9
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Characterization of Renal Injury and Inflammation in an Experimental Model of Intravascular Hemolysis

      research-article
      1 , 2 , 3 , 1 , 2 , 3 , 1 , 2 , 3 , 1 , 2 , 3 , 4 , 1 , 2 , 3 , 1 , 2 , 3 , 1 , 2 , 3 , 1 , 2 , 3 , 1 , 5 , 6 , 5 , 6 , 7 , 8 , 8 , 1 , 2 , 3 , 9 , 9 , 3 , 10 , 11 , 9 , 1 , 2 , 3 , 1 , 2 , 3 , *
      Frontiers in Immunology
      Frontiers Media S.A.
      hemolysis, heme, kidney injury, endothelial activation, inflammation, hemopexin, phenylhydrazine, experimental model of intravascular hemolysis

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Intravascular erythrocyte destruction, accompanied by the release of pro-oxidative and pro-inflammatory components hemoglobin and heme, is a common event in the pathogenesis of numerous diseases with heterogeneous etiology and clinical features. A frequent adverse effect related to massive hemolysis is the renal injury and inflammation. Nevertheless, it is still unclear whether heme––a danger-associated molecular pattern––and ligand for TLR4 or upstream hemolysis-derived products are responsible for these effects. Well-characterized animal models of hemolysis with kidney impairment are needed to investigate how hemolysis drives kidney injury and to test novel therapeutic strategies. Here, we characterized the pathological processes leading to acute kidney injury and inflammation during massive intravascular hemolysis, using a mouse model of phenylhydrazine (PHZ)-triggered erythrocyte destruction. We observed profound changes in mRNA levels for markers of tubular damage (Kim-1, NGAL) and regeneration (indirect marker of tubular injury, Ki-67), and tissue and vascular inflammation (IL-6, E-selectin, P-selectin, ICAM-1) in kidneys of PHZ-treated mice, associated with ultrastructural signs of tubular injury. Moreover, mass spectrometry revealed presence of markers of tubular damage in urine, including meprin-α, cytoskeletal keratins, α-1-antitrypsin, and α-1-microglobulin. Signs of renal injury and inflammation rapidly resolved and the renal function was preserved, despite major changes in metabolic parameters of PHZ-injected animals. Mechanistically, renal alterations were largely heme-independent, since injection of free heme could not reproduce them, and scavenging heme with hemopexin in PHZ-administered mice could not prevent them. Reduced overall health status of the mice suggested multiorgan involvement. We detected amylasemia and amylasuria, two markers of acute pancreatitis. We also provide detailed characterization of renal manifestations associated with acute intravascular hemolysis, which may be mediated by hemolysis-derived products upstream of heme release. This analysis provides a platform for further investigations of hemolytic diseases and associated renal injury and the evaluation of novel therapeutic strategies that target intravascular hemolysis.

          Related collections

          Most cited references52

          • Record: found
          • Abstract: found
          • Article: not found

          The clinical sequelae of intravascular hemolysis and extracellular plasma hemoglobin: a novel mechanism of human disease.

          The efficient sequestration of hemoglobin by the red blood cell membrane and the presence of multiple hemoglobin clearance mechanisms suggest a critical need to prevent the buildup of this molecule in the plasma. A growing list of clinical manifestations attributed to hemoglobin release in a variety of acquired and iatrogenic hemolytic disorders suggests that hemolysis and hemoglobinemia should be considered as a novel mechanism of human disease. Pertinent scientific literature databases and references were searched through October 2004 using terms that encompassed various aspects of hemolysis, hemoglobin preparations, clinical symptoms associated with plasma hemoglobin, nitric oxide in hemolysis, anemia, pulmonary hypertension, paroxysmal nocturnal hemoglobinuria, and sickle-cell disease. Hemoglobin is released into the plasma from the erythrocyte during intravascular hemolysis in hereditary, acquired, and iatrogenic hemolytic conditions. When the capacity of protective hemoglobin-scavenging mechanisms has been saturated, levels of cell-free hemoglobin increase in the plasma, resulting in the consumption of nitric oxide and clinical sequelae. Nitric oxide plays a major role in vascular homeostasis and has been shown to be a critical regulator of basal and stress-mediated smooth muscle relaxation and vasomotor tone, endothelial adhesion molecule expression, and platelet activation and aggregation. Thus, clinical consequences of excessive cell-free plasma hemoglobin levels during intravascular hemolysis or the administration of hemoglobin preparations include dystonias involving the gastrointestinal, cardiovascular, pulmonary, and urogenital systems, as well as clotting disorders. Many of the clinical sequelae of intravascular hemolysis in a prototypic hemolytic disease, paroxysmal nocturnal hemoglobinuria, are readily explained by hemoglobin-mediated nitric oxide scavenging. A growing body of evidence supports the existence of a novel mechanism of human disease, namely, hemolysis-associated smooth muscle dystonia, vasculopathy, and endothelial dysfunction.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Characterization of heme as activator of Toll-like receptor 4.

            Heme is an ancient and ubiquitous molecule present in organisms of all kingdoms, composed of an atom of iron linked to four ligand groups of porphyrin. A high amount of free heme, a potential amplifier of the inflammatory response, is a characteristic feature of diseases with increased hemolysis or extensive cell damage. Here we demonstrate that heme, but not its analogs/precursors, induced tumor necrosis factor-alpha (TNF-alpha) secretion by macrophages dependently on MyD88, TLR4, and CD14. The activation of TLR4 by heme is exquisitely strict, requiring its coordinated iron and the vinyl groups of the porphyrin ring. Signaling of heme through TLR4 depended on an interaction distinct from the one established between TLR4 and lipopolysaccharide (LPS) since anti-TLR4/MD2 antibody or a lipid A antagonist inhibited LPS-induced TNF-alpha secretion but not heme activity. Conversely, protoporphyrin IX antagonized heme without affecting LPS-induced activation. Moreover, heme induced TNF-alpha and keratinocyte chemokine but was ineffective to induce interleukin-6, interleukin-12, and interferon-inducible protein-10 secretion or co-stimulatory molecule expression. These findings support the concept that the broad ligand specificity of TLR4 and the different activation profiles might in part reside in its ability to recognize different ligands in different binding sites. Finally, heme induced oxidative burst, neutrophil recruitment, and heme oxygenase-1 expression independently of TLR4. Thus, our results presented here reveal a previous unrecognized role of heme as an extracellular signaling molecule that affects the innate immune response through a receptor-mediated mechanism.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Heme Oxygenase-1 Mitigates Ferroptosis in Renal Proximal Tubule Cells.

              Ferroptosis is an iron-dependent form of regulated, non-apoptotic cell death, which contributes to damage in models of acute kidney injury (AKI). Heme oxygenase-1 (HO-1) is a cytoprotective enzyme induced in response to cellular stress, and is protective against AKI due to its anti-apoptotic and anti-inflammatory properties. However, the role of HO-1 in regulating ferroptosis is unclear. The purpose of this study was to elucidate the role of HO-1 in regulating ferroptotic cell death in renal proximal tubule cells (PTCs). Immortalized PTCs obtained from HO-1+/+ and HO-1-/- mice were treated with erastin or RSL3, ferroptosis inducers, in the presence or absence of anti-oxidants, an iron source, or an iron chelator. Cells were assessed for changes in morphology and metabolic activity as an indicator of cell viability. Treatment of HO-1+/+ PTCs with erastin resulted in a time- and dose-dependent increase in HO-1 gene expression and protein levels compared to vehicle-treated controls. HO-1-/- cells showed increased dose-dependent erastin- or RSL3-induced cell death in comparison to HO-1+/+ PTCs. Iron supplementation with ferric ammonium citrate in erastin-treated cells decreased cell viability further in HO-1-/- PTCs compared with HO-1+/+ cells. Co-treatment with ferrostatin-1 (ferroptosis inhibitor), deferoxamine (iron chelator), or N-acetyl-L-cysteine (glutathione replenisher) significantly increased cell viability and attenuated erastin-induced ferroptosis in both HO-1+/+ and HO-1-/- PTCs. These results demonstrate an important anti-ferroptotic role of HO-1 in renal epithelial cells.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Immunol
                Front Immunol
                Front. Immunol.
                Frontiers in Immunology
                Frontiers Media S.A.
                1664-3224
                01 March 2018
                2018
                : 9
                : 179
                Affiliations
                [1] 1INSERM, UMR_S 1138, Centre de Recherche des Cordeliers , Paris, France
                [2] 2Sorbonne Universités, UPMC Univ Paris 06 , Paris, France
                [3] 3Université Paris Descartes, Sorbonne Paris Cité , Paris, France
                [4] 4Assistance Publique – Hôpitaux de Paris, Service de néphrologie, Hôpital Européen Georges Pompidou , Paris, France
                [5] 5INSERM, UMR 995 , Lille, France
                [6] 6University of Lille, CHU Lille, Nephrology Department , Lille, France
                [7] 7CSL Behring, R&D , Kankakee, IL, United States
                [8] 8CSL Behring, Research Bern , Bern, Switzerland
                [9] 9Université Paris-Saclay, CNRS, CEA, Univ Evry, Laboratoire Analyse et Modélisation pour la Biologie et l’Environnement , Evry, France
                [10] 10Paris Center for Cardiovascular Research, INSERM UMR_S 970 , Paris, France
                [11] 11Assistance Publique – Hôpitaux de Paris, Service de pathologie, Hôpital Necker enfants malades , Paris, France
                Author notes

                Edited by: Janos G. Filep, Université de Montréal, Canada

                Reviewed by: Mariya Hristova, University College London, United Kingdom; Daniel Ricklin, University of Basel, Switzerland

                *Correspondence: Lubka T. Roumenina, lubka.roumenina@ 123456crc.jussieu.fr

                Specialty section: This article was submitted to Molecular Innate Immunity, a section of the journal Frontiers in Immunology

                Article
                10.3389/fimmu.2018.00179
                5839160
                29545789
                34359565-a408-43d9-a599-835b91109fa4
                Copyright © 2018 Merle, Grunenwald, Figueres, Chauvet, Daugan, Knockaert, Robe-Rybkine, Noe, May, Frimat, Brinkman, Gentinetta, Miescher, Houillier, Legros, Gonnet, Blanc-Brude, Rabant, Daniel, Dimitrov and Roumenina.

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 04 October 2017
                : 19 January 2018
                Page count
                Figures: 6, Tables: 1, Equations: 0, References: 66, Pages: 13, Words: 8840
                Funding
                Funded by: Agence Nationale de la Recherche 10.13039/501100001665
                Award ID: ANR JCJC––INFLACOMP 2015-2018 ANR-15-CE15-0001, ANR JCJC––COBIG ANR-13-JSV1-0006, ANR-11-IDEX-05-02 Recherche-USPC “HEMIR”
                Funded by: CSL Behring 10.13039/100008322
                Funded by: Institut National de la Santé et de la Recherche Médicale 10.13039/501100001677
                Funded by: Centre National de la Recherche Scientifique 10.13039/501100004794
                Categories
                Immunology
                Original Research

                Immunology
                hemolysis,heme,kidney injury,endothelial activation,inflammation,hemopexin,phenylhydrazine,experimental model of intravascular hemolysis

                Comments

                Comment on this article