological assessment and targeted support. ABSTRACT Chronic mountain sickness (CMS) is a maladaptation syndrome encountered at high altitude (HA) characterised by severe hypoxaemia that carries a higher risk of stroke and migraine and is associated with increased morbidity and mortality. The present cross-sectional study examined to what extent exaggerated systemic oxidative-inflammatory-nitrosative stress (OXINOS), defined by an increase in free radical formation and corresponding decrease in vascular nitric oxide (NO) bioavailability, is associated with impaired cerebrovascular function, accelerated cognitive decline and depression in CMS. Venous blood was obtained from healthy male lowlanders (80 m, n = 17), and age- and gender-matched HA dwellers born and bred in La Paz, Bolivia (3600 m) with (CMS+, n = 23) and without (CMS-, n = 14) CMS. We sampled blood for oxidative (electron paramagnetic resonance spectroscopy, HPLC), nitrosative (ozone-based chemiluminescence) and inflammatory (fluorescence) biomarkers. We employed transcranial Doppler ultrasound to measure cerebral blood flow (CBF) and reactivity. We utilised psychometric tests and validated questionnaires to assess cognition and depression. Highlanders exhibited elevated systemic OXINOS (P < 0.05 vs. lowlanders) that was especially exaggerated in the more hypoxaemic CMS+ patients (P < 0.05 vs. CMS-). OXINOS was associated with blunted cerebral perfusion and vasoreactivity to hypercapnia, impaired cognition and, in CMS+, symptoms of depression. Collectively, these findings are the first to suggest that a physiological continuum exists for hypoxaemia-induced OXINOS in HA dwellers that when excessive is associated with accelerated cognitive decline and depression, helping identify those in need of specialist neurological assessment and support.
Collapse
Affiliation(s)
- Damian M. Bailey
- Neurovascular Research LaboratoryFaculty of Life Sciences and Education, University of South WalesUK
| | - Julien V. Brugniaux
- Neurovascular Research LaboratoryFaculty of Life Sciences and Education, University of South WalesUK
- HP2 Laboratory, INSERM U1042Grenoble Alpes UniversityGrenobleFrance
| | - Teresa Filipponi
- Neurovascular Research LaboratoryFaculty of Life Sciences and Education, University of South WalesUK
| | - Christopher J. Marley
- Neurovascular Research LaboratoryFaculty of Life Sciences and Education, University of South WalesUK
| | - Benjamin Stacey
- Neurovascular Research LaboratoryFaculty of Life Sciences and Education, University of South WalesUK
| | - Rodrigo Soria
- Department of Cardiology and Clinical ResearchUniversity HospitalBernSwitzerland
| | - Stefano F. Rimoldi
- Department of Cardiology and Clinical ResearchUniversity HospitalBernSwitzerland
| | - David Cerny
- Department of Cardiology and Clinical ResearchUniversity HospitalBernSwitzerland
| | - Emrush Rexhaj
- Department of Cardiology and Clinical ResearchUniversity HospitalBernSwitzerland
| | | | | | | | | | - Jonathan D. Smirl
- Centre for Heart, Lung and Vascular Health, School of Health and Exercise ScienceUniversity of British Columbia OkanaganKelownaBritish ColumbiaCanada
| | | | | | - Urs Scherrer
- Department of Cardiology and Clinical ResearchUniversity HospitalBernSwitzerland
- Facultad de Ciencias, Departamento de BiologíaUniversidad de TarapacáAricaChile
| | - Claudio Sartori
- Department of Internal MedicineUniversity HospitalUNIL‐LausanneSwitzerland
| |
Collapse
|
117
|
Mennerich D, Kellokumpu S, Kietzmann T. Hypoxia and Reactive Oxygen Species as Modulators of Endoplasmic Reticulum and Golgi Homeostasis. Antioxid Redox Signal 2019; 30:113-137. [PMID: 29717631 DOI: 10.1089/ars.2018.7523] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
SIGNIFICANCE Eukaryotic cells execute various functions in subcellular compartments or organelles for which cellular redox homeostasis is of importance. Apart from mitochondria, hypoxia and stress-mediated formation of reactive oxygen species (ROS) were shown to modulate endoplasmic reticulum (ER) and Golgi apparatus (GA) functions. Recent Advances: Research during the last decade has improved our understanding of disulfide bond formation, protein glycosylation and secretion, as well as pH and redox homeostasis in the ER and GA. Thus, oxygen (O2) itself, NADPH oxidase (NOX) formed ROS, and pH changes appear to be of importance and indicate the intricate balance of intercompartmental communication. CRITICAL ISSUES Although the interplay between hypoxia, ER stress, and Golgi function is evident, the existence of more than 20 protein disulfide isomerase family members and the relative mild phenotypes of, for example, endoplasmic reticulum oxidoreductin 1 (ERO1)- and NOX4-knockout mice clearly suggest the existence of redundant and alternative pathways, which remain largely elusive. FUTURE DIRECTIONS The identification of these pathways and the key players involved in intercompartmental communication needs suitable animal models, genome-wide association, as well as proteomic studies in humans. The results of those studies will be beneficial for the understanding of the etiology of diseases such as type 2 diabetes, Alzheimer's disease, and cancer, which are associated with ROS, protein aggregation, and glycosylation defects.
Collapse
Affiliation(s)
- Daniela Mennerich
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu , Oulu, Finland
| | - Sakari Kellokumpu
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu , Oulu, Finland
| | - Thomas Kietzmann
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu , Oulu, Finland
| |
Collapse
|
118
|
Stanford KR, Ajmo JM, Bahia PK, Hadley SH, Taylor-Clark TE. Improving redox sensitivity of roGFP1 by incorporation of selenocysteine at position 147. BMC Res Notes 2018; 11:827. [PMID: 30466490 PMCID: PMC6249920 DOI: 10.1186/s13104-018-3929-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 11/13/2018] [Indexed: 12/19/2022] Open
Abstract
Objective Redox-sensitive green fluorescent protein (roGFP) is a genetically-encoded redox-sensitive protein used to detect cellular oxidative stress associated with reactive oxygen species production. Here we replaced the cysteine at position 147 of roGFP1 (variant of roGFP) with selenocysteine in order to increase redox sensitivity of the redox reporter. Results Expression of roGFP1 selenoprotein (roGFP1-Se147) in HEK293 cells required the presence of a selenocysteine insertion sequence and was augmented by co-expression with SBP2. roGFP1-Se147 demonstrated a similar excitation and emission spectra to roGFP1. Although expression of roGFP1-Se147 was limited, it was sufficient enough to perform live cell imaging to evaluate sensitivity to oxidation and reduction. roGFP1-Se147 exhibited a 100-fold increase in sensitivity to oxidation with H2O2 in comparison to roGFP1 as well as a 20-fold decrease in the EC50 of H2O2. Furthermore, roGFP1-Se147, unlike roGFP1, was able to detect oxidation caused by the mitochondrial electron transport complex III inhibitor antimycin A. Unfortunately roGFP-Se147 exhibited a diminished dynamic range and photoinstability. Electronic supplementary material The online version of this article (10.1186/s13104-018-3929-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Katherine R Stanford
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, USA
| | - Joanne M Ajmo
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, USA
| | - Parmvir K Bahia
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, USA
| | - Stephen H Hadley
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, USA
| | - Thomas E Taylor-Clark
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, USA.
| |
Collapse
|
119
|
Smith KA, Schumacker PT. Sensors and signals: the role of reactive oxygen species in hypoxic pulmonary vasoconstriction. J Physiol 2018; 597:1033-1043. [PMID: 30091476 DOI: 10.1113/jp275852] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 07/20/2018] [Indexed: 12/12/2022] Open
Abstract
When lung cells experience hypoxia, the functional response, termed hypoxic pulmonary vasoconstriction, activates a multitude of pathways with the goal of optimizing gas exchange. While previously controversial, overwhelming evidence now suggests that increased reactive oxygen species - produced at complex III of the mitochondrial electron transport chain and released into the intermembrane space - is the cellular oxygen signal responsible for triggering hypoxic pulmonary vasoconstriction. The increased reactive oxygen species (ROS) activate many downstream targets that ultimately lead to increased intracellular ionized calcium concentration and contraction of pulmonary arterial smooth muscle cells. While the specific targets of ROS signals are not completely understood, it is clear that this signalling pathway is critical for development and for normal lung function in newborns and adults.
Collapse
Affiliation(s)
- Kimberly A Smith
- Department of Pediatrics, Division of Neonatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Paul T Schumacker
- Department of Pediatrics, Division of Neonatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
120
|
Bailey DM, Rasmussen P, Evans KA, Bohm AM, Zaar M, Nielsen HB, Brassard P, Nordsborg NB, Homann PH, Raven PB, McEneny J, Young IS, McCord JM, Secher NH. Hypoxia compounds exercise-induced free radical formation in humans; partitioning contributions from the cerebral and femoral circulation. Free Radic Biol Med 2018; 124:104-113. [PMID: 29859345 DOI: 10.1016/j.freeradbiomed.2018.05.090] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 05/19/2018] [Accepted: 05/29/2018] [Indexed: 12/19/2022]
Abstract
This study examined to what extent the human cerebral and femoral circulation contribute to free radical formation during basal and exercise-induced responses to hypoxia. Healthy participants (5♂, 5♀) were randomly assigned single-blinded to normoxic (21% O2) and hypoxic (10% O2) trials with measurements taken at rest and 30 min after cycling at 70% of maximal power output in hypoxia and equivalent relative and absolute intensities in normoxia. Blood was sampled from the brachial artery (a), internal jugular and femoral veins (v) for non-enzymatic antioxidants (HPLC), ascorbate radical (A•-, electron paramagnetic resonance spectroscopy), lipid hydroperoxides (LOOH) and low density lipoprotein (LDL) oxidation (spectrophotometry). Cerebral and femoral venous blood flow was evaluated by transcranial Doppler ultrasound (CBF) and constant infusion thermodilution (FBF). With 3 participants lost to follow up (final n = 4♂, 3♀), hypoxia increased CBF and FBF (P = 0.041 vs. normoxia) with further elevations in FBF during exercise (P = 0.002 vs. rest). Cerebral and femoral ascorbate and α-tocopherol consumption (v < a) was accompanied by A•-/LOOH formation (v > a) and increased LDL oxidation during hypoxia (P < 0.043-0.049 vs. normoxia) implying free radical-mediated lipid peroxidation subsequent to inadequate antioxidant defense. This was pronounced during exercise across the femoral circulation in proportion to the increase in local O2 uptake (r = -0.397 to -0.459, P = 0.037-0.045) but unrelated to any reduction in PO2. These findings highlight considerable regional heterogeneity in the oxidative stress response to hypoxia that may be more attributable to local differences in O2 flux than to O2 tension.
Collapse
Affiliation(s)
- Damian M Bailey
- Neurovascular Research Laboratory, Faculty of Life Sciences and Education, University of South Wales, UK.
| | - Peter Rasmussen
- Department of Anesthesia, Rigshospitalet, University of Copenhagen, Denmark
| | - Kevin A Evans
- Neurovascular Research Laboratory, Faculty of Life Sciences and Education, University of South Wales, UK
| | - Aske M Bohm
- Department of Anesthesia, Rigshospitalet, University of Copenhagen, Denmark
| | - Morten Zaar
- Department of Anesthesia, Rigshospitalet, University of Copenhagen, Denmark
| | - Henning B Nielsen
- Department of Anesthesia, Rigshospitalet, University of Copenhagen, Denmark
| | - Patrice Brassard
- Department of Kinesiology, Faculty of Medicine, Université Laval, Québec, Canada
| | - Nikolai B Nordsborg
- Faculty of Science, Department of Nutrition, Exercise and Sports, University of Copenhagen, Denmark
| | | | - Peter B Raven
- Department of Integrative Physiology and Anatomy, University of North Texas Health Science Center, TX, USA
| | - Jane McEneny
- Centre for Public Health, Queen's University Belfast, Northern Ireland, UK
| | - Ian S Young
- Centre for Public Health, Queen's University Belfast, Northern Ireland, UK
| | - Joe M McCord
- Department of Medicine, Division of Pulmonary Science and Critical Care Medicine, University of Colorado at Denver, Denver, CO, USA
| | - Niels H Secher
- Department of Anesthesia, Rigshospitalet, University of Copenhagen, Denmark
| |
Collapse
|
121
|
Culley MK, Chan SY. Mitochondrial metabolism in pulmonary hypertension: beyond mountains there are mountains. J Clin Invest 2018; 128:3704-3715. [PMID: 30080181 DOI: 10.1172/jci120847] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Pulmonary hypertension (PH) is a heterogeneous and fatal disease of the lung vasculature, where metabolic and mitochondrial dysfunction may drive pathogenesis. Similar to the Warburg effect in cancer, a shift from mitochondrial oxidation to glycolysis occurs in diseased pulmonary vessels and the right ventricle. However, appreciation of metabolic events in PH beyond the Warburg effect is only just emerging. This Review discusses molecular, translational, and clinical concepts centered on the mitochondria and highlights promising, controversial, and challenging areas of investigation. If we can move beyond the "mountains" of obstacles in this field and elucidate these fundamental tenets of pulmonary vascular metabolism, such work has the potential to usher in much-needed diagnostic and therapeutic approaches for the mitochondrial and metabolic management of PH.
Collapse
Affiliation(s)
- Miranda K Culley
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Stephen Y Chan
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
122
|
Grimm A, Cummins N, Götz J. Local Oxidative Damage in the Soma and Dendrites Quarantines Neuronal Mitochondria at the Site of Insult. iScience 2018; 6:114-127. [PMID: 30240605 PMCID: PMC6137705 DOI: 10.1016/j.isci.2018.07.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 06/11/2018] [Accepted: 07/17/2018] [Indexed: 12/19/2022] Open
Abstract
Neurons are highly dependent on mitochondria, but little is known about how they react to a local mitochondrial oxidative insult. We therefore developed a protocol in primary hippocampal cultures that combines the photosensitizer mito-KillerRed with fluorescent biosensors and photoactivatable GFP. We found in both the soma and dendrites that neurons restrict the local increase in mitochondria-derived reactive oxygen species and the decrease in ATP production to the damaged compartment, by quarantining mitochondria. Although the cytosol of both the soma and dendrites became oxidized after mito-KillerRed activation, dendrites were more sensitive to the oxidative insult. Importantly, the impaired mitochondria exhibited decreased motility and fusion, thereby avoiding the spread of oxidation throughout the neuron. These results establish how neurons manage oxidative damage and increase our understanding about the somatodendritic regulation of mitochondrial functions after a local oxidative insult. An oxidative insult is contained locally to the damaged region of a neuron ATP levels decrease only in the damaged region of the soma or dendrite ATP levels increase in the regions distal to the oxidative insult Stressed mitochondria are fragmented, with a decreased motility and fusion rate
Collapse
Affiliation(s)
- Amandine Grimm
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland, St Lucia Campus, Brisbane, QLD 4072, Australia
| | - Nadia Cummins
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland, St Lucia Campus, Brisbane, QLD 4072, Australia
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland, St Lucia Campus, Brisbane, QLD 4072, Australia.
| |
Collapse
|
123
|
Arias-Mayenco I, González-Rodríguez P, Torres-Torrelo H, Gao L, Fernández-Agüera MC, Bonilla-Henao V, Ortega-Sáenz P, López-Barneo J. Acute O 2 Sensing: Role of Coenzyme QH 2/Q Ratio and Mitochondrial ROS Compartmentalization. Cell Metab 2018; 28:145-158.e4. [PMID: 29887397 DOI: 10.1016/j.cmet.2018.05.009] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 04/04/2018] [Accepted: 05/09/2018] [Indexed: 12/20/2022]
Abstract
Acute O2 sensing by peripheral chemoreceptors is essential for mammalian homeostasis. Carotid body glomus cells contain O2-sensitive ion channels, which trigger fast adaptive cardiorespiratory reflexes in response to hypoxia. O2-sensitive cells have unique metabolic characteristics that favor the hypoxic generation of mitochondrial complex I (MCI) signaling molecules, NADH and reactive oxygen species (ROS), which modulate membrane ion channels. We show that responsiveness to hypoxia progressively disappears after inducible deletion of the Ndufs2 gene, which encodes the 49 kDa subunit forming the coenzyme Q binding site in MCI, even in the presence of MCII substrates and chemical NAD+ regeneration. We also show contrasting effects of physiological hypoxia on mitochondrial ROS production (increased in the intermembrane space and decreased in the matrix) and a marked effect of succinate dehydrogenase activity on acute O2 sensing. Our results suggest that acute responsiveness to hypoxia depends on coenzyme QH2/Q ratio-controlled ROS production in MCI.
Collapse
Affiliation(s)
- Ignacio Arias-Mayenco
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Avenida Manuel Siurot s/n, Seville, Spain; Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Patricia González-Rodríguez
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Avenida Manuel Siurot s/n, Seville, Spain
| | - Hortensia Torres-Torrelo
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Avenida Manuel Siurot s/n, Seville, Spain; Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Lin Gao
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Avenida Manuel Siurot s/n, Seville, Spain; Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - M Carmen Fernández-Agüera
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Avenida Manuel Siurot s/n, Seville, Spain
| | - Victoria Bonilla-Henao
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Avenida Manuel Siurot s/n, Seville, Spain; Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Patricia Ortega-Sáenz
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Avenida Manuel Siurot s/n, Seville, Spain; Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - José López-Barneo
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Avenida Manuel Siurot s/n, Seville, Spain; Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| |
Collapse
|
124
|
Sgarbi G, Gorini G, Liuzzi F, Solaini G, Baracca A. Hypoxia and IF₁ Expression Promote ROS Decrease in Cancer Cells. Cells 2018; 7:E64. [PMID: 29933600 PMCID: PMC6071258 DOI: 10.3390/cells7070064] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 06/15/2018] [Accepted: 06/19/2018] [Indexed: 12/11/2022] Open
Abstract
The role of reactive oxygen species (ROS) in the metabolic reprogramming of cells adapted to hypoxia and the interplay between ROS and hypoxia in malignancy is under debate. Here, we examined how ROS levels are modulated by hypoxia in human cancer compared to untransformed cells. Short time exposure (20 min) of either fibroblasts or 143B osteosarcoma cells to low oxygen tension down to 0.5% induced a significant decrease of the cellular ROS level, as detected by the CellROX fluorescent probe (−70%). Prolonging the cells’ exposure to hypoxia for 24 h, ROS decreased further, reaching nearly 20% of the normoxic value. In this regard, due to the debated role of the endogenous inhibitor protein (IF₁) of the ATP synthase complex in cancer cell bioenergetics, we investigated whether IF₁ is involved in the control of ROS generation under severe hypoxic conditions. A significant ROS content decrease was observed in hypoxia in both IF₁-expressing and IF₁- silenced cells compared to normoxia. However, IF₁-silenced cells showed higher ROS levels compared to IF1-containing cells. In addition, the MitoSOX Red-measured superoxide level of all the hypoxic cells was significantly lower compared to normoxia; however, the decrease was milder than the marked drop of ROS content. Accordingly, the difference between IF₁-expressing and IF₁-silenced cells was smaller but significant in both normoxia and hypoxia. In conclusion, the interplay between ROS and hypoxia and its modulation by IF₁ have to be taken into account to develop therapeutic strategies against cancer.
Collapse
Affiliation(s)
- Gianluca Sgarbi
- Department of Biomedical and Neuromotor Sciences, Laboratory of Biochemistry and Mitochondrial Pathophysiology, University of Bologna, Bologna 40126, Italy.
| | - Giulia Gorini
- Department of Biomedical and Neuromotor Sciences, Laboratory of Biochemistry and Mitochondrial Pathophysiology, University of Bologna, Bologna 40126, Italy.
- Department of Biomedical, Experimental, and Clinical Sciences "Mario Serio", University of Florence, Florence 50121, Italy.
| | - Francesca Liuzzi
- Department of Biomedical and Neuromotor Sciences, Laboratory of Biochemistry and Mitochondrial Pathophysiology, University of Bologna, Bologna 40126, Italy.
| | - Giancarlo Solaini
- Department of Biomedical and Neuromotor Sciences, Laboratory of Biochemistry and Mitochondrial Pathophysiology, University of Bologna, Bologna 40126, Italy.
| | - Alessandra Baracca
- Department of Biomedical and Neuromotor Sciences, Laboratory of Biochemistry and Mitochondrial Pathophysiology, University of Bologna, Bologna 40126, Italy.
| |
Collapse
|
125
|
Nunes SC, Lopes-Coelho F, Gouveia-Fernandes S, Ramos C, Pereira SA, Serpa J. Cysteine boosters the evolutionary adaptation to CoCl 2 mimicked hypoxia conditions, favouring carboplatin resistance in ovarian cancer. BMC Evol Biol 2018; 18:97. [PMID: 29921232 PMCID: PMC6011206 DOI: 10.1186/s12862-018-1214-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 06/07/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Ovarian cancer is the second most common gynaecologic malignancy and the most common cause of death from gynaecologic cancer, especially due to diagnosis at an advanced stage, when a cure is rare. As ovarian tumour grows, cancer cells are exposed to regions of hypoxia. Hypoxia is known to be partially responsible for tumour progression, metastasis and resistance to therapies. These suggest that hypoxia entails a selective pressure in which the adapted cells not only have a fitness increase under the selective environment, but also in non-selective adverse environments. In here, we used two different ovarian cancer cell lines - serous carcinoma (OVCAR3) and clear cell carcinoma (ES2) - in order to address the effect of cancer cells selection under normoxia and hypoxia mimicked by cobalt chloride on the evolutionary outcome of cancer cells. RESULTS Our results showed that the adaptation to normoxia and CoCl2 mimicked hypoxia leads cells to display opposite strategies. Whereas cells adapted to CoCl2 mimicked hypoxia conditions tend to proliferate less but present increased survival in adverse environments, cells adapted to normoxia proliferate rapidly but at the cost of increased mortality in adverse environments. Moreover, results suggest that cysteine allows a quicker response and adaptation to hypoxic conditions that, in turn, are capable of driving chemoresistance. CONCLUSIONS We showed that cysteine impacts the adaptation of cancer cells to a CoCl2 mimicked hypoxic environment thus contributing for hypoxia-drived platinum-based chemotherapeutic agents' resistance, allowing the selection of more aggressive phenotypes. These observations support a role of cysteine in cancer progression, recurrence and chemoresistance.
Collapse
Affiliation(s)
- Sofia C. Nunes
- Centro de Estudos de Doenças Crónicas (CEDOC), NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Campo Mártires da Pátria 130, 1169-056 Lisbon, Portugal
- Unidade de Investigação em Patobiologia Molecular do Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto, 1099-023 Lisbon, Portugal
| | - Filipa Lopes-Coelho
- Centro de Estudos de Doenças Crónicas (CEDOC), NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Campo Mártires da Pátria 130, 1169-056 Lisbon, Portugal
- Unidade de Investigação em Patobiologia Molecular do Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto, 1099-023 Lisbon, Portugal
| | - Sofia Gouveia-Fernandes
- Unidade de Investigação em Patobiologia Molecular do Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto, 1099-023 Lisbon, Portugal
| | - Cristiano Ramos
- Unidade de Investigação em Patobiologia Molecular do Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto, 1099-023 Lisbon, Portugal
| | - Sofia A. Pereira
- Centro de Estudos de Doenças Crónicas (CEDOC), NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Campo Mártires da Pátria 130, 1169-056 Lisbon, Portugal
| | - Jacinta Serpa
- Centro de Estudos de Doenças Crónicas (CEDOC), NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Campo Mártires da Pátria 130, 1169-056 Lisbon, Portugal
- Unidade de Investigação em Patobiologia Molecular do Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto, 1099-023 Lisbon, Portugal
| |
Collapse
|
126
|
Zhang J, Chen C, Li L, Zhou HJ, Li F, Zhang H, Yu L, Chen Y, Min W. Endothelial AIP1 Regulates Vascular Remodeling by Suppressing NADPH Oxidase-2. Front Physiol 2018; 9:396. [PMID: 29731721 PMCID: PMC5921534 DOI: 10.3389/fphys.2018.00396] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 04/04/2018] [Indexed: 12/31/2022] Open
Abstract
Objective: AIP1 expression is downregulated in human atherosclerotic plaques and global deletion of AIP1 in mice exacerbates atherosclerosis in ApoE-KO mouse models. However, the direct role of AIP1 in endothelium, vascular remodeling and associated vascular diseases has not been determined. Approach and Results: We used endothelial cell (EC)-specific AIP1-deficient (AIP1-ECKO) mice to define the role of AIP1 in vascular remodeling and intima-media thickening in a mouse carotid artery ligation model characterized by both neointimal hyperplasia and inward vessel remodeling. Compared to WT littermates, AIP1-ECKO mice had 2.2-fold larger intima area and 4.4-fold thicker intima as measured by intima/media ratio in arteries with more proliferating vascular smooth muscle cells (VSMCs) at week 2-4 post-injury. Increased reactive oxygen species (ROS) in endothelium at early time points induced inflammation and vessel dysfunction in AIP1-ECKO prior to VSMC accumulations. Moreover, knockdown of AIP1 in human EC enhanced ROS generation which was attenuated by co-silencing of NOX2. Mechanistically, AIP1 via its proline-rich region binds to the SH3 domain of cytosolic subunit p47phox to disrupt formation of an active NOX2 complex, attenuating ROS production. Conclusion: Our study supports that AIP1 regulates vascular remodeling with intima-media thickening by suppressing endothelial NOX2-dependent oxidative stress. Highlights: •In a carotid ligation model, endothelial cell (EC)-specific AIP1-deficient (AIP1-ECKO) mice had much larger media area, thicker vessel wall and augmented neointima formation.•Increased production of reactive oxygen species in vascular EC at early time points concomitant with vessel dysfunction in AIP1-ECKO.•AIP1 via its proline-rich region binds to the SH3 domain of cytosolic subunit p47phox to disrupt formation of an active NOX2 complex, attenuating ROS production.
Collapse
Affiliation(s)
- Jiqin Zhang
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Pathology and The Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, United States
| | - Chaofei Chen
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Pathology and The Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, United States
| | - Li Li
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huanjiao J. Zhou
- Department of Pathology and The Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, United States
| | - Fenghe Li
- Department of Pathology and The Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, United States
| | - Haifeng Zhang
- Department of Pathology and The Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, United States
| | - Luyang Yu
- Institute of Genetics, Institute of Genetics and Regenerative Biology, College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Yuxin Chen
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Wang Min
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Pathology and The Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
127
|
A Brief Overview of Nitric Oxide and Reactive Oxygen Species Signaling in Hypoxia-Induced Pulmonary Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 967:71-81. [PMID: 29047082 DOI: 10.1007/978-3-319-63245-2_6] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Pulmonary hypertension (PH) is characterized by increased vasoconstriction and smooth muscle cell hyperplasia driving pathological vascular remodeling of arterial vessels. In this short review, we discuss the primary source of reactive oxygen species (ROS) and nitric oxide (NO) relevant to PH and the mechanism by which dysregulation of their production contributes to PH. Specifically, hypoxia-induced PH is associated with diminished endothelial nitric oxide synthase (eNOS)-derived NO production and increased production of superoxide (O2•-) through eNOS uncoupling and defective mitochondrial respiration. This drives the inhibition of the NO/soluble guanylate cyclase (sGC) pathway and activation of the transcription factor hypoxia-inducible factor-1α (HIF-1α) with consequential dysregulation of the pulmonary vasculature. Therapeutics aimed at increasing NO or cGMP bioavailabilities are amenable to hypoxia disease-induced PH. Similarly, strategies targeting HIF-1α are now considered. Overall, pulmonary hypertension including hypoxia-induced PH offers unique opportunities for the rational development of therapeutics centered on modulating redox signaling.
Collapse
|
128
|
Hydrogen Sulfide as an O 2 Sensor: A Critical Analysis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 967:261-276. [PMID: 29047091 DOI: 10.1007/978-3-319-63245-2_15] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
There is increasing interest in the physiological actions and therapeutic potential of the gasotransmitter hydrogen sulfide (H2S). In addition to exerting antihypertensive, anti-inflammatory, antioxidant, and pro-angiogenic effects, H2S has been suggested to play a central and ubiquitous role in O2 sensing. According to this concept, because H2S is metabolized by oxidation, its cellular concentration varies inversely with the ambient pO2 such that hypoxia causes a rise in intracellular [H2S]; this then acts to induce appropriate cellular responses. In particular, it has been proposed that H2S underpins O2 sensing in the carotid body, which triggers increases in ventilation in response to hypoxemia, and also in pulmonary arteries, which constrict in response to local alveolar hypoxia. This process, termed hypoxic pulmonary vasoconstriction (HPV), acts to divert blood to better-oxygenated regions of the lung, thereby maintaining the ventilation-perfusion ratio and minimizing hypoxia-induced falls in blood O2 saturation. In this chapter, we present a critical review of the evidence supporting and questioning this model in both HPV and the carotid body.
Collapse
|
129
|
Bain AR, Ainslie PN, Hoiland RL, Barak OF, Drvis I, Stembridge M, MacLeod DM, McEneny J, Stacey BS, Tuaillon E, Marchi N, Fayd'Herbe De Maudave A, Dujic Z, MacLeod DB, Bailey DM. Competitive apnea and its effect on the human brain: focus on the redox regulation of blood-brain barrier permeability and neuronal-parenchymal integrity. FASEB J 2018; 32:2305-2314. [PMID: 29191963 DOI: 10.1096/fj.201701031r] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Static apnea provides a unique model that combines transient hypertension, hypercapnia, and severe hypoxemia. With apnea durations exceeding 5 min, the purpose of the present study was to determine how that affects cerebral free-radical formation and the corresponding implications for brain structure and function. Measurements were obtained before and following a maximal apnea in 14 divers with transcerebral exchange kinetics, measured as the product of global cerebral blood flow (duplex ultrasound) and radial arterial to internal jugular venous concentration differences ( a-vD). Apnea increased the systemic (arterial) and, to a greater extent, the regional (jugular venous) concentration of the ascorbate free radical, resulting in a shift from net cerebral uptake to output ( P < 0.05). Peroxidation (lipid hydroperoxides, LDL oxidation), NO bioactivity, and S100β were correspondingly enhanced ( P < 0.05), the latter interpreted as minor and not a pathologic disruption of the blood-brain barrier. However, those changes were insufficient to cause neuronal-parenchymal damage confirmed by the lack of change in the a-vD of neuron-specific enolase and human myelin basic protein ( P > 0.05). Collectively, these observations suggest that increased cerebral oxidative stress following prolonged apnea in trained divers may reflect a functional physiologic response, rather than a purely maladaptive phenomenon.-Bain, A. R., Ainslie, P. N., Hoiland, R. L., Barak, O. F., Drvis, I., Stembridge, M., MacLeod, D. M., McEneny, J., Stacey, B. S., Tuaillon, E., Marchi, N., De Maudave, A. F., Dujic, Z., MacLeod, D. B., Bailey, D. M. Competitive apnea and its effect on the human brain: focus on the redox regulation of blood-brain barrier permeability and neuronal-parenchymal integrity.
Collapse
Affiliation(s)
- Anthony R Bain
- Centre for Heart, Lung, and Vascular Health, School of Health and Exercise Science, University of British Columbia, Kelowna, British Columbia, Canada
- Integrative Physiology, University of Colorado, Boulder, Colorado, USA
| | - Philip N Ainslie
- Centre for Heart, Lung, and Vascular Health, School of Health and Exercise Science, University of British Columbia, Kelowna, British Columbia, Canada
| | - Ryan L Hoiland
- Centre for Heart, Lung, and Vascular Health, School of Health and Exercise Science, University of British Columbia, Kelowna, British Columbia, Canada
| | - Otto F Barak
- School of Medicine, University of Split, Split, Croatia
- Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| | - Ivan Drvis
- School of Kinesiology, University of Zagreb, Zagreb, Croatia
| | - Mike Stembridge
- Cardiff Centre for Exercise & Health, Cardiff Metropolitan University, Cardiff, United Kingdom
| | | | - Jane McEneny
- Centre for Public Health, Queen's University Belfast, Belfast, Northern Ireland, United Kingdom
| | - Benjamin S Stacey
- Neurovascular Research Laboratory, Faculty of Life Sciences and Education, University of South Wales, Pontypridd, United Kingdom
| | - Eduoard Tuaillon
- Unit Mixte de Recherche (UMR), INSERM l'Etablissement Français du Sang (EFS), Université Montpellier 1, Montpellier, France
| | - Nicola Marchi
- UMR, Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Department of Neuroscience, Centre National de la Recherche Scientifique (CNRS), INSERM, Institute of Functional Genomics, Montpellier, France; and
| | - Alexis Fayd'Herbe De Maudave
- UMR, Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Department of Neuroscience, Centre National de la Recherche Scientifique (CNRS), INSERM, Institute of Functional Genomics, Montpellier, France; and
| | - Zeljko Dujic
- School of Medicine, University of Split, Split, Croatia
| | - David B MacLeod
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Damian M Bailey
- Neurovascular Research Laboratory, Faculty of Life Sciences and Education, University of South Wales, Pontypridd, United Kingdom
| |
Collapse
|
130
|
Socodato R, Portugal CC, Rodrigues A, Henriques J, Rodrigues C, Figueira C, Relvas JB. Redox tuning of Ca 2+ signaling in microglia drives glutamate release during hypoxia. Free Radic Biol Med 2018; 118:137-149. [PMID: 29501565 DOI: 10.1016/j.freeradbiomed.2018.02.036] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 01/31/2018] [Accepted: 02/27/2018] [Indexed: 02/07/2023]
Abstract
Hypoxia causes oxidative stress and excitotoxicity, culminating in neuronal damage during brain ischemia. Hypoxia also activates microglia, the myeloid resident cells of the brain parenchyma. Upon activation, microglia release high amounts of the neurotransmitter glutamate, contributing for neuronal excitotoxicity during brain insults. Here, we reveal a signaling pathway controlling glutamate release from human microglia during hypoxia. We show that hypoxia-mediated redox imbalance promotes the activation of endoplasmic reticulum inositol 1,4,5-trisphosphate (InsP3) receptors leading to Ca2+ mobilization into the cytosol. Increasing cytosolic Ca2+ signaling in microglia activates the non-receptor protein tyrosine kinase Src at the plasma membrane. Src activation enhances the permeability of microglial gap junctions promoting the release of glutamate during hypoxia. Preventing the hypoxia-triggered redox imbalance, using the dietary antioxidants neochlorogenic acid or vitamin C, inhibits InsP3-dependent Ca2+ signaling and abrogates the release of glutamate. Overall, modulating microglial Ca2+ signaling in response to changes in the redox microenvironment might be critical for controlling glutamate excitotoxicity during hypoxia.
Collapse
Affiliation(s)
- Renato Socodato
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular (IBMC) and The Discoveries Centre for Regeneration and Precision Medicine - Porto campus, Universidade do Porto, Porto, Portugal.
| | - Camila C Portugal
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular (IBMC) and The Discoveries Centre for Regeneration and Precision Medicine - Porto campus, Universidade do Porto, Porto, Portugal
| | - Artur Rodrigues
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular (IBMC) and The Discoveries Centre for Regeneration and Precision Medicine - Porto campus, Universidade do Porto, Porto, Portugal
| | - Joana Henriques
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular (IBMC) and The Discoveries Centre for Regeneration and Precision Medicine - Porto campus, Universidade do Porto, Porto, Portugal
| | - Carla Rodrigues
- Diverge, Grupo Nabeiro Innovation Center, Alameda dos Oceanos 65, 1.1, 1900-208 Lisbon, Portugal
| | - Cláudia Figueira
- Diverge, Grupo Nabeiro Innovation Center, Alameda dos Oceanos 65, 1.1, 1900-208 Lisbon, Portugal
| | - João B Relvas
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular (IBMC) and The Discoveries Centre for Regeneration and Precision Medicine - Porto campus, Universidade do Porto, Porto, Portugal.
| |
Collapse
|
131
|
Pak O, Scheibe S, Esfandiary A, Gierhardt M, Sydykov A, Logan A, Fysikopoulos A, Veit F, Hecker M, Kroschel F, Quanz K, Erb A, Schäfer K, Fassbinder M, Alebrahimdehkordi N, Ghofrani HA, Schermuly RT, Brandes RP, Seeger W, Murphy MP, Weissmann N, Sommer N. Impact of the mitochondria-targeted antioxidant MitoQ on hypoxia-induced pulmonary hypertension. Eur Respir J 2018; 51:1701024. [PMID: 29419444 DOI: 10.1183/13993003.01024-2017] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 01/10/2018] [Indexed: 12/18/2022]
Abstract
Increased mitochondrial reactive oxygen species (ROS), particularly superoxide, have been suggested to mediate hypoxic pulmonary vasoconstriction (HPV), chronic hypoxia-induced pulmonary hypertension and right ventricular remodelling.We determined ROS in acute and chronic hypoxia, and investigated the effect of the mitochondria-targeted antioxidant MitoQ under these conditions.The effect of MitoQ or its inactive carrier substance, decyltriphenylphosphonium, on acute HPV (1% O2 for 10 min) was investigated in isolated blood-free perfused mouse lungs. Mice exposed to chronic hypoxia (10% O2 for 4 weeks) or after banding of the main pulmonary artery were treated with MitoQ or decyltriphenylphosphonium (50 mg·kg-1·day-1).Total cellular superoxide and mitochondrial ROS levels were increased in pulmonary artery smooth muscle cells but decreased in pulmonary fibroblasts in acute hypoxia. MitoQ significantly inhibited HPV and acute hypoxia-induced rise in superoxide concentration. ROS was decreased in pulmonary artery smooth muscle cells, while it increased in the right ventricle after chronic hypoxia. Correspondingly, MitoQ did not affect the development of chronic hypoxia-induced pulmonary hypertension but attenuated right ventricular remodelling after chronic hypoxia as well as after pulmonary arterial banding.Increased mitochondrial ROS of pulmonary artery smooth muscle cells mediate acute HPV, but not chronic hypoxia-induced pulmonary hypertension. MitoQ may be beneficial under conditions of exaggerated acute HPV.
Collapse
Affiliation(s)
- Oleg Pak
- Excellence Cluster Cardio-Pulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Giessen, Germany
- These two authors contributed equally to this work
| | - Susan Scheibe
- Excellence Cluster Cardio-Pulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Giessen, Germany
- These two authors contributed equally to this work
| | - Azadeh Esfandiary
- Excellence Cluster Cardio-Pulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Giessen, Germany
| | - Mareike Gierhardt
- Excellence Cluster Cardio-Pulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Giessen, Germany
| | - Akylbek Sydykov
- Excellence Cluster Cardio-Pulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Giessen, Germany
| | | | - Athanasios Fysikopoulos
- Excellence Cluster Cardio-Pulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Giessen, Germany
| | - Florian Veit
- Legal Medicine, University of Giessen, Giessen, Germany
| | - Matthias Hecker
- Excellence Cluster Cardio-Pulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Giessen, Germany
| | - Florian Kroschel
- Excellence Cluster Cardio-Pulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Giessen, Germany
| | - Karin Quanz
- Excellence Cluster Cardio-Pulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Giessen, Germany
| | - Alexandra Erb
- Excellence Cluster Cardio-Pulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Giessen, Germany
| | - Katharina Schäfer
- Excellence Cluster Cardio-Pulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Giessen, Germany
| | - Mirja Fassbinder
- Excellence Cluster Cardio-Pulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Giessen, Germany
| | - Nasim Alebrahimdehkordi
- Excellence Cluster Cardio-Pulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Giessen, Germany
| | - Hossein A Ghofrani
- Excellence Cluster Cardio-Pulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Giessen, Germany
| | - Ralph T Schermuly
- Excellence Cluster Cardio-Pulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Giessen, Germany
| | - Ralf P Brandes
- Institut für Kardiovaskuläre Physiologie, Goethe-Universität, German Center for Cardiovascular Research (DZHK), Partner Site Rhein-Main, Frankfurt am Main, Germany
| | - Werner Seeger
- Excellence Cluster Cardio-Pulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Giessen, Germany
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | | | - Norbert Weissmann
- Excellence Cluster Cardio-Pulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Giessen, Germany
- These two authors contributed equally to this work
| | - Natascha Sommer
- Excellence Cluster Cardio-Pulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Giessen, Germany
- These two authors contributed equally to this work
| |
Collapse
|
132
|
Gandra PG, Shiah AA, Nogueira L, Hogan MC. A mitochondrial-targeted antioxidant improves myofilament Ca 2+ sensitivity during prolonged low frequency force depression at low PO2. J Physiol 2018; 596:1079-1089. [PMID: 29334129 DOI: 10.1113/jp275470] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 01/08/2018] [Indexed: 12/22/2022] Open
Abstract
KEY POINTS Skeletal muscle contractile activity is associated with an enhanced reactive oxygen species (ROS) generation. At very low PO2, ROS generation by mitochondria can be elevated in intact cells. An elevated intracellular oxidant activity may affect muscle force development and recovery from fatigue. We treated intact single muscle fibres with a mitochondrial antioxidant and stimulated the fibres to contract at a low extracellular PO2 that is similar to the intracellular PO2 that is observed during moderate to intense exercise in vivo. The mitochondrial antioxidant prevented a sustained decrease in the myofibrillar Ca2+ sensitivity and improved muscle submaximal force development after fatigue at low extracellular PO2. ABSTRACT Skeletal muscle can develop a prolonged low frequency-stimulation force depression (PLFFD) following fatigue-inducing contractions. Increased levels of reactive oxygen species (ROS) have been implicated in the development of PLFFD. During exercise the skeletal muscle intracellular PO2 decreases to relatively low levels, and can be further decreased when there is an impairment in O2 diffusion or availability, such as in certain chronic diseases and during exercise at high altitude. Since ROS generation by mitochondria is elevated at very low PO2 in cells, we tested the hypothesis that treatment of muscle fibres with a mitochondrial-targeted antioxidant at a very low, near hypoxic, PO2 can attenuate PLFFD. We treated intact single fibres from mice with the mitochondrial-specific antioxidant SS31, and measured force development and intracellular [Ca2+ ] 30 min after fatigue at an extracellular PO2 of ∼5 Torr. After 30 min following the end of the fatiguing contractions, fibres treated with SS31 showed significantly less impairment in force development compared to untreated fibres at submaximal frequencies of stimulation. The cytosolic peak [Ca2+ ] transients (peak [Ca2+ ]c ) were equally decreased in both groups compared to pre-fatigue values. The combined force and peak [Ca2+ ]c data demonstrated that myofibrillar Ca2+ sensitivity was diminished in the untreated fibres 30 min after fatigue compared to pre-fatigue values, but Ca2+ sensitivity was unaltered in the SS31 treated fibres. These results demonstrate that at a very low PO2, treatment of skeletal muscle fibres with a mitochondrial antioxidant prevents a decrease in the myofibrillar Ca2+ sensitivity, which alleviates the fatigue induced PLFFD.
Collapse
Affiliation(s)
- Paulo G Gandra
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (Unicamp), Campinas, Brazil.,Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Amy A Shiah
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Leonardo Nogueira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Michael C Hogan
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
133
|
Muggiolu G, Simon M, Lampe N, Devès G, Barberet P, Michelet C, Delville MH, Seznec H. In Situ Detection and Single Cell Quantification of Metal Oxide Nanoparticles Using Nuclear Microprobe Analysis. J Vis Exp 2018:55041. [PMID: 29443063 PMCID: PMC5912329 DOI: 10.3791/55041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Micro-analytical techniques based on chemical element imaging enable the localization and quantification of chemical composition at the cellular level. They offer new possibilities for the characterization of living systems and are particularly appropriate for detecting, localizing and quantifying the presence of metal oxide nanoparticles both in biological specimens and the environment. Indeed, these techniques all meet relevant requirements in terms of (i) sensitivity (from 1 up to 10 µg.g-1 of dry mass), (ii) micrometer range spatial resolution, and (iii) multi-element detection. Given these characteristics, microbeam chemical element imaging can powerfully complement routine imaging techniques such as optical and fluorescence microscopy. This protocol describes how to perform a nuclear microprobe analysis on cultured cells (U2OS) exposed to titanium dioxide nanoparticles. Cells must grow on and be exposed directly in a specially designed sample holder used on the optical microscope and in the nuclear microprobe analysis stages. Plunge-freeze cryogenic fixation of the samples preserves both the cellular organization and the chemical element distribution. Simultaneous nuclear microprobe analysis (scanning transmission ion microscopy, Rutherford backscattering spectrometry and particle induced X-ray emission) performed on the sample provides information about the cellular density, the local distribution of the chemical elements, as well as the cellular content of nanoparticles. There is a growing need for such analytical tools within biology, especially in the emerging context of Nanotoxicology and Nanomedicine for which our comprehension of the interactions between nanoparticles and biological samples must be deepened. In particular, as nuclear microprobe analysis does not require nanoparticles to be labelled, nanoparticle abundances are quantifiable down to the individual cell level in a cell population, independently of their surface state.
Collapse
Affiliation(s)
- Giovanna Muggiolu
- Centre d'Etudes Nucléaires Bordeaux Gradignan (CENBG), Université de Bordeaux; Centre d'Etudes Nucléaires Bordeaux Gradignan (CENBG), CNRS
| | - Marina Simon
- Centre d'Etudes Nucléaires Bordeaux Gradignan (CENBG), Université de Bordeaux; Centre d'Etudes Nucléaires Bordeaux Gradignan (CENBG), CNRS
| | - Nathanael Lampe
- Centre d'Etudes Nucléaires Bordeaux Gradignan (CENBG), Université de Bordeaux; Centre d'Etudes Nucléaires Bordeaux Gradignan (CENBG), CNRS
| | - Guillaume Devès
- Centre d'Etudes Nucléaires Bordeaux Gradignan (CENBG), Université de Bordeaux; Centre d'Etudes Nucléaires Bordeaux Gradignan (CENBG), CNRS
| | - Philippe Barberet
- Centre d'Etudes Nucléaires Bordeaux Gradignan (CENBG), Université de Bordeaux; Centre d'Etudes Nucléaires Bordeaux Gradignan (CENBG), CNRS
| | - Claire Michelet
- Centre d'Etudes Nucléaires Bordeaux Gradignan (CENBG), Université de Bordeaux; Centre d'Etudes Nucléaires Bordeaux Gradignan (CENBG), CNRS
| | - Marie-Hélène Delville
- Institut de Chimie de la Matière Condens é e de Bordeaux (ICMCB), CNRS; Institut de Chimie de la Matière Condens é e de Bordeaux (ICMCB), Université de Bordeaux
| | - Hervé Seznec
- Centre d'Etudes Nucléaires Bordeaux Gradignan (CENBG), Université de Bordeaux; Centre d'Etudes Nucléaires Bordeaux Gradignan (CENBG), CNRS;
| |
Collapse
|
134
|
Musharaf I, Hinton M, Yi M, Dakshinamurti S. Hypoxic challenge of hyperoxic pulmonary artery myocytes increases oxidative stress due to impaired mitochondrial superoxide dismutase activity. Pulm Pharmacol Ther 2018; 48:195-202. [DOI: 10.1016/j.pupt.2017.12.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 12/06/2017] [Accepted: 12/11/2017] [Indexed: 01/14/2023]
|
135
|
Yang Z, Wang Y, Zhang Y, He X, Zhong CQ, Ni H, Chen X, Liang Y, Wu J, Zhao S, Zhou D, Han J. RIP3 targets pyruvate dehydrogenase complex to increase aerobic respiration in TNF-induced necroptosis. Nat Cell Biol 2018; 20:186-197. [PMID: 29358703 DOI: 10.1038/s41556-017-0022-y] [Citation(s) in RCA: 209] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 12/05/2017] [Indexed: 01/22/2023]
Abstract
Receptor-interacting protein kinase 3 (RIP3)-regulated production of reactive oxygen species (ROS) positively feeds back on tumour necrosis factor (TNF)-induced necroptosis, a type of programmed necrosis. Glutamine catabolism is known to contribute to RIP3-mediated ROS induction, but the major contributor is unknown. Here, we show that RIP3 activates the pyruvate dehydrogenase complex (PDC, also known as PDH), the rate-limiting enzyme linking glycolysis to aerobic respiration, by directly phosphorylating the PDC E3 subunit (PDC-E3) on T135. Upon activation, PDC enhances aerobic respiration and subsequent mitochondrial ROS production. Unexpectedly, mixed-lineage kinase domain-like (MLKL) is also required for the induction of aerobic respiration, and we further show that it is required for RIP3 translocation to meet mitochondria-localized PDC. Our data uncover a regulation mechanism of PDC activity, show that PDC activation by RIP3 is most likely the major mechanism activated by TNF to increase aerobic respiration and its by-product ROS, and suggest that RIP3-dependent induction of aerobic respiration contributes to pathologies related to oxidative stress.
Collapse
Affiliation(s)
- Zhentao Yang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yan Wang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yingying Zhang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Xiadi He
- State Key Laboratory of Genetic Engineering, School of Life Science and Institute of Biomedical Sciences, The Obstetrics & Gynecology Hospital of Fudan University, Shanghai, China
| | - Chuan-Qi Zhong
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Hengxiao Ni
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Xin Chen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yaoji Liang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Jianfeng Wu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Shimin Zhao
- State Key Laboratory of Genetic Engineering, School of Life Science and Institute of Biomedical Sciences, The Obstetrics & Gynecology Hospital of Fudan University, Shanghai, China
| | - Dawang Zhou
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Jiahuai Han
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
136
|
Lu H, Wang R, Li W, Xie H, Wang C, Hao Y, Sun Y, Jia Z. Plasma proteomic study of acute mountain sickness susceptible and resistant individuals. Sci Rep 2018; 8:1265. [PMID: 29352170 PMCID: PMC5775437 DOI: 10.1038/s41598-018-19818-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 12/20/2017] [Indexed: 12/20/2022] Open
Abstract
Although extensive studies have focused on the development of acute mountain sickness (AMS), the exact mechanisms of AMS are still obscure. In this study, we used isobaric tags for relative and absolute quantitation (iTRAQ) proteomic analysis to identify novel AMS-associated biomarkers in human plasma. After 9 hours of hypobaric hypoxia the abundance of proteins related to tricarboxylic acid (TCA) cycle, glycolysis, ribosome, and proteasome were significantly reduced in AMS resistant (AMS-) group, but not in AMS susceptible (AMS+) group. This suggested that AMS- individuals could reduce oxygen consumption via repressing TCA cycle and glycolysis, and reduce energy consumption through decreasing protein degradation and synthesis compared to AMS+ individuals after acute hypoxic exposure. The inflammatory response might be decreased resulting from the repressed TCA cycle. We propose that the ability for oxygen consumption reduction may play an important role in the development of AMS. Our present plasma proteomic study in plateau of the Han Chinese volunteers gives new data to address the development of AMS and potential AMS correlative biomarkers.
Collapse
Affiliation(s)
- Hui Lu
- Key Laboratory of the plateau of environmental damage control, Lanzhou General Hospital of Lanzhou Military Command, No. 333 Binhe South Road, Lanzhou, 730050, Gansu, China
| | - Rong Wang
- Key Laboratory of the plateau of environmental damage control, Lanzhou General Hospital of Lanzhou Military Command, No. 333 Binhe South Road, Lanzhou, 730050, Gansu, China.
| | - Wenbin Li
- Key Laboratory of the plateau of environmental damage control, Lanzhou General Hospital of Lanzhou Military Command, No. 333 Binhe South Road, Lanzhou, 730050, Gansu, China
| | - Hua Xie
- Key Laboratory of the plateau of environmental damage control, Lanzhou General Hospital of Lanzhou Military Command, No. 333 Binhe South Road, Lanzhou, 730050, Gansu, China
| | - Chang Wang
- Key Laboratory of the plateau of environmental damage control, Lanzhou General Hospital of Lanzhou Military Command, No. 333 Binhe South Road, Lanzhou, 730050, Gansu, China
| | - Ying Hao
- Key Laboratory of the plateau of environmental damage control, Lanzhou General Hospital of Lanzhou Military Command, No. 333 Binhe South Road, Lanzhou, 730050, Gansu, China
| | - Yuhuan Sun
- Key Laboratory of the plateau of environmental damage control, Lanzhou General Hospital of Lanzhou Military Command, No. 333 Binhe South Road, Lanzhou, 730050, Gansu, China
| | - Zhengping Jia
- Key Laboratory of the plateau of environmental damage control, Lanzhou General Hospital of Lanzhou Military Command, No. 333 Binhe South Road, Lanzhou, 730050, Gansu, China.
| |
Collapse
|
137
|
Erard M, Dupré-Crochet S, Nüße O. Biosensors for spatiotemporal detection of reactive oxygen species in cells and tissues. Am J Physiol Regul Integr Comp Physiol 2018; 314:R667-R683. [PMID: 29341828 DOI: 10.1152/ajpregu.00140.2017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Redox biology has become a major issue in numerous areas of physiology. Reactive oxygen species (ROS) have a broad range of roles from signal transduction to growth control and cell death. To understand the nature of these roles, accurate measurement of the reactive compounds is required. An increasing number of tools for ROS detection is available; however, the specificity and sensitivity of these tools are often insufficient. Furthermore, their specificity has been rarely evaluated in complex physiological conditions. Many ROS probes are sensitive to environmental conditions in particular pH, which may interfere with ROS detection and cause misleading results. Accurate detection of ROS in physiology and pathophysiology faces additional challenges concerning the precise localization of the ROS and the timing of their production and disappearance. Certain ROS are membrane permeable, and certain ROS probes move across cells and organelles. Targetable ROS probes such as fluorescent protein-based biosensors are required for accurate localization. Here we analyze these challenges in more detail, provide indications on the strength and weakness of current tools for ROS detection, and point out developments that will provide improved ROS detection methods in the future. There is no universal method that fits all situations in physiology and cell biology. A detailed knowledge of the ROS probes is required to choose the appropriate method for a given biological problem. The knowledge of the shortcomings of these probes should also guide the development of new sensors.
Collapse
Affiliation(s)
- Marie Erard
- Université Paris-Sud, Université Paris-Saclay , Orsay , France.,Centre National de la Recherche Scientifique, Laboratoire de Chimie Physique , Orsay , France
| | - Sophie Dupré-Crochet
- Université Paris-Sud, Université Paris-Saclay , Orsay , France.,Centre National de la Recherche Scientifique, Laboratoire de Chimie Physique , Orsay , France
| | - Oliver Nüße
- Centre National de la Recherche Scientifique, Laboratoire de Chimie Physique , Orsay , France
| |
Collapse
|
138
|
Lack of PINK1 alters glia innate immune responses and enhances inflammation-induced, nitric oxide-mediated neuron death. Sci Rep 2018; 8:383. [PMID: 29321620 PMCID: PMC5762685 DOI: 10.1038/s41598-017-18786-w] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 12/15/2017] [Indexed: 12/20/2022] Open
Abstract
Neuroinflammation is involved in the pathogenesis of Parkinson’s disease (PD) and other neurodegenerative disorders. We show that lack of PINK1- a mitochondrial kinase linked to recessive familial PD – leads to glia type-specific abnormalities of innate immunity. PINK1 loss enhances LPS/IFN-γ stimulated pro-inflammatory phenotypes of mixed astrocytes/microglia (increased iNOS, nitric oxide and COX-2, reduced IL-10) and pure astrocytes (increased iNOS, nitric oxide, TNF-α and IL-1β), while attenuating expression of both pro-inflammatory (TNF-α, IL-1β) and anti-inflammatory (IL-10) cytokines in microglia. These abnormalities are associated with increased inflammation-induced NF-κB signaling in astrocytes, and cause enhanced death of neurons co-cultured with inflamed PINK1−/− mixed glia and neuroblastoma cells exposed to conditioned medium from LPS/IFN-γ treated PINK1−/− mixed glia. Neuroblastoma cell death is prevented with an iNOS inhibitor, implicating increased nitric oxide production as the cause for enhanced death. Finally, we show for the first time that lack of a recessive PD gene (PINK1) increases α-Synuclein-induced nitric oxide production in all glia types (mixed glia, astrocytes and microglia). Our results describe a novel pathogenic mechanism in recessive PD, where PINK1 deficiency may increase neuron death via exacerbation of inflammatory stimuli-induced nitric oxide production and abnormal innate immune responses in glia cells.
Collapse
|
139
|
He X, Song S, Ayon RJ, Balisterieri A, Black SM, Makino A, Wier WG, Zang WJ, Yuan JXJ. Hypoxia selectively upregulates cation channels and increases cytosolic [Ca 2+] in pulmonary, but not coronary, arterial smooth muscle cells. Am J Physiol Cell Physiol 2018; 314:C504-C517. [PMID: 29351410 DOI: 10.1152/ajpcell.00272.2017] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Ca2+ signaling, particularly the mechanism via store-operated Ca2+ entry (SOCE) and receptor-operated Ca2+ entry (ROCE), plays a critical role in the development of acute hypoxia-induced pulmonary vasoconstriction and chronic hypoxia-induced pulmonary hypertension. This study aimed to test the hypothesis that chronic hypoxia differentially regulates the expression of proteins that mediate SOCE and ROCE [stromal interacting molecule (STIM), Orai, and canonical transient receptor potential channel TRPC6] in pulmonary (PASMC) and coronary (CASMC) artery smooth muscle cells. The resting cytosolic [Ca2+] ([Ca2+]cyt) and the stored [Ca2+] in the sarcoplasmic reticulum were not different in CASMC and PASMC. Seahorse measurement showed a similar level of mitochondrial bioenergetics (basal respiration and ATP production) between CASMC and PASMC. Glycolysis was significantly higher in PASMC than in CASMC. The amplitudes of cyclopiazonic acid-induced SOCE and OAG-induced ROCE in CASMC are slightly, but significantly, greater than in PASMC. The frequency and the area under the curve of Ca2+ oscillations induced by ATP and histamine were also larger in CASMC than in PASMC. Na+/Ca2+ exchanger-mediated increases in [Ca2+]cyt did not differ significantly between CASMC and PASMC. The basal protein expression levels of STIM1/2, Orai1/2, and TRPC6 were higher in CASMC than in PASMC, but hypoxia (3% O2 for 72 h) significantly upregulated protein expression levels of STIM1/STIM2, Orai1/Orai2, and TRPC6 and increased the resting [Ca2+]cyt only in PASMC, but not in CASMC. The different response of essential components of store-operated and receptor-operated Ca2+ channels to hypoxia is a unique intrinsic property of PASMC, which is likely one of the important explanations why hypoxia causes pulmonary vasoconstriction and induces pulmonary vascular remodeling, but causes coronary vasodilation.
Collapse
Affiliation(s)
- Xi He
- Department of Pharmacology, Xi'an Jiaotong University Health Science Center, Xi'an, Shannxi Province, China.,Division of Translational and Regenerative Medicine, Department of Medicine, The University of Arizona College of Medicine , Tucson, Arizona
| | - Shanshan Song
- Division of Translational and Regenerative Medicine, Department of Medicine, The University of Arizona College of Medicine , Tucson, Arizona
| | - Ramon J Ayon
- Division of Translational and Regenerative Medicine, Department of Medicine, The University of Arizona College of Medicine , Tucson, Arizona
| | - Angela Balisterieri
- Division of Translational and Regenerative Medicine, Department of Medicine, The University of Arizona College of Medicine , Tucson, Arizona
| | - Stephen M Black
- Division of Translational and Regenerative Medicine, Department of Medicine, The University of Arizona College of Medicine , Tucson, Arizona.,Department of Physiology, The University of Arizona College of Medicine , Tucson, Arizona
| | - Ayako Makino
- Division of Translational and Regenerative Medicine, Department of Medicine, The University of Arizona College of Medicine , Tucson, Arizona.,Department of Physiology, The University of Arizona College of Medicine , Tucson, Arizona
| | - W Gil Wier
- Department of Physiology, The University of Arizona College of Medicine , Tucson, Arizona
| | - Wei-Jin Zang
- Department of Pharmacology, Xi'an Jiaotong University Health Science Center, Xi'an, Shannxi Province, China
| | - Jason X-J Yuan
- Division of Translational and Regenerative Medicine, Department of Medicine, The University of Arizona College of Medicine , Tucson, Arizona.,Department of Physiology, The University of Arizona College of Medicine , Tucson, Arizona
| |
Collapse
|
140
|
Esposito S, Masala A, Sanna S, Rassu M, Pimxayvong V, Iaccarino C, Crosio C. Redox-sensitive GFP to monitor oxidative stress in neurodegenerative diseases. Rev Neurosci 2018; 28:133-144. [PMID: 28030361 DOI: 10.1515/revneuro-2016-0041] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 10/02/2016] [Indexed: 02/06/2023]
Abstract
Redox processes are key events in the degenerative cascade of many adult-onset neurodegenerative diseases (NDs), but the biological relevance of a single redox change is often dependent on the redox couple involved and on its subcellular origin. The biosensors based on engineered fluorescent proteins (redox-sensitive GFP [roGFP]) offer a unique opportunity to monitor redox changes in both physiological and pathological contexts in living animals and plants. Here, we review the use of roGFPs to monitor oxidative stress in different three adult-onset NDs: Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). Despite the many differences spanning from incidence to onset, the hypotheses on biological processes underlying both sporadic and familiar ND forms in humans outline a model in which noncompeting mechanisms are likely to converge in various unsuccessful patterns to mediate the selective degeneration of a specific neuronal population. roGFPs, targeted to different cell compartments, are successfully used as specific markers of cell toxicity, induced by expression of causative genes linked to a determined ND. We also report the use of roGFP to monitor oxidative stress induced by the expression of the ALS-causative gene SOD1.
Collapse
|
141
|
Krylatov AV, Maslov LN, Voronkov NS, Boshchenko AA, Popov SV, Gomez L, Wang H, Jaggi AS, Downey JM. Reactive Oxygen Species as Intracellular Signaling Molecules in the Cardiovascular System. Curr Cardiol Rev 2018; 14:290-300. [PMID: 29962348 PMCID: PMC6300799 DOI: 10.2174/1573403x14666180702152436] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 06/13/2018] [Accepted: 06/20/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Redox signaling plays an important role in the lives of cells. This signaling not only becomes apparent in pathologies but is also thought to be involved in maintaining physiological homeostasis. Reactive Oxygen Species (ROS) can activate protein kinases: CaMKII, PKG, PKA, ERK, PI3K, Akt, PKC, PDK, JNK, p38. It is unclear whether it is a direct interaction of ROS with these kinases or whether their activation is a consequence of inhibition of phosphatases. ROS have a biphasic effect on the transport of Ca2+ in the cell: on one hand, they activate the sarcoplasmic reticulum Ca2+-ATPase, which can reduce the level of Ca2+ in the cell, and on the other hand, they can inactivate Ca2+-ATPase of the plasma membrane and open the cation channels TRPM2, which promote Ca2+-loading and subsequent apoptosis. ROS inhibit the enzyme PHD2, which leads to the stabilization of HIF-α and the formation of the active transcription factor HIF. CONCLUSION Activation of STAT3 and STAT5, induced by cytokines or growth factors, may include activation of NADPH oxidase and enhancement of ROS production. Normal physiological production of ROS under the action of cytokines activates the JAK/STAT while excessive ROS production leads to their inhibition. ROS cause the activation of the transcription factor NF-κB. Physiological levels of ROS control cell proliferation and angiogenesis. ROS signaling is also involved in beneficial adaptations to survive ischemia and hypoxia, while further increases in ROS can trigger programmed cell death by the mechanism of apoptosis or autophagy. ROS formation in the myocardium can be reduced by moderate exercise.
Collapse
Affiliation(s)
| | - Leonid N. Maslov
- Address correspondence to this author at the Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of
Science, Tomsk, Russia; Tel: 3822 262174; Fax: 3822 555057;
E-mail:
| | | | | | | | | | | | | | | |
Collapse
|
142
|
Zhou J, Zhang J, Lu Y, Huang S, Xiao R, Zeng X, Zhang X, Li J, Wang T, Li T, Zhu L, Hu Q. Mitochondrial transplantation attenuates hypoxic pulmonary vasoconstriction. Oncotarget 2017; 7:31284-98. [PMID: 27121314 PMCID: PMC5058756 DOI: 10.18632/oncotarget.8893] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 04/02/2016] [Indexed: 01/15/2023] Open
Abstract
Hypoxia triggers pulmonary vasoconstriction, however induces relaxation of systemic arteries such as femoral arteries. Mitochondria are functionally and structurally heterogeneous between different cell types. The aim of this study was to reveal whether mitochondrial heterogeneity controls the distinct responses of pulmonary versus systemic artery smooth muscle cells to hypoxia. Intact mitochondria were transplanted into Sprague-Dawley rat pulmonary artery smooth muscle cells in culture and pulmonary arteries in vitro. Mitochondria retained functional after transplantation. The cross transplantation of mitochondria between pulmonary and femoral artery smooth muscle cells reversed acute hypoxia-induced alterations in cell membrane potential, [Ca2+]i signaling in smooth muscle cells and constriction or relaxation of arteries. Furthermore, the high or low amount of reactive oxygen species generation from mitochondria and their divergent (dis-)abilities in activating extracellular Ca2+-sensing receptor in smooth muscle cells were found to cause cell membrane potential depolarization, [Ca2+]i elevation and constriction of pulmonary arteries versus cell membrane potential hyperpolarization, [Ca2+]i decline and relaxation of femoral arteries in response to hypoxia, respectively. Our findings suggest that mitochondria necessarily determine the behaviors of vascular smooth muscle cells in response to hypoxia.
Collapse
Affiliation(s)
- Juan Zhou
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.,Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.,Current address: Department of Clinical Laboratory of Xuzhou Central Hospital, Xuzhou 221009, China
| | - Jiwei Zhang
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yankai Lu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.,Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.,Department of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Songling Huang
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.,Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Rui Xiao
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.,Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xianqin Zeng
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.,Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiuyun Zhang
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.,Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jiansha Li
- Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.,Department of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tao Wang
- Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.,Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tongfei Li
- Department of Pathology, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, China
| | - Liping Zhu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.,Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qinghua Hu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.,Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
143
|
Pastore N, Vainshtein A, Klisch TJ, Armani A, Huynh T, Herz NJ, Polishchuk EV, Sandri M, Ballabio A. TFE3 regulates whole-body energy metabolism in cooperation with TFEB. EMBO Mol Med 2017; 9:605-621. [PMID: 28283651 PMCID: PMC5412821 DOI: 10.15252/emmm.201607204] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
TFE3 and TFEB are members of the MiT family of HLH–leucine zipper transcription factors. Recent studies demonstrated that they bind overlapping sets of promoters and are post‐transcriptionally regulated through a similar mechanism. However, while Tcfeb knockout (KO) mice die during early embryonic development, no apparent phenotype was reported in Tfe3 KO mice. Thus raising the need to characterize the physiological role of TFE3 and elucidate its relationship with TFEB. TFE3 deficiency resulted in altered mitochondrial morphology and function both in vitro and in vivo due to compromised mitochondrial dynamics. In addition, Tfe3 KO mice showed significant abnormalities in energy balance and alterations in systemic glucose and lipid metabolism, resulting in enhanced diet‐induced obesity and diabetes. Conversely, viral‐mediated TFE3 overexpression improved the metabolic abnormalities induced by high‐fat diet (HFD). Both TFEB overexpression in Tfe3 KO mice and TFE3 overexpression in Tcfeb liver‐specific KO mice (Tcfeb LiKO) rescued HFD‐induced obesity, indicating that TFEB can compensate for TFE3 deficiency and vice versa. Analysis of Tcfeb LiKO/Tfe3 double KO mice demonstrated that depletion of both TFE3 and TFEB results in additive effects with an exacerbation of the hepatic phenotype. These data indicate that TFE3 and TFEB play a cooperative, rather than redundant, role in the control of the adaptive response of whole‐body metabolism to environmental cues such as diet and physical exercise.
Collapse
Affiliation(s)
- Nunzia Pastore
- Jan and Dan Duncan Neurological Research Institute, Texas Children Hospital, Houston, TX, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Anna Vainshtein
- Jan and Dan Duncan Neurological Research Institute, Texas Children Hospital, Houston, TX, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Tiemo J Klisch
- Jan and Dan Duncan Neurological Research Institute, Texas Children Hospital, Houston, TX, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Andrea Armani
- Department of Biomedical Science, University of Padova, Padova, Italy
| | - Tuong Huynh
- Jan and Dan Duncan Neurological Research Institute, Texas Children Hospital, Houston, TX, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Niculin J Herz
- Jan and Dan Duncan Neurological Research Institute, Texas Children Hospital, Houston, TX, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Elena V Polishchuk
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli (Naples), Italy
| | - Marco Sandri
- Department of Biomedical Science, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Andrea Ballabio
- Jan and Dan Duncan Neurological Research Institute, Texas Children Hospital, Houston, TX, USA .,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.,Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli (Naples), Italy.,Medical Genetics, Department of Medical and Translational Sciences, Federico II University, Naples, Italy
| |
Collapse
|
144
|
Zhu L, Lu Y, Zhang J, Hu Q. Subcellular Redox Signaling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 967:385-398. [DOI: 10.1007/978-3-319-63245-2_25] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
145
|
Byrne MB, Leslie MT, Patel HS, Gaskins HR, Kenis PJA. Design considerations for open-well microfluidic platforms for hypoxic cell studies. BIOMICROFLUIDICS 2017; 11:054116. [PMID: 29152027 PMCID: PMC5659862 DOI: 10.1063/1.4998579] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 10/03/2017] [Indexed: 05/24/2023]
Abstract
Regions of hypoxia are common in solid tumors and are associated with enhanced malignancy, metastasis, and chemo/radio resistance. Real-time hypoxic cellular experimentation is challenging due to the constant need for oxygen control. Most microfluidic platforms developed thus far for hypoxic cell studies are burdened by complex design parameters and are difficult to use for uninitiated investigators. However, open-well microfluidic platforms enable short and long term hypoxic cell studies with an ease of use workflow. Specifically, open-well platforms enable manipulation and addition of cells, media, and reagents using a micropipette for hypoxic cell studies in tunable dissolved oxygen concentrations as low 0.3 mg/l. We analyzed design considerations for open-well microfluidic platforms such as media height, membrane thickness, and impermeable barriers to determine their effects on the amount of dissolved oxygen within the platform. The oxygen concentration was determined by experimental measurements and computational simulations. To examine cell behavior under controlled oxygen conditions, hypoxia-induced changes to hypoxia inducible factor activity and the mitochondrial redox environment were studied. A fluorescent reporter construct was used to monitor the stabilization of hypoxia inducible factors 1α and 2α throughout chronic hypoxia. Reporter construct fluorescence intensity inversely correlated with dissolved oxygen in the medium, as expected. Additionally, the glutathione redox poise of the mitochondrial matrix in living cancer cells was monitored throughout acute hypoxia with a genetically encoded redox probe and was observed to undergo a reductive response to hypoxia. Overall, these studies validate an easy to use open-well platform suitable for studying complex cell behaviors in hypoxia.
Collapse
Affiliation(s)
| | | | - Heeral S Patel
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | | | | |
Collapse
|
146
|
Strielkov I, Pak O, Sommer N, Weissmann N. Recent advances in oxygen sensing and signal transduction in hypoxic pulmonary vasoconstriction. J Appl Physiol (1985) 2017; 123:1647-1656. [PMID: 28751366 DOI: 10.1152/japplphysiol.00103.2017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Hypoxic pulmonary vasoconstriction (HPV) is a physiological reaction, which adapts lung perfusion to regional ventilation and optimizes gas exchange. Impaired HPV may cause systemic hypoxemia, while generalized HPV contributes to the development of pulmonary hypertension. The triggering mechanisms underlying HPV are still not fully elucidated. Several hypotheses are currently under debate, including a possible decrease as well as an increase in reactive oxygen species as a triggering event. Recent findings suggest an increase in the production of reactive oxygen species in pulmonary artery smooth muscle cells by complex III of the mitochondrial electron transport chain and occurrence of oxygen sensing at complex IV. Other essential components are voltage-dependent potassium and possibly L-type, transient receptor potential channel 6, and transient receptor potential vanilloid 4 channels. The release of arachidonic acid metabolites appears also to be involved in HPV regulation. Further investigation of the HPV mechanisms will facilitate the development of novel therapeutic strategies for the treatment of HPV-related disorders.
Collapse
Affiliation(s)
- Ievgen Strielkov
- Excellence Cluster Cardiopulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University, Giessen , Germany
| | - Oleg Pak
- Excellence Cluster Cardiopulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University, Giessen , Germany
| | - Natasha Sommer
- Excellence Cluster Cardiopulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University, Giessen , Germany
| | - Norbert Weissmann
- Excellence Cluster Cardiopulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University, Giessen , Germany
| |
Collapse
|
147
|
Pluripotent Stem Cell Metabolism and Mitochondria: Beyond ATP. Stem Cells Int 2017; 2017:2874283. [PMID: 28804500 PMCID: PMC5540363 DOI: 10.1155/2017/2874283] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 05/07/2017] [Indexed: 12/19/2022] Open
Abstract
Metabolism is central to embryonic stem cell (ESC) pluripotency and differentiation, with distinct profiles apparent under different nutrient milieu, and conditions that maintain alternate cell states. The significance of altered nutrient availability, particularly oxygen, and metabolic pathway activity has been highlighted by extensive studies of their impact on preimplantation embryo development, physiology, and viability. ESC similarly modulate their metabolism in response to altered metabolite levels, with changes in nutrient availability shown to have a lasting impact on derived cell identity through the regulation of the epigenetic landscape. Further, the preferential use of glucose and anaplerotic glutamine metabolism serves to not only support cell growth and proliferation but also minimise reactive oxygen species production. However, the perinuclear localisation of spherical, electron-poor mitochondria in ESC is proposed to sustain ESC nuclear-mitochondrial crosstalk and a mitochondrial-H2O2 presence, to facilitate signalling to support self-renewal through the stabilisation of HIFα, a process that may be favoured under physiological oxygen. The environment in which a cell is grown is therefore a critical regulator and determinant of cell fate, with metabolism, and particularly mitochondria, acting as an interface between the environment and the epigenome.
Collapse
|
148
|
Oxidative Stress Gene Expression Profile Correlates with Cancer Patient Poor Prognosis: Identification of Crucial Pathways Might Select Novel Therapeutic Approaches. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:2597581. [PMID: 28770020 PMCID: PMC5523271 DOI: 10.1155/2017/2597581] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 05/30/2017] [Indexed: 12/17/2022]
Abstract
The role of altered redox status and high reactive oxygen species (ROS) is still controversial in cancer development and progression. Intracellular levels of ROS are elevated in cancer cells suggesting a role in cancer initiation and progression; on the contrary, ROS elevated levels may induce programmed cell death and have been associated with cancer suppression. Thus, it is crucial to consider the double-face of ROS, for novel therapeutic strategies targeting redox regulatory mechanisms. In this review, in order to derive cancer-type specific oxidative stress genes' profile and their potential prognostic role, we integrated a publicly available oxidative stress gene signature with patient survival data from the Cancer Genome Atlas database. Overall, we found several genes statistically significant associated with poor prognosis in the examined six tumor types. Among them, FoxM1 and thioredoxin reductase1 expression showed the same pattern in four out of six cancers, suggesting their specific critical role in cancer-related oxidative stress adaptation. Our analysis also unveiled an enriched cellular network, highlighting specific pathways, in which many genes are strictly correlated. Finally, we discussed novel findings on the correlation between oxidative stress and cancer stem cells in order to define those pathways to be prioritized in drug development.
Collapse
|
149
|
Sgarbi G, Gorini G, Costanzini A, Barbato S, Solaini G, Baracca A. Hypoxia decreases ROS level in human fibroblasts. Int J Biochem Cell Biol 2017; 88:133-144. [DOI: 10.1016/j.biocel.2017.05.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 03/24/2017] [Accepted: 05/04/2017] [Indexed: 12/31/2022]
|
150
|
Plecitá-Hlavatá L, Tauber J, Li M, Zhang H, Flockton AR, Pullamsetti SS, Chelladurai P, D'Alessandro A, El Kasmi KC, Ježek P, Stenmark KR. Constitutive Reprogramming of Fibroblast Mitochondrial Metabolism in Pulmonary Hypertension. Am J Respir Cell Mol Biol 2017; 55:47-57. [PMID: 26699943 DOI: 10.1165/rcmb.2015-0142oc] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Remodeling of the distal pulmonary artery wall is a characteristic feature of pulmonary hypertension (PH). In hypoxic PH, the most substantial pathologic changes occur in the adventitia. Here, there is marked fibroblast proliferation and profound macrophage accumulation. These PH fibroblasts (PH-Fibs) maintain a hyperproliferative, apoptotic-resistant, and proinflammatory phenotype in ex vivo culture. Considering that a similar phenotype is observed in cancer cells, where it has been associated, at least in part, with specific alterations in mitochondrial metabolism, we sought to define the state of mitochondrial metabolism in PH-Fibs. In PH-Fibs, pyruvate dehydrogenase was markedly inhibited, resulting in metabolism of pyruvate to lactate, thus consistent with a Warburg-like phenotype. In addition, mitochondrial bioenergetics were suppressed and mitochondrial fragmentation was increased in PH-Fibs. Most importantly, complex I activity was substantially decreased, which was associated with down-regulation of the accessory subunit nicotinamide adenine dinucleotide reduced dehydrogenase (ubiquinone) Fe-S protein 4 (NDUFS4). Owing to less-efficient ATP synthesis, mitochondria were hyperpolarized and mitochondrial superoxide production was increased. This pro-oxidative status was further augmented by simultaneous induction of cytosolic nicotinamide adenine dinucleotide phosphate reduced oxidase 4. Although acute and chronic exposure to hypoxia of adventitial fibroblasts from healthy control vessels induced increased glycolysis, it did not induce complex I deficiency as observed in PH-Fibs. This suggests that hypoxia alone is insufficient to induce NDUFS4 down-regulation and constitutive abnormalities in complex I. In conclusion, our study provides evidence that, in the pathogenesis of vascular remodeling in PH, alterations in fibroblast mitochondrial metabolism drive distinct changes in cellular behavior, which potentially occur independently of hypoxia.
Collapse
Affiliation(s)
- Lydie Plecitá-Hlavatá
- 1 Department of Membrane Transport Biophysics, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Jan Tauber
- 1 Department of Membrane Transport Biophysics, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Min Li
- 2 Developmental Lung Biology and Cardiovascular Pulmonary Research Laboratory, University of Colorado, Denver, Colorado
| | - Hui Zhang
- 2 Developmental Lung Biology and Cardiovascular Pulmonary Research Laboratory, University of Colorado, Denver, Colorado.,3 Department of Pediatrics, Shengjing Hospital of China Medical, University, Shenyang, China
| | - Amanda R Flockton
- 2 Developmental Lung Biology and Cardiovascular Pulmonary Research Laboratory, University of Colorado, Denver, Colorado
| | - Soni Savai Pullamsetti
- 4 Department of Lung Development and Remodeling, University of Giessen and Marburg Lung Center, Bad Nauheim, Germany; and
| | - Prakash Chelladurai
- 4 Department of Lung Development and Remodeling, University of Giessen and Marburg Lung Center, Bad Nauheim, Germany; and
| | | | - Karim C El Kasmi
- 6 Pediatric Gastroenterology, University of Colorado, Denver, Colorado
| | - Petr Ježek
- 1 Department of Membrane Transport Biophysics, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Kurt R Stenmark
- 2 Developmental Lung Biology and Cardiovascular Pulmonary Research Laboratory, University of Colorado, Denver, Colorado
| |
Collapse
|