26
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Ribosome Inactivating Proteins: From Plant Defense to Treatments against Human Misuse or Diseases

      editorial
      , *
      Toxins
      MDPI

      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

          Ribosome inactivating proteins (RIPs) form a vast family of hundreds of toxins from plants, fungi, algae, and bacteria. RIP activities have also been detected in animal tissues. They exert an N-glycosydase catalytic activity that is targeted to a single adenine of a ribosomal RNA, thereby blocking protein synthesis and leading intoxicated cells to apoptosis. In many cases, they have additional depurinating activities that act against other nucleic acids, such as viral RNA and DNA, or genomic DNA. Although their role remains only partially understood, their functions may be related to plant defense against predators and viruses, plant senescence, or bacterial pathogenesis. In this Special Issue, a review by Fabbrini and colleagues [1] addresses our current knowledge about the function and mechanisms of action of plant type I and type II RIPs. In particular, they emphasize the diversity found in their mechanisms of action, although they share sequence and structural identities in their catalytic chain. In another review, De Zaeytijd and Van Damme focus on the heterogeneity of cereal RIPs from an evolutionary perspective, their differences from non-cereal RIPs, and their variety of roles in addition to defense against pathogens and insects [2]. Most RIPs are no threat to human or animal health. However, several bacterial RIPs are major virulence factors involved in severe epidemic diseases such as dysentery or the hemolytic uremic syndrome that may occur in patients suffering from Shiga toxin-producing entero-hemorrhagic Escherichia coli infection. A few RIPs synthesized in plant seeds such as ricin toxin, abrin, or sarcin have been or may be involved in accidental or criminal poisonings, political intimidation, or bio-suicides. In this Special Issue, four contributions address the most recent advances in understanding the three major steps of the intoxication process of cells by Shiga toxins, ricin, and/or sarcin: receptor-binding and triggering of endocytosis, the components of the intracellular trafficking machinery involved in intoxication and binding, and depurination of the ribosome. Johannes describes how the pentavalent binding of Shiga toxins to Gb3 gangliosides in lipid rafts induces membrane structural changes and stress leading to the internalization of the toxin-receptor complexes [3]. Becker and colleagues set up an elegant screening method in yeast, enabling them to not only to confirm the importance of the GARP complex and other protein partners in ricin A chain intracellular trafficking, but also to identify seven new proteins involved along the pathway [4]. This method can now be applied to identify trafficking components used by other RIPs. Li and Tumer analyze the differences in ribosome binding and catalytic activities of the non-activated and activated Shiga and ricin holotoxins, showing opposite behaviors for these two toxins [5]. Finally, Shi et al. describe the structure of the complex of ricin A chain with the C-terminal peptide of the ribosome stalk protein P2 [6]. They discuss the differences in this interaction with that of other RIPs with the same ribosomal target. The pathophysiology of intoxication by the most dangerous RIPs, such as Shiga toxins and ricin toxin, seem much more complicated than a sole link to circulation in the bloodstream and cell death, and is still far from being understood. Diana Karpman and her group review their recent work that brings important progress in the understanding of the mechanisms underlying the hemolytic uremic syndrome provoked by Shiga toxins [7]. They show that Shiga toxins are internalized by red blood cells and then released in microvesicles. It is these toxin-containing microvesicles that participate in the prothrombotic lesions, hemolysis, and transfer of the toxin from the circulation into the kidney, that are characteristic of this deadly syndrome. Furthermore, a research article from the groups of Lee and Park describes for the first time the apoptotic processes induced by Shiga toxins in human retinal pigment epithelial cells, suggesting the mechanisms leading to blindness in severe cases of hemolytic and uremic syndrome [8]. Gal et al. extend their characterization of the crucial role of inflammation in ricin toxin pathogenesis by showing that total body irradiation of mice decreases inflammation markers and extends time to death [9]. Second after ricin toxin, the plant RIP abrin is considered an increasing risk of malevolent and suicidal use. Thus, there is a need for detection and decontamination tools. Tam et al. set up a two-monoclonal antibody-based ELISA that can detect as low as 1 ng/mL of abrin and that shows no false positive detection of other plant RIPs [10]. The same team showed that while various pH treatments of the toxin did not affect its activity, heating above 74 °C completely inactivated its capacity to kill cells and mice. However, they showed that this treatment affects the lectin part of the toxin rather than its catalytic chain [11]. Interestingly, this article sets a correspondence between cytotoxicity testing and the mouse bioassay, which should help reduce the use of the mouse model for the evaluation of abrin and other RIPs. Due to sporadic but recurrent cases of biosuicide and biothreats with ricin toxin, there is an urgent need for a treatment of human intoxication. The group of Kronman gives us a thorough review of existing data on potential countermeasures and treatment strategies, although none are approved for medical use [12]. While antibodies represent the most realistic approach in the case of early post-intoxication intervention, the review stresses the importance of not only eliminating the toxin but also downregulating the explosive inflammatory response triggered by the toxin, as additionally described in a research article by the same group [9]. Interestingly, Whitfield and colleagues describe in detail an F(ab’)2 polyclonal ovine antitoxin and its performance in a mouse model of ricin inhalation that is intended to be pharmaceutically qualified for human use [13]. Protection is mediated both by reducing the amount of circulating toxin and blocking its intracellular trafficking to the Golgi apparatus. Many studies in the past showed that a fraction only of the antibodies generated in the course of an immune response against ricin toxin was neutralizing. Here, the group of Nicholas Mantis identifies the presence of a supercluster of neutralizing epitopes at the interface between the A and B chains of the toxin by analyzing a series of VHH camelid antibody fragments from a phage library generated against ricin toxin [14]. Interestingly, these antibodies do not interfere with the binding of the toxin to the galactose and N-acetyl-galactosamine residues of cell surface glycosylation. This is a step forward in understanding the basis for antibody-mediated protection against this toxin. Hall et al. review the attempts to develop antibodies or other antitoxin strategies to treat the hemolytic and uremic syndrome caused by Shiga toxins, none of which have reached approval [15]. They suggest that the rarity of this disease is a major limit to achieving the necessary clinical trials. Then they advocate the development of drugs targeting the unfolded protein response and the ribotoxic stress response triggered by Shiga toxins as these pathways are involved in many other conditions, which may decrease the barriers to commercial development. The final aspect of research on RIPs covered by this Special Issue concerns their use in the engineering of immunotoxins to target cancer or cells infected by Human immunodeficiency virus (HIV) by conjugation of antibodies or other targeting moieties. Two reviews by Fabbrini et al. and Rust et al. discuss the difficulties that have been encountered in the development of several generations of immunotoxins, none of which have been approved after clinical trials [1,16]. They also present the future trends of immunotoxin development. Two examples of the complexities of such development are given in the articles of Polito et al. and Au et al. The former analyzes the difference in the mechanism of killing of two closely related saporin-containing immunotoxins targeting different markers on B-cell lymphomas, CD20 and CD22 [17]. The latter addresses the effect of PEGylation on the pharmacology, biological activity, and antibody induction of a TAT-maize RIP construction designed to target HIV-infected cells [18]. Overall, this Special Issue of Toxins presents the most recent data on all aspects of RIPs, including function, diversity, evolution, as well as mechanism, pathophysiology, medical countermeasures, and engineering into anticancer drugs.

          Related collections

          Most cited references16

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          Shiga Toxin—A Model for Glycolipid-Dependent and Lectin-Driven Endocytosis

          The cellular entry of the bacterial Shiga toxin and the related verotoxins has been scrutinized in quite some detail. This is due to their importance as a threat to human health. At the same time, the study of Shiga toxin has allowed the discovery of novel molecular mechanisms that also apply to the intracellular trafficking of endogenous proteins at the plasma membrane and in the endosomal system. In this review, the individual steps that lead to Shiga toxin uptake into cells will first be presented from a purely mechanistic perspective. Membrane-biological concepts will be highlighted that are often still poorly explored, such as fluctuation force-driven clustering, clathrin-independent membrane curvature generation, friction-driven scission, and retrograde sorting on early endosomes. It will then be explored whether and how these also apply to other pathogens, pathogenic factors, and cellular proteins. The molecular nature of Shiga toxin as a carbohydrate-binding protein and that of its cellular receptor as a glycosylated raft lipid will be an underlying theme in this discussion. It will thereby be illustrated how the study of Shiga toxin has led to the proposal of the GlycoLipid-Lectin (GL-Lect) hypothesis on the generation of endocytic pits in processes of clathrin-independent endocytosis.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            Treatments for Pulmonary Ricin Intoxication: Current Aspects and Future Prospects

            Ricin, a plant-derived toxin originating from the seeds of Ricinus communis (castor beans), is one of the most lethal toxins known, particularly if inhaled. Ricin is considered a potential biological threat agent due to its high availability and ease of production. The clinical manifestation of pulmonary ricin intoxication in animal models is closely related to acute respiratory distress syndrome (ARDS), which involves pulmonary proinflammatory cytokine upregulation, massive neutrophil infiltration and severe edema. Currently, the only post-exposure measure that is effective against pulmonary ricinosis at clinically relevant time-points following intoxication in pre-clinical studies is passive immunization with anti-ricin neutralizing antibodies. The efficacy of this antitoxin treatment depends on antibody affinity and the time of treatment initiation within a limited therapeutic time window. Small-molecule compounds that interfere directly with the toxin or inhibit its intracellular trafficking may also be beneficial against ricinosis. Another approach relies on the co-administration of antitoxin antibodies with immunomodulatory drugs, thereby neutralizing the toxin while attenuating lung injury. Immunomodulators and other pharmacological-based treatment options should be tailored according to the particular pathogenesis pathways of pulmonary ricinosis. This review focuses on the current treatment options for pulmonary ricin intoxication using anti-ricin antibodies, disease-modifying countermeasures, anti-ricin small molecules and their various combinations.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Microvesicle Involvement in Shiga Toxin-Associated Infection

              Shiga toxin is the main virulence factor of enterohemorrhagic Escherichia coli, a non-invasive pathogen that releases virulence factors in the intestine, causing hemorrhagic colitis and, in severe cases, hemolytic uremic syndrome (HUS). HUS manifests with acute renal failure, hemolytic anemia and thrombocytopenia. Shiga toxin induces endothelial cell damage leading to platelet deposition in thrombi within the microvasculature and the development of thrombotic microangiopathy, mostly affecting the kidney. Red blood cells are destroyed in the occlusive capillary lesions. This review focuses on the importance of microvesicles shed from blood cells and their participation in the prothrombotic lesion, in hemolysis and in the transfer of toxin from the circulation into the kidney. Shiga toxin binds to blood cells and may undergo endocytosis and be released within microvesicles. Microvesicles normally contribute to intracellular communication and remove unwanted components from cells. Many microvesicles are prothrombotic as they are tissue factor- and phosphatidylserine-positive. Shiga toxin induces complement-mediated hemolysis and the release of complement-coated red blood cell-derived microvesicles. Toxin was demonstrated within blood cell-derived microvesicles that transported it to renal cells, where microvesicles were taken up and released their contents. Microvesicles are thereby involved in all cardinal aspects of Shiga toxin-associated HUS, thrombosis, hemolysis and renal failure.
                Bookmark

                Author and article information

                Journal
                Toxins (Basel)
                Toxins (Basel)
                toxins
                Toxins
                MDPI
                2072-6651
                18 April 2018
                April 2018
                : 10
                : 4
                : 160
                Affiliations
                Service d’Ingénierie Moléculaire des Protéines (SIMOPRO), CEA, Université Paris-Saclay, LabEx LERMIT, 91191 Gif-sur-Yvette, France; julien.barbier@ 123456cea.fr
                Author notes
                [* ]Correspondence: Daniel.gillet@ 123456cea.fr ; Tel.: +33-1-69-08-76-46
                Author information
                https://orcid.org/0000-0003-2187-4973
                https://orcid.org/0000-0003-0477-3599
                Article
                toxins-10-00160
                10.3390/toxins10040160
                5923326
                29669991
                7eda9874-b72e-460d-a40b-cbcd8fd1fe23
                © 2018 by the authors.

                Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license ( http://creativecommons.org/licenses/by/4.0/).

                History
                : 11 April 2018
                : 13 April 2018
                Categories
                Editorial

                Molecular medicine
                Molecular medicine

                Comments

                Comment on this article