Vittorio, Serena

University of Messina (Italy)

Serena Vittorio graduated in Medicinal Chemistry and Technology at University of Messina in March 2016. In the same year she received a post-graduate fellowship “Fondazione Prof. Antonio Imbesi” working in part at University of Messina under the supervision of Professor Laura De Luca and for nine months in the research group of Professor Thierry Langer at University of Vienna. In 2018 she started her doctoral studies at University of Messina spending six-months as visiting PhD student at the University of Vienna. Her research is focused on the application of computational approaches for the design of new therapeutic agents.


Abstract:

In silico modeling of small molecules as α-synuclein aggregation inhibitors

Topic:

The search for a cure of Parkinson’s disease (PD) represents a challenging task in the pharmaceutical research field. To date, the
available therapies are addressed to restore dopamine levels thus reducing the motor symptoms related to PD, such as rest
tremor, bradykinesia and muscular rigidity. Recently, the inhibition of α-synuclein (α-syn) aggregation has emerged as promising
strategy to slow or halt the neurodegenerative process. The α-syn is a 140 aa presynaptic protein implicated in the regulation of
neurotransmitter release from the synaptic vesicles. In PD α-syn aggregates into toxic oligomers and fibrils forming Lewi bodies
that represent the hallmark of this neurological disorder. In order to identify new small molecules as α-syn aggregation
inhibitors, we generated a ligand-based pharmacophore model to be used as filter to virtually screen two distinct chemical
libraries: i) our in-house database CHIME 2.0 and ii) the MyriaScreen Diversity Library II. By means of this virtual screening we
selected small molecules that were tested in vitro thus leading to the identification of the 3-(cinnamylsulfanyl)-5-(4-pyridinyl)-
1,2,4-triazol-4-amine as promising hit compound for the development of new α-syn aggregation inhibitors. Therefore, few
structural modifications were carried out thus obtaining a new series of small molecules that were synthesized and tested in
order to investigate the biological profile. Finally, the binding mode of these new inhibitors was elucidated by molecular docking
studies.


Back to speaker overview Back to Oral- and Flash presentations overview

Miller, Gavin

School of Chemical and Physical Sciences Manchester (UK)

Gavin read Chemistry at UMIST and was awarded an MChem in Medicinal Chemistry.


Abstract:

Chemoenzymatic synthesis of sugar nucleotide chemical biology tools to explore the GDP-D-mannose dehydrogenase from Pseudomonas aeruginosa

Topic:

The opportunistic human pathogen Pseudomonas aeruginosa (PA) causes chronic bacterial infections in cystic fibrosis patients, contributing to a reduction in lung function and increased mortality rates. The lung environment induces a switch of P. aeruginosa to its mucoid phenotype, which is characterised by an overproduction of the exopolysaccharide alginate. Composed of β-D-mannuronic acid and its C5 epimer α-L-guluronic acid, alginate is a key component in the formation of a bacterial biofilm, which increases persistence of the bacteria in the airways and retards antimicrobial treatments. Inspection of the PA biosynthetic pathway reveals a key enzyme involved in alginate production, GDP-mannose dehydrogenase (GMD), which catalyses an NAD+-dependent oxidation of GDP-D-Man to GDP-D-ManA: the alginate feedstock monosaccharide. We have designed and synthesised a series of GDP-Man probes to interact with the GMD active site, providing mechanistic insight and identifying the first sugar nucleotide inhibitor of GMD.


Back to speaker overview Back to Oral- and Flash presentations overview

Rubio-Sánchez, Roger

University of Cambridge (UK)

Abstract:

A Modular, Dynamic, DNA-based Platform for Regulating Cargo Distribution and Transport between Lipid Domains

Topic:

Biological membranes feature highly evolved proteo-lipid machinery able to co-localise in lipid rafts, nano-scaled assemblies believed to underpin signal transduction [1], amongst other cellular processes. Bottom-up synthetic biology aims to replicate life-like behaviours in model artificial cells [2], often using synthetic lipid bilayers as passive enclosures that lack the functional complexity associated to their biological analogues. DNA nanotechnology has emerged as a popular choice for biomimicry, coupling bio-inspired nano-devices with model membranes using amphiphilic oligonucleotides [3]. In fact, amphiphilic DNA nanostructures also undergo partitioning in lipid domains [4], evoking the affinity of proteins for raft microenvironments.

Here, we regulate the lateral distribution of DNA nanostructures in phase-separated membranes by exploiting the tendency of cholesterol and tocopherol motifs to respectively enrich liquid-ordered (Lo) and liquid-disordered (Ld) domains. By prescribing combinations of multiple anchors, changes to nanostructure topology, and size, our DNA architectures are programmed to achieve partitioning states that span the energy landscape. In addition, the functionality of our approach is showcased with a responsive biomimetic DNA device that dynamically achieves ligand-induced reconfiguration and mediates cargo transport between lipid domains. Our synergistic platform [5] paves the way for the development of next-generation biomimetic DNA-based architectures, that can achieve sensing and communication in synthetic cellular systems

 


Back to speaker overview Back to Oral- and Flash presentations overview

Nakashima, Karina

Radboud University ( The Netherlands)

Liquid-liquid phase separation plays an important role in cellular organization, and it is an emerging alternative in compartment-first hypotheses for the origin of life. Control over phase separation by enzymatic reactions is essential in order to use droplets as organelle mimics or protocells. To elucidate the physicochemical principles that govern the nucleation, growth and coarsening of biomimetic droplets, we use ATP-based complex coacervate droplets that we control by a kinase reaction. We track the coacervates by microscopy and follow their active growth at a single-droplet level. We quantify the partitioning of all components in our system by HPLC and fluorescence labelling to support our results with a kinetic comparison. We show that droplet size increases as a result of the chemical reaction, an active behavior that is a plausible mechanism for protocellular growth. Moreover, growth rate can be averaged over the entire droplet population, and is significantly affected by environmental conditions and droplet composition. We also find that Ostwald ripening is suppressed in complex coacervates, and therefore these compartments, although membraneless, are more stable than it is usually speculated in the literature. Our findings show that the behavior of active droplets, obtained through coupling phase separation to enzymatic reactions, can be quantified and explained in terms of chemical principles.


Abstract:

Active coacervate droplets: protocells that grow and survive

Topic:

I obtained my BSc (2012) and MSc (2015) Chemistry degrees at the University of São Paulo. I came to the Netherlands in 2017 for my PhD to investigate how chemical reactions can control coacervate droplets and vice-versa, as a small step in understanding cells and the chemical origin of life


Back to speaker overview Back to Oral- and Flash presentations overview

Morretta, Elva

University of Salerno (Italy)

Elva Morretta (EM) obtained a Master’s degree in Pharmaceutical Chemistry and Technology (110/110 cum laude) in July 2016 at Salerno University (bio-organic chemistry thesis, tutor Prof. Monti). In September, EM obtained a fellowship for the Drug Discovery and Development Ph.D. program (tutor Prof. Casapullo). In 2018, EM spent six months at Prof. Heck Biomolecular Mass Spectrometry and Proteomics Lab. On March 2020, EM defended her Ph.D., discussing a thesis entitled “Interactome Analysis of Bioactive Molecules: Optimization of a Functional Proteomics Platform”. From January 16th 2020, EM works as a Post-Doctoral Researcher at the Pharmacy Department of Salerno University.


Abstract:

Label-free functional proteomics links the anti-angiogenic properties of the pyrazolyl-urea GeGe-3 to Calreticulin binding

Topic:

n the last twenty years, 5-pyrazolyl-ureas have been largely investigated for their poly-pharmacological potential. In this scenario, ethyl 1-(2-hydroxypentyl)-5-(3-(3-(trifluoromethyl) phenyl)ureido)-1H-pyrazole-4-carboxylate (GeGe-3) emerged as a promising anti-angiogenic compound, inhibiting Human Umbilical Vein Endothelial Cells (HUVEC cells) proliferation and endothelial tube formation, impairing inter-segmental angiogenesis during zebrafish embryos development and blocking tumour growth in transplanted subcutaneous Lewis Lung Carcinomas. Regrettably, although different primary targets implicated in cell division and/or calcium homeostasis have been hypothesized for this compound, all the binding tests gave negative results. Thus, to link GeGe-3 anti-angiogenic potential to a suitable protein partner, the molecule interactome has been deeply investigated in HUVEC cells through label-free functional proteomics approaches, namely Drug Affinity Responsive Target Stability (DARTS) and targeted Limited Proteolysis coupled to Multiple Reaction Monitoring Mass Spectrometry (t-LiPMRM). These approaches share the principle that, interacting with a molecule, a protein undergoes conformational changes that result in its lower sensitivity to limited proteolysis, when performed in native conditions. Thus, in a first step, the coupling of DARTS with high resolution mass spectrometry allowed the identification of GeGe-3 most reliable interacting protein, Calreticulin, as later on validated by Western Blotting. Subsequently t-LiP-MRM, which allows to discover the target protein structural alterations due to complex formation with GeGe-3, served the purpose of pinpointing Calreticulin regions directly or distally involved in the interaction with the compound. T-LiP-MRM obtained results were corroborated by molecular docking analyses. Calreticulin is a major Ca2+ binding protein involved in intracellular Ca2+ homeostasis, cells adhesion, migration, proliferation, differentiation and apoptosis, as well as in cell-cell interactions. To shed light on the biological consequences of GeGe-3 interaction with such an interesting protein partner, in cell assays were performed. The obtained results disclosed GeGe-3 potential mechanism of action as anti-angiogenic factor: due to its binding to calreticulin, the molecule is able to alter Ca2+ intracellular shift in HUVEC cells, consequently modifying their cytoskeletal proteins organization.


Back to speaker overview Back to Oral- and Flash presentations overview

Roth, Lukas

University of Sydney (Australia)

Abstract:

Immobilized Metal Affinity Chromatography as a Potential Drug Discovery Platform

Topic:

In 1990, approximately 80% of medicines approved in the U.S. were either natural products or their derivatives. In the early 2000s there was a significant drop in the number of natural products in clinical studies, coinciding with the expansion of high throughput screening (HTS) techniques. However, the limited structural diversity inherent to HTS and the emerging threat of antimicrobial resistance has reinvigorated the focus on exploiting natural products for the discovery and development of new medicines. Immobilized metal affinity chromatography (IMAC), a technique originally designed for the isolation of histidinetagged proteins, has shown promise in isolating and purifying bioactive compounds. IMAC relies on the fundamentals of coordination chemistry to reversibly retain compounds with known metal ion affinity. Although originally developed for recombinant protein purification, this simple method has been shown to readily purify hydroxamic acid siderophores, such as desferrioxamine B (DFOB) and other clinical agents, from bacterial cultures. Most IMAC work, in the context of siderophore isolation and purification, has utilized Ni(II) as the metal ion, but there is potential in substituting Ni(II) with other metal ions, such as Cu(II), Fe(III), Ga(III) and Zn(II). The modified IMAC resin beds may consequently act as metalloenzyme surrogates and select for different metabolites as directed by distinct coordination chemistries. This could open up a new platform to discover metalloenzyme inhibitor drug candidates as the isolated metabolites, by virtue of their metal binding affinity, may demonstrate activity against various metalloenzymes. As an initial proof of concept, we have exposed a mixture of in use metalloenzyme inhibitors to IMAC columns charged with various biologically relevant metals with promising results. The IMAC ligand-metal complex is a reasonable surrogate of the active site of a metalloenzyme and the method is capable of reversibly binding a variety of antihypertensive, anti-inflammatory and anticancer drugs.


Back to speaker overview Back to Oral- and Flash presentations overview

Walczewska-Szewc, Katarzyna

Nicolaus Copernicus University (Poland)

Katarzyna Walczewska-Szewc is an assistant professor in the Theoretical Biophysics group, Nicolaus Copernicus University in Toruń, Poland. In her research she uses a number of computer-aided methods to model biomolecular interactions and to reveal the structural mechanisms of action of medically important proteins. She obtained her PhD degree in biology and physics jointly from the Australian National University and University of Gdansk, Poland. Katarzyna’s PhD project involved the development and use of numerical and simulation methods to design and interpret Förster resonance energy transfer (FRET) experiments in physics and biology


Abstract:

The changes in prolyl oligopeptidase structure upon inhibition modify its ability to decrease alpha-synuclein aggregation

Topic:

The formation of extended misfolded protein aggregates is one of the main reasons for neuronal malfunction and, eventually, brain damage in many neurodegenerative diseases. In Parkinson’s disease alpha-synucleins are implicated in the accumulation of the aggregates. The origin of such aggregation is not yet known, however, there is a compelling evidence that it can be reduced by inhibition of prolyl oligopeptidase (PREP). This effect cannot be simply related to the inhibition of the catalytic function of the enzyme, as not all PREP inhibitors stop the alpha-synuclein aggregation. Finding differences in the dynamics of the enzyme inhibited with diverse compounds would allow us to pinpoint the regions of the protein involved in the interaction between PREP and alpha-synuclein. Here, we study the action of three PREP inhibitors, each of which affects alphasynuclein aggregation to different extent. Using molecular dynamics modelling, we determine molecular mechanisms underlying the PREP inhibition and identify structural differences in each inhibitor-PREP system. We suggest that even subtle differences in the dynamics of the enzyme affect its interactions with alpha-synucleins. Thus, identification of these regions may be crucial in preventing formation of alpha-synuclein aggregates. Acknowledgement: The computational results were obtained using the facilities of the Interdisciplinary Centre for Modern Technologies, NCU, Poland


Back to speaker overview Back to Oral- and Flash presentations overview

Arrabito*, Giuseppe

University of Palermo (Italy)

Topic:

Printing Biology: where printing meets synthetic biology

Abstract:

The assembly of life-like artificial systems is an emerging topic that contributes to the fundamental understanding of the molecular origins of life, and fuels the development of life-inspired platforms usable in different fields (e.g. molecular sensing, artificial biology, tissue engineering) [1]. The implementation of these platforms depends upon the ability to recapitulate the structural and functional features of biological systems, including multi-scale organization, adaptivity to environmental stimuli, collective behaviors [2]. The resulting research efforts have led to the recent definition of Printing Biology [3], a field resulting from the intersection between printing and the bottom up Synthetic Biology. Printing Biology aims at realizing reconfigurable multiscale systems (from nanometers to millimeters) with bespoke molecular composition, allowing for the determination of molecular interactions and features in conditions mimicking those of the living systems. As representative examples, the reproducible fabrication process of stable fL-scale compartments (the size selected by Nature for the formation of organelles and molecular condensates) by inkjet printing (IJP) and microcantilever spotting (μCS) will be shown.

The molecular composition of the compartments will be varied with the final aim to demonstrate the activity retention of different classes of the encapsulated biomolecules. Three different model applications will be shown, including DNA, proteins and phospholipids ink printable formulations. At first, the mechanism of DNA oligonucleotides ink imbibition by μCS into nylon porous supports is demonstrated (Figure 1). Subsequently, the immobilized DNA oligonucleotides (printed at different concentrations) are hybridized with a fluorolabeled complementary sequence permits to demonstrate the retaining of biological function [4], and the optical detection of oligonucleotides down to few tents of zeptomoles. As a second application, the retaining of CYP2E1 enzymatic activity from IJP compartments mimicking mitochondria is shown, highlighting the possibility to further induce spatial organization of the reaction products (Figure 2) [5]. Finally, preliminary experiments showing the realization of ordered phospholipids compartments containing fluorescein tagged phospholipids by μCS onto glass surfaces are reported. These systems allow for the realization of artificial platforms that could find applications in membrane-protein interaction studies.


Back to speaker overview Back to Oral- and Flash presentations overview

Hadian, Kamyar

Helmholtz Zentrum München (Germany)

Dr. Kamyar Hadian is the Head of the Research Group ‘Assay Development and Screening Platform’ at the Helmholtz Zentrum München in Munich/Germany. Dr. Hadian’s laboratory focuses on two main research aspects: (i) unraveling mechanisms that drive or block ferroptosis, a regulated cell-death pathway and (ii) identifying and characterizing protein interactions and regulators of ubiquitin signaling pathways. In addition, his research group has deep understanding in employing biochemical high-throughput and phenotypic high-content imaging approaches combined with machine learning methods to identify and validate small molecule modulators for treatment of distinct diseases related to the fields of Cancer, Virology and Immunology.

Dr. Hadian studied Biology at the Technical University of Munich (TUM) and received his PhD at the Helmholtz Zentrum München/Ludwig Maximilians University (LMU) in the field of Virology (HIV research). After a short PostDoc period in the field of NF-kB signaling, he was appointed the Head of ‘Assay Development and Screening Platform’ at the HelmholtzZentrum München in 2010. In 2015 he became a tenured Principal Investigator. From 2016-2017 he had a parallel appointment as an Adjunct Associate Research Scientist at the Columbia University in New York/USA, where he joined Dr. Stockwell’s lab to establish a long-lasting transatlantic collaboration. Dr. Hadian is the author of >50 publications, 5 patent applications and 2 issued patents.


Topic:

Acriflavine, a clinically aproved drug, inhibits SARS-CoV-2 and other betacoronaviruses

Abstract:

The COVID-19 pandemic caused by SARS-CoV-2 has been socially and economically devastating. Despite an unprecedented research effort and availability of vaccines, effective therapeutics are still missing to limit severe disease and mortality. Using high-throughput screening, we identified acriflavine as a potent papain-like protease (PLpro) inhibitor. NMR titrations and a co-crystal structure confirm that acriflavine blocks the PLpro catalytic pocket in an unexpected binding mode. We show that the drug inhibits viral replication at nanomolar concentration in cellular models, as well as in vivo in mice and ex vivo in human airway epithelia, with broad range activity against SARS-CoV-2 and other betacoronaviruses. Considering that acriflavine is an inexpensive drug approved in some countries, it may be immediately tested in clinical trials and play an important role during the current pandemic and future outbreaks.


Back to speaker overview Back to Oral- and Flash presentations overview

Garcia-Moreno, Maria Isabel

University of Seville (Spain)

María Isabel García Moreno works in the research group Carbohydrate Bioorganic Chemistry at the University of Seville. She was appointed Tenured Professor of Organic Chemistry in 2009. Her outstanding contributions revolve around the development of sp2-iminosugars as specific inhibitors of glycosylhydrolases. Ongoing projects include the design of glycomimetics for moderate the activity of glycosidases and the development of new therapies, with special attention to lysosomal storage diseases, inflammation and cancer


Abstract:

GalNAc-related sp2-iminosugars as mutant lysosomal β-hexosaminidase A activity enhancers in late-onset Tay-Sachs disease patients’ fibroblasts.

Topic:

Dysfuntion of human β-hexosaminidase A (Hex A) results in Tay-Sachs disease (TSD), an autosomal recessive lysosomal storage disorder (LSD) condition associated with phenotypic neurodegeneration, for which no effective treatment options are available. Since many of the TSD-causative mutations do not compromise the catalytic site of Hex A, the development of pharmacological chaperones (PCs) that can stabilize the native folding of the protein despite its anomalous conformation and restore activity appears attractive. Most reported PCs developed for LSDs are competitive inhibitors of the target enzyme; they however exert an effector action by dissociating from the corresponding mature enzyme:inhibitor complex in the presence of an excess of substrate in the lysosomes of patient cells. A main problem is that Hex A inhibitors oftentimes also inhibit the related enzyme O-linked N-acetylglucosaminidase (GlcNAcase; OGA), which represents a serious drawback for translation into the clinics. Based on structural information and the known substrate selectivity profile of HexA and OGA, we have addressed this problem by designing sp2-iminosugar glycomimetics closely related to N-acetylgalactosamine (GalNAc). The new candidates feature either a neutral piperidine-derived thiourea or a basic piperidine-thiazolidine bicyclic core and are accessed through a structure diversity-oriented approach. Compounds behaving as selective nanomolar competitive inhibitors of human Hex A at pH 7, with high Hex A/OGA selectivity, and displaying a ten-fold lower inhibitory potency at pH 5 were identified, which should facilitate the dissociation of the Hex A-glycomimetic complex at the lysosome, were the Hex A substrate (namely GM2 ganglioside) accumulates. In agreement with this notion the selected candidates specifically increased the levels of lysosomal Hex A activity in patient fibroblasts having the G269S mutation, the one with the highest prevalence in late-onset Tay-Sachs disease.


Back to speaker overview Back to Oral- and Flash presentations overview