• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 2
  • Tagged with
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 1
  • 1
  • 1
  • 1
  • 1
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
1

A Comparative Study On The Sensitivity Of Cells Of Different Lineages To Plant Ribosome Inactivating Protein - Abrin

Bora, Namrata 09 1900 (has links)
Proteins with selective toxicity have been investigated for use in many ways. One class of proteins, ribosome-inactivating proteins (RIPs), is found throughout the plant kingdom as well as in lower organisms like certain fungi and bacteria. These are a group of proteins that has the property of damaging the ribosomes in an irreversible manner. They are N-glycosidases that modify the 28S rRNAs to render them incapable of sustaining further translation. RIPs have been divided into two groups, i.e. type I RIPs, which are single polypeptide chains and type II RIPs, which are heterodimeric. Abrin is a type II RIP, isolated from the seeds of Abrus precatorius plant commonly known as jequirity plant. It is a heterodimeric glycoprotein consisting of an A and a B subunit linked together by a single disulfide bond. The toxicity of the protein comes from the A subunit harboring the RNA-N- glycosidase activity which catalyses the depurination of a specific adenine residue at position 4324 on the 28S rRNA. The depurination of the adenine prevents the formation of a critical stem loop structure to which the elongation factor -2 (EF-2) binds during the translocation step of the translation, thus stalling the translation machinery of the cells. The B subunit of abrin is a galactose specific lectin. The lectin activity enables the protein toxin to bind to the cell surface glycoproteins and/or glycolipids. Binding of abrin is followed by internalization of the protein by receptor mediated endocytosis and transport to the Endoplasmic reticulum (ER) by the retrograde transport pathway. Inside the ER, the single disulfide bond linking the two subunits, is reduced which is important for the A subunit toxicity. The A subunit then translocates into the cytosol using the ER-associated degradation (ERAD) pathway and cleaves the specific adenine residue on the 28S rRNA of the 60 S ribosome involved in active translation and thereby inhibiting the protein synthesis. In addition to its ability to inhibit translation, abrin induces apoptosis in cells. Earlier work from our laboratory has shown that abrin-induced apoptosis follows the intrinsic pathway of apoptotic cell death. The treated cells show mitochondrial membrane potential loss followed by caspases -9 and -3 activation and DNA fragmentation. RIPs have been used primarily in immunotherapy because of their toxicity at very low concentrations (picomolar). With the development of monoclonal antibodies as tool for targeting cell surface markers, the possibility to couple antibodies to RIPs and thus deliver the toxic protein directly to specific cells becomes feasible. Abrin, as one such potent RIP, has gained interest in the field of medicine and immunotherapeutics. Abrin can also be a candidate for use in bioterrorism and warfare. Therefore, it is very important to first understand the inhibitory effect of abrin and the extent of its toxicity on cells. Earlier studies from our laboratory have focused on the sensitivity and mechanism of cell death induced by abrin in Jurkat cells, a T –cell line. In the present study, we attempted to investigate the overall toxicity of the molecule with respect to both properties, inhibition of protein translation and induction of apoptosis, in different lineages of cells. We have carried out a comparative study on abrin toxicity on human cell lines from two different cell lineages namely hematopoietic and epithelial. The thesis is divided into introduction and two chapters. In the introduction, we have presented the general properties of this family of proteins, with a brief history; classification and distribution of plant RIPs and their enzymatic properties. The chapter also deals with possible usage of these proteins, mainly in the field of immunotherapy. We have introduced, abrin, the protein of our interest in this chapter. The structure of abrin is described and also the biological effects of the toxin are discussed in brief. The chapter one deals with the translation inhibitory property of the protein, abrin. As mentioned earlier, abrin inhibits protein synthesis via the RNA-N-glycosidase activity residing in its A-chain. We have presented the general cytotoxic pathway of type II RIPs in this chapter. It deals with the internalization and transport of the toxin to their site of action, the cytosol. As reported earlier, our results confirmed that abrin inhibited protein synthesis in all cells. Abrin mediated inhibition of translation was dose dependent. Though the inhibition was common to all the cells from both the lineages, the sensitivity of the cells towards the toxin and kinetics of this inhibition event differed significantly. The kinetics of inhibition of protein synthesis is faster in case of hematopoietic cells as compared to the epithelial cells even at lower doses of the toxin. These differences were not due to variations in the ability of protein synthesis of cells. The chapter also discusses binding of the protein to cells. Our data suggest that binding of abrin to the cells is not responsible for the variations observed in the translation inhibitory property of the protein except in Raji cells. The B-cell line Raji was found to be least sensitive towards the toxin. Our studies show that due to presence of high sialic acid residues on the surface of these cells, Raji cells are refractory to abrin mediated inhibition of protein synthesis. The second chapter presents our data on cell death upon abrin treatment. This part is divided into an introduction and two sections, A and B. In the introduction, different cell death modalities are discussed along with recent findings in the field of programmed cell death. Section A deals with abrin induced apoptosis in epithelial cells. We have compared the extent of abrin-triggered apoptosis in these cells. Some of the early events known in the apoptotic cascade of abrin are compared. Though apoptosis is observed in these cells, our data suggest a delay in the apoptotic trigger in the epithelial cells showing that epithelial cells can survive the stress induced by abrin for a longer time. When treated with other apoptotic agents, like etoposide, these cells are found to be resistant. Therefore, though there is a delay in the trigger of apoptosis, we have shown that the cells tested from the epithelial lineage undergo apoptosis on abrin treatment. Section B, discusses the ability of the protein to induce cell death in hematopoietic cells. We have presented studies on cell death other than apoptosis, detected in these cells upon abrin treatment. We found that some of the cell lines tested undergoes more necrosis than apoptosis with abrin treatment. When the status of the mitochondria was checked, we found that in U266B1 cells, a B-cell line, there was mitochondrial stress as well as reactive oxygen species (ROS) production. But these cells died by necrosis. The data obtained from this study show the involvement of lysosomes and cathepsins in abrin induced cell death in U266B1 cells. Though other cells also undergo necrosis, these events were unique to U266B1 cells.
2

Mechanism of Abrin-Induced Apoptosis and Insights into the Neutralizing Activity of mAb D6F10

Mishra, Ritu January 2014 (has links) (PDF)
Abrin is a potent toxin obtained from the seeds of Abrus precatorius. It is a heterodimeric glycoprotein consisting of an A and a B subunit linked together by a disulfide bond. The toxicity of the protein comes from the A subunit harboring RNA-N-glycosidase activity which cleaves the glycosidic bond between the ribose sugar and the adenine at position 4324 in 28S rRNA. The depurination of a specific adenine residue at position 4324 results in loss of conformation of the 28S rRNA at the α sarcin/ricin loop to which elongation factor-2 (EF-2) binds, during the transloction step of translation, leading to inhibition of protein synthesis. The B subunit of abrin is a galactose specific lectin. The lectin activity enables the toxin to gain entry inside cells on binding to receptors with terminal galactose. After entering cells, a few molecules of abrin reach the endoplasmic reticulum (ER) via the retrograde transport, where the disulfide bond between the A and the B subunits gets cleaved. Then the A chain escapes into the cytosol where it binds to its target, the α-sarcin loop of the 28S ribosomal RNA and inhibits protein synthesis. Apart from inhibition of protein synthesis, exposure of cells to abrin leads to the loss of mitochondrial membrane potential (MMP) resulting in the activation of caspases and finally apoptosis. However, whether apoptosis is dependent on the inhibition of protein synthesis has not been elucidated. The major objectives of this study are therefore to delineate the signaling pathways involved in abrin-induced apoptosis. The thesis is divided into 4 Chapters: Chapter 1. provides a overview of the general properties of RIPs, with a brief history, classification, trafficking and biological activities of the toxins. This chapter also discusses their potential use in bio-warfare and the treatments available for management of toxicity. Chapter 2 and 3 discuss the results obtained on studies aimed at gaining insights into the signaling pathways involved in abrin-induced apoptosis. Chapter 4 focuses on the research carried out to understand the mechanisms of neutralization of abrin by the mAb D6F10. Towards the first objective, chapter 2 elucidates the role of endoplasmic reticulum (ER) stress signaling in abrin-induced apoptosis using the human T-cell line, Jurkat as a model system. It could be concluded that the inhibition of protein synthesis by the catalytic A subunit of abrin could result in accumulation of unfolded proteins in the ER leading to ER stress which triggers the unfolded protein response (UPR) pathway. The ER resident trans-membrane sensors IRE1 (Inositol-requiring enzyme 1), PERK (PKR-like ER kinase) and ATF6 (Activating transcription factor 6) are the important players of UPR in mammalian cells. These sensors inhibit translation and increase the levels of chaperones to restore protein homeostasis. However, if the ER stress is prolonged, apoptotic pathways get activated to remove severely damaged cells in which protein folding defects cannot be resolved. Recent studies have shown that endoplasmic reticulum (ER) stress induces apoptosis by activating initiater caspases such as caspase-2 and -8 which eventually trigger mitochondrial membrane potential loss and activation of downstream effector capases-9 and -3. Phosphorylation of eukaryotic initiation factor 2α and upregulation of CHOP [CAAT/enhancer binding protein (C/EBP) homologous protein], important players involved in ER stress signaling by abrin, suggested activation of ER stress in the cells. ER stress is also known to induce apoptosis via stress kinases such as p38 MAPK and JNK. Activation of both the pathways was observed upon abrin treatment and found to be upstream of the activation of caspases. However, abrin-induced apoptosis was found be dependent on p38 MAPK but not JNK. We also observed that abrin induced activation of caspase-2 and caspase-8 and triggered Bid cleavage leading to mitochondrial membrane potential loss and thus connecting the signaling events from ER stress to mitochondrial death machinery. Few toxins belonging to the family of ribosome inactivating proteins such as Shiga toxin have been observed to induce DNA damage in human endothelial cells and activate p53/ATM-dependent signaling pathway in mammalian cells. To further investigate the role of abrin on activation of DNA damage signaling pathway, we analysed the phosphorylation of H2AX and ATM, which are markers for double strand DNA breaks. We observed phosphorylation of H2AX and ATM upon abrin treatment but not when cells were pretreated with the broad spectrum pan caspase inhibitor. This study suggested that the DNA damage observed was an indirect effect of caspase-activated DNase. We concluded from the studies in chapter 2 that inhibition of protein synthesis by abrin can trigger endoplasmic reticulum stress leading to mitochondria-mediated apoptosis. Further studies were conducted to understand the dependence of ER stress on inhibition of protein synthesis and are presented in chapter 3. For this study, we have used an active site mutant of abrin A chain (R167L) which exhibits lower protein synthesis inhibitory activity than the wild type abrin A chain. Recombinant wild type and mutant abrin A chains were expressed in E.coli and purified. Since, abrin A chain requires the B chain for internalization into cells, both wild type and mutant abrin A chains were conjugated to native ricin B chain to generate a hybrid toxin. Next, we have compared the toxic effects of the two conjugates in cells. The rate of inhibition of protein synthesis mediated by the mutant ricin B-rABRA (R167L) conjugate was slower than that of the wild type ricin B-rABRA conjugate but it could trigger ER stress leading to mitochondrial mediated apoptosis in cells though delayed, suggesting that inhibition of protein synthesis is the major factor contributing to abrin-mediated apoptosis. Abrin is extremely lethal and considered as a potential agent for use in biological warfare. Currently, there are no antidotes or effective therapies available for abrin poisoning. Antibody based antitoxins function by either preventing toxin binding to cell surface receptors or by translocation. Antibodies against the B chain of RIPs function by inhibiting the binding of B chain of the toxin to cells, whereas the exact mechanism by which antibodies against A chain function is still not clear. The only known neutralizing monoclonal antibody against abrin A chain, namely, D6F10, was generated in our laboratory and was shown to rescue cells and mice from abrin intoxication. Earlier experiments with confocal microscopy suggested that mAb D6F10 could internalize in HeLa cells along with abrin, suggesting that the antibody can function intracellularly. Chapter 4 discusses the work carried out to delineate the mechanism of intracellular neutralization of abrin by the mAb D6F10. We observed significant reduction in binding and delay in abrin internalization in the presence of the neutralizing monoclonal antibody (mAb) D6F10. Considering that the majority of the abrin after internalization is removed by lysosomal degradation, we studied the fate of abrin in the presence of mAb D6F10. Confocal images did not show any difference in the distribution of abrin in the lysosomes in the absence or presence of antibody. However, the antibody remained persistently colocalized with abrin in the cells, suggesting that the antibody might inhibit enzymatic activity of abrin at its cellular site of action.

Page generated in 0.065 seconds