Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Am J Physiol Lung Cell Mol Physiol ; 326(5): L524-L538, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38375572

ABSTRACT

Lung surfactant collectins, surfactant protein A (SP-A) and D (SP-D), are oligomeric C-type lectins involved in lung immunity. Through their carbohydrate recognition domain, they recognize carbohydrates at pathogen surfaces and initiate lung innate immune response. Here, we propose that they may also be able to bind to other carbohydrates present in typical cell surfaces, such as the alveolar epithelial glycocalyx. To test this hypothesis, we analyzed and quantified the binding affinity of SP-A and SP-D to different sugars and glycosaminoglycans (GAGs) by microscale thermophoresis (MST). In addition, by changing the calcium concentration, we aimed to characterize any consequences on the binding behavior. Our results show that both oligomeric proteins bind with high affinity (in nanomolar range) to GAGs, such as hyaluronan (HA), heparan sulfate (HS) and chondroitin sulfate (CS). Binding to HS and CS was calcium-independent, as it was not affected by changing calcium concentration in the buffer. Quantification of GAGs in bronchoalveolar lavage (BAL) fluid from animals deficient in either SP-A or SP-D showed changes in GAG composition, and electron micrographs showed differences in alveolar glycocalyx ultrastructure in vivo. Taken together, SP-A and SP-D bind to model sulfated glycosaminoglycans of the alveolar epithelial glycocalyx in a multivalent and calcium-independent way. These findings provide a potential mechanism for SP-A and SP-D as an integral part of the alveolar epithelial glycocalyx binding and interconnecting free GAGs, proteoglycans, and other glycans in glycoproteins, which may influence glycocalyx composition and structure.NEW & NOTEWORTHY SP-A and SP-D function has been related to innate immunity of the lung based on their binding to sugar residues at pathogen surfaces. However, their function in the healthy alveolus was considered as limited to interaction with surfactant lipids. Here, we demonstrated that these proteins bind to glycosaminoglycans present at typical cell surfaces like the alveolar epithelial glycocalyx. We propose a model where these proteins play an important role in interconnecting alveolar epithelial glycocalyx components.


Subject(s)
Calcium , Glycocalyx , Glycosaminoglycans , Pulmonary Alveoli , Pulmonary Surfactant-Associated Protein A , Pulmonary Surfactant-Associated Protein D , Animals , Humans , Mice , Alveolar Epithelial Cells/metabolism , Bronchoalveolar Lavage Fluid , Calcium/metabolism , Glycocalyx/metabolism , Glycosaminoglycans/metabolism , Heparitin Sulfate/metabolism , Mice, Inbred C57BL , Protein Binding , Pulmonary Alveoli/metabolism , Pulmonary Surfactant-Associated Protein A/metabolism , Pulmonary Surfactant-Associated Protein D/metabolism
2.
Nucleic Acids Res ; 48(11): 6210-6222, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32365182

ABSTRACT

The ribotoxin α-sarcin belongs to a family of ribonucleases that cleave the sarcin/ricin loop (SRL), a critical functional rRNA element within the large ribosomal subunit (60S), thereby abolishing translation. Whether α-sarcin targets the SRL only in mature 60S subunits remains unresolved. Here, we show that, in yeast, α-sarcin can cleave SRLs within late 60S pre-ribosomes containing mature 25S rRNA but not nucleolar/nuclear 60S pre-ribosomes containing 27S pre-rRNA in vivo. Conditional expression of α-sarcin is lethal, but does not impede early pre-rRNA processing, nuclear export and the cytoplasmic maturation of 60S pre-ribosomes. Thus, SRL-cleaved containing late 60S pre-ribosomes seem to escape cytoplasmic proofreading steps. Polysome analyses revealed that SRL-cleaved 60S ribosomal subunits form 80S initiation complexes, but fail to progress to the step of translation elongation. We suggest that the functional integrity of a α-sarcin cleaved SRL might be assessed only during translation.


Subject(s)
Endoribonucleases/metabolism , Fungal Proteins/metabolism , Ribosome Subunits, Large, Eukaryotic/chemistry , Ribosome Subunits, Large, Eukaryotic/metabolism , Ricin/metabolism , Saccharomyces cerevisiae/metabolism , Active Transport, Cell Nucleus , Cell Nucleolus/drug effects , Cell Nucleolus/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Endoribonucleases/pharmacology , Fungal Proteins/pharmacology , Protein Biosynthesis , RNA, Ribosomal/metabolism , Ricin/chemistry , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/growth & development
3.
Int J Biol Macromol ; 136: 625-631, 2019 Sep 01.
Article in English | MEDLINE | ID: mdl-31220501

ABSTRACT

Ribotoxins are fungal proteins that serve as weapons against parasites and insects. They are strongly toxic due to their ability to enter host cells and inactivate ribosomes. Ageritin is the prototype of a new ribotoxin-like protein family present in basidiomycetes. We demonstrate that this enzyme has peculiar binding and enzymatic features. Different from other ribotoxins, its ribonucleolytic activity requires the presence of divalent cations, with a maximum activation in the presence of zinc ions, for which Ageritin exhibits the strongest affinity of binding. We modeled the catalytic metal binding site of Ageritin, made of the putative triad Asp68, Asp70 and His77. This report highlights that Ageritin has the structure and function of an RNase but a Mg2+/Zn2+-dependent mechanism of action, a new finding for ribotoxins. As a zinc-dependent toxin, Ageritin can be classified among the arsenal of zinc-binding proteins involved in fungal virulence.


Subject(s)
Agrocybe/enzymology , Ribonucleases/chemistry , Ribonucleases/metabolism , Zinc/metabolism , Catalytic Domain , Colicins/metabolism , Models, Molecular , Protein Binding
4.
Biochim Biophys Acta Gen Subj ; 1862(12): 2888-2894, 2018 12.
Article in English | MEDLINE | ID: mdl-30262416

ABSTRACT

Ageritin has been recently described as the first ribotoxin-like from Basidiomycota division (mushroom Agrocybe aegerita) with known antitumor activity (BBA 2017, 1861: 1113-1121). By investigating structural, catalytic and binding properties, we demonstrate that Ageritin is a unique ribotoxin-like protein. Indeed, typical of the ribotoxin family, it shows the specific ribonucleolytic activity against the ribosomal Sarcin-Ricin Loop in a rabbit reticulocytes assay. However, it displays several elements of novelty, as this activity is strongly metal-dependent and completely suppressed in the presence of EDTA, different from other representative members of the ribotoxin family. Consistently, we prove that Ageritin is able to bind magnesium ions with low micromolar affinity. We also show that Ageritin is significantly more stable than other ribotoxins in thermal and chemical denaturation experiments. These peculiar features make Ageritin the prototype of a new ribotoxin-like family present in basidiomycetes. Finally, given its high stability, this enzyme is a promising candidate as a new tool in immunoconjugates and nanoconstructs.


Subject(s)
Agrocybe/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Magnesium/metabolism , Ribonucleases/chemistry , Ribonucleases/pharmacology , Ribosomes/drug effects , Toxins, Biological/chemistry , Toxins, Biological/pharmacology , Animals , Calorimetry , Circular Dichroism , Electrophoresis, Polyacrylamide Gel , Protein Binding , Protein Stability , Protein Structure, Secondary , Rabbits , Ribosomes/metabolism , Spectrophotometry, Ultraviolet
5.
Arch Biochem Biophys ; 619: 45-53, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28283321

ABSTRACT

Fungal ribotoxins are highly specific extracellular RNases which cleave a single phosphodiester bond at the ribosomal sarcin-ricin loop, inhibiting protein biosynthesis by interfering with elongation factors. Most ribotoxins show high degree of conservation, with similar sizes and amino acid sequence identities above 85%. Only two exceptions are known: hirsutellin A and anisoplin, produced by the entomopathogenic fungi Hirsutella thompsonii and Metarhizium anisopliae, respectively. Both proteins are similar but smaller than the other known ribotoxins (130 vs 150 amino acids), displaying only about 25% sequence identity with them. They can be considered minimized natural versions of their larger counterparts, best represented by α-sarcin. The conserved α-sarcin active site residue Tyr48 has been replaced by the geometrically equivalent Asp, present in the minimized ribotoxins, to produce and characterize the corresponding mutant. As a control, the inverse anisoplin mutant (D43Y) has been also studied. The results show how the smaller versions of ribotoxins represent an optimum compromise among conformational freedom, stability, specificity, and active-site plasticity which allow these toxic proteins to accommodate the characteristic abilities of ribotoxins into a shorter amino acid sequence and more stable structure of intermediate size between that of other nontoxic fungal RNases and previously known larger ribotoxins.


Subject(s)
Fungal Proteins/chemistry , Fungi/enzymology , Metarhizium/enzymology , Ribonucleases/chemistry , Catalytic Domain , Endoribonucleases/chemistry , Escherichia coli/metabolism , Mutation , Peptide Elongation Factors/chemistry , Protein Biosynthesis , Protein Conformation , Ribosomes/metabolism , Spectrophotometry, Ultraviolet , Tyrosine/chemistry
6.
Toxins (Basel) ; 9(2)2017 02 21.
Article in English | MEDLINE | ID: mdl-28230789

ABSTRACT

Fungi establish a complex network of biological interactions with other organisms in nature. In many cases, these involve the production of toxins for survival or colonization purposes. Among these toxins, ribotoxins stand out as promising candidates for their use in biotechnological applications. They constitute a group of highly specific extracellular ribonucleases that target a universally conserved sequence of RNA in the ribosome, the sarcin-ricin loop. The detailed molecular study of this family of toxic proteins over the past decades has highlighted their potential in applied research. Remarkable examples would be the recent studies in the field of cancer research with promising results involving ribotoxin-based immunotoxins. On the other hand, some ribotoxin-producer fungi have already been studied in the control of insect pests. The recent role of ribotoxins as insecticides could allow their employment in formulas and even as baculovirus-based biopesticides. Moreover, considering the important role of their target in the ribosome, they can be used as tools to study how ribosome biogenesis is regulated and, eventually, may contribute to a better understanding of some ribosomopathies.


Subject(s)
Fungal Proteins , Fungi/enzymology , Mycotoxins , Ribonucleases , Animals , Biotechnology , Fungal Proteins/metabolism , Fungal Proteins/toxicity , Humans , Mycotoxins/metabolism , Mycotoxins/toxicity , Ribonucleases/metabolism , Ribonucleases/toxicity , Ribosomes
7.
Biol Chem ; 398(1): 135-142, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27472070

ABSTRACT

Metarhizium anisopliae is an entomopathogenic fungus relevant in biotechnology with applications like malaria vector control. Studies of its virulence factors are therefore of great interest. Fungal ribotoxins are toxic ribonucleases with extraordinary efficiency against ribosomes and suggested as potential insecticides. Here we describe this ribotoxin characteristic activity in M. anisopliae cultures. Anisoplin has been obtained as a recombinant protein and further characterized. It is structurally similar to hirsutellin A, the ribotoxin from the entomopathogen Hirsutella thompsonii. Moreover, anisoplin shows the ribonucleolytic activity typical of ribotoxins and cytotoxicity against insect cells. How Metarhizium uses this toxin and possible applications are of interest.


Subject(s)
Metarhizium , Ribonucleases/chemistry , Ribonucleases/toxicity , Toxins, Biological/chemistry , Toxins, Biological/toxicity , Amino Acid Sequence , Animals , Sf9 Cells , Spodoptera
8.
Biol Chem ; 397(2): 135-45, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26584355

ABSTRACT

Ribotoxins are cytotoxic members of the family of fungal extracellular ribonucleases best represented by RNase T1. They share a high degree of sequence identity and a common structural fold, including the geometric arrangement of their active sites. However, ribotoxins are larger, with a well-defined N-terminal ß-hairpin, and display longer and positively charged unstructured loops. These structural differences account for their cytotoxic properties. Unexpectedly, the discovery of hirsutellin A (HtA), a ribotoxin produced by the invertebrate pathogen Hirsutella thompsonii, showed how it was possible to accommodate these features into a shorter amino acid sequence. Examination of HtA N-terminal ß-hairpin reveals differences in terms of length, charge, and spatial distribution. Consequently, four different HtA mutants were prepared and characterized. One of them was the result of deleting this hairpin [Δ(8-15)] while the other three affected single Lys residues in its close spatial proximity (K115E, K118E, and K123E). The results obtained support the general conclusion that HtA active site would show a high degree of plasticity, being able to accommodate electrostatic and structural changes not suitable for the other previously known larger ribotoxins, as the variants described here only presented small differences in terms of ribonucleolytic activity and cytotoxicity against cultured insect cells.


Subject(s)
Fungal Proteins/chemistry , Fungal Proteins/pharmacology , Insecticides/chemistry , Insecticides/pharmacology , Lysine/metabolism , Spodoptera/cytology , Spodoptera/drug effects , Amino Acid Sequence , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Fungal Proteins/genetics , Models, Molecular , Molecular Sequence Data , Mutation , Protein Conformation , Static Electricity , Structure-Activity Relationship
9.
Toxicon ; 96: 1-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25598497

ABSTRACT

Ribotoxins are a family of fungal ribosome-inactivating proteins displaying highly specific ribonucleolytic activity against the sarcin/ricin loop (SRL) of the larger rRNA, with α-sarcin as its best-characterized member. Their toxicity arises from the combination of this activity with their ability to cross cell membranes. The involvement of α-sarcin's loops 2 and 3 in SRL and ribosomal proteins recognition, as well as in the ribotoxin-lipid interactions involving cell penetration, has been suggested some time ago. In the work presented now different mutants have been prepared in order to study the role of these loops in their ribonucleolytic and lipid-interacting properties. The results obtained confirm that loop 3 residues Lys 111, 112, and 114 are key actors of the specific recognition of the SRL. In addition, it is also shown that Lys 114 and Tyr 48 conform a network of interactions which is essential for the catalysis. Lipid-interaction studies show that this Lys-rich region is indeed involved in the phospholipids recognition needed to cross cell membranes. Loop 2 is shown to be responsible for the conformational change which exposes the region establishing hydrophobic interactions with the membrane inner leaflets and eases penetration of ribotoxins target cells.


Subject(s)
Endoribonucleases/chemistry , Endoribonucleases/toxicity , Fungal Proteins/chemistry , Fungal Proteins/toxicity , Models, Molecular , Protein Synthesis Inhibitors/toxicity , Ribosomes/drug effects , Absorption, Physicochemical , Amino Acid Sequence , Animals , Catalysis , Cell Line , Circular Dichroism , Cloning, Molecular , DNA, Complementary/genetics , Endoribonucleases/genetics , Escherichia coli , Fungal Proteins/genetics , Molecular Sequence Data , Mutagenesis , Oligonucleotides/genetics , Phospholipids/metabolism , Protein Binding , Protein Conformation , Sequence Alignment , Spectrophotometry , Spodoptera
10.
Toxicon ; 83: 69-74, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24631599

ABSTRACT

Ribotoxins are fungal extracellular ribonucleases highly toxic due to their ability to enter host cells and their effective ribonucleolytic activity against the ribosome. The natural role of these proteins in the producing fungi is still unsolved. Nevertheless, recent studies showing the insecticidal properties of two ribotoxins from different origin support their involvement in defense mechanisms. Thus, it seems that not just the entomopathogen Hirsutella thompsonii expresses the ribotoxin hirsutellin A as a virulence factor but also Aspergillus, the main ribotoxin producer, does so. In this review we focus on this little known aspect of this family of proteins, their toxicity against insects, from the point of view of its biological relevance and its potential biotechnological applications.


Subject(s)
Acaricides/chemistry , Cytotoxins/chemistry , Fungal Proteins/chemistry , Hypocreales/chemistry , Pest Control, Biological , Ribonucleases , Acaricides/isolation & purification , Amino Acid Sequence , Cytotoxins/isolation & purification , Fungal Proteins/isolation & purification , Fungal Proteins/physiology , Molecular Sequence Data , Sequence Alignment
11.
Biochemistry ; 53(10): 1545-7, 2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24568582

ABSTRACT

The ribosomal sarcin/ricin loop (SRL) is the target of ribosome-inactivating proteins like the N-glycosidase ricin and the fungal ribotoxin α-sarcin. The eukaryotic ribosomal stalk directly interacts with several members of the N-glycosidase family, favoring their disruption of the SRL. Here we tested this hypothesis for the ribotoxin α-sarcin. Experiments with isolated ribosomes, cell-free translation systems, and viability assays with Saccharomyces cerevisiae strains defective in acidic stalk proteins showed that the inactivation exerted by α-sarcin is independent of the composition of the ribosomal stalk. Therefore, α-sarcin, with the same ribosomal target as ricin, seems to access the SRL by a different pathway.


Subject(s)
Endoribonucleases/metabolism , Fungal Proteins/metabolism , Protein Biosynthesis , Ribosomal Proteins/metabolism , Ribosomes/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Ribosomal Proteins/genetics , Ribosomes/genetics , Ricin/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
12.
Insects ; 4(3): 339-56, 2013 Jul 09.
Article in English | MEDLINE | ID: mdl-26462423

ABSTRACT

The fungal pathogen Hirsutella thompsonii produces an insecticidal protein named hirsutellin A (HtA), which has been described to be toxic to several species of mites, insect larvae, and cells. On the other hand, on the basis of an extensive biochemical and structural characterization, HtA has been considered to be a member of the ribotoxins family. Ribotoxins are fungal extracellular ribonucleases, which inactivate ribosomes by specifically cleaving a single phosphodiester bond located at the large rRNA. Although ribotoxins were brought to light in the 1960s as antitumor agents, their biological function has remained elusive. Thus, the consideration of hirsutellin A, an insecticidal protein, as a singular ribotoxin recalled the idea of the biological activity of these toxins as insecticidal agents. Further studies have demonstrated that the most representative member of the ribotoxin family, α-sarcin, also shows strong toxic action against insect cells. The determination of high resolution structures, the characterization of a large number of mutants, and the toxicity assays against different cell lines have been the tools used for the study of the mechanism of action of ribotoxins at the molecular level. The aim of this review is to serve as a compilation of the facts that allow identification of HtA as a paradigmatic example of the insecticidal function of fungal ribotoxins.

13.
Insect Biochem Mol Biol ; 43(1): 39-46, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23153726

ABSTRACT

Fungal ribotoxins were discovered almost 50 years ago as extracellular ribonucleases (RNases) with antitumoral properties. However, the biological function of these toxic proteins has remained elusive. The discovery of the ribotoxin HtA, produced by the invertebrates pathogen Hirsutella thompsonii, revived the old proposal that insecticidal activity would be their long searched function. Unfortunately, HtA is rather singular among all ribotoxins known in terms of sequence and structure similarities. Thus, it was intriguing to answer the question of whether HtA is just an exception or, on the contrary, the paradigmatic example of the ribotoxins function. The work presented uses HtA and α-sarcin, the most representative member of the ribotoxins family, to show their strong toxic action against insect larvae and cells.


Subject(s)
Endoribonucleases/isolation & purification , Fungal Proteins/isolation & purification , Insecticides/isolation & purification , Mycotoxins/isolation & purification , Animals , Endoribonucleases/pharmacology , Fungal Proteins/pharmacology , Insecticides/pharmacology , Moths , Mycotoxins/pharmacology , Ribosomes/drug effects , Sf9 Cells
14.
FEBS J ; 279(17): 3022-32, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22748038

ABSTRACT

Within the last 10 years, the use of different RNases as therapeutic agents for various diseases has been pursued. Furthermore, the advancements of recombinant technology have allowed the assembly of proteins with different functions. In this regard, immunoribonucleases (immunoRNases) stand out as some of the most promising therapeutic candidates given their enzymatic and non-mutagenic character. Accordingly, the work reported here describes fusing RNase T1, one of the most studied members of the microbial RNase family, to the single-chain variable fragment (scFv) of a monoclonal antibody that targets the glycoprotein A33 antigen (GPA33) from human colon cancer cells. A heterologous production system, which employs the yeast Pichia pastoris, has been optimized to produce this immunoRNase (scFvA33T1) with yields of ∼ 5-10 mg · L(-1). The purified protein appears to be correctly folded as it retains its antigen specificity and ribonucleolytic activity. Finally, it also shows specific binding to, internalization into and toxicity against GPA33-positive cell lines compared with the control, GPA33-negative cells. Overall, it can be concluded that scFvA33T1 is a promising therapeutic fusion protein with the additional advantage that presumably it can be produced and purified in large amounts using an easily scalable yeast-based system.


Subject(s)
Colonic Neoplasms/enzymology , Ribonuclease T1/metabolism , Single-Chain Antibodies , Circular Dichroism , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Electrophoresis, Polyacrylamide Gel , Endocytosis , Humans , Spectrophotometry, Ultraviolet
15.
Protein Eng Des Sel ; 25(8): 425-35, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22718791

ABSTRACT

A single-chain fusion protein that directed the cytolytic activity of α-sarcin to A33 tumor antigen expressing cells was constructed and shown to effectively kill targeted cells. Glycoprotein A33 (GPA33) is a well-known colon cancer marker and a humanized antibody against it was used to target the α-sarcin. The fungal ribotoxin α-sarcin is one of the most potent and specific toxins known. It is small, protease resistant, thermostable and highly efficient towards the inactivation of ribosomes. This work describes the production and characterization of an immunotoxin resulting from fusing the single-chain variable fragment (scFv) of the monoclonal antibody that targets GPA33 to fungal α-sarcin. This chimeric protein (scFvA33αsarcin), produced in Pichia pastoris and purified in high yield was proven to be properly folded, active, specific and stable. It showed high specific toxicity against GPA33-positive tumoral cell lines providing scientific evidence to sustain that scFvA33αsarcin is a good immunotherapeutic candidate against GPA33-positive colon carcinomas.


Subject(s)
Colonic Neoplasms/metabolism , Endoribonucleases/chemistry , Fungal Proteins/chemistry , Immunotoxins/chemistry , Membrane Glycoproteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Single-Chain Antibodies/chemistry , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/immunology , Endoribonucleases/genetics , Endoribonucleases/metabolism , Flow Cytometry , Fungal Proteins/genetics , Fungal Proteins/metabolism , Humans , Immunotoxins/genetics , Immunotoxins/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Microscopy, Fluorescence , Pichia/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Single-Chain Antibodies/genetics , Single-Chain Antibodies/metabolism
16.
Biol Chem ; 393(6): 449-56, 2012 May.
Article in English | MEDLINE | ID: mdl-22628308

ABSTRACT

Ribotoxins are a family of toxic proteins that exert a highly specific cleavage at the universally conserved sarcin/ricin loop (SRL) of the larger rRNA molecule. Before this ribonucleolytic action, passage through the cell membrane is a necessary step for ribotoxin internalization and the limiting factor for cytotoxicity. Although extensive knowledge of their ribonucleolytic activity and substrate recognition has been accumulated, little is known about the mechanisms of cell entry of ribotoxins. Hirsutellin A (HtA) is a recently described member of this family, which accommodates the main abilities of previously characterized ribotoxins into a shorter sequence, but exhibits some differences regarding membrane interaction properties. This work investigates the contribution of tryptophan (Trp) residues 71 and 78 to both endoribonucleolytic activity and cellular toxicity of this ribotoxin. Substitution mutants W71F and W78F, as well as the double mutant W71/78F, were obtained and assayed against isolated ribosomes, synthetic SRL, and human tumor cells. The results provide evidence that cell membrane passage and internalization, as well as substrate-specific recognition, require the participation of the region involving both Trp 71 and Trp 78. Additionally, the mutant W71/78F is the first non-cytotoxic but specific ribosome-cleaving ribotoxin mutant obtained to date.


Subject(s)
Cytotoxins/chemistry , Cytotoxins/toxicity , Fungal Proteins/chemistry , Fungal Proteins/toxicity , RNA, Ribosomal/metabolism , Ribonucleases/chemistry , Ribonucleases/metabolism , Tryptophan/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Conserved Sequence , Cytotoxins/genetics , Cytotoxins/metabolism , Endoribonucleases/chemistry , Fungal Proteins/genetics , Fungal Proteins/metabolism , Humans , Models, Molecular , Mutation , Protein Conformation , Protein Synthesis Inhibitors/chemistry , Protein Synthesis Inhibitors/metabolism , Protein Synthesis Inhibitors/toxicity , Protein Transport , Ribonucleases/genetics , Ribonucleases/toxicity , Ricin/chemistry , Substrate Specificity , Tryptophan/genetics
17.
Biochimie ; 94(2): 427-33, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21871524

ABSTRACT

Ribotoxins are fungal extracellular ribonucleases that specifically cleave ribosomes leading to cell-death via apoptosis. α-Sarcin is the ribotoxin studied in deepest detail, and therefore constitutes the referential protein for the whole family. It has been demonstrated that ribotoxin activity depends on a very precise structural microenvironment in which electrostatic interactions among residues in the active site are of the highest importance. Hirsutellin A (HtA) has been recently described as the smallest ribotoxin known to date, encompassing all the abilities of previously characterized members of this family into a shorter sequence. Comparison of HtA and α-sarcin three-dimensional structures suggested that residues presumably forming the catalytic triad of HtA would be His 42, Glu 66, and His 113. Within this same idea, the presence of an Asp residue (Asp 40) in a position equivalent to α-sarcin Tyr 48 is highlighted as a novelty in this field. In this work, substitution mutants H42Q, E66Q and H113Q, as well as double and triple mutants in all possible combinations, are studied regarding their ribonucleolytic activity and cytotoxicity. Implication of these three residues in the ribotoxin activity of HtA is confirmed, though none of them is strictly essential for ribosomal cleavage. Studies with mutants D40N and D40N/E66Q demonstrate an important role for Asp 40 in the activity of HtA and establish a new set of electrostatic interactions different from the one described for already known ribotoxins.


Subject(s)
Aspartic Acid/metabolism , Fungal Proteins/genetics , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors/pharmacology , Ribosomes/metabolism , Aspartic Acid/genetics , Catalytic Domain , Cloning, Molecular , Endoribonucleases/chemistry , Endoribonucleases/genetics , Endoribonucleases/pharmacology , Fungal Proteins/chemistry , Fungal Proteins/pharmacology , Models, Molecular , Mutagenesis, Site-Directed , Mutation , Protein Conformation , Protein Synthesis Inhibitors/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Static Electricity
18.
Biochim Biophys Acta ; 1814(10): 1377-82, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21767671

ABSTRACT

Fungal ribotoxins are toxic secreted ribonucleases that cleave a conserved single phosphodiester bond located at the sarcin/ricin loop of the larger rRNA. This cleavage inactivates ribosomes leading to protein biosynthesis inhibition and cell death. It has been proposed that interactions other than those found at the active site of ribotoxins are needed to explain their exquisite specific activity. The study presented shows the ability of a catalytically inactive α-sarcin mutant (H137Q) to bind eukaryotic ribosomes and interfere with in vitro protein biosynthesis. The results obtained are compatible with previous observations that α-sarcin can promote cell death by a mechanism that is independent of rRNA cleavage, expanding the potential set of activities performed by this family of toxins.


Subject(s)
Endoribonucleases/pharmacology , Fungal Proteins/metabolism , Fungal Proteins/pharmacology , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors/pharmacology , Animals , Endoribonucleases/genetics , Endoribonucleases/metabolism , Fungal Proteins/genetics , Models, Biological , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Mutant Proteins/pharmacology , Protein Binding , Protein Synthesis Inhibitors/metabolism , Rabbits , Ribosomes/metabolism , Saccharomyces cerevisiae
19.
Biochim Biophys Acta ; 1808(9): 2275-88, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21621507

ABSTRACT

Actinoporins constitute a group of small and basic α-pore forming toxins produced by sea anemones. They display high sequence identity and appear as multigene families. They show a singular behaviour at the water-membrane interface: In aqueous solution, actinoporins remain stably folded but, upon interaction with lipid bilayers, become integral membrane structures. These membranes contain sphingomyelin, display phase coexistence, or both. The water soluble structures of the actinoporins equinatoxin II (EqtII) and sticholysin II (StnII) are known in detail. The crystalline structure of a fragaceatoxin C (FraC) nonamer has been also determined. The three proteins fold as a ß-sandwich motif flanked by two α-helices, one of them at the N-terminal end. Four regions seem to be especially important: A cluster of aromatic residues, a phosphocholine binding site, an array of basic amino acids, and the N-terminal α-helix. Initial binding of the soluble monomers to the membrane is accomplished by the cluster of aromatic amino acids, the array of basic residues, and the phosphocholine binding site. Then, the N-terminal α-helix detaches from the ß-sandwich, extends, and lies parallel to the membrane. Simultaneously, oligomerization occurs. Finally, the extended N-terminal α-helix penetrates the membrane to build a toroidal pore. This model has been however recently challenged by the cryo-EM reconstruction of FraC bound to phospholipid vesicles. Actinoporins structural fold appears across all eukaryotic kingdoms in other functionally unrelated proteins. Many of these proteins neither bind to lipid membranes nor induce cell lysis. Finally, studies focusing on the therapeutic potential of actinoporins also abound.


Subject(s)
Porins/chemistry , Water/chemistry , Amino Acid Sequence , Animals , Cnidarian Venoms/chemistry , Cnidarian Venoms/metabolism , Cryoelectron Microscopy/methods , Lipid Bilayers/chemistry , Membranes, Artificial , Molecular Conformation , Molecular Sequence Data , Phospholipids/chemistry , Protein Folding , Protein Structure, Secondary , Protein Structure, Tertiary , Sea Anemones , Sequence Homology, Amino Acid
20.
Nucleic Acids Res ; 38(12): 4108-19, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20215430

ABSTRACT

Ribotoxins are potent inhibitors of protein biosynthesis and inactivate ribosomes from a variety of organisms. The ribotoxin alpha-sarcin cleaves the large 23S ribosomal RNA (rRNA) at the universally conserved sarcin-ricin loop (SRL) leading to complete inactivation of the ribosome and cellular death. The SRL interacts with translation factors that hydrolyze GTP, and it is important for their binding to the ribosome, but its precise role is not yet understood. We studied the effect of alpha-sarcin on defined steps of translation by the bacterial ribosome. alpha-Sarcin-treated ribosomes showed no defects in mRNA and tRNA binding, peptide-bond formation and sparsomycin-dependent translocation. Cleavage of SRL slightly affected binding of elongation factor Tu ternary complex (EF-Tu*GTP*tRNA) to the ribosome. In contrast, the activity of elongation factor G (EF-G) was strongly impaired in alpha-sarcin-treated ribosomes. Importantly, cleavage of SRL inhibited EF-G binding, and consequently GTP hydrolysis and mRNA-tRNA translocation. These results suggest that the SRL is more critical in EF-G than ternary complex binding to the ribosome implicating different requirements in this region of the ribosome during protein elongation.


Subject(s)
Endoribonucleases/metabolism , Fungal Proteins/metabolism , Peptide Elongation Factor G/metabolism , Peptide Elongation Factor Tu/metabolism , Protein Synthesis Inhibitors/metabolism , RNA, Ribosomal, 23S/metabolism , Escherichia coli/genetics , Nucleic Acid Conformation , Peptide Elongation Factor G/chemistry , Peptide Elongation Factor Tu/chemistry , Protein Biosynthesis , RNA, Ribosomal, 23S/chemistry , Ribosomes/chemistry , Ribosomes/metabolism , Ricin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...