Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Christos G. Savva is active.

Publication


Featured researches published by Christos G. Savva.


FEBS Journal | 2011

Molecular basis of toxicity of Clostridium perfringens epsilon toxin

Monika Bokori-Brown; Christos G. Savva; Sérgio P. Fernandes da Costa; Claire E. Naylor; Ajit K. Basak; Richard W. Titball

Clostridiumu2003perfringensε‐toxin is produced by toxinotypes B and D strains. The toxin is the aetiological agent of dysentery in newborn lambs but is also associated with enteritis and enterotoxaemia in goats, calves and foals. It is considered to be a potential biowarfare or bioterrorism agent by the US Government Centers for Disease Control and Prevention. The relatively inactive 32.9u2003kDa prototoxin is converted to active mature toxin by proteolytic cleavage, either by digestive proteases of the host, such as trypsin and chymotrypsin, or by C.u2003perfringensλ‐protease. Inu2003vivo, the toxin appears to target the brain and kidneys, but relatively few cell lines are susceptible to the toxin, and most work has been carried out using Madin–Darby canine kidney (MDCK) cells. The binding of ε‐toxin to MDCK cells and rat synaptosomal membranes is associated with the formation of a stable, high molecular weight complex. The crystal structure of ε‐toxin reveals similarity to aerolysin from Aeromonasu2003hydrophila, parasporin‐2 from Bacillusu2003thuringiensis and a lectin from Laetiporusu2003sulphureus. Like these toxins, ε‐toxin appears to form heptameric pores in target cell membranes. The exquisite specificity of the toxin for specific cell types suggests that it binds to a receptor found only on these cells.


The EMBO Journal | 2013

Structure of a bacterial type IV secretion core complex at subnanometre resolution

Angel Rivera-Calzada; Rémi Fronzes; Christos G. Savva; Vidya Chandran; Pei W. Lian; Toon Laeremans; Els Pardon; Jan Steyaert; Han Remaut; Gabriel Waksman; Elena V. Orlova

Type IV secretion (T4S) systems are able to transport DNAs and/or proteins through the membranes of bacteria. They form large multiprotein complexes consisting of 12 proteins termed VirB1‐11 and VirD4. VirB7, 9 and 10 assemble into a 1.07 MegaDalton membrane‐spanning core complex (CC), around which all other components assemble. This complex is made of two parts, the O‐layer inserted in the outer membrane and the I‐layer inserted in the inner membrane. While the structure of the O‐layer has been solved by X‐ray crystallography, there is no detailed structural information on the I‐layer. Using high‐resolution cryo‐electron microscopy and molecular modelling combined with biochemical approaches, we determined the I‐layer structure and located its various components in the electron density. Our results provide new structural insights on the CC, from which the essential features of T4S system mechanisms can be derived.


Journal of Biological Chemistry | 2013

Molecular architecture and functional analysis of NetB, a pore-forming toxin from Clostridium perfringens

Christos G. Savva; P. Fernandes da Costa; Monika Bokori-Brown; Claire E. Naylor; Ambrose R. Cole; David S. Moss; Richard W. Titball; Ajit K. Basak

Background: Clostridium perfringens toxin NetB is a key factor in avian necrotic enteritis. Results: NetB forms heptameric pores structurally similar to Staphylococcus aureus toxins but lacks a phosphocholine binding pocket. NetB activity is enhanced by cholesterol. Conclusion: NetB has distinct binding specificity, and cholesterol may act as a receptor. Significance: The structure of NetB will facilitate development of control measures against necrotic enteritis. NetB is a pore-forming toxin produced by Clostridium perfringens and has been reported to play a major role in the pathogenesis of avian necrotic enteritis, a disease that has emerged due to the removal of antibiotics in animal feedstuffs. Here we present the crystal structure of the pore form of NetB solved to 3.9 Å. The heptameric assembly shares structural homology to the staphylococcal α-hemolysin. However, the rim domain, a region that is thought to interact with the target cell membrane, shows sequence and structural divergence leading to the alteration of a phosphocholine binding pocket found in the staphylococcal toxins. Consistent with the structure we show that NetB does not bind phosphocholine efficiently but instead interacts directly with cholesterol leading to enhanced oligomerization and pore formation. Finally we have identified conserved and non-conserved amino acid positions within the rim loops that significantly affect binding and toxicity of NetB. These findings present new insights into the mode of action of these pore-forming toxins, enabling the design of more effective control measures against necrotic enteritis and providing potential new tools to the field of bionanotechnology.


Nature Communications | 2016

Cryo-EM structure of lysenin pore elucidates membrane insertion by an aerolysin family protein

Monika Bokori-Brown; Thomas G. Martin; Claire E. Naylor; Ajit K. Basak; Richard W. Titball; Christos G. Savva

Lysenin from the coelomic fluid of the earthworm Eisenia fetida belongs to the aerolysin family of small β-pore-forming toxins (β-PFTs), some members of which are pathogenic to humans and animals. Despite efforts, a high-resolution structure of a channel for this family of proteins has been elusive and therefore the mechanism of activation and membrane insertion remains unclear. Here we determine the pore structure of lysenin by single particle cryo-EM, to 3.1u2009Å resolution. The nonameric assembly reveals a long β-barrel channel spanning the length of the complex that, unexpectedly, includes the two pre-insertion strands flanking the hypothetical membrane-insertion loop. Examination of other members of the aerolysin family reveals high structural preservation in this region, indicating that the membrane-insertion pathway in this family is conserved. For some toxins, proteolytic activation and pro-peptide removal will facilitate unfolding of the pre-insertion strands, allowing them to form the β-barrel of the channel.


Vaccine | 2013

Protection against avian necrotic enteritis after immunisation with NetB genetic or formaldehyde toxoids

Sérgio P. Fernandes da Costa; Dorien Mot; Monika Bokori-Brown; Christos G. Savva; Ajit K. Basak; Filip Van Immerseel; Richard W. Titball

Highlights • NetB from Clostridium perfringens is the major virulence factor in avian necrotic enteritis.• Vaccination with a NetB genetic or formaldehyde toxoid protects chicken in an in vivo disease model.• NetB toxoids could form the bases of an efficient vaccine against necrotic enteritis.


Protein Science | 2013

Clostridium perfringens epsilon toxin H149A mutant as a platform for receptor binding studies.

Monika Bokori-Brown; Maria C. Kokkinidou; Christos G. Savva; Sérgio P. Fernandes da Costa; Claire E. Naylor; Ambrose R. Cole; David S. Moss; Ajit K. Basak; Richard W. Titball

Clostridium perfringens epsilon toxin (Etx) is a pore‐forming toxin responsible for a severe and rapidly fatal enterotoxemia of ruminants. The toxin is classified as a category B bioterrorism agent by the U.S. Government Centres for Disease Control and Prevention (CDC), making work with recombinant toxin difficult. To reduce the hazard posed by work with recombinant Etx, we have used a variant of Etx that contains a H149A mutation (Etx‐H149A), previously reported to have reduced, but not abolished, toxicity. The three‐dimensional structure of H149A prototoxin shows that the H149A mutation in domain III does not affect organisation of the putative receptor binding loops in domain I of the toxin. Surface exposed tyrosine residues in domain I of Etx‐H149A (Y16, Y20, Y29, Y30, Y36 and Y196) were mutated to alanine and mutants Y30A and Y196A showed significantly reduced binding to MDCK.2 cells relative to Etx‐H149A that correlated with their reduced cytotoxic activity. Thus, our study confirms the role of surface exposed tyrosine residues in domain I of Etx in binding to MDCK cells and the suitability of Etx‐H149A for further receptor binding studies. In contrast, binding of all of the tyrosine mutants to ACHN cells was similar to that of Etx‐H149A, suggesting that Etx can recognise different cell surface receptors. In support of this, the crystal structure of Etx‐H149A identified a glycan (β‐octyl‐glucoside) binding site in domain III of Etx‐H149A, which may be a second receptor binding site. These findings have important implications for developing strategies designed to neutralise toxin activity.


Vaccine | 2014

Clostridium perfringens epsilon toxin mutant Y30A-Y196A as a recombinant vaccine candidate against enterotoxemia

Monika Bokori-Brown; Charlotte A. Hall; Charlotte Vance; Sérgio P. Fernandes da Costa; Christos G. Savva; Claire E. Naylor; Ambrose R. Cole; Ajit K. Basak; David S. Moss; Richard W. Titball

Highlights • Etx mutant Y30A-Y196A showed markedly reduced cytotoxicity towards MDCK.2 cells.• Y30A-Y196A is inactive in mice after intraperitoneal administration.• Y30A-Y196A is able to induce a specific antibody response in rabbits.• Y30A-Y196A polyclonal antibody is able to induce protective immunity in vitro.• Y30A-Y196A could form the basis of a recombinant vaccine against enterotoxemia.


Journal of Molecular Biology | 2014

Structure of a C. perfringens Enterotoxin mutant in complex with a Modified Claudin-2 Extracellular Loop 2

Tamas S. Yelland; Claire E. Naylor; Tannya Bagoban; Christos G. Savva; David S. Moss; Bruce A. McClane; Ingolf E. Blasig; Michel R. Popoff; Ajit K. Basak

CPE (Clostridium perfringens enterotoxin) is the major virulence determinant for C. perfringens type-A food poisoning, the second most common bacterial food-borne illness in the UK and USA. After binding to its receptors, which include particular human claudins, the toxin forms pores in the cell membrane. The mature pore apparently contains a hexamer of CPE, claudin and, possibly, occludin. The combination of high binding specificity with cytotoxicity has resulted in CPE being investigated, with some success, as a targeted cytotoxic agent for oncotherapy. In this paper, we present the X-ray crystallographic structure of CPE in complex with a peptide derived from extracellular loop 2 of a modified, CPE-binding Claudin-2, together with high-resolution native and pore-formation mutant structures. Our structure provides the first atomic-resolution data on any part of a claudin molecule and reveals that claudins CPE-binding fingerprint (NPLVP) is in a tight turn conformation and binds, as expected, in CPEs C-terminal claudin-binding groove. The leucine and valine residues insert into the binding groove while the first residue, asparagine, tethers the peptide via an interaction with CPEs aspartate 225 and the two prolines are required to maintain the tight turn conformation. Understanding the structural basis of the contribution these residues make to binding will aid in engineering CPE to target tumor cells.


PLOS ONE | 2013

Structural Insights into Clostridium perfringens Delta Toxin Pore Formation

Jessica Huyet; Claire E. Naylor; Christos G. Savva; Michel R. Popoff; Ajit K. Basak

Clostridium perfringens Delta toxin is one of the three hemolysin-like proteins produced by C. perfringens type C and possibly type B strains. One of the others, NetB, has been shown to be the major cause of Avian Nectrotic Enteritis, which following the reduction in use of antibiotics as growth promoters, has become an emerging disease of industrial poultry. Delta toxin itself is cytotoxic to the wide range of human and animal macrophages and platelets that present GM2 ganglioside on their membranes. It has sequence similarity with Staphylococcus aureus β-pore forming toxins and is expected to heptamerize and form pores in the lipid bilayer of host cell membranes. Nevertheless, its exact mode of action remains undetermined. Here we report the 2.4 Å crystal structure of monomeric Delta toxin. The superposition of this structure with the structure of the phospholipid-bound F component of S. aureus leucocidin (LukF) revealed that the glycerol molecules bound to Delta toxin and the phospholipids in LukF are accommodated in the same hydrophobic clefts, corresponding to where the toxin is expected to latch onto the membrane, though the binding sites show significant differences. From structure-based sequence alignment with the known structure of staphylococcal α-hemolysin, a model of the Delta toxin pore form has been built. Using electron microscopy, we have validated our model and characterized the Delta toxin pore on liposomes. These results highlight both similarities and differences in the mechanism of Delta toxin (and by extension NetB) cytotoxicity from that of the staphylococcal pore-forming toxins.


Toxins | 2014

Identification of a Key Residue for Oligomerisation and Pore-Formation of Clostridium perfringens NetB

Sérgio P. Fernandes da Costa; Christos G. Savva; Monika Bokori-Brown; Claire E. Naylor; David S. Moss; Ajit K. Basak; Richard W. Titball

Necrotic enteritis toxin B (NetB) is a β-pore-forming toxin produced by Clostridium perfringens and has been identified as a key virulence factor in the pathogenesis of avian necrotic enteritis, a disease causing significant economic damage to the poultry industry worldwide. In this study, site-directed mutagenesis was used to identify amino acids that play a role in NetB oligomerisation and pore-formation. NetB K41H showed significantly reduced toxicity towards LMH cells and human red blood cells relative to wild type toxin. NetB K41H was unable to oligomerise and form pores in liposomes. These findings suggest that NetB K41H could be developed as a genetic toxoid vaccine to protect against necrotic enteritis.

Collaboration


Dive into the Christos G. Savva's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge