International One Health Symposium 2024

Europe/Berlin
https://hotel-moa-berlin.de/?lang=en (Mercure Hotel MOA Berlin)

https://hotel-moa-berlin.de/?lang=en

Mercure Hotel MOA Berlin

Stephanstr. 41 10559 Berlin
Description

 

Welcome to the International One Health Symposium 2024 which will take place under the highlight topic "One Health: connecting infectious diseases with ecological challenges "

 

Date: October 17th - October 19th, 2024

Venue: Mercure Hotel MOA Berlin, Stephanstraße 41, 10559 Berlin

 

Participation fees:

€ 80 for undergraduate student, graduate student, PhD student

€ 250 for postdocs, senior scientists, public health workers, etc.

 

We are delighted that 6 great keynote speakers have confirmed their participation in the first International One Health Symposium 2024.

 

The Call for Abstracts has been extended until June 30th, 2024.

Please submit your contribution on one of the following topics and read the instructions carefully:

  • Biodiversity and Human Health
  • Climate change, Water and Plant Health
  • Climate change and Human health
  • Environmental pollution, Water and Plant Health
  • Infectiouse diseases and Zoonoses
  • Data modelling and AI
  • Genomics and Proteomics
  • One Health and Public Health
  • Vectors
  • Ehical Stewardship

- The review results were sent to all submitters by e-mail on Friday, September 6th. -

 

Conference language: English                                                                 

Please keep an eye on your e-mail for further announcements.

 


Funded by:

                 

_______________________________________________________________________________________________________

Sponsored by:

_______________________________________________________________________________________________________

Registration
1. Registration for Organizers and Keynote Speakers (€ 0)
2. Registration for participants from Germany and non-EU countries (€ 80 and € 250)
3. Registration for participants from a country within the European Union (except Germany) (€ 80 and € 250)
    • Registration & Snacks Atrium

      Atrium

    • Welcome Note MOA 4 + 5

      MOA 4 + 5

    • Keynote I: Matthias Liess MOA 4 + 5

      MOA 4 + 5

    • 14:00
      Break Atrium

      Atrium

    • Session 1: Infectious Diseases and Zoonoses I MOA 4 + 5

      MOA 4 + 5

      • 1
        Non-invasive surveillance of avian influenza virus and environmental pathogens

        Avian influenza virus (AIV) is causing significant mortality in wild birds, poultry, and mammals, posing a global health threat due to wild bird migration that facilitates AIV transmission and evolution. We developed a genomics-based surveillance protocol using portable nanopore sequencing for rapid AIV profiling from non-invasively collected environmental samples. Using a well-defined AIV strain, we benchmarked portable RNA extraction and detection, compared DNA- and RNA-nanopores, and evaluated computational pipelines for viral sequence creation and analysis. We validated our setup with environmental samples from a duck farm in France, a “vulture restaurant”, and a wetland in Spain. Additionally, we used non-invasive water and air samples to profile the RNA virome and antimicrobial resistome, offering a broader pathogen view. Our results show that converting viral RNA to cDNA for amplification is effective for low-concentration samples. Non-invasive passive and active techniques, including air sampling via liquid impingement and water sampling with torpedo-shaped devices, outperformed traditional methods, detecting AIV in challenging environments and enabling comprehensive RNA virome and antimicrobial resistome characterization. This approach holds promise for monitoring wildlife pathogens, understanding their evolution and transmission, and informing One Health risks at the intersection of human, animal, and environmental health systems.

        Speaker: Dr Albert Perlas (Helmholtz AI, Helmholtz Zentrum Muenchen, Neuherberg, Germany )
      • 2
        The MEK1/2 inhibitor ZMN efficiently blocks HPAIV infections

        In 2021 highly pathogenic avian influenza strain H5N1 clade 2.3.4.4b spread in domestic and wild birds, now considered to be a panzoonosis. Spillover events in marine mammals, mink farms and dairy cattle farms posing the risk that the virus gains the ability to be transferred between mammals and may also jump to humans. Besides prophylactic vaccinations, antiviral drugs are needed to treat acute infections. Direct acting antivirals (DAA) target viral components, making them highly effective, but prolonged treatment bears the risk of resistance selection. Alternative approaches are host-targeted antivirals (HTA), like the MEK1/2 inhibitor Zapnometinib (ZMN), interfering with cellular mechanisms misused by viruses. In this study, we evaluated the efficacy of ZMN against HPAIV including H5N1 clade 2.3.4.4b. We could show, that ZMN reduces the production of progeny viral particles of several H5Nx and H7Nx viruses. This antiviral mode of action is not restricted to the commonly used A549 cell line, but could be shown for different cell lines (Calu-3, MDCK II, VeroE6). The inhibitory effect is caused by a nuclear retention of the viral genomes in the later stage of the infection. Additionally, ZMN can act in a synergistic mode of action, with the DAA´s Oseltamivir carboxylate (OTC) and Baloxavir acid (BXA) providing an alternative treatment strategy by combining DAAs and HTAs. These results demonstrate that ZMN represents an effective drug against H5N1 and other HPAIV infections.

        Speaker: Dr André Schreiber
      • 3
        H7 highly pathogenic avian influenza A virus genomes from the first half of the 20th century

        Highly pathogenic avian influenza viruses (HPAIV) cause high mortality in poultry and significant economic losses. Since 1902, H7 and H5 subtypes have been the most prevalent in poultry, posing potential health risks to humans, albeit with only sporadic infections so far.

        Despite their importance, the genomic diversity of HPAIV before the advent of large-scale systematic surveillance in the 1980s remains poorly understood. To date, only 15 complete genomes from before 1950 have been reconstructed. Historical pathology collections offer a unique window into the evolutionary history of pathogens and provide a valuable resource for studying ancient HPAIV.

        We analyzed 11 formalin-fixed avian specimens from the 1920s to 1950s from Vetmeduni Vienna. High-throughput sequencing yielded IAV reads from five specimens, allowing for the assembly of two high-quality genomes of H7N1 and H7N7 from 1927 and an undated specimen, and lower quality genomes from three specimens collected between 1942 and 1949.

        We are currently enhancing the genomic coverage of these last specimens. Subsequent extensive phylodynamic analyses will allow us to explore the evolution of H7 HPAIV during the first half of the 20th century and understand their contribution to the genetic landscape of later IAV lineages.

        Speaker: Dr Annika Graaf-Rau (Department of Pathogen Evolution, Helmholtz Institut für One Health (HIOH))
      • 4
        Combining natural products, probiotics and vaccines to decolonize multidrug-resistant Enterobacterales from the intestine: An integrative strategy to combat antimicrobial resistance in the One Health context

        The rise of antimicrobial resistance (AMR) in Enterobacterales like Escherichia coli (EC) and Klebsiella pneumoniae (KP) poses a significant threat to human, animal, and environmental health. This calls for innovative strategies that go beyond the traditional use of antibiotics.
        The project MDR-Dekol employs a multifaceted approach combining natural products, probiotics and vaccines to effectively decolonize multidrug-resistant (MDR) EC/KP from chicken intestines. To avoid unwanted disruption of the commensal flora, an advanced pangenomic approach was used to identify surface markers of high-risk (HR) EC/KP lineages for targeted vaccine development. In addition, we employ an in vitro model to identify the most promising prebiotic-probiotic combinations that hinder bacterial biofilm formation and adhesion of HR EC/KP strains to epithelial cells but do not affect commensals.
        We have already identified 3 promising surface markers each for EC and KP. Suitable vaccine candidates are currently being developed. Several natural products with (so far unknown) biofilm eradication properties and promising effects on decolonization especially of HR EC/KP strains were discovered. The combinatorial effects of natural products and probiotics are still under investigation, with some combinations showing synergistic effects.
        In summary, our project offers an alternative strategy to control MDR EC/KP in chickens that is also relevant for other animals, humans and potentially, the environment.

        Speaker: Dr Elias Eger (Epidemiology and Ecology of Antimicrobial Resistance, Helmholtz Institute for One Health, Helmholtz Centre for Infection Research)
      • 5
        ZooNotify - New online portal provides data on the occurrence of zoonotic agents and their antibiotic resistance along the food chain in Germany

        Data on the occurrence of zoonotic agents and related antibiotic resistance in the food chain in Germany are mainly available in reports. This leads to limited accessibility and reusability of this data in science and for risk assessment. ZooNotify makes existing zoonoses data findable, accessible, interoperable and reusable (FAIR).
        ZooNotify currently includes data from 2012 to 2022 collected in accordance with the Directive 2003/99/EC and the Regulation 2160/2003/EC. Data is available on prevalence, antibiotic resistance and typing of zoonotic agents and originates from samples taken at various stages of the food chain, such as primary production, slaughterhouses and retail. The majority of the data is related to livestock and foods of animal origin. However, data is also available on feed, wild animals and plant-based foods.
        ZooNotify allows customized data searches based on user interests. The selected data can be downloaded for further use in a standardized way. ZooNotify also offers quick and easy data visualisation including time-based evaluations. Each section of ZooNotify features clear and concise explanations, ensuring the tool is user-friendly and facilitates a comprehensive understanding of the data.
        The continuous development of ZooNotify supports the EU strategy for open data and forms an important basis for improved risk assessment and the protection of animal and consumer health.

        Speaker: Tasja Crease
    • Session 2: Environmental pollution, climate change and water MOA 6

      MOA 6

      • 6
        Dangerous hitchhikers! - Cigarette filters promote the spread of high-risk bacterial communities in aquatic ecosystems

        In addition to direct human health risks of smoking cigarettes, discarded cigarette filters are an important urban pollutant. Around 37% of filters end up in aquatic environments where they provide novel colonizable surfaces for microbes. We here propose that the toxic compounds entrapped on cigarette filters lead to a selective colonization by bacteria thriving under stressful conditions. These might include pathogens and increased levels of antimicrobial resistant genes (ARGs), hence creating additional indirect health hazards.
        To test this, used cigarette filters enriched with toxicants and unused control filters were submerged in a sewer. Filter colonizing communities were analyzed for ARGs and mobile genetic elements (MGEs). Further, a live pathogenicity model, Galleria mellonella (wax moth larvae) was injected with communities extracted from used and unused filters, with larval survival monitored for 24 hours.
        Larvae exposed to used filter communities had significantly higher mortality rates. Thus, filters enriched with toxicants were selectively colonized by communities with higher pathogenicity. Moreover, >30 tested ARGs and MGEs were selectively enriched on used filters.
        In conclusion, high-risk communities are colonizing discarded filters and can hitchhike large distances to novel environments, where they can negatively affect environmental and human health. This emphasizes the need for improved waste management strategies to mitigate the impact of cigarette filters.

        Speaker: Diala Konyali (TU Dresden, Institute of Hydrobiology)
      • 7
        Tradeoffs of increasing temperatures through climate change on the spread of antimicrobial resistance in river biofilms

        River microbial communities regularly act as the first barrier against the spread of antimicrobial resistance genes (ARGs) that enter through wastewater. However, how invasion dynamics of these ARGs into river biofilm communities shift due to climate change remains unknown. Here, we aimed to elucidate effects of increasing temperatures on both, the natural river biofilm resistome, and the invasion success of foreign ARGs entering through wastewater. Natural biofilms were grown in a low-anthropogenic impact river and transferred to laboratory recirculation flume systems operated at 20, 25, and 30°C. After 1 week of temperature acclimatization, significant increases in abundance of naturally occurring ARGs were detected at higher temperatures. After this acclimatization, biofilms were exposed to a single pulse of wastewater, and invasion dynamics of wastewater-borne ARGs were analysed. After 1 day, wastewater-borne ARGs were able to invade the biofilms successfully with no effect of temperature on their relative abundance. Thereafter, ARGs were lost at far increased rates at 30°C, with ARG levels dropping to the initial natural levels after 14 days. Contrary at lower temperatures, ARGs were either lost slower or able to establish in biofilms with stable abundances above natural levels. Hence, higher temperatures come with contrary effects on river biofilm resistomes: naturally occurring ARGs increase in abundance, while foreign, invading ARGs are lost at elevated speed.

        Speaker: Uli Klümper (TU Dresden)
      • 8
        Building Knowledge in Urban Agriculture Water Supply in São Paulo - The Context of Irrigation, Foodborne Pathogens and Antimicrobial Resistance

        The serious increase of antimicrobial resistance (AMR) in the urban food sectors threatens global health. Urbanization in many developing countries is linked to rising poverty, polluted environments, and growing food insecurity. The World Health Organization estimates unsafe food causes 600 million cases of foodborne diseases and 420,000 deaths annually, mostly due to inadequate sanitation, poor hygiene, and unclean water. Urban agriculture improves local economies, health conditions, and food supply. São Paulo, as one of the world's largest metropolitan areas, is a reference for research on whether irrigation water management can diminish foodborne diseases and reduce the occurrence of AMR. From June 2023 to February 2024, a comparative case-control study on 1500 farms was conducted on 150 urban farms in São Paulo. We interviewed face to face 200 lead farmers using quantitative questionnaire to analyze socio-demographic, health, and environmental data related to their agricultural practices. Water, food and fecal samples were collected, for micro- and molecular-biological (quantitative polymerase chain reaction - qPCR) evaluation of pathogens along the food chain. Preliminary results indicate irrigation water as a contamination source with pathogenic presence varying by sources. Inequalities in technical assistance and water access affect farming decision. Understanding contamination mechanisms in the urban water system, through food into the human microbiome, aims to develop evidence-based guidelines and hygiene recommendations for disease prevention.

        Speaker: Mr Manuel Glass
      • 9
        Intensive land usage induces shift in the bacterial communities and subsequent enrichment of antibiotic resistance genes at different trophic levels of grassland ecosystems

        The intensive use of fertilizers significantly impacts grassland ecosystems, inducing selective enrichment of microbial taxa, which can reduce biodiversity and promote the spread of antibiotic resistance, posing serious health concerns. This study examines the effects of various fertilization regimes on the diversity and function of microbiomes across different trophic levels in grassland ecosystems using metabarcoding. Additionally, the functional potential of these bacterial communities was inferred through imputed metagenomics. Our analysis revealed significant differences in bacterial diversity among grasslands fertilized with biogas, cow manure, and pig slurry, as measured by the Shannon index. These differences were consistent across target organisms, including earthworms, voles, and bumblebees, representing various trophic levels. Metagenomic imputation indicated that gene families related to streptomycin and tetracycline biosynthesis were commonly present and particularly abundant in voles, suggesting an enrichment of antibiotic-resistance genes at higher trophic levels. This study highlights the detrimental effects of intensive land use on microbial communities across multiple trophic levels in grassland ecosystems. The proliferation of allochthonous microbial taxa due to fertilization regimes and the subsequent enrichment of antibiotic-resistance genes at higher trophic levels have serious implications for animal and public health, warranting detailed investigation.

        Speaker: Dr Kunal Jani (Institute of Evolutionary Ecology and Conservation Genomics, University of Ulm)
      • 10
        Pesticide pollution and Schistosoma mansoni tolerance: implications for Schistosomiasis risk in Western Kenya

        Schistosomiasis is a neglected tropical disease caused by trematodes of the genus Schistosoma. The pathogen is transmitted via freshwater snails, which are highly tolerant to agricultural pesticides and indirectly benefit from pesticide exposure due to adverse effects on their less tolerant competitors. Our field studies show that pesticide concentrations deemed “safe” in environmental risk assessments can indirectly affect human health by increasing populations of Schistosoma-host snails. Pesticides in surface waters may thus elevate the risk of schistosomiasis transmission unless they also affect the pathogen. To address this concern, we also tested the tolerance of the free-swimming life stages (miracidia and cercariae) of Schistosoma mansoni to insecticides diazinon and imidacloprid, which were frequently detected in inland waters of Kenya. Additionally, we investigated whether these pesticides impair the ability of miracidia to infect and develop as sporocysts within the host snail Biomphalaria pfeifferi. We showed that these insecticides did not affect the performance of S. mansoni at environmentally relevant concentrations. Particularly within its host snail, the pathogen can escape exposure peaks that have been shown to affect other sensitive invertebrates and their biological control of host snails. Our findings suggest that freshwater pollution with agricultural pesticides increases the risk of schistosomiasis, illustrating the need to integrate environmental and public health risk assessment and management.

        Speaker: Dr Naeem Shahid (Helmholtz Centre for Environmental Research - UFZ)
    • 15:45
      Coffee Break Atrium

      Atrium

    • Keynote II: Lutz Merbold MOA 4 + 5

      MOA 4 + 5

    • Posterslam I MOA 4 + 5

      MOA 4 + 5

    • 17:30
      Break Atrium

      Atrium

    • Session 3: Genomics and Proteomics MOA 4 + 5

      MOA 4 + 5

      • 11
        Significance of the hatchery in the introduction of antibiotic-resistant Enterobacterales into an organic broiler farm

        Despite efforts to minimize antibiotics in conventional broiler husbandry, usage remains high, resulting in high prevalence of antibiotic-resistant bacteria in the animals and meat thereof. With growing dietary awareness, the demand for organic broiler meat and the number of organic broiler farms are increasing. To address knowledge gaps on the occurrence and transmission of antibiotic-resistant Enterobacterales, we conducted a longitudinal study in several organic broiler farms in Germany, monitoring four consecutive fattening periods with five sampling points each. We identified one farm from which ESBL-producing K. pneumoniae and quinolone-, ampicillin-, and/or tetracycline-resistant E. coli were continuously isolated. Resistant isolates were already collected from chick papers of 1-day-old chicks and at later stages of three fattening periods. Transmission dynamics differed between K. pneumoniae and E. coli. K. pneumoniae were clonally related and carried a conjugative IncFIB/FII plasmid harboring genes for resistances against four antibiotic classes. E. coli isolates were genetically highly diverse and carried different resistance plasmids, whose transmissibility is currently being investigated.
        For the fourth fattening period, chicks were purchased from a different hatchery and predominantly susceptible Enterobacterales were isolated highlighting the role of organic certified hatcheries in introducing antibiotic-resistant bacteria into the organic broiler food chain.

        Speaker: Dr Ulrike Binsker (Department Biological Safety, German Federal Institute for Risk Assessment, Berlin, Germany)
      • 12
        MetagenomeWatch: Harnessing Big Data for Pathogen Surveillance and Genome Discovery

        With the rapid advancement in Next-Generation Sequencing technology, the amount of publicly available metagenome sequencing data has increased exponentially. The Sequence Read Archive (SRA) comprises over 9 million genomic sequencing datasets from a variety of environments around the globe, from sources such as human, animal, wastewater, and others. This data provides a unique opportunity to better understand microbial communities and their impact on public health. The ability to search the SRA would improve the surveillance, and characterisation of pathogens, facilitating outbreak detection, and disease spread prevention. Analyzing petabyte-scale datasets such as the SRA has been challenging due to both storage as well as computational requirements. However, with recent advances in compression techniques such as sketching, which uses only a fraction of the original data, being stochastically sufficient to determine whether a given sequence is likely to be contained in a dataset, this is now feasible within a reasonable amount of time and storage space. We introduce MetagenomeWatch, a web interface to the state-of-the-art computational methods implemented by the tool “sourmash”. It offers pathogen experts the ability to search publicly available metagenome databases in real-time for viruses, bacteria, and eukaryotic pathogens. With the option to set up automated watches that report on database updates, it will be possible to monitor and track outbreaks on a global scale.

        Speaker: Dr Daniel Desirò (Robert Koch Institute)
      • 13
        RIBAP: A comprehensive bacterial core genome annotation pipeline for pangenome calculation beyond species level and applications in environmental and wastewater metagenomics

        Microbial pangenome analysis reveals gene presence and absence across prokaryotic genomes. Traditional tools struggle with species exhibiting high sequence diversity or in genus/family-level analyses. RIBAP (Roary ILP Bacterial Core Annotation Pipeline) addresses these issues with an integer linear programming (ILP) approach to refine gene clusters and accurately identify core genes, even in diverse genomes and bacterial communities.

        RIBAP can annotate genomes, calculate pangenomes with Roary, refine gene clusters using ILPs, and visualize results in interactive formats. We tested RIBAP on diverse datasets (Brucella, Chlamydia, Enterococcus, Klebsiella) and metagenome-assembled genomes from wastewater communities. We consistently identified more comprehensive core genomes when analyzing bacterial isolates. In a metagenomic and wastewater context, RIBAP’s performance is challenged by the heavy ILP calculations but can yield core gene connections not detectable by other tools. Our results highlight RIBAP's ability to handle high sequence diversity and accurately predict core genes.

        Thus, RIBAP provides an advanced framework for pangenome analysis, especially for diverse or genus-level datasets, offering a more accurate view of core genetic components such as ARGs. Additionally, RIBAP shows promise for analyzing core genes in microbial communities from environmental samples, such as wastewater metagenomics data.

        Speaker: Dr Martin Hölzer (RKI)
      • 14
        Pan-Proteomics for a Better Understanding of Host Adaptation and Antibiotic Resistance in Public Health-Relevant Bacteria

        The rise of antimicrobial resistance (AMR) in Neisseria gonorrhoeae is a significant threat to public health. The understanding of AMR-driven dynamics is of utmost importance to tackle this challenge. However, current genomic approaches are limited due to predetermined loci in the form of MLST schemes. Pangenomics solves this by extending beyond traditional genomics analyzing the entire gene pool of a species, providing novel information that a single genome cannot. This approach allows us to identify different subclades based on genetic differences and gain insights into horizontal gene transfer, which plays a crucial role in bacterial adaptation and antibiotic resistance. Recent development in the field of bacterial proteomics enabled fast and precise detection of AMR. In this study, we are using state-of-the-art proteomics workflows to gain complementary protein expression and function that can provide additional information about the genes and their clusters beyond their mere presence. Pangenomics and proteomics, each of which provides unique insights into microbial behavior and evolution, are both important fields in the study of bacterial dynamics. By integrating both datasets into Pan-Proteomics, the goal is to uncover mechanisms of host adaptation and resistance that are not apparent when these fields are studied in isolation. This could potentially lead to improved strategies for managing infectious diseases - a significant step in the fight against AMR.

        Speaker: Lena Buchmann (Robert Koch Institut (RKI), MF1 Genome Competence Center, Berlin)
      • 15
        Decoding Bacterial Secrets: Unraveling DNA Modifications through Nanopore Sequencing for Public Health Insights

        Investigating bacterial methylation profiles is critical for assessing the potential influence that DNA modifications have on virulence, antibiotic resistance, and bacteria’s ability to evade the immune system. Real-time Nanopore sequencing and recent advances in basecalling algorithms allow the accurate identification of modified bases from the raw signal data. However, decoding methylation signals for various bacteria remains challenging and needs species-specific investigations.
        Here, we focus on public health-relevant, multi-drug-resistant strains to analyze methylation profiles and extract methylation motifs. Our dataset includes samples from S. aureus, L. monocytogenes, E. faecium, and K. pneumoniae, which were sequenced using the Nanopore GridION platform with recent advances in flow cell chemistry (R10.4.1) and modification basecalling (Dorado).
        Our investigation aims to reveal distinct methylation profiles among the bacterial strains and species, highlighting species-specific modification patterns and shedding light on the most heavily modified genes. Ongoing efforts aim to connect active methyltransferases with methylated motifs, enhancing our understanding of bacterial DNA modifications. Additionally, we evaluate error rates and improve assembly accuracy with the latest basecalling models.
        The primary goal of our study is to explore the potential of recent advanced tools for modification detection and make them accessible to the scientific community.

        Speaker: Valentina Galeone (RKI)
      • 16
        First comprehensive analysis of Wǔhàn sharpbelly bornavirus in grass carp cell lines: A Multi-method Approach

        The family Bornaviridae, within the order Mononegavirales, contains three genera: Orthobornavirus, Carbovirus, and Cultervirus. Orthobornaviruses have the broadest host range, infecting avian, reptilian, and mammalian species, whereas carboviruses and culterviruses have been found in reptiles and fish, respectively.
        Our recent study using in silico data mining identified novel fish culterviruses, including Wǔhàn sharpbelly bornavirus (WhSBV), which was discovered in grass carp kidney and liver cell lines. Further next-generation sequencing of fish cell lines from the cell culture bank of the Friedrich Loeffler Institute revealed nearly identical WhSBV genomes in two grass carp swim bladder cell lines.
        We developed RT-qPCR assays to detect all WhSBV genes and determined the 5' and 3' ends of the WhSBV genome using RACE. In addition, we obtained electron microscopic image of WhSBV in grass carp cell lines. Infection experiments indicated persistent WhSBV infection in cell lines from Cyprinidae and Leuciscidae. Mass spectrometry detected peptides corresponding to the predicted N, X, P, G and M proteins of WhSBV. Northern blot analysis revealed that the X, P, and G genes are expressed from polycistronic mRNA, whereas the N gene is transcribed from monocistronic mRNA.
        Our multi-method approach provides the first molecular characterization of WhSBV and provides critical data for a better understanding of the genetic structure, replication mechanism, and host spectrum of culterviruses.

        Speaker: Mirette Eshak (Friedrich-Loeffler-Institute)
    • Session 4: One Health & Public Health I MOA 6

      MOA 6

      • 17
        Increasing preparedness by networking: 20-year network “Rodent-borne pathogens”

        The network was established as a platform for an interdisciplinary collaboration of scientists in mammalogy, ecology, genetics, immunology, toxicology, epidemiology, virology, microbiology, parasitology and human and veterinary medicine.
        Over the last 20 years, about 30,000 rodents and other small mammals were collected by network collaborators from forests, grasslands, zoological gardens, urban landscapes and pet rat breedings and other husbandries. A variety of pathogen-specific, generic and open view molecular methods were used to detect and identify known, and yet undiscovered, pathogens.
        The network was involved in the discovery of numerous rodent- and shrew-specific herpes- and polyomaviruses. Based on clinical cases in humans, domestic, wild and zoo animals, novel zoonotic and animal pathogens were identified, e.g. variegated squirrel bornavirus 1, squirrel adenovirus and rustrela virus. The network enables the detection of several viruses of unknown zoonotic potential, e.g. novel hepeviruses in rats, common voles and shrews, and the identification of the rodent reservoirs of rustrela virus and lymphocytic choriomeningitis virus. In addition to hantaviruses in wildlife, the zoonotic Seoul hantavirus was recently identified in pet rats in Germany.
        In conclusion, the network provides an important infrastructure for interdisciplinary scientific work in a One Health perspective and allows a holistic view on environment-host-pathogen interactions.

        Speaker: Rainer Ulrich (Friedrich-Loeffler-Institut Bundesforschungsinstitut für Tiergesundheit)
      • 18
        Molecular-based profiling of the gastrointestinal resistome in wild mice

        Antimicrobial resistance represents a global challenge, primarily driven by antibiotic misuse, leading to the spread of bacteria carrying antimicrobial resistance genes in various environments. However, the dynamics and maintenance of antimicrobial resistance genes within host-derived microbiomes remain to be fully understood, especially in hosts with close interactions with the environment and humans. Here, we conducted a molecular profiling of the collection of all antimicrobial resistance genes (resistome) along the gastrointestinal tract of house mice from their natural environment. Utilizing metagenomic sequencing of intestinal content, we assessed the richness and composition of the microbiome and resistome. Microbiome composition accounted for approximately 23% of resistome composition, with gastrointestinal and geographical distance as additional significant predictors. We traced antimicrobial resistance genes associated with transmission risk and anthropogenic impact in the different GI segments. Notably, we identified a high prevalence of genes associated with resistance to glycopeptides, specially the vancomycin resistance genes vanA and vanR, with abundance that steadily increased along the gastrointestinal tract. Our findings highlight the impact of the microbial composition of intestinal niches on antimicrobial resistance gene content. The results suggest a crucial role of interspecies bacterial interactions in maintaining pre-existing resistance independent of direct antibiotic exposure. Microbiomes of mice from their natural environment are a reservoir for resistant bacteria linked to human exposure and an experimentally traceable model.

        Speaker: Dr Víctor Hugo Jarquín-Díaz (Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany)
      • 19
        Applied One Health research on Borna disease virus 1 (BoDV-1) – Introducing the ZooBoFo project

        Borna disease virus 1 (BoDV-1) is known as the causative agent of Borna disease in animals for many decades. However, it was only proven in 2018 that BoDV-1 is also transmissible to humans, causing severe, usually fatal encephalitis. While the reservoir host for BoDV-1, the bicolored white-toothed shrew (Crocidura leucodon), is evidenced, transmission routes remain unknown. After investigating the first detected local cluster of this rare disease in Bavaria in 2022, we continued our research using an applied One Health approach. Our interdisciplinary project “Zoonotic Bornavirus Focalpoint Bavaria” (ZooBoFo) aims to gain further insights into BoDV-1 in order to be able to make more targeted prevention recommendations in the future. Further objectives are to narrow down transmission routes, to gain further insights into the reservoir host and BoDV-1 environmental stability, to precisely describe the regional occurrence of the virus in Bavaria and to further determine clinical symptoms of human BoDV-1 infections. Among other things, a shrew model was established to carry out infection and tenacity tests for this purpose. Moreover, shrews sent in by concerned individuals are continuously tested and if a BoDV-1-positive shrew is detected, an environmental investigation is triggered and we have been able to detect BoDV-1-RNA at several locations. Another aim of ZooBoFo is to raise awareness of BoDV-1 among physicians/veterinarians and to create a platform for informing the public.

        Speaker: Dr Merle M. Böhmer (1 Department for Infectious Disease Epidemiology, Bavarian Health and Food Safety Authority, Munich, Germany 2 Institute of Social Medicine and Health Systems Research, Otto-von-Guericke-University, Magdeburg, Germany)
      • 20
        Education for Sustainable Development as a relevant field for OneHealth: an interdisciplinary approach

        Climate change and environmental degradation alter ecosystems. This generates health risks for human and non-human life. These risks are unevenly distributed across geographical space, human populations, and plant, animal, bacterial, and viral species (Githeko et al. 2000). Global health approaches based on One Health principles thus must focus jointly on human and more-than-human health (cf. Gruetzmacher et al., 2021). Education in regions with high transmission risks for infectious diseases are often characterized by colonial histories and subsequent imbalances of power and knowledge (Tuhebwe et al., 2023). Moreover, knowledge transmitted through schools and colleges is human-centered. It perpetuates non-systemic, dualistic models of thinking about health risks associated with climate and environmental change (Ress et al., 2022). The proposed paper introduces education for sus-tainable development (ESD) in postcolonial constellations. ESD plays a key role in tackling OneHealth challenges globally. Based on general insights about learning, it can help shape interdisciplinary collaborations (e.g., human and veterinary medicine) by conceptualizing such collaborations as spaces of learning in the context of uncertainty (cf. Taube & Ress, forthcoming). The proposed paper reflects a model for interdisciplinary learning. It constitutes itself an interdisciplinary collaboration between scholars in epidemiology and education sciences.

        Speaker: Susanne Ress
      • 21
        Development of a standardized One Health surveillance of antimicrobial resistance and heavy metal tolerance in wild birds and surface waters of the Baltic Sea

        The spread of antimicrobial resistance (AMR) and heavy metal tolerance (HMT) in microorganisms of wildlife and aquatic environments is an often underestimated but detrimental threat to human, animal, and environmental health. Therefore standardized and feasible strategies are needed for comprehensive monitoring of AMR/HMT bacteria and related genes.

        The project BALTIC-AMR employs a longitudinal surveillance approach combining molecular (e.g. genomics) and phenotyping (e.g. culturomics) techniques to characterize AMR/HMT Enterobacterales and Pseudomonadales from the Baltic Sea from four littoral states. In addition, the occurrence of other water-borne pathogens such as Vibrio spp. will also be investigated. To study migratory birds as a connected reservoir in the Baltic region, we also examine fecal samples of selected gull and duck species and include movement data in our comprehensive analysis.

        A protocol for reliable AMR/HMT monitoring has been established by the interdisciplinary consortium. Although the longitudinal sampling is ongoing, we have already isolated a large number of AMR/HMT strains from surface waters and wild bird droppings, many exhibit resistance to at least three clinically used antibiotic classes and thus exhibit a multidrug-resistant phenotype.
        In summary, this is a proof-of-concept study that demonstrates how a standardized and reliable AMR surveillance strategy can be established within the One Health context.

        Speaker: Max Sittner
      • 22
        Building up One Health Surveillance systems in the African tropics and in Germany

        The Helmholtz Institute for One Health (HIOH) is dedicated to addressing the interdependence of human, animal, and ecosystem health and how they are affected by climate change.
        The One Health Surveillance (OHS) Core Unit is at the heart of the institute. Similar to weather stations that record various parameters over many years, the OHS systematically collects and analyzes a broad spectrum of samples and data – from climate and environment to health and social data – at high resolution in defined indicator regions. The integration of this data from multiple disciplines will contribute to pandemic prevention and preparedness. In addition to the indicator regions in sub-Saharan Africa (Côte d’Ivoire (CIV) and Central African Republic (CAR)), the OHS will be active at the doorstep of the HIOH in northeast Germany.
        Clinical Surveillance: To determine which pathogens are relevant to the populations in the indicator regions, we are establishing clinical surveillance in collaboration with local hospitals and health centers.
        Wildlife Disease Monitoring: We are running veterinary programs to detect wildlife diseases in CIV and CAR using non-invasive methods such as the analysis of urine and fecal samples, or necropsies of wild animals found dead.
        One Health Cohorts: Currently we are setting up cohort studies with human volunteers whose health we will follow over the course of many years. In addition, we systematically survey associated livestock and small mammal vectors, biodiversity and micro - and macro climate.

        Speaker: Fee Zimmermann (Helmholtz Institut für One Health)
    • Snacks & Poster Viewing Atrium

      Atrium

    • Registration Atrium

      Atrium

    • Keynote III: Simone Sommer MOA 4 + 5

      MOA 4 + 5

    • Session 5: Biodiversity & Human Health MOA 4 + 5

      MOA 4 + 5

      • 23
        Macaques, humans & oil palm: an integrative approach to foster ‘One Health’ in Malaysian plantations

        One major factor contributing to biodiversity loss is the conversion of rainforest into oil palm plantation. In Malaysia, pig-tailed macaques (Macaca nemestrina) have adapted to this new environment, daily entering plantations adjacent to their remaining forest habitat. There, macaques feed on palm fruits but also provide an ecosystem service by reducing plantation rats, the main pest species of oil palm crops. Our research uncovered significant impacts on macaque fitness in this landscape matrix. Recently, we reported a high mean infant mortality rate (~60% over 10 years), with prolonged plantation visits increasing risks of infant death. Our results suggest that the exposure to agricultural chemicals may play a significant role. Some herbicides gradually accumulate in mammals, increasing pesticide exposure for developing offspring, especially those born to first-time mothers and after prolonged interbirth intervals. Indeed, our data show changes in macaques’ chemical profiles following herbicide exposure. Further, human-primate interactions pose risks to humans through the potential transmission of zoonotic diseases. Gut microbiome data of macaques revealed links between frequent human interactions and the diversity and richness of potentially pathogenic bacteria. Our future work aims at developing an ‘One Health’ project to assess the impact of anthropogenic activities on the health of wildlife, humans, and the environment, exemplified by the shared oil palm landscape.

        Speaker: Anja Widdig
      • 24
        The ecology of species-specific and ubiquitous rodent-borne pathogens in the network of mammalian biodiversity and seasonality

        Many rodent species are reservoirs for zoonotic pathogens. Biodiversity could be closely related to pathogen prevalence through direct and indirect effects and all actors are probably affected by seasonality (vegetative period, food, shelter, competition, predation etc.). Increased biodiversity should modulate transmission of pathogens in the small mammal host species. It can minimize transmission of species-specific pathogens (dilution) and enhance the transmission of ubiquitous pathogens (amplification).
        We investigated the relative impact of population-level regulation of the prevalence of species-specific Puumala orthohantavirus (PUUV) in bank voles and Tula orthohantavirus (TULV) in common voles and the ubiquitous Leptospira in the small mammal community. Rodents were trapped in grasslands and forests, necropsied and tissue and blood samples analyzed for pathogen infections. The biodiversity of large terrestrial mammals was monitored with wildlife cameras.
        There was a strong influence of seasonality on vole host abundance and pathogen prevalence. For instance, in some seasons, shrew abundance was negatively correlated to bank vole abundance. Host abundance promoted PUUV seroprevalence. Increased abundance of non-host small mammals reduced PUUV seroprevalence in the host. Vole abundance in summer was a strong driver of TULV prevalence. In summer, predator abundance decreased and mammal biodiversity increased Leptospira prevalence in the community. The results indicate the expected dilution and amplification effects and suggest a complex network that considerably depends on season.

        Speaker: Jens Jacob
      • 25
        Extensive diversity of viruses identified in mosquitoes from Southwestern Uganda

        Mosquitoes are vectors for pathogenic viruses, such as Dengue or Zika viruses. Although over 3,700 mosquito species exist, only about 200 have been tested for viral infections, mostly anthropophilic mosquitoes as Aedes aegypti or Ae. albopictus. In addition, knowledge about viruses emerging from enzootic to epidemic transmission cycles remains limited. Our study aimed to assess the genetic diversity of viruses circulating in mosquitoes in primary and adjacent rural ecosystems in Uganda and to analyse emergence patterns.
        In 2019 and 2020, we collected 39,834 mosquitoes from lowland rainforest, savannah gallery forest, montane rainforest, and adjacent rural areas in southwestern Uganda. An extraordinary richness of 139 mosquito species was found, including many species that have never been tested for viruses before. We observed that mosquito communities significantly differed between the three ecosystems and for each ecosystem between undisturbed and disturbed areas. Testing for viral infections revealed 709 viruses belonging to 158 species. Most of these showed less than 90% nucleotide identity to known viruses, suggesting the detection of numerous previously unknown viruses belonging to the families Toga-, Flavi-, Mesoni-, Peribunya-, Reo-, Rhabdo- and Phenuiviridae.
        Intriguingly, diversity of mosquitoes and viruses was found to be temperature-dependent and varied significantly among ecosystems with highest species richness found in the warmest undisturbed primary ecosystems.

        Speaker: Selina Graff
      • 26
        CiFly – a fly-based Citizien Science project with pupils to monitor mammal biodiversity and anti-microbial resistance (AMR)

        In this project, we develop and evaluate an educational measure to integrate One Health and hands-on science into the curriculum of a local high school in the Vorpommern region. The measure is designed as a sustainable citizen science project with pupils to investigate the impact of urbanisation and agriculture on wild mammal biodiversity and antimicrobial resistance in the environment. Together with scientists, pupils develop a fly-based monitoring of wildlife and AMR in Vorpommern-Greifswald based on state-of-the-art methods in the field of environmental DNA and bacteriology. As flies thrive in many environments and feed, amoung others, on mammal faeces or carcasses, ingesting DNA from different sources, we use them as samplers of DNA from wildlife and bacteria. The project aims to improve the pupils' general understanding of science, convey the basics of the One Health concept and generate scientific data. The first group of pupils started the project in March 2024, caught 222 flies and will complete 12 lessons of lectures and experimental sessions with a final lesson, presenting their results to a panel of scientists and their teacher. To assess the project’s impact on the pupils' attitudes and confidence in science, they completed a questionnaire before the project began, which will be repeated at the end of the project.

        Speaker: Dr Johanna Maria Eberhard (Helmholtz Institute for One Health, Helmholtz Centre for Infection Research )
      • 27
        Effects of tree diversity on human health – results from the Dr.Forest project

        Forest risks and benefits to human health are widely recognised. Yet, the role of forest characteristics – such as tree species diversity or canopy density – driving health effects remains underexplored. Based on data from a European forest network (Dr.Forest), we quantified causal pathways relating different forest types to physical and mental health. Specifically, we determined i) mental wellbeing via visual cues and ii) via auditory cues; iii) thermal comfort; iv) polyphenol content of medicinal plants; v) nutritive qualities of mushrooms, vi) air quality, and vii) ticks and Lyme disease.
        Results show that forests generate net health benefits regardless of their ecological characteristics, except for the high tick prevalence compared to other ecosystems. Canopy density and tree species diversity emerge as key drivers, but their effect size and directionality are strongly pathway-dependent. Changes in canopy density can generate trade-offs; e.g., forests optimised for heat buffering may enhance Lyme disease prevalence. Tree diversity effects were weaker but more consistently positive.
        Forest management may enhance health benefits and mitigate risks. Even when modulation effects are small, this can translate to considerable impact at the public health level. Importantly, management should account for trade-offs to tailor forest biodiversity and functioning to local public health needs of priority.

        Speaker: Michael Scherer-Lorenzen
      • 28
        Frontiers in using environmental DNA to explore disease ecology and the sources of disease outbreaks

        The detection of environmental DNA (eDNA), the traces of DNA that animals leave in their environments, has revolutionized animal monitoring in aquatic ecosystems over the last decade. Recent advances are making eDNA from a diversity of substrates (e.g., air, vegetation swabs, carrion flies) increasingly relevant for the detection of animals in terrestrial ecosystems. In some cases, these same substrates also allow for the (co-)detection of pathogens and animals, creating tools to study the distribution, genetic diversity, and even host ranges of pathogens. We present a brief review our own work on this topic, as well as an exploration of the broader literature, to highlight that eDNA is now a tool ripe for studying the ecology of pathogens, as well as the origins of zoonotic disease outbreaks. Indeed, we argue that eDNA has already played a role in our understanding of the origins of two of the major emerging zoonotic infectious diseases outbreaks of the last decade, the West African Ebola virus epidemic and SARS-CoV-2. We suggest the time is right for eDNA to become a stable part of the One Health practitioner’s toolkit.

        Speaker: Dr Jan Frederik Gogarten (Helmholtz Institute for One Health, Helmholtz-Centre for Infection Research (HZI))
    • Session 6: Vectors I MOA 6

      MOA 6

      • 29
        Urban Distribution and Abundance of Aedes albopictus in Response to Climate Change: A Model Incorporating Temperature and Land-Cover

        Mosquitoes, particularly Aedes albopictus, pose health and economic risks. Recently, this invasive species has spread to central Europe, including Germany, raising concerns for uninvaded areas like Bavaria due to climate change. The mosquito’s survival and reproduction can fluctuate across fine spatial resolutions, often overlooked in studies. Urban areas show this variation in habitat suitability. With Ae. albopictus favoring urban habitats, understanding their urban distribution is crucial for public health.
        Eight Bavarian cities with similar climates, helping to isolate the effect of land-cover, and high populations were selected for study: Munich, Nuremberg, Augsburg, Regensburg, Ingolstadt, Fürth, Bayreuth, and Würzburg. All are along a highway.The modeling was based on the mechanistic model dynamAedes (Da Re et al., 2022). Random Forest machine learning approaches were employed to model air surface temperature based on MODIS Land surface temperature (Moderate Resolution Imaging Spectroradiometer) for the study areas as well as producing novel land-cover maps (grids 30 x 30 m, validated by confusion matrix and an out of bag score) as input data. A weighting of the land-cover classes based on an expert survey was applied.
        The study revealed a positive correlation between the probability of Ae. albopictus establishment and the proportion of urban green spaces within city boundaries. Climate change was found to intensify this correlation, further favoring urban green spaces over built-up areas for Ae. albopictus establishment. Moreover, climate change was observed to increase the overall abundance of Ae. albopictus across all study areas. The disparity in both abundance and establishment probabilities between the climate scenario RCP 2.6 and RCP 8.5 is significant. In fact, transitioning from RCP 8.5 to RCP 2.6 could potentially result in a more than five-fold reduction in mosquito abundance numbers.
        The current absence of Ae. albopictus from these cities despite their suitability for the species highlights the importance of continued monitoring programs to prevent populations from establishing or reduce their abundance.

        Speaker: Stephanie Thomas
      • 30
        Analyzing the Complexity: interactions of arboviruses and insect-specific viruses in vector competence studies with two invasive Aedes species

        The global spread of invasive mosquito species increases the risk of arbovirus infections in humans and lifestook. Besides Aedes albopictus; Aedes koreicus and Aedes japonicus have recently spread within Central Europe. Extensive information on the vector competence of the latter two species is missing. Therefore, we evaluated the vector competence of field caught Ae. koreicus and Ae. japonicus mosquitoes from Germany for different arboviruses. Additionally, the virome of the examined mosquitoes was investigated, and the impact of co-infection with arbovirus and insect-specific virus (ISV) on vector competence was evaluated.

        Mosquitoes were orally infected and incubated under different temperature conditions (27 ± 5◦C; 24 ± 5◦C, 21 ± 5◦C), to evaluate the influence of temperature on transmission dynamics. A salivation assay was performed to investigate the vector competence. Using a whole virome analysis via NGS, novel insect-specific viruses (ISVs) were identified.

        Ae. koreicus had the potential to transmit Chikungunya virus (CHIKV) and Zika virus (ZIKV) but not West nile virus (WNV). Transmission was only observed at the higher temperature with low transmission efficiency. Ae. japonicus was able to transmit CHIKV, Sindbis virus (SINV) and western equine encephalitis virus (WEEV). The risk of CHIKV transmission by Ae. japonicus was also rather low and exclusively observed at the higher temperature. Transmission of SINV and WEEV was observed at all investigated temperature profiles with high transmission efficiency.

        While we detected only one ISV in Ae. koreicus, we could identify seven ISVs in Ae. japonicus. Correlating the ISV infection status of individual specimens with their transmission capability for specific arboviruses revealed a potential interference of ISVs on arbovirus infection, characterized by highly complex dynamics.

        Jansen S, Cadar D, Lühken R, et al. Vector Competence of the Invasive Mosquito Species Aedes koreicus for Arboviruses and Interference with a Novel Insect Specific Virus. Viruses. 2021;13(12):2507. Published 2021 Dec 14. doi:10.3390/v13122507

        Speaker: Stephanie Jansen (UHH/BNITM)
      • 31
        Recent and future ecological niche changes of major Anopheles species in the European and Mediterranean areas

        Anopheles species are the main vectors of malaria, and changes in their habitat due to climate or land-use also alter the disease risk. Especially in the Mediterranean, a climate hotspot, changes in temperature and precipitation could substantially modify habitat suitability in the coming decades. In addition, ongoing land-use changes such as urbanization, deforestation and irrigation have also significant impacts e.g., on host availability and breeding sites.
        38 climate and 14 land-use predictors were considered to model the recent and future ecological niches of An. atroparvus, An. labranchiae, An. messeae, An. sacharovi, An. sergentii, and An. superpictus using Boosted Regression Trees (BRT). For each species we established 16 different models based on four different sets of predictors and four different background selection methods. Changes in the ecological niches are assessed by a model ensemble and the best model in terms of model skill and model transferability. Furthermore, the most important predictors for vector occurrences and the predictors that are responsible for changes in the ecological niches were identified for each species.
        Results show that, on a large scale, climate predictors are generally more important than land-use predictors for the establishment of the species. The expected changes will help some species to expand, while leaving others unable to cope. Depending on the species, habitat suitability depends on different subgroups of climatic predictors.

        Speaker: Christian Merkenschlager (Universität Augsburg)
      • 32
        Genotypic and Phylogeographic Insights into a Pre-Epidemic Variant of Wesselsbron Virus detected in sylvatic Aedes mcintoshi from Semuliki Forest, Uganda

        Wesselsbron virus (WSLV) is a neglected mosquito-borne virus within the yellow fever sub-group of the Orthoflavivirus genus in the Flaviviridae family. Primarily a veterinary pathogen causing stillbirths, congenital malformations, and mortality in ruminants, WSLV also infects humans, causing a self-limiting febrile illness with rare neurological complications. WSLV causes sporadic outbreaks in Southern Africa but findings in other African countries suggest a wider distribution. We detected and isolated WSLV from an Aedes mcintoshi mosquito in Semuliki National Park, Uganda. The novel strain showed impaired infectivity across various host cell lines compared to an epidemic reference strain from South Africa. Complete genome sequencing and phylogenetic analysis revealed that the virus grouped with strains from South Africa and Senegal. Phylogeographic reconstruction of WSLV’s spatial and temporal spread combined with displacement of the origin of the newly detected strain suggest that WSLV may be widely distributed across Africa. Our data show that the geographic distribution of WSLV and its impact on human and animal health is likely underestimated.

        Speaker: Mr Georg Joachim Eibner (Institut für Virologie - Charité Universitätsmedizin Berlin)
      • 33
        Effects of rodent control in sewer systems on zoonotic pathogen composition and prevalence in Norway rats (Rattus norvegicus) - a pilot study

        Several rodent species can be problematic in cities and agricultural landscapes and they are carriers of zoonotic pathogens. Some of these pathogens can cause serious disease in humans, livestock and pet animals. Commensal rodents in urban areas pose a considerable threat to human health by transmitting zoonotic pathogens. Rodent management – often using anticoagulant rodenticides – is often applied in such situations to reduce their abundance. However, management itself may disturb rodent population structure and cause influx of new individuals, potentially with new suits of pathogens. We monitored pathogen occurrence in Norway rats (Rattus norvegicus) caught in the Berlin sewer systems and above ground before and after regular rodent management action. We included viruses [hepatitis E virus (HEV), Seoul hantavirus, intestinal viruses, Mpox virus], bacteria [Leptospira spp., Bartonella spp., Salmonella spp., Campylobacter spp., pathogenic Escherichia coli] and endoparasites [Babesia spp., Toxoplasma gondii]. Prevalences were low for HEV (5%), Toxoplasma (2%) and Mpox (1%). Seoul virus, Shigatoxin-producing E. coli and Salmonella were absent, enteropathogenic E. coli prevalence was 10% and Campylobacter jejuni prevalence was about 20%. More results including habitat effects will be presented. Our study contributes to a better understanding of the presence and transmission risk of rodent-borne pathogens in urban areas in the context of rodent management actions.

        Speaker: Mr Florian Hüls (Julius Kühn-Institut, Bundesforschungsinstitut für Kulturpflanzen, Institut für Epidemiologie und Pathogendiagnostik, Nagetierforschung)
      • 34
        Bluetongue virus serotype 3 (BTV-3) and Schmallenberg virus in Culicoides biting midges, collected in western Germany during the BTV-3 outbreak in neighbouring Netherlands, autumn 2023

        After the outbreak of bluetongue virus serotype 8 (BTV-8) from 2006-2009, bluetongue disease occurred in central Europe only sporadically. By contrast, Schmallenberg virus (SBV) has become enzootic since its first detection in 2011. Both viruses are transmitted by Culicoides biting midges (Diptera: Ceratopogonidae). In September 2023, BTV-3 emerged for the first time in continental Europe. From an initially affected four farms in the Netherlands, it rapidly spread, soon reaching Belgium, the UK and Germany. During September 24-26, i.e. before any of BTV-3 cases had been reported from Germany, we installed UV-light traps in cattle, sheep and goat stables on 18 farms (1 trap per farm) in the federal states of Lower Saxony and North Rhine-Westphalia to collect putative BTV-3 vectors and test them for virus RNA. Captured biting midges were morphologically identified as C. obsoletus group, C. pulicaris complex, and other Culicoides. Quantitative real-time PCRs were used to screen pools of ≤50 individuals for BTV, SBV and epizootic hemorrhagic disease virus (EHDV). BTV-3 was detected in a pool of C. obsoletus group biting midges collected on October 12, 2023, in the same district where the first infected sheep was observed. SBV-RNA was found in numerous Culicoides pools, reflecting extensive regional circulation with substantial virus loads. All pools tested negative for EHDV. BTV-3 circulation must be expected to pick up speed during spring and summer 2024 in Germany.

        Speaker: Anja Voigt (Leibniz-Centre for Agricultural Landscape Research, Müncheberg, Germany)
    • 11:00
      Coffee Break & Poster Viewing Atrium

      Atrium

    • One Health Career get-together to be announced

      to be announced

    • Keynote IV: James Hassell MOA 4 + 5

      MOA 4 + 5

    • Posterslam II MOA 4 + 5

      MOA 4 + 5

    • 12:45
      Lunch & Poster Viewing Atrium

      Atrium

    • Session 7: Data modeling and AI MOA 4 + 5

      MOA 4 + 5

      • 35
        Computer vision and artificial intelligence as emerging diagnostic tools for highly accurate classification of clinical fungi

        Background
        Artificial intelligence (AI) based on deep-learning (DL) approaches enables computers to learn from large datasets to replicate human intelligence. DL circumscribes a diverse set of algorithms, which have demonstrated state-of-the-art performance in biomedical applications for accurate classification of image data. In the advent of a perpetual AI revolution taking place at an intangible pace, microbial diagnostics utilizing DNA and proteins as primary data sources to characterize fungal pathogens, would greatly benefit from computer vision to increase precision, reduce costs, and to further democratize accurate diagnostics in low-income countries. The methodology offers a supplementary (developmental) diagnostic opportunity also universally expandable to prokaryotes, to contribute to a successful long-term implementation of the One Health approach.
        Methods
        We developed a supervised learning classifier based on convolutional neural networks, with pre-sets of transfer learning, to identify fungal pathogens directly from high-resolution petri-dish image data. Core of the dataset is the yet largest and most precisely photographed set of medical fungi, derived from the Atlas of Clinical Fungi encyclopedia. The tested image subset comprised >100 species as a proof of principle.
        Results
        Utilizing different transfer learning approaches, and building on neural network architectures, such as VGG16 or Efficientnet trained on millions of random images, we successfully classify all target pathogens with a varying degree of accuracy, and depending on experimental settings, of up to >95%. Our results successfully benchmarks a simple, powerful method, and is the yet most comprehensive study in microbial diagnostics utilizing computer vision to date.
        Conclusion
        The AI revolution will enhance and open possibilities for clinical diagnostics at a yet unseen pace. We intend to further train and fine tune our classifier on all known fungal pathogens and opportunists to create a functional tool for identification of clinical fungi.

        Speaker: Dr Johannes Benjamin Stielow (Helmholtz Institute for One Health)
      • 36
        Progress towards predicting significant changes in wastewater microbial communities: A machine learning approach using time series data

        Changes in the human microbiome can be an indicator of the onset of an infectious disease such as sepsis whereas fluctuations of the microbes observed e.g. in wastewater might indicate a disease outbreak in the respective catchment area. The capacity to distinguish these significant changes from naturally occurring fluctuations in the microbiome could facilitate the early detection of potentially harmful diseases. We aim to take a step toward this prediction of microbial abundance trends based on analysis of 16S rRNA data. To this end, we apply Long Short-Term Memory (LSTM) models to publicly available long-term microbial time series data from two healthy subjects. To increase the explanatory power of the model, Shapley Additive Explanations (SHAP) is used for feature significance analysis.
        Thus far, the model has demonstrated superior performance compared to other tested model architectures, including VARMA and Random Forest. It has also exhibited promising results in predicting the normal abundances in a sewer in the Ruhr area. Further testing of additional architectures is planned. Overall, the aim is to provide a treatment advantage to physicians and patients by providing a system for the early detection of potential health risks. In an environmental context, a machine learning-based early warning and monitoring system could assist with decision-making in the event of public health concerns.

        Speaker: Ann-Kathrin Dörr
      • 37
        Exploring the human gut microbiome from over 500 thousand genomes: Insights into functional potentials and biodiversity via Dense Deep Clustering

        We compiled 509,610 metagenome-assembled genomes (MAGs) and their metadata to create human gut microbiome fingerprints, distinguishing functional potential by country of origin and disease condition. We selected 14,082 metagenomes from the HumanMetagenomeDB, recovering over 300,000 MAGs. Our database included 154,000 MAGs from Pasolli et al. (Cell, 2019) and 60,000 from Nayfach et al. (Nature, 2019). Metadata was unified into a standardized dataset, and MAGs were dereplicated into 6,794 species. Gene annotation was performed using MuDoGeR and Prokka. After one-hot encoding of MAGs from adult gut samples, we had 426,648 MAGs and 40,424 non-redundant genes. An autoencoder neural network with dense layers and ReLU activation was applied, followed by DenMune clustering to capture the metagenomic fingerprint. We analyzed MAG distribution in the new feature space by taxonomy, geographical origin, and host medical condition. The embedded space revealed different density profiles, capturing relevant microbiome metadata. These profiles led to new findings, such as distinct profiles between samples from healthy subjects and cancer patients (PERMANOVA < 0.05). The embedded space also separated libraries by country, demonstrating robustness. MAG clusters based on functional potential identified taxonomical groups diverging from their majority cluster, indicating differences in microbial strains. Our findings could be the basis for developing predictive models for health and disease.

        Speaker: Dr Ulisses Rocha (Helmholtz Centre for Environmental Research)
      • 38
        From Variance to Unity: A NFDI4OneHealth Data Infrastructure Proposal

        The increasing complexity of global health challenges necessitates a comprehensive and integrated approach, recognizing the intricate interdependencies between human, animal, and environmental health. This proposal seeks to establish a robust national research data infrastructure that bridges these domains, facilitating seamless data integration and collaborative research.
        NFDI4OneHealth will increase interoperability and integrate various data standards from different fields, leveraging existing efforts within the NFDI framework, including NFDI4Health, NFDI4Microbiota, NFDI4DataScience, NFDI4Biodiversity, and NFDI4Earth. This synergy will enable the creation of machine-actionable data repositories, standardized data models, and advanced analytical tools capable of handling the high variance in data types inherent to One Health research. Emphasizing FAIR (Findable, Accessible, Interoperable, Reusable) data principles, NFDI4OneHealth will ensure that data is optimally managed and utilized.
        Key challenges such as developing interoperable One Health data models and implementing combined time series and multilevel multi-factor analyses across diverse research areas will be central to this initiative. The interdisciplinary approach will enhance data handling capabilities, crucial for rapid responses to global health crises. The NFDI4OneHealth will provide a comprehensive platform, ultimately contributing to a more sustainable and resilient global health ecosystem.

        Speaker: Dominic Bläsing (Helmholtz Institute for One Health )
    • Session 8: Vectors II MOA 6

      MOA 6

      • 39
        West Nile virus infection in mosquitoes from Berlin, 2023

        West Nile virus (WNV) is an arbovirus that is maintained in nature through a transmission cycle involving birds and mosquitoes. Zoonotic human infection can lead to West Nile Fever and West Nile neuroinvasive disease. Since 2018, WNV is endemic in eastern Germany including Berlin. Here, we aimed to get insight into urban areas that favour WNV amplification in Berlin.
        We collected mosquitoes in Berlin at five sites (< 1 km distance): a sponge city site (S), a residential courtyard (RB), a park-like residential area (RA), a park-like cemetery (C) and a natural conservation area (N). Mosquitoes were collected for four consecutive nights once per month from Jun-Sept 2023, identified morphologically and tested for WNV infection by qPCR.
        In total, 13.627 mosquitoes were collected, with highest densities at N (n=4202) and C (n=3927), and lowest at RB (n=669). For all sites, the main vectors of WNV were the most prominent (up to 92%). WNV was detected in 78 out of 1625 mosquito pools. While mosquito density peaked in July, WNV infection rates peaked in August, with the highest minimum infection rate (MIR) at C (2.08 %) and RA (1.46%), and surprisingly low MIR at N (0.26% in August). All detected WNV strains contained unique Berlin-specific nucleotide variants and emerged from local amplification.
        An unexpected high WNV infection rate in mosquitoes at RA and C in Berlin in 2023 was found, compatible with infection rates detected during outbreak situations in other European countries.

        Speaker: Dr Corinna Patzina-Mehling (Charité Universitätsmedizin Berlin)
      • 40
        Quo vadis West Nile virus? – Auguring viral expansion through wild bird monitoring

        West Nile virus (WNV) is an arthropod-borne flavivirus with almost worldwide distribution and an increasing number of human infections in Europe in recent years. WNV circulates in an enzootic cycle between Culex mosquitoes and birds with occasional transmission to dead-end hosts like humans, where it can cause severe disease and even death in some cases.
        In 2021 and 2022, more than 2300 blood and 3100 sets of organ samples were collected from birds by members of the wild bird-associated zoonoses network (WBA-Zoo) in order to monitor the distribution of WNV and the closely related Usutu virus (USUV) in the German (wild) bird population. Specimens were analysed by molecular and serological methods.
        Both WNV and USUV increased in circulation in 2022 compared to 2021. This enabled WNV, hitherto considered to be enzootic only in the central-east of Germany, to emerge e.g. further north in Hamburg. Phylogenetic analyses revealed a continuous dominance (95% of generated sequences) of one subcluster of WNV lineage 2, which has maintained the infection process over the last five years. New serological data also confirm a low level of enzootic transmission of the virus outside the endemic regions, without detection of acute infections in animals or humans so far.
        In conclusion, WNV is “on the move” and wild bird surveillance constitutes a suitable early-warning system for potential zoonotic spillover events in a One Health approach.

        Speaker: Franziska Schopf (Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Institute of Novel and Emerging Infectious Diseases, Greifswald-Insel Riems, Germany)
      • 41
        Spatial and temporal analysis of West Nile Fever in Germany using a process-based epidemic model

        Since the first autochthonous transmission of West NileVirus was detected in Germany, it has become endemic in several parts of the country and has been spreading due to the attainment of a suitable environment for vector occurrence and pathogen transmission. Increasing temperature associated a changing climate has been identified as a potential driver of mosquito-borne disease in temperate regions. We developed a process-based epidemiological model driven by environmental and epidemiological data. Functional traits of mosquitoes and birds of interest were used to parameterize our model appropriately.Air temperature, precipitation, and relative humidity were the key climatic forcings used to replicate the fundamental niche responsible for supporting mosquito population and infection transmission risks in the study area. An inverse calibration method used to optimize our parameter selection. The epidemiological model was used to simulate spatial and temporally explicit vector population,basic reproductive number (R0)maps showing dynamics of WNV infection on birds, number of infected birds and humans across Germany. Epidemiological data for human infections sourced from Robert Koch Institute and animal cases collected from the Animal Disease Information System(TSIS) of the Friedrich-Loeffler-Institute were used to validate model-simulated transmission and infection rates. This study presents a path for developing an early warning system for vector-borne diseases driven by climate change.

        Speaker: Mr Oliver Mbaoma (University of Bayreuth)
      • 42
        Experimental Infection of Mosquito Larvae: A Novel Approach for Vector Competence Studies

        Vector competence studies are a valuable method in arthropod-borne virus (arbovirus) research, as they can provide knowledge about transmission routes and virus-vector-interactions. Due to the geographical co-circulation of arboviruses, studies on co-infections in mosquitoes have become increasingly important, but are difficult to implement because of the low feeding and survival rates of mosquitoes under laboratory conditions. An alternative method could be the experimental infection of mosquito larvae, resulting in the emergence of infected adults that can be used for further infection studies.
        A total of 300 Culex pipiens biotype molestus larvae were placed for 24 hours on mammalian cells (Vero, n=150) or mosquito cells (C6/36, n=150) infected with Usutu virus (USUV). Larvae were then raised to adults. 15 days after infection, saliva was collected from all living females. Mosquito bodies and saliva were then investigated for USUV RNA via RT-qPCR.
        The results showed that the mosquito larvae acquired USUV from the infected cells and were very susceptible to an infection. A total of 19/22 (Vero) and 39/40 (C6/36) adult females were infected with USUV. Viral RNA was also detected in the saliva samples of 17/22 (Vero) and 31/40 (C6/36) animals. An experimental infection of mosquitoes in the larval stage thus offers the opportunity to produce infected adult animals, which can be used for vector competence studies to investigate viral interactions in the mosquito vector.

        Speaker: Christin Körsten (Friedrich-Löffler-Institut, Institut für Infektionsmedizin)
    • 14:45
      Break Atrium

      Atrium

    • Members Assembly OHP & Election SAB MOA 4 + 5

      MOA 4 + 5

    • 16:45
      Break https://hotel-moa-berlin.de/?lang=en

      https://hotel-moa-berlin.de/?lang=en

      Mercure Hotel MOA Berlin

      Stephanstr. 41 10559 Berlin
    • Panel Discussion (in German) MOA 4 + 5

      MOA 4 + 5

    • 18:30
      Break & Poster Viewing Atrium

      Atrium

    • Social Dinner MOA 8 + 9

      MOA 8 + 9

    • Keynote V: Chadia Wannous MOA 4 + 5

      MOA 4 + 5

    • Session 10: Infectious Diseases and Zoonoses II MOA 4 + 5

      MOA 4 + 5

      • 43
        Targeting viral and host factors against zoonotic RNA viruses

        Zoonotic diseases caused by RNA viruses have gained huge public interest since the SARS-COV-2 pandemic. Currently, avian influenza viruses (AIV) of Subtype H5N1 are being detected in many different mammalian species worldwide, which may increase the chance of a virus adaptation to humans. Most of these viruses show already different degrees of resistance against the common antiviral drugs. In 2023, the highly pathogenic Nipah Virus, a member of the genus Henipavirus which has been reported to infect pigs and humans since 1998 with fatality rates of 40-75%, re-emerged and caused human cases in India. Up to now there are no drugs or vaccines available against Nipah virus. Both RNA viruses could potentially play a role in future pandemic outbreaks which would require therapeutic interventions. To counteract these pathogens, we are exploiting antiviral drug combinations that not only rely on viral targets, but also on host factors. We have therefore combined different nucleoside analogues with pyrimidine synthesis inhibitors targeting enzymes which are essential for the intracellular generation of CTP to achieve exquisite synergy against virus propagation. AIVs of H5 subtypes as well as different porcine influenza viruses with zoonotic potential have been tested in vitro for their sensitivity against different drug combinations. So far, we found all tested strains to be sensitive to drug treatments at varying degrees. Similar results could be found for Cedar virus, a low pathogenic henipavirus. To further validate this drug efficacy in vivo, animal trials in ferrets with H5N1 are currently ongoing. In conclusion, this approach may lead to new therapeutic strategies against important zoonotic pathogens with decreased chance of antiviral resistance.

        Speaker: David Scheibner (Friedrich-Loeffler-Institut)
      • 44
        Effectiveness of rabies vaccination of dogs using oral baits - results from the largest field trial

        Rabies is a prime example of One Health interventions. Oral rabies vaccination (ORV) of dogs has become a novel strategy in the global effort to control and eventually eliminate dog rabies, the source of rabies cases in humans. ORV involves the delivery of vaccines through baits specifically designed to be ingested by dogs and targets particularly free-roaming and stray dogs, which are often difficult to capture for injection. We wanted to assess the in-field practicability, effectiveness and vaccination coverage in the Zambezi region, a remote part in Namibia’s Northeast. We used standardized egg-flavoured baits filled with a third-generation vaccine with the highest safety profile. Veterinary staff of the region was trained in bait handling, vaccination and data capturing. Baits were then offered by teams to local free-roaming dogs and data was captured using a mobile-phone app. During the campaign in June 2024, 7,625 dogs were vaccinated within four and a half working days with ten teams with two people each. Teams managed to vaccinate, on average, 22.44 dogs/hour with a range of 17.45 and 27.8 dogs/hour, despite remote rural areas and using a door-to-door vaccination approach. Interestingly, five teams had working hours in which they vaccinated more than 50 dogs. These favourable results in terms of acceptance, field applicability and effectiveness further support the great potential of ORV in dog rabies control programmes to reach the goal of Zero by Thirty.

        Speaker: Dr Thomas Müller
      • 45
        Analyzing the Zoonosis Risk Along the Bushmeat Value-Chain in the Democratic Republic of the Congo

        Wild animals hunted and sold as bushmeat are a vital protein source in some African regions but also pose a risk as potential reservoirs of pathogens transmissible to humans. Previous research on bushmeat has primarily focused on the zoonotic origin of HIV and related retroviruses such as Simian T-lymphotropic virus (STLV). However, for acute disease-causing pathogens like Ebola virus and Mpox, the connections have mostly been epidemiological.
        To enhance our understanding of zoonotic risks associated with bushmeat in the Democratic Republic of the Congo, we studied the trade chain from Salonga National Park to rural and urban markets. We collected 716 tissue and swab samples from various species including primates, rodents, ungulates, pangolins, and civets. These samples were analyzed using PCR systems targeting coronaviruses, filoviruses, retroviruses, orthopoxviruses, and anthrax-causing Bacillus cereus biovar anthracis (BCBVA). PCR-positive samples underwent further analysis through target enrichment, whole genome sequencing, and phylogenetic placement.
        Our findings confirmed the presence of known pathogens such as STLV and, for the first time, identified agents capable of causing acute disease, including orthopoxviruses and BCBVA. The next phase of our research will correlate these findings with local hunting practices, meat processing, transportation, and consumption habits. Engaging with local communities to discuss these results will be crucial for insights into risk perception, historical outbreak records, and countermeasures previously employed. This community-engaged approach will enhance in-country pandemic preparedness and underscore the need for integrating social sciences to bolster pandemic prevention at the source.

        Speaker: Jasmin Schlotterbeck (Ecology and Emergence of Zoonotic Diseases - Helmholtz Institute for One Health)
      • 46
        A strain typing approach to investigate the transmission barrier of TSE field isolates

        Transmissible spongiform encephalopathies (TSEs) encompass human and animal diseases with infectious, hereditary or sporadic aetiology, including Scrapie in small ruminants, Chronic Wasting Disease (CWD) in cervids and bovine spongiform encephalopathy (BSE) in cattle. The pathognomonic spongiform lesions and amyloid plaque accumulation in the central nervous system are caused by the host encoded prion protein (PrPC) converting into a pathologic isoform (PrPSc). Different phenotypical characteristics of PrPSc indicate the presence of prion strains. Besides a strain’s distinct PrPSc and lesion profile, it is important to define its ability to overcome the transmission barrier and adapt to a new host. BSE easily overcomes the transmission barrier of several species including humans. However, for Scrapie and CWD prions the infectious interspecies potential is not yet fully understood. Therefore, we aimed to investigate the transmission barrier of several Scrapie and CWD field cases, with BSE as reference. All isolates were intracerebrally inoculated into the well-established TgshpIX mouse model, which overexpresses ovine PrPC. The resulting phenotype was characterized by using biochemical, histopathological and immunohistochemical methods. This study will help to further characterize the infectious nature of different TSEs and evaluates the potential threat of cross-species transmission between wildlife, livestock and humans due to environmental contamination.

        Speaker: Sonja Ernst (Friedrich-Loeffler-Insitut)
      • 47
        Genomic context of SARS-CoV-2 in farmed mink outbreaks in Spain during pandemic: unveiling host adaptation mechanisms

        After the onset of the SARS-CoV-2 pandemic, mink susceptibility to the virus has been well reported. During the 2020-2021 season, 117 respiratory samples from 18 farm outbreaks were sequenced and analyzed. To obtain the complete genome, we used an in-house approach combining SISPA amplification with the ARTIC protocol. Specific primers targeting the spike protein were used to complete the sequences of the main antigen. The binding kinetics between mink spike protein and various ACE-2 receptors were measured using Biacore technology. Antigenic characterization involved pseudovirus neutralization and competition assays.
        We obtained 68 complete SARS-CoV-2 sequences and 6 complete spike sequences from 5 different lineages. Sequencing revealed numerous non-lineage-defining mutations. Outbreaks were associated with groups of mutations rather than specific lineages especially in the ORF1a polyprotein and spike protein.
        Genomic analysis of spike proteins from different outbreaks revealed mutations in key antigenic sites, such as Y453F and Y453F plus M869T in lineage B.1.1777, and N501T with F486V, N501T, D796H in lineage B.1. These changes, previously unassociated with these lineages, were present in various variants of concern. Functional analysis showed these mutations increased affinity for the mink ACE-2 receptor, demonstrating host adaptation. However, these spikes did not significantly impact vaccinated individuals' serum. The spread of SARS-CoV-2 in farmed mink is a potential environment where the rapid transmission can facilitate the emergence of new variants.

        Speaker: Dr María de la Montaña Iglesias Caballero (National Centre of Microbiology)
      • 48
        Lymphocytic choriomeningitis virus: A neglected zoonotic pathogen

        Lymphocytic choriomeningitis virus (LCMV) is a globally distributed zoonotic arenavirus. In humans, LCMV infection can cause meningitis and encephalitis, while prenatal infections can lead to hydrocephaly, birth defects or miscarriage. In New World primates (NWP), LCMV causes callitrichid hepatitis. Despite the global distribution of its reservoir host, the house mouse (Mus musculus), and the severity of disease caused by infection, LCMV is under-reported.
        Here, we investigate the outbreak and ongoing localized presence of LCMV in captive NWP and wild house mice in a zoo in Germany. LCMV was not detected in any mice collected in the zoo in 2009, but in about half of all 280 tested mice sampled between 2021-2023. For the first time, two lineages of LCMV (lineages I and II) were found in the same population of wild mice, but only lineage II in diseased NWP.
        Additionally, more than 2,100 small mammals from seven European countries (2005-2023) were screened for LCMV RNA. A single LCMV lineage I positive house mouse from the Netherlands and a novel, divergent lineage of LCMV was detected in two wood mice (Apodemus sylvaticus) from Germany.
        The re-emergence and localized persistence of LCMV in wild rodents in Germany may have significant implications for public health. Moreover, the newly discovered virus lineage indicates a greater diversity of susceptible hosts, which may influence the distribution and transmission of LCMV.

        Speaker: Calvin Mehl (Friedrich-Loeffler Institute)
      • 49
        Biofilm formation and persistence of various anthrax causing bacteria on leaves

        Bacillus cereus biovar anthracis (Bcbva) is a novel anthrax causing agent infecting animals in African rainforest areas. Human infections were not yet confirmed, but exposure is likely due to hunting and consumption of ’bush meat’ and was also evidenced by seroprevalence studies in humans living in affected regions.

        It is still largely unknown how animals get infected with Bcbva. One route of infection might be similar to that of Bacillus anthracis via spores released into the soil by dead animals. However, also monkey species living only in trees get regularly infected by Bcbva. Thus, we assume that carrion flies are able to transmit the disease and indeed Bcbva has been confirmed in carrion flies. These flies could potentially spread Bcbva and its spores from infected carcasses to leaves and fruits which in turn get consumed by various monkey species.

        In our study, we use in vivo models to show that Bcbva is capable of persisting and even replicating on leaves and fruits as biofilm.
        To gain insight into biofilm formation, we use confocal laser scanning as well as electron microscopy. Multiplication and spore formation of Bcbva are confirmed by quantification methods such as colony forming unit calculation and quantitative PCR. At last, we aim to characterize and quantify biofilm matrix composition by labeling with specific fluorescent markers and photometric measurement.

        Speaker: Leonard Borst
    • Session 9: One Health & Public Health II MOA 4 + 5

      MOA 4 + 5

      • 50
        Micropollutants and trace substances as quality control parameters for wastewater-based epidemiology

        Since the coronavirus pandemic, the focus has shifted to wastewater-based epidemiology (WBE) for monitoring pathogens in the population. In line with the One Health approach, the German Infection Protection Act (2023) and the revision of the Urban Wastewater Directive (2024), a research field of health data generation by wastewater and environmental samples has been emerging.
        In Germany, the project AMELAG, Wastewater Monitoring for Epidemiological Situation Assessment, is currently implementing the establishment of the WBE at 170 wastewater treatment plants. One significant challenge in the interpretation of quantitative data sets is posed by site-dependent factors and fluctuations in flow rate and wastewater composition. Consequently, normalisation must be carried out with further parameters, such as the daily flow rate, ammonia content or control viruses (surrogate viruses). Furthermore, municipal wastewater contains a plethora of chemical substances, such as pharmaceuticals, food ingredients and human metabolites.
        The presentation will provide an overview of the suitability of various micropollutants and trace substances as normalisation parameters in various wastewater treatment plants. The results indicate that carbamazepine and caffeine yield more favourable outcomes than diclofenac and isolithocholic acid in the process of normalisation. The analysis of different normalisation models shows that such human markers are suitable for normalising WBE data.

        Speaker: Thomas Exner (Department III 2.5 Wastewater analysis, monitoring methods, Umweltbundesamt (UBA))
      • 51
        Isolate- and metagenome-based detection of antimicrobial resistance in Klebsiella pneumoniae from wastewater

        Klebsiella pneumoniae (KPN) is a significant healthcare-associated pathogen responsible for severe infections. The rise of antimicrobial resistance (AMR) in these bacteria poses a critical challenge due to their rapid acquisition of multidrug resistance genes (MDR). Wastewater (WW) surveillance emerges as an effective method for monitoring pathogens, offering community-level insights, acting as an early warning system, highlighting environmental reservoirs, and being a cost-effective approach for continuous monitoring of trends. Within the AMELAG project our research involved collecting and culturing WW samples to isolate KPN strains at the Umweltbundesamt, Germany. We performed genome reconstruction and resistance screening with short and long read sequencing. This approach identified various resistance patterns, like MDR genes and their host organism. Nanopore sequencing allowed the assembly of complete chromosomes and plasmids, accurately pinpointing resistance genes. Metagenomic sequencing of the same WW samples enabled detection of the broader microbiome and its resistome, allowing comparison of MDR genes in a metagenomic context to specific isolates. The integration of isolate-based genomic detection and metagenomic sequencing provides valuable insights into the dynamics of AMR in WW. Thus offering valuable insights into community-level AMR trends and environmental reservoirs, highlighting its potential as a critical complementary component of AMR monitoring frameworks.

        Speaker: Dr Christian Blumenscheit (Robert Koch Institut (RKI), MF1 Genome Competence Center, Berlin)
      • 52
        Identification and full genome sequencing of sandfly-borne phleboviruses using a newly established capture-based next generation sequencing approach

        Sandflies are vectors of phleboviruses that cause febrile illness and neuro-invasive infections in humans. We recently discovered five novel viruses in sandflies in Kenya indicating that sandfly-borne phleboviruses circulate beyond the Mediterranean. Here, we aimed to develop a capture-based next-generation sequencing (NGS) approach for phlebovirus genome sequencing.
        For specific capture and enrichment of sequences in NGS libraries, a bait set was designed based on sandfly-borne phlebovirus genomes. The bait set was established using infectious cell culture supernatants from eleven phleboviruses and tested using phlebovirus-positive sandfly specimens collected in Kenya.
        Complete genomes were retrieved for all eleven reference viruses with mean genome coverages ranging from 378 to 440,018 reads. Further, coding-complete sequences of four viruses with mean genome coverages ranging from 334 to 9764 reads were retrieved from the sandfly specimens whereas conventional NGS was not successful. Sequence analyses showed the presence of Embossos, Bogoria and Kiborgoch viruses, as well as the detection of three previously unknown viruses, the latter showing 25-31% nucleotide distances to viruses of the Salehabad serocomplex, as well as to Embossos and Bogoria viruses.
        We successfully established a capture-based NGS approach for known and previously unknown sandfly-borne phleboviruses. Our findings further expand the genetic diversity of sandfly-borne phleboviruses circulating in Kenya.

        Speaker: Dr Inga Slothouwer (Institute of Virologie, Charité Berlin, Germany)
      • 53
        Today’s challenges demand modern solutions: innovative pipelines for reliable antimicrobial resistance surveillance within a One Health framework

        Antimicrobial resistance (AMR) is a global threat and a major focus of the World Health Organization. Multidrug-resistant Enterobacterales such as Escherichia coli and Klebsiella pneumoniae are among the most critical bacterial pathogens, making AMR surveillance crucial not only for human and livestock sectors but also for the environment and wild animals.
        Our group applies a global One Health approach, addressing all these sectors to detect the dynamics of pathogen transmission and contribute to outbreak identification. To achieve this, we combine state-of-the-art genomic and phenomic techniques, employing them on a variety of different samples including water, soil, migratory birds, and feces from humans and wildlife in sub-Saharan Afrika and northeast Germany.
        To investigate these samples, establishing a standardized and innovative pipeline is essential. The InnoDia project is developing reliable sampling strategies alongside methods for preprocessing, bacterial cultivation, and DNA extraction. This includes a biobanking approach for long-term storage. An automated metagenomic pipeline will be established as well. The innovative methods being developed will combine specificity and sensitivity even during the upscaling process to a high-throughput system.
        In summary, the developed innovative pipelines will be a valuable tool for global AMR surveillance, providing a standardized sample process for detecting multidrug-resistant bacteria within a global One Health framework.

        Speaker: Alexandra Dürwald (Epidemiology and Ecology of Antimicrobial Resistance, Helmholtz Institute for One Health, Greifswald)
      • 54
        A One-Health Rapid Diagnostic Pipeline for Emergency Transboundary Infectious Disease

        It has become apparent that the ease of travelling, global warming, and changes in the environment accelerate the spread of infectious diseases of zoonotic origin. International collaboration is essential for controlling disease epidemics, and as the COVID pandemic has shown, rapid diagnostics are one of the main pillars. The Pipeline for Rapid Diagnostics of Emergency Transboundary Infectious Diseases (PREPARE-TID) project targets infectious agents of three types: an ongoing neglected epidemic, an ongoing emerging epidemic, and an ongoing elimination drive. The aim is to develop the necessary in vitro diagnostic tools for preparedness needed in case of the emergence of infectious agent X. PREPARE-TID is a multidisciplinary research consortium drawn from 16 European and 4 international research organizations and enterprises. Biomedical researchers are working alongside epidemiologists, clinicians, veterinarians, and software engineers to implement a pipeline from a mobile suitcase sequencing laboratory with a simple bioinformatics to a fieldable rapid nucleic acid extraction procedure, a mobile smartphone linked molecular point-of-care and a multiplex PCR one-health surveillance platform, and a one-health digital platform. These are providing innovative diagnostic solutions for cross-border health threats at international level. PREPARE-TID increases the accessibility to novel diagnostics for detection of pathogens with pandemic potential. The consortium delivers point-of-care and mobile diagnostics, which can be easily deployed within advanced surveillance programs based on a comprehensive digital “One Health” platform, and mass produced in case of an epidemic or pandemic.

        Speaker: Paula Schweizer (Institute of Animal Hygiene and Veterinary Public Health, Leipzig University, Leipzig, Germany)
      • 55
        One Health in heat-health action planning: status quo in North-Rhine Westphalia and perspectives

        As climate change exacerbates the frequency and intensity of extreme heat periods, it is critical that adaptation efforts take integrated approaches that consider the interconnected health impacts on humans, animals and the environment. The One Health approach provides a comprehensive framework for this, as research at the interface of One Health and climate adaptation is increasingly recognised as essential for addressing complex health challenges related to climate change (e.g. UNEP & ILRI 2020; Zingsstag et al. 2018 ). However, its consideration in municipal heat-health action planning in the state of North-Rhine Westphalia remains underexplored.

        This research aims to explore the extent to which the One Health approach can be considered in heat-health action planning as well as its current role in municipal heat-health action plans in the state of North-Rhine Westphalia. By attempting to answer the following two questions, the research contributes to a deeper understanding of the need for a One Health approach in climate adaptation strategies that address extreme heat.

        • Which aspects of the One Health approach could be included in heat-health action planning processes?
        • How much One Health is included in existing municipal heat-health action plans?

        This research will fill a gap in the literature field of One Health by exploring possibilities and perspectives of the interconnectedness of One Health and heat-health action planning.

        Speaker: Dr Thomas Claßen (NRW Centre for Health (LZG.NRW))
    • 11:30
      Lunch & Poster Viewing Atrium

      Atrium

    • Keynote VI: Katharina Schaufler MOA 4 + 5

      MOA 4 + 5

    • Poster Prize, Fare well MOA 4 + 5

      MOA 4 + 5