MECHANISMS AND EVOLUTION OF INTERGENERATIONAL CHANGE 24-26 September 2019 ABSTRACT BOOK ABSTRACT Name:

Mechanisms and Evolution of Intergenerational Change

Wellcome Genome Campus Conference Centre, Hinxton, Cambridge, UK 24-26 September 2019

Scientific Programme Committee:

Jennifer Brisson University of Rochester, USA

Paula Brunton University of Edinburgh, UK

Nick Burton , UK

Ben Dantzer University of Michigan, USA

Tweet about it: #IntergenChange19

@ACSCevents /ACSCevents /c/WellcomeGenomeCampusCoursesandConferences

1

Wellcome Genome Campus Scientific Conferences Team:

Rebecca Twells Treasa Creavin Nicole Schatlowski Head of Advanced Courses and Scientific Programme Scientific Programme Scientific Conferences Manager Officer

Jemma Beard Lucy Criddle Zoey Willard Conference and Events Conference and Events Conference and Events Organiser Organiser Organiser

Laura Wyatt Conference and Events Manager

2

Dear colleague,

I would like to offer you a warm welcome to the Wellcome Genome Campus Advanced Courses and Scientific Conferences: Mechanisms and Evolution of Intergenerational Change. I hope you will find the talks interesting and stimulating, and find opportunities for networking throughout the schedule.

The Wellcome Genome Campus Advanced Courses and Scientific Conferences programme is run on a not-for-profit basis, heavily subsidised by the .

We organise around 50 events a year on the latest biomedical science for research, diagnostics and therapeutic applications for human and animal health, with world-renowned scientists and clinicians involved as scientific programme committees, speakers and instructors.

We offer a range of conferences and laboratory-, IT- and discussion-based courses, which enable the dissemination of knowledge and discussion in an intimate setting. We also organise invitation-only retreats for high-level discussion on emerging science, technologies and strategic direction for select groups and policy makers. If you have any suggestions for events, please contact me at the email address below.

The Wellcome Genome Campus Scientific Conferences team are here to help this meeting run smoothly, and at least one member will be at the registration desk between sessions, so please do come and ask us if you have any queries. We also appreciate your feedback and look forward to your comments to continually improve the programme.

Best wishes,

Dr Rebecca Twells Head of Advanced Courses and Scientific Conferences [email protected]

3

General Information

Conference Badges Please wear your name badge at all times to promote networking and to assist staff in identifying you.

Scientific Session Protocol Photography, audio or video recording of the scientific sessions, including poster session is not permitted.

Social Media Policy To encourage the open communication of science, we would like to support the use of social media at this year’s conference. Please use the conference hashtag #IntergenChange19. You will be notified at the start of a talk if a speaker does not wish their talk to be open. For posters, please check with the presenter to obtain permission.

Internet Access Wifi access instructions:  Join the ‘ConferenceGuest’ network  Enter your name and email address to register  Click ‘continue’ – this will provide a few minutes of wifi access and send an email to the registered email address  Open the registration email, follow the link ‘click here’ and confirm the address is valid  Enjoy seven days’ free internet access!  Repeat these steps on up to 5 devices to link them to your registered email address

Presentations Please provide an electronic copy of your talk to a member of the AV team who will be based in the meeting room.

Poster Sessions Posters will be displayed throughout the conference. Please display your poster in the Conference Centre on arrival. There will be two poster sessions during the conference:  Odd number poster assignments will be presenting in poster session 1, which takes place on Tuesday, 24 September at 18:10 – 19:20.  Even number poster assignments will be presenting in poster session 2, which takes place on Wednesday, 25 September, at 17:45 – 19:00.

The page number of your abstract in the abstract book indicates your assigned poster board number. An index of poster numbers appears in the back of this book.

Conference Meals and Social Events Lunch and dinner will be served in the Hall, apart from on Tuesday, 24 September when it will be served in the Conference Centre. Please refer to the conference programme in this book as times will vary based on the daily scientific presentations. Please note there are no lunch or dinner facilities available outside of the conference times.

All conference meals and social events are for registered delegates. Please inform the conference organiser if you are unable to attend the either of the dinners.

The Hall Bar (cash bar) will be open from 19:00 – 23:00 each day.

Dietary Requirements If you have advised us of any dietary requirements, you will find a coloured dot on your badge. Please make yourself known to the catering team and they will assist you with your meal request.

4

If you have a gluten or nut allergy, we are unable to guarantee the non-presence of gluten or nuts in dishes, even if they are not used as a direct ingredient. This is due to gluten and nut ingredients being used in the kitchen.

For Wellcome Genome Campus Conference Centre Guests Check in If you are staying on site at the Wellcome Genome Campus Conference Centre, you may check into your bedroom from 14:00. The Conference Centre reception is open 24 hours.

Breakfast Your breakfast will be served in the Hall restaurant from 07:30 – 09:00.

Telephone If you are staying on-site and would like to use the telephone in your room, you will need to contact the Reception desk (Ext. 5000) to have your phone line activated – they will require your credit card details to do so.

Departures You must vacate your room by 10:00 on the day of your departure. Please ask at reception for assistance with luggage storage in the Conference Centre.

Taxis Please find a list of local taxi numbers on our website. The Conference Centre reception will also be happy to book a taxi on your behalf.

Return Ground Transport Complimentary return transport has been arranged for 13:30 on Thursday, 26 September to Cambridge station and city centre (Downing Street), and Stansted and Heathrow airports.

A sign-up sheet will be available at the conference registration desk from 15:40 on Tuesday, 24 September. Places are limited so you are advised to book early.

Please allow a 30-40-minute journey time to both Cambridge city and Stansted airport, and up to 3 hours to Heathrow airport.

Messages and Miscellaneous Lockers are located outside the Conference Centre toilets and are free of charge.

All messages will be available for collection from the registration desk in the Conference Centre.

A variety of toiletry and stationery items are available for purchase at the Conference Centre reception. Cards for our self-service laundry are also available.

Certificate of Attendance A certificate of attendance can be provided. Please request one from the conference organiser based at the registration desk.

Contact numbers Wellcome Genome Campus Conference Centre – 01223 495000 (or Ext. 5000) Wellcome Genome Campus Conference Organiser (Zoey) – 07747 024256

If you have any queries or comments, please do not hesitate to contact a member of staff who will be pleased to help you.

5

Conference Summary

Tuesday, 24 September 2019 12:00 – 13:00 Registration with lunch 13:00 – 13:10 Welcome and introduction 13:10 – 14:10 Keynote lecture Pat Monaghan, University of Glasgow, UK 14:10 – 15:40 Session 1: Evolutionary modelling of intergenerational effects 15:40 – 16:10 Afternoon tea 16:10 – 17:55 Session 2: Evolutionary models of intergenerational change in invertebrates 17:55 – 18:10 Lightning talks for poster session abstracts 18:10 – 19:20 Poster session and drinks reception 19:20 Buffet dinner

Wednesday, 25 September 2019 09:00 – 10:30 Session 3: Evolutionary models of intergenerational change in plants 10:30 – 11:00 Morning coffee 11:00 – 12:30 Session 4: Evolutionary paradigms of intergenerational change in vertebrates 12:30 – 14:00 Lunch 14:00 – 15:30 Session 5: Foetal programming models in mammals I 15:30 – 16:00 Afternoon tea 16:00 – 17:30 Session 6: Foetal programming models in mammals II 17:30 – 17:45 Lightning talks for poster session abstracts 17:45 – 19:00 Poster session and drinks reception 19:00 Served conference dinner

Thursday, 26 September 2019 09:00 – 10:30 Session 7: Foetal programming in humans and animal models 10:30 – 11:00 Morning coffee 11:00 – 12:00 Keynote lecture Kent Thornburg, Oregon Health and Science University, USA 12:00 – 12:10 Closing remarks 12:10 – 13:30 Lunch 13:30 Coaches depart to Cambridge City Centre and Train Station, and Heathrow Airport via Stansted Airport

6

Conference Sponsors

7

Mechanisms and Evolution of Intergenerational Change

Wellcome Genome Campus Conference Centre, Hinxton, Cambridge

24 – 26 September 2019

Lectures to be held in the Francis Crick Auditorium Lunch and dinner to be held in the Hall Restaurant Poster sessions to be held in the Conference Centre

Spoken presentations - If you are an invited speaker, or your abstract has been selected for a spoken presentation, please give an electronic version of your talk to the AV technician.

Poster presentations – If your abstract has been selected for a poster, please display this in the Conference Centre on arrival.

Conference programme

Tuesday, 24 September 2019

12:00 - 13:00 Registration with lunch

13:00 - 13:10 Welcome and introduction Programme Committee: Nick Burton, University of Cambridge, UK

13:10 - 14:10 Keynote lecture I Chair: Paula Brunton, University of Edinburgh, UK

Effect of parental state on offspring longevity; time scales, trade-offs and telomeres Pat Monaghan University of Glasgow, UK

14:10 - 15:40 Session 1: Evolutionary modelling of intergenerational effects Chair: Sinead English, University of Bristol, UK

14:10 Parental effects in evolutionary Tobias Uller Lund University, Sweden

14:40 How genomic conflict shapes the evolution of parental effects Bram Kuijper University of Exeter, UK

15:10 Extended heredity and the extended synthesis: an attempt to put recent developments of evolutionary theory into perspective Troy Day Queen's University, Canada

8

15:25 General pathways to specific outcomes: stress links maternal environment and offspring phenotypes Ahva Potticary University of Arizona, USA

15:40 - 16:10 Afternoon tea

16:10 - 17:55 Session 2: Evolutionary models of intergenerational change in invertebrates Chair: Jennifer Brisson, University of Rochester, USA

16:10 Maternally transmitted nematode mutualists modify the microbial communities of their beetle hosts Cris Ledón-Rettig Indiana University, USA

16:40 Insulin/IGF signaling and vitellogenin provisioning mediate intergenerational adaptation to nutrient stress Ryan Baugh Duke University, USA

17:10 Intergenerational inheritance of metabolic state Heidi Lempradl Van Andel Institute, USA

17:40 Energy-sensing pathways mediate intergenerational inheritance Andre Pires University of Warwick, UK

17:55 - 18:10 Lightning talks Chair: Paula Brunton, University of Edinburgh, UK

18:10 - 19:20 Poster session 1 (odd numbers) with drinks reception

19:20 Buffet dinner

Wednesday, 25 September 2019

09:00 - 10:30 Session 3: Evolutionary models of intergenerational change in plants Chair: Cris Ledón-Rettig, Indiana University, USA

09:00 Primed plants do not forget: mechanisms and ecological drivers of transgenerational acquired resistance in Arabidopsis thaliana Jurriaan Ton University of Sheffield, UK

09:30 Transgenerational environmental effects in variable environments: plant responses to environments within and across generations Kathleen Donohue Duke University, USA

9

10:00 EMBO Young Investigator Lecture: DNA methylation reprogramming in plant germlines Xiaoqi Feng John Innes Centre, UK

10:30 - 11.00 Morning coffee

11:00 - 12:30 Session 4: Evolutionary paradigms of intergenerational change in vertebrates Chair: Heidi Lempradl, Van Andel Institute, USA

11:00 Stress and survival in vertebrates: transgenerational effects of stress, environmental context, and why it matters Kirsty MacLeod Lund University, Sweden

11:30 Mums the word: trans-generational transmission of phenotype programmed by early-life stress in birds Karen Spencer University of St. Andrews, UK

12:00 The role of epigenetic modifications in intergenerational changes induced by captive rearing Sofia Consuegra Swansea University, UK

12:15 Maternal stress exposure leads to diversification of offspring behaviour in annual freshwater fish Agnieszka Magierecka University of Glasgow, UK

12:30 - 14:00 Lunch

14.00 - 15:30 Session 5: Foetal programming models in mammals I Chair: Paula Brunton, University of Edinburgh, UK

14:00 Parental delivery: somatic signals impacting neurodevelopment Tracy Bale University of Maryland, USA

14:30 Experimental evidence for fetal programming via epi-sensitive regions of the genome Rosalind John Cardiff University, UK

15:00 Improving neurobehavioural outcomes following exposure to stressful events in pregnancy Jonathan Hirst University of Newcastle, Australia

10

15:15 The prenatal maternal response to immune activation predicts offspring cognitive dysfunction in a rat model of schizophrenia Harry Potter University of Manchester, UK

15:30 - 16:00 Afternoon tea

16:00 - 17:30 Session 6: Foetal programming models in mammals II Chair: Nick Burton, University of Cambridge, UK

16:00 Programming by maternal over-nutrition: a plentiful problem Susan Ozanne University of Cambridge, UK

16:30 Epigenetic modulation of repeat elements Anne Ferguson-Smith University of Cambridge, UK

17:00 Effects of maternal obesity on oocyte methylome are reflected on blastocyst epigenome Antonio Galvao Babraham Institute, UK

17:15 Effects of prenatal stress on lactation and offspring development: linking evolutionary models with human cohort studies Sinead English University of Bristol, UK

17:30 - 17:45 Lightning talks Chair: Jennifer Brisson, University of Rochester, USA

17:45 - 19:00 Poster session 2 (even numbers) with drinks reception

19:00 Conference dinner

11

Thursday, 26 September 2019

09:00 - 10:30 Session 7: Foetal programming in humans and animal models Chair: Susan Ozanne, University of Cambridge, UK

09:00 Transgenerational transmission of susceptibility – evidence from the Dutch famine Tessa Roseboom Amsterdam UMC, The Netherlands

09:30 Heart disease link to fetal hypoxia: an intergenerational perspective Dino Giussani University of Cambridge, UK

10:00 Early life epigenetic predictors of cardiometabolic health in childhood Toby Mansell Murdoch Children's Research Institute, Australia

10:15 Intra-uterine growth restriction secondary to maternal obesity and hyperglycaemia is associated with altered glucose metabolism and expression of the DNA methyltransferases in the offspring brain Kahyee Hor University of Edinburgh, UK

10:30 - 11:00 Morning coffee

11:00 - 12:00 Keynote lecture II Chair: Jennifer Brisson, University of Rochester, USA

Enduring consequences of fetal stress Kent Thornburg Oregon Health and Science University, USA

12.00 - 12:10 Closing remarks Programme Committee: Nick Burton, University of Cambridge, UK

12.10 - 13:30 Lunch

13:30 Coaches depart to Cambridge city centre and train station, and Heathrow Airport via Stansted Airport

12

These abstracts should not be cited in bibliographies. Materials contained herein should be treated as personal communication and should be cited as such only with consent of the author.

13

Notes

14

Spoken Presentations

Effect of parental state on offspring longevity: time scales, trade-offs and telomeres

Pat Monaghan

Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow, UK

It is now well recognised that the early environment is not simply permissive of development, but can also shape the phenotype in ways that have long term consequences for performance and fitness. There are many routes, both direct and indirect, whereby such environmental effects on phenotypic development can come about and span generations. Of considerable importance in this context is parental state, which can influence offspring via effects operating at many stages, from parents’ germ cells through to the developmental environment parents provide for their offspring. Offspring growth, development and life history can then be altered in adaptive and non-adaptive ways. In this talk, I will focus particularly on effects on offspring of parental age and of early exposure to stress, and discuss the consequences over varying time scales. I will present illustrative data from unmanipulated natural populations, and from a range of taxa in which conditions have been experimentally manipulated in both the lab and the field. I will also examine some potential mechanisms that might mediate effects that can occur over relatively long time scales, including changes in stress sensitivity and telomere dynamics.

S1

Notes

S2

Parental effects in evolutionary biology

Tobias Uller

Professor of Evolutionary Biology, Department of Biology, Lund University, Sweden

There is a bewildering diversity of perspectives on intergenerational change. One reason for this is that effects of parents on offspring (‘parental effects’) are mediated by a diversity of mechanisms, and these mechanisms are themselves evolving. Another reason is that there are many possible evolutionary consequences of these mechanisms, which means researchers use different ways to represent them. My aim here is to structure and make sense of this diversity of perspectives. Firstly, I will place intergenerational change into context by considering it to primary be about cause-effect relations in development. Second, I will show why this helps to formulate questions about the evolutionary causes and consequences of parental effects. Third, I will illustrate how models can be used to answer some of those questions.

S3

Notes

S4

How genomic conflict shapes the evolution of parental effects

Bram Kuijper

Centre for Ecology and Conservation, University of Exeter, Penryn Campus, UK

A growing number of studies highlight the presence of adaptive parental effects, which allow parents to inform their young about the current environment. Few studies have considered, however, the prevalence of conflicts of evolutionary interest between mothers and fathers, as well as between parents and offspring, when making predictions about parental effects. I therefore model the coevolution of adaptive maternal and paternal effects as well as offspring sensitivity to signals of both parents, while accounting for conflicts of interest within the family. I find that offspring often evolve sensitivity to either paternal effects or maternal effects, but not to both effects simultaneously. Moreover, the degree of sex-biased dispersal and the type of mating system have strong impacts: when females are the philopatric sex or the mating system is polyandrous, adaptive paternal effects should predominate. By contrast, if males are the philopatric sex or the mating system is polygynous, adaptive maternal effects should prevail. Finally, I also predict that for some forms of transgenerational effects (e.g., DNA methylation and small RNAs, but not for social learning), one parent can evolve signals which undermine the other parent's signals. Overall, this model suggests that genetic conflicts have a major role to play in the evolution of adaptive paternal vs maternal effects.

S5

Notes

S6

Extended heredity and the extended synthesis: an attempt to put recent developments of evolutionary theory into perspective

Troy Day, Russell Bonduriansky

Queen's University, Canada

In recent years there it has become increasingly apparent that non-genetic forms of heredity exist in a wide variety of organisms. Furthermore, these "extended" forms of heredity can have interesting and important effects on how evolution by natural selection proceeds. Parallel to these findings has been the development of ideas from evo-devo, niche construction theory, and theory related to other "constructive" processes in evolution, with many of the researchers involved now calling for a revision or extension of the Modern Synthesis of Evolutionary Biology. In this talk I will give my own view of these issues by attempting to put all of the recent arguments within a common theoretical perspective. My goal with doing so is to clarify what both sides of the debate are saying so that a more product dialogue can be had.

S7

Notes

S8

General pathways to specific outcomes: stress links maternal environment and offspring phenotypes

Ahva Potticary, Renee Duckworth

University of Arizona, Department of Ecology and Evolutionary Biology, USA

Context-specific maternal effects - those that shift offspring phenotype to match conditions in a particular environment - are often contrasted with passive maternal effects that result from a lack of buffering of offspring from maternal stress. Yet, maternal experience of stress can itself act as a cue to induce adaptive offspring phenotypes. Here, we investigate the idea that maternal stress pathways, which are responsive to a wide variety of environmental contexts, are coopted to transmit a general message to offspring: whether natal environmental quality is high or low. We test this idea using a known adaptive maternal effect in western bluebirds, where mothers influence sex-biased lay order to shift offspring dispersal. Combining seventeen years of hormone sampling in diverse environmental contexts with experimental manipulation of competitive environment, we show that multiple factors influenced maternal stress, and this consistently resulted in the same maternal effect expression: females with high corticosterone levels altered sex-biased laying order in the direction that is known to produce more dispersive offspring. These results support the idea that multiple stressors interchangeably induce a maternal effect in this system and that offspring have evolved to respond to maternal stress in an adaptive way.

S9

Notes

S10

Maternally transmitted nematode mutualists modify the microbial communities of their beetle hosts

Cristina C. Ledón-Rettig, Armin P. Moczek, Erik J. Ragsdale

Indiana University, USA

Maternally transmitted factors have the potential to play an important role in evolutionary and ecological processes. While much attention has been paid to the microbial communities that are passed between parents and offspring, other, larger organisms can in principle also be inherited. Such “mesobiotic” partners can modify the developmental environments of their hosts and therefore both the phenotypes expressed by and the selective regimes experienced by offspring. Here, we present our investigation of the nematode Diplogastrellus monhysteroides, which was been found to be in close association with the adults and offspring of dung beetles, specifically the bull-headed dung beetle, Onthophagus taurus. We demonstrate that this nematode associate is both sexually transmitted between adults and vertically transmitted to offspring. Then, by rearing beetle offspring in artificial chambers where the presence of nematodes can be manipulated, we show that D. monhysteroides enhances the growth of beetle offspring. Using microbial profiling, we find that D. monhysteroides nematodes influence the bacterial and fungal communities in the developmental environments of their beetle hosts, specifically by increasing the abundances of taxa known to be involved in biomass degradation. Thus, nematodes possibly benefit developing beetles by “engineering” the microbial communities that beetle offspring experience, allowing larval beetles to better access their plant-based diet. We have now begun to further investigate mesobiotic community effects on the nematode-beetle mutualism. Specifically, we have found that two other maternally inherited nematode species – (i) a strict microbivore and (ii) a facultative predator of other nematodes – sometimes also inhabit the broodball with D. monhysteroides and consequently have their own effects on their host’s microbionts. Given nematodes’ association with many insects, particularly those with parental care, the conditionally positive effects nematodes have on their hosts’ fitness may be a more widespread feature of insect health.

S11

Notes

S12

Insulin/IGF signaling and vitellogenin provisioning mediate intergenerational adaptation to nutrient stress

L. Ryan Baugh

Duke University, USA

The roundworm C. elegans reversibly arrests larval development during starvation, but early- life starvation reduces reproductive success. We report that extended starvation in first-stage (L1) larvae followed by unrestricted feeding results in a variety of developmental abnormalities in the reproductive system, including germ-cell tumors and uterine masses, resulting in decreased fecundity. However, maternal dietary restriction buffers progeny from starvation as young larvae, preserving reproductive success. We found that maternal dietary restriction and reduced maternal insulin/IGF signaling increase oocyte provisioning of vitellogenin lipoprotein, reducing penetrance of starvation-induced abnormalities in progeny. Furthermore, we show that maternal dietary restriction and reduced maternal insulin/IGF signaling reduce insulin/IGF signaling in progeny. daf-16/FoxO and skn-1/Nrf, transcriptional effectors of insulin/IGF signaling, are required in progeny for maternal dietary restriction and increased vitellogenin provisioning to suppress starvation-induced abnormalities. daf- 16/FoxO activity in somatic tissues is sufficient to suppress starvation-induced abnormalities, suggesting cell-nonautonomous regulation of reproductive system development. This work reveals that early-life starvation compromises reproductive development and that vitellogenin-mediated intergenerational insulin/IGF-to-insulin/IGF signaling mediates adaptation to nutrient availability.

S13

Notes

S14

Intergenerational inheritance of metabolic state

Heidi Lempradl

Van Andel Institute, Grand Rapids, MI, USA

Given the sharp increase in the prevalence of heritable diseases such as obesity, it is exceedingly important to begin a systematic examination into non-genetic mechanisms of inheritance. We established a Drosophila model of paternal-diet-induced Inter-Generational Metabolic Reprogramming (IGMR). Intriguingly, as little as two days of dietary intervention in fathers can stably reprogram offspring physiology, lifelong, confirming the responsiveness of mature sperm to physiological cues. Paternal sugar leads to H3K9/K27me3 dependent alterations that affect expression of metabolic genes in two distinct germline and zygotic windows. Novel findings provide evidence towards the involvement of the piRNA pathway in IGMR. Mutant analysis reveals, that an intact piRNA pathway is critical for proper IGMR. Using multiple methods, we can detect all PIWI clade proteins in mature sperm suggesting the presence of a functional piRNA pathway. Importantly, small RNA sequencing of piwi bound RNAs in mature sperm reveals the presence of functional piRNAs, and fathers carrying a heterozygous mutation in key enzymes of the piRNA pathway give rise to reprogrammed offspring.

S15

Notes

S16

Energy-sensing pathways mediate intergenerational inheritance

Andre Pires, Pedro Robles, Anisa Turner

University of Warwick, School of Life Sciences, Coventry, UK

Information sensed by the maternal generation may pass to the offspring to increase their fitness. However, the mechanisms of inheritance of adaptive maternal effects through the germline are largely unknown. In the free-living nematode Auanema freiburgensis, the maternal environment determines stress-resistance and sex of the following generation: mothers kept in isolation produce mostly female offspring, whereas mothers exposed to high population densities produce stress-resistant larvae (dauers) that later develop into adult hermaphrodites. Ablation of maternal sensory neurons prevents the formation of F1 dauers and hermaphrodites, even when mothers are in high-density conditions. Exposure to high- density conditions correlates with histone modifications in the germline nuclei, such as low levels of H3K9me3 and high levels of H3 and H4 acetylation. Inhibition of demethylases involved in H3K9me3 demethylation prevents the formation of F1 dauers/hermaphrodite in all conditions. On the other hand, histone deacetylase inhibitors promote the formation of dauer/hermaphrodite formation in F1 in low-density conditions. Modification of chromatin structure may occur in response to conserved energy-sensing pathways, such as AMP- activated protein kinase (AMPK). High population densities increase the levels of PAR- 4/LKB1, a kinase that activates AMPK. Furthermore, maternal activation of AMPK increases the production of dauer/hermaphrodite offspring in low crowding conditions. Conversely, inhibition of AMPK in crowding conditions prevents the formation of dauer/hermaphrodite offspring. In summary, these results indicate the role of AMPK signalling, a highly conserved master regulator of metabolism, is involved in intergenerational inheritance through the germline.

S17

Notes

S18

Primed plants do not forget: mechanisms and ecological drivers of transgenerational acquired resistance in Arabidopsis thaliana

Joost Stassen1, Leonardo Furci1, Sam Wilkinson1, Ana Lopez1, Ritushree Jain, Lisa Smith1, Katia Symeonidi2, Frank Johannes2, and Jurriaan Ton1

1P3 Institute of Plant and Soil Biology, Animal and Plant Sciences, University of Sheffield, UK 2Technical University of Munich, Department of Plant Sciences, Hans Eisenmann-Zentrum for Agricultural Sciences, Germany

Plants can prime their immune system in response to hostile environmental signals and develop acquired resistance. In Arabidopsis, priming of jasmonic acid (JA)- and salicylic acid (SA)-dependent is a long-lasting acquired resistance responds that can lasts weeks. Evidence for an epigenetic basis of immune priming came from our discovery that progeny from heavily diseased Arabidopsis develops transgenerational acquired resistance (TAR), which requires active DNA de-methylation at/around transposable elements (TEs). TAR can be elicited by both biotrophic and necrotrophic pathogens, and its intensity and durability over stress-free generations are determined by the level stress encountered in the parent generation. TAR is associated with costs from increased susceptibility other (a)biotic stresses than the TAR-eliciting stress to which the parent plant had been exposed. Bisulfite- sequencing of independent lines that for >10 generations had been exposed to disease or SA treatment showed increased DNA de-methylation rates at TEs. To identify TAR- regulating epigenetic loci, we screened epigenetic recombinant inbred lines (epiRILs) for downy mildew resistance, and identified four TE-rich hypo-methylated pericentromeric loci controlling heritable disease resistance. Transcriptome analysis of selected epiRILs suggests that DNA de-methylation of TEs at these loci trans-control global defence gene priming.

S19

Notes

S20

Transgenerational environmental effects in variable environments: plant responses to environments within and across generations

Donohue, Kathleen; Alvarez, Mariano; Bleich, Andrew

Duke University, USA

All organisms develop in response to the environment they themselves experience, but they also respond to the environment of their parents and even grandparents. What is the contribution of these different generations—grandparents, parents, and individuals themselves—to developmental responses to environments, and how are responses to any given environment modified by exposure to past or present environments? We exposed the genetic model plant, Arabidopsis thaliana, to three generations of environmental manipulations to quantify how thermal environments experienced in consecutive generations influences plant development. First, we found that A. thaliana responds to environments experienced by themselves, their parents, and their grandparents. Responses to ancestral environments do not merely dissipate across generations, but effects of prior environments can be stronger than those of present environments and can even be in the opposite direction from them. Second, natural genotypes differed in the effects of ancestral environments, indicating that developmental responses to past environments have a genetic basis and may potentially evolve. Third, environments experienced across consecutive generations interact to produce developmental phenotypes, and genotypes differ in these interactions: specifically, individuals can respond to their own environment only if they experienced certain ancestral environments, but not others. Likewise, responses to ancestral environments are only manifest when individuals themselves experience certain environments, but not others. In particular, maternal environments that induce a form of developmental arrest (seed dormancy) enable progeny to respond to their own environments, suggesting that developmental arrest is an important mechanism that enables responses to immediate environments. In contrast, responses to maternal environments were manifest more strongly in favorable progeny environments that allowed differential developmental rates to be expressed. Finally, preliminary analysis suggests that environmental effects are transmitted across generations not by the direct transmission of methylation states, but by some other transmitted factor that regulates methylation states in progeny. In summary, developmental responses to ancestral environments vary in their temporal persistence across generations and they vary in their stability across present environments. Genetic variation in both aspects of transgenerational environmental effects suggests that the temporal persistence and environmental stability of transgenerational effects have a genetic basis and can evolve in response to natural selection imposed by diverse modes of environmental variation.

S21

Notes

S22

EMBO Young Investigator Lecture: DNA methylation reprogramming in plant germlines

Xiaoqi Feng, James Walker, Jingyi Zhang, Martin Vickers

John Innes Centre, UK

The existence of DNA methylation reprogramming in plant germlines was long debated, particularly because of the strong evidence for transgenerational methylation inheritance in plants. We found methylation reprogramming occurs throughout the male germline in Arabidopsis, and revealed the RNA-directed DNA methylation pathway (RdDM), which targets transposons in soma, induces methylation of genes specifically in the germline. This germline-specific methylation signature facilitates the splicing of a key meiosis gene in the meiotic cells, thereby regulating meiosis. My laboratory further investigated the presence of germline-specific genic methylation across land plants to ascertain the prevalence of this phenomenon and whether it relates to gametophytic function or sexual reproduction. We observed strong genic methylation specifically in rice and Brassica rapa germlines, and the meiotic sporophytes of Marchantia polymorpha, a species from the most basal lineage of extant land plants. This result demonstrated that germline methylation reprogramming is prevalent among land plants. Furthermore, Marchantia's special life cycle, in which meiosis and gametogenesis are well separated into different generations, allowed us to demonstrate that specific genic methylation relates to reproduction rather than gametophytic function.

S23

Notes

S24

Stress and survival in vertebrates: transgenerational effects of stress, environmental context, and why it matters

Kirsty MacLeod

Marie Skłodowska Curie Action (MSCA) Research Fellow, Lund University, Sweden

Ecological stressors such as predation can shape ecosystems, driving prey population and community dynamics through indirect, non-consumptive effects, such as long-lasting neurological, physiological and behavioural changes. I show that predation risk in a wild mammal (the snowshoe hare) can have lethal transgenerational effects with likely consequences for group size and population dynamics: in addition to reducing adult survival, predator exposure during gestation also reduced offspring survival after the period of risk had ended. I explore potential hormonal mechanisms for such effects using a different system, the eastern fence lizard, and demonstrate that short term elevations of glucocorticoid hormones at the level of a predator encounter can induce similar effects on adult survival and reproductive success. I will also discuss the importance of considering the ecological context in which maternal effects occur for determining their evolutionary importance across species and taxa.

S25

Notes

S26

Mums the word: trans-generational transmission of phenotype programmed by early- life stress in birds

Karen A .Spencer

School of Psychology & Neuroscience, University of St Andrews, Scotland, UK

The traditional view of developmental stress is one of constraint; many studies have shown that animals exposed to stressful environments during early life have a higher propensity to several pathologies, and that these negative consequences can be transmitted to future generations. However, an alternative adaptive hypothesis recognises the potential for developmental programming of behaviour, which enhances fitness if programmed environments match those experienced later in life. One intriguing possibility is also that these ‘positive’ effects are carried through into the next generation, also increasing fitness. Members of the Mechanisms of Behaviour group at St Andrews have been addressing this research question using an avian model – the Japanese quail. Avian species represent an excellent comparative model for understanding developmental programming due to the production of the egg, which receives discrete transfer of material for embryonic development, including glucocorticoid (GC) hormones. Importantly the levels of GCs can be experimentally manipulated within physiologically relevant limits to mimic those deposited by stressed mothers. This provides a powerful experimental tool to quantify individual responses to elevated GCs in developing young.

Here, I present experimental data on the neuroendocrine and behavioural effects of early life stress both within and across generations in our study system. We simulated both pre- and post-natal stressors via direct GC injection into the egg yolk and unpredictable food regimes in post-natal life, respectively. F1 birds that experienced pre-natal stress exhibited attenuated corticosterone responses to acute stress, more efficient feedback mechanisms within the HPA axis and increased motivation to explore a novel environment. F1 individuals that experienced stress at both pre- and post-natal development also showed heighted exploration behaviour in novel environments. Each F1 female was then allowed to breed twice, once under ad libitum food and once under unpredictable availability to create an F2 population. F2 Individuals from a pre-natally stressed mother showed the same behavioural responses to novel environments as their mothers. These individuals also exhibited elevated hippocampal expression of intracellular receptors that increase the negative feedback efficiency of the HPA axis resulting in attenuated physiological stress responses. Individuals from mothers exposed to both early-life stresses also showed increased exploration behaviour, but also consumed the most food during exploration trials. These results suggest that behavioural and neuroendocrine phenotypic traits programmed by early-life stress can be transmitted to offspring and this may enhance their ability to cope with stressful environments. However, these altered phenotypic traits may also have costs and the potential trade-offs are also discussed.

S27

Notes

S28

The role of epigenetic modifications in intergenerational changes induced by captive rearing

Sofia Consuegra, Deiene Rodriguez-Barreto, Waldir Miron Berbel, Carlos Garcia de Leaniz

Swansea University, UK

We are investigating the potential role of DNA methylation in the transmission to the offspring of behavioural and physiological changes caused by the early rearing environment of the parents. For this we are comparing wild and farmed Atlantic salmon and mangrove killifish reared in captivity with and without environmental enrichment. Interbreeding between hatchery-reared and wild fish, through deliberate stocking or escapes from fish farms, can result in rapid phenotypic and gene expression changes in hybrids, leading to maladaptation to the wild, but the underlying mechanisms are unknown. We assessed if one generation of captive breeding was sufficient to generate inter- and/or transgenerational epigenetic modifications in Atlantic salmon. We found that the sperm of wild and captive-reared males differed in methylated regions consistent with early epigenetic signatures of domestication. Some of the epigenetic marks that differed between hatchery and wild males affected genes related to transcription, neural development, olfaction and aggression, and were maintained in the offspring beyond developmental reprogramming. Our findings suggest that rearing in captivity may trigger epigenetic modifications in the sperm of hatchery fish that could explain the rapid phenotypic and genetic changes observed among hybrid fish[1]. Thus, epigenetic introgression via fish sperm represents a previously unappreciated mechanism that could compromise locally adapted fish populations. Taking advantage of the self-fertilising nature of the mangrove killifish Kryptolebias marmoratus we also investigated the effects of the rearing environment in parents and offspring by comparing neophobia, metabolic rate and brain DNA methylation patterns of genetically-identical fish reared in enriched or barren environments. Parental fish reared in enriched environments had lower cortisol levels, lower metabolic rates and were more active and neophobic than those reared in barren environments. They also differed in 584 methylated cytosines (DMCs). Offspring activity and neophobia were determined by the parental environment. We found evidence of both putative intergenerational and transgenerational parental effects on the DNA methylation patterns of the offspring, the first being more prominent. Among the DMCs of the parents, 55 followed the same methylation patterns in the offspring, five of which were significantly influenced by parental environments. Our results suggest that the environment experienced by the parents influences behaviour and brain DNA methylation patterns of the offspring[2].

1. Rodriguez Barreto D., Garcia de Leaniz C., Verspoor E., Sobolewska H., Coulson M., Consuegra S. 2019 DNA methylation changes in the sperm of captive-reared fish: a route to epigenetic introgression in wild populations. Molecular Biology and Evolution.

S29

Notes

S30

Maternal stress exposure leads to diversification of offspring behaviour in annual freshwater fish

Agnieszka Magierecka, Neil B. Metcalfe

Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, UK

Exposure of females to stressful conditions during the period of egg production or embryo formation can have a profound effect on the behaviour of their offspring. This effect is mediated by factors such as maternally derived stress hormones and mRNAs, which play an important organisational role during neurogenesis and neurodevelopment. As a result, various aspects of the post-natal behavioural phenotype (e.g. aggression, boldness, anxiety- like behaviour) may show altered patterns. These changes may be in terms of the consistency of behavioural patterns as well as in their average level of performance. Whilst the repeatability of individual behaviour is well documented, including in the context of maternal effects, few studies have quantified the consistency of offspring behavioural traits between related individuals, particularly in response to maternal stress. In this study, I attempted to answer this open question using an annual population of three-spined sticklebacks (Gasterosteus aculeatus). In the period leading up to spawning, I exposed adult females to an unpredictable chronic stress protocol, in which they experienced a range of environmental and husbandry stressors. I then subjected the offspring from the successive breeding attempts (clutches) of these females to a number of behavioural trials and quantified their activity, sheltering and anxiety-like behaviour. I calculated within- (siblings) and between-clutch (half-siblings) repeatability scores for these behaviours, which were translated into an intra-family variability, where high repeatability indicated low variability between siblings or half-siblings, and vice versa. Exposure of a mother to a period of chronic stress resulted in diversification in the behaviour of her offspring, as indicated by an increased within-family variability in behavioural traits such as activity levels and use of a shelter. This effect was consistent both within and between the clutches of the same female. At the same time, control females produced offspring that showed significant consistency of behaviour between siblings and half-siblings. This result may be an effect of a bet-hedging strategy adopted by female sticklebacks in a stressful and unpredictable environment. In such environments, it would be beneficial to increase maternal fitness by producing offspring differing in behavioural phenotype, and thus increasing the chance that some of these offspring will be able to better cope with the prevailing environmental conditions.

S31

Notes

S32

Parental delivery: somatic signals impacting neurodevelopment

Tracy L. Bale1, J. Chan1, C. Morgan1, N.A. Leu3, S. Ament1, B. Garcia3, M. Kane2, C.N. Epperson4

Departments of Pharmacology & Psychiatry and Center for Epigenetic Research in Child Health and Brain Development, University of Maryland School of Medicine1, School of Pharmacy2, Baltimore, MD; University of Pennsylvania3, and University of Colorado School of Medicine4, USA

Parental lifetime exposures to perturbations such as stress, infection, malnutrition, and advanced age have been linked with an increased risk for offspring disease, including a strong association with neurodevelopmental disorders. Our studies have focused on identifying the causal biological mechanisms whereby information in the environment can be transmitted in sperm. In these studies, we demonstrate a causal role for somatic-to-germline transmission of stress information capable of altering fetal neurodevelopment via extracellular vesicles (EVs) in a preclinical model, and we further establish the translational potential of this model in a human cohort. We identified broad histone and transcriptomic alterations in mouse epididymal epithelial cells (EECs) in vivo, with corresponding persistent changes in miRNA and proteomic extracellular vesicle cargo secreted from pure caput EEC populations in vitro. In these studies, the transmission of paternal stress and changes in epigenetic marks only occur following a stress recovery period suggesting a cellular allostatic shift in chromatin programming. Using a transgenic approach, we demonstrated in vivo that the EEC glucocorticoid receptor, a central node of cellular stress mechanisms, serves a key role in this cellular programming where genetic knockdown of this receptor rescued the germline transmission of the offspring stress phenotype. We further established the causal involvement of EEC EVs utilizing intracytoplasmic sperm injection of caput sperm incubated with EVs secreted following stress treatment recovery. Translationally, we have examined similar stress programming changes in human sperm miRNA content. We utilized within and between human subject comparisons of sperm samples and neuropsychiatric perceived stress reports collected repeatedly over six months to identify a distinct miRNA expression pattern from subjects showing a post-stress recovery state. Together, these studies demonstrate in both mice and humans a clear involvement of stress in the environment to impact sperm miRNA content, and a novel role for reproductive tract EVs to harness changes in the paternal milieu and integrate into germ cell signals.

S33

Notes

S34

Experimental evidence for fetal programming via epi-sensitive regions of the genome

Rosalind John, Matt van de Pette, Grainne McNamara, Simon Tunster, Anthony Isles, Mandy Fisher

RJ, GM and AI - Cardiff University; MvP and MF - Imperial College; ST - Cambridge University, UK

Early life adversity, especially during pregnancy, is linked to fetal growth restriction and significantly poorer health outcomes for children in later life including increased risk of metabolic and mental health disorders. Epigenetic inheritance is a highly promising mechanism linking these early life exposure to later outcomes as epigenetic marks, unlike DNA sequence, can respond reversibly to environmental cues. Imprinted genes have been highlighted as potential targets of fetal programming because these remarkable genes are monoallelically expressed and precisely regulated as a consequence of epigenetic mechanisms, and because studies in mice have shown that modest alterations in the expression of individual imprinted genes can have a disproportionate impact on fetal growth, placental development, metabolism and behaviour. We developed an in vivo mouse reporter strain in which the bioluminescence enzyme luciferase was targeted into the 3' UTR of maternally expressed gene cyclin dependent kinase inhibitor 1c (Cdkn1c). These mice were used to demonstrate that dietary protein restriction during gestation results in a loss of imprinting of Cdkn1c with expression detected from the paternal allele in utero through live imaging (Van de Pette et al., 2017). This altered epigenetic state persisted in animals as they matured, even when a normal diet was resumed. At a molecular level, loss of paternal silencing was associated with erosion of DNA methylation at the somatic differentially methylated region, and prevented by folate supplement in pregnancy. Exclusive LOI of Cdkn1c results in low birth weight (Andrews et al., 2007), metabolic disturbances (Van de Pette et al., 2018; Van De Pette et al., 2016), altered behaviour and changes in dopamine circuitry (McNamara et al., 2018a; McNamara et al., 2016; McNamara et al., 2018b). These proof-in-principle experiments support the hypothesis that fetal programming, in part, acts via epi-sensitive regions of the genome (imprinted loci).

S35

Notes

S36

Improving neurobehavioural outcomes following exposure to stressful events in pregnancy

Jonathan J Hirst, Hannah K Palliser, Julia C Shaw, David W Walker, Gabrielle K Crombie

Mothers and Babies Research Centre, Hunter Medical Research Institute, School of Biomedical Sciences and Pharmacy, Newcastle, Australia; and School of Health & Biomedical Sciences RMIT University, Melbourne, Australia

Background: Extensive evidence shows that stressful events in pregnancy can affect neurodevelopment outcomes. These pregnancy compromises lead to life-long programming that markedly increase the risk of emotional and behavioural disorders in later life. Neurosteroids have a key role in brain development particularly through promotion of myelination. Chronic stress exposure in pregnancy has been shown to decrease neurosteroid synthesis, decrease myelination and disrupt GABAergic pathways in the fetal brain. This results in an increased risk of behavioural deficits including anxiety and attention deficit hyperactivity disorder (ADHD). We propose that stimulation of neurosteroid synthesis through administration of a ligand (Emapunil) of the mitochondrial translocator protein (TSPO) will reduce these adverse outcomes.

Methods: Pregnant guinea pigs were exposed to control-handling or light stress (strobe light exposure for 2hrs/day on gestational age 50, 55, 60 and 65 (term 70 days). In the neonatal period, offspring were allocated to receive emapunil (0.3mg/kg daily) or vehicle (45% β- cyclodextrin) on postnatal day (PND) 2-8 and underwent open field and elevated plus maze testing on PND 28. Mature myelination was assessed by myelin basic protein immunohistochemistry and GABAA receptor α2, α4, and α5 subunit expression was assessed with RT-PCR.

Results: At PND28 the prenatal stress exposure group showed a hyperactive phenotype when compared to control in the open field arena, displaying increased line crossings (p=0.038), entries into the inner zone of the arena (p=0.046) and distance in the inner zone (p=0.035). This hyperactive phenotype was also displayed in the elevated plus maze with the prenatal stress group displaying increased distance travelled in the open arm (p=0.013) and closed arm of the maze (p=0.028). The prenatal stressed neonates that were treated with emapunil did not show these hyperactive patterns of behaviour. Myelination was decreased at PND30 in the prenatal stress group while the emapunil-treated stress group displayed control levels of myelination. Similarly, only the prenatal stress group displayed increased mRNA expression of the GABAA receptor subunits.

Conclusions: This study further supports the negative role of prenatal stress in programming for negative neurobehavioural outcomes in the offspring. This work also shows that therapeutic intervention during the early neonatal period can reverse these negative outcomes. The mechanisms involved may include changes in GABAergic pathways. Postnatal stimulation of the neurosteroid synthesis pathway with TSPO ligand emapunil may be a viable treatment option following exposure to marked prenatal stress, thus warrants further investigation.

S37

Notes

S38

The prenatal maternal response to immune activation predicts offspring cognitive dysfunction in a rat model of schizophrenia

Harry Potter, Rebecca Woods, Grace Revill, Hager Kowash, Jocelyn Glazier, Joanna Neill, Reinmar Hager

Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK

Converging evidence from epidemiological studies and animal models implicates maternal immune activation (mIA) as a risk factor for neurodevelopmental disorders such as schizophrenia in the adult offspring. The transient increase in maternal pro-inflammatory cytokines following mIA is thought to perturb fetal neurodevelopment and results in cognitive deficits. Here, we employed a split-litter cross-fostering design to investigate how the prenatal and postnatal maternal environments interact to predict offspring cognitive deficits in a rodent model of mIA.

20 female Wistar rats were timed-mated and treated with a single intraperitoneal injection of 10mg/kg bodyweight polyinosinic-polycytidylic acid (poly I:C; low-molecular weight, InvivoGen) or vehicle (endotoxin-free 0.9% saline) on gestational day 15 (GD15). Offspring were culled to 10 pups/litter on postnatal day 1 (PD1) and either crossed to a dam in the opposite treatment group or remained in their home litter. Offspring ultrasonic vocalisations (USVs) were recorded on PD6, 10, and 14 and analysed using the open-source MATLAB script MUPET. Offspring were tested on the attentional set-shifting task (ASST) in adulthood. Global DNA methylation in the frontal cortices of offspring at a prenatal (GD21) and postnatal (PD21) time-point were measured by ELISA (Enzo Life Sciences). Statistical analyses were carried out in SPSS using General Linear Mixed Models (GLMM).

Prenatal exposure to poly I:C significantly increased the number of syllables emitted by female (GLMM, F1,19=6.26, p=0.022, n=24-28) and male (GLMM, F1,20=8.87, p=0.008, n=25-31) pups with no effect of cross-fostering. In adulthood, offspring exposed to poly I:C showed a deficit in the ASST as evidenced by a significant increase in the intra-extra dimensional shift, regardless of sex (GLMM, F1,49=7.42, p=0.009, n=5-7). Poly I:C caused a significant and sex-specific increase in global DNA methylation in males only at GD21 (GLMM, F1,8=18.97, p=0.002), but increased DNA methylation in females only at PD21 (GLMM, F1,8=6.39, p=0.035).

Our data suggests that the prenatal maternal response to poly I:C, rather than the postnatal maternal environment, is critical for the developmental programming of behavioural and cognitive deficits associated with schizophrenia in a rat model of mIA. Using a split-litter cross-fostering design we show that mIA by poly I:C on GD15 causes an increase in number and duration of vocalisations in early postnatal life followed by a clinically-relevant deficit in cognitive flexibility in adulthood, regardless of sex or the postnatal maternal environment. mIA may induce these developmental changes through an epigenetically-regulated mechanism which acts in a sex- and age-specific manner.

S39

Notes

S40

Programming by maternal over-nutrition: a plentiful problem

Susan Ozanne

University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, UK

Obesity prevalence is increasing in both the developed and developing world in all age groups within the population including women of childbearing age. In many countries, such as the UK, over half of women are now overweight or obese during pregnancy. This is of concern as evidence suggests that developing in utero in an obesogenic environment not only has short term detrimental effects for both mother and baby but also has long-term effects on the cardio-metabolic health of the child later in life, including their risk of obesity. Therefore, the current obesity epidemic is priming the next generation for a lifetime of poor health. This has been termed the Developmental Origins of Health and Disease and has been supported by studies in humans and in a range of animal models (from drosophila to non-human primates). We have used a mouse model of maternal diet-induced obesity to define the mechanisms by which obesity during pregnancy impacts on the long-term cardio- metabolic health of the offspring that could help in the design of rationale intervention strategies. Maternal obesity is induced by feeding females a high fat diet supplemented with condensed milk. Our studies have demonstrated that young adult offspring of obese dams develop insulin resistance, pathological left ventricular cardiac hypertrophy, cardiac dysfunction, hypertension, fatty liver and age-associated increased adiposity when weaned onto a healthy diet. They are also more susceptible to diet-induced obesity. Our studies have identified maternal insulin as a key ‘programming” factor and highlight it as an important target for intervention studies such as those involving increased maternal physical activity or pharmacological interventions. Our findings suggest that effective interventions to reduce the burden of cardio-metabolic disease and obesity should start early in life and not delayed until adulthood when disease has become manifest.

S41

Notes

S42

Epigenetic modulation of repeat elements

Anne C. Ferguson-Smith

Department of Genetics, University of Cambridge, UK

Genetic models of epigenetic inheritance can provide useful insights into the mechanisms, stability and heritability of modified states. Endogenous retroviruses (ERVs) are a class of LTR retrotransposons representing around one quarter of the repetitive elements in the murine genome. Most are epigenetically silenced. However, in two classic mouse models, Agouti viable yellow (Avy) and Axin fused (Axin(Fu)), a member of the IAP class of ERV has inserted in the vicinity of the agouti and axin genes respectively, and been variably DNA methylated between individuals; this is associated with transcriptional variability of the associated genes, and non-genetically conferred phenotypic variation which can be transmitted across generations. Such alleles are known as metastable epialleles. We have conducted a genome-wide systematic screen for metastable epialleles at ERVs using mouse strain-specific datasets that we generated as part of the BLUEPRINT reference epigenome project (EUFP7 BLUEPRINT grant HEALTH-F5-2011-282510). The properties, impact and heritability of these novel variably methylated IAPs provides useful insights into the impact of the epigenetic control of repetitive elements on the mammalian genome and transgenerational epigenetic inheritance. In particular, the variable methylation state is locus- specific within an individual and very few act as heterologous promoters for adjacent genes. Of interest, variably methylated IAPs are enriched for the methylation-sensitive DNA binding factor CTCF which may influence the establishment/maintenance of the metastable state as well as contribute longer range functional effects. Variably methylated IAPs are reprogrammed after fertilization and re-established as variable loci in the next generation indicating reconstruction of metastable epigenetic states. Finally, we explore the sensitivity of metastable epialleles to environmental effects to determine the extent to which they can act as biosensors of environmental compromise and mediators of environmental effects on genome function.

S43

Notes

S44

Effects of maternal obesity on oocyte methylome are reflected on blastocyst epigenome

A. Galvão1,2, S. Clark1, K. Wolodko1, Juan Castillo-Fernandez1, G. Kelsey1

1 Epigenetics Programme, The Babraham Institute, Cambridge, UK 2 Institute of Animal Reproduction and Food Research, Polish Academy of Science, Olsztyn, Poland

The oocyte epigenome has the potential to control initial reprogramming events in the early embryo, as well as metabolic outcomes in offspring. We are investigating the impact of maternal obesity on the oocyte and blastocyst methylome and transcriptome.

Using a combined method we performed single-cell RNA-sequencing and single-cell bisulfite sequencing in MII-oocytes from C57Bl/6J (B6) mice fed chow diet (CD) or high-fat diet (HFD) for 16 weeks. Subsequently, we performed in vitro fertilisation on oocytes and addressed how metabolically-driven changes in the oocyte epigenome could affect the E3.5 embryo methylome and transcriptome at a single blastocyst level.

DESeq2 analysis identified 195 differently expressed genes in oocytes (p<0.05). Interestingly, transcripts affecting early development, such as Dppa3 and Plac1, were significantly increased in HFD oocytes. Unbiased CpG methylation analysis of the oocytes revealed 450 differently methylated regions (DMRs), with an absolute cut-off of 20% (p<0.05): 61% of these DMRs overlapped genes and lost methylation (p<0.05), whereas the remaining 39% were intragenic and gained methylation in HFD oocytes (p<0.05). Whole genome methylome analysis in blastocysts revealed 205 DMRs (p<0.05), with 10% of blastocyst DMRs coinciding with those in the oocyte, suggesting potential longer-term consequences of methylation changes induced in oocytes. In conclusion, we give the first critical evidence that the oocyte epigenome senses maternal metabolic changes and show that methylation changes can persist at least during preimplantation development.

S45

Notes

S46

Effects of prenatal stress on lactation and offspring development: linking evolutionary models with human cohort studies

Sinead English [1], India Wright [2], Verity Ashburn [2], Gemma Ford [2], Doretta Caramaschi [3]

1. School of Biological Sciences, University of Bristol, UK 2. Reproduction and Development Programme, Bristol Medical School, Translations Health Sciences, University of Bristol, UK 3. MRC Integrative Research Unit, Bristol Medical School, Population Health Sciences, University of Bristol, UK

There is current debate on whether maternal stress primarily imposes costs on offspring development (constraint hypothesis) or serves as a cue to prepare them for stressful environments (programming hypothesis). These different hypotheses generate several predictions on how maternal stress may influence offspring development. Much research in testing the link between stress and development has focused on exposure in utero, yet maternal stress can have delayed effects through reduced investment during lactation. Moreover, while there have been several studies exploring the link between prenatal stress, maternal investment and offspring development across mammals - mainly primate and rodent laboratory models - fewer studies have explored these processes in humans using cohort studies. Here, we present, first, an evolutionary framework to generate testable predictions on how maternal stress might impose constraints on, or adaptively programme, offspring development and we then interrogate these predictions using a UK birth cohort dataset.

S47

Notes

S48

Transgenerational transmission of susceptibility – evidence from the Dutch famine

Tessa Roseboom

Amsterdam UMC, The Netherlands

In this presentation the findings of studies among men and women who were born around the time of the Dutch famine of 1944-1945 will be presented, demonstrating effects of undernutrition during critical periods of development on later health and disease. The Dutch famine was remarkable in several ways and its unique features have allowed scientists to investigate the long term consequences of prenatal undernutrition in humans. The effects of undernutrition depended on its timing during gestation, and the organs and tissues undergoing critical periods of development at that time. Early gestation appeared to be the most vulnerable. The effects of famine were widespread and affected the structure and function of many organs and tissues, resulted in altered behaviour and increased susceptibility to chronic degenerative diseases, which in turn led to reduced participation in the labour market and increased mortality. Also, the effects of famine were independent of size at birth, which suggests that programming may occur without altering size at birth. We found indications that the effects of prenatal famine exposure may not be restricted to those exposed to it prenatally: their offspring also seems to be affected, which might be mediated through epigenetic mechanisms.

Studies in other settings show that those faced with undernutrition during the critical earliest stages of development have increased rates of chronic generative disease in adult life. This suggests that these findings reflect biologically fundamental processes that describe human plasticity. These findings teach us the fundamental importance of a good start in life.

S49

Notes

S50

Heart disease link to fetal hypoxia: an intergenerational perspective

Dino A. Giussani

Department of Physiology, Development & Neuroscience, University of Cambridge, UK

The quality of the intrauterine environment interacts with our genetic makeup to shape the risk of developing disease in later life. Fetal hypoxia is a common complication of pregnancy. It programmes cardiac and endothelial dysfunction in the offspring in adult life. However, the mechanisms via which this occurs remain elusive, precluding the identification of potential therapy. Using an integrative approach in large and small animal species at the in vivo, isolated organ, cellular and molecular levels, my programmes of work have raised the hypothesis that oxidative stress in the fetal heart and vasculature underlies the mechanism via which prenatal hypoxia programmes cardiovascular dysfunction in later life. We have shown that developmental hypoxia independent of changes in maternal nutrition promotes fetal growth restriction and induces changes in the cardiovascular, metabolic and endocrine systems of the adult offspring, which are normally associated with disease states during ageing. Treatment with antioxidants of animal pregnancies complicated with reduced oxygen delivery to the fetus prevents the alterations in fetal growth, and the cardiovascular, metabolic and endocrine dysfunction in the fetal and adult offspring. Interestingly, the risk of programmed heart disease by adverse pregnancy can be transmitted across generations via the paternal line. Conversely, the mother can transmit onto her offspring protection against future heart disease via mitochondrial mechanisms. We can therefore modify the risk of heart disease not only in our children but also in their children and their children’s children.

Funding by The British Heart Foundation

S51

Notes

S52

Early life epigenetic predictors of cardiometabolic health in childhood

Toby Mansell (1,2), Anne-Louise Ponsonby (1,2,3), Fiona Collier (1,4,5), David Burgner (1,2,6), Peter Vuillermin (1,4,5), Joanne Ryan (1,7), Richard Saffery (1,2), Barwon Infant Study Investigator Team

1 Murdoch Children’s Research Institute, Parkville, Australia 2 Department of Paediatrics, University of Melbourne, Parkville, Australia 3 The Florey Institute of Neuroscience and Mental Health, Parkville, Australia 4 School of Medicine, Deakin University, Geelong, Australia 5 Child Health Research Unit, Barwon Health, Geelong, Australia 6 Department of Paediatrics, Monash University, Clayton, Australia 7 School of Public Health & Preventive Medicine, Monash University, Melbourne, Australia

It is now clear that cardiovascular and metabolic (cardiometabolic) conditions have their origins early in life, potentially even prior to birth. A limited number of studies suggest a role of epigenetic markers, including DNA methylation, as potential mediators of this risk, but compelling evidence is lacking. The relationship of epigenetics with cardiometabolic health in early life is generally poorly understood, but there is the potential for early life epigenetic variation to associate with cardiovascular development and metabolic health in childhood. Understanding these relationships may both contribute to unravelling the molecular aetiology of cardiometabolic disease risk and also provide additional predictive biomarkers to identify those most at risk of poor cardiometabolic outcomes earlier in the life course, creating the opportunity for more effective intervention.

As part of the Barwon Infant Study, an Australian population-based longitudinal birth cohort, we investigated the relationship between pregnancy exposures, infant birth outcomes and infant methylation of two genes linked to adult BMI and obesity (hypoxia-inducible factor 3α, HIF3A; and leptin, LEP) in blood from birth and 12 months of age. We then considered the potential association between early life methylation of these genes and cardiometabolic measures at four years of age, and the impact of infant genetic variation on these relationships.

To date we have identified associations between early life exposures (gestational diabetes, pre-eclampsia, infant sex, gestational age, and HIF3A genetic variation) and cord blood methylation of HIF3A, and evidence that this methylation predicts systolic and diastolic blood pressure at four years of age, primarily in males.

Infant sex, pre-eclampsia, birth weight and genetic variation were independently associated with LEP cord blood methylation. Adiposity measures at birth predicted 12-month LEP methylation in a genotype-dependent manner. We also found higher methylation of a specific CpG site at 12-months of age was associated with decreased weight at four years.

While the relationships between early life methylation and four-year cardiometabolic measures persisted for both these genes after adjustment for anthropometric measures at birth and 12 months of age, our analysis indicated that the DNA methylation markers had comparatively limited predictive utility in isolation. Future studies examining the relationship between longitudinal metabolomic profile with similar outcome measures are underway.

S53

Notes

S54

Intra-uterine growth restriction secondary to maternal obesity and hyperglycaemia is associated with altered glucose metabolism and expression of the DNA methyltransferases in the offspring brain

Kahyee Hor, Giles Hardingham and Amanda Drake

KH and AD - Centre for Cardiovascular Science, University of Edinburgh. GH - Centre for Discovery Brain Sciences, University of Edinburgh, UK

Neurodevelopment is particularly vulnerable to adverse influences during fetal life. Emerging evidence demonstrates that offspring of obese or hyperglycaemic women have an increased risk of neurodevelopmental disorders although the mechanisms underlying this observation remain unclear. I hypothesise that in utero exposure to maternal obesity and hyperglycaemia alters offspring brain metabolic and epigenetic landscape, with downstream neurodevelopmental effects.

Using an established murine diet-induced obesity model, C57BL/6 dams were commenced on either a high fat (HFD) or control diet (CON) at 3 weeks of age. C57BL/6 males on standard chow were used for mating. Dams underwent 2 mating cycles to ensure breeding ability and offspring from the second pregnancy were sacrificed on postnatal day 1 for analyses.

Dams in the HFD group (n=7) were significantly heavier (p<0.0001) with higher random blood glucose levels (p=0.02) than CON group (n=6) throughout the pre-mating, pregnant and post-natal period. HFD offspring had a significantly lower birthweight (p=0.0003) and blood glucose(p=0.018) but higher brain:body weight ratio (p=0.0163).

RT-qPCR on candidate genes was performed on offspring frontal cortex and cerebellum. Although there was no difference in cortical or cerebellar GLUT1 expression, cerebellar Hexokinase 1 (Hk1) was downregulated in HFD offspring (p=0.025). Cerebellar DNMT1 gene expression was lower in HFD offspring (p=0.015) and a similar trend was observed in DNMT3a gene expression in cerebellum (p=0.067) and cortex (p=0.054).

The reduction in expression of the rate-limiting enzyme Hk1 suggests that maternal obesity/hyperglycaemia associates with altered glucose metabolism in the offspring brain. DNA methylation is a key epigenetic modification for normal neurodevelopment and perturbed cerebellar DNA methylation has been implicated in disorders including schizophrenia. The reduction in DNMT1 and 3a expression among HFD offspring suggests that there could be effects on DNA methylation.

Further experiments are in progress to interrogate the implications of these findings in order to understand the impact of maternal obesity and hyperglycaemia on fetal neurodevelopment.

S55

Notes

S56

Enduring consequences of fetal stress

Kent L. Thornburg, Ph.D.

M. Lowell Edwards Chair, Professor of Medicine Director, Center for Developmental Health, Knight Cardiovascular Institute Director, Bob and Charlee Moore Institute for Nutrition & Wellness Oregon Health & Science University, Portland, OR, USA

Once it was clear that low birthweight was associated with lethal heart disease as described originally by Professor David Barker and colleagues, a number of other chronic diseases and medical conditions were found to be related to fetal growth patterns. In some instances, chronic disease was related to a fetal stressor that did not affect birthweight. Thousands of epidemiological and animal studies have now uncovered many aspects of developmental accommodations which underlie the enduring propensity for chronic disease over the lifecourse. Of all the possible stressors underlying poor intrauterine growth, four stand out as most prevalent and pervasive. These include poor maternal nutrition before and during pregnancy and lactation, severe maternal social stress, excessive exposure to toxic chemicals and poor fetal oxygenation-- usually derived from poor placentation. In addition to these, several common maternal conditions including preeclampsia and diabetes mellitus underlie disease risk in offspring in unknown fashion. There is increasing evidence that often ignored parental social conditions affect placental growth and function and lead to offspring vulnerability for disease. These stressors affect the fetoplacental unit by compromising developing organ structure, biochemical regulatory pathways and altered gene expression patterns through epigenetic mechanisms. The result is a degradation of anti-oxidant defenses, anti-inflammatory processes, immune function as well as apparent dysregulation of stem cell quality and numbers that underpin fetal vulnerability for disease. While the placental size and shape are clearly associated with specific disease outcomes, predicting disease outcome based on placental phenotype is not yet possible. Nevertheless, it is clear that fetuses who have endured stresses and made physiological compromises have high risks for chronic diseases if they experience a postnatal life full of adversity.

S57

Notes

S58

Poster Presentations

Are glucocorticoids involved in the transmitting the programming effects of maternal stress to the fetus?

Paula J Brunton, Joana Fernandes, Ying Sze

Centre for Discovery Brain Sciences, University of Edinburgh, UK

Chronic maternal social stress during pregnancy exerts negative 'programming' effects on the developing fetus, resulting in altered offspring physiology and behaviour in later life. The mechanism through which this programming occurs is unclear, although a long-held assumption is that excess maternal glucocorticoid transfer to the fetus plays a role. However, the presence of the enzyme, 11β-hydroxysteroid dehydrogenase type 2 (11βHSD2), which converts corticosterone into the inactive metabolite 11- dehydrocorticosterone (11-DHC), in the placenta acts as a 'protective barrier'. 11βHSD2 and 11βHSD1 (which catalyses the reverse reaction) are also expressed in the fetal brain, where both enzymes act together to regulate local glucocorticoid concentrations.

Here, we aimed to establish whether maternal glucocorticoids are directly transferred from the maternal to fetal circulation following chronic social stress, and whether the placental 11βHSD2 barrier is compromised by chronic stress exposure. Pregnant rats were exposed to 10 min social stress/day from gestational day (GD) 16-20 and were killed on GD20 after the final stress exposure. Corticosterone and 11-DHC concentrations in the maternal and fetal plasma, placenta, fetal brain and liver were quantified using liquid chromatography- mass spectrometry. In situ hybridisation was used to quantify placental 11βHSD2 and glucocorticoid receptor (GR) mRNA expression, which may indicate placental glucocorticoid sensitivity. In the foetal hippocampus, 11βHSD2 and 11βHSD1 gene expression was also quantified.

Both plasma corticosterone and 11-DHC concentrations were markedly greater in stressed pregnant rats compared to controls. Maternal stress resulted in a modest increase in circulating corticosterone concentrations in the female fetuses, but had no effect in the males. Corticosterone concentrations were significantly greater in the liver of both male and female fetuses following maternal stress; but no changes were detected in the fetal brain. 11-DHC concentrations were not different in fetal plasma, brain or liver. In the placenta, stress resulted in greater corticosterone concentrations and 11βHSD2 mRNA expression was increased in males, but not females. There were no changes in placental GR, nor hippocampal 11βHSD1 and 11βHSD2 gene expression in the fetal brain in either sex.

In conclusion, although repeated social stress increases maternal corticosterone secretion, the placental barrier appears intact, especially in the males, and direct transfer of corticosterone from the maternal to fetal circulation is minimal. The data suggest maternal glucocorticoids are not directly involved in transmitting the programming effects of maternal stress to the fetuses, but may act in an indirect sex-dependent manner.

Funding: BBSRC, BSN (PJB). UoE Principal's Career Development PhD Scholarship (SY).

P1

NFkB signalling is implicated in a novel mouse model of early life stress

Eamon Fitzgerald 1, Megan C Holmes 1, James P Boardman 2, Amanda J Drake 1

1 University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, The Queen’s Medical Research Institute, 47 Little France Crescent, Edinburgh, UK; 2 MRC Centre for Reproductive Health, University of Edinburgh, The Queen’s Medical Research Institute, 47 Little France Crescent, Edinburgh, UK

Preterm birth is a major risk factor for neurodevelopmental disorders (e.g. schizophrenia, autism spectrum disorders (ASD)), but the mechanisms behind this remain poorly understood. Many preterm infants experience physiological and environmental stress (e.g. excessive sensory stimuli and inflammation). To mimic this, we developed a novel model of early-life stress (modified maternal separation or MMS) to include maternal separation and gentle agitation, with or without prior LPS exposure. Given that accurate spatial and temporal cortical gene expression is crucial for normal development, and cortical transcriptomes are altered in schizophrenia and ASD, we hypothesized that the MMS paradigm would alter the cortical transcriptome and that this would be potentiated by LPS exposure.

C57/Bl6 male mice were divided into 4 groups: 1) MMS for 1.5 hours/day for 3 days (P4-P7) 2) 1mg/kg LPS by IP injection 24 hours prior to MMS 3) LPS alone 4) controls (littermates) which were left in the home cage. Pups were weighed daily. At P7, all mice were killed by decapitation, blood collected, and the brain removed and halved along the midline: one half was embedded for paraffin sectioning and immunohistochemistry, the cortex was dissected from the remaining half and snap-frozen for RNA extraction. Plasma corticosterone was measured by ELISA and the cortical transcriptome profiled using 3'RNA sequencing (N of 3/group). Sequencing data were analysed using the Voom (sample weights) method, all other data were analysed using two-way ANOVA.

LPS reduced weight gain (p<0.001) but there was no effect of MMS (p=0.35) and no interaction between LPS and MMS (p=0.87). There were no differences in populations of Iba1+ (p=0.45, 0.89 and 0.77 for effect of MMS, LPS, interaction between MMS/LPS respectively), Olig2+ (p=0.29, 0.86, 0.51) or GFAP+ (p=0.77, 0.32, 0.48) cells. NR3C1 expression was reduced after MMS (p=0.03) with no independent effect of LPS (p=0.73) or interaction (p=0.85). There were no effects of MMS (p=0.29) or LPS (p=0.42) on plasma corticosterone levels. 3'RNA sequencing revealed 1043 genes with ≥2-fold expression changes following MMS, with a significant enrichment of genes regulated by NFkB signalling. In contrast, when LPS was given before MMS, only 228 genes showed a >2-fold expression change, with no larger role for NFkB signalling than MMS alone.

In conclusion, here we present a novel mouse model of early-life stress with relevance to preterm birth. We see a transcriptomic signature associated with increased NFkB signalling following MMS which is not potentiated with prior LPS exposure.

P2

Why are genetically identical worms growing in the same environment phenotypically different?

Mirko Francesconi (1), Marcos Francisco Perez (2), Ben Lehner (2,3,4)

1 Laboratory of biology and modelling of the cell (LBMC) ENS-Lyon 46 allee d’Italie 69394 LYON CEDEX 07, France. 2 Systems Biology Program, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology (BIST) Dr. Aiguader 88, 08003 Barcelona, Spain.3 Universitat Pompeu Fabra (UPF), Dr. Aiguader 88, 08003 Barcelona, Spain.4 Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain

Genetically identical individuals growing in the same environment often show substantial phenotypic variation. The reasons for this phenotypic variation are usually unknown. We found that isogenic C. elegans growing in the same environment differ in developmental speed, including the relative developmental rate of the soma and the germline (soma- germline heterochrony), fecundity and many other fitness important traits such as size and resistance to early starvation. We have previously shown that a change in maternal provisioning of yolk to the embryo, which increases with age, explains some but not all of these phenotypic differences, for example it does not explain differences in soma-germline heterochrony and fecundity. We now show that these differences are induced by parental exposure to pheromone: offspring of pheromone exposed individuals have a comparatively delayed germline development but a higher fecundity, that are not explained by a change in yolk provisioning. Pheromone conveys information about the environment such as population density and food quantity/quality. These observations therefore suggest that worms might have evolved a way to tune their progeny development and life history strategy to adapt to environment they are likely to encounter.

P3

Maternal age and maternal diet affect offspring lifespan, fitness, and response to caloric restriction

Dr Kristin Gribble, Martha Bock, George Jarvis, Emily Corey, Emily Stone

Marine Biological Laboratory, UK

"Maternal effects" occur when a mother's phenotype or response to the environment causes a change in offspring phenotype without a change in genotype. To better understand the role of maternal effects in aging, we examined the influence of maternal age, maternal diet, and offspring diet on offspring lifespan, fecundity, mitochondrial phenotype and response to interventions in the rotifer, Brachionus manjavacas. Rotifers are microscopic, aquatic invertebrates with many advantages as a model system for the biology of aging, including ease of culture, transparency, large maternal investment in offspring, direct development with no larval stage, greater genome homology with humans than in established invertebrate models, sequenced genomes and transcriptomes, and RNAi. We found that lifespan, fecundity, and stress resistance decline with increasing maternal age. The decreased fitness of old-mother offspring is associated with increased mtDNA content, decreased mitochondrial inner membrane folding, changes in ROS, and altered protein carbonylation, suggesting these deleterious maternal age effects may be mediated via inherited mitochondrial dysfunction. Caloric restriction increases lifespan in offspring from all maternal ages, but the magnitude of lifespan extension is greater in old-mother offspring. The trade- off between reproduction and lifespan extension under low food conditions expected by life history theory is observed in young mother offspring, but not in old-mother offspring. Age- related changes in maternal resource allocation to reproduction do not appear to drive changes in offspring fitness or plasticity under caloric restriction in B. manjavacas. Our results suggest that the declines in reproduction in old-mother offspring may negate the evolutionary fitness benefits of lifespan extension under caloric restriction.

P4

The linker histone H1, beyond a structural chromatin component

Shengbo He, Martin Vickers, Jingyi Zhang, and Xiaoqi Feng

Department of Cell and Developmental Biology, John Innes Centre, Norwich, UK

Not only is H1 a linker between two adjacent nucleosomes, but H1 regulates many important biological processes in animals, including transposon expression, DNA replication, cancer cell proliferation, and apoptosis and so on. Despite its importance, little is known about H1 function in plants. Our study shows that H1 expression is dramatically reduced in male gametophyte of Arabidopsis thaliana, which correlates with global CG methylation increase in male germline compared to soma where CG methylation is not perfectly maintained. This CG methylation reprogramming in sex cells, which is probably mediated by H1 downregulation as H1 is shown to inhibit CG methylation in soma, can explain how CG methylation is faithfully inherited across generations. We further found that H1 is specifically expressed in sperm cells in pollen but not in sperm companion cell - vegetative cell (VC). Intriguingly, transposable elements (TEs), which are epigenetically silenced in soma, are activated in VC. However, the extent and mechanism of this activation are unknown. Our work shows that about 100 heterochromatic TEs are activated in VCs, mostly by DEMETER- catalyzed DNA demethylation. We further demonstrate that DEMETER access to some of these TEs is permitted by the natural depletion of linker histone H1 in VCs. Ectopically expressed H1 suppresses TEs in VCs by reducing DNA demethylation and via a methylation-independent mechanism. We demonstrate that H1 is required for heterochromatin condensation in plant cells and show that H1 overexpression creates heterochromatic foci in the VC progenitor cell. Taken together, our results demonstrate that the natural depletion of H1 during male gametogenesis facilitates DEMETER-directed DNA demethylation, heterochromatin relaxation, and TE activation.

P5

Inter and intra-generational effects of insulin/IGF signaling on early-life starvation- induced pathology

Jordan, J.M., Shaul, N.C., Webster, A.K.,Chitrakar, R., Baugh, L.R.

Duke University, Durham, North Carolina, USA

C. elegans provision additional yolk when dietarily restricted or when insulin/IGF signaling (IIS) is reduced. Increased yolk provisioning reduces IIS in developing progeny, buffering them from pathology induced by early-life starvation (Jordan et al., 2019). We addressed the question of how early-life starvation followed by unrestricted feeding compromises developmental fidelity leading to development of germline tumors and how reduction of IIS can improve reproductive outcomes using RNAseq on young adults. RNAseq analysis comparing previously starved to control worms revealed that of the 2447 differentially expressed genes, 1965 (80%) are upregulated. Of these, 887 (45%) are downregulated with IIS knockdown during recovery from starvation. Expression of these genes is associated with development of germline tumors, and they include conserved oncogenes. Members of the Wnt and Hedgehog families are particularly enriched, and knockdown of some of these genes ameliorates starvation-induced abnormalities. Further, gene set enrichment analysis strongly implicates membrane biology in pathogenesis. Lipidomic analysis shows that lipid composition is altered by early-life starvation. Starved animals have more phosphatidylcholine (PC) and its precursor diacylglycerol (DAG) than control animals at egg laying onset. Furthermore, reducing IIS during recovery mitigated the difference in DAG and PC levels following starvation. These results suggest early-life starvation promotes pathology by inducing expression of growth-promoting genes and disrupting membrane homeostasis. Reducing IIS negatively regulates the expression of these genes and restores membrane homeostasis, thus reducing pathological outcomes of early-life starvation.

Jordan, J.M., Hibshman, J.D., Webster, A.K., Kaplan, R.E.W., Leinroth, A., Guzman, R., Maxwell, C.S., Chitrakar, R., Bowman, E.A., Fry, A.L., et al. (2019). Insulin/IGF Signaling and Vitellogenin Provisioning Mediate Intergenerational Adaptation to Nutrient Stress. Curr Biol.

P6

The impact of elevated maternal CORT on F1 sperm quality

Nicola Khan, Kylie Robert

University of St Andrews, La Trobe University, UK

Environmental conditions during development play an important role in an individual's subsequent life history. Corticosterone (CORT), the primary glucocorticoid in birds, maintains homeostasis and energy balance by mediating physiological and behavioural responses to stress events, such as anthropogenic disturbance, elevated predation risk, or limited resource availability. How an individual copes with stress events is a likely determinant of their health and well-being, and maternal stress can significantly affect offspring physiology, behaviour and reproductive performance. One area that has been little studied is the impact of maternal CORT on the quality of their sons' sperm. Only 0.001% of sperm from the ejaculate are able to fertilise ova, and sperm morphology is associated with fertilisation success. For example, both sperm length and sperm mobility have positive relationships with fertilisation success, and smaller spermatozoa survive longer in the female sperm storage tubules than larger sperm.

This study aimed to determine how increased maternal stress during ovulation affects their sons as adults, by comparing sperm morphology between males exposed to maternally- derived CORT (hereafter 'MC') and control groups (hereafter 'unstressed'). Optimal sperm, i.e. the 'fertilising set', for zebra finches have relatively short heads, coupled with longer tails and reduced morphological variability. Interestingly, we found that MC birds showed the exact opposite traits than the fertilising set; sperm from MC males had longer heads, which is associated with decreased fertilisation success, and significantly higher variance in flagellum and total length than unstressed counterparts, indicating poor control of spermatogenesis. This is concerning for at-risk populations, as it shows that a relatively short period of elevated CORT during ovulation can have ongoing consequences for their offspring by potentially impacting their sons' reproductive success as adults.

P7

Effects of predation risk on egg steroid profiles across multiple populations of stickleback

Katie McGhee, Ryan Paitz, John Baker, Susan Foster, Alison Bell

Department of Biology, The University of the South, Sewanee, TN, USA, School of Biological Sciences, Illinois State University, Normal, IL USA, Department of Biology, Clark University, Worcester, MA, USA, School of Integrative Biology, University of Illinois, Urbana-Champaign, Urbana, IL, USA

Although there is evidence for parallel evolution of prey behavior and morphology in response to predation regime, mechanistic traits underlying these phenotypes seldom show similarly strong parallelism. Thus, it is unclear whether the steroid hormones that underlie prey responses to predators and mediate maternal effects will show consistent patterns across populations that vary in predation risk. We investigated how predation risk affects maternally-derived steroids in threespine stickleback eggs across nine Alaskan populations that vary in whether predatory trout are native, stocked, or absent. Using liquid chromatography coupled with mass spectroscopy (LC-MS/MS), we detected 20 steroids within eggs and these tended to cluster together within steroid classes, emphasizing the interconnectedness of the endocrine response. Surprisingly, predator regime did not significantly affect egg steroid profiles. Despite being the most abundant steroid, cortisol was also not affected by predation regime. Predators may indeed affect maternal stress and thus, embryonic exposure to maternal steroids, but this pattern is clearly more complex than a simple one-to-one relationship between predation risk and the steroids that mothers transfer to their eggs. Our results suggest that different multivariate steroid combinations might give rise to equivalent phenotypes, and thus, it might be difficult to detect consistent intergenerational consequences of maternal stress.

P8

Analysing mothers’ milk for factors programming offspring body composition

Kim Moorwood, Valentyna Varenko, Mike Cowley, Sasi Saminathan, Andrew Ward

Department of Biology and Biochemistry, University of Bath, UK

Imprinted Grb10 encodes an adaptor of receptor tyrosine kinases (including the insulin receptor) regulating fetal growth, body proportion and composition. Although widespread during mouse fetal development, Grb10 expression is restricted to specific sites (eg. pancreatic beta cells) in adulthood. Interestingly, one of these sites is the lactating mammary gland in which the maternal allele is activated.

Our mouse cross-fostering studies have shown that Grb10KO females (themselves large and muscular) support reduced offspring growth and fat mass between birth and weaning in comparison to wild-type females. Now, we show that these changes have physiological implications for offspring in later life. Further, we have begun to investigate the co-adaptive mechanism by which the nursing mother's Grb10 gene might program the body composition of the next generation.

Preliminary analytical comparisons of candidate lactocrine signalling factors in milk produced by Grb10KO and wild-type females so far suggest that epidermal growth factor and a set of milk-borne miRNAs are not involved in programming offspring physiology and an unbiased approach might be necessary.

P9

Egg development plasticity and the transmission of maternal effects in Daphnia

Stew Plaistow, Megan Hasoon, Roy Goodacre, Howbeer Muhamadali, Alan Reynolds

University of Liverpool, UK

Environment-induced non-genetic inheritance (NGI) may play a key role in facilitating rapid adaptation to changing environments. However, the mechanisms underpinning NGI remain obscure and we currently know little about how genetic, non-genetic and environmental cues are integrated. The egg phenotype is critical in this respect because it provides many routes for information transfer from one generation to the next and defines the initial environment that inherited genes are exposed to. Parthenogenetic organisms such as Daphnia are ideal models for investigating the integration of developmental cues because large numbers of genetically identical individuals can be reared in different environments in parental and offspring generations, making it easy to separate genetic and non-genetic influences. However, variation in egg phenotypes is typically only ever studied at a simplistic level. Here we use metabolomics to compare the egg phenotypes produced by mothers exposed to different environments, or mothers occupying different states (sexual vs asexual, old vs young). We then use confocal microscopy to compare the embryology of offspring from mothers from different environments, or different states. Our results suggest that (1) changes in the maternal environment or state result in changes in the egg phenotype, and (2) that changes in the egg phenotype results in egg development plasticity that can explain life-history variation in adults. We conclude that offspring provisioning is likely to be a key factor explaining how genetic, non-genetic and environmental cues are integrated.

P10

Epigenomic regulation of germ cell development in fish: a target for environmental stressors?

Paige Robinson, Hannah Littler, Ronny van Aerle, Emma Dempster, Jon Mill, Eduarda Santos

College of Life & Environmental Sciences, University of Exeter, UK International Centre of Excellence for Aquatic Animal Health, Cefas Weymouth Laboratory, Dorset, UK University of Exeter Medical School, University of Exeter, UK

Vertebrate embryos undergo extensive epigenetic reprogramming events during early development, allowing the formation of totipotent cells and avoiding inheritance of many deleterious alterations accrued during the parent's lifetime. We hypothesise that the epigenome of developing embryos is particularly sensitive to environmental stressors during these reprogramming periods, with the potential for epigenetic alterations induced during this period to persist throughout an organism's life and have lasting phenotypic consequences. In mammals, two rounds of reprogramming have been documented - firstly in somatic cells and a second reprogramming window discovered in germ cells. In early development in fish, a window of incomplete reprogramming of somatic cells has also been identified; however, the germ cell epigenome during this period remains unstudied.

The first aim of my PhD is to document the methylome of germ cells across early development in zebrafish to identify sensitive periods of reprogramming, during which maternal effects may potentially cause epigenetic alterations that persist into later life or future generations.

The second aim of my PhD is to explore how exposure to environmental stressors, particularly toxic metals, during these susceptible periods of development may alter the epigenome, and whether these alterations can persist into later life or in future generations in the absence of continued exposure. A particular area of focus in the multigenerational studies will be identifying whether toxicant-induced epigenetic alterations are able to escape reprogramming in order to be transgenerationally inherited from parents to offspring via the germ cells.

In my current work I am screening for the potential of environmental stressors to disrupt the epigenome of zebrafish embryos during the reprogramming period which occurs prior to gastrulation. I am also developing methodologies to isolate germ cells during development, in order to document the dynamics of the transcriptome and epigenome of these cells during germ cell migration and gonadal formation. This information will facilitate further studies to determine how environmental chemicals disrupt the physiology of these cells.

P11

Do prenatal thyroid hormones program energy metabolism and ageing physiology?

Suvi Ruuskanen, Bin-Yan Hsu1, Coline Marciau1, Nina Cossin Sevrin2, Pierre Bize3, Lars Gustafsson4, Antoine Stier1,5

University of Turku, Finland; 2 University of Strasbourg3, University of Aberdeen, UK; 4 Uppsala University, Sweden; 5 University of Glasgow, UK

The effect of early-life environment on ageing rates and energy metabolism has been repeatedly observed in human and non-human vertebrates, but the mechanisms underlying this association have been poorly characterized. Thyroid hormones (THs) are key hormones regulating metabolism and growth, and vertebrates are exposed prenatally to varying levels of THs of maternal origin. We predict that energy metabolism by the cell powerhouses, mitochondria, could be positively influenced by prenatal exposure to THs, whereas hallmarks of ageing (oxidative status and telomere length) could be negatively affected - both of which could lead to long-lasting programming effects on offspring. We experimentally elevated egg THs in wild populations of passerine species widely used in eco-evolutionary studies (collared flycatchers, Ficedula albicollis) and great tits (Parus major) and studied the effects on offspring development (growth rate), ageing (telomere length and shortening) and energy metabolism (mitochondrial density and respiration). As predicted, in great tits THs increased telomere shortening at nestling stage and also later in life, whereas in collared flycatchers, nestlings from TH eggs had longer telomeres than control nestlings at both early and late nestling stage. There was no indication of effects of prenatal THs on mitochondrial density, yet effects on respiration remain still under investigation. Given that individual differences in telomere length in early life tend to be carried until adulthood, effects of prenatal THs on telomeres may have long-lasting effects on senescence.

P12

Fetal androgen excess determines adult male health via hepatic dysfunction and dyslipidaemia

Kasia Siemienowicz, Panagiotis Filis3, Sophie Shaw3, Alex Douglas3, Jennifer Thomas1, Forbes Howie2, Paul Fowler3, Colin Duncan2 and Mick Rae1

1 School of Applied Sciences, Edinburgh Napier University, UK; 2 MRC Centre for Reproductive Health, University of Edinburgh, UK; 3Institute of Medical Sciences, University of Aberdeen, UK

Altered intrauterine environments can 'programme' sub-optimal postnatal health. Clinical linkage to excess androgen exposure in utero includes polycystic ovary syndrome (PCOS) and congenital adrenal hyperplasia, but perhaps also nutritional restriction, since nutrition alters fetal testosterone secretion. Studies on male offspring from androgen excess pregnancies are scarce, however adult males from PCOS pregnancies develop dyslipidaemia with elevated triglycerides (TG), increased total and LDL-cholesterol (LDL-C), and hyperinsulinaemia.

We hypothesised that poorer adult health noted in first degree male relatives of PCOS patients was partially attributable to excess androgen exposure during development.

Testosterone was directly administered to developing male ovine fetuses on day62 and 82 of gestation (term is day147). Such in utero androgen excess recreated the dyslipidaemia and hormonal profile (elevated insulin and AMH) observed in sons of women with PCOS. After applying RNAseq and proteomics to liver biopsies from control (n=14) and testosterone (n=14; prenatal androgen group (PA), 15,134 and 2,766 genes and proteins respectively were quantified. 1,084 genes and 408 proteins were altered in male offspring liver during adolescence, attributable to fetal androgen excess. This altered liver function predisposed to adolescent development of an intrahepatic cholestasis-like condition, with attendant hepatic bilirubin accumulation and increased circulating bilirubin. An emergent pro-fibrotic, pro- oxidative stress gene and protein expression profile was evident in liver and circulation, after application of plasma proteomics to adolescent offspring blood samples. analysis confirmed this emerging disease state, and further, indicated overall increased chronic disease risk, including atherosclerosis, associated with prenatal androgen excess in males. Our data demonstrate aetiological mechanisms of such prenatally programmed hepatic dysfunction, strongly paralleling human cholestasis development, whilst also suggesting likely inefficacy of standard treatment regimens to ameliorate the dyslipidaemic profile observed. We conclude that excess prenatal androgen exposure is a previously unrecognised determinant of lifelong male health.

Supported by MRC Project Grants G0901807 (MTR / WCD) and MR/P011535/1 (MTR / PAF / WCD)

P13

Micronutrient supplementation and the effect on hepatic gene expression and DNA methylation in Atlantic salmon

Kaja Helvik Skjaerven, Takaya Saito, Paul Whatmore, Anne-Catrin Adam, Marit Espe

Institute of Marine Research, Norway

Nutritional micronutrient status throughout life is a key environmental variable to target through feed in order to permanently increase growth performance and quality. Especially, during periods of rapid growth, vertebrates show great developmental plasticity regulated by nutritional stimuli. We have earlier described that the zebrafish parental micronutrient status alters the embryonic gene expression of immune-, lipid and apolipoprotein genes already at the sensitive and conserved embryonic stages of development. Nutritional stores and/or maternal mRNA transcripts deposited in the yolk during oogenesis also influenced epigenetic DNA methylation, gene transcription and liver lipid accumulation to a fatty-liver- like phenotype in mature zebrafish offspring. Hence, in salmon aquaculture, attention towards the micronutrient composition in feed raw material increased after the inclusion changed from marine to plant based raw material. When feeding plant-based ingredients the micronutrient requirement for especially b-vitamins increased to avoid a fatty liver like phenotype. B-vitamins are especially interesting in terms of epigenetic regulation of gene expression as B-vitamins like folate, vitamin B12 and B6 and the amino acid methionine are important for the regulation of the methylgroups used for DNA methylation. As such, we hypothesised that the gene expression in the liver were regulated differently due to the b- vitamin inclusion in feed. We found specifically downregulation of genes encoding transcripts important for cholesterol synthesis including the Acetyl-CoA carboxylase 1 encoded by the acaca gene between the lowest and highest supplemented diet. DNA methylation patterns showed specifically multiple differentially methylated (DM) CpGs found in the acaca gene promoter region. These DM CpGs are hyper-methylated while the mRNA expression is downregulated in supplemented fish. We believe those datasets should be useful for further studies to compare with other treatments and tissue types, and also understand the effect of micronutrients at the molecular level for Atlantic Salmon and other vertebrates.

P14

Intergenerational inheritance through pheromone detection and a stress response pathway in the nematode A. freiburgensis

Pedro Robles Naharro, Anisa Turner, Giusy Zuco, Andre Pires da Silva

University of Warwick, UK

Auanema freiburgensis is a free-living nematode that can be used as a model system to study mechanisms of intergenerational change. Hermaphrodite mothers of A. freiburgensis sense environmental cues for high population densities and respond by shifting from the default production of female and male offspring, under low density conditions, to the production of hermaphrodites and males. These mothers are therefore receiving a signal from the environment that is transferred to the germline in order to adaptively alter the phenotype of their offspring. Resources can be expected to be low for the offspring in the immediate environment, but hermaphrodites go through an obligate stage of arrested development ('dauer'), enabling them to survive 'predicted' difficult conditions for prolonged periods and migrate to new habitats. It is important to discover the processes involved in adaptive intergenerational responses, from sensing of the environment to resultant F1 phenotypes, in order to understand how such systems can evolve.

Ascarosides (pheromones released by many nematodes) appear to be sensed by hermaphrodite mothers in order to influence the sex of their offspring. The most common ascaroside found to be present in cultures of high-density A. freiburgensis populations is ascaroside #18, which therefore appears to be a good candidate for the environmental cue responsible for intergenerational change in this species. In our experiments, hermaphrodites were exposed to this ascaroside by placing single individuals on NGM plates treated with ascaroside #18 for 24 hours before collecting eggs for three days. The progeny was allowed to develop to adulthood as isolated individuals which were then sexed to find the ratio of hermaphrodites to females. We found that exposure to ascaroside #18 results in the production of a high number of hermaphrodites in low-density conditions. However, other ascarosides and secreted chemicals may also be involved in the intergenerational response and further testing will help uncover the precise nature of the environmental signal. Furthermore, pharmacological manipulation of stress-response signalling pathways can also alter F1 sex ratios. Pathways that are known to regulate the conserved Nrf2/SKN-1 transcription factor, which mediates various cellular stress responses, have been tested by exposure to pharmacological compounds. The results show an elevation in the production of hermaphrodites upon activation of Nrf2/SKN-1, as well as immunostaining showing its activation in intestinal cells upon exposure to 'crowding' cues. Our data suggest a role, in A. freiburgensis, for stress response regulation in bringing about the intergenerational plastic response to the pheromone signal.

P15

Transgenerational effects of extended dauer diapause on starvation survival and gene expression plasticity in C. elegans

Amy K. Webster, James M. Jordan, Jonathan D. Hibshman, Rojin Chitrakar, L. Ryan Baugh

Duke University, Biology Department, USA

Phenotypic plasticity is facilitated by epigenetic regulation, and remnants of such regulation may persist after plasticity-inducing cues are gone. However, the relationship between plasticity and transgenerational epigenetic memory is not understood. Dauer diapause in Caenorhabditis elegans provides an opportunity to determine how a plastic response to the early-life environment affects traits later in life and in subsequent generations. We report that, after extended diapause, postdauer worms initially exhibit reduced reproductive success and greater interindividual variation. In contrast, F3 progeny of postdauers display increased starvation resistance and lifespan, revealing potentially adaptive transgenerational effects. Transgenerational effects are dependent on the duration of diapause, indicating an effect of extended starvation. In agreement, RNA-seq demonstrates a transgenerational effect on nutrient-responsive genes. Further, postdauer F3 progeny exhibit reduced gene expression plasticity, suggesting a trade-off between plasticity and epigenetic memory. This work reveals complex effects of nutrient stress over different time scales in an animal that evolved to thrive in feast and famine.

P16

Understanding inherited immunity using a C. elegans model of microsporidia infection

Alexandra Willis, Winnie Zhao, Aaron Reinke

University of Toronto, Canada

It has emerged that some animals can pass on memory of previous infection to their offspring, thereby enhancing immunity their progeny. Inherited immunity is an example of epigenetic inheritance, whereby the parental environment modulates gene expression in the germline. Though inherited immunity can provide resistance to bacterial, viral and fungal pathogens, the molecular mechanisms underlying protection are unknown.

Studies in the simple and genetically tractable nematode C. elegans have enabled key advances in both immunity and epigenetics. Microsporidia are poorly-characterised fungal pathogens, and Nematocida parisii is a microsporidial parasite of C. elegans. New data shows that the progeny of N. parisii-infected worms exhibit robust immunity against microsporidia. Though microsporidia replicate in the intestinal cells of naïve larvae, parasitism is not seen in our immune-primed larvae.

Using the N. parisii-C. elegans infection model, we are now characterising inherited immunity in vivo. In particular, we are assaying the strength and duration of resistance to microsporidia in immune-primed worms, using studies of parasite invasion to reveal mechanisms of protection, and performing gene expression analyses to reveal putative immune effectors. We are also testing the tolerance of our primed worms to different environmental stresses, and examining the role of epigenetic factors in the transmission of immunity from parent to progeny.

Our study offers unique insight into the immune reactivity of the genome. Epigenetic factors mediating inherited immunity in C. elegans may have homologues in higher organisms; this would have major implications for health and evolution. Whilst microsporidia are important opportunistic pathogens of humans and parasitize almost all animal species, very little is known of their infection biology. Study of this medically and environmentally relevant pathogen will suggest new ways to treat infection.

P17

Assessing coral stress responses to environmental changes using microarray

Seonock Woo, Seungshic Yum

Korea Institute of Ocean Science and Technology, South Korea

The Kuroshio is a warm current running from tropical Philippines through subtropical Taiwan to the temperate region of Japan and Korea and it transfers heat from lower to higher latitudes. It warmed most rapidly in 1981-1998, when sea surface temperatures rose by 1.5°C (0.9°C/decade), almost 7 times the global rate. The reef building corals in the region have rapidly expanded their range northward in response to warming temperatures. Furthermore, coral reefs of Taiwan and Japan were closely linked by the Kuroshio Current and inevitably, the coral population and its distribution in Korea could be affected by warming currents, similar to the situation in Japan and Taiwan. In this study, we chose Eleutherobia rubra which is a azooxanthellate soft coral around Korean peninsula to investigate its gene expression change against the environmental stress and constructed microarray. The probes in microarray was based on the noble and known genes sequences from NGS technology. Approximately 75,000 genes from E. rubra were used and we carried out heat stress experiment using microarray.

This study focused on the identification of functional genes in coral to assess the molecular level responses to environmental stress like as seawater temperature change. Climate change and rising seawater temperature are placing spatially divergent stresses on the world's coral ecosystem through increasing ocean surface temperatures and ocean acidification. To assess the coral health condition as well as coral stress responses, we purposed to identify molecular level stress responses using gene expression change as well as functional genes responding to external stress like increased seawater temperature. Approximately 200 genes including Kinesin-like protein, guanine-nucleotide releasing factor, caspase, apoptotic cysteine protease, etc changed their gene expressions after heat shock stress. These isolated genes were clustered into functional groups like information storage and processing, cellular processes and signaling and metabolism. Over 70 % of isolated genes were related to the signal transduction.

Acknowledgments This study was supported by NRF-2017R1A2B2012541

P18

Notes

P19

Notes

P20

Notes

P21

Speaker and Delegate List

Tracy Bale Sinead English University of Maryland University of Bristol [email protected] [email protected]

Ryan Baugh Xiaoqi Feng Duke University John Innes Centre [email protected] [email protected]

Fabian Braukmann Anne FergusonSmith , University of Cambridge University of Cambridge [email protected] [email protected]

Jenn Brisson Panagiotis Filis University of Rochester University of Aberdeen [email protected] [email protected]

Paula Brunton Benjamin Fisher University of Edinburgh University of Cambridge [email protected] [email protected]

Nick Burton Eamon Fitzgerald University of Cambridge University of Edinburgh [email protected] [email protected]

Graham Burton Paul Fowler University of Cambridge University of Aberdeen [email protected] [email protected]

Tim CluttonBrock Mirko Francesconi University of Cambridge Ecole Normale Superieure de Lyon [email protected] [email protected]

Sofia Consuegra Giulia Furlan Swansea university Gurdon Institute [email protected] [email protected]

Alexandra Dallaire Antonio Galvao Wellcome Trust/Cancer Research UK Babraham Institute Gurdon Institute [email protected] [email protected] George Gericke Troy Day Ampath National Pathology Queen's University [email protected] [email protected] Dino Giussani Kathleen Donohue University of Cambridge Duke University [email protected] [email protected]

Kristin Gribble Heidi Lempradl Marine Biological Laboratory Van Andel Institute [email protected] [email protected]

Jamie Hackett Kirsty MacLeod EMBL Lund University [email protected] [email protected]

Shengbo He Agnieszka Magierecka John Innes Centre University of Glasgow [email protected] [email protected]

Jonathan Hirst Toby Mansell University of Newcastle Murdoch Children's Research Institute [email protected] [email protected]

Kahyee Hor Rahia Mashoodh University of Edinburgh University of Cambridge [email protected] [email protected]

Rosalind John Katie McGhee Cardiff University Sewanee: The University of the South [email protected] [email protected]

James Jordan Eric Miska Duke University University of Cambridge [email protected] [email protected]

Leea KeskiNisula Pat Monaghan Kuopio University Hospital Professor [email protected] [email protected]

Nicola Khan Kim Moorwood University of St Andrews University of Bath [email protected] [email protected]

Bram Kuijper Sue Ozanne University of Exeter University of Cambridge [email protected] [email protected]

Lisa Lampersberger Christian Page The Gurdon Institute Norwegian Institute of Public Health [email protected] [email protected]

Cristina Ledón Rettig Andre Pires Indiana University University of Warwick [email protected] [email protected]

Stewart Plaistow Kasia Siemienowicz University of Liverpool Edinburgh Napier University [email protected] [email protected]

Harry Potter Kaja Helvik Skjaerven University of Manchester Institute of Marine Research [email protected] [email protected]

Ahva Potticary Karen Spencer University of Arizona University of St Andrews [email protected] [email protected]

Jonathan Price Kent Thornburg University of Cambridge Oregon Health & Science University [email protected] [email protected]

Audrey Putman Jurriaan Ton University of Cambridge University of Sheffield [email protected] [email protected]

Mick Rae Anisa Turner Edinburgh Napier University University of Warwick [email protected] [email protected]

Lisa Rhuman Tobias Uller Oregon Health & Science Univ Lund University [email protected] [email protected]

Paige Robinson Tamsyn Uren Webster University of Exeter Swansea University [email protected] [email protected]

Tessa Roseboom Silvia Vogl Amsterdam UMC German Federal Institute for Risk Assessment [email protected] [email protected]

Suvi Ruuskanen Andrew Ward University of Turku University of Bath [email protected] [email protected]

Gilbert Schoenfelder Amy Webster German Federal Institute for Risk Duke University Assessement (BfR) [email protected] [email protected] Priscilla Wehi Gemma Sharp Manaaki Whenua Landcare Research University of Bristol [email protected] [email protected]

Alexandra Willis University of Toronto [email protected]

Seonock Woo Korea Institute of Ocean Science and Technology [email protected]

Index

Bale, T S33 Robinson, P P11 Baugh, R S13 Roseboom, T S49 Brunton, P P1 Ruuskanen, S P12

Consuegra, S S29 Siemienowicz, K P13 Skjaerven, K H P14 Day, T S7 Spencer, K S27 Donohue, K S21 Thornburg, K S57 English, S S47 Ton, J S19 Turner, A P15 Feng, X S23 Ferguson-Smith, A S43 Uller, T S3 Fitzgerald, E P2 Francesconi, M P3 Webster, A P16 Willis, A P17 Galvao, A S45 Woo, S P18 Giussani, D S51 Gribble, K P4

He, S P5 Hirst, J S37 Hor, K S55

John, R S35 Jordan, J P6

Khan, N P7 Kuijper, B S5

Ledón-Rettig, C S11 Lempradl, H S15

MacLeod, K S25 Magierecka, A S31 Mansell, T S53 McGhee, K P8 Monaghan, P S1 Moorwood, K P9

Ozanne, S S41

Pires, A S17 Plaistow, S P10 Potter, H S39 Potticary, A S9   @ACSCevents  Wellcome Genome Campus Courses and Conferences wellcomegenomecampus.org /coursesandconferences