Skip to main content

University of Exeter Medical School

Professor Soojin Ryu

Professor Soojin Ryu

Mireille Gillings Professor of Neurobiology

 S.Ryu@exeter.ac.uk

 7580

 +44 (0) 1392 727580

 Living Systems Institute T03.11

 

Living Systems Institute, University of Exeter, Stocker Road, Exeter, EX4 4QD


Overview

As a recently appointed Mireille Gillings Professor of Neurobiology (awarded to women academics developing the next generation of female leaders in medicine, science, leadership and business) Soojin Ryu joined the University of Exeter College of Medicine and Health and the Living Systems Institute in January 2020.

She received her undergraduate degree in Biology from Harvard University (John Harvard Award for Excellence 1994) and her PhD from the University of California at Berkeley in the laboratory of Professor Robert Tjian. During her studies at Berkeley as a Howard Hughes Medical Institute Predoctoral Fellow, she identified a novel protein complex which plays a key role in gene regulation.

Soojin joined Professor Wolfgang Driever’s group as a Human Frontiers Long term Postdoctoral Fellow at the University of Freiburg. Here her work focused on molecular mechanisms for neuronal fate specification using zebrafish as a model organism.

In 2008, she was appointed as a Max Planck Independent Research Group Leader and joined the Max Planck Institute for Medical Research in Heidelberg where she stayed until 2016. During this period, Soojin’s lab contributed significantly to establishing zebrafish as a new model organism for stress research.

Until 2019, Soojin was a part of the medical faculty of the University of Mainz in Germany as a Professor where her work firmly established zebrafish models of human stress-induced disorders.

Role of Stress Hormones on Animal Behaviour

Soojin is deeply interested in how stress hormones shape animal behavior including the ways in which  prolonged stress exposure leads to behavioral dysfunctions. Her long-term goal is to use this knowledge to identify novel therapeutic targets for stress-induced disorders in humans.

Qualifications

  • B.A. in Biology, Harvard University
  • Ph.D. in Molecular and Cell Biology, University of California Berkeley

Back to top


Research

Research interests

Stress on Brain and Behaviour

Prolonged or severe exposure to stress hormones can lead to behavioural dysfunctions.  I lead a group developing detailed and sophisticated models relating to the effects of stress on brain and behavior using zebrafish. We are particularly interested in understanding how prolonged or severe exposure to stress hormones lead to behavioral dysfunctions. Our current research centers around the key stress hormones produced by our body’s main stress response system, the Hypothalamo-Pituitary-Adrenal (HPA) axis. The HPA axis coordinates diverse aspects of stress response in all vertebrates and the hormones produced by the HPA axis are remarkably conserved throughout vertebrate kingdom. Strikingly, while critical for the survival and coping capacity of the animal in fluctuating environment, the prolonged exposure to the HPA axis hormones is harmful and the dysregulation of the HPA axis is strongly implicated in a number of stress-induced disorders in humans including depression and anxiety disorders.

Pioneering Work with Zebrafish

Although it has long been recognised that humans share 70% of their genes with zebrafish the latter's application to particular areas of medical research is a developing science.  Our lab has successfully applied advanced research techniques to establish zebrafish as a model organism with which to study the stress reponse and HPA axis function.  This method enables us to unravel complex underlying mechanisms by which the HPA axis hormones exert their functions both in normal and compromised individuals, our lab has invested significant effort in establishing zebrafish as a model organism to study HPA axis function. As a part of this effort we have developed a method that allows modulation of the HPA axis hormone levels at will in a living animal.

Using such manipulations combined with robust behavioral analysis and comprehensive molecular and cellular profiling, our primary goal is to identify hitherto unknown mechanisms that mediate stress hormone’s striking control of animal behavior. Given the high degree of conservation of the stress system throughout vertebrates, the mechanistic insights we gain in zebrafish will be invaluable for understanding human stress biology as well.

Potential Therapeutic Applications

Recognising the potential for more sophisticated and specific medical interventions in the areas of depression and other stress-induced disorders our work is designed to harness the advantages for high-throughput analysis opportunities that zebrafish offers.  To this end our second major goal is to screen for novel molecules that could ameliorate detrimental effects of stress as potential new therapeutic avenues for depression and other stress-induced disorders.

Research projects

  • How does stress rapidly alter an animal’s behavior?

Using optogenetic manipulation, we have recently demonstrated the crucial modulatory actions of pituitary corticotroph cell activity on locomotion, avoidance and stimulus responsiveness directly after the onset of stress. We combine state-of-the-art optogenetic and genetic manipulation with sophisticated behavioral analysis to unravel mechanisms by which HPA axis rapidly alters behaviour.

  • How does stress exposure during development alter neural circuit structure and function?

We want to better understand how early life stress exposure shapes an animal’s future stress response and behavior. While some exposure to stress during development can improve future performance, prolonged or severe stress exposure in early life can lead to an individual with compromised coping capacity. We combine defined early life stress exposure with in-depth anatomical, physiological, molecular and cellular analyses to study new cellular and molecular mechanisms leading to adult fitness.

  • Which molecules can regulate an animal’s stress response?

We believe that our zebrafish model provides a unique opportunity to screen for molecules that can restore normal HPA function following prolonged or severe stress exposure. Such molecules represent promising targets for restoring proper stress response in humans as well. We are actively pursuing identifying modulators of the HPA axis function in zebrafish in order to develop new molecular treatments for stress-induced disorders in humans.


Grants/Funding:

  • 2020-2025  Mireille Gillings Professorial Fellowship   (£1 million)
  • 2017-2020  Boehringer Ingelheim Foundation "Neurobehavioral mechanisms of stress inoculation – a translational approach" (€138.300)
  • 2017-2020  Boehringer Ingelheim Foundation Grant "Extinction learning as a neurobiological resilience mechanism in zebrafish" (€162.300)
  • 2016-2020  German Research Foundation Grant CRC1193 “Neurobiology of resilience to stress-related mental dysfunction” (€361.600 - shared by two PIs)
  • 2016-2019 German Research Foundation Priority Program “Next Generation Optogenetics” SPP 1926 (€197.600)
  • 2015-2020  German Ministry for Education and Research Grant (BMBF), “The regulation of developing neural circuits by stress” (€2.459.815)
  • 2010-2016  German Research Foundation Grant- "Protein-based photoswitches" (€436.700)
  • 2013-2016  University of Heidelberg, Excellence Initiative program (€80.700)
  • 2010-2013  EU FP7 PEOPLE  "Neuroendocrine influences on aging" (€150.000)
  • 2010-2013  University of Heidelberg Frontiers Innovation Fund (€78.000)
  • 2009-2012  Behrens-Weise Foundation (€150.800)

Back to top


Publications

Journal articles

Eachus H, Choi M-K, Tochwin A, Kaspareit J, Ho M, Ryu S (2024). Elevated glucocorticoid alters the developmental dynamics of hypothalamic neurogenesis in zebrafish. Commun Biol, 7(1). Abstract.  Author URL.
Eachus H, Ryu S (2024). Glucocorticoid effects on the brain: from adaptive developmental plasticity to allostatic overload. Journal of Experimental Biology, 227(Suppl_1). Abstract.
Cook A, Beckmann H, Azap R, Ryu S (2023). Acute Stress Modulates Social Approach and Social Maintenance in Adult Zebrafish. eNeuro, 10(9), eneuro.0491-eneu22.2023.
Herget U, Ryu S, De Marco RJ (2023). Altered glucocorticoid reactivity and behavioral phenotype in rx3-/- larval zebrafish. Frontiers in Endocrinology, 14
Ayash S, Lingner T, Ramisch A, Ryu S, Kalisch R, Schmitt U, Müller MB (2023). Fear circuit–based neurobehavioral signatures mirror resilience to chronic social stress in mouse. Proceedings of the National Academy of Sciences, 120(17). Abstract.
Jacobs EAK, Ryu S (2023). Larval zebrafish as a model for studying individual variability in translational neuroscience research. Frontiers in Behavioral Neuroscience, 17
Eugenin von Bernhardi J, Biechl D, Miek L, Herget U, Ryu S, Wullimann MF (2022). A versatile transcription factor: Multiple roles of orthopedia a (otpa) beyond its restricted localization in dopaminergic systems of developing and adult zebrafish (Danio rerio) brains. J Comp Neurol, 530(14), 2537-2561. Abstract.  Author URL.
Albert L, Nagpal J, Steinchen W, Zhang L, Werel L, Djokovic N, Ruzic D, Hoffarth M, Xu J, Kaspareit J, et al (2022). Bistable Photoswitch Allows in Vivo Control of Hematopoiesis. ACS CENTRAL SCIENCE, 8(1), 57-66.  Author URL.
Gemmer A, Mirkes K, Anneser L, Eilers T, Kibat C, Mathuru A, Ryu S, Schuman E (2022). Oxytocin receptors influence the development and maintenance of social behavior in zebrafish (Danio rerio). Sci Rep, 12(1). Abstract.  Author URL.
Anneser L, Gemmer A, Eilers T, Alcantara IC, Loos A-Y, Ryu S, Schuman EM (2022). The neuropeptide Pth2 modulates social behavior and anxiety in zebrafish. iScience, 25(3), 103868-103868.
Eachus H, Ryu S, Placzek M, Wood J (2022). Zebrafish as a model to investigate the CRH axis and interactions with DISC1. Current Opinion in Endocrine and Metabolic Research, 26
Gemmer A, Mirkes K, Anneser L, Eilers T, Kibat C, Mathuru A, Ryu S, Schuman E (2021). Oxytocin receptors influence the development and maintenance of social behavior in zebrafish (<i>Danio rerio</i>). Abstract.
Ayash S, Lingner T, Ryu S, Kalisch R, Schmitt U, Müller M (2021). Reconceptualising Resilience Within a Translational Framework is Supported by Unique and Brain-Region Specific Transcriptional Signatures in Mice. Biological Psychiatry, 89(9).
Eachus H, Choi M-K, Ryu S (2021). The Effects of Early Life Stress on the Brain and Behaviour: Insights from Zebrafish Models. Front Cell Dev Biol, 9 Abstract.  Author URL.
Anneser L, Gemmer A, Eilers T, Alcantara IC, Loos A-Y, Ryu S, Schuman EM (2021). The neuropeptide Pth2 modulates social behavior and anxiety in zebrafish. Abstract.
Ayash S, Lingner T, Ryu S, Kalisch R, Schmitt U, Müller MB (2020). Reconceptualising resilience within a translational framework is supported by unique and brain-region specific transcriptional signatures in mice. Abstract.
Anneser L, Alcantara IC, Gemmer A, Mirkes K, Ryu S, Schuman EM (2020). The neuropeptide Pth2 dynamically senses others via mechanosensation. Nature, 588(7839), 653-657.
Castillo-Ramírez LA, Ryu S, De Marco RJ (2019). Active behaviour during early development shapes glucocorticoid reactivity. Sci Rep, 9(1). Abstract.  Author URL.
Nagpal J, Herget U, Choi MK, Ryu S (2019). Anatomy, development, and plasticity of the neurosecretory hypothalamus in zebrafish. Cell Tissue Res, 375(1), 5-22. Abstract.  Author URL.
Langebeck-Jensen K, Shahar OD, Schuman EM, Langer JD, Ryu S (2019). Larval Zebrafish Proteome Regulation in Response to an Environmental Challenge. Proteomics, 19(14). Abstract.  Author URL.
Herzog DP, Beckmann H, Lieb K, Ryu S, Müller MB (2018). Understanding and Predicting Antidepressant Response: Using Animal Models to Move Toward Precision Psychiatry. Front Psychiatry, 9 Abstract.  Author URL.
Beretta CA, Dross N, Guglielmi L, Bankhead P, Soulika M, Gutierrez-Triana JA, Paolini A, Poggi L, Falk J, Ryu S, et al (2017). Early Commissural Diencephalic Neurons Control Habenular Axon Extension and Targeting. Curr Biol, 27(2), 270-278. Abstract.  Author URL.
Biechl D, Tietje K, Ryu S, Grothe B, Gerlach G, Wullimann MF (2017). Identification of accessory olfactory system and medial amygdala in the zebrafish. Sci Rep, 7 Abstract.  Author URL.
Ryu S, De Marco RJ (2017). Performance on innate behaviour during early development as a function of stress level. Sci Rep, 7(1). Abstract.  Author URL.
Herget U, Gutierrez-Triana JA, Salazar Thula O, Knerr B, Ryu S (2017). Single-Cell Reconstruction of Oxytocinergic Neurons Reveals Separate Hypophysiotropic and Encephalotropic Subtypes in Larval Zebrafish. eNeuro, 4(1). Abstract.  Author URL.
De Marco RJ, Thiemann T, Groneberg AH, Herget U, Ryu S (2016). Optogenetically enhanced pituitary corticotroph cell activity post-stress onset causes rapid organizing effects on behaviour. Nat Commun, 7 Abstract.  Author URL.
Vom Berg-Maurer CM, Trivedi CA, Bollmann JH, De Marco RJ, Ryu S (2016). The Severity of Acute Stress is Represented by Increased Synchronous Activity and Recruitment of Hypothalamic CRH Neurons. J Neurosci, 36(11), 3350-3362. Abstract.  Author URL.
Gutierrez-Triana JA, Mateo JL, Ibberson D, Ryu S, Wittbrodt J (2016). iDamIDseq and iDEAR: an improved method and computational pipeline to profile chromatin-binding proteins. Development, 143(22), 4272-4278. Abstract.  Author URL.
Herget U, Ryu S (2015). Coexpression analysis of nine neuropeptides in the neurosecretory preoptic area of larval zebrafish. Front Neuroanat, 9 Abstract.  Author URL.
Gutierrez-Triana JA, Herget U, Castillo-Ramirez LA, Lutz M, Yeh C-M, De Marco RJ, Ryu S (2015). Manipulation of Interrenal Cell Function in Developing Zebrafish Using Genetically Targeted Ablation and an Optogenetic Tool. Endocrinology, 156(9), 3394-3401. Abstract.  Author URL.
Groneberg AH, Herget U, Ryu S, De Marco RJ (2015). Positive taxis and sustained responsiveness to water motions in larval zebrafish. Front Neural Circuits, 9 Abstract.  Author URL.
Gutierrez-Triana J, Herget U, Lichtner P, Castillo-Ramírez LA, Ryu S (2014). A vertebrate-conserved cis -regulatory module for targeted expression in the main hypothalamic regulatory region for the stress response. BMC Developmental Biology, 14(1).
Gutierrez-Triana JA, Herget U, Lichtner P, Castillo-Ramírez LA, Ryu S (2014). A vertebrate-conserved cis-regulatory module for targeted expression in the main hypothalamic regulatory region for the stress response. BMC Dev Biol, 14 Abstract.  Author URL.
Preuss SJ, Trivedi CA, vom Berg-Maurer CM, Ryu S, Bollmann JH (2014). Classification of object size in retinotectal microcircuits. Curr Biol, 24(20), 2376-2385. Abstract.  Author URL.
Gasser C, Taiber S, Yeh C-M, Wittig CH, Hegemann P, Ryu S, Wunder F, Möglich A (2014). Engineering of a red-light-activated human cAMP/cGMP-specific phosphodiesterase. Proc Natl Acad Sci U S A, 111(24), 8803-8808. Abstract.  Author URL.
Herget U, Wolf A, Wullimann MF, Ryu S (2014). Molecular neuroanatomy and chemoarchitecture of the neurosecretory preoptic-hypothalamic area in zebrafish larvae. J Comp Neurol, 522(7), 1542-1564. Abstract.  Author URL.
Herget U, Wolf A, Wullimann MF, Ryu S (2014). Molecular neuroanatomy and chemoarchitecture of the neurosecretory preoptic‐hypothalamic area in zebrafish larvae. The Journal of Comparative Neurology, 522(13), 3139-3139.
De Marco RJ, Groneberg AH, Yeh C-M, Treviño M, Ryu S (2014). The behavior of larval zebrafish reveals stressor-mediated anorexia during early vertebrate development. Front Behav Neurosci, 8 Abstract.  Author URL.
Yeh C-M, Glöck M, Ryu S (2013). An optimized whole-body cortisol quantification method for assessing stress levels in larval zebrafish. PLoS One, 8(11). Abstract.  Author URL.
De Marco RJ, Groneberg AH, Yeh C-M, Castillo Ramírez LA, Ryu S (2013). Optogenetic elevation of endogenous glucocorticoid level in larval zebrafish. Front Neural Circuits, 7 Abstract.  Author URL.
Wolf A, Ryu S (2013). Specification of posterior hypothalamic neurons requires coordinated activities of Fezf2, Otp, Sim1a and Foxb1.2. Development, 140(8), 1762-1773. Abstract.  Author URL.
Beretta CA, Dross N, Guiterrez-Triana JA, Ryu S, Carl M (2012). Habenula circuit development: past, present, and future. Front Neurosci, 6 Abstract.  Author URL.
Gabriel JP, Trivedi CA, Maurer CM, Ryu S, Bollmann JH (2012). Layer-specific targeting of direction-selective neurons in the zebrafish optic tectum. Neuron, 76(6), 1147-1160. Abstract.  Author URL.
Tay TL, Ronneberger O, Ryu S, Nitschke R, Driever W (2011). Comprehensive catecholaminergic projectome analysis reveals single-neuron integration of zebrafish ascending and descending dopaminergic systems. Nat Commun, 2 Abstract.  Author URL.
Kastenhuber E, Kratochwil CF, Ryu S, Schweitzer J, Driever W (2010). Genetic dissection of dopaminergic and noradrenergic contributions to catecholaminergic tracts in early larval zebrafish. J Comp Neurol, 518(4), 439-458. Abstract.  Author URL.
Löhr H, Ryu S, Driever W (2009). Zebrafish diencephalic A11-related dopaminergic neurons share a conserved transcriptional network with neuroendocrine cell lineages. Development, 136(6), 1007-1017. Abstract.  Author URL.
Ryu S, Mahler J, Acampora D, Holzschuh J, Erhardt S, Omodei D, Simeone A, Driever W (2008). Orthopedia Homeodomain Protein is Essential for Diencephalic Dopaminergic Neuron Development. Current Biology, 18(4).
Meng S, Ryu S, Zhao B, Zhang D-Q, Driever W, McMahon DG (2008). Targeting retinal dopaminergic neurons in tyrosine hydroxylase-driven green fluorescent protein transgenic zebrafish. Mol Vis, 14, 2475-2483. Abstract.  Author URL.
Filippi A, Dürr K, Ryu S, Willaredt M, Holzschuh J, Driever W (2007). Expression and function of nr4a2, lmx1b, and pitx3 in zebrafish dopaminergic and noradrenergic neuronal development. BMC Dev Biol, 7 Abstract.  Author URL.
Ryu S, Mahler J, Acampora D, Holzschuh J, Erhardt S, Omodei D, Simeone A, Driever W (2007). Orthopedia homeodomain protein is essential for diencephalic dopaminergic neuron development. Curr Biol, 17(10), 873-880. Abstract.  Author URL.
Brockschmidt A, Todt U, Ryu S, Hoischen A, Landwehr C, Birnbaum S, Frenck W, Radlwimmer B, Lichter P, Engels H, et al (2007). Severe mental retardation with breathing abnormalities (Pitt-Hopkins syndrome) is caused by haploinsufficiency of the neuronal bHLH transcription factor TCF4. Hum Mol Genet, 16(12), 1488-1494. Abstract.  Author URL.
Dürr K, Holzschuh J, Filippi A, Ettl A-K, Ryu S, Shepherd IT, Driever W (2006). Differential roles of transcriptional mediator complex subunits Crsp34/Med27, Crsp150/Med14 and Trap100/Med24 during zebrafish retinal development. Genetics, 174(2), 693-705. Abstract.  Author URL.
Ryu S, Holzschuh J, Mahler J, Driever W (2006). Genetic analysis of dopaminergic system development in zebrafish. J Neural Transm Suppl(70), 61-66. Abstract.  Author URL.
Ryu S, Driever W (2006). Minichromosome maintenance proteins as markers for proliferation zones during embryogenesis. Cell Cycle, 5(11), 1140-1142. Abstract.  Author URL.
Kastenhuber E, Ryu S, Driever W (2006). [P240]: Guidance of dopaminergic axonal projections in zebrafish. International Journal of Developmental Neuroscience, 24(8), 598-598.
Chapouton P, Adolf B, Leucht C, Tannhäuser B, Ryu S, Driever W, Bally-Cuif L (2006). her5 expression reveals a pool of neural stem cells in the adult zebrafish midbrain. Development, 133(21), 4293-4303. Abstract.  Author URL.
Ryu S, Holzschuh J, Erhardt S, Ettl A-K, Driever W (2005). Depletion of minichromosome maintenance protein 5 in the zebrafish retina causes cell-cycle defect and apoptosis. Proc Natl Acad Sci U S A, 102(51), 18467-18472. Abstract.  Author URL.
Holzschuh J, Ryu S, Aberger F, Driever W (2001). Dopamine transporter expression distinguishes dopaminergic neurons from other catecholaminergic neurons in the developing zebrafish embryo. Mech Dev, 101(1-2), 237-243. Abstract.  Author URL.
Ryu S, Tjian R (1999). Purification of transcription cofactor complex CRSP. Proc Natl Acad Sci U S A, 96(13), 7137-7142. Abstract.  Author URL.
Ryu S, Zhou S, Ladurner AG, Tjian R (1999). The transcriptional cofactor complex CRSP is required for activity of the enhancer-binding protein Sp1. Nature, 397(6718), 446-450. Abstract.  Author URL.
Näär AM, Ryu S, Tjian R (1998). Cofactor requirements for transcriptional activation by Sp1. Cold Spring Harb Symp Quant Biol, 63, 189-199.  Author URL.

Chapters

Ryu S, De Marco RJ (2017). Optogenetic Interpellation of Behavior Employing Unrestrained Zebrafish Larvae. In  (Ed) Optogenetics: a Roadmap, Springer Nature, 117-131.

Back to top


External Engagement and Impact

Awards

  • Mireille Gillings Professorial Fellowship, 2019
  • BMBF Independent Neuroscience Group Award, 2015
  • Behrens-Weise Foundation Award, 2009
  • Human Frontiers Science Program Postdoctoral Fellowship, 12/2000 - 12/2003
  • Alexander von Humboldt Foundation Postdoctoral Fellowship, 6/2000  - 11/2000
  • Howard Hughes Medical Institute Predoctoral Fellowship, 1995 - 2000
  • John Harvard Award for Academic Excellence, 1994
  • Fourth Place National Winner for Westinghouse Science Talent Search, 1989

Committee/panel activities

  • 2017- 2019            Institute Council member, German Resilience Center
  • 2017- 2019            External advisor, Korean Institute of Science and Technology  Europe, Saarbrucken, Germany
  • 2015- 2019            Steering committee member, German Research Foundation Priority Program “Next Generation Optogenetics” (SPP 1926)
  • 2015                      Evaluation referee, the International Max Planck Research School for Neural Circuits, Frankfurt
  • 2012-2015             Steering committee member, Interdisciplinary Center for Neuroscience, University of Heidelberg
  • 2012-2015             Assembly member, Excellence Cluster CellNetworks, University of Heidelberg
  • 2011                      Second phase preparation committee member, Excellence Cluster CellNetworks, University of Heidelberg

Invited lectures

  • Southwest Zebrafish Meeting, Bristol, UK, September 2019
  • International Congress of Neuroendocrinology, Toronto, Canada, Summer 2018
  • 4th Medaka Principal Investigator Meeting, Heidelberg, Germany, Spring 2018
  • The 5th European Zebrafish Principal Investigators Meeting, Trento, Italy, Spring 2018
  • Living Systems Institute, University of Exeter, Exeter, UK, Spring 2018
  • Bio Korea in Europe 3rd Symposium, London, UK, Fall 2017
  • The Gulbekian Institute, Oeiras, Portugal, Fall 2017
  • 4th Imaging Structure and Function in the Zebrafish Brain Conference, Martinsried, Germany, Winter 2016
  • Korean Institute of Science and Technology Europe, Saarbrücken, Germany, Spring 2016
  • EpiStressNet Conference, Sheffield UK, Winter 2016
  • Bio Korea in Europe Symposium, Saarbrücken, Germany, Spring 2016
  • German Research Foundation (DFG) Roundtable Discussion Photoreceptors, Frauenchiemsee, Germany, Fall 2015
  • Max Planck Society Neuroscience Retreat, Frankfurt, Germany, Spring 2015
  • European Neuroscience Conference for Doctoral Students, Lisbon, Portugal 2015
  • Europe-Korea Conference on Science and Technology, Strasbourg, France 2015
  • Rhein-Main Neuroscience Network Annual Retreat, Oberwesel, Germany, 2014
  • Dept. Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, USA, 2014
  • DFG Roundtable Meeting „Optogenetics“ Bad Homburg, Germany, Spring 2014
  • 3th Imaging Structure and Function in the Zebrafish Brain Conference, Paris, France, Winter 2014
  • Helmholtz Center, Munich, Germany, Winter 2014
  • Max Planck Institute for Psychiatry, Munich, Germany, Winter 2014
  • 9th FENS Symposium, Milan, Italy, Summer 2014
  • Channelrhodopsin Meeting, Würzburg, Germany, Fall 2014
  • Dept. Developmental Biology, University of Sheffield, UK 2013
  • DFG Roundtable Discussion Photoreceptors, Schloss Ringberg, Germany, Fall 2013
  • Janelia Farm Zebrafish Workshop, Ashburn, Virginia USA 2013
  • DFG Forschergruppe 1279 Optogenetics Conference, Würzburg, Germany,  2012
  • 2nd Imaging Structure and Function in the Zebrafish Brain Conference, London, UK, Winter 2012
  • DFG Forschergruppe 1279 Optogenetics Conference, Munich, Germany, 2011
  • Neuroendocrine Influence on Aging Symposium, Munich, Germany,  2011
  • Imaging Structure and Function in the Zebrafish Brain Conference, Lisbon, Portugal, Winter 2010
  • Max Planck Institute for Psychiatry annual Retreat, Schloss Ringberg, Germany, 2010
  • 8th World Congress on Neuorhypophysial Hormones, Kitakyushu, Japan, 2009
  • Heidelberg Forum, Heidelberg, 2009
  • Dept. of Biology, Seoul National University, Seoul, Korea 2009
  • Max Planck Institute for Psychiatry, Munich, Germany, 2008

Media Coverage

  • 2019 BBC spotlight
  • 2018 Spiegel Online

Back to top


Teaching

2017-18 - Organizer and Lecturer, “Stress Resilience: From Molecular Mechanism to Behavior” (for medical students) University Medical School, Johannes Gutenberg University Mainz

2017 -  Lecturer, The Gulbenkian Integrative Biology and Biomedicine PhD Program, Neurobiology-Brian and Behavior Module, Instituto Gulbenkian de Ciencia, Portugal

2017 - Lecturer, “Zebrafish in Neuroscience,” Institute of Molecular Biology International Summer School Block Lecture, Mainz

2014 -  Lecturer, Model Systems Course, IBioBA (Biomedicine Research Institute of Buenos Aires), Argentina

2013 - Undergraduate intensive practical course on “Analysis of the Zebrafish Nervous System Development,” University of Heidelberg

2013 - Undergraduate intensive seminar course on “How to Analyze Scientific Publications,” University of Heidelberg

2012/13 - Intensive practical course on “Imaging Nervous System Development” for Bachelors Students, University of Heidelberg

2012 - Lecture course on Developmental Neurobiology, Max Planck Institute for Medical Research

2010 - Graduate laboratory course on Gene Regulation Analysis for PhD students, Hartmut-Hoffmann-Berling International Graduate School, Heidelberg

2009 - Graduate lecture course on Developmental Neurobiology for PhD students, Max Planck Institute for Medical Research, Heidelberg

2008/9 - Graduate Developmental Neurobiology seminar course for Master students, Masters in Biotechnology Program, University of Heidelberg

Back to top


Supervision / Group

Research Fellows

Research Technicians

Back to top


Edit Profile