Network


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

Hotspot


Dive into the research topics where Eleni Douni is active.

Publication


Featured researches published by Eleni Douni.


Human Molecular Genetics | 2012

A RANKL G278R mutation causing osteopetrosis identifies a functional amino acid essential for trimer assembly in RANKL and TNF

Eleni Douni; Vagelis Rinotas; Eleni Makrinou; Jochen Zwerina; Josef M. Penninger; Elias Eliopoulos; Georg Schett; George Kollias

Receptor activator of nuclear factor-κB ligand (RANKL), a trimeric tumor necrosis factor (TNF) superfamily member, is the central mediator of osteoclast formation and bone resorption. Functional mutations in RANKL lead to human autosomal recessive osteopetrosis (ARO), whereas RANKL overexpression has been implicated in the pathogenesis of bone degenerative diseases such as osteoporosis. Following a forward genetics approach using N-ethyl-N-nitrosourea (ENU)-mediated random mutagenesis, we generated a novel mouse model of ARO caused by a new loss-of-function allele of Rankl with a glycine-to-arginine mutation at codon 278 (G278R) at the extracellular inner hydrophobic F β-strand of RANKL. Mutant mice develop severe osteopetrosis similar to Rankl-deficient mice, whereas exogenous administration of recombinant RANKL restores osteoclast formation in vivo. We show that RANKL(G278R) monomers fail to assemble into homotrimers, are unable to bind and activate the RANK receptor and interact with wild-type RANKL exerting a dominant-negative effect on its trimerization and function in vitro. Since G278 is highly conserved within the TNF superfamily, we identified that a similar substitution in TNF, G122R, also abrogated trimerization, binding to TNF receptor and consequently impaired TNF biological activity. Notably, SPD304, a potent small-molecule inhibitor of TNF trimerization that interacts with G122, also inhibited RANKL activity, suggesting analogous inhibitory mechanisms. Our results provide a new disease model for ARO and identify a functional amino acid in the TNF-like core domain essential for trimer formation both in RANKL and in TNF that could be considered a novel potential target for inhibiting their biological activities.


Journal of Bone and Mineral Research | 2014

Novel Genetic Models of Osteoporosis by Overexpression of Human RANKL in Transgenic Mice

Vagelis Rinotas; Alexandra Niti; Romain Dacquin; Nicolas Bonnet; Marina Stolina; Chun-Ya Han; Paul J. Kostenuik; Pierre Jurdic; Serge Livio Ferrari; Eleni Douni

Receptor activator of NF‐κB ligand (RANKL) plays a key role in osteoclast‐induced bone resorption across a range of degenerative bone diseases, and its specific inhibition has been recently approved as a treatment for women with postmenopausal osteoporosis at high or increased risk of fracture in the United States and globally. In the present study, we generated transgenic mice (TghuRANKL) carrying the human RANKL (huRANKL) genomic region and achieved a physiologically relevant pattern of RANKL overexpression in order to establish novel genetic models for assessing skeletal and extraskeletal pathologies associated with excessive RANKL and for testing clinical therapeutic candidates that inhibit human RANKL. TghuRANKL mice of both sexes developed early‐onset bone loss, and the levels of huRANKL expression were correlated with bone resorption and disease severity. Low copy Tg5516 mice expressing huRANKL at low levels displayed a mild osteoporotic phenotype as shown by trabecular bone loss and reduced biomechanical properties. Notably, overexpression of huRANKL, in the medium copy Tg5519 line, resulted in severe early‐onset osteoporosis characterized by lack of trabecular bone, destruction of the growth plate, increased osteoclastogenesis, bone marrow adiposity, increased bone remodeling, and severe cortical bone porosity accompanied by decreased bone strength. An even more severe skeletal phenotype developed in the high copy Tg5520 founder with extensive soft tissue calcification. Model validation was further established by evidence that denosumab, an antibody that inhibits human but not murine RANKL, fully corrected the hyper‐resorptive and osteoporotic phenotypes of Tg5519 mice. Furthermore, overexpression of huRANKL rescued osteopetrotic phenotypes of RANKL‐defective mice. These novel huRANKL transgenic models of osteoporosis represent an important advance for understanding the pathogenesis and treatment of high‐turnover bone diseases and other disease states caused by excessive RANKL.


PLOS ONE | 2014

A Splicing Mutation in the Novel Mitochondrial Protein DNAJC11 Causes Motor Neuron Pathology Associated with Cristae Disorganization, and Lymphoid Abnormalities in Mice

Fotis Ioakeimidis; Christine Ott; Vera Kozjak-Pavlovic; Foteini Violitzi; Vagelis Rinotas; Eleni Makrinou; Elias Eliopoulos; Costas Fasseas; George Kollias; Eleni Douni

Mitochondrial structure and function is emerging as a major contributor to neuromuscular disease, highlighting the need for the complete elucidation of the underlying molecular and pathophysiological mechanisms. Following a forward genetics approach with N-ethyl-N-nitrosourea (ENU)-mediated random mutagenesis, we identified a novel mouse model of autosomal recessive neuromuscular disease caused by a splice-site hypomorphic mutation in a novel gene of unknown function, DnaJC11. Recent findings have demonstrated that DNAJC11 protein co-immunoprecipitates with proteins of the mitochondrial contact site (MICOS) complex involved in the formation of mitochondrial cristae and cristae junctions. Homozygous mutant mice developed locomotion defects, muscle weakness, spasticity, limb tremor, leucopenia, thymic and splenic hypoplasia, general wasting and early lethality. Neuropathological analysis showed severe vacuolation of the motor neurons in the spinal cord, originating from dilatations of the endoplasmic reticulum and notably from mitochondria that had lost their proper inner membrane organization. The causal role of the identified mutation in DnaJC11 was verified in rescue experiments by overexpressing the human ortholog. The full length 63 kDa isoform of human DNAJC11 was shown to localize in the periphery of the mitochondrial outer membrane whereas putative additional isoforms displayed differential submitochondrial localization. Moreover, we showed that DNAJC11 is assembled in a high molecular weight complex, similarly to mitofilin and that downregulation of mitofilin or SAM50 affected the levels of DNAJC11 in HeLa cells. Our findings provide the first mouse mutant for a putative MICOS protein and establish a link between DNAJC11 and neuromuscular diseases.


ACS Medicinal Chemistry Letters | 2013

Solvent Selection for Insoluble Ligands, a Challenge for Biological Assay Development: A TNF-α/SPD304 Study

Christos P. Papaneophytou; Anthi Mettou; Vagelis Rinotas; Eleni Douni; George Kontopidis

Many active compounds may be excluded from biological assays due to their low aqueous solubility. In this study, a simple method for the determination of the solubility of compounds containing aromatic rings is proposed. In addition to DMSO, five organic solvents for screening experiments of TNF-α inhibitors were explored. DMSO and PEG3350 were the most suitable for both protein stability and ligand-binding experiments. In addition, glycerol is a promising solvent for the screening of other compounds for which it might provide acceptable solubilization, due to its strong tendency to preserve the protein. Moreover, a fluorescence binding assay was developed using the TNF-α/SPD304 system, and a K d of 5.36 ± 0.21 μM was determined. The results of this study could be used for the future screening of potential TNF-α inhibitors, while the protocols developed in this work could be applied to other proteins.


PLOS Computational Biology | 2017

Cheminformatics-aided discovery of small-molecule Protein-Protein Interaction (PPI) dual inhibitors of Tumor Necrosis Factor (TNF) and Receptor Activator of NF-κB Ligand (RANKL)

Georgia Melagraki; Evangelos Ntougkos; Vagelis Rinotas; Christos Papaneophytou; Georgios Leonis; Thomas Mavromoustakos; George Kontopidis; Eleni Douni; Antreas Afantitis; George Kollias

We present an in silico drug discovery pipeline developed and applied for the identification and virtual screening of small-molecule Protein-Protein Interaction (PPI) compounds that act as dual inhibitors of TNF and RANKL through the trimerization interface. The cheminformatics part of the pipeline was developed by combining structure–based with ligand–based modeling using the largest available set of known TNF inhibitors in the literature (2481 small molecules). To facilitate virtual screening, the consensus predictive model was made freely available at: http://enalos.insilicotox.com/TNFPubChem/. We thus generated a priority list of nine small molecules as candidates for direct TNF function inhibition. In vitro evaluation of these compounds led to the selection of two small molecules that act as potent direct inhibitors of TNF function, with IC50 values comparable to those of a previously-described direct inhibitor (SPD304), but with significantly reduced toxicity. These molecules were also identified as RANKL inhibitors and validated in vitro with respect to this second functionality. Direct binding of the two compounds was confirmed both for TNF and RANKL, as well as their ability to inhibit the biologically-active trimer forms. Molecular dynamics calculations were also carried out for the two small molecules in each protein to offer additional insight into the interactions that govern TNF and RANKL complex formation. To our knowledge, these compounds, namely T8 and T23, constitute the second and third published examples of dual small-molecule direct function inhibitors of TNF and RANKL, and could serve as lead compounds for the development of novel treatments for inflammatory and autoimmune diseases.


Protein Expression and Purification | 2013

A statistical approach for optimization of RANKL overexpression in Escherichia coli: Purification and characterization of the protein

Christos P. Papaneophytou; Vagelis Rinotas; Eleni Douni; George Kontopidis

Receptor activator of nuclear factor-κB (RANK) and its cognate ligand (RANKL) is a member of the TNF superfamily of cytokines which is essential in osteobiology and its overexpression has been implicated in the pathogenesis of bone degenerative diseases such as osteoporosis. Therefore, RANKL is considered a major therapeutic target for the suppression of bone resorption in bone metabolic diseases such as rheumatoid arthritis and cancer metastasis. To evaluate the inhibitory effect of potential RANKL inhibitors a sufficient amount of protein is required. In this work RANKL was cloned for expression at high levels in Escherichia coli with the interaction of changing cultures conditions in order to produce the protein in a soluble form. In an initial step, the effect of expression host on soluble protein production was investigated and BL21(DE3) pLysS was the most efficient one found for the production of RANKL. Central composite design experiment in the following revealed that cell density before induction, IPTG concentration, post-induction temperature and time as well as their interactions had a significant influence on soluble RANKL production. An 80% increase of protein production was achieved after the determination of the optimum induction conditions: OD600nm before induction 0.55, an IPTG concentration of 0.3mM, a post-induction temperature of 25°C and a post-induction time of 6.5h. Following RANKL purification the thermal stability of the protein was studied. The interaction of RANKL with SPD304, a patented small-molecule inhibitor of TNF-α, was also studied in a fluorescence binding assay resulting in a Kd value of 14.1 ± 0.5 μM.


Laboratory Animals | 2013

FELASA guidelines for the refinement of methods for genotyping genetically-modified rodents: A report of the Federation of European Laboratory Animal Science Associations Working Group

Dolores Bonaparte; Paolo Cinelli; Eleni Douni; Yann Herault; Alex Maas; Pirjo Pakarinen; Matti Poutanen; Mirentxu Santos Lafuente; Ferdinando Scavizzi

The use of genetically-modified (GM) animals as research models continues to grow. The completion of the mouse genome sequence, together with the high-throughput international effort to introduce mutations across the mouse genome in the embryonic stem (ES) cells (www.knockoutmouse.org) facilitates an efficient way to obtain mutated mouse strains as research models. The increasing number of available mutated mouse strains and their combinations, together with the increasing complexity in the targeting approaches used, reinforces the need for guidelines that will provide information about the mouse strains and the robust and reliable methods used for their genotyping. This information, however, should be obtained with a method causing minimal discomfort to the experimental animals. We have, therefore, compiled the present document which summarizes the currently available methods for obtaining genotype information. It provides updated guidelines concerning animal identification, DNA sampling and genotyping, and the information to be kept and distributed for any mutated rodent strain.


Bioorganic & Medicinal Chemistry | 2014

2,2'-Dihydroxybenzophenones and their carbonyl N-analogues as inhibitor scaffolds for MDR-involved human glutathione transferase isoenzyme A1-1.

Fereniki Perperopoulou; Petros G. Tsoungas; Trias Thireou; Vagelis Rinotas; Eleni Douni; Elias Eliopoulos; Nikolaos E. Labrou; Yannis D. Clonis

The MDR-involved human GSTA1-1, an important isoenzyme overexpressed in several tumors leading to chemotherapeutic-resistant tumour cells, has been targeted by 2,2-dihydroxybenzophenones and some of their carbonyl N-analogues, as its potential inhibitors. A structure-based library of the latter was built-up by a nucleophilic cleavage of suitably substituted xanthones to 2,2-dihydroxy-benzophenones (5-9) and subsequent formation of their N-derivatives (oximes 11-13 and N-acyl hydrazones 14-16). Screening against hGSTA1-1 led to benzophenones 6 and 8, and hydrazones 14 and 16, having the highest inhibition potency (IC₅₀ values in the range 0.18 ± 0.02 to 1.77 ± 0.10 μM). Enzyme inhibition kinetics, molecular modeling and docking studies showed that they interact primarily at the CDNB-binding catalytic site of the enzyme. In addition, the results from cytotoxicity studies with human colon adenocarcinoma cells showed low LC₅₀ values for benzophenone 6 and its N-acyl hydrazone analogue 14 (31.4 ± 0.4 μM and 87 ± 1.9 μM, respectively), in addition to the strong enzyme inhibition profile (IC₅₀(6)=1,77 ± 0.10 μM; IC₅₀(14)=0.33 ± 0.05 μM). These structures may serve as leads for the design of new potent mono- and bi-functional inhibitors and pro-drugs against human GTSs.


Archiv Der Pharmazie | 2014

Rationally Designed Less Toxic SPD‐304 Analogs and Preliminary Evaluation of Their TNF Inhibitory Effects

Polyxeni Alexiou; Athanasios Papakyriakou; Evangelos Ntougkos; Christos P. Papaneophytou; Fotini Liepouri; Anthi Mettou; Ioannis A. Katsoulis; Anna Maranti; Katerina Tsiliouka; Alexandros Strongilos; Sotiria Chaitidou; Eleni Douni; George Kontopidis; George Kollias; Elias A. Couladouros; Elias Eliopoulos

SPD‐304 was discovered as a promising tumor necrosis factor alpha (TNF) antagonist that promotes dissociation of TNF trimers and therefore blocks the interaction of TNF and its receptor. However, SPD‐304 contains a potentially toxic 3‐alkylindole moiety, which can be bioactivated to a reactive electrophilic intermediate. A series of SPD‐304 analogs was synthesized with the aim to diminish its toxicophore groups while maintaining the binding affinity for TNF. Incorporation of electron‐withdrawing substituents at the indole moiety, in conjunction with elimination of the 6′‐methyl group of the 4‐chromone moiety, led to a significantly less toxic and equally potent TNF inhibitor.


Cytokine & Growth Factor Reviews | 2015

The unbearable lightness of bone marrow homeostasis

Dimitrios Agas; Luigi Marchetti; Eleni Douni; Maria Giovanna Sabbieti

The anatomical and functional dimensions of bone marrow topography have been at the forefront of modern bone and immunological research for many years and remain a source of complexity and perplexity due to the multitude of microhabitats within this microenvironment. In fact, research has uncovered fascinating functional aspects of bone marrow residents, and the bone marrow niche has been identified as the foremost reservoir of a variety of cells including hematopoietic, skeletal and endothelial stem/progenitor cells. The physical interactions of the marrow residents, combined with the release of cytokines and growth factors, organize well-defined operative compartments, which preserve bone and immune cell homeostasis. In a simplistic view, both the hematopoietic and bone marrow stromal (mesenchymal) stem/progenitor cell populations dwell at the interface between the endosteum and the bone marrow area (endosteal niche) and in the perivascular space (vascular niche). Indeed, the tantalizing hypothesis of bone marrow regulatory dependency on these niches is supported by current research insofar as the increase in the number of osteoblasts results in a concomitant increase in the hematopoietic population, indicating that the osteoblasts and the endosteal niche are key components of HSC maintenance. On the other hand, impaired function of the vascular niche compromises the endosteal niches ability to support hematopoiesis. These fascinating discoveries indicate that there are strong ties between bone marrow inhabitants within the confines of the bone marrow itself. When these ties fail, niche-niche communication suffers and results in reduced bone formation, enfeebled hematopoiesis and unrestrained HSC migration through blood circulation. This study focused on the extraordinary homeostatic equilibrium and function of both bone and immune cells within the spatially defined microenvironment of bone marrow. But how important is the anatomically outlined scenery in which the bone marrow entity supports and hosts the hematopoietic elements?

Collaboration


Dive into the Eleni Douni's collaboration.

Top Co-Authors

Avatar

Vagelis Rinotas

Agricultural University of Athens

View shared research outputs
Top Co-Authors

Avatar

Elias Eliopoulos

Agricultural University of Athens

View shared research outputs
Top Co-Authors

Avatar

George Kollias

Alexander Fleming Biomedical Sciences Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alexandros Strongilos

Agricultural University of Athens

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Elias A. Couladouros

Agricultural University of Athens

View shared research outputs
Top Co-Authors

Avatar

Fotis Ioakeimidis

Agricultural University of Athens

View shared research outputs
Researchain Logo
Decentralizing Knowledge