MASARYK UNIVERSITY Faculty of Science

National Centre for Biomolecular Research

PROTEINS INVOLVED IN HOST-PATHOGEN RECOGNITION

EVA FUJDIAROVÁ

Ph.D. Thesis

Supervisor: Prof. RNDr. Michaela Wimmerová, Ph.D.

Brno 2020

BIBLIOGRAFICKÝ ZÁZNAM

Autor: MVDr. Eva Fujdiarová Masarykova univerzita, Přírodovědecká fakulta Národní centrum pro výzkum biomolekul,

Název práce: Proteiny zapojené do rozpoznávání patogenu hostitelem Studijní program: Biomolekulární chemie a bioinformatika

Vedoucí práce: Prof. RNDr. Michaela Wimmerová, Ph.D. Přírodovědecká fakulta, Masarykova univerzita Ústav biochemie a Národní centrum pro výzkum biomolekul - NCBR, Středoevropský technologický institut – CEITEC, Masarykova univerzita

Akademický rok: 2019/2020

Počet stran: 138+165

Klíčová slova: Lektiny, sacharidy, Photorhabuds laumondii, reaktivní formy kyslíku, fenoloxidáza, multivalence

BIBLIOGRAPHIC ENTRY

Author: MVDr. Eva Fujdiarová National Centre for Biomolecular Research, Faculty of Science, Masaryk University

Title of Thesis: Proteins involved in host-pathogen interaction

Degree programme: Biomolecular chemistry and bioinformatics

Field of Study: Biomolecular chemistry and bioinformatics

Supervisor: Prof. RNDr. Michaela Wimmerová, Ph.D. Masaryk University, Faculty of Science, Department of Biochemistry and National Centre for Biomolecular Research Central European Institute of Technology, Masaryk University

Academic year: 2019/2020

Number of Pages: 138+165

Keywords: Lectins, saccharides, laumondii, reactive oxygen species, phenoloxidase, multivalency

ACKNOWLEDGMENT

My journey to biomolecular chemistry was not straightforward. I studied to be a veterinarian. Although I did enjoy the biochemistry classes, I threw away all my notes shortly after graduation thinking I would never need them again. I remember myself during all those lab practical seminaries thinking – I am not studying such a demanding field only to be pipetting in a lab. I wanted to be a surgeon, to save animals and gain people’s respect – as all of us did. And look at me now 10 years later – happily pipetting. 

In this place, I would like to thank everyone who has been there for me in this chapter of my life. I thank my parents and family for supporting my crazy idea to move to Brno and switch fields, for hiding their fears and doubts from me and telling only: you can do it, and there is always a place for you here. Their belief in me and the feeling of having a safe place to return helped me to face all challenges that doctoral study brings. I thank my supervisor professor Wimmerová for allowing me to learn and grow independently in her lab while watching my progress from distance, letting me go through all the necessary mistakes one needs to do in order to learn. But I thank her also for stepping forward whenever necessary, providing support, counsel, and insight I did not have, not letting me to walk in circles for too long. I thank Janča Vičanová and my shifu Dou Wanchun for English corrections of this text. I thank Pepa, Lenka, Janča, Jitka, Majka and all members of the Glyco group for creating the environment where it is not a shame to ask stupid questions and where co-workers become friends. I thank my friends for simply being. For traditions we have created during the years that I love so much – the movie club, Travný expeditions, TMOU!, boat trips… A special thanks to Hanka for our Dominion sessions, Andula for many hiking memories, and all people connected to “Mycelium” for making me feel I belong somewhere. Thank you all for reminding me that there are more ways to see the world and that it is important to keep things in perspective, especially when you are digging in proteins on an atomic level. And one special note for my little nephew: MITO MITO!

I hereby declare that the thesis “Proteins involved in host-pathogen recognition“, was written by me under the guidance of the thesis supervisor and with the use of literature sources.

Eva Fujdiarová

ABSTRAKT

Lektiny jsou skupina proteinů neimunitního původu, které rozeznávají sacharidy s neobyčejně vysokou specifitou. Díky této vlastnosti jsou lektiny ideálním nástrojem pro čtení “cukerného kódu”, který se nachází na povrchu všech buněk zapsán do struktury specifických cukerných epitopů. Lektiny zprostředkovávají buněčnou komunikaci na molekulární úrovni a jsou zapojeny do mnoha fyziologických i patofyziologických procesů. Patogenní bakterie a viry využívají lektiny k přichycení na hostitelskou tkáň, což je jeden z předpokladů pro rozvoj infekce. Blokace adheze patogenů specifickými lektinovými inhibitory je základem anti-adhezivní terapie, alternativního přistupu k léčbě infekcí způsobených multirezistentními bakteriálními kmeny. Tato dizertační práce je zaměřena převážně na studium lektinů z bakterie Photorhabdus laumondii. Tato bakterie má komplexní životní cyklus, který zahrnuje fázi mutualismu s mikroskopickou hlísticí rodu a také fázi patogenity vůči hmyzu. Bakterie ve svém genomu kóduje lektiny, jejichž přesná role v životním cyklu bakterie není dosud známá. Detailní charakterizace lektinů, jejich struktury a vazebných vlastností, stejně tak i studium delečních bakteriálních mutantů, může na tuto otázku odpovědět. Obsahem dizertační práce je také testování účinnosti multivalentních cukerných inhibitorů, zacílených na blokaci lidských patogenů jako Pseudomonas aeruginosa, Burkholderia cenocepacia a Photorhabdus asymbiotica. Text dizertační práce je rozdělen na teoretický úvod a praktickou část. Teoretický úvod je zaměřený na problematiku lektinů, antiadhezivní terapie a rodu Photorhabdus. V praktické části práce jsou popsány tři projekty, do kterých jsem byla v rámci studia zapojena a jsou v ní shrnuty dosažené výsledky.

ABSTRACT

Lectins are a group of proteins of non-immune origin that recognize carbohydrates with extremely high specificity. Due to this property, lectins are the ideal tool for reading a glyco code which is found on the surface of every cell and is encoded in the structure of specific sugar epitopes. Lectins mediate cellular communication at the molecular level and are involved in many physiological and pathophysiological processes. Pathogenic and viruses use lectins to attach to host tissues, which is one of the prerequisites for infection development. Blocking the adhesion of pathogens by specific lectin inhibitors is the basis of anti-adhesion therapy, an alternative approach to the treatment of infections caused by multi-resistant bacterial strains. This thesis is focused mainly on the study of lectins from Photorhabdus laumondii. This bacterium has a complex life cycle that includes the phase of mutualism with the microscopic nematode of the genus Heterorhabditis and also the phase of pathogenicity towards . The bacterium encodes lectins in its genome. Their exact role in the bacterial life cycle is not yet known. The detailed characterization of lectins, their structure, and binding properties, as well as the study of deletion bacterial mutants, may answer this question. The content of this thesis is also testing the effectiveness of multivalent sugar inhibitors aimed at blocking human pathogens such as Pseudomonas aeruginosa, Burkholderia ceanocepatia, and Photorhabdus asymbiotica. The text of the thesis is divided into a theoretical introduction and a practical part. The theoretical introduction is focused on lectins, anti-adhesion therapy, and Photorhabdus genus. In the practical part of the thesis, there are three projects, which I was involved in during the study, described and the results are summarized.

ABBREVIATIONS

3OMG 3-O-methyl-D-glucose AFP alpha-fetoprotein AUC analytical ultracentrifugation CA15-3 carbohydrate antigen associated with breast cancer CRDs carbohydrate-binding domains CRP C-reactive protein CuAAC copper-catalyzed azide-alkyne cycloaddition DMSO dimethyl sulfoxide EPNs entomopathogenic nematodes fMLF N-formyl-L-methionyl-L-leucyl-L-phenylalanine HA hemagglutinin HBSS Hank`s balanced salt solution HIA hemagglutination inhibition assay IJ infective juvenile IPTG isopropyl-β-D-1-thiogalactoside ITC isothermal titration calorimetry LCA Lens culinaris agglutinin MIC minimal inhibitory concentration MBL mannose-binding lectin MGMR 3,6-O-Me2-D-Glcβ1-4(2,3-O-Me2)-L-Rhaα MRSA methicillin-resistant Staphylococcus aureus RNA ribonucleic acid ROS reactive oxygen species SAP serum amyloid P SNR signal to noise ratio sp2 linker -O-(p-C6H4)-O-CH2CH2NH2 SPR surface plasmon resonance STD NMR saturation transfer difference nuclear magnetic resonance P1 primary variant of Photorhabdus (phase one) P2 secondary variant of Photorhabdus (phase two) PCR polymerase chain reaction PHA Phaseolus vulgaris agglutinin PMA phorbol 12-myristate 13-acetate PRSA penicillin-resistant Staphylococcus aureus PO phenoloxidase pPO prophenoloxidase UTIs urinary tract infections VRSA vancomycin-resistant Staphylococcus aureus WGA wheat germ agglutinin

TABLE OF CONTENT

BIBLIOGRAFICKÝ ZÁZNAM ...... 3 BIBLIOGRAPHIC ENTRY ...... 5 ACKNOWLEDGMENT ...... 7 ABSTRAKT ...... 11 ABSTRACT ...... 13 ABBREVIATIONS ...... 15 TABLE OF CONTENT ...... 17 1. LECTINS ...... 21 1.1. History ...... 21 1.2. Basic characterization and classification ...... 23 1.2.1. Microbial and viral lectins ...... 25 1.2.2. Plant lectins ...... 28 1.2.3. Animal lectins ...... 29 1.3. Lectin applications ...... 30 1.3.1. Research ...... 30 1.3.2. Agriculture ...... 31 1.3.3. Medicine ...... 32 2. ANTI-ADHESION THERAPY ...... 34 2.1. Introduction ...... 34 2.2. Approaches to anti-adhesion therapy ...... 35 2.2.1. Inhibition of pilus assembly ...... 36 2.2.2. Usage of adhesin analogs ...... 36 2.2.3. Dietary supplements ...... 36 2.2.4. Carbohydrates as lectin inhibitors ...... 37 2.2.5. Other approaches ...... 39 2.3. Perspectives of anti-adhesion therapy ...... 40 3. PHOTORHABDUS GENUS ...... 41 3.1. History and ...... 41 3.2. Basic characterization ...... 43 3.3. Life cycle ...... 44 3.4. Lectins from Photorhabdus spp...... 45 3.5. Utilization of entomopathogenic complex ...... 47 THE AIM OF THE THESIS ...... 48 4. CHARACTERIZATION OF LECTINS FROM PHOTORHABDUS LAUMONDII ...... Chyba! Záložka není definována. 4.1. Project overview ...... Chyba! Záložka není definována. 4.2 Novel lectins from P. laumondii ...... Chyba! Záložka není definována. 4.3. Materials and methods ...... Chyba! Záložka není definována. 4.3.1. Protein production ...... Chyba! Záložka není definována. 4.3.2. Protein purification ...... Chyba! Záložka není definována. 4.3.3. Glycan microarray ...... Chyba! Záložka není definována. 4.3.4. Isothermal titration calorimetry (ITC) ... Chyba! Záložka není definována. 4.3.5. Surface plasmon resonance (SPR) ...... Chyba! Záložka není definována. 4.3.6. Analytical ultracentrifugation (AUC) .... Chyba! Záložka není definována. 4.3.7. Crystallization ...... Chyba! Záložka není definována. 4.3.8. Data collection and structure determination ...... Chyba! Záložka není definována. 4.3.9 Reactive oxygen species (ROS) production in human blood ...... Chyba! Záložka není definována. 4.3.10. Phenoloxidase (PO) activity in haemolymph Chyba! Záložka není definována. 4.4. Results and discussion ...... Chyba! Záložka není definována. 4.4.1. Protein production and purification ...... Chyba! Záložka není definována. 4.4.2. Glycan microarray ...... Chyba! Záložka není definována. 4.4.3. Isothermal titration calorimetry (ITC) ... Chyba! Záložka není definována. 4.4.4. Surface plasmon resonance (SPR) ...... Chyba! Záložka není definována. 4.4.5. Analytical Ultracentrifugation (AUC) ... Chyba! Záložka není definována. 4.4.6. Crystallization and X-ray structure determination..... Chyba! Záložka není definována. 4.4.7. Reactive oxygen species (ROS) production in human blood ...... Chyba! Záložka není definována. 4.4.8. Phenoloxidase (PO) activity in insect haemolymph ... Chyba! Záložka není definována. 4.5. Conclusion ...... Chyba! Záložka není definována. 5. PREPARATION OF KNOCK-OUT MUTANTS OF PHOTORHABDUS LAUMONDII ...... Chyba! Záložka není definována. 5.1. Project overview ...... Chyba! Záložka není definována. 5.2. Materials and methods ...... Chyba! Záložka není definována. 5.2.1. Design of vector sequences and primers Chyba! Záložka není definována. 5.2.2. Transformation of E. coli XL1 cells ...... Chyba! Záložka není definována. 5.2.3. Molecular cloning ...... Chyba! Záložka není definována. 5.2.4. Transformation of E. coli S17-1 cells ... Chyba! Záložka není definována. 5.2.5. Gene knock-out via bacterial conjugation ...... Chyba! Záložka není definována. 5.2.6. Identification of knock-out mutants ...... Chyba! Záložka není definována. 5.2.7. Phenotype variant identification ...... Chyba! Záložka není definována. 5.3. Results and discussion ...... Chyba! Záložka není definována. 5.3.1. Design of vector sequences and primers Chyba! Záložka není definována. 5.3.1. Molecular cloning ...... Chyba! Záložka není definována. 5.3.2. Gene knock-out via bacterial conjugation ...... Chyba! Záložka není definována. 5.3.3. Colony PCR...... Chyba! Záložka není definována. 5.3.4. Phenotype variant identification ...... Chyba! Záložka není definována. 6. DEVELOPMENT OF SYNTHETIC MULTIVALENT INHIBITORS OF LECTINS ...... Chyba! Záložka není definována. 6.1. Project overview ...... Chyba! Záložka není definována.

6.2. Synthesis of α-L-fucoside-presenting glycoclusters and investigation of their interaction with Photorhabdus asymbiotica lectin (PHL) ...... Chyba! Záložka není definována. 6.3. Selectivity of original C-hexopyranosyl calix[4]arene conjugates towards lectins of different origin ...... Chyba! Záložka není definována. 6.4. Synthesis of β-D-galactopyranoside-presenting glycoclusters, investigation of their interactions with Pseudomonas aeruginosa lectin A (PA-IL) and evaluation of their anti-adhesion potential ...... Chyba! Záložka není definována. 7. OTHER PROJECT ...... Chyba! Záložka není definována. SCIENTIFIC PUBLICATIONS ...... 49 REFERENCES ...... 52 CURRICULUM VITAE ...... 71 APPENDIX ...... 76

1. LECTINS

1.1. History

Lectins are a group of saccharide recognizing proteins with a characteristic ability to agglutinate cells (e.g. erythrocytes). Although the term ”lectin” first appeared in the mid 20th century [1], the research on this topic started several decades earlier. The first mention of hemagglutination activity appeared in 1860 when Silas Weil Mitchel from the University of Philadelphia observed agglutination of the pigeon blood after mixing it with rattlesnake venom [2]. Nevertheless, the generally acknowledged start of “lectinology” was almost 30 years later, in 1888 at the University of Dorpat (now Tartu, Estonia). In his doctoral study (Fig. 1), Peter Hermann Stillmark isolated and described the first hemagglutinin – ricin, a toxic substance in the crude extract of the castor bean seeds (Ricinus communis) [3]. Because of its toxicity, there were some attempts to use ricin as a weapon during World War II. Fortunately, the ricin bomb tested by the British military had never been finished [4]. Subsequently, H. Helin, also from the Tartu University, described another toxic hemagglutinin called abrin found in the extract of the jequirity bean (Abrus precatorius) [4]. In the light of their hemagglutinating activity, these Figure 1: Frontispiece of the doctoral proteins were called hemagglutinins, or thesis of Peter Hermann Stillmark phytohemagglutinins, because they were found mostly in plants. The first pure hemagglutinin, concanavalin A, was isolated from jack bean (Canavalia ensiformis) by James B. Sumner at Cornell University, New York in 1919 [5]. Concanavalin A was reported to agglutinate erythrocytes and yeasts and it also precipitated glycogen from the solution. In 1936, James B. Sumner and Howell showed that the activity of concanavalin A can

21 be inhibited by sucrose [6]. This experiment was the first demonstration of the sugar specificity of hemagglutinins. The hemagglutination activity was thoroughly studied in the 1950s. Olavi Mäkelä, a doctoral student at the University of Helsinki, examined seed extracts from 743 plant species from Leguminosae family. He detected hemagglutination activity in more than one-third of them, and almost 10% showed blood type specificity [7]. Hemagglutinins played a crucial role in the investigation of ABO blood group antigens. Walter J. T. Morgan and Winifried M. Watkins at the Lister Institute in London found that hemagglutination of different blood groups can be inhibited by different monosaccharides [8], thus concluded the blood group antigens are determined by distinct sugar epitopes. This work was one of the first evidences for the presence of the saccharides on the cell surface and their potential role as the identity markers. The interaction of hemagglutinins with other cell types was also studied in the 1960s. Peter C. Nowell discovered that the lymphocyte mitosis can be stimulated upon binding of phytohemagglutinin (PHA) from red kidney bean (Phaseolus vulgaris) [9]. Several other lectins were proven to have mitogenic activity in a short time, among them also concanavalin A. An inhibition of the mitogenic activity of concanavalin A by monosaccharides, e.g. D-mannose, was of special importance [10]. This experiment proved that lymphocytes are stimulated to mitosis by the binding of agglutinins to the cell surface of glycans. This was one of the earliest demonstrations of a biological role of cell surface glycans. The name ”lectin” was proposed by Boyd and Shapleigh in a short article published in Science, 1954 [1]. The name originates from Latin legere, “to pick out” or “to choose”. It is based on the ability of plant agglutinins to distinguish among different blood groups. The name was later generalized to embrace all saccharide specific proteins of non-immune origin without catalytic activity. With the advances of the recombinant techniques in the 1970s, the studies of lectins were intensified. The major contribution to the development of the modern age of ”lectinology” was a work of Nathan Sharon and Halina Lis who purified multiple lectins [4]. Nathan Sharon devoted his work to lectin research and contributed tremendously to the current state of the art. The first lectin with a determined primary sequence and also the first lectin with a 3D structure solved by x-ray crystallography was concanavalin A in 1972 [11,12]

22

(Fig. 2A). The number of identified lectins has been continuously growing to this day. Today there are 536 distinct lectin sequences with solved 3D structure (Fig. 2B) [13]. Lectins have grown into a heterogeneous group of proteins with a ubiquitous prevalence in nature and with various roles in organisms, mediating the cell-cell communication at a molecular level. The progress in whole genome sequencing and modern bioinformatic tools allows us to accelerate searching for new potential lectins. [14]

Figure 2: A) The overall structure of concanavalin A (PDB: 3CNA). Individual monomers are color coded (green, red, orange and cyan). Divalent cations are depicted as spheres – Ca2+ green and Mn2+ violet. B) Distribution of solved lectin 3D structures in the nature according to Glyco3D database [14], last update 06.12.2019.

1.2. Basic characterization and classification

As was mentioned before, lectins are a group of proteins capable of binding saccharides. Lectins differ from other groups of carbohydrate recognizing proteins, e.g. carbohydrate-specific antibodies and enzymes, in several important aspects. In contrast to antibodies, lectins are not a product of the immune system and are present also in organisms incapable of conducting immune reaction (e.g. microorganisms). Lectins are structurally diverse, whereas antibodies are structurally similar. The high specificity of lectins makes them akin to enzymes, but in contrast to enzymes, lectins lack catalytic activity and they leave the ligand unaffected [15].

23

The lectin-saccharide interaction is mainly mediated via the formation of hydrogen bonds with sugar hydroxyl groups. However, other binding forces can be involved too. Hydrophobic interactions and van der Waals interactions often contribute to the binding [16]. The presence of divalent cation, usually Ca2+ or Mn2+, is required for saccharide binding for the whole group of leguminous lectins [17], animal C-type lectins [15] and in exceptional cases for bacterial lectins [18,19]. Recently, the importance of CH-π interactions between aromatic amino acids and the apolar part of carbohydrate molecules is becoming the center of interest [20–22]. Due to their exceptionally high saccharide specificity, lectins are a perfect tool to interpret the glyco code. The information about organism type, cell line and intracellular processes of the individual cell is reflected in the structure of oligosaccharides present on the surface of every cell [23]. The coding capacity of saccharides derives from variability in multiple sugar properties and their combination: a number and type of different monosaccharides, a monosaccharide linkage (usually three to four hydroxyls are available for covalent bond per monosaccharide), an anomeric position (α or β anomers) and a ring size (furanose or pyranose). Potential covalent modification (e.g. methylation, sulphation, phosphorylation) and the possibility of creating branched chains increase the diversity of possible formed molecules even further (Fig. 3) [17,24–26]. As a result, 3.55 x 104 unique tetrasaccharides can be created from four different monosaccharides, whereas only 24 distinct tetranucleotides can be formed from four bases. Classification of lectins has been a challenge and it has been difficult to find a mutual agreement. Therefore, lectins are divided according to multiple aspects. On the bases of their specificity, lectins can be sorted into 5 groups according to the monosaccharide they preferentially bind: D-mannose, L-fucose, D-galactose/N-acetyl-D-galactosamine, N-acetylneuraminic acid, N-acetyl-D-glucosamine [15,17]. Among all numerous saccharides present in nature, those five monosaccharides are typical constituents of eukaryotic cell surface glycosylation. However, this classification masks the fact that lectins often prefer oligosaccharides as their ligands. Lectin-monosaccharide interaction has usually weak affinity, with the dissociation constants in the millimolar range. However, the affinity towards oligosaccharides can be as much as 1000-fold higher. Moreover, lectins from the same

24

Figure 3: The structural variation of glycans is a result of differences in (a) anomeric stereochemistry; (b) variability of linkages; (c) ring size; and (d) covalent modification of hydroxyl groups [26]. monosaccharide specificity group may differ markedly in their affinities towards oligosaccharides [17]. According to their structural properties, lectins can be sorted into three classes: 1. simple, 2. mosaic (or multidomain) and 3. macromolecular assemblies. The simple lectins are composed of small numbers of subunits. This group covers almost all known plant lectins as well as animal galectins. The mosaic lectins are diverse in molecular weight and are composed of several kinds of domains, but only one of them includes a sugar-binding site. This group includes viral hemagglutinins as well as C-, P- and I- type animal lectins. Lectins from the macromolecular assemblies group are common in bacteria, where they form bacterial fimbriae or pili [17]. The most general classification of lectins is according to their origin and they are described in this chapter following this classification.

1.2.1. Microbial and viral lectins Microbial lectins are often considered to be the virulence factors of pathogenic microorganisms. They facilitate the pathogen adhesion on the host tissues (Fig. 4), which is one of the prerequisites of infection initiation [27]. Lectins involved in the microbe attachment to the host tissue can be present on the

25 surface of the bacterial cells in a form of fimbriae or pili. A typical fimbria consists of a major subunit forming the stem (shaft) and a minor subunit responsible for binding activity and sugar specificity. The fimbrial carbohydrate specificity varies among and also within bacterial species [28]. The most studied fimbriae are from the family Enterobacteriaceae. The mannose-specific type 1 fimbria with lectin FimH is produced by Escherichia coli, Klebsiella pneumoniae, Salmonella typhimurium and others [29]. Escherichia coli uses the type 1 fimbriae for adhesion to the urinary tract tissues and subsequently causes urinary tract infections. (UTIs) [30].

Figure 4: Schematic representation of lectin-mediated adhesion. Bacteria (ovals) and viruses (crystals) are adhering to the host cell via interaction with surface sugar epitopes (black dots).

In addition to fimbrial lectins, bacterial lectins can be located directly on the bacterial surface. Soluble surface lectins are transported to the cell surface by an often unknown mechanism but they are not actively excreted to the environment [31]. Examples of this type of lectins are BabA and SabA lectins from Helicobacter pylori. This bacterium is the causative agent of peptic ulcer and the lectins serve as adhesins to the gastric tissues. BabA is more involved in the initial stage of the infection, whereas SabA mediates the adhesion to the inflamed mucosa in the later stages of the infection [32–34]. Lectins PA-IL and PA-IIL from Pseudomonas aeruginosa [35,36], the human pathogen associated with cystic fibrosis, are responsible for

26 the bacterial attachment to the lung tissues and are involved in the creation of the biofilm [37,38]. The intriguing saccharide specificity of pathogen adhesins directly influences the host-pathogen specificity. Examples are the influenza viruses hemagglutinins (HA). HA of all the influenza strains recognize sialic acid but are extremely specific towards the linkage between sialic acid and galactose. Human strains of influenza recognize α2-6 linkage (Neu5Acα2–6Gal) that is expressed in human trachea. Avian and equine strains of influenza specifically recognize α2-3 linkage (Neu5Acα2–3Gal) which is present in horse trachea and avian intestine. Finally, porcine influenza strains are able to recognize both linkages and pig trachea expresses both saccharides [31,39]. Influenza HA is also the major immunogenic antigen and changes in this antigen contribute to the outbreaks of viral epidemics [31]. Another example of lectins facilitating the host specificity of pathogenic microbe is a bacterial strain Escherichia coli K99. This strain binds glycolipids containing N-glycolylneuraminic acid (Neu5Gc) which is expressed in piglet’s intestine. Therefore, the bacteria can cause lethal diarrhea to piglets, but they are not able to infect humans who lack N-glycolylneuraminic acid (Neu5Ac). Humans express N-acetylneuraminic acid, and even though the difference is only in one acyl group, Escherichia coli K99 is not able to recognize this glycan [17]. Another group of microbial lectins are secreted toxins. These lectins lack catalytic activity, but they form domains or subunits in the complex with enzymes involved in pathogenesis and they serve as recognition and targeting agents. A typical example of these types of proteins are members of the AB5 family. Proteins consist of one A subunit responsible for the toxic effect and five B subunits with the lectin activity. B subunit is responsible for binding to the cell surface receptors and the transport of the toxic A subunit to the cell [40]. AB5 toxins are employed by major bacterial pathogens causing several lethal human diseases such as whooping cough (pertussis toxin from Bordetella pertussis) [41], cholera (cholera toxin from Vibrio cholerae) [42] and copious watery diarrhea (heat-labile toxin from enterotoxigenic Escherichia coli) [43].

27

1.2.2. Plant lectins Plant lectins form a well-studied group consisting of several hundreds of lectins. Plant lectins are usually located in seeds, but they were isolated also from other plant tissues such as leaves, roots, fruits, and stems [17]. The function of plant lectins is intensively studied, but not yet thoroughly understood. The general theory proposes that plant lectins act as defense agents against phytopathogens (e.g. fungi, insects or invertebrates) [44,45]. The second theory assumes that lectins facilitate the symbioses between leguminous plants and nitrogen-fixing bacteria (mostly rhizobia) [45,46]. Recently it has been discovered that rhizobium-plant specificity is mediated by the interaction between rhizobial lipo-chitin oligosaccharides (Nod-factors) and LysM receptor kinase in the leguminous plant [47]. Nevertheless, gain-of- function experiments show that lectins are involved in the symbioses facilitating the bacterial attachment rather than directing the symbioses [45]. The largest family of plant lectins are lectins from leguminous plants. Leguminous lectins are mostly found in seeds. They are all structurally similar, with approx. 20 % of amino acids conserved in the sequence [17]. Residues involved in sugar binding are conserved (an asparagine, an aspartic acid, and an aromatic amino acid) and they occupy an identical spatial position in all these lectins. However, the sugar specificity of leguminous lectins differ. Discrimination between closely related saccharide ligands is achieved by structural variability of the unconserved amino acids lining and surrounding the binding pocket. The presence of Ca2+ or Mn2+ ion is essential for sugar binding [15]. Typical leguminous lectins are concanavalin A from jack bean (Canavalia ensiformis) and phytohemagglutinin (PHA) from red kidney bean (Phaseolus vulgaris). Another group of plant lectins is that from cereals. In contrast to leguminous lectins, lectins from cereals are exceptionally rich in cysteine and they do not require cations for saccharide binding. Cereal lectins are all specific to N-acetyl-D-glucosamine or N-acetylneuraminic acid. A typical example is a wheat germ agglutinin (WGA) [15]. Another plant lectin worth mentioning is ricin (Fig. 5) from beans of castor bean (Ricinus communis). Ricin is one of the deadliest poisons known. It consists of two domains linked by the S-S bridge. Globular domain B, specific to galactose and lactose, mediate ricin attachment to the cell surface.

28

Domain A possesses the cytotoxic activity by enzymatically inactivating the ribonucleic acid (RNA) involved in protein synthesis. According to some estimates, a single ricin molecule is enough to kill a cell [15].

Figure 5: Structure of ricin (PDB 2AAI). Chain A responsible for toxicity is colored green, chain B with lectin activity is colored red.

1.2.3. Animal lectins Animal lectins, including the human lectins, are a wide group of proteins with various functions in organisms. They are classified in several groups, each characterized by its own carbohydrate-binding domain (CRD) with highly conserved amino acids involved in saccharide binding. Galectins are a family of soluble lectins, specific to β-galactosides such as lactose or N-acetyllactosamine [15]. Interestingly, the tertiary structure of galectins may exhibit the jelly-roll fold typical for legume lectins without any significant sequence similarity between the two lectin families [48]. Galectins are found inside the cytoplasm and in the nucleus, occasionally on the cell surface. Their expression is regulated and they are believed to be essential for normal tissue development [17]. They also play a role in protein trafficking [49], inflammation [50], tumor metastasis [51] and apoptosis [52]. C-type lectins are the most widespread animal lectins with a broad range of biological functions. They require Ca2+ ions for binding. They are mosaic molecules in which CRD is attached to a variable number of protein domains.

29

The C-type lectins are divided into sixteen groups based on their structure and phylogenetic relationship [53]. The two most studied families are selectins and collectins. The selectins mediate the selective contact between leucocytes and endothelial cells through sialyl Lewis X recognition, thus mediating the migration of leukocytes to the site of infection [15]. The collectins are soluble lectins present in blood serum of mammals and birds. An important member of collectins is MBL – mannose-binding lectin which can activate the complement cascade – an important part of the innate immune system [54,55]. Pentraxins are evolutionary highly conserved proteins characterized by a multimeric (usually pentameric) structure. The classic examples are C-reactive protein (CRP) and serum amyloid P component (SAP). They are acute-phase proteins in humans produced by the liver in response to pro- inflammatory signals, most prominently interleukin 6 (IL-6) [56,57].

1.3. Lectin applications

Lectins stand out as important tools in various areas of research, medicine, and agriculture where their saccharide binding abilities and biological properties are utilized [15,58].

1.3.1. Research

Lectins are employed in detection, isolation, and characterization of glycoconjugates in the tissue sections, and on the cells and the subcellular organelles. For these purposes, lectins can be derivatized with a fluorescent dye, gold nano-particles or enzymes [15]. A novel platform emerging in recent years is lectin microarray (Fig. 6). A series of lectins (or carbohydrate specific antibodies) immobilized on a glass slide greatly facilitate the analysis of an extensive range of glycoconjugates present on the cell surface. Various procedures for rapid glycan profiling have been developed for glycan-related biomarkers connected to cancer or chronic diseases [59,60]. Lectins covalently bound to resin are used for purification and isolation of glycoproteins, glycopeptides, and oligosaccharides by affinity chromatography [15].

30

Figure 6: A basic concept of lectin microarray. Multiple lectins are immobilized on the glass slide, organized in a grid where one spot contains one lectin. Prelabelled glycoproteins or cells are then allowed to interact with lectins. Positive spots are identified by illumination under an appropriate scanner. Modified from [59].

1.3.2. Agriculture Numerous lectins are tested as tools for the development of the integrated insect pest control strategies due to their insecticidal activity. The insecticidal lectins are often used in the form of sprayings. The coding genes for such lectins can also be engineered into a variety of crops (e.g. wheat, rice, tobacco, and potatoes) to enhance the plant resistance against insect pests [61]. The example of lectins successfully used this way are Bt toxins from Bacillus thuringiensis. They act by binding to glycolipid receptors conserved among nematodes and insects, but absent in vertebrates, and generate pores to the host cell membrane [31]. Another example of lectin tested for these purposes is Orysata, a mannose-specific lectin found in rice seedlings (Orysum sativum) [62]. It was successfully expressed in the transgenic tobacco and its insecticidal activity against important pest insect was studied. The larvae of the beet armyworm (Spodoptera exigua) and the green peach aphid (Myzus persicae) fed on the transformed plants showed higher mortality compared to the larvae fed on the wild type plants [63]. Introducing transgenic plants does not seem to have any negative effect on the environment. Moreover, it is beneficial to the environment due to the lower consumption of chemical insecticides and herbicides [64]. Unfortunately, the occurrence of insects resistant against Bt-crops has been reported already [65].

31

1.3.3. Medicine The usage of lectins in medicine is heavily investigated and several strategies based on lectin properties have been developed. However, lectins are more frequently used in medical research and in vitro studies than in clinical practice. Numerous lectins display an anti-tumor activity and are tested as potential anti- tumor drugs. In in vitro studies, multiple lectins showed antiproliferative activity on the cancer cells or they elicit the cancer cell apoptosis [66]. From numerous examples we can mention the lectin from dark red kidney bean (Phaseolus vulgaris) with an antiproliferative activity towards leukemia L1210 cells [67], the lectin from Sophora flavescens which induces apoptosis in HeLa cell line (cervical cancer) [68] or BIL, a galactose-binding C-type lectin from Bothrops leucurus snake venom which induces death of melanoma cells [69]. Lectins are also tested for cancer therapy in vivo. A lectin from mistletoe (Viscum album) has been used to improve the quality of life in breast cancer patients [70], however, the results of clinical trials are highly inconsistent [71]. In order to improve the mistletoe lectin efficacy and use in clinical practice, an improved delivery system is being developed [72]. Lectins have been investigated as detection agents in cancer diagnosis due to their extremely high saccharide specificity. It is known that altered glycosylation occurs in cancer development and aberrantly expressed proteins are used as biomarkers of cancer development [60,73,74]. The diagnosis of hepatocellular carcinoma (HCC) is based on the detection of alpha-fetoprotein (AFP) in blood serum combined with ultrasonography. Elevation of AFP levels is associated not only with HCC but also with other types of cancer (stomach, pancreas or biliary tree), chronic liver disease or pregnancy [75]. In cases where the characteristic AFP blood levels overlap, the microheterogeneity of protein glycosylation is targeted to improve the diagnoses. The AFP-L3 isoform with the increased core fucosylation is related to hepatocellular carcinoma. This isoform is targeted by Lens culinaris agglutinin (LCA) isolated from the lentil seeds [74]. A commercial kit for hepatocellular carcinoma diagnosis based on LCA was developed [76] and it is widely used in the USA as a valuable alternative to more expensive diagnostics methods [77]. However, only two proteins, previously mentioned AFP and antigen CA15-3 specific for breast cancer, are clinically monitored for their glycan changes. All other cancer biomarkers are monitored for their total protein

32 levels. Recently, lectin microarrays are employed in the search for the new glycan-based cancer markers [60]. There have been attempts to develop a lectin-based drug delivery system over the past 40 years [78,79]. The rationale of the system is again based on the altered glycosylation of the diseased cells and the lectin ability to specifically target these unusual glycosylations. Lectins are used as carriers of the drug and allow a tissue-specific effect of the treatment. A tomato lectin was investigated as a potential intestinal bioadhesive agent which could slow down the intestinal transit of the oral drugs and increase their bioavailability. The tomato lectin was also proven to cross the intestinal mucosa in vitro [80]. The concept of lectin-mediated drug absorption enhancement can be applied also to other biological barriers, such as nasal mucosa, lungs or the blood-brain barrier [78]. Problems with toxicity and immunogenicity still need to be addressed, nevertheless, the lectin delivery system represents a promising way of drug administration. Several other clinical applications of lectins could be mentioned. The usage of a soybean agglutinin for the purging of human bone marrow for transplantation is one of them. Mature T cells present in the marrow are selectively agglutinated to prevent the lethal graft-versus-host reaction [15]. Mitogenic stimulation by lectins (e.g. concanavalin A or PHA) provides means to improve the immunocompetence of the immunocompromised patients, such as patients with AIDS or patients with immunosuppressive therapy [81]. Blood typing can be performed with the L-fucose specific lectins from Lotus tetragonolobus and Ulex europaeus which are able to select blood type-O cells [15]. Last but not least, the usage of lectins as a target in anti-adhesion therapy should be mentioned. This novel therapeutic approach focused on infections caused by the antibiotic-multiresistant bacterial strains will be discussed in a more detailed way in the next chapter.

33

2. ANTI-ADHESION THERAPY

2.1. Introduction

Since the discovery of penicillin in 1928 [82] and its usage in clinical practice since the 1940s, antibiotics have been the main way how to fight bacterial infections [83]. Unfortunately, the golden era of antibiotics is slowly coming to an end. Bacteria have found a way how to adapt to the new conditions and resistant bacterial strains are appearing and rapidly spreading. These strains are usually present in an environment with a frequent antibiotic administration, especially in hospitals, and nosocomial infections are becoming a serious problem of the 21st century. The situation can be demonstrated on the example of Staphylococcus aureus, an important human pathogen. S. aureus rapidly acquired resistance against novel types of antibiotics straight after they were introduced to the market (Fig. 7). Nowadays, methicillin-resistant strains of S. aureus (MRSA) are spread worldwide [84] and MRSA caused an estimated 80 400 cases of infection in the USA in 2011 [85]. For the MRSA strains, one effective antibiotic, glycopeptide vancomycin, remained. Despite careful administration of vancomycin as the antibiotic of the last choice. Clinical isolates of S. aureus with complete vancomycin resistance have already been reported [84,86,87].

Figure 7: Timeline of the advent of the antibiotic therapy and emergence of resistant strains of Staphylococcus aureus. PRSA = penicilin resistant Staphylococcus aureus, MRSA = methicillin resistant Staphylococcus aureus, VRSA = vancomycin resistant Staphlycoccus aureus. Modified from [84].

34

Due to the rise of the antibiotic-resistant bacterial strains, alternative approaches to treat bacterial infections are needed. The anti-adhesion therapy represents one of the promising ways how to deal with the multi-resistant bacterial strains.

2.2. Approaches to anti-adhesion therapy

The anti-adhesion therapy targets the attachment of the pathogen to the host cells or tissues, which is one of the first steps during the bacterial infection. Pathogens unable to adhere to the host tissues are restricted in growth and the formation of a protective biofilm is also prevented. This helps the natural clearance mechanisms of the body (such as urine flow in the urinary tract, airflow and mucociliary removal in the airways or shedding of upper epithelial cell layers) and the host immune system to eliminate the pathogen (Fig. 8) [88,89]. Another advantage is that the anti-adhesion therapy does not kill the pathogen, therefore a selective pressure is not created and the resistance to the anti-adhesives is not developed [90]. The most important adhesins expressed by numerous bacteria are lectins, present on the bacterial surface in the form of fimbriae/pili or as soluble surface lectins (see chapter 1.2.1.). Several strategies on how to interfere with lectin- mediated pathogen adhesion are being developed.

Figure 8: Schematic representation of the principle of the anti-adhesion therapy. Bacteria are adhering to the host cells via interaction with surface sugar epitopes (black dots). In the presence of inhibitor (red dots) bacteria lack free lectins and the adhesion is prevented.

35

2.2.1. Inhibition of pilus assembly The assembly of pili in Gram-negative bacteria is coordinated by the chaperone-usher pathway, a very well conserved assembly and secretion system [91]. Small organic molecules interfering with this system – so-called pilicides – are being developed [92]. The pilicide compounds were shown to bind on chaperons [93], strongly bind to uropathogenic Escherichia coli [94] and interfere with the pili assembly of the uropathogenic Escherichia coli strains in a dose-dependent manner [95].

2.2.2. Usage of adhesin analogs This strategy is based on the assumption that an isolated pathogen adhesin or its fragment binds to the host receptor and blocks the bacterial adhesion competitively. This approach needs to deal with potential toxicity and immunogenicity of the chosen adhesin [89]. Nevertheless, partial successes in experiments with Streptococcus mutans, the main cause of dental caries, were achieved. A synthetic 20-residue peptide based on the S. mutans adhesin was able to inhibit the S. mutans adhesion to immobilized salivary receptors in in vitro studies [96]. The re-colonization of the peptide pre-treated teeth with S. mutans was retarded. Nevertheless, this promising result needs to be treated carefully, because the adhesion of S. mutans may be mediated also by other adhesins [89].

2.2.3. Dietary supplements Empirical observations suggested that certain food may have a beneficial effect in the protection against bacterial infections. The well-known example of such nutrient is human maternal milk. The maternal milk is rich in oligosaccharides capable of the inhibition of various surface lectins of enteric bacteria and it was proven to protect breast-fed infants against diarrhoea [97–99]. Special attention is now paid to the fucosylated oligosaccharides (e.g. fucosyllactose) that are inhibitors of Campylobacter jejuni, the major cause of the diarrhoea worldwide [100]. Also, lactoferrin, a glycoprotein in breast milk, is able to inhibit the adhesion of several bacterial species (e.g. Actinobacillus actinomycetemcomitans, Prevotella intermedia or Escherichia coli) and thus contribute to the infant protection [101–103].

36

Another nutritive that is known to have an anti-adhesive effect is cranberry (Vaccinium macrocarpon). The beneficiary effect of drinking cranberry juice in the therapy of the urinary tract infections (UTIs) is long known folk wisdom. The scientific research on this topic dates to the 1960s [104,105]. Nowadays, the inhibition of biofilm formation and adhesion of uropathogenic Escherichia coli by urine of the person treated with cranberry juice was shown in vitro [106] and reduction of bacteriuria after drinking cranberry juice was proven in several small double-blinded placebo-controlled clinical studies [107,108]. The main active substances are proposed to be proanthocyanidins [109], which are now available in the synthetic form as a nutrition supplement to prevent the urinary tract infection. Despite some positive results, meta-analyses of data from 24 studies did not show any significant effect of the cranberry juice on the urinary tract infection compared to placebo. Only the cranberry products (pills) with a high content of active substances might have a positive effect, although further study is needed [110].

2.2.4. Carbohydrates as lectin inhibitors Usage of saccharides to block the lectin-mediated bacterial adhesion is inspired by the protective effect of oligosaccharides present in human maternal milk. There were some experiments with monosaccharides conducted and some of their results were promising. The positive effect of D-mannose on the therapy of the UTIs has been known since 1979 when the co-administration of methyl α-D-mannoside together with type 1 fimbriated Escherichia coli into the mice urinary bladder was tested. Methyl α-D-mannoside reduced the rate of mice urinary tract infections by two thirds, while methyl α-glucoside had no effect [111]. A more recent pilot study conducted in Rome in 2014 suggests that D-mannose can be effective support at acute UTIs in women [112]. The effect of other saccharides on ongoing infections was explored as well. A study with rhesus monkeys infected by Helicobacter pylori suggests that sialyl-3- lactose could be a safe anti-adhesive agent against H. pylori, albeit the study included only a small number of animals [113]. Also, a mixture of three monosaccharides (D-mannose, L-rhamnose, and D-galactose) showed a promising result in preventing the adhesion of Pseudomonas aeruginosa on canine corneocytes [114]. The last study mentioned was supported by a pharmaceutic company Virbac which included this patented saccharide

37 mixture in a commercial product Epi-otic III, used in veterinary medicine as prevention of otitis externa in dogs and cats. Despite the optimistic results of in vitro and in vivo studies, the usage of monosaccharides in the therapy of bacterial infections remains less effective compared to other active substances (e.g. antibiotics). This might be caused by the nature of lectin-monosaccharide interaction which is generally in the low- affinity range. An effective anti-adhesion therapy requires a high-affinity ligand. Therefore synthetic multivalent glycosides or glycomimetics are being developed where several copies of a monosaccharide are bound together on a synthetic scaffold [115]. Large series of multivalent glycoconjugates were designed in the last decades with different valencies and topological properties (Fig. 9). They can be divided into two distinct groups:

• Glycoclusters and glycodendrimers are limited in a number of epitopes (usually under 20), but they provide a systematic control over the number of sugar epitopes [115,116]. A large number of scaffolds have been developed as a synthetic core, such as fullerenes [117,118], calixarenes [119–121], porphyrins [122], cyclodextrins [123] or functionalized oligoethylene glycols [124]. On this core, a number of linker arms with one or more monosaccharides on the top is attached. Individual glycocompounds differ in type and number of bound monosaccharides, length of linker arms and the inner flexibility of the compound.

• Glycopolymers or glyconanoparticles can include a large number of sugar-epitopes, but with less control over their number [115,116]. Carbon nanotubes [125,126] or poly(p-phenylene ethylene) [127] can be used as the core of the linear glycopolymers. Compared to the linear polymers, glyconanoparticles display a globular shape. Besides a large number of sugar epitopes, they provide also variability in ligand density on the surface. Glyconanoparticles are in rapid development nowadays and encouraging results have been reported recently [128,129]. The examples of glyconanoparticle types are gold glyconanoparticles [130], silver glyconanoparticles [131], diamond glyconanoparticles [132], semiconductor glyco-quantum dots [133] and magnetic glyconanoparticles [134].

38

Figure 9: The example of different synthetic multivalent glycoconjugates [115].

2.2.5. Other approaches For the sake of completeness, other anti-adhesive therapy approaches that are not directly associated with lectins or carbohydrates should be mentioned. Somewhat controversial is the administration of sublethal doses of antibiotics. Some studies show this strategy reduces pathogen adherence to various substrates [135,136], whereas other studies report an increase in pathogen adhesion instead [137,138]. Nevertheless, this issue is important to study, because there is a high likelihood of periods of sublethal antibiotic concentration in the system of the antibiotic-treated patients due to the poor compliance in drug administration. The development of adhesin-based vaccines is a relatively new approach that uses a conserved region of adhesin as a vaccine agent [139,140]. The active means of anti-adhesin immunization faces the challenging need of increasing the mucosal IgA-mediated immunity, where active immunization, in general, promotes preferably systemic IgG- mediated immunity. Although the active immunization with adhesins in animal

39 models provides both IgG- and IgA-mediated protection (reviewed in [137]), the passive means of immunization may be more useful. The administration of antibodies in milk is known to protect suckling piglets and calves against diarrhea [141]. The use of probiotics as anti-adhesive agents is convenient and inexpensive. The probiotic bacterial strains compete with the pathogens for nutrients and life-space [142]. Healthy microflora is a natural body protection against infection, and it can be supported by healthy nutrition and administration of probiotic supplements.

2.3. Perspectives of anti-adhesion therapy

The prevalence of the antibiotic-resistant bacterial strains is rapidly increasing and the need for the alternative therapeutic approaches is more and more acute. An anti-adhesion therapy represents one of the promising strategies in how to cope with this threat. Despite the potential advantages and partial successes of the anti-adhesion strategy, it still serves the purpose of mostly being only a support for the treatment with other active substances. The major challenge in the improvement of the anti-adhesion therapy lies in the search for an effective agent to block bacterial adhesion. Such an agent needs to have high affinity towards a variety of bacterial adhesins with different specificities and it cannot be immunogenic, nor toxic. A better understanding of bacterial adhesins, their properties, specificities, and receptor-ligand interactions is needed to develop such compounds. Once these compounds become available, they may become an optional drug to treat multiple infectious diseases.

40

3. PHOTORHABDUS GENUS

3.1. History and taxonomy

The genus Photorhabdus embraces Gram-negative bacteria symbiotic with entomopathogenic nematodes (EPNs) from the family Heterorhabditidae. The first described bacterium living in symbiosis with (EPNs) was large, Gram-negative rod-shaped bacterium Achromobacter nematophilus, discovered by Gerard Thomas and Gorge Poinar in 1965 [143]. 14 years later the bacterium was transferred to a newly established genus Xenorhabdus in the family Enterobacteriaceae and it was renamed to Xenorhabdus nematophilus. Another member of Xenorhabdus genus was Xenorhabdus luminescens, a bacterium with a unique ability of [144]. Distinct differences between two bacteria, not only the bioluminescence but also the catalase activity, pigment production, DNA sequence and type of symbiont, led later to the establishment of a new genus, Photorhabdus (the name means “glowing rod”) [145]. More bacterial species symbiotic with Heterorhabditidae nematodes were assigned to the genus Photorhabdus, whereas bacteria symbiotic with the EPNs from the family Steirnernematidae belong to the genus Xenorhabdus. Recent comparative genomic analyses of the members of the order Enterobacteriales and phylogenic reconstruction of this order led to the establishment of six new families, and the genus Photorhabdus was assigned to the novel family [146]. The genus Photorhabdus consisted of four species with numerous subspecies [147–149] until the revisit of Photorhabdus phylogeny based on the whole- genome sequencing resulted in the elevation of numerous subspecies to the species level in 2018 [150]. Now, the genus consists of 19 species, which are listed in Table 1.

41

Table 1: Taxonomy of the genus Photorhabdus (ncbi.nlm.nih.gov). Four “original” species described before 2018 are highlighted in bold.

Superkingdom Bacteria Phylum Class Order Enterobacteriales Family Morganellaceae Genus Photorhabdus Species Photorhabdus arkhustii Photorhabdus asymbiotica Photorhabdus australis Photorhabdus bodei Photorhabdus caribbeanensis Photorhabdus cinerea Photorhabdus hainanensis Photorhabdus heterorhabditis Photorhabdus kayaii Photorhabdus khanii Photorhabdus kleinei Photorhabdus laumondii Photorhabdus luminescens Photorhabdus namnaonensis Photorhabdus noenieputensis Photorhabdus stackebrandtii Photorhabdus tasmaniensis Photorhabdus temperata Photorhabdus thracensis

42

3.2. Basic characterization

The genus Photorhabdus is formed by Gram-negative, highly motile, rod-shaped, facultatively anaerobic bacteria with the lengths from 2 to 10 µm [146]. All species of the Photorhabdus genus are pathogenic towards insect [151] and two species, P. asymbiotica, and P. australis, are also emerging human pathogens [152]. Bacteria cannot be found in the environment, as they cannot survive in soil on their own. Their natural habitat is the intestine of host nematode, or the tissues of the host during the infection stage of the life cycle (see chapter 3.3.). Photorhabdus spp. bacteria and Heterorhabditis spp. nematode form a highly effective entomopathogenic complex with each other. Two phenotypic variations of Photorhabdus bacteria have been described [153]. The primary variant (P1) is naturally present in the EPNs and can be isolated from them. The secondary variant (P2) can be generated by in vitro cultivation in low-nutrient media. The P2 variant differs from the P1 variant in metabolite production, it is still highly infective towards insect but it no longer supports the symbiosis with the nematode [151,154]. The interesting ability of bioluminescence is possible thanks to the lux operon. Around 30 other bacterial species are capable of bioluminescence, but they are all marine species, making the genus Photorhabdus the only known terrestrial bioluminescent bacteria [155]. Visible blue-green light (wavelength ~490 nm) is produced in a reaction catalyzed by the enzyme luciferase (Fig. 10) [155,156]. The biological purpose of the bioluminescence is still not clear. The three general hypotheses are: (i) it represents a signal within bacterial population or from bacteria to nematode to synchronize the symbiosis; (ii) it is a visual warning for nocturnal scavengers to avoid a cadaver, thus saving the tissues for Photorhabdus and the symbiotic nematode; or (iii) it attracts more insect to the site of the cadaver, and thus provides further prey. It could also serve as a sink for molecular oxygen, which could cause starvation of the host immune cells [151].

43

Figure 10: A scheme of the bacterial luciferase reaction. A long-chain fatty aldehyde (RCHO) is converted into the corresponding fatty acid (RCOOH) in the presence of

reduced flavin mononucleotide (FMNH2) and molecular oxygen (O2). Besides

RCOOH, water (H2O), oxidized flavin mononucleotide (FMN) and the light (hν) are being produced.

Photorhabdus spp. is highly pathogenic towards insect hosts. Only 50-200 bacterial cells are sufficient to cause lethal septicemia in insect prey [151]. This fact was supported by the whole genome sequencing of P. laumonidii subsp. laumnodii TT01, where more toxin genes than in any other sequenced bacterial genome were predicted [157]. Produced toxins and other metabolites (e.g. adhesins, hemolysins, proteases and antibiotics) serve the bacteria as virulence factors to overcome the host defense during the infection and also prevent other bacterial species from proliferating in the host tissues, thus saving the tissues for the symbiotic nematode [158].

3.3. Life cycle

The life cycle of the entomopathogenic Photorhabdus-Heterorhabditis complex (Fig. 11) is described very well [151,158–160]. In the soil bacteria can be found only inside the intestine of an infective juvenile (IJ), a specialized larval stage of the nematode. This larval stage has a non-functional digestive tract and it does not ingest. IJs actively search for the insect prey and when they find it, they penetrate the host body through natural openings (mouth, anus or spiracles). Unlike other nematodes, IJs of Heterorhabditis bacteriophora can enter the host body also actively, thanks to a tooth-like cuticle protuberance that can be used to disrupt the insect cuticle [161]. After entering the insect’s body, IJs regurgitate the bacteria into the insect hemocoel. The bacteria rapidly proliferate, produce toxins and kill the prey within 48 hours [151]. Meanwhile, the IJs undergo the process of recovery and starts to evolve into the adult [162]. The insect cadaver is bioconverted into the nutrients and protected from other scavengers by Photorhabdus, while Heterorhabditis nematodes feast on the

44

Figure 11: Schematic representation of the life cycle of the entomopathogenic Photorhabdus-Heterorhabditis complex. L1-L4 stand for larval stages of the nematode, IJ stands for infective juvenile. insect tissues as well as bacterial biomass and undergo one to three cycles of sexual reproduction. Once the host tissues are depleted, the sexual reproduction of the nematode is disrupted and new infective juveniles are produced inside the maternal uterus during a process called endotokia matricida [163]. IJs are re-associated with the bacteria and leave the cadaver to search for new insect prey.

3.4. Lectins from Photorhabdus spp.

Lectins, in general, are involved in various processes including cell to cell communication [15,27]. Lectins produced by entomopathogens with a complex life cycle like the genus Photorhabdus could play a role in three basic processes: (i) the interaction with host tissues, (ii) the interaction with nematode symbionts and (iii) the interactions within the bacterial population. Several lectins were identified and described in the genome of Photorhabdus.

45

The first described lectin from the genus Photorhabdus was lectin PLL from Photorhabdus luminescens [164] (now P. laumondii). Its close homolog PHL was described shortly after in Photorhabdus asymbiotica [165]. Both lectins recognize L-fucose and unusual O-methylated disaccharide 3,6-O-Me2-D-Glcβ1-4(2,3-O-Me2)-L-Rhaα present in the Mycobacterium leprae glycolipid PGL-1 [166,167]. Proteins share the monomeric fold, a seven-bladed β-propeller, but they differ in the oligomeric state, PLL being a tetramer [164] and PHL a dimer [165]. 7 potential fucose-binding sites were described in PLL, 3 of them were occupied in the crystal structure of PLL in the complex with L-fucose [164]. Besides the set of 7 potential fucose- binding sites, another set of binding sites was discovered in PHL. This second set of binding sites recognized D-galactose [165]. The proposed function of these lectins is to help the bacteria to overcome the insect defense by interfering with the insect immune system. Both proteins were shown to bind insect hemocytes and, moreover, PHL affected the production of reactive oxygen species and phenoloxidase activity – two ways of insect primary defense against pathogens [164,165]. Both bacterial species, P. laumondii and P. asymbiotica, code other potential lectins homologous to the PLL and PHL which are being investigated right now. The characterization of lectin homologs of PLL is included in the practical part of this thesis. The reason for having multiple homologous proteins in the genome as well as the possibility of lectin cooperation with each other remains unclear. Another lectin described in P. laumondii is PllA [168]. This protein is not structurally related to PLL nor PHL, but it is a homolog of the PA-IL (LecA) lectin from Pseudomonas aeruginosa with a 37% identity. The lectin was shown to recognize α-galactoside terminated glycocompounds. This property can be used for the identification of the α-Gal epitopes on the porcine cells, responsible for hyperacute rejection of pig to primate organ xenotransplant [169]. The biological function of this lectin in Photorhabdus life cycle remains unclear, too.

46

3.5. Utilization of entomopathogenic complex

The high toxicity for insects makes EPNs a promising agent in the biological control of insect pests in agriculture [170]. Mostly, the two genera (Steinernematidae and Heterorhabditidae) with their symbiotic bacteria (Xenorhabdus and Photorhabdus species, respectively) are being studied for this purpose. The long-term perspective is to use EPNs as a replacement of pesticides in agriculture. Unfortunately, promising results from the laboratory do not always translate into success in the field [171,172] and there are still several barriers preventing expansion to massive agricultural use. The main challenges represent the high cost of EPNs production, difficult optimization of the EPNs mass production and sub-optimal efficacy of transmission to the insect pest [173,174]. Prior to the intensive introduction of EPN to the environment, its host specificity also needs to be considered. To avoid unintentional damage in the population of beneficial soil insects, nematode species with a narrow range of insect hosts are being selected. Nevertheless, EPNs were successfully produced in smaller amounts and commercial products are already available worldwide [170,175,176]. These products achieve great success in the protection of smaller areas such as gardens or greenhouses.

47

THE AIM OF THE THESIS

This thesis is composed of three projects. It deals with bacterial lectins, their functional and structural characterization (project 1); deciphering their role in the bacterial life cycle (projects 1 and 2); and development of their inhibitors (project 3). The main focus of the presented work is on the novel lectins discovered in the bacterium Photorhabdus laumondii, an effective insect pathogen. The goals of the individual projects, in brief, are:

Project 1: Characterization of lectins from Photorhabdus laumondii. Characterization of newly identified hypothetical lectins P. laumondii – PLL2, PLL3, PLL4, and PLL5, which includes:

• Production of lectins in a recombinant form. • Functional analysis - specificity and affinity (glycan microarray, surface plasmon resonance, and isothermal titration calorimetry). • Determining the structure (analytical ultracentrifugation and X-ray crystallography) • Revealing lectins influence on the host innate immune system (phenoloxidase activity, production of reactive oxygen species)

Project 2: Preparation of knock-out mutants of Photorhabdus laumondii

• Preparation of a library of P. laumondii knock-out mutants with deleted genes for selected lectins – PLL, PLL2, PLL3, PLL4, PLL5, and PLU1 - in various combinations.

Project 3: Development of synthetic multivalent inhibitors of lectins

• Test the ability of selected multivalent synthetic glycocompounds to interact with a complex environment of the bacterial surface.

48

SCIENTIFIC PUBLICATIONS

Manuscript 1 (Appendix I)

Eva Fujdiarová, Josef Houser, Pavel Dobeš, Gita Paulíková, Nikolay Kondakov, Leonid Kononov, Pavel Hyršl, Michaela Wimmerová. Heptabladded β-propeller lectins PLL2 and PHL from Photorhabdus spp. recognize O-methylated sugars and influence the host immune system. Submitted to FEBS Journal, after 1st review, in revision. Author’s contribution Manuscript writing, investigation (protein expression and purification, glycan microarray, ITC, SPR, crystallization).

Article 1 (Appendix II)

Lukáš Faltinek*, Eva Fujdiarová*, Filip Melicher, Josef Houser, Martina Kašáková, Nikolay Kondakov, Leonid Kononov, Kamil Parkan, Sébastien Vidal, Michaela Wimmerová. Lectin PLL3, a novel monomeric member of seven-bladed β-propeller lectin family. * These authors contributed equally Molecules, 2019, 24, 4540 DOI: 10.3390/molecules24244540 Author’s contribution Manuscript writing, investigation (protein expression and purification, glycan microarray, ITC, SPR).

49

Article 2 (Appendix III)

Gita Jančaříková, Mihály Herczeg, Eva Fujdiarová, Josef Houser, Katalin E. Köver, Anikó Borbás, Michaela Wimmerová, and Magdolna Csávás.

Synthesis of α-L-fucoside-presenting glycoclusters and investigation of their interaction with Photorhabdus asymbiotica lectin (PHL). Chemistry: A European Journal, 2018, 24(16), 4055-4068 DOI: 10.1002/chem.201705853 Author’s contribution Investigation (P. asymbiotica cross-linking experiments), manuscript writing (P. asymbiotica growth, cell aggregation assay).

Article 3 (Appendix IV)

Martina Kašáková, Lenka Malinovská, Tomáš Klejch, Martina Hlaváčová, Hana Dvořáková, Eva Fujdiarová, Zdeňka Rottnerová, Olga Maťátková, Pavel Lhoták, Michaela Wimmerová, Jitka Moravcová. Selectivity of original C-hexopyranosyl calix[4]arene conjugates towards lectins of different origin. Carbohydrate Research, 2018, 469, 60-72. DOI: 10.1016/j.carres.2018.08.012 Author’s contribution Investigation (B. cenocepacia cross-linking experiments).

50

Article 4 (Appendix V)

Lenka Malinovská, Son Thai Le, Mihály Herczeg, Michaela Vašková, Josef Houser, Eva Fujdiarová, Jan Komárek, Petr Hodek, Anikó Borbás, Michaela Wimmerová, Magdolna Csávás.

Synthesis of β-D-galactopyranoside-presenting glycoclusters, investigation of their interactions with Pseudomonas aeruginosa lectin A (PA-IL) and evaluation of their anti-adhesion potential. Biomolecules, 2019, 9(11). DOI: 10.3390/biom9110686. Author’s contribution Investigation (P. aeruginosa cross-linking experiments).

Article 5 (Appendix VI)

Petra Sýkorová, Jitka Novotná, Gabriel Demo, Guillaume Pompidor, Eva Dubská, Jan Komárek, Eva Fujdiarová, Josef Houser, Lucia Hároníková, Annabelle Varrot, Nadezhda Shilova, Anne Imberty, Nicolai Bovin, Martina Pokorná, Michaela Wimmerová. Characterization of novel lectins from Burkholderia pseudomallei and Chromobacterium violaceum with seven-bladed β-propeller fold. International Journal of Biological Macromolecules, 2019, in press. DOI: 10.1016/j.ijbiomac.2019.10.200 Author’s contribution Investigation (glycan microarray experiments), manuscript writing (P. aeruginosa cross-linking assay).

51

REFERENCES

1. Boyd WC, Shapleigh E. Specific Precipitating Activity of Plant Agglutinins (Lectins). Science. 1954;119: 419–419. doi:10.1126/science.119.3091.419 2. Mitchell SW. Researches upon the venom of the rattlesnake : with an investigation of the anatomy and physiology of the organs concerned / By S. Weir Mitchell. Washington : Smithsonian Institution,; 1860. doi:10.5962/bhl.title.45454 3. Stillmark H. Über Ricin, ein giftiges Ferment aus den Samen von Ricinus comm. L. und einigen anderen Euphorbiaceen. 1888. Available: http://hdl.handle.net/10062/2332 4. Sharon N, Lis, H. History of lectins: from hemagglutinins to biological recognition molecules. Glycobiology. 2004;14: 53R-62R. doi:10.1093/glycob/cwh122 5. Sumner JB. The globulins of jack bean, Canavalia ensiformis. J Biol Chem. 1919;37: 137–142. 6. Sumner JB, Howell SF. Identification of Hemagglutinin of Jack Bean with Concanavalin A. J Bacteriol. 1936;32: 227–237. 7. Makela O. Studies in hemagglutinins of leguminosae seeds. Ann Med Exp Biol Fenn. 1957;35: 1–133. 8. Morgan WTJ, Watkins WM. The inhibition of the haemagglutinins in plant seeds by human blood group substances and simple sugars. Br J Exp Pathol. 1953;34: 94–103. 9. Nowell PC. Phytohemagglutinin: an initiator of mitosis in cultures of normal human leukocytes. Cancer Res. 1960;20: 462–466. 10. Powell AE, Leon MA. Reversible interaction of human lymphocytes with the mitogen concanavalin A. Exp Cell Res. 1970;62: 315–325. 11. Edelman GM, Cunningham BA, Reeke GN, Becker JW, Waxdal MJ, Wang JL. The covalent and three-dimensional structure of concanavalin A. Proc Natl Acad Sci USA. 1972;69: 2580–2584. 12. Hardman KD, Ainsworth CF. Structure of concanavalin A at 2.4-Ang resolution. Biochemistry. 1972;11: 4910–4919. doi:10.1021/bi00776a006 13. Bonnardel F, Mariethoz J, Salentin S, Robin X, Schroeder M, Perez S, et al. UniLectin3D, a database of carbohydrate binding proteins with curated information on 3D structures and interacting ligands. Nucleic Acids Research. 2019;47: D1236–D1244. doi:10.1093/nar/gky832 14. Pérez S, Sarkar A, Rivet A, Breton C, Imberty A. Glyco3D: A Portal for Structural Glycosciences. In: Lütteke T, Frank M, editors. Glycoinformatics. New York, NY: Springer New York; 2015. pp. 241–258. doi:10.1007/978-1- 4939-2343-4_18

52

15. Sharon N. Lectins. In: John Wiley & Sons, Ltd, editor. eLS. Chichester, UK: John Wiley & Sons, Ltd; 2009. p. a0000708.pub2. doi:10.1002/9780470015902.a0000708.pub2 16. Komath SS, Kavitha M, Swamy MJ. Beyond carbohydrate binding: new directions in plant lectin research. Org Biomol Chem. 2006;4: 973. doi:10.1039/b515446d 17. Lis H, Sharon N. Lectins: Carbohydrate-Specific Proteins That Mediate Cellular Recognition. Chem Rev. 1998;98: 637–674. 18. Cioci G, Mitchell EP, Gautier C, Wimmerová M, Sudakevitz D, Pérez S, et al. Structural basis of calcium and galactose recognition by the lectin PA-IL of Pseudomonas aeruginosa. FEBS Letters. 2003;555: 297–301. doi:10.1016/S0014-5793(03)01249-3 19. Mitchell EP, Sabin C, Šnajdrová L, Pokorná M, Perret S, Gautier C, et al. High affinity fucose binding of Pseudomonas aeruginosa lectin PA-IIL: 1.0 Å resolution crystal structure of the complex combined with thermodynamics and computational chemistry approaches. Proteins. 2004;58: 735–746. doi:10.1002/prot.20330 20. Wimmerová M, Kozmon S, Nečasová I, Mishra SK, Komárek J, Koča J. Stacking Interactions between Carbohydrate and Protein Quantified by Combination of Theoretical and Experimental Methods. Srinivasan N, editor. PLoS ONE. 2012;7: e46032. doi:10.1371/journal.pone.0046032 21. Hudson KL, Bartlett GJ, Diehl RC, Agirre J, Gallagher T, Kiessling LL, et al. Carbohydrate–Aromatic Interactions in Proteins. J Am Chem Soc. 2015;137: 15152–15160. doi:10.1021/jacs.5b08424 22. Spiwok V. CH/π Interactions in Carbohydrate Recognition. Molecules. 2017;22. doi:10.3390/molecules22071038 23. Gabius H-J, Roth J. An introduction to the sugar code. Histochem Cell Biol. 2017;147: 111–117. doi:10.1007/s00418-016-1521-9 24. Gabius H-J, Siebert H-C, André S, Jiménez-Barbero J, Rüdiger H. Chemical Biology of the Sugar Code. ChemBioChem. 2004;5: 740–764. doi:10.1002/cbic.200300753 25. Gabius H-J. Glycans: bioactive signals decoded by lectins. Biochm Soc Trans. 2008;36: 1491–1496. doi:10.1042/BST0361491 26. Hevey R. Strategies for the Development of Glycomimetic Drug Candidates. Pharmaceuticals. 2019;12: 55. doi:10.3390/ph12020055 27. Imberty A, Varrot A. Microbial recognition of human cell surface glycoconjugates. Current Opinion in Structural Biology. 2008;18: 567–576. doi:10.1016/j.sbi.2008.08.001 28. Soto GE, Hultgren SJ. Bacterial adhesins: common themes and variations in architecture and assembly. J Bacteriol. 1999;181: 1059–1071.

53

29. Sharon N. Bacterial lectins, cell-cell recognition and infectious disease. FEBS Lett. 1987;217: 145–157. 30. Edén CS, Hansson HA. Escherichia coli pili as possible mediators of attachment to human urinary tract epithelial cells. Infect Immun. 1978;21: 229–237. 31. Nizet V, Varki A, Aebi M. Microbial Lectins: Hemagglutinins, Adhesins, and Toxins. 3rd ed. In: Varki A, Cummings RD, Esko JD, Stanley P, Hart GW, Aebi M, et al., editors. Essentials of Glycobiology. 3rd ed. Cold Spring Harbor (NY): Cold Spring Harbor Laboratory Press; 2015. Available: http://www.ncbi.nlm.nih.gov/books/NBK453032/ 32. Magalhães A, Reis CA. Helicobacter pylori adhesion to gastric epithelial cells is mediated by glycan receptors. Braz J Med Biol Res. 2010;43: 611–618. doi:10.1590/S0100-879X2010007500049 33. Ansari S, Yamaoka Y. Helicobacter pylori BabA in adaptation for gastric colonization. WJG. 2017;23: 4158. doi:10.3748/wjg.v23.i23.4158 34. Benktander J, Barone A, Johansson MM, Teneberg S. Helicobacter pylori SabA binding gangliosides of human stomach. Virulence. 2018;9: 738–751. doi:10.1080/21505594.2018.1440171 35. Gilboa-Garber N. Pseudomonas aeruginosa lectins. Meth Enzymol. 1982;83: 378–385. 36. Imberty A, wimmerová M, Mitchell EP, Gilboa-Garber N. Structures of the lectins from Pseudomonas aeruginosa: insight into the molecular basis for host glycan recognition. Microbes Infect. 2004;6: 221–228. 37. Diggle SP, Stacey RE, Dodd C, Camara M, Williams P, Winzer K. The galactophilic lectin, LecA, contributes to biofilm development in Pseudomonas aeruginosa. Environ Microbiol. 2006;8: 1095–1104. doi:10.1111/j.1462-2920.2006.001001.x 38. Tielker D. Pseudomonas aeruginosa lectin LecB is located in the outer membrane and is involved in biofilm formation. Microbiology. 2005;151: 1313–1323. doi:10.1099/mic.0.27701-0 39. Suzuki Y. Sialobiology of Influenza: Molecular Mechanism of Host Range Variation of Influenza Viruses. Biological & Pharmaceutical Bulletin. 2005;28: 399–408. doi:10.1248/bpb.28.399 40. Beddoe T, Paton AW, Le Nours J, Rossjohn J, Paton JC. Structure, biological functions and applications of the AB5 toxins. Trends in Biochemical Sciences. 2010;35: 411–418. doi:10.1016/j.tibs.2010.02.003 41. Zlamy M. Rediscovering Pertussis. Front Pediatr. 2016;4. doi:10.3389/fped.2016.00052 42. Chinnapen DJ-F, Chinnapen H, Saslowsky D, Lencer WI. Rafting with cholera toxin: endocytosis and trafficking from plasma membrane to ER.

54

FEMS Microbiol Lett. 2007;266: 129–137. doi:10.1111/j.1574- 6968.2006.00545.x 43. Turner SM, Scott-Tucker A, Cooper LM, Henderson IR. Weapons of mass destruction: virulence factors of the global killer Enterotoxigenic Escherichia coli. FEMS Microbiology Letters. 2006;263: 10–20. doi:10.1111/j.1574- 6968.2006.00401.x 44. Murdock LL, Shade RE. Lectins and protease inhibitors as plant defenses against insects. J Agric Food Chem. 2002;50: 6605–6611. 45. De Hoff PL, Brill LM, Hirsch AM. Plant lectins: the ties that bind in root symbiosis and plant defense. Mol Genet Genomics. 2009;282: 1–15. doi:10.1007/s00438-009-0460-8 46. Sharon N, Lis H. Lectins as cell recognition molecules. Science. 1989;246: 227–234. 47. Oldroyd GED, Downie JA. Coordinating Nodule Morphogenesis with Rhizobial Infection in Legumes. Annu Rev Plant Biol. 2008;59: 519–546. doi:10.1146/annurev.arplant.59.032607.092839 48. Lobsanov YD, Gitt MA, Leffler H, Barondes SH, Rini JM. X-ray crystal structure of the human dimeric S-Lac lectin, L-14-II, in complex with lactose at 2.9-A resolution. J Biol Chem. 1993;268: 27034–27038. doi:10.2210/pdb1hlc/pdb 49. Delacour D, Koch A, Jacob R. The role of galectins in protein trafficking. Traffic. 2009;10: 1405–1413. doi:10.1111/j.1600-0854.2009.00960.x 50. Brinchmann MF, Patel DM, Iversen MH. The Role of Galectins as Modulators of Metabolism and Inflammation. Mediators of Inflammation. 2018;2018: 1–11. doi:10.1155/2018/9186940 51. Danguy A, Camby I, Kiss R. Galectins and cancer. Biochim Biophys Acta. 2002;1572: 285–293. 52. Hsu DK, Yang R-Y, Liu F-T. Galectins in apoptosis. Meth Enzymol. 2006;417: 256–273. doi:10.1016/S0076-6879(06)17018-4 53. Cummings RD, McEver RP. C-Type Lectins. 3rd ed. In: Varki A, Cummings RD, Esko JD, Stanley P, Hart GW, Aebi M, et al., editors. Essentials of Glycobiology. 3rd ed. Cold Spring Harbor (NY): Cold Spring Harbor Laboratory Press; 2015. Available: http://www.ncbi.nlm.nih.gov/books/NBK453028/ 54. Garred P. Mannose-binding lectin genetics: from A to Z: Table 1. Biochm Soc Trans. 2008;36: 1461–1466. doi:10.1042/BST0361461 55. Turner MW. The role of mannose-binding lectin in health and disease. Mol Immunol. 2003;40: 423–429. 56. Bottazzi B, Garlanda C, Salvatori G, Jeannin P, Manfredi A, Mantovani A. Pentraxins as a key component of innate immunity. Current Opinion in Immunology. 2006;18: 10–15. doi:10.1016/j.coi.2005.11.009

55

57. Gabius HJ. Animal lectins. Eur J Biochem. 1997;243: 543–576. 58. Lagarda-Diaz I, Guzman-Partida A, Vazquez-Moreno L. Legume Lectins: Proteins with Diverse Applications. IJMS. 2017;18: 1242. doi:10.3390/ijms18061242 59. Hirabayashi J, Yamada M, Kuno A, Tateno H. Lectin microarrays: concept, principle and applications. Chem Soc Rev. 2013;42: 4443–4458. doi:10.1039/c3cs35419a 60. Hashim OH, Jayapalan JJ, Lee C-S. Lectins: an effective tool for screening of potential cancer biomarkers. PeerJ. 2017;5: e3784. doi:10.7717/peerj.3784 61. Macedo MLR, Oliveira CFR, Oliveira CT. Insecticidal activity of plant lectins and potential application in crop protection. Molecules. 2015;20: 2014– 2033. doi:10.3390/molecules20022014 62. Peumans WJ, Barre A, Houles Astoul C, Rovira P, Roug P, Proost P, et al. Isolation and characterization of a jacalin-related mannose-binding lectin from salt-stressed rice (Oryza sativa) plants. Planta. 2000;210: 970–978. doi:10.1007/s004250050705 63. Al Atalah B, Smagghe G, Van Damme EJM. Orysata, a jacalin-related lectin from rice, could protect plants against biting-chewing and piercing-sucking insects. Plant Science. 2014;221–222: 21–28. doi:10.1016/j.plantsci.2014.01.010 64. Kumar S, Chandra A, Pandey KC. Bacillus thuringiensis (Bt) transgenic crop: an environment friendly insect-pest management strategy. J Environ Biol. 2008;29: 641–653. 65. Tabashnik BE, Carrière Y, Dennehy TJ, Morin S, Sisterson MS, Roush RT, et al. Insect Resistance to Transgenic Bt Crops: Lessons from the Laboratory and Field. Journal of Economic Entomology. 2003;96: 1031–1038. doi:10.1093/jee/96.4.1031 66. Lam SK, Ng TB. Lectins: production and practical applications. Appl Microbiol Biotechnol. 2011;89: 45–55. doi:10.1007/s00253-010-2892-9 67. Xia L, Ng TB. A hemagglutinin with mitogenic activity from dark red kidney beans. Journal of Chromatography B. 2006;844: 213–216. doi:10.1016/j.jchromb.2006.07.042 68. Liu Z, Liu B, Zhang Z-T, Zhou T-T, Bian H-J, Min M-W, et al. A mannose- binding lectin from Sophora flavescens induces apoptosis in HeLa cells. Phytomedicine. 2008;15: 867–875. doi:10.1016/j.phymed.2008.02.025 69. Aranda-Souza MA, Rossato FA, Costa RAP, Figueira TR, Castilho RF, Guarniere MC, et al. A lectin from Bothrops leucurus snake venom raises cytosolic calcium levels and promotes B16-F10 melanoma necrotic cell death via mitochondrial permeability transition. Toxicon. 2014;82: 97–103. doi:10.1016/j.toxicon.2014.02.018

56

70. Semiglazov VF, Stepula VV, Dudov A, Schnitker J, Mengs U. Quality of life is improved in breast cancer patients by Standardised Mistletoe Extract PS76A2 during chemotherapy and follow-up: a randomised, placebo- controlled, double-blind, multicentre clinical trial. Anticancer Res. 2006;26: 1519–1529. 71. Ernst E, Schmidt K, Steuer-Vogt MK. Mistletoe for cancer?: A systematic review of randomised clinical trials. Int J Cancer. 2003;107: 262–267. doi:10.1002/ijc.11386 72. Lyu S-Y, Kwon Y-J, Joo H-J, Park W-B. Preparation of alginate/chitosan microcapsules and enteric coated granules of mistletoe lectin. Arch Pharm Res. 2004;27: 118–126. doi:10.1007/BF02980057 73. Varki A, Kannagi R, Toole B, Stanley P. Glycosylation Changes in Cancer. 3rd ed. In: Varki A, Cummings RD, Esko JD, Stanley P, Hart GW, Aebi M, et al., editors. Essentials of Glycobiology. 3rd ed. Cold Spring Harbor (NY): Cold Spring Harbor Laboratory Press; 2015. Available: http://www.ncbi.nlm.nih.gov/books/NBK453023/ 74. S. Coulibaly F, C. Youan B-B. Current status of lectin-based cancer diagnosis and therapy. AIMS Molecular Science. 2017;4: 1–27. doi:10.3934/molsci.2017.1.1 75. Bialecki ES, Di Bisceglie AM. Diagnosis of hepatocellular carcinoma. HPB. 2005;7: 26–34. doi:10.1080/13651820410024049 76. Shimizu K, Taniichi T, Satomura S, Matsuura S, Taga H, Taketa K. Establishment of assay kits for the determination of microheterogeneities of alpha-fetoprotein using lectin-affinity electrophoresis. Clin Chim Acta. 1993;214: 3–12. 77. Leerapun A, Suravarapu SV, Bida JP, Clark RJ, Sanders EL, Mettler TA, et al. The Utility of Lens Culinaris Agglutinin-Reactive α-Fetoprotein in the Diagnosis of Hepatocellular Carcinoma: Evaluation in a United States Referral Population. Clinical Gastroenterology and Hepatology. 2007;5: 394–402. doi:10.1016/j.cgh.2006.12.005 78. Bies C, Lehr C-M, Woodley JF. Lectin-mediated drug targeting: history and applications. Advanced Drug Delivery Reviews. 2004;56: 425–435. doi:10.1016/j.addr.2003.10.030 79. Lehr CM. Lectin-mediated drug delivery: the second generation of bioadhesives. J Control Release. 2000;65: 19–29. 80. Naisbett B, Woodley J. The potential use of tomato lectin for oral drug delivery. 1. Lectin binding to rat small intestine in vitro. International Journal of Pharmaceutics. 1994;107: 223–230. doi:10.1016/0378-5173(94)90438-3 81. Carvalho EVMM, Oliveira WF, Coelho LCBB, Correia MTS. Lectins as mitosis stimulating factors: Briefly reviewed. Life Sciences. 2018;207: 152– 157. doi:10.1016/j.lfs.2018.06.003

57

82. Fleming A. On the antibacterial action of cultures of a Penicillium, with special reference to their use in the isolation of B.influenzae. Br J Exp Pathol. 1929;10: 226–36. 83. Gaynes R. The Discovery of Penicillin—New Insights After More Than 75 Years of Clinical Use. Emerg Infect Dis. 2017;23: 849–853. doi:10.3201/eid2305.161556 84. McGuinness WA, Malachowa N, DeLeo FR. Vancomycin Resistance in Staphylococcus aureus. Yale J Biol Med. 2017;90: 269–281. 85. Dantes R. National Burden of Invasive Methicillin-Resistant Staphylococcus aureus Infections, United States, 2011. JAMA Intern Med. 2013 [cited 19 Jun 2019]. doi:10.1001/jamainternmed.2013.10423 86. Hiramatsu K, Aritaka N, Hanaki H, Kawasaki S, Hosoda Y, Hori S, et al. Dissemination in Japanese hospitals of strains of Staphylococcus aureus heterogeneously resistant to vancomycin. The Lancet. 1997;350: 1670–1673. doi:10.1016/S0140-6736(97)07324-8 87. Gardete S, Tomasz A. Mechanisms of vancomycin resistance in Staphylococcus aureus. J Clin Invest. 2014;124: 2836–2840. doi:10.1172/JCI68834 88. Asadi A, Razavi S, Talebi M, Gholami M. A review on anti-adhesion therapies of bacterial diseases. Infection. 2019;47: 13–23. doi:10.1007/s15010-018-1222-5 89. Ofek I, Hasty DL, Sharon N. Anti-adhesion therapy of bacterial diseases: prospects and problems. FEMS Immunology & Medical Microbiology. 2003;38: 181–191. doi:10.1016/S0928-8244(03)00228-1 90. Sharon N. Carbohydrates as future anti-adhesion drugs for infectious diseases. Biochimica et Biophysica Acta (BBA) - General Subjects. 2006;1760: 527– 537. doi:10.1016/j.bbagen.2005.12.008 91. Busch A, Waksman G. Chaperone-usher pathways: diversity and pilus assembly mechanism. Philosophical Transactions of the Royal Society B: Biological Sciences. 2012;367: 1112–1122. doi:10.1098/rstb.2011.0206 92. Åberg V, Almqvist F. Pilicides—small molecules targeting bacterial virulence. Org Biomol Chem. 2007;5: 1827–1834. doi:10.1039/B702397A 93. Svensson A, Larsson A, Emtenäs H, Hedenström M, Fex T, Hultgren SJ, et al. Design and evaluation of pilicides: potential novel antibacterial agents directed against uropathogenic Escherichia coli. Chembiochem. 2001;2: 915– 918. 94. Chorell- E, Pinkner JS, Bengtsson C, Edvinsson S, Cusumano CK, Rosenbaum E, et al. Design and Synthesis of Fluorescent Pilicides and Curlicides: Bioactive Tools to Study Bacterial Virulence Mechanisms. Chem Eur J. 2012;18: 4522–4532. doi:10.1002/chem.201103936

58

95. Åberg V, Fällman E, Axner O, Uhlin BE, Hultgren SJ, Almqvist F. Pilicides regulate pili expression in E. coli without affecting the functional properties of the pilus rod. Mol BioSyst. 2007;3: 214–218. doi:10.1039/B613441F 96. Kelly CG, Younson JS, Hikmat BY, Todryk SM, Czisch M, Haris PI, et al. A synthetic peptide adhesion epitope as a novel antimicrobial agent. Nat Biotechnol. 1999;17: 42–47. doi:10.1038/5213 97. Morrow AL, Ruiz-Palacios GM, Jiang X, Newburg DS. Human-Milk Glycans That Inhibit Pathogen Binding Protect Breast-feeding Infants against Infectious Diarrhea. The Journal of Nutrition. 2005;135: 1304–1307. doi:10.1093/jn/135.5.1304 98. Newburg DS, Ruiz-Palacios GM, Morrow AL. HUMAN MILK GLYCANS PROTECT INFANTS AGAINST ENTERIC PATHOGENS. Annu Rev Nutr. 2005;25: 37–58. doi:10.1146/annurev.nutr.25.050304.092553 99. Newburg DS, Ruiz-Palacios GM, Altaye M, Chaturvedi P, Meinzen-Derr J, Guerrero M de L, et al. Innate protection conferred by fucosylated oligosaccharides of human milk against diarrhea in breastfed infants. Glycobiology. 2004;14: 253–263. doi:10.1093/glycob/cwh020 100. Yu Z-T, Nanthakumar NN, Newburg DS. The Human Milk Oligosaccharide 2′-Fucosyllactose Quenches Campylobacter jejuni–Induced Inflammation in Human Epithelial Cells HEp-2 and HT-29 and in Mouse Intestinal Mucosa. The Journal of Nutrition. 2016;146: 1980–1990. doi:10.3945/jn.116.230706 101. Alugupalli KR, Kalfas S. Characterization of the lactoferrin-dependent inhibition of the adhesion of Actinobacilllus actinomycetemcomitans, Prevotella intermedia and Prevotella nigrescens to fibroblasts and to a reconstituted basement membrane. APMIS. 1997;105: 680–688. doi:10.1111/j.1699-0463.1997.tb05071.x 102. Giugliano LG, Ribeiro STG, Vainstein MH, Ulhoa CJ. Free secretory component and lactoferrin of human milk inhibit the adhesion of enterotoxigenic Escherichia coli. Journal of Medical Microbiology. 1995;42: 3–9. doi:10.1099/00222615-42-1-3 103. Wakabayashi H, Yamauchi K, Kobayashi T, Yaeshima T, Iwatsuki K, Yoshie H. Inhibitory Effects of Lactoferrin on Growth and Biofilm Formation of Porphyromonas gingivalis and Prevotella intermedia. Antimicrobial Agents and Chemotherapy. 2009;53: 3308–3316. doi:10.1128/AAC.01688-08 104. Moen DV. Observations on the effectiveness of cranberry juice in urinary infections. Wis Med J. 1962;61: 282–283. 105. Papas PN, Brusch CA, Ceresia GC. Cranberry juice in the treatment of urinary tract infections. Southwest Med. 1966;47: 17–20. 106. Tao Y, Pinzón-Arango PA, Howell AB, Camesano TA. Oral Consumption of Cranberry Juice Cocktail Inhibits Molecular-Scale Adhesion of Clinical

59

Uropathogenic Escherichia coli. Journal of Medicinal Food. 2011;14: 739– 745. doi:10.1089/jmf.2010.0154 107. Howell AB, Botto H, Combescure C, Blanc-Potard A-B, Gausa L, Matsumoto T, et al. Dosage effect on uropathogenic Escherichia coli anti- adhesion activity in urine following consumption of cranberry powder standardized for proanthocyanidin content: a multicentric randomized double blind study. BMC Infect Dis. 2010;10: 94. doi:10.1186/1471-2334-10-94 108. Avorn J, Monane M, Gurwitz JH, Glynn RJ, Choodnovskiy I, Lipsitz LA. Reduction of bacteriuria and pyuria after ingestion of cranberry juice. JAMA. 1994;271: 751–754. 109. Blumberg JB, Camesano TA, Cassidy A, Kris-Etherton P, Howell A, Manach C, et al. Cranberries and Their Bioactive Constituents in Human Health. Advances in Nutrition. 2013;4: 618–632. doi:10.3945/an.113.004473 110. Jepson RG, Williams G, Craig JC. Cranberries for preventing urinary tract infections. Cochrane Kidney and Transplant Group, editor. Cochrane Database of Systematic Reviews. 2012 [cited 1 Jul 2019]. doi:10.1002/14651858.CD001321.pub5 111. Aronson M, Medalia O, Schori L, Mirelman D, Sharon N, Ofek I. Prevention of Colonization of the Urinary Tract of Mice with Escherichia coli by Blocking of Bacterial Adherence with Methyl -D-Mannopyranoside. Journal of Infectious Diseases. 1979;139: 329–332. doi:10.1093/infdis/139.3.329 112. Domenici L, Monti M, Bracchi C, Giorgini M, Colagiovanni V, Muzii L, et al. D-mannose: a promising support for acute urinary tract infections in women. A pilot study. Eur Rev Med Pharmacol Sci. 2016;20: 2920–2925. 113. Mysore JV, Wigginton T, Simon PM, Zopf D, Heman-Ackah LM, Dubois A. Treatment of Helicobacter pylori infection in rhesus monkeys using a novel antiadhesion compound. Gastroenterology. 1999;117: 1316–1325. doi:10.1016/S0016-5085(99)70282-9 114. McEwan NA, Rme CA, Gatto H, Nuttall TJ. Monosaccharide inhibition of adherence by Pseudomonas aeruginosa to canine corneocytes. Veterinary Dermatology. 2008;19: 221–225. doi:10.1111/j.1365-3164.2008.00678.x 115. Cecioni S, Imberty A, Vidal S. Glycomimetics versus Multivalent Glycoconjugates for the Design of High Affinity Lectin Ligands. Chem Rev. 2015;115: 525–561. doi:10.1021/cr500303t 116. Deniaud D, Julienne K, Gouin SG. Insights in the rational design of synthetic multivalent glycoconjugates as lectin ligands. Org Biomol Chem. 2011;9: 966–979. doi:10.1039/C0OB00389A 117. Durka M, Buffet K, Iehl J, Holler M, Nierengarten J-F, Taganna J, et al. The functional valency of dodecamannosylated fullerenes with Escherichia

60

coli FimH—towards novel bacterial antiadhesives. Chem Commun. 2011;47: 1321–1323. doi:10.1039/C0CC04468G 118. Isobe H, Mashima H, Yorimitsu H, Nakamura E. Synthesis of Fullerene Glycoconjugates through Sulfide Connection in Aqueous Media. Org Lett. 2003;5: 4461–4463. doi:10.1021/ol0357705 119. Cecioni S, Lalor R, Blanchard B, Praly J-P, Imberty A, Matthews S, et al. Achieving High Affinity towards a Bacterial Lectin through Multivalent Topological Isomers of Calix[4]arene Glycoconjugates. Chem Eur J. 2009;15: 13232–13240. doi:10.1002/chem.200901799 120. Neri P, Sessler JL, Wang M-X. Calixarenes and Beyond. Cham: Springer International Publishing; 2016. doi:10.1007/978-3-319-31867-7 121. Calvo-Flores FG, Isac-García J, Hernández-Mateo F, Pérez-Balderas F, Calvo-Asín JA, Sanchéz-Vaquero E, et al. 1,3-Dipolar Cycloadditions as a Tool for the Preparation of Multivalent Structures. Org Lett. 2000;2: 2499– 2502. doi:10.1021/ol006175v 122. Sol V, Chaleix V, Champavier Y, Granet R, Huang Y-M, Krausz P. Glycosyl bis-porphyrin conjugates: Synthesis and potential application in PDT. Bioorganic & Medicinal Chemistry. 2006;14: 7745–7760. doi:10.1016/j.bmc.2006.08.004 123. Bouckaert J, Li Z, Xavier C, Almant M, Caveliers V, Lahoutte T, et al. Heptyl α- D -Mannosides Grafted on a β-Cyclodextrin Core To Interfere with Escherichia coli Adhesion: An In Vivo Multivalent Effect. Chem Eur J. 2013;19: 7847–7855. doi:10.1002/chem.201204015 124. Gouin SG, Wellens A, Bouckaert J, Kovensky J. Synthetic Multimeric Heptyl Mannosides as Potent Antiadhesives of Uropathogenic Escherichia coli. ChemMedChem. 2009;4: 749–755. doi:10.1002/cmdc.200900034 125. Gorityala BK, Ma J, Wang X, Chen P, Liu X-W. Carbohydrate functionalized carbon nanotubes and their applications. Chem Soc Rev. 2010;39: 2925. doi:10.1039/b919525b 126. Gu L, Elkin T, Jiang X, Li H, Lin Y, Qu L, et al. Single-walled carbon nanotubes displaying multivalent ligands for capturing pathogens. Chem Commun. 2005; 874. doi:10.1039/b415015e 127. Disney MD, Zheng J, Swager TM, Seeberger PH. Detection of Bacteria with Carbohydrate-Functionalized Fluorescent Polymers. J Am Chem Soc. 2004;126: 13343–13346. doi:10.1021/ja047936i 128. Murray RA, Qiu Y, Chiodo F, Marradi M, Penadés S, Moya SE. A Quantitative Study of the Intracellular Dynamics of Fluorescently Labelled Glyco-Gold Nanoparticles via Fluorescence Correlation Spectroscopy. Small. 2014;10: 2602–2610. doi:10.1002/smll.201303604 129. de la Fuente JM, Penadés S. Glyconanoparticles: Types, synthesis and applications in glycoscience, biomedicine and material science. Biochimica et

61

Biophysica Acta (BBA) - General Subjects. 2006;1760: 636–651. doi:10.1016/j.bbagen.2005.12.001 130. de la Fuente JM, Barrientos AG, Rojas TC, Rojo J, Cañada J, Fernández A, et al. Gold Glyconanoparticles as Water-Soluble Polyvalent Models To Study Carbohydrate Interactions. Angew Chem Int Ed Engl. 2001;40: 2257– 2261. doi:10.1002/1521-3773(20010618)40:12<2257::AID- ANIE2257>3.0.CO;2-S 131. Veerapandian M, Lim SK, Nam HM, Kuppannan G, Yun KS. Glucosamine-functionalized silver glyconanoparticles: characterization and antibacterial activity. Anal Bioanal Chem. 2010;398: 867–876. doi:10.1007/s00216-010-3964-5 132. Barras A, Martin FA, Bande O, Baumann J-S, Ghigo J-M, Boukherroub R, et al. Glycan-functionalized diamond nanoparticles as potent E. coli anti- adhesives. Nanoscale. 2013;5: 2307. doi:10.1039/c3nr33826f 133. Chen Y, Ji T, Rosenzweig Z. Synthesis of Glyconanospheres Containing Luminescent CdSe−ZnS Quantum Dots. Nano Lett. 2003;3: 581–584. doi:10.1021/nl034086g 134. Gallo J, García I, Padro D, Arnáiz B, Penadés S. Water-soluble magnetic glyconanoparticles based on metal-doped ferrites coated with gold: Synthesis and characterization. J Mater Chem. 2010;20: 10010. doi:10.1039/c0jm01756f 135. Mortensen NP, Fowlkes JD, Maggart M, Doktycz MJ, Nataro JP, Drusano G, et al. Effects of sub-minimum inhibitory concentrations of ciprofloxacin on enteroaggregative Escherichia coli and the role of the surface protein dispersin. International Journal of Antimicrobial Agents. 2011;38: 27–34. doi:10.1016/j.ijantimicag.2011.03.011 136. Wojnicz D, Jankowski S. Effects of subinhibitory concentrations of amikacin and ciprofloxacin on the hydrophobicity and adherence to epithelial cells of uropathogenic Escherichia coli strains. International Journal of Antimicrobial Agents. 2007;29: 700–704. doi:10.1016/j.ijantimicag.2007.01.007 137. Ofek I, Doyle RJ. Bacterial Adhesion to Cells and Tissues. Boston, MA: Springer US; 1994. doi:10.1007/978-1-4684-6435-1 138. Balague C. Effect of ciprofloxacin on adhesive properties of non-P mannose- resistant uropathogenic Escherichia coli isolates. Journal of Antimicrobial Chemotherapy. 2003;51: 401–404. doi:10.1093/jac/dkg048 139. Langermann S. Prevention of Mucosal Escherichia coli Infection by FimH- Adhesin-Based Systemic Vaccination. Science. 1997;276: 607–611. doi:10.1126/science.276.5312.607 140. Wizemann TM, Adamou JE, Langermann S. Adhesins as Targets for Vaccine Development. Emerg Infect Dis. 1999;5: 395–403. doi:10.3201/eid0503.990310

62

141. Moon HW, Bunn TO. Vaccines for preventing enterotoxigenic Escherichia coli infections in farm animals. Vaccine. 1993;11: 213–220. doi:10.1016/0264-410X(93)90020-X 142. Candela M, Perna F, Carnevali P, Vitali B, Ciati R, Gionchetti P, et al. Interaction of probiotic Lactobacillus and Bifidobacterium strains with human intestinal epithelial cells: Adhesion properties, competition against enteropathogens and modulation of IL-8 production. International Journal of Food Microbiology. 2008;125: 286–292. doi:10.1016/j.ijfoodmicro.2008.04.012 143. Poinar GO, Thomas GM. A new bacterium, Achromobacter nematophilus sp. nov. (Achromobacteriaceae: Eubacteriales) associated with a nematode. International Bulletin of Bacteriological Nomenclature and Taxonomy. 1965;15: 249–252. doi:10.1099/00207713-15-4-249 144. Thomas GM, Poinar GO. Xenorhabdus gen. nov., a Genus of Entomopathogenic, Nematophilic Bacteria of the Family Enterobacteriaceae. International Journal of Systematic Bacteriology. 1979;29: 352–360. doi:10.1099/00207713-29-4-352 145. Boemare NE, Akhurst RJ, Mourant RG. DNA Relatedness between Xenorhabdus spp. (Enterobacteriaceae), Symbiotic Bacteria of Entomopathogenic Nematodes, and a Proposal To Transfer Xenorhabdus luminescens to a New Genus, Photorhabdus gen. nov. International Journal of Systematic Bacteriology. 1993;43: 249–255. doi:10.1099/00207713-43-2-249 146. Adeolu M, Alnajar S, Naushad S, S Gupta R. Genome-based phylogeny and taxonomy of the “Enterobacteriales”: proposal for ord. nov. divided into the families Enterobacteriaceae, Erwiniaceae fam. nov., Pectobacteriaceae fam. nov., Yersiniaceae fam. nov., Hafniaceae fam. nov., Morganellaceae fam. nov., and Budviciaceae fam. nov. Int J Syst Evol Microbiol. 2016;66: 5575–5599. doi:10.1099/ijsem.0.001485 147. Akhurst RJ. Taxonomy of Australian clinical isolates of the genus Photorhabdus and proposal of Photorhabdus asymbiotica subsp. asymbiotica subsp. nov. and P. asymbiotica subsp. australis subsp. nov. INTERNATIONAL JOURNAL OF SYSTEMATIC AND EVOLUTIONARY MICROBIOLOGY. 2004;54: 1301–1310. doi:10.1099/ijs.0.03005-0 148. Fischer-Le Saux M, Viallard V, Brunel B, Normand P, Boemare NE. Polyphasic classification of the genus Photorhabdus and proposal of new taxa: P. luminescens subsp. luminescens subsp. nov., P. luminescens subsp. akhurstii subsp. nov., P. luminescens subsp. laumondii subsp. nov., P. temperata sp. nov., P. temperata subsp. temperata subsp. nov. and P. asymbiotica sp. nov. International Journal of Systematic Bacteriology. 1999;49: 1645–1656. doi:10.1099/00207713-49-4-1645

63

149. Ferreira T, van Reenen CA, Endo A, Tailliez P, Pages S, Sproer C, et al. Photorhabdus heterorhabditis sp. nov., a symbiont of the entomopathogenic nematode Heterorhabditis zealandica. INTERNATIONAL JOURNAL OF SYSTEMATIC AND EVOLUTIONARY MICROBIOLOGY. 2014;64: 1540–1545. doi:10.1099/ijs.0.059840-0 150. Machado RAR, Wüthrich D, Kuhnert P, Arce CCM, Thönen L, Ruiz C, et al. Whole-genome-based revisit of Photorhabdus phylogeny: proposal for the elevation of most Photorhabdus subspecies to the species level and description of one novel species Photorhabdus bodei sp. nov., and one novel subspecies Photorhabdus laumondii subsp. clarkei subsp. nov. International Journal of Systematic and Evolutionary Microbiology. 2018;68: 2664–2681. doi:10.1099/ijsem.0.002820 151. Waterfield NR, Ciche T, Clarke D. Photorhabdus and a Host of Hosts. Annu Rev Microbiol. 2009;63: 557–574. doi:10.1146/annurev.micro.091208.073507 152. Mulley G, Beeton ML, Wilkinson P, Vlisidou I, Ockendon-Powell N, Hapeshi A, et al. From Insect to Man: Photorhabdus Sheds Light on the Emergence of Human Pathogenicity. Skurnik M, editor. PLoS ONE. 2015;10: e0144937. doi:10.1371/journal.pone.0144937 153. Boemare NE, Akhurst RJ. Biochemical and Physiological Characterization of Colony Form Variants in Xenorhabdus spp. (Enterobacteriaceae). Microbiology. 1988;134: 751–761. doi:10.1099/00221287-134-3-751 154. Han R, Ehlers R-U. Effect of Photorhabdus luminescens phase variants on the in vivo and in vitro development and reproduction of the entomopathogenic nematodes Heterorhabditis bacteriophora and Steinernema carpocapsae. FEMS Microbiology Ecology. 2001;35: 239–247. doi:10.1111/j.1574- 6941.2001.tb00809.x 155. Dunlap P. Biochemistry and Genetics of Bacterial Bioluminescence. In: Thouand G, Marks R, editors. Bioluminescence: Fundamentals and Applications in Biotechnology - Volume 1. Berlin, Heidelberg: Springer Berlin Heidelberg; 2014. pp. 37–64. doi:10.1007/978-3-662-43385-0_2 156. Brodl E, Winkler A, Macheroux P. Molecular Mechanisms of Bacterial Bioluminescence. Computational and Structural Biotechnology Journal. 2018;16: 551–564. doi:10.1016/j.csbj.2018.11.003 157. Duchaud E, Rusniok C, Frangeul L, Buchrieser C, Givaudan A, Taourit S, et al. The genome sequence of the entomopathogenic bacterium Photorhabdus luminescens. Nat Biotechnol. 2003;21: 1307–1313. doi:10.1038/nbt886 158. Hinchliffe SJ. Insecticidal Toxins from the Photorhabdus and Xenorhabdus Bacteria. TOTNJ. 2013;3: 101–118. doi:10.2174/1875414701003010101

64

159. Forst S, Dowds B, Boemare N, Stackebrandt E. XENORHABDUS AND PHOTORHABDUS SPP.:Bugs That Kill Bugs. Annu Rev Microbiol. 1997;51: 47–72. doi:10.1146/annurev.micro.51.1.47 160. Clarke DJ. Photorhabdus : a model for the analysis of pathogenicity and mutualism. Cellular Microbiology. 2008;10: 2159–2167. doi:10.1111/j.1462- 5822.2008.01209.x 161. Bedding RA, Molyneux AS. Penetration of Insect Cuticle By Infective Juveniles of Heterorhabditis Spp. (Heterorhabditidae: Nematoda). Nematol. 1982;28: 354–359. doi:10.1163/187529282X00402 162. Ciche TA, Ensign JC. For the Insect Pathogen Photorhabdus luminescens, Which End of a Nematode Is Out? Applied and Environmental Microbiology. 2003;69: 1890–1897. doi:10.1128/AEM.69.4.1890-1897.2003 163. Ciche TA, Kim K -s., Kaufmann-Daszczuk B, Nguyen KCQ, Hall DH. Cell Invasion and Matricide during Photorhabdus luminescens Transmission by Heterorhabditis bacteriophora Nematodes. Applied and Environmental Microbiology. 2008;74: 2275–2287. doi:10.1128/AEM.02646-07 164. Kumar A, Sýkorová P, Demo G, Dobeš P, Hyršl P, Wimmerová M. A Novel Fucose-binding Lectin from Photorhabdus luminescens (PLL) with an Unusual Heptabladed β-Propeller Tetrameric Structure. J Biol Chem. 2016;291: 25032–25049. doi:10.1074/jbc.M115.693473 165. Jančaříková G, Houser J, Dobeš P, Demo G, Hyršl P, Wimmerová M. Characterization of novel bangle lectin from Photorhabdus asymbiotica with dual sugar-binding specificity and its effect on host immunity. Schneider DS, editor. PLoS Pathog. 2017;13: e1006564. doi:10.1371/journal.ppat.1006564 166. Zhang J, Chatterjee D, Brennan PJ, Spencer JS, Liav A. A modified synthesis and serological evaluation of neoglycoproteins containing the natural disaccharide of PGL-I from Mycobacterium leprae. Bioorganic & Medicinal Chemistry Letters. 2010;20: 3250–3253. doi:10.1016/j.bmcl.2010.04.072 167. Hunter SW, Fujiwara T, Brennan PJ. Structure and antigenicity of the major specific glycolipid antigen of Mycobacterium leprae. J Biol Chem. 1982;257: 15072–15078. 168. Beshr G, Sikandar A, Jemiller E-M, Klymiuk N, Hauck D, Wagner S, et al. Photorhabdus luminescens lectin A (PllA): A new probe for detecting α- galactoside–terminating glycoconjugates. J Biol Chem. 2017;292: 19935– 19951. doi:10.1074/jbc.M117.812792 169. Cooper DKC, Ezzelarab MB, Hara H, Iwase H, Lee W, Wijkstrom M, et al. The pathobiology of pig-to-primate xenotransplantation: a historical review. Xenotransplantation. 2016;23: 83–105. doi:10.1111/xen.12219 170. Zyl C van, Malan AP. The Role of Entomopathogenic Nematodes as Biological Control Agents of Insect Pests, with Emphasis on the History of

65

Their Mass Culturing and in vivo Production. African Entomology. 2014;22: 235–249. doi:10.4001/003.022.0222 171. Memari Z, Karimi J, Kamali S, Hossein Goldansaz S, Hosseini M. Are Entomopathogenic Nematodes Effective Biological Control Agents Against the Carob Moth, Ectomyelois ceratoniae? Journal of Nematology. 2016;48: 261–267. doi:10.21307/jofnem-2017-034 172. Wilson MJ, Wilson DJ, Rodgers A, Gerard PJ. Developing a strategy for using entomopathogenic nematodes to control the African black beetle (Heteronychus arator) in New Zealand pastures and investigating temperature constraints. Biological Control. 2016;93: 1–7. doi:10.1016/j.biocontrol.2015.11.002 173. Ehlers R-U. Mass production of entomopathogenic nematodes for plant protection. Applied Microbiology and Biotechnology. 2001;56: 623–633. doi:10.1007/s002530100711 174. Shapiro-Ilan DI, Han R, Dolinksi C. Entomopathogenic nematode production and application technology. J Nematol. 2012;44: 206–217. 175. Lacey LA, Georgis R. Entomopathogenic nematodes for control of insect pests above and below ground with comments on commercial production. J Nematol. 2012;44: 218–225. 176. Rezaei N, Karimi J, Hosseini M, Goldani M, Campos-Herrera R. Pathogenicity of Two Species of Entomopathogenic Nematodes Against the Greenhouse Whitefly, Trialeurodes vaporariorum (Hemiptera: Aleyrodidae), in Laboratory and Greenhouse Experiments. J Nematol. 2015;47: 60–66. 177. Sievers F, Wilm A, Dineen D, Gibson TJ, Karplus K, Li W, et al. Fast, scalable generation of high-quality protein multiple sequence alignments using Clustal Omega. Molecular Systems Biology. 2014;7: 539–539. doi:10.1038/msb.2011.75 178. Wiseman T, Williston S, Brandts JF, Lin LN. Rapid measurement of binding constants and heats of binding using a new titration calorimeter. Anal Biochem. 1989;179: 131–137. doi:10.1016/0003-2697(89)90213-3 179. Schuck P. Size-distribution analysis of macromolecules by sedimentation velocity ultracentrifugation and lamm equation modeling. Biophys J. 2000;78: 1606–1619. doi:10.1016/S0006-3495(00)76713-0 180. Mueller U, Darowski N, Fuchs MR, Förster R, Hellmig M, Paithankar KS, et al. Facilities for macromolecular crystallography at the Helmholtz-Zentrum Berlin. J Synchrotron Radiat. 2012;19: 442–449. doi:10.1107/S0909049512006395 181. Kabsch W. XDS. Acta Crystallogr D Biol Crystallogr. 2010;66: 125–132. doi:10.1107/S0907444909047337 182. Krug M, Weiss MS, Heinemann U, Mueller U. XDSAPP : a graphical user interface for the convenient processing of diffraction data using XDS. Journal

66

of Applied Crystallography. 2012;45: 568–572. doi:10.1107/S0021889812011715 183. Winn MD, Ballard CC, Cowtan KD, Dodson EJ, Emsley P, Evans PR, et al. Overview of the CCP 4 suite and current developments. Acta Crystallographica Section D Biological Crystallography. 2011;67: 235–242. doi:10.1107/S0907444910045749 184. Vagin A, Teplyakov A. Molecular replacement with MOLREP. Acta Crystallographica Section D Biological Crystallography. 2010;66: 22–25. doi:10.1107/S0907444909042589 185. Murshudov GN, Skubák P, Lebedev AA, Pannu NS, Steiner RA, Nicholls RA, et al. REFMAC 5 for the refinement of macromolecular crystal structures. Acta Crystallographica Section D Biological Crystallography. 2011;67: 355– 367. doi:10.1107/S0907444911001314 186. Emsley P, Lohkamp B, Scott WG, Cowtan K. Features and development of Coot. Acta Crystallographica Section D Biological Crystallography. 2010;66: 486–501. doi:10.1107/S0907444910007493 187. Lebedev AA, Young P, Isupov MN, Moroz OV, Vagin AA, Murshudov GN. JLigand: a graphical tool for the CCP4 template-restraint library. Acta Crystallogr D Biol Crystallogr. 2012;68: 431–440. doi:10.1107/S090744491200251X 188. Laughton AM, Siva-Jothy MT. A standardised protocol for measuring phenoloxidase and prophenoloxidase in the honey bee, Apis mellifera. Apidologie. 2011;42: 140–149. doi:10.1051/apido/2010046 189. Becker DJ, Lowe JB. Fucose: biosynthesis and biological function in mammals. Glycobiology. 2003;13: 41R-53R. doi:10.1093/glycob/cwg054 190. Walski T, De Schutter K, Van Damme EJM, Smagghe G. Diversity and functions of protein glycosylation in insects. Insect Biochemistry and Molecular Biology. 2017;83: 21–34. doi:10.1016/j.ibmb.2017.02.005 191. Wohlschlager T, Butschi A, Grassi P, Sutov G, Gauss R, Hauck D, et al. Methylated glycans as conserved targets of animal and fungal innate defense. Proc Natl Acad Sci USA. 2014;111: E2787–E2796. doi:10.1073/pnas.1401176111 192. Staudacher E. Methylation – an uncommon modification of glycans. Biological Chemistry. 2012;393. doi:10.1515/hsz-2012-0132 193. Hillyer JF. Insect immunology and hematopoiesis. Developmental & Comparative Immunology. 2016;58: 102–118. doi:10.1016/j.dci.2015.12.006 194. Almond A. Hyaluronan. Cell Mol Life Sci. 2007;64: 1591–1596. doi:10.1007/s00018-007-7032-z 195. Scott D, Harding E, Rowe A. A Brief Introduction to the Analytical Ultracentrifugation of Proteins for Beginners. Analytical Ultracentrifugation.

67

Cambridge: Royal Society of Chemistry; 2007. pp. 1–25. doi:10.1039/9781847552617-00001 196. Jančaříková G, Herczeg M, Fujdiarová E, Houser J, Kövér KE, Borbás A, et al. Synthesis of α-L-Fucopyranoside-Presenting Glycoclusters and Investigation of Their Interaction with Photorhabdus asymbiotica Lectin (PHL). Chemistry. 2018;24: 4055–4068. doi:10.1002/chem.201705853 197. Zug R, Hammerstein P. Wolbachia and the insect immune system: what reactive oxygen species can tell us about the mechanisms of Wolbachia-host interactions. Front Microbiol. 2015;6: 1201. doi:10.3389/fmicb.2015.01201 198. Dubovskii IM, Grizanova EV, Chertkova EA, Slepneva IA, Komarov DA, Vorontsova YaL, et al. Generation of reactive oxygen species and activity of antioxidants in hemolymph of the moth larvae (L.) (Lepidoptera: Piralidae) at development of the process of encapsulation. J Evol Biochem Phys. 2010;46: 35–43. doi:10.1134/S0022093010010044 199. Gay NJ, Symmons MF, Gangloff M, Bryant CE. Assembly and localization of Toll-like receptor signalling complexes. Nat Rev Immunol. 2014;14: 546– 558. doi:10.1038/nri3713 200. Makni-Maalej K, Chiandotto M, Hurtado-Nedelec M, Bedouhene S, Gougerot-Pocidalo M-A, Dang PM-C, et al. Zymosan induces NADPH oxidase activation in human neutrophils by inducing the phosphorylation of p47phox and the activation of Rac2: involvement of protein tyrosine kinases, PI3Kinase, PKC, ERK1/2 and p38MAPkinase. Biochem Pharmacol. 2013;85: 92–100. doi:10.1016/j.bcp.2012.10.010 201. González-Santoyo I, Córdoba-Aguilar A. Phenoloxidase: a key component of the insect immune system: Biochemical and evolutionary ecology of PO. Entomologia Experimentalis et Applicata. 2012;142: 1–16. doi:10.1111/j.1570-7458.2011.01187.x 202. Cerenius L, Söderhäll K. The prophenoloxidase-activating system in invertebrates. Immunol Rev. 2004;198: 116–126. 203. Zhao P, Lu Z, Strand MR, Jiang H. Antiviral, anti-parasitic, and cytotoxic effects of 5,6-dihydroxyindole (DHI), a reactive compound generated by phenoloxidase during insect immune response. Insect Biochem Mol Biol. 2011;41: 645–652. doi:10.1016/j.ibmb.2011.04.006 204. Hyrsl P, Dobes P, Wang Z, Hauling T, Wilhelmsson C, Theopold U. Clotting factors and eicosanoids protect against nematode infections. J Innate Immun. 2011;3: 65–70. doi:10.1159/000320634 205. Wang Z, Wilhelmsson C, Hyrsl P, Loof TG, Dobes P, Klupp M, et al. Pathogen entrapment by transglutaminase--a conserved early innate immune mechanism. PLoS Pathog. 2010;6: e1000763. doi:10.1371/journal.ppat.1000763

68

206. Šulák O, Cioci G, Lameignère E, Balloy V, Round A, Gutsche I, et al. Burkholderia cenocepacia BC2L-C Is a Super Lectin with Dual Specificity and Proinflammatory Activity. Saper MA, editor. PLoS Pathog. 2011;7: e1002238. doi:10.1371/journal.ppat.1002238 207. Philippe N, Alcaraz J-P, Coursange E, Geiselmann J, Schneider D. Improvement of pCVD442, a suicide plasmid for gene allele exchange in bacteria. Plasmid. 2004;51: 246–255. doi:10.1016/j.plasmid.2004.02.003 208. Ye J, Coulouris G, Zaretskaya I, Cutcutache I, Rozen S, Madden TL. Primer-BLAST: A tool to design target-specific primers for polymerase chain reaction. BMC Bioinformatics. 2012;13: 134. doi:10.1186/1471-2105-13-134 209. https://www.sigmaaldrich.com/content/dam/sigma- aldrich/docs/Sigma/Bulletin/pln70bul.pdf 210. https://pws288.byu.edu/Portals/89/Docs/Qiagen%20Clean- Up%20of%20Gel%20Fragments/Qiagen'sMineluteHandbook.pdf 211. Llosa M, Gomis-Ruth FX, Coll M, Cruz F de la. Bacterial conjugation: a two-step mechanism for DNA transport. Mol Microbiol. 2002;45: 1–8. doi:10.1046/j.1365-2958.2002.03014.x 212. Johnson CM, Grossman AD. The Composition of the Cell Envelope Affects Conjugation in Bacillus subtilis. Christie PJ, editor. J Bacteriol. 2016;198: 1241–1249. doi:10.1128/JB.01044-15 213. Webb M. The Influence of Magnesium on Cell Division: The Effect of Magnesium on the Growth of Bacteria in Simple Chemically Defined Media. Journal of General Microbiology. 1949;3: 418–424. doi:10.1099/00221287-3- 3-418 214. Groisman EA, Hollands K, Kriner MA, Lee E-J, Park S-Y, Pontes MH. Bacterial Mg 2+ Homeostasis, Transport, and Virulence. Annu Rev Genet. 2013;47: 625–646. doi:10.1146/annurev-genet-051313-051025 215. Lorenz TC. Polymerase Chain Reaction: Basic Protocol Plus Troubleshooting and Optimization Strategies. JoVE. 2012; 3998. doi:10.3791/3998 216. https://www.agilent.com/cs/library/usermanuals/public/600870.pdf 217. Folkesson A, Jelsbak L, Yang L, Johansen HK, Ciofu O, Høiby N, et al. Adaptation of Pseudomonas aeruginosa to the cystic fibrosis airway: an evolutionary perspective. Nat Rev Microbiol. 2012;10: 841–851. doi:10.1038/nrmicro2907 218. Thai Le S, Malinovska L, Vašková M, Mező E, Kelemen V, Borbás A, et al. Investigation of the Binding Affinity of a Broad Array of L-Fucosides with Six Fucose-Specific Lectins of Bacterial and Fungal Origin. Molecules. 2019;24: 2262. doi:10.3390/molecules24122262 219. Chemani C, Imberty A, de Bentzmann S, Pierre M, Wimmerova M, Guery BP, et al. Role of LecA and LecB Lectins in Pseudomonas aeruginosa-

69

Induced Lung Injury and Effect of Carbohydrate Ligands. Infection and Immunity. 2009;77: 2065–2075. doi:10.1128/IAI.01204-08

70

CURRICULUM VITAE

Personal details Name: Eva Fujdiarová Date of birth: 14.12.1983 Nationality: Czech Contact: [email protected]

Employment 2015-present Researcher at Glycobiochemistry group, CEITEC, and NCBR, Masaryk University, Brno, Czech Republic 2012-2013 Veterinary inspector in veterinary hygiene department, Regional Veterinary Administration, Nový Jičín 2009-2012 Veterinarian, Veterinary clinics for small animals (dogs, cats, and small rodents) Čáslav, Brno, and Příbor

Education since 9/2014 Doctoral study of Biomolecular chemistry and bioinformatics, National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno 2003-2009 Master study of Veterinary medicine, University of Veterinary and Pharmaceutical Sciences, Brno

71

Research experience Glycobiology with the focus on bacterial lectins, their interaction with carbohydrates and structural properties. Experience with following methods: bioinformatics, molecular biology methods (gene cloning, PCR, colony PCR, gene knockout, CRISPR), protein expression and purification, biophysical methods related to biomolecular interaction measurements (ITC, SPR, MST), methods for macromolecular structure determination (DLS, AUC), protein crystallography.

Teaching activities Co-supervisor of two bachelor theses Lukáš Faltinek, Agglutination of Photorhabdus luminescens cells using multivalent saccharides compounds (2015-2017) Šárka Hanáková, Purification of lectins from Photorhabdus luminescens (2015-2017) Consultant of two diploma theses Filip Melicher, Structural studies of lectins from Photorhabdus luminescens (2017-2018) Lukáš Faltinek, Characterization of lectins from Photorhabdus spp. (2018- 2019)

Publications Lukáš Faltinek*, Eva Fujdiarová*, Filip Melicher, Josef Houser, Martina Kašáková, Nikolay Kondakov, Leonid Kononov, Kamil Parkan, Sébastien Vidal, Michaela Wimmerová: Lectin PLL3, a novel monomeric member of seven-bladed β-propeller lectin family. Molecules, 2019, 24, 4540, doi.org/10.3390/molecules24244540 Lenka Malinovská, Son Thai Le, Mihály Herczeg, Michaela Vašková, Josef Houser, Eva Fujdiarová, Jan Komárek, Petr Hodek, Anikó Borbás, Michaela Wimmerová, Magdolna Csávás. Synthesis of β-D-galactopyranoside- presenting glycoclusters, investigation of their interactions with Pseudomonas aeruginosa lectin A (PA-IL) and evaluation of their

72 anti-adhesion potential. Biomolecules, 2019, 9(11), DOI: 10.3390/biom9110686. Petra Sýkorová, Jitka Novotná, Gabriel Demo, Guillaume Pompidor, Eva Dubská, Jan Komárek, Eva Fujdiarová, Josef Houser, Lucia Hároníková, Annabelle Varrot, Nadezhda Shilova, Anne Imberty, Nicolai Bovin, Martina Pokorná, Michaela Wimmerová. Characterization of novel lectins from Burkholderia pseudomallei and Chromobacterium violaceum with seven- bladed β-propeller fold. International Journal of Biological Macromolecules, 2019, in press, DOI: 10.1016/j.ijbiomac.2019.10.200. Martina Kašáková, Lenka Malinovská, Tomaš Klejch, Martina Hlavačková, Hana Dvořaková, Eva Fujdiarová, Zdeňka Rottnerová, Olga Maťatková, Pavel Lhoták, Michaela Wimmerová, Jitka Moravcová. Selectivity of original C- hexopyranosyl calix[4]arene conjugates towards lectins of different origin. Carbohydrate research, 2018, ISSN 1873-426X, doi: 10.1016/j.carres.2018.08.012. Gita Jančaříková, Mihály Herczeg, Eva Fujdiarová, Josef Houser, Katalin E. Kövér, Anikó Borbás, Michaela Wimmerová, and Magdolna Csávás. Synthesis of a-Lfucopyranoside-presenting glycoclusters and investigation of their interaction with Photorhabdus asymbiotica lectin (PHL). Chemistry: A European Journal, 2018, ISSN 1521-3765, DOI: 10.1002/chem.201705853.

International fellowships Department of Microbiology, University College Cork, Ireland. 08 - 21 February 2016. I was trained in the laboratory of microbiology of professor David Clark. During the fellowship, I was introduced to the theory of bacterial genome modification. On the example of Photorhabdus laumondii, I became familiar with the method of gene knock-out using the bacteriophage derived vector pDS132, transferred by the bacterial conjugation process. I also independently conducted experiments leading to the construction of the desired P. laumondii mutant.

73

Active conference participation ▪ XVI Discussions in Structural Molecular Biology, 21 – 23 March 2019, Nové Hrady. Influence of glucose 3-O-methylation on binding properties towards PLL2 lectin (poster presentation) ▪ 29th International Carbohydrate Symposium, 15 – 19 July 2018, Lisbon. Glycoclusters as a tool for inhibition of lectin-mediated bacterial adhesion (poster presentation) ▪ The 43rd FEBS Congress, 07 – 13 July 2018, Prague. Study of lectins from Photorhabdus luminescens bacterium (speed talk + poster presentation) ▪ XVIII Meeting of Biochemists and Molecular Biologists, 14 – 15 November 2017, Brno. Study of Photorhabdus luminescens lectins to reveal their function in the entomopathogenic life cycle (student talk) ▪ XXV Biochemical Congress, 13 - 16 September 2016, Praha. Characterization of lectins from Photorhabdus luminescens to reveal their function in an entomopathogenic complex with Heterorhabditis bacteriophora (student talk)

Selected workshops and courses Macromolecular X-ray crystallography ▪ Advanced diffraction techniques for biology. 19-22 November 2019, Institute de Biologie Structurale, Grenoble, France. ▪ Workshop on data collection and structure solving in macromolecular X-ray diffraction. 12-16 July 2019, Jerzy Haber Institute of Catalysis and Surface Chemistry, Krakow, Poland. ▪ HERCULES European school. 25 February – 03 March 2018, European Synchrotron Radiation Facility (ESRF), Grenoble, France. ▪ Biomacromolecular crystallization workshop. 23 – 26 October 2017, CEITEC, Masaryk University, Brno. ▪ Tutorial in Molecular Crystallography. 06 – 10 March 2017, Institute de Biologie Structurale (IBS), Grenoble, France. ▪ Structural glycoscience – workshop. 28 June – 1 July 2016, Université Grenoble Alpes, Grenoble, France.

74

Protein interaction and characterization ▪ SPR Workshop Application in life-science. 15 - 16 November 2018, CEITEC, Masaryk University, Brno. ▪ How to characterize your sample and check its quality. 23 - 25 May 2018, CEITEC, Masaryk University, Brno. ▪ Differential Scanning Calorimetry. 03 – 04 October 2017, CEITEC, Masaryk University, Brno. ▪ Workshop on Surface Plasmon Resonance – theory and hands-on. 22 – 24 May 2017, CEITEC, Masaryk University, Brno. ▪ Microscale thermophoresis and differential scanning fluorimetry workshop. 12 – 13 November 2015, CEITEC, Masaryk University, Brno.

Molecular biology ▪ Modern Techniques in Molecular Biology and Genome Engineering Workshop. 17 – 20 July 2017, Vienna biocentre Core Facility, Vienna, Austria.

75

APPENDIX

76

APENDIX I

Eva Fujdiarová, Josef Houser, Pavel Dobeš, Gita Paulíková, Nikolay Kondakov, Leonid Kononov, Pavel Hyršl, Michaela Wimmerová. Heptabladded β-propeller lectins PLL2 and PHL from Photorhabdus spp. recognize O-methylated sugars and influence the host immune system. Submitted to FEBS Journal

APENDIX II

Lukáš Faltinek*, Eva Fujdiarová*, Filip Melicher, Josef Houser, Martina Kašáková, Nikolay Kondakov, Leonid Kononov, Kamil Parkan, Sébastien Vidal, Michaela Wimmerová. Lectin PLL3, a novel monomeric member of seven-bladded β-propeller lectin family. * These authors contributed equally Molecules, 2019, 24, 4540 doi.org/10.3390/molecules24244540

APENDIX III

Gita Jančaříková, Mihály Herczeg, Eva Fujdiarová, Josef Houser, Katalin E. Köver, Anikó Borbás, Michaela Wimmerová, and Magdolna Csávás.

Synthesis of α-L-fucoside-presenting glycoclusters and investigation of their interaction with Photorhabdus asymbiotica lectin (PHL). Chemistry: A European Journal, 2018, 24(16), 4055-4068 DOI: 10.1002/chem.201705853 Full text is used with permission of John Wiley & Sons in printed and electronic version (Licence number: 4334690785003)

APENDIX IV

Martina Kašáková, Lenka Malinovská, Tomáš Klejch, Martina Hlaváčová, Hana Dvořáková, Eva Fujdiarová, Zdeňka Rottnerová, Olga Maťátková, Pavel Lhoták, Michaela Wimmerová, Jitka Moravcová. Selectivity of original C-hexopyranosyl calix[4]arene conjugates towards lectins of different origin. Carbohydrate Research, 2018, 469, 60-72. DOI: 10.1016/j.carres.2018.08.012

APENDIX V

Lenka Malinovská, Son Thai Le, Mihály Herczeg, Michaela Vašková, Josef Houser, Eva Fujdiarová, Jan Komárek, Petr Hodek, Anikó Borbás, Michaela Wimmerová, Magdolna Csávás.

Synthesis of β-D-galactopyranoside-presenting glycoclusters, investigation of their interactions with Pseudomonas aeruginosa lectin A (PA-IL) and evaluation of their anti-adhesion potential. Biomolecules, 2019, 9(11). DOI: 10.3390/biom9110686.

APENDIX VI

Petra Sýkorová, Jitka Novotná, Gabriel Demo, Guillaume Pompidor, Eva Dubská, Jan Komárek, Eva Fujdiarová, Josef Houser, Lucia Hároníková, Annabelle Varrot, Nadezhda Shilova, Anne Imberty, Nicolai Bovin, Martina Pokorná, Michaela Wimmerová. Characterization of novel lectins from Burkholderia pseudomallei and Chromobacterium violaceum with seven-bladed β-propeller fold. International Journal of Biological Macromolecules, 2019, in press. DOI: 10.1016/j.ijbiomac.2019.10.200