Principal Investigator:

Prof. Dr. Stephan Becker

Institut für Virologie
Philipps-Universität Marburg
Hans-Meerwein-Str. 2
35043 Marburg
Phone: +49 (0)6421-28 66253
E-Mail: becker(at)staff.uni-marburg(dot)de


Electronmicroscopic picture of Ebola virus leaving an infected cell (red arrow). ©Schauflinger

Project description

The Ebola virus (EBOV) causes severe fever with extraordinarily high fatality rates. The matrix protein VP40 of EBOV plays key roles for the virus replication cycle and is regulated by homooligomerization. VP40 dimerization is crucial for the protein’s transport towards the plasma membrane where the dimers polymerize resulting in filament formation which enables virus budding. VP40 octamerization results in the down-regulation of viral RNA synthesis. Due to their central role as building blocks of the higher-order oligomers, dimers represent a promising target for antiviral intervention. A fragment-based approach was used to identify hits binding to VP40 crystals which will be developed into lead compounds in order to inhibit VP40 oligomerization.

Scientific goal:

High-resolution crystal Structure of Ebola virus VP40 Dimers. ©Anke Werner

The project aims to develop lead molecules into antiviral compounds using structure-based drug design – a combination of protein crystallography with in silico methods – as well as cell culture experiments under BSL4- conditions for validation.

DRUID collaboration partners:

B1 Diederich/Kolb lab, A4 Heine/Reuter lab, D1 Steinmetzer lab, E3 Rahlfs/Przyborski lab


References A1: 1. *Hartlieb et al. (2007) PNAS 104: 624-9 2. *Hartlieb et al., (2003) J. Biol. Chem. 278: 41830-6 3. *Hoenen et al. (2005) J Virol. 79: 1898-905 4. *Möller et al. 79, 14876-86 (2005) J Virol. 5. Hoenen et al. (2010) J Virol 84: 7053-63. 6. *Gomis-Ruth et al. (2003) Structure 11: 423-33.
* own project-specific preliminary work

Principal Investigator:

Prof. Dr. Arnold Grünweller

Philipps-Universität Marburg
Bau C
Marbacher Weg 6
35032 Marburg
Tel.: +49 (0)6421-2825849
Fax: +49 (0)6421-28 25854
E-Mail: arnold.gruenweller(at)staff.uni-marburg(dot)de


Project description

The cellular RNA helicase eIF4A is an excellent target for the development of broad-spectrum antivirals. During initiation of viral protein synthesis, many viruses, especially corona viruses, rely on this enzyme, which can efficiently and specifically be inhibited by rocaglates. In project A2, we would like to further develop rocaglates for potential clinical trials by e.g. nebulizing these compounds for local application into the respiratory tract and by creating a detailed side effect profile. In addition, new eIF4A inhibitors will be screened and characterized.  The systematic mutagenesis of known coronavirus sequences should allow a prediction of rocaglate sensitivity in newly emerging coronaviruses. Finally, the therapeutic relevance of rocaglates in different DRUID-relevant pathogens expressing eIF4A variants will be investigated.

RNA clamping onto the surface of eIF4A by the rocaglate Silvestrol and evidence of inhibition of (corona)viral protein synthesis by the rocaglate CR-31-B (-).

Scientific goal:

Rocaglates will be further developed for their testing in clinical trials and the possibility to predict rocaglate sensitivity in emerging corona viruses and other DRUID-relevant pathogens will be evaluated.

 

DRUID Collaboration partners:

B2 Ziebuhr lab, B4 Grevelding lab, B5 Schlitzer lab, C6 Häberlein lab, D1 Friebertshäuser/Steinmetzer lab, D3 van Zandbergen lab, D4 Hermosilla/Mazurek/Taubert lab, E4 Spengler lab, E6 Schiffmann lab


References A2: 1.*Biedenkopf et al., (2017), Antiviral Res. 137: 76-81; 2. *Müller et al., (2018), Antiviral Res. 150:123-129; 3. *Elgner et al., (2018), Viruses. 10(4): 149; 4. *Glitscher et al., (2018), Viruses.  10(6): 301; 5. *Henß et al., (2018), Viruses.  10(11): 592; 6. *Müller et al., (2020), Antiviral Res. 175:104706; *7. *Müller et al., (2021), Antiviral Res. 186: 105012; 8. *Blum et al., (2020), J Cell Mol Med. 24(12): 6988-6999; [*own publications].

Principal Investigator:

Prof. Dr. Friedemann Weber

Institut für Virologie
FB Veterinärmedizin
Justus-Liebig-Universität Gießen
Schubertstraße 81
35392 Gießen
Phone: +49 (0)641-99 38350
E-Mail: friedemann.weber(at)vetmed.uni-giessen(dot)de


Project description

Due to their small genome, viruses are highly dependent on functions of the host organism. Many of those functions are regulated by cell-encoded posttranslational protein modifications for which a substantial number of pharmaceutical inhibitors are available.

Rift Valley Fever Virus (RVFV) is a mosquito-borne zoonotic pathogen endemic in parts of Africa. In large and devastating outbreaks, it typically kills thousands of farm animals and hundreds of humans. In the preceding funding period, we used a high-throughput genetic screen and identified a pro-viral host cell factor for RVFV that binds to posttranslational protein modifications. Inhibition of this factor in a human organoid model reduced viral RNA synthesis and progeny particle production. In addition, proteomic analyses showed that a viral protein is modified is a manner that the host cell factor can bind, and mutation of the relevant site led to a reduction of viral RNA synthesis.

 

Scientific goal:

We aim to elucidate the molecular mechanism and exploit it to specifically inhibit RVFV infection. Moreover, we will test available pharmaceutical inhibitors, and also screen for other pathogenic RNA viruses that may depend on this mechanism.

 

DRUID Collaboration partners:

A1 Becker, A2 Grünweller, B2 Ziebuhr, C1 Bender/Hildt, D1 Friebertshäuser/Steinmetzer, E3 Rahlfs/ Przyborski, E4 Spengler, E6 Schiffmann, E7P Krijnse Locker


References A3: 1. Wuerth & Weber (2016) Viruses 8, 174*. 2. Barr, Weber, Schmaljohn (2020) Fields Virology, vol 1, p 706-749*

Principal Investigator:

Prof. Dr. Andreas Heine

Institut für Pharmazeutische Chemie
Marbacher Weg 6
35032 Marburg
Tel.: +49 (0)6421-28 21313
Fax: +49 (0)6421-28 28994
E-Mail: heinea(at)staff.uni-marburg(dot)de


Principal Investigator:

Prof. Dr. Klaus Reuter

Institut für Pharmazeutische Chemie
Philipps-Universität Marburg
Marbacher Weg 6
35032 Marburg
Tel.: +49 (0)6421-28 25845
Fax: +49 (0)6421-28 28994
E-Mail: reuterk(at)staff.uni-marburg(dot)de


Project description

Bacteria of the genus Shigella invade the epithelial cells of the colon, which results in the severe inflammation of the large intestine. Known as bacterial dysentery or Shigellosis, this causes a large number of deaths, foremost in developing countries. The Shigella specific chaperone IpgC interacts with numerous further pathogenicity factors and is prerequisite for the virulence of this organism. In the absence of a “substrate protein”, IpgC forms a homodimer, which is essential for its stability. We use IpgC as a target protein for the structure-based design of compounds against Shigellosis by preventing IpgC homodimer formation and/or binding to substrate proteins. By now, we have established a protocol which reproducibly yields excellently diffracting IpgC crystals. Using a fragment-based approach, we have identified a number of IpgC “binders”, some of which we were able to expand significantly. In addition to protein crystallography, we use “Microscale Thermophoresis”, Isothermal Titration Calorimetry” and a “Thermal Shift” assay to study the influence of such molecules on homodimer formation, on the ability to interact with “substrates” and on stability.

Crystal struc-ture of ho-modimeric IpgC. ©Klaus Reuter

“Follow up” compound bound to IpgC. ©Marina Gardonyi

Scientific goal:

In addition to further structural information on IpgC, our main goal is the optimization of the compounds identified so far using them as lead structures in the development of anti-Shigellosis compounds.

 

DRUID Collaboration partners:

A1 Stephan Becker, B1 Wibke Diederich / Peter Kolb, B7 Franco Falcone, D1 Eva Friebertshäuser / Torsten Steinmetzer


References A4: [1] Agerberth et al. (2005) World Health Organ [2] Williams & Berkley (2018) Paediatr Int Child Health 38:50-65. [3] Sansonetti (2001) Am J Physiol Liver Physiol 280:319-323. [4] Parsot et al. (2003) Curr Opin Microbiol 6:7-14. [5] Lunelli et al. (2009) Proc Natl Acad Sci USA 106:9661-9666.

Principal Investigator:

Dr. Ross Douglas

Biomedical Research Center Seltersberg (BFS)
Molecular Infections Biology
Justus Liebig Universität Giessen
Schubertstrasse 81
35392 Gießen
Tel.: +49 (0)641-99 39145
Fax: +49 (0)641-99 39129
E-Mail: ross.g.douglas(at)ernaehrung.uni-giessen(dot)de


Project description

Malaria remains one of the most devastating diseases and is caused by single celled parasites called Plasmodium. These parasites are transmitted between people by Anopheles mosquitoes. The parasite needs a set of diverse proteins that enable it to transmit to the mosquito vector, including members of the highly divergent actin cytoskeleton and its regulators. The parasite cytoskeleton has unique properties in order to transmit and, given its essential nature in various parasite processes at different life cycle stages, contains promising targets for novel malaria therapies. We make use of target validation approaches to identify and characterize novel transmission blocking targets that could be used to control disease spread.

Target validation approach has identified novel transmission blocking targets.

Scientific goal:

We aim to characterize identified proteins of interest using a variety of in vitro and in vivo methods with a view to identify novel compounds that selectively target these proteins and thus serve as transmission blocking drug candidates.

 

DRUID Collaboration partners:

A7 Przyborski, B1 Diederich/Kolb, B7 P Falcone, E3 Rahlfs/Przyborski


References A6: [1] Douglas et al. (2018) PLOS Bio e2005345; [2] Douglas et al. (2018) Malaria J 17:3191898-905; [3] Douglas et al. (2015) Trends Parasitol 31(8):357-362.

Principal Investigator:

Prof. Dr. Jude Przyborski

Interdisziplinäres Forschungszentrum (iFZ)
Justus-Liebig-Universität Gießen
Heinrich-Buff-Ring 26-32
35392 Gießen
Tel.: +49 (0)641-99 39114
E-Mail: jude.przyborski(at)ernaehrung.uni-giessen(dot)de

Homepage


Project description

Overview of assay design

Malaria parasites invade and live within mature human red blood cells RBC). To enable their survival, the parasite renovates it’s chosen host cell to its own advantage. Infected red blood cells become sticky and adhere to the lining of small blood vessels, and also coat themselves with proteins which enable them to become invisible the immune system. This unfortunately causes disease in the patient, and eventually leads to death. We have recently identified a number of important molecular players which are essential for this renovation process, including members of the so-called HSP70 and HSP40

families. It is the goal of this project to block the function of HSP40 and HSP70. If we can do this, parasites are likely to be cleared from the bloodstream, relieving the severity of disease. To do this, we will establish a number of assays to measure the activity of HSP40/HSP70, and use these to search for compounds which reduce this interaction. Promising compounds will then be tested directly on parasites for their ability to reduce host cell modification.


References A7: 1. Diehl et al. (2021) PLoS Pathogens 17:e1009969 2. Zhang et al. (2017) Sci Rep 7: 42188 3. Charnaud et al. (2017) PLoS One 12: e0181656 4. Külzer et al. (2012) Cell Micro 14: 1784-95 5. Külzer et al. (2010) Cell Micro 12: 1398-1420

Principal Investigator:

Prof. Dr. Wibke Diederich

Institut für Pharmazeutische Chemie
Zentrum für Tumor- und Immunbiologie
Philipps-Universität Marburg
Hans-Meerwein-Straße 3
35043 Marburg
Tel.: +49 (0)6421-28 25810
Fax: +49 (0)6421-28 26254
E-Mail: wibke.diederich(at)staff.uni-marburg(dot)de

Homepage


Principal Investigator:

Prof. Dr. Peter Kolb

Institut für Pharmazeutische Chemie
Philipps-Universität Marburg
Marbacher Weg 8
35032 Marburg
Tel.: +49 (0)6421-28 25908
Fax: +49 (0)6421-28 26652
E-Mail: peter.kolb(at)uni-marburg(dot)de

Homepage


Project description:

Infections with the dengue virus have reached a new high in recent years, with around 50-100 million new infections per year. In the vast majority of cases, the infection progresses with mild, flu-like symptoms, but a small percentage of those affected, often children, develop hemorrhagic fever, which is fatal if severe. Although a vaccine is now available, since this is only approved for a very limited group of people, the development of agents that efficiently suppress the multiplication of the virus is essential. Our work focuses on the virus’s own serine protease NS3/NS2B, which cleaves the viral precursor protein into functional proteins and is essential for the maturation of the virus.

Abb. B1. Kristallstruktur der NS3/NS2B Protease DENV 3.1

Scientific goal:

The aim of the project is to further develop allosteric inhibitors of the viral serine protease NS3/NS2B, not only with respect to their affinity, but also with respect to their pharmacokinetic properties and toxicity (hit-to-lead development) using a combined approach of computer-aided design, synthesis, biological assays and crystal structure analysis. In addition, inhibitors of the related Zika protease will be developed based on the knowledge gained.

 

DRUID Collaboration partners:

A1 Becker, A2 Grünweller, B3 Rahlfs/Kolb/van Zandbergen, XY Herker, E6 Schiffmann/Laux


References B1: [1] Noble et al. (2012) J Virol 86(1):438-446; *[2] Chevillard et al. (2015), J Chem Inf Model 55(9):1824-1835; *[3] Chevillard et al. (2018) J Med Chem 61(3):1118-1129

Principal Investigator:

Prof. Dr. John Ziebuhr

Institut für Medizinische Virologie
Biomedizinisches Forschungszentrum
Seltersberg (BFS)
Justus-Liebig-Universität Gießen
Schubertstraße 81
35392 Gießen
Tel.: +49 (0)641-99 41200
Fax: +49 (0)641-99 41209
E-Mail: John.Ziebuhr(at)viro.med.uni-giessen(dot)de


Project description:

Coronaviruses are important human and animal pathogens. They are mainly associated with respiratory and intestinal infections and have significant zoonotic potential, resulting in several outbreaks of severe respiratory infections in humans over the past 2 decades including the SARS-CoV-2 pandemic starting in 2019. Therapeutic options to treat severe forms of COVID-19 and other coronavirus infections are very limited, indicating a high priority for the development of novel antiviral drugs. To address this need, project B2 focuses on two coronaviral proteins that are conserved among all coronaviruses: the coronavirus macrodomain (macD) in nonstructural protein 3 (nsp3) and the recently discovered nucleotidyltransferase (NiRAN) which is linked to the viral RNA-dependent RNA polymerase domain in nsp12 and was recently shown to be essential for coronavirus replication.

Coronavirus replicase polyprotein 1ab. Proteolytic processing by two or three viral proteases (PL, 3CL) results in the release of up to 16 nonstructural proteins (nsp) with numerous enzymatic and other functions.

Scientific goal:

The project aims to comprehensively characterize the biochemical properties of two coronavirus proteins/enzymes (macD, NiRAN) and their functions in the viral replication cycle using appropriate cell culture systems. Based on the conserved substrate specificity of the NiRAN domain, HTS assays will be developed and used to identify potential inhibitors.

 

DRUID Collaboration partners:

A2 Grünweller, A3 Weber, C5 Kraiczy, E3 Rahlfs/Przyborski


References B2: 1. *Slanina et al. (2021) Proc Natl Acad Sci USA 118: e2022310118 2. *Krichel et al. (2021) Sci. Adv. 7: eabf1004  3. *Müller et al. (2021) Antiviral Res 175: 1004706 4. *Snijder et al. (2016) Adv Virus Res 96: 59-126 5. *Putics et al. (2005) J Virol 79: 12721-31.

Principal Investigator:

Dr. Stefan Rahlfs

Biochemie und Molekularbiologie
Justus-Liebig-Universität Gießen
Heinrich-Buff-Ring 26-32
35390 Gießen
Tel.: +49 (0)641-99 39117
E-Mail: stefan.rahlfs(at)ernaehrung.uni-giessen(dot)de


Principal Investigator:

Prof. Dr. Ger van Zandbergen

Abteilung Immunologie
Paul-Ehrlich-Institut
Paul-Ehrlich-Str. 51-59
63225 Langen
Tel.: +49 (0)6103-77 2005
E-Mail: Ger.Zandbergen(at)pei(dot)de


Principal Investigator:

Prof. Dr. Peter Kolb

Institut für Pharmazeutische Chemie
Philipps-Universität Marburg
Marbacher Weg 8
35032 Marburg
Tel.: +49 (0)6421-28 25908
Fax: +49 (0)6421-28 26652
E-Mail: peter.kolb(at)uni-marburg(dot)de

Homepage


Project description:

Cellular redox balance plays an essential role in pathogenic microorganisms. Enzymes of the NAD(P)H-dependent glutathione and thioredoxin system1,2, as well as the glucose 6-phosphate dehydrogenase (G6PD) and 6-phosphogluconate dehydrogenase (6PGD), which significantly contribute to the NADPH and ribose 5-phosphate pool via the pentose phosphate pathway3, are centrally involved. G6PD of the malaria parasite Plasmodium falciparum and P. vivax (GluPho), present as a bifunctional enzyme, differs functionally and structurally from the human host enzyme4 and is essential for malaria parasites5. Together with the Sanford-Burnham Institute/UCSD, La Jolla, we have established a high-throughput compatible assay for PfGluPho and have screened about 400,000 compounds (i.a. NIH MLSMR Collection)6. Following structure-activity relationship studies and lead optimization, we identified the highly selective PfGluPho inhibitor SBI-750, which is active in the lower nanomolar range7. The concept could already be transferred to Leishmania; the 3D crystal structure of Leishmania donovani G6PDs and PGDs could be solved and based on this a first in silico screening of small molecules could be realized. In addition, a high-throughput compatible assay for the recombinant G6PDs and 6PGDs from Leishmania was established to screen for inhibitors at the Novartis FAST lab in Cambridge (USA).

Workflow-Diagramm zur Inhibitor Identifizierung gegen G6PDs/PGDs

3D-Kristallstruktur LdG6PD ©Isabell Berneburg

Scientific goal:

The aim of this project is to functionally and structurally characterize the enzymes G6PD and 6PGD from Leishmania and Plasmodium as targets for drug development, and to identify and further develop inhibitors against these enzymes (in silico and HTS). The concept will also be transferred to other pathogens within the DRUID consortium, such as Schistosoma.

 

DRUID Collaboration partners:

B1 Diederich/Kolb; B4 Grevelding; D3 van Zandbergen; E1 Grevelding/Häberlein; E3 Rahlfs/Przyborski; E4 Spengler; E6 Schiffmann/Laux


References B3: 1. Fritz-Wolf et al. (2011) Nature Comm. 2:383* 2. Koncarevic et al. (2009) PNAS 106: 13323-8* 3. Bozdech and Ginsburg (2005) Malaria J 3:23 4. Jortzik et al. (2011) Biochem J Energy 436:641-50* 5. Allen et al. (2015) FEBS J 282:3808-23*
6. Preuss et al. (2012) J Med Chem 55:7262-72* 7. Berneburg et al. (2022) Antimicrob Agents Chemother (accepted)*

*own puplications

Principal Investigator:

Prof. Dr. Christoph Grevelding

BFS, Institut für Parasitologie
Justus-Liebig-Universität Gießen
Schubertstraße 81
35392 Gießen
Tel.: +49 (0)641-99 38466
Fax: +49 (0)641-99 38469
E-Mail: Christoph.Grevelding(at)vetmed.uni-giessen(dot)de


Project description:

Background: In the human and animal parasite Schistosoma mansoni, the sexual maturation of the female depends on a continuous pairing contact with the male (Abb. 1). Pairing is a prerequisite for egg produc-tion and as such decisive for the pathogenesis of schistosomiasis, which is induced by the eggs. Our research showed that different classes of molecules are involved in organizing reproductive processes in schisto-somes.1 Among others, experiments with kinase inhibitors and by kinase RNA interference demonstrated that kinases regulate cell division, sper-matogenesis, oogenesis, egg production and vitality of schistosomes. For instance, the Abl-tyrosine kinase inhibitor imatinib (cancer drug Glivec) negatively affected reproduction and caused the degradation of the gastrodermis of adult S. mansoni in vitro with lethal consequences.2,3

Besides kinases, which we study in cooperation with the Falcone group4, we investigate further enzymes such as aldehyde dehydrogenases (ALDHs) and an aldehyde reductase (AR). In schistosomes, as in other organisms, these molecules are putatively involved in regulating responses to molecular stress. Inhibiting these molecules might devitalize the parasite. First in vitro experiments with inhibitors against these molecules, like the ALDH inhibitor disulfiram, caused morphologic alterations, a decrease of pairing stability, reduced vitality and finally, the death of adult schistosomes within days in vitro. Helicases might also represent interesting targets as suggested by studies using the RNA helicase-specific inhibitor Silvestrol.5 In adult S. mansoni, Silvestrol reduced the vitality of adult worms in vitro but also stem cell-proliferation in gonad cells (Abb. 2). Together with the working groups of Prof. Schlitzer and Prof. Grünweller (Marburg), we will investigate these potential target molecules and develop synthetic inhibitors6,7 to study their physiological und morphological effects, first in vitro. Further, top candidates will be used for in vivo tests in a rodent infection model.

Abb. 1. Bright-field microscopy of a S. mansoni couple. During the constant pairing contact, the female (arrow) resides within the ventral groove of the male. Pairing is the essential pre-requisite for the production of eggs (stars).

Abb. 2. EdU-staining of female S. mansoni before (left) and after Silvestrol treatment (right). The num-ber of proliferating stem cells (green) is significantly reduced (right).

Scientific goal:

We will clone and characterize two RNA helicases of S. mansoni at the molecular and biochemical levels. In different cooperations within DRUID (see below), we will test the recombinant proteins in enzyme assays against Silvestrol and synthetic derivatives of this substance. Furthermore, we continue our analyses of the recombinantly expressed enzymes SmALDH312 and SmAR, further potential target molecules, and perform enzyme tests with inhibitors. Among these are disulfiram derivatives, which exhibited anti-schistosomal effects in vitro as shown in previous experiments. In addition, we plan to crystallize these molecules for structure analyses. Aim is to develop substances with inhibitor activity against the selected target molecules with high specificity, bioavailability and reduced toxicity for the host.8 Finally, results of the in vitro and in vivo experiments provide a basis for using this knowledge also for other parasitic systems, in which effective compounds will be tested (platform projekt E1).

 

DRUID Collaboration partners:

A2 AG Grünweller, B3 AG Rahlfs/Kolb/van Zandbergen, B5 AG Schlitzer, E1 Platformprojekt/Häberlein, E3 AG Rahlfs/Przyborski, E5 AG Czermak/Salzig lab


References B4: 1Beckmann et al. (2010) PLoS Pathog 6:e1000769; 2Beckmann et al. (2010) Int J Parasitol 40:521-6; 3Beckmann et al. (2012) Curr Pharm Des 18:3579-94; 4Moreira et al. (2022) Molecules (in press); 5Taroncher-Oldenburg et al. (2021) Microorganisms 9(3):540; 6Peter Ventura et al. (2021) ChemMedChem 14(21):1856-62; 7Peter Ventura et al. (2012) Arch Pharm (Weinheim) 354(12): e2100259; 8Mäder et al. (2018) ChemMedChem 13(22):2374-89.

Principal Investigator:

Prof. Dr. Martin Schlitzer

Institut für Pharmazeutische Chemie
Fachbereich Pharmazie
Philipps-Universität Marburg
Marbacher Weg 6
35037 Marburg
Tel.: +49 (0)6421 28-25840
E-Mail: schlitzer(at)staff.uni-marburg(dot)de


Project description:

Helminthic infections represent a major problem in large parts of the world. In comparison to the burden the arsenal of anthelminthic drugs is rather limited. Using successive cycles of design, synthesis and testing the novel class of dithiocarbamates shall be optimized regarding activity towards different helminths, host toxicity and drug-likeness. Derivatives are synthesized, tested against different helminths and human cell lines. For promising derivatives ADME-parameters are determined and selected compounds could be tested in vivo.

Different viruses pathogenic to humans, as for instance the Ebola- or the SARS-CoV-2 virus, are using host factors for intracellular replication. These host factors therefor represent a target for anti-viral drug design. One of these host factors is the RNA-helicase eIF4A. Based on the crystal structure of the eIF4A-RNA complex appropriate ligands are designed, evaluated by docking and in case of proper fit synthesized. Compounds are tested in regard to their effect on translation efficiency. Active derivatives are subsequently evaluated regarding their effect on virus replication in cell cultures.

Scientific goal:

Both projects should yield compounds which possess the quality of a lead structure or possibly a development candidate

 

DRUID Collaboration partners:

A2, A3, A4, A7, B2, B7P, C6 NWG, D4, E1, E4, E6


References B5:

Principal Investigator:

Prof. Dr. Eva Herker

Institut für Virologie
Philipps-Universität Marburg
Hans-Meerwein-Str. 2
35043 Marburg
Tel.: +49 (0)6421-28-64525
E-Mail: eva.herker(at)uni-marburg(dot)de


Project description:

The human pathogenic flaviviruses Dengue (DENV), Yellow Fever (YFV), Zika (ZIKV), West Nile (WNV) and tick-borne encephalitis virus (TBEV) cause acute infections with severe complications. Fundamental steps in flavivirus replication are closely linked to cellular lipids. These include, among other things, membrane reorganizations for the formation of replication vesicles or capsid envelopment. Flaviviruses alter the lipid composition of the host cell, and the activity of various lipid-metabolizing enzymes is essential for successful replication. For this reason, enzymes from different lipid metabolism pathways represent interesting targets for (pan-) antiflaviviral therapy. However, it has not yet been clarified in detail whether the above-mentioned flaviviruses are dependent on different or similar branches of lipid metabolism.

Immunofluorescence microscopy of cells infected with different flaviviruses; red: viral E protein; green: lipid droplets.

Scientific goal:

The project aims to identify antiviral targets in the cellular lipid metabolism. The role of various key enzymes in de novo fatty acid and cholesterol biosynthesis, in phospholipid and neutral lipid metabolism, and of lipid remodeling enzymes in flavivirus replication is analyzed using an shRNA-based screen. In addition, various inhibitors are tested. The results are validated in different cell types and the molecular mechanisms are examined in detail

 

DRUID Collaboration partners:

B1 Diederich/Kolb, D1 Böttcher-Friebertshäuser/Steinmetzer, E7P Krijnse-Locker, E4 Spengler


References B6P: Herker et al. (2010) Nat Med 16: 1295 2. Harris et al. (2011) J Biol Chem 286: 42615 3. Herker et al. (2012) J Biol Chem 287: 2280 4. Rosch et al. (2016) Cell Rep 16: 3219 5. Hofmann et al. (2018) Biochim Biophys Acta Mol Cell Biol Lipids 1863: 1041 6. Schobel et al. (2018) Sci Rep 8: 3893 7. Lassen et al. (2019) J Cell Sci 132: jcs.217042 8. Bley et al. (2020) Int J Mol Sci 21:  9. Herker et al. (2021) Trends Cell Biol 31: 345 10. Nguyen-Dinh et al. (2021) Cells 10: 2407

Principal Investigator:

Prof. Dr. Franco Falcone

Institut für Parasitologie
BFS - Biomedizinisches Forschungszentrum Seltersberg
Justus-Liebig-Universität Gießen
Schubertstraße 81
35392 Gießen
Tel.: +49 (0)641-99 38030
E-Mail: franco.falcone(at)vetmed.uni-giessen(dot)de


Project description:

Echinococcosis and Schistosomiasis are two co-called Neglected Tropical Diseases, which have a severe impact on the health of affected individuals in endemic countries. Our group is developing new tools that can be used for a better control of these two diseases. For schistosomiasis, we are developing new drugs that target the schistosomal Mitogen Activated Protein Kinases [1],  in an effort to find new drugs, which are larvicidal as well as adulticidal. For Echinococcosis, we are developing a novel diagnostic platform based on detection of parasite-specific IgE using humanized IgE reporter cell lines [2-4], which can be used in a variety of formats. We are also working on extending this diagnostic platform to other parasitic infections (e.g. Fasciola, Cysticercosis, Clonorchis, Opisthorchis).

Larvicidal and adulticidal activity of compound 38_8, chosen from a compound library using our bioinformatic approach.

Structural prediction of S. mansoni JNK compared to the human orthologue.

Scientific goal:

The Schistosome project aims to identify lead kinase inhibitor molecules using a bioinformatics in silico analysis pipeline and further develop these into suitable drugs, while in the Echinococcus project we are pursuing a better understanding of the protective immune responses against echinococci and aim to incorporate this knowledge into the development of vaccines and greatly improved, innovative diagnostic technologies.

 

DRUID Collaboration partners:

A4 Heine lab, B4 Grevelding lab, C6 NWG Häberlein lab, E5 Czermak/Salzig lab


References B7P: [1] *Pereira-Moreira et al. (2020) ACS Omega 5:9064-9070 [2] Kalli M et al. (2020) Sci Rep. 10:18208 [3] Kalli M. et al. (2020) Methods Mol Biol.;2163:155-162. [4] Prakash PS et al. (2021)  Parasitol Res.

Principal Investigator:

Dr. Daniela Bender

Paul-Ehrlich-Institut
Bundesinstitut für Impfstoffe
und biomedizinische Arzneimittel
Paul-Ehrlich-Straße 51-59
63225 Langen
Tel.: +49 (0)6103-77 5411
E-Mail: Daniela.Bender(at)pei(dot)de

Homepage


Principal Investigator:

Prof. Dr. Eberhard Hildt

Bundesinstitut für Impfstoffe
und biomedizinische Arzneimittel
Paul-Ehrlich-Institut
Paul-Ehrlich-Straße 51-59
63225 Langen
Tel.: +49 (0)6103-77 2140
Fax: +49 (0)6103-77 1234
E-Mail: Eberhard.Hildt(at)pei(dot)de

Homepage


Project description:

Zika viruses (ZIKV) are arboviruses, which belong to the flaviviridae family. During the ZIKV outbreak in Brazil in 2016, where a considerable number of microcephaly cases in newborns was associated with ZIKV infection during pregnancy. The WHO declared a public health emergency of international concern (PHEIC). At present neither a preventive vaccine or antiviral drugs are available. By inhibition of virus replication in an early phase of the viral life cycle if applicable as a temporary preventive approach, the viral load could be significantly reduced.  Thereby, the spread of the virus would be impaired and the risk of an intrauterine infection would be reduced.  During the first funding period target structures could be identified.

Intracellular distribution of tetherin (red) and ZIKV envelope protein (green) in cells that were infected either by the Uganda or the French Polynesia isolate.

Scientific goal:

Based on already identified targets and further targets, antiviral strategies are developed, the underlying mechanisms will be investigated and the effect on additional members of the flaviviridae family will be studied.

 

DRUID Collaboration partners:

A2 Grünweller lab, B1 Diderich/Kolb lab, B6P Herker lab, C2 Kempf lab, C5 Glebe/Geyer lab, D1 Steinmetzer lab, E6 Schiffmann/Laux lab


References C1: 1. Herrlein et al. (2021) J Virol. doi: 10.1128/jvi.02117-2  2. Sabino et al (2021)., J Virol. doi: 10.1128 3. Maddaluno et al., 2020 EMBO Mol Med. doi: 10.15252/emmm.201911793 4. Basic et al. 2019 Antiviral Res. doi: 10.1016/j.antiviral.2019.1046445. Akhras et al. (2019) Viruses doi: 10.3390/v11080748. 6.  Sabino et al., (2019) doi: 10.3390/v11060524 7. Elgner et al (2018) Viruses doi: 10.3390/v10040149

Principal Investigator:

Prof. Dr. Volkhard A. J. Kempf

Universitätsklinikum Frankfurt
Institut für Medizinische Mikrobiologie
und Krankenhaushygiene
Goethe-Universität Frankfurt/Main
Paul-Ehrlich-Str. 40
60596 Frankfurt/Main
Tel.: +49 (0)69-6301 5019
Fax: +49 (0)69-6301 83431
E-Mail: volkhard.kempf(at)kgu(dot)de


Project description

Bartonella bacilliformis is the causative agent of Carrións disease, a vector borne illness restricted to the South American Andes. The bacteria cause severe hemolytic fever with high fatality rates. The inhibition of hemolysis represents a promising therapeutic approach. Two pathogenicity factors play a crucial role in hemolysis of which at least one represents a promising drug target. Humans are the only known reservoir host for B. bacilliformis and represent the source of new outbreaks. It is therefore of particular importance for disease control to identify asymptomatic carriers. For this, an alpha version of a B. bacilliformis IgG ELISA and line blot were developed.

Scanning electron microscopy of infected human erythrocytes with B. bacilliformis.

B. bacilliformis NovaLisa® KIT.

Scientific goal:

The project aims to analyze the role of the two pathogenicity factors in the process of hemolysis to develop a novel, anti-virulence-based therapeutic strategy. Furthermore, the alpha version of the B. bacilliformis IgG ELISA and line blot will be evaluated in field studies in cooperation with our partners from Lima/Peru.

 

DRUID Collaboration partners:

A7 Przyborski lab, C1 Bender / Hildt, E7 Locker lab, Gold Standard Diagnostics (formerly NovaTec)


References C2: [1] Garcia-Quintanilla et al. (2019) Parasites&Vectors 12(1):141, [2] Riess et al. (2004) J Exp Med 200:1267-78, [3] Dichter et al. (2019) Microbiology Res Announc, DOI: 10.1128/MRA.01377-19, [4] Dichter et al. (2021) Lancet Microbe 2:e685–94.

Principal Investigator:

Prof. Dr. Peter Kraiczy

Goethe-Universität Frankfurt/Main
Institut für Medizinische Mikrobiologie und Krankenhaushygiene
Universitätsklinikum Frankfurt
Paul-Ehrlich-Str. 40
60596 Frankfurt am Main
Tel.: +49 (0)69-6301 7165
Fax: +49 (0)69-6301 83431
E-Mail kraiczy(at)em.uni-frankfurt(dot)de


Project description:

Borrelia recurrentis is considered as a “neglected arthropod-borne pathogen” and the causative agent of louse-borne relapsing fever. If left untreated, the case-fatality rate of this epidemic disease can exceed >40%. The hematogenous dissemination suggests that borreliae developed efficient immune evasion strategies to overcome innate immunity, in particular complement. Proteins interacting with complement are known to play a crucial role in host-pathogen interaction. Bioinformatic analyses revealed a cluster of five proteins all of which inhibit complement at different activation levels. Of importance, molecules interacting with the immune system represent promising candidates for the development of in vitro diagnostics. Two test systems (Line immunoblot and ELISA) for the diagnosis of louse-borne relapsing fever have already been developed and evaluated.

Immunofluorescene microscopy of B. recurrentis.

Schematic representation of the complement inactivation on the outer surface of B. recurrentis.

Scientific goal:

The focus of this project deals with the functional and structural characterization of additional complement-inhibiting proteins of Borrelia recurrentis as well as the optimization of the evaluated test systems and the development of a point-of-care antigen test for the diagnosis of relapsing fever.

 

DRUID Collaboration partners:

B2 Ziebuhr lab, C2 Kempf lab, C4 Steinhoff lab, D1 Steinmetzer lab, E3 Rahlfs/Przyborski, Gold Standard Diagnostics (formerly NovaTec)


References C3: [1] Cordes et al. (2005) Nat Struct Mol Biol 12:276-277; [2] Röttgerding et al. (2017) Sci Rep 7:303; [3] Nguyen et al. (2018) Front Cell Infect Microbiol. 8:23; [4] Walter et al. (2019) Front Immunol 10:2722; [5] Röttgerding and Kraiczy (2020) Front Immunol 11:1560; [6] Schmidt et al. (2021) Sci Rep 11:4964.

Principal Investigator:

Prof. Dr. Ulrich Steinhoff

Institut für Medizinische Mikrobiologie und Krankenhaushygiene
Philipps-Universität Marburg
Hans-Meerwein-Straße 2
35043 Marburg
Tel.: +49 (0)6421-28 66134
Fax: +49 (0)6421-58 66420
E-Mail: ulrich.steinhoff(at)staff.uni-marburg(dot)de


Project description:

A reliable diagnosis and treatment of humans and dogs (reservoir host) suffering from visceral leishmaniasis (VL) is crucial for the control of this infection. Currently available diagnostic tests based on the antibody reaction with the Leishmania kinesin protein show poor sensitivity in some endemic areas. We have developed and patented a kinesin antigen (rKLi8.3) with an improved diagnostic performance in humans and animals. This was possible by developing a kinesin antigen with an optimized kinesin structure (repeats) and sequence. Currently various test formats with the rKLi 8.3 antigen are produced and tested with sera from infected humans and dogs.

The treatment of VL is problematic in terms of effectiveness and side effects. An inhibitor (GNF-6702) that selectively inhibits the proteasome of the Kinetoplastida (T. brucei, T. cruzei and L. donovani) has been developed. GNF-6702 binds to a proteasome subunit of Kinetoplastida (beta 4 subunit) that is structurally distinct from humans. The specific bind-ing to the kinetoplast proteasome seems to be the reason for the very low toxicity in mam-malian cells. We could demonstrate that the proteasomal beta 4 subunit is conserved in all Leishmania isolates, tested to date. Thus, the proteasome is a promising therapeutic target.

 

Scientific goal:

In the diagnostic part of the project, various formats of a sero-diagnostic VL rapid test will be manufactured and tested in cooperation with our industrial partner. In the treatment part of the project, we validate the specificity, effectiveness and toxicity of the new, kinetoplastid-specific proteasome inhibitors in cell culture experiments of Leishmania infected macrophages.

 

DRUID Collaboration partners:

B1 Diederich/Kolb lab, C2 Kempf lab, D3 van Zandbergen lab


References C4: [1] Abass et al. (2013) PLoS Negl Trop Dis. 18;7; [2] Abass et al. (2015) PLoS One. 3;10; [3] Martínez Abad et al. (2017) Acta Trop. 166:133-138; [4] Pereira et al. (2020) Eur J Microbiol Immunol 27;10:165-171. [5] Khare et al. (2016) Nature 537: 229-233

Principal Investigator:

Prof. Dr. Dieter Glebe

Institut für Medizinische Virologie
Justus-Liebig-Universität Gießen
Schubertstraße 81
35392 Gießen
Phone: +49 (0)641-99 41246
E-Mail: dieter.glebe(at)viro.med.uni-giessen(dot)de


Principal Investigator:

Prof. Dr. Joachim Geyer

Institut für Pharmakologie und Toxikologie
Justus-Liebig-Universität Gießen
Schubertstraße 81
35392 Gießen
Phone: +49 (0)641-99 38404
E-Mail: joachim.m.geyer(at)vetmed.uni-giessen(dot)de


Project description:

Infections with the Hepatitis B (HBV) and D (HDV) viruses are the main cause of hepatocellular carcinoma and liver cirrhosis as a consequence of chronic hepatitis. Although an effective prophylactic vaccine is available, therapeutic options are highly limited, in particular for HDV. A promising novel drug target to block HBV/HDV virus entry into hepatocytes is represented by the hepatic bile acid carrier NTCP (Na+/taurocholate co-transporting polypeptide) that has been identified as the bona fide hepatic receptor for HBV/HDV. So far, more than 200 compounds from different compound classes have been tested and corresponding 3D structure-activity-relationship (QSAR) models have been generated. Furthermore, a pharmacophore model for HBV/HDV entry inhibitors was established. Virtual compound libraries have already been screened with these models and further effective hits have been identified.

NTCP is a physiological bile acid transporter in the plasma membrane of liver cells. NTCP is also the hepatic receptor for HBV and HDV. HBV/HDV entry inhibitors should selectively block NTCP viral receptor function. 3D QSAR and pharmacophore models help identify novel NTCP inhibitors.

 

Scientific goal:

Development of oral and selective HBV/HDV entry inhibitors that specifically block virus binding to NTCP, without tackling its physiological bile acid transport function. Hit compounds will be further developed into lead structures by means of molecular drug design.

 

DRUID Collaboration partners:

B1 Diederich/Kolb, B3 Rahlfs/Kolb/van Zandbergen, D2 Pfeiffer/Zeuzem/Hildt, E3 Rahlfs/Przyborski, E6 Schiffmann/Laux


References C5:1. *Kirstgen et al. (2020) Sci Rep 10:21772 2. *Grosser et al. (2021) Front Mol Biosci 8:689757 3. *Kirstgen et al. (2021) Viruses 13:666 4. *Kirstgen et al. (2021) Viruses 13:1489.

Principal Investigator:

PD Dr. Simone Häberlein

Justus-Liebig-Universität Gießen
BFS, Institut für Parasitologie
Schubertstraße 81
35392 Gießen
Tel.: +49 (0)641-99 38476
Fax: +49 (0)641-99 38469
E-Mail:

Homepage


Project description:

Protein kinases regulate a vast variety of cellular processes and represent promising targets not only for therapy of cancer, but also infections with parasites. One common feature of both disease types is the involvement of stem cells in tissue growth. We pursue the hypothesis that inhibition of protein kinases can be employed as a new therapeutic option against the liver fluke Fasciola hepatica, a globally prevalent zoonotic and NTD-associated pathogen. The project involves three aspects: (1) bioinformatical identification of putative drug targets within the Fasciola kinome and their genetic validation, (2) identification of kinase inhibitors with activity against liver flukes; and (3) characterization of the mode of action of potent kinase inhibitors by using biochemical and imaging-based methods.

In cooperation with the Spengler lab (project E4) we established AP-MALDI mass spectrometry imaging to achieve “drug imaging” within liver fluke tissue, which allows to study the route of drug uptake, its kinetic and tissue tropism of a drug.

Strategy to identify protein kinase inhibitors as drug candidates against the liver fluke Fasciola hepatica. ©Simone Häberlein

Scientific goal:

This projects aims to identify protein kinase inhibitors that can serve as new drug candidates for the therapy of fasciolosis.

 

DRUID Collaboration partners:

A2 Grünweller lab, B4 Schlitzer lab, B5 Grevelding lab, B7 Falcone lab, E4 Spengler lab


References C6: 1. Houhou et al. (2019) Sci Rep 9:15867. 2. Li et al. (2019) Parasitol Res 118(3):881-890. 3. Morawietz et al. (2020) Front Vet Sci 7:611270. 4. Mokosch et al. Anal Bioanal Chem 413(10): 2755-2766. 5. Morawietz et al. (2022) Parasitol Res (online ahead of print) doi: 10.1007/s00436-021-07388-1

Principal Investigator:

Prof. Dr. Eva Friebertshäuser

Institut für Virologie
Philipps-Universität Marburg
Hans-Meerwein-Str. 2
35043 Marburg
Tel.: +49 (0)6421-28 66019
Fax: +49 (0)6421-28 68962
E-Mail: friebertshaeuser(at)staff.uni-marburg(dot)de


Principal Investigator:

Prof. Dr. Thorsten Steinmetzer

Institut für Pharmazeutische Chemie
Philipps-Universität Marburg
Marbacher Weg 10
35032 Marburg
Tel.: +49 (0)6421-28 25900
Fax: +49 (0)6421-28 25901
E-Mail: torsten.steinmetzer(at)staff.uni-marburg(dot)de


Project description:

Cleavage of viral envelope proteins by host proteases is essential for the infectivity of many human pathogenic viruses. Among others, the surface glycoproteins of highly pathogenic avian influenza viruses (e.g. H5N1), chikungunya virus or dengue, West Nile and Zika viruses are activated by furin-like serine proteases. The surface glycoproteins hemagglutinin of zoonotic H7N9 and seasonal influenza A viruses or the spike protein S of many coronaviruses (CoV) are cleaved by the membrane-bound trypsin-like serine protease TMPRSS2. Recently, we were able to show that SARS-CoV-2 S is activated by both furin and TMPRSS2. Therefore, these host proteases are promising targets for the development of novel broad-spectrum antiviral agents.

Crystal structure of furin in complex with inhibitor MI-1851

Crystal structure of TMPRSS2 superimposed with inhibitors MI-1904 (yellow) and MI-432 (orange).

 

Scientific goal:

Structure-based development of effective inhibitors targeting virus-activating host proteases; determination of their potency and selectivity by enzyme kinetic studies; structural characterization of their binding mode in complex with relevant host proteases; testing of their antiviral efficacy against significant human pathogenic viruses in cell cultures, tissue cultures and animal models.

 

DRUID Collaboration partners:

A1 Becker lab, A2 Grünweller lab, B6 Herker lab, C1 Hildt lab


References D1: 1. Böttcher et al. (2006) J Virol 80: 9896-8 3. Becker et al. (2012) J Biol Chem 287: 21992-03 4. Böttcher-Friebertshäuser et al. (2012) Vaccine 30: 7374-80 5. Ivanova et al. (2017) ChemMedChem 12: 1953-68 6. Lam van et al. (2019) ChemMedChem 14, 673-85 7. Bestle et al. (2020) LSA 3: e202000786  8. Bestle et al. (2021) J Virol 95: e0090621 9. Lam van et al. (2021) ACS Med Chem Lett 12: 426-32.

Principal Investigator:

Dr. Kai-Henrik Peiffer

Goethe-Universität Frankfurt am Main
Universitätsklinikum
Zentrum der Inneren Medizin
Medizinische Klinik I
Theodor-Stern-Kai 7
60596 Frankfurt am Main
E-Mail: kai-henrik.peiffer(at)kgu(dot)de


Principal Investigator:

Prof. Dr. Eberhard Hildt

Bundesinstitut für Impfstoffe
und biomedizinische Arzneimittel
Paul-Ehrlich-Institut
Paul-Ehrlich-Straße 51-59
63225 Langen
Tel.: +49 (0)6103-77 2140
Fax: +49 (0)6103-77 1234
E-Mail: Eberhard.Hildt(at)pei(dot)de

Homepage


Principal Investigator:

Prof. Dr. Stefan Zeuzem

Department of Medicine
Goethe University Hospital
Theodor-Stern-Kai 7
60590 Frankfurt am Main
Tel.: +49 (0)69-6301 4544
E-Mail: zeuzem(at)em.uni-frankfurt(dot)de


Project description:

HEV infects more than 20 Mio people per year  and concerns industrial nations as well as developing countries. At present there is no specific therapy available.As non-enveloped virus HEV release depends on endodomal processes which represent a promising target for antivirals. In the first funding period the lysosmal degradation of HEV in the endosomal system was identified as a central anchor point for antivirals. In addition to innate immunity cholesterol homeostasis is a a central factor affecting HEV reelase. Based on this the underlying mechanisms  are analysed in detalil to identifiy further targets  which can be addressed by drug repurposing. Animal models will help to broaden the insight in immunological processes controlling HEV life cycle.

3D reconstruction of subcellular distribution of HEV (green) , GBP-1 (cyan) and lysosomes (red) in interferon -treated cells.

CLSM-based 3D reconstruction of sub-cellular distribution of HEV (green) and lysosomes (cyan) after induction of and cholesterol accumulation (magenta).

 

Scientific goal:

Inhibtion of the endosomal life cycle of HEV by modulation of cholesterol-dependent regulated target structurs via application of identified bioactive compounds (drug repurposing).

 

DRUID Collaboration partners:

A2 Grünweller lab, A4 Heine/Reuter lab, B1 Diederich/Kolb lab, B6P Herker lab, C5 Glebe/Geyer lab, D1 Steinmetzer lab


References D2:  [1] Glitscher et al. (2018) Viruses 10(6):301; [2] Müller et al. (2020) Antiviral Res 174:104706; [3] Basic et al. (2019) Antiviral Res 172:104644; [4] Glitscher et al. (2021) J Virol doi: 10.1128/JVI.01564-20; [5] Glitscher et al. (2021) Cell Mol Gastroenterol Hepatol, doi:10.1016 /j.jcmgh.2021.02.002; [6] Himmelsbach et al. (2018) Emerg Microbes Infect 7(1):196. [7] Glitscher et al. (2021) Cell Microbiol. 16:e13379

Principal Investigator:

Prof. Dr. Ger van Zandbergen

Abteilung Immunologie
Paul-Ehrlich-Institut
Paul-Ehrlich-Str. 51-59
63225 Langen
Tel.: +49 (0)6103-77 2005
E-Mail: Ger.Zandbergen(at)pei(dot)de


Project description:

Leishmaniasis is a neglected tropical disease caused by the parasite Leishmania spp, which is endemic in almost 100 countries. We could demonstrate that apoptotic Leishmania major (L. major) promastigotes are both responsible for the infectivity and the survival of parasites in host cells. Apoptotic parasites induce an anti-inflammatory response in human macrophages that does not lead to an effective T-cell response against Leishmania. Even if L. major parasites show all typical characteristics of apoptosis, typical eukaryotic apoptosis regulating proteins are not present in Leishmania.

Staining of fragmented DNA (TUNEL staining) in L. major Cas9/T7 in which apoptosis was induced by Miltefosin (TUNEL: green; DAPI: blue; anti-Lm-serum: red. © Ger van Zandbergen)

 

Scientific goal:

For a novel vaccination approach and to kill parasites we aim to identify apoptosis regulating proteins in Leishmania as potential drug targets as well as attenuated Leishmania strains without anti-inflammatory properties.

 

DRUID Collaboration partners:

B1 Kolb, B3 Rahlfs/Kolb, E3 Rahlfs/Przyborski


References D3: [1] Arens et al. (2018) Front Immunol 31(9):1772. [2] Crauwels et al. (2019) Front Immunol 22(10):2697. Further publications within DRUID: [3] Turoňová et al. (2020) Science 370(6513):203-208.

Principal Investigator:

Prof. Dr. Sybille Mazurek

Institut für Veterinär-Physiologie und -Biochemie
Fachbereich Veterinärmedizin
Justus-Liebig-Universität Gießen
Frankfurter Str. 100
35392 Gießen
Tel.: + 49 (0)641-99 38182
E-Mail Sybille.Mazurek(at)vetmed.uni-giessen(dot)de

Website


Principal Investigator:

Prof. Dr. Carlos Hermosilla

Institut für Parasitologie/Institut für Veterinär-Physiologie und -Biochemie
Justus-Liebig-Universität Gießen
Schubertstr. 81/Frankfurter Str. 100
Tel.: +49 (o)641-99 38461/99 38182
E-Mail: Carlos.R.Hermosilla(at)vetmed.uni-giessen(dot)de

Homepage


Principal Investigator:

Prof. Dr. Anja Taubert

BFS, Institut für Parasitologie
Justus-Liebig-Universität Gießen
Schubertstraße 81
35392 Gießen
Tel.: +49 (0)641-99 38460
Fax: +49 (0)641-99 38469
E-Mail: Anja.Taubert(at)vetmed.uni-giessen(dot)de

Website


Project description:

Cryptosporidium spp. are intestinal parasites which cause severe diarrhoea in humans, especially in HIV patients and young children. Especially in developing countries these protozoan infections induce high morbiditiy and mortality. So far, epidemiological factors contributing to cryptosporidiosis in underdeveloped countries – such as Cameroon – have insufficiently been studied. Currently available therapeutics show insufficient efficacies in the above mentioned risk groups. Cryptosporidium spp. are obligate intracellular protozoa which own minimal metabolic capacities. Consequently, to sustain their intracellular proliferation, these parasites have to modulate the host cellular metabolism. By characterizing metabolic signatures of C. parvum-infected host cells, we recently identified host cell metabolic reactions and pathways that are essential for parasite proliferation.

Cryptosporidium parvum- (yellow) infected host cells (nuclei: blue), tomographic microscopie © Juan Vélez

Inhibition of Cryptosporidium parvum via exemplary metabolic blockers (Vélez et al. 2021c)

 

Scientific goal:

This projects targets specific metabolic pathways of host cells (mainly glycolysis and glutaminolysis) by using new metabolic inhibitors or combinatory treatments. Moreover, a One Health study on several epidemiological factors of cryptosporidiosis in Cameroon is conducted.

 

DRUID Collaboration partners:

E4 Spengler lab


References D4: 1. *Vélez et al. (2021) Pathogens 11(1):49.  2.  * Vélez et al. (2021) Biology 10(10):963 3. ** Vélez et al. (2021) Biology 10(1):60

Principal Investigator:

PD Dr. Simone Häberlein

Justus-Liebig-Universität Gießen
BFS, Institut für Parasitologie
Schubertstraße 81
35392 Gießen
Tel.: +49 (0)641-99 38476
Fax: +49 (0)641-99 38469
E-Mail:

Homepage


Principal Investigator:

Prof. Dr. Christoph Grevelding

BFS, Institut für Parasitologie
Justus-Liebig-Universität Gießen
Schubertstraße 81
35392 Gießen
Tel.: +49 (0)641-99 38466
Fax: +49 (0)641-99 38469
E-Mail: Christoph.Grevelding(at)vetmed.uni-giessen(dot)de


Project description:

Targets of antiparasitic compounds are often conserved, which opens the possibility to achieve therapy of different parasitic infections with the same active compound. In platform project E1, we focus on parasitic worms (helminths), for which the discovery of new active compounds is particularly important because of resistances or suboptimal therapeutic successes of existing drugs. To this end, we implement in vitro screenings of compounds originating from DRUID projects to test their efficacy against the blood fluke Schistosoma mansoni (causing schistosomiasis) and the liver fluke Fasciola hepatica (causing fascioliasis). The platform project cooperates with several national and international partners, including colleagues in endemic countries in Asia and Africa, who open us test possibilities against numerous other helminths species.

Strategy for identifying active compounds with broad antiparasitic efficacy against globally important helminths species. ©Simone Häberlein

 

We also investigate the mechanism of action for selected test compounds by applying various in vitro culture-based and imaging methods.

Test spectrum provided by the platform project to identify and characterize anthelminthic compounds. ©Simone Häberlein, Miray Tonk-Rügen

Scientific goal:

Aim of the project is the identification of substances with a broad anti-parasitic efficacy and the discovery of conserved modes of action.

 

DRUID Collaboration partners:

A2 Grünweller lab, A6 Douglas lab, A7 Przyborski lab, B4 Schlitzer lab, B5 Grevelding lab, B7 Falcone lab, E4 Spengler lab


References E1: 1. Peter-Ventura et al. (2019) ChemMedChem 14(21):1856-1862. 2. Houhou et al. (2019) Sci Rep 9:15867. 3. Li et al. (2019) Parasitol Res 118(3):881-890. 4. Morawietz et al. (2020) Front Vet Sci 7:611270. 5. Kellershohn et al. (2019) PLOS Negl Trop Dis 13:e0007240. 6. Tonk et al. (2020) Antibiotics 9:664. 7. Mokosch et al. Anal Bioanal Chem 413(10): 2755-2766. 8. Mughal et al. (2021a) Int J Parasitol 51(7):571-585. 9. Mughal et al. (2021b) Int J Parasitol S0020-7519(21)00312-X. 10. Gallinger et al. (2022) Pharmaceuticals 15(2): 119. 11. Morawietz et al. (2022) Parasitol Res (online ahead of print) doi: 10.1007/s00436-021-07388-1

Principal Investigator:

Prof. Dr. Jude Przyborski

Interdisziplinäres Forschungszentrum (iFZ)
Justus-Liebig-Universität Gießen
Heinrich-Buff-Ring 26-32
35392 Gießen
Tel.: +49 (0)641-99 39114
E-Mail: jude.przyborski(at)ernaehrung.uni-giessen(dot)de

Homepage


Principal Investigator:

Dr. Stefan Rahlfs

Biochemie und Molekularbiologie
Justus-Liebig-Universität Gießen
Heinrich-Buff-Ring 26-32
35390 Gießen
Tel.: +49 (0)641-99 39117
E-Mail: stefan.rahlfs(at)ernaehrung.uni-giessen(dot)de


Project description:

Within the framework of this platform project, we offer methods for protein production, for the development of high-throughput assays, for crystallisation and structural analysis as well as for interaction analyses. A variety of optimisation options for cloning/expression and purification of native proteins (affinity chromatography, gel filtration, ion exchange) are available. Once recombinant protein has been obtained, provide support in the development of assay systems up to high throughput formats. Furthermore, the platform proves access to the crystallisation platform at the iFZ. A pipetting and a crystallisation robot as well as starter kits and various additive screens for optimisation are available for crystallisation screens on the nl scale. The system is currently being updated with funding from the latest DRUID funding round. Dr Fritz-Wolf acts as crystallographer and contact person and can carry out the first tests on the crystals. The X-ray sources of the Max Planck Institute for Medical Research (Heidelberg) and the AG Klebe/Heine [A5] (Marburg) are available for this purpose.

Crystal structure of sfroGFP (Heimsch et al. 2022)

DRUID Collaboration partners:

Protein-Expression: A1, A3, A6NWG, A7, B2, B3, B4, C3
Crystallisation: B2, B3, C3
Assay-Devlopment: A7, B3


References E3: 1. Heimsch et al. (2022) Antioxid & Redox Signal doi: 10.1089/ars.2021.0234 2. Harnischfeger et al. (2020) Electronic Journal of Biotechnology 3. Fritz-Wolf et al. (2011) Nat Commun 2:383 4. Koncarevic et al. (2009) Proc Natl Acad Sci USA 106:13323-8

Principal Investigator:

Prof. Dr. Bernhard Spengler

Institut für Anorganische und Analytische Chemie
Justus-Liebig-Universität Gießen
Heinrich-Buff-Ring 17
35392 Gießen
Tel.: +49 (0)641-99 34801
E-Mail: bernhard.spengler(at)ac.jlug(dot)de

Homepage


Project description:

High resolution mass spectrometry imaging provides numerous opportunities to aid in the characterization of pathogens, host-pathogen interactions and the analysis of drug molecules. The sample is analyzed with a laser in a rasterized fashion, the step size is only a few micrometers. The sample material is ablated and ionized, allowing for the parallel detection of hundreds of chemical compounds as well as the creation of images that show the distribution of the analytes throughout the sample.  Identification of the molecules is carried out using exact mass determination and is supported by fragmentation experiments. Continued improvements in instrumentation allow for analysis and graphical representation of smaller structures while additional sample preparation steps like in-situ derivatization provide the opportunity to detect analytes at very low concentrations.

 

Scientific goal:

The goal of the project is the improvement and application of the available methods for the demands of the DRUID center. Especially the distribution of drug compounds, the lipidomic and metabolomic characterization of pathogens and the interaction of pathogens and hosts are important research areas.

 

DRUID Collaboration partners:

A2 Grünweller lab, B4 Grevelding lab, B5 Schlitzer lab, D4 Taubert lab, E1 Häberlein lab, W3 Herker lab


References E4: [1] Kadesch et al. (2020) PLoS Negl. Trop. Dis. 14(5): e0008145; [2] Morawietz et al. (2020), Front. Vet. Sci. 7, 611270; [3] Mokosch et al. (2021)  Anal Bioanal Chem 413, 2755–2766; [4] Müller et al. (2021) J. Am. Soc. Mass Spectrom.,3,32(2),465-472; [5] Spengler et al. (2015) Anal Chem 87 (1) 64-82.

Principal Investigator:

Prof. Dr. Ing. Peter Czermak

Institut für Bioprozesstechnik
Technische Hochschule Mittelhessen
Wiesenstraße 14
35390 Gießen
Tel.: +49 (0)641-309 2551
E-Mail: peter.czermak(at)lse.thm(dot)de


Principal Investigator:

Prof. Dr. Ing. Denise Salzig

Institut für Bioverfahrenstechnik
und Pharmazeutische Technologie
Technische Hochschule Mittelhessen
Gutfleischstr. 3-5
Tel.: +49 (0)641-309 2630
Fax: +49 (0)641-309 2553
E-Mail: denise.salzig(at)lse.thm(dot)de


Project description:

For a translation of research results from the DRUID consortium into the clinic or industry, robust production processes, which represent more than just scaling up, are essential. These processes must follow the guidelines of good manufacturing practice (cGMP) and process analytical technology (PAT). The following work is planned: i) process intensification and expansion of the BEVS production platform as well as implementation of a continuous process control, ii) integration of online PAT technology, e.g., impedance spectroscopy for an automated determination of the optimal point in time for harvesting and automated harvesting, iii) expression (BEVS) and purification of S. mansoni kinases as well as study of the kinases and putative inhibitors, and iv) study of novel transfection reagents for transient protein production on a bioreactor scale.

Optimization of the transient transfection process using statistical design of experiments. ©IBPT

Production concept with integrated PAT technology. ©IBPT

 

Scientific goal:

The work on production process development and process control will be further adapted to the questions of the center, expanded, and offered to the entire consortium for use. It is planned to further intensify and automate the established BEVS (Baculovirus Expression System) platform. In addition, the second production platform – the transient production with HEK-293T cells – is also being further developed.

 

DRUID Collaboration partners:

B4 Schlitzer Lab, B5 Grevelding lab, B7 P Falcone Lab, C6 NWG Häberlein, E1 Platform Grevelding/ Häberlein


References E5: Biotechnol, DOI: 10.1016/j.ejbt.2021.08.002, 3. Lothert et al. (2020) Methods Mol Biol 2183:217-248, 4. Dekevic et al. (2022) J Biotechn 346 23-34, 5. Schwarz et al. (2021) Elec J Biotechnol, DOI: 10.1016/j.ejbt.2022.01.003; 6. Barekzai et al. (2020) New Adv Ferm Processes, DOI: 10.5772/intechopen.90029, 7. Eckhardt et al. (2021) Sep Sci Technol, 57 (6) 886-897

Principal Investigator:

Dr. Susanne Schiffmann

Fraunhofer Institut für Translationale Medizin und Pharmakologie ITMP
Theodor-Stern-Kai 7
60596 Frankfurt am Main
Tel.: +49 (0)69-8700 25060
E-Mail: susanne.schiffmann(at)itmp.fraunhofer(dot)de

Homepage


Project description:

The SARS-CoV2 pandemic clearly demonstrates that there is an unmet medical need for anti-pathogenic drugs. In order to identify new treatment options, promising substances from other working groups will be characterized preclinical. This includes the generation of a safety profile and/or bioavailability testing of the drug candidates. Since supportive therapy with immune-modulating drugs is a proven therapeutic approach, an interaction of the drug candidates with the immune system will be tested.

Surface marker expression of treated M2 macrophages ©Marina Henke

Cytokine expression of treated M2 macrophages. ©Leonard Blum

Scientific goal:

As part of the project, a safety and/or immunomodulatory profile will be created for small molecules/peptides that have a potential antiviral, antiparasitic or antibacterial property. Furthermore, human proteins (e.g. TMPRSS2) which represent new targets for combating pathogens will be investigated.

 

DRUID Collaboration partners:

A1 Becker lab, A2 Grünweller lab, A3 Weber lab, B1 Diederich/Kolb lab, B3 Rahlfs/Kolb/van Zandbergen, B5 Schlitzer lab, D1 Friebertshäuser lab


References E6: 1. Blum et al. (2021) J Mol Med 99(2):261-72; 2. Blum et al. (2020) Sci Rep 10(1):7534; 3. Blum et al. (2020) J Cell Mol Med 24(12):6988-99.

Principal Investigator:

Prof. Dr. Jacomina Krijnse Locker

Elektronenmikroskopie von Pathogenen
Paul-Ehrlich-Institut
Paul-Ehrlich-Straße 51-59
63225 Langen
Tel.: +49 (0)6103-77 2011
E-Mail: Jacomina.KrijnseLocker(at)pei(dot)de


Project description:

We apply imaging techniques to understand how pathogens interact with cells to cause disease, focusing on electron microscopy (EM) and correlated light- and electron microscopy (CLEM). To generate robust protocols that can easily be adapted, we use our model virus, the large DNA-virus vaccinia (VACV).

The SARS CoV2 pandemic illustrates how viruses have a world-wide socio-economical impact with dramatic consequences for the neglected tropical diseases (NTDs). There is an urgent need to contain the SARS CoV2 pandemic and refocus resources on NTDs. In a collaborative effort we showed that the spike-surface protein (S) of SARS COV2, that mediates infection, shows unexpected flexibility. The latter could facilitate receptor binding and cell entry of the virus, which remains to demonstrated.

Cryo-electron tomography and molecular dynamic simulation of SARS-CoV-2 spike shows unexpected flexibility of the stalk.

Upon budding at intracellular membranes coronaviruses leave the cell in a poorly explored way. Vesicular trafficking as well as regulated lysosomal exocytosis have been proposed.

 

Scientific goal:

The project aims to interfere with the flexibility of S and test for the outcome of infection using antibodies, drugs and genetics. Virus release from infected cells will be studied by a CLEM approach, combining live cell imaging with EM.  The expertise gained from our VACV- and SARS CoV2 host system is then applied to answer questions related to virus-host systems used in other teams of the DRUID-consortium.

 

DRUID Collaboration partners:

A3 Weber lab, B2 Ziebuhr lab, B6 P Herker lab, C1 Hildt lab, D3 van Zandbergen lab, E5 Czermak lab


References E7 P: Turoňová, B., et al. (2020). Science (80-. ). 370, 203–208. Blanco-Rodriguez, G., et al. (2020). J. Virol. 94 e00135-20. Quemin, E.R.,  et al. (2018). J. Mol. Biol.430, 1714-1724. Chlanda, P., and Krijnse Locker, J. (2017). Biochem. J. 474, 1041–1053. Sartori-Rupp, A., et al. (2019). Nat. Commun. 10, 342.