The community site for and by
developmental and stem cell biologists

The people behind the papers – Lisa Conrad, Steve Runser, Roman Vetter and Dagmar Iber

Posted by , on 26 July 2021

This interview, the 97th in our series, was published in Development earlier this year

Epithelial tubes perform crucial functions in various organs, providing routes for the transport of fluids and gases. A new paper in Development addresses the question of how epithelial tubes elongate during development, using a combination of mouse organ culture and mathematical modelling. To find out more about the work, we met four of its authors: PhD students Lisa Conrad and Steven Runser, senior scientist Roman Vetter, and their supervisor Dagmar Iber, Professor in the Department of Biosystems Science and Engineering at ETH Zurich.

Lisa (top left), Steve (top right), Roman (bottom left) and Dagmar (bottom right).

Dagmar, can you give us your scientific biography and the questions your lab is trying to answer?

DI: I studied mathematics and biology, and did Masters degrees and PhDs in Cambridge and Oxford in each field. Since the very start, I have been interested in using mathematical modelling to uncover biological mechanisms. A Junior Research Fellowship at St. John’s College in Oxford gave me the freedom to pursue my own ideas and develop more precise, data-based models than the established conceptual models. Initially, I worked in immunology, but then switched to cell differentiation in bacteria, as the required quantitative data to build and validate models was available only for such simple organisms. These days, my group focuses on developmental mechanisms all the way up to human, and although we mostly collaborate with experimental groups, ETH allows me to also run a small wet lab to generate data and test ideas. The lab largely focuses on mouse lung and kidney development, but we maintain a rather broad interest in fundamental patterning mechanisms, and also collaborate with clinicians.

Roman, can you tell us a little about your research history and how you ended up working on developmental questions?

RV: I’m a computational physicist by training, and did my studies and PhD at ETH Zurich. With a few exceptions, my interest has always been in explaining nature’s wealth of emergent complex behaviour from simple, fundamental principles. During the earlier stages of my academic career, I found these problems mainly in the physical disciplines – from snow metamorphism to filament packing to crumpled shells and even fuel cell simulations. If you go through life with an open mind, open eyes and open ears, there’s an interesting question calling for a quantitative explanation virtually everywhere. My attention is drawn easiest by questions that combine geometrical shapes with mechanics and patterning. Lately, I have found such inspiration more and more in biology, where fundamental aspects of morphogenesis and development require taking new vantage points to advance further. I joined Dagmar’s group 2 years ago when she was looking for a senior biophysics modeller and I was looking to enter the field of computational biology to find new puzzles to solve – it was an instant match. I started as a postdoctoral researcher and recently became a senior scientist and lecturer in her group.

Lisa and Steve – how did you come to work in Dagmar’s lab and what drives your research today?

LC: I studied biology at the University of Freiburg and molecular medicine at Uppsala University. Developmental biology has fascinated me from the beginning; besides the interesting questions and methods in this field, the beauty of development is just so captivating! During a variety of lab projects, I noticed how much I enjoy projects that bring together expertise from different scientific backgrounds. By applying to the Life Science Zurich Graduate School, I found out about Dagmar’s group and got curious about their multidisciplinary approach to developmental biology. I started as the first PhD student with an experimental focus in the group’s then recently opened wet lab, eager to build on my experimental and research skills, while learning about new ways on how to tackle developmental questions from a different angle. It has been challenging to keep up with the aspects of the group’s research that are far from what I studied, but it’s also immensely rewarding when we can join forces to find new ways to better understand organ morphogenesis!

SR: I started my studies in cellular and molecular biology at the University of Strasbourg, but I quickly deviated towards more computational fields. I have always been interested in the design and application of mechanical simulations for the study of biological systems. To respond to this interest and to learn more about numerical approaches, I applied to Dagmar’s lab to do my master’s thesis. After completion of the thesis a little less than 2 years ago, Dagmar offered me the opportunity to continue in this field as a doctoral student. Since then, I have been developing and using different types of simulation models to study organ growth and morphogenesis.

How did you come to study tube elongation?

DI: The group had long been interested in lung and kidney branching morphogenesis. Although our ligand-receptor-based Turing mechanism could nicely predict where new branch points would form during lung and kidney branching morphogenesis, we noticed that the branch shapes that emerged in our simulations looked nothing like in the embryo because we were missing that bias in outgrowth that lets epithelial tubes of embryonic lungs and kidneys lengthen more than they widen.

Before this project, what mechanisms had been proposed for biased tube elongation?

DI: In lungs and kidneys, chemotactic movement towards a source of FGF10 or GDNF had long been noticed, and the extracellular matrix is thinner at the bud tips. In mammary glands, a constricting force had been proposed. In plants, hoop stress is a popular theory to explain their biased growth. We tested all those ideas, but none could explain the biased outgrowth of embryonic lung or kidney tubules. In fact, when checking the potential of hoop stress, we noticed that the lumen of the tubes is mostly very narrow, whereas the wall, the epithelium, is comparably thick. Although this is inconsistent with a role of hoop stress, it gave us the idea that fluid flow-induced shear stress may play a role.

Fluid flow (arrows) from tip to base in the lumen (green) of developing lungs causes shear stress levels strong enough to be sensed by epithelial cells (magenta), giving them a direction in which to preferentially grow. This discovery provides a new explanation for the stereotypical directional bias in tube outgrowth observed during the development of branched organs such as lungs and kidneys.

Can you give us the key results of the paper in a paragraph?

SR: The key result of the paper is that the observed bias in epithelial tube outgrowth, the accompanying bias in the apical cell shape and the resulting biased orientation of cell divisions, can be explained with fluid-flow driven shear stress. After having ruled out all the previously proposed mechanisms, Roman used a Finite Element method to demonstrate that the collapse of the tubes in itself was very unlikely to result in a bias in outgrowth. Instead, the narrow luminal region meant that a fluid flow in the epithelial tubes might cause a significant level of shear stress on the cell walls. To prove the existence of a flow, Lisa injected micro-beads in the lung lumens and observed their movements over time. We simulated the effect that a flow with the measured velocity would have on the cell walls of a similar lung tube geometry. The shear stress levels thus calculated were well within the range of what epithelial cells can sense. Shear stress is well known to result in the elongation of endothelial cells along the flow direction in blood vessels. I used a cell-based vertex model to investigate the impact of such an elongation on epithelial tissues. Once parameterized based on quantitative data, the model was able to recapitulate all the measured features of the lung and kidney tube epithelium. The bias in cell division orientations was well in line with what had been measured, and similarly the bias in outgrowth of the tissue matched with the experimental observations.

How do you think lung and kidney cells sense shear stress, and how is this sensing translated into biased growth at the tissue level?

DI: Epithelial cells can sense shear stress with their cilium. How they translate this into a change in cell shape, and how the extent of the cell shape change relates to the shear stress level is not known. Microfluidic experiments may help to resolve this.

Do you have any ideas for what causes tube collapse in early development?

RV: Indeed we do have some ideas, and we’re working toward testing possible theoretical explanations with detailed computer simulations by Steve, and toward validating them with experimental data from our wet lab, together with Lisa and other group members. Tube shape and collapse is an exciting topic of ongoing research in our group, and we’re looking forward to telling you more once we have conclusive answers.

When doing the research, did you have any particular result or eureka moment that has stuck with you?

LC, SR: The project really picked up momentum when Harold Gómez, who is also shared first author, noticed, through careful examination of his beautiful lightsheet microscopy data, that the lumen is very narrow in many parts of the lung. But the most exciting moment was when we simulated the shear stress produced by the fluid flow, which we had finally succeeded in measuring experimentally, and realized that it was within the range cells can sense.

The most exciting moment was when we simulated the shear stress produced by the fluid flow.

And what about the flipside: any moments of frustration or despair?

DI: For me, certainly the first response by the referees. For years, I had asked experimental colleagues how to measure fluid flow in lung or kidney tubules and discussed strategies with my team. Yet, no one in my group felt they could pull this off. So, I decided to send the paper to Development, hoping that it would inspire some experimental lab to do that last experiment. After all, through a combination of (simple) experiments and simulations (that encompassed many different sophisticated techniques) we had excluded all previous proposals, suggested a new one, and provided convincing evidence for it. Yet, the referees would not have it. They not only insisted on that last experiment, but also saw little value in the paper as it was. I have seen it more than once that junior members got driven away from science by referee reports that had failed to recognise the value of their work. But not so Lisa: realising that those fluid-flow measurements were the make-or-break, she decided to just give it a try – despite all COVID restrictions. She remembered that Renato Paro had left an injector to our department when retiring, which no one used. She got it to work with the help of his former technician – and demonstrated fluid flow at the required level in the developing lungs.

LC: For me, the biggest source of frustration is when an experiment fails due to technical issues and the tissue samples that I harvested from an animal go to waste. Regarding the referee reports, Dagmar encouraged us to take on the challenge, and celebrating the small successes along the way kept spirits up! In the end, confirming our proposed mechanism was a really good experience.

How has your research been affected by the COVID-19 pandemic?

LC: Last year in March, ETH went into lockdown to help slow down the spread of the virus and our lab was closed for about 6 weeks. The reopening was done in several phases, so we had to work in shifts for a while and it took some time to start up experiments again. Working in shared facilities is still restricted, but most of the time we can figure that out by communicating with colleagues and working around each other’s schedules. In general, everything needs a bit more planning now. For the paper revisions, we had to set up a new experiment in the lab while COVID-19 restrictions were still in place, which of course provided an extra challenge. Having a colleague take a look at your (failed) experiment usually speeds up optimization and trouble-shooting and there are fewer opportunities for spontaneous exchange with other groups. Despite the additional work and stress brought by the pandemic, I feel like everyone at BSSE is supporting each other; especially Makiko Seimiya and Tom Lummen (BSSE Single Cell Facility), who have been awesome in sharing their advice and equipment, and helping me with many tries at the spinning disk confocal, which was a new microscopy system for me.

DI, RV, SR: As theoreticians, we have been forced into a home office for more than a year now. We miss the personal interactions, but remote work is otherwise straightforward for us.

What next for you two after this paper?

LC: I am currently finishing up a project that compares branching morphogenesis in lungs and kidneys. In a collaboration with Roman and other team members, we’re also looking at tube formation, where I’m using nephrogenesis in kidney organoids as a model.

SR: The 2D vertex model used in this paper offers many possibilities for the study of tightly packed cell sheets. However, numerous developmental events can only correctly be represented in three dimensions. With the advent of high performance computing, new computational frameworks representing cells more realistically in 3D can now be developed. I am currently developing such a model specifically designed for the simulation of epithelial tissues.

Where will this story take the Iber lab?

DI: The mechanism that defines the aspect ratio of tubes is one important puzzle piece to explain how complex organs are shaped and how organ-specific differences arise. There are many other fundamental questions concerning tubulogenesis, epithelial organisation, the physics of budding, the role of the mesenchyme and the final reorganisation of the lung epithelial tree into a fractal-like architecture. As a group, we are interested more generally in self-organisation during development.

Finally, let’s move outside the lab – what do you like to do in your spare time in Basel?

LC: I picked up diving a few years back and it’s so much fun! For a local spot, I would recommend the Bodensee; it’s really amazing to dive into a whole new world ‘at home’, now that travelling hasn’t really been an option. Basel has lots of relaxing greenery and I often go for walks along the Rhine. Since the start of the pandemic, I have also rediscovered the fun in crafting and caring for plants on my balcony.

SR: I enjoy a lot of different activities, which range from reading historical novels to playing football.

RV: Spare time comes and goes in phases. There are more things I’d enjoy doing than I could possibly fit into a full day – cycling across the country is one of many.

DI: I enjoy the Swiss mountains, swimming in the Rhine, the tennis court in front of my house – and the preparation for SoLa, the yearly running relay race, which we participate in with the entire group.

Thumbs up (No Ratings Yet)
Loading...

Tags:
Categories: Interview

Navigate the archive

Use our Advanced Search tool to search and filter posts by date, category, tags and authors.

The Sustainable Conferencing Initiative

Posted by , on 23 July 2021

Hello all! I am Viktoria, the Sustainable Conferencing Officer of The Company of Biologists. I am here to quickly answer some questions about our Sustainable Conferencing Initiative. 

What is the aim of the Initiative? 

As biologists become increasingly aware of their environmental and social impact, questions about the environmental sustainability and social responsibility of our events have become more prominent. The Company of Biologists launched the Sustainable Conferencing Initiative in October 2020 to provide guidance and support on the sustainability of events. Furthermore, acknowledging the changes that the pandemic has brought on virtual communications, we are also here to offer insights into technologies that can be used for virtual and hybrid events. Our main aim is to facilitate the discussion of sustainability issues and innovative technologies in our community. 

How will you share information? 

We have our own website! You can visit sustainability.biologists and browse through our different pages. There is a Blog page where our team publishes pieces of information based on the questions coming from our community, and a Resources page that lists useful links regarding sustainability and innovative technologies. Also, in June our Forum became live. Its purpose is to offer a space to our community where ideas, best practices and questions can be shared, and we can all learn from and help each other. 

Is there another form of support available? 

Sustainable Conferencing Grants are available to fund innovative ideas that enable biologists to collaborate productively while minimising their impact on the environment. We accept applications from organisers of meetings, workshops, conferences, seminars, training events, and a wide range of activities in the fields covered by our journals. The events can be in-person, virtual or hybrid. Moreover, applicants for Scientific Meeting Grants may be awarded an additional £1,000 if they can demonstrate efforts have been made to reduce the environmental footprint of their event. 

How can anyone join the discussion? 

Visit our site to explore the information we share. If you register for an account, you will automatically gain access to our Forum where you can ask questions, offer advice, and engage with the community on sustainability and new technologies issues. You can also join our mailing list to receive our newsletters (we promise no spam emails). 

Furthermore, you can find us on Twitter as @COB_Sustainable  Come join us there and share ideas or questions by using our hashtag #Sustainable_Conferencing. Of course, you can always contact us via email at sustainability@biologists.com

We are looking forward to connecting with our community and starting the discussion! 

Thumbs up (No Ratings Yet)
Loading...

Categories: Uncategorized

An interview with Zhiyi Lv

Posted by , on 22 July 2021

Over on The Company of Biologists WeChat channel, we’re enjoying getting to know the growing community. We recently published an interview with Dr Zhiyi Lv, a member of Professor Bo Dong’s group at Ocean University China in Qingdao, China. The Dong lab was established in 2014 and is interested in uncovering the cellular, mechanical and biochemical signalling networks that interact to drive the diverse morphogenetic processes during organ formation and tissue regression using marine ascidians and flies as models. You can find out more about Bo’s research in a ‘The people behind the papers’ interview published in Development last year and a recent Development presents… talk by Hongzhe Peng, a doctoral student in the lab.

Having gained his undergraduate and Master degrees at Northwestern A&F University in China, Dr Lv moved to Germany to obtain his PhD at the University of Göttingen. He remained there for his first postdoc position before relocating back to China to join Professor Bo Dong’s lab at Ocean University of China. Here, he tells us more about his scientific journey, including why he finds the dev bio field amazing and how labs differ between Europe and China.

When did you first become interested in science?

I don’t think there was a specific timepoint that where I thought, ‘aha, now I am interested in science!’ All kids are curious about the unknown world and they are always trying to explore the surroundings. In this way, scientists and children have a lot in common. Fortunately, I did not lose this curiosity as I grew up.  

What attracted you to the field of developmental biology?

How a simple-structured (relatively) fertilised egg becomes a complex adult with head and legs attracted so many people since Aristotle’s time. Then, people realised that genes controlled the development. Now, increasing evidence suggests that mechanical forces contribute morphogenesis actively. It is amazing, isn’t it?

You gained your PhD at the University of Göttingen in Germany and stayed in the same lab for your postdoc but switched from biochemistry to biophysics of morphogenesis – can you tell us more?

I got my PhD under the supervision of Professor Grosshans. I worked on the regulatory mechanism of actin polymerisation. At that time, we identified that a F-BAR protein, Cip4, inhibits actin polymerisation by inactivating Diaphanous, which is an actin nucleator. We got very exciting data, which was published in Journal of Cell Science. Biophysics of embryogenesis has been an important topic in the Grosshans lab. I was impressed by my biophysical colleagues’ talks during our seminars. Professor Grosshans was very nice and always encouraged us to explore a new area. Two projects were running in the lab. One was mechano-transduction at cell-cell contact, and the other one was nuclear array self-organisation in a Drosophila syncytial embryo. I chose the second one for my postdoctoral project.

What are the differences in the lab between Europe and China?

The biggest difference is that experienced postdoc researchers are the main power in the biological labs in Europe. However, most bench work is done by the master and doctoral students. In this case, we need more time and effort to train the students.

Also, some labs in Europe are quite small – one PI tends to lead several postdocs and PhD students, although there are also big labs in Germany. In China, most labs, especially productive labs, are large!

How did you come to work in Professor Dong’s lab?

I met Professor Dong when we were in an EMBO symposium in Heidelberg, Germany in 2018. I was attracted by his work and also by his personality. We share similar scientific interests, and he asked whether I was willing to join to his group. Why not? It was a spontaneous decision.

You have been back in China for a couple of years now – what was it like coming home?

I have experienced ‘reverse culture shock’! For example, when we go to dinner with friends or colleagues, we do not split the bill in China. The leader or the senior person pays for all. As time passes, I will get used to Chinese culture again.

What question is your research currently trying to answer? The origin and the regulation of forces driving morphogenesis, and the crosstalk between genetic cascade and mechanical forces.

What are the main advantages and drawbacks of the model systems you work with, Drosophila and Ciona?

Some students in the lab often misunderstand that Drosophila is a user-friendly model compared to Ciona. The reason behind this might be that Drosophila is easier for genetic manipulation. But in my opinion, this is totally wrong – CRIPSR/Cas9 can also generate the mutant we want in Ciona. I think the advantage of Drosophila is that you can keep the stocks in the lab and you can do experiments whenever you want. The drawback of this model is that you have to take care the animals frequently. We need to collect Ciona from sea. So, the material is limited during early spring and late winter. We need to set up the inbred line in the lab. This is what we are currently doing. Ciona embryos and its larvae are smaller than Drosophila, which is a big advantage for imaging.

What is next for you?

I plan to focus more on Ciona embryogenesis research, and hope to involve myself in the Ciona community more actively.

Follow us on WeChat for exclusive interviews and research highlights written by the community, as well as useful resources to help navigate the publishing process.


Find out more about the Company’s efforts to engage with Chinese researchers

Thumbs up (No Ratings Yet)
Loading...

Categories: Interview

The people behind the papers – Vincent Mouilleau, Célia Vaslin and Stéphane Nedelec

Posted by , on 19 July 2021

This interview, the 96th in our series, was published in Development earlier this year

As the vertebrate body axis extends, HOX genes are sequentially activated in axial progenitors to specify their identity. A new paper in Development addresses what regulates the tempo of this HOX expression in human progenitors. To hear more about the story, we caught up with the paper’s two first authors, Vincent Mouilleau and Célia Vaslin, and their supervisor Stéphane Nedelec, Group Leader at the Institut du Fer à Moulin in Paris.

Vincent, Célia and Stéphane (L to R)

Stéphane, can you give us your scientific biography and the questions your lab is trying to answer?

SN: I studied Biology in Rennes and then Paris, where I did my PhD in the Department of Biology of the Ecole Normale Supérieure. I had the chance to work in the group of Alain Prochiantz under the supervision of Alain Trembleau, where I studied local protein synthesis in neurons. To follow up on this research I then joined Hynek Wichterle, who had recently started his lab at Columbia University. Hynek is a pioneer of in vitro differentiation of pluripotent stem cells (PSCs) to study development and diseases. It was very exciting to work in this environment in the early days of the field with great colleagues and so many things to explore, and this time at Columbia had a profound impact on my scientific career since I was still working on spinal cord development using in vitro approaches. I then moved back to France, working again in a very stimulating environment with Cécile Martinat at the I-STEM institute in Evry. There, I started projects aiming at studying human developmental biology using human PSCs (hPSCs). We developed a powerful approach to assess how extrinsic cues control cell fate and discovered pathways sufficient to convert hPSCs into distinct neuronal subtypes, including spinal motor neurons (MNs). Building on this work, I started a new lab in Paris, at the Institut du Fer à Moulin: a very dynamic and collaborative Neuroscience institute. The current story was largely developed there in collaboration with both the Hynek and Cécile groups, which was very satisfying.

We currently use spinal cord development as a model system to address two interrelated questions. First, the mechanisms by which a limited number of extrinsic factors control human spinal neuronal diversity and morphogenesis – in vitro differentiation of hPSCs is a unique model to approach this question. Second, the mechanisms by which mutations in ubiquitous genes perturb developmental programs to impair selective neuronal populations and cause MN diseases – here we take advantage of developmental studies to improve cell and tissue engineering.

Vincent and Célia – how did you come to work in Stéphane’s lab and what drives your research today?

VM: During my bachelor’s degree in Nantes I became fascinated by stem cells and neurodevelopment. I thus decided to join a Master’s program in Paris focusing on these two topics. The possibility of generating, in vitro, a specific subtype of human cells with the right ‘recipes’ fascinated me, and for my internship I joined the I-STEM institute and Stéphane’s lab. I then moved on to do a PhD, and helped Stéphane set up the new lab in Paris while continuing working between the two institutes. It was an intense but enriching experience.

CV: During my undergraduate studies at Sorbonne Université, I quickly became interested in developmental biology and neuroscience. During a first internship in the lab of Jean Livet in Paris, I studied neural lineages in the chick embryo spinal cord, which confirmed my interest in these two fields. This led me to join Stéphane’s lab, first as an intern and then a PhD student, to investigate molecular mechanisms controlling spinal cord development. The power of the in vitro approaches used in the lab allowed me to decipher signalling mechanisms controlling spinal neural diversification – a subject that fascinates me.

How has your research been affected by the COVID-19 pandemic?

VM, CV & SN: Our lab was completely shut down for 2 months last spring. Afterwards, we worked part-time on site to limit the number of people, which obviously significantly delayed the projects. However, working mostly with in vitro models helped, as it was easier to stop and restart the experiments. Also, the lockdown forced us to focus on writing and planning experiments, which was a positive side effect. Overall, this pandemic has most certainly delayed the progress of our research, but we were fortunate to return to the lab fairly quickly and be able to work in good conditions thanks to the heads of our institute, who did a fantastic job in dealing with the situation.

Before this project, what was understood about the relative influence of extrinsic and intrinsic factors on the pacing of the human HOX clock?

VM, CV & SN: We already knew a lot about the extrinsic and intrinsic mechanisms controlling the sequential induction of HOX genes during axial elongation. However, several aspects remain obscure; notably, the mechanisms pacing the clock within axial progenitors, in particular in humans. It was well established that cis-regulatory sequences within and outside the complexes are important for HOX gene sequential induction, and that progressive changes in chromatin structure along the complexes accompany the progression of the HOX clock. On the other hand, extrinsic factors such as retinoic acid (RA), Wnts, FGFs and GDF11 were shown to induce HOX gene collinear expression or modulate HOX gene expression patterns. However, whether these extrinsic factors were pacing the sequential activation within axial progenitors or were actuating an intrinsic timer was unclear.

Effects of modulating the duration of retinoic acid exposure on HOX gene expression during in vitro motor neuron differentiation.

Can you give us the key results of the paper in a paragraph?

VM, CV & SN: We first characterized the expression profile of HOX transcription factors and MN subtype markers in the human embryonic spinal cord, so we could assess the functional consequences of HOX regulation in axial progenitors and properly define the identity of in vitro-generated human MNs. Using MN subtype as a readout, as well as transcriptional analysis of the axial progenitor stages, we showed that HOX genes undergo a temporal collinear activation in hPSC-derived axial progenitors that, upon differentiation, generate MN subtypes found in progressively more caudal regions of the spinal cord. Analysis of the transcriptomic data showed that the sequential activation of HOX genes was paralleled by an increase in FGF ligands and markers of active FGF signalling. This FGF activity was necessary for the HOX clock to proceed, and precociously increasing FGF levels hastened the expression of HOX genes expressed normally later on. The HOX clock was further accelerated with a rapid rise of the very caudal HOX10 genes when FGF was combined with GDF11, another extrinsic factor known to control the expression pattern of caudal thoracic and lumbar HOX genes in mouse and chick embryos. Slowing down or accelerating the clock in axial progenitors was always paralleled by a shift in MN subtype specification within the same time line of differentiation. These results demonstrated that the pace of HOX gene activation within axial progenitors is regulated by sequences of extrinsic factors. This observation argues against a solely intrinsic, chromatin-based, pacing mechanism. However, even in the most accelerating/caudalizing conditions, HOX genes are still expressed in a largely collinear sequence, which suggests that cell-intrinsic mechanisms likely ensure the order of expression. In addition, our work provides for the first time a method to efficiently generate well-defined MN subtypes for basic and translation approaches.

The pace of HOX gene activation within axial progenitors is regulated by sequences of extrinsic factors

Do you have any idea what controls the onset and duration of FGF signalling in hPSC cultures and in the embryo?

VM, CV & SN: The onset and duration of FGF signalling in axial progenitors are certainly controlled by extrinsic factors both in vivo and in vitro. Work from different labs has indicated that FGF and Wnt signals, provided in vivo by the primitive streak and the surrounding epiblast, and in vitro by addition of agonists in the medium, specify axial progenitors, which in turn induce different FGF and Wnt ligands. Thereby, a positive-feedback loop is generated, which likely contributes to an increase in FGF signalling overtime. Accordingly, in this study we observed a temporal induction of FGF ligands and of well-recognized downstream target genes in hPSC-derived axial progenitors. Then, in the neuronal lineage, the duration of FGF signalling in axial progenitors depends on the rate of their differentiation in neural progenitors. In vivo, neurogenesis-promoting RA from the abutting somites can repress FGF gene expression and pathway activity. In our study, the duration of FGF signalling is also likely controlled by the moment at which axial progenitors are exposed to RA. Of note, we also showed that FGF concentration can be integrated by axial progenitors so, in addition to the duration of FGF signalling, a progressive increase in FGF concentration might play a role in pacing the HOX clock. Whether this occurs in embryos is currently unclear.

The dynamics of intracellular signalling downstream of FGFs might also play an important role in rostro-caudal patterning. We showed that FGF activity on HOX genes requires activation of the MAPK pathway. In other models, this pathway adopts distinct signalling activity dynamics in response to variations in concentration or in duration of extrinsic factors. Whether changes in intracellular signalling dynamics downstream of FGFs play a role in HOX clock regulation is an interesting avenue to pursue.

How do you think your findings will impact clinical or bioengineering efforts?

VM, CV & SN: It’s another important aspect of the paper. Studying developmental principles using hPSC differentiation helps optimize differentiation strategies so specific cell or tissues types can then be used for disease modelling, drug screening or cell therapy approaches.

In our case, one consequence of the discovery of the HOX pacing mechanisms is the ability to efficiently and synchronously generate MN subtypes found at different positions in the human spinal cord. MN diseases, such as amyotrophic lateral sclerosis or spinal muscular atrophies, differentially impact MN subpopulations, but the basis of this differential vulnerability remains largely unknown. Providing the community with the ability to generate these MN subtypes might stimulate research on these currently incurable diseases.

Finally, considering the iterative use of HOX transcription factors to induce cell diversity in many lineages, it will be interesting to explore whether our strategy could help refine the production of other cell types, such as somite or neural crest derivatives.

When doing the research, did you have any particular result or eureka moment that has stuck with you?

VM: First seeing the efficient induction of caudal HOX genes while preserving MN induction was a particularly important moment. I’m also happy that we characterized HOX expression patterns and MN subtype markers in human embryos in collaboration with Gist and Mackenzie, who initiated that at Columbia. I think it will be an important resource.

CV: I tested different concentrations of Wnt agonist and I was particularly thrilled when I finally understood why a specific cell line required higher concentration of this agonist to generate caudal MNs. It’s a result that is a bit hidden in the manuscript but might have important consequences when people want to use these protocols with their favourite cell line. I also remember when I assembled images and graphs for the first time to organize the figures, I realized the accomplished work, even if much remained to be done at this time, and I was very proud of all the work we did together with Vincent.

And what about the flipside: any moments of frustration or despair?

CV: Experimentally, it has not always been smooth and easy: hPSCs can be tricky to deal with and are always demanding. However, I had the chance to work very closely with Vincent and constantly support each other, which helped a lot, both scientifically and personally. And finally, discovering such interesting results always cheered us up.

VM: As Celia said, working with hPSCs has its pros and cons as cells need to be taken care of almost every day, and all products and reagents need to be carefully calibrated. We had coating issues at some points and still unexplained cell death at another. These periods were very frustrating.

What next for you two after this paper?

CV: I am currently exploring the signalling mechanisms downstream FGF using reporters of signalling pathway activities. I will defend my thesis in a few weeks. Then I’m favouring a career in a biotech or industry.

VM: After defending my thesis I wanted to look for jobs abroad but the pandemic delayed this project. In the meantime, I’m helping with the national effort to test and track Covid patients while exploring future plans.

Where will this story take the Nedelec lab?

SN: On one hand, as we always try to combine developmental biology with cell engineering, we are exploring what’s downstream of the extrinsic factors that pace the HOX clock, the mechanisms by which they signal to the genome to induce distinct cell fates, and how they control spinal cord morphogenesis (using a new type of organoid model). 3D in vitro differentiation provides an experimentally accessible model for fine modulations of signalling pathways that can be coupled to genomic analysis while tracking consequences on cell fate and tissue shape. In collaboration, we are implementing optogenetic approaches and genomic approaches to address these questions.

On the other hand, we use the products of these developmental studies to study the basis of the differential vulnerability of MN subtypes in different forms of paediatric MN diseases called spinal muscular atrophies. For that, we have created a very stimulating network of collaborators, including clinicians and cell biologists.

Finally, let’s move outside the lab – what do you like to do in your spare time in Paris?

CV: A long time ago, before the pandemic, I really enjoyed living in Paris and often went to the cinema or museum, and I loved to discover new restaurants. But nowadays, it’s more biking or walking in the city and around, when the weather is nice!

VM: I like to walk randomly and get lost in the maze of Paris, discovering new streets and monuments randomly. When it was still possible, I particularly enjoyed waking up early on weekends to go to the Louvre to walk around the museum with almost nobody around. I also really enjoyed going to bars with co-workers to share problems and discuss projects. While I reduced this activity during my PhD, I’m also a big fan of Aikido.

SN: As Célia and Vincent mentioned, Paris will, hopefully, soon be Paris again, so we can enjoy the theatres, museums and the terraces. I also like rock climbing and, while it might sound surprising, Paris is not such a bad place for it. The nearby forest of Fontainebleau is a fantastic bouldering spot with endless possibilities. As my daughter has started to really enjoy it as well, I try to go as much as I can.

Thumbs up (No Ratings Yet)
Loading...

Tags:
Categories: Interview

How exploring single cell dynamics in the early embryo revealed a protective system based on mechanical cell cooperation

Posted by , on 15 July 2021

by Esteban Hoijman and Verena Ruprecht

The immune system is able to deal with cellular anomalies and invading pathogens. However, it has remained enigmatic how vertebrate embryos handle stress conditions before such an immune system develops. We recently found that embryos are able to perform innate immune functions as early as in blastula stage of development, using the surface epithelium as a phagocytic clearance system of defective cells1. This epithelium, the trophoblast in mammalian embryos, is the first differentiated tissue that forms during embryonic development, and our study revealed that it has unrecognized important functions for the survival of the early embryo.

The story began when Esteban joined as a postdoctoral researcher the recently established lab of Verena at the Centre for Genomic Regulation in Barcelona, Spain. With the aim to study early vertebrate embryo development under native and stress conditions we started to acquire movies of single cell dynamics at high spatiotemporal resolution in the zebrafish blastula. Interestingly, we noticed that epithelial cells of the embryo surface (termed enveloping layer in zebrafish) were able to ingest fragments of internal stem cells, forming vacuolar like structures in epithelial cells (Figure 1). We further observed that epithelial uptake was specifically increased in stressed embryos that showed signs of malformations or tissue damage caused by mechanical perturbation, suggesting that the ingested cells originated from cellular failures or events of cell death. In fact, we were able to confirm that the surface epithelium specifically ingested dead cells, as the ones generated in normal embryo development by errors in mitosis. This led us to think that the epithelial tissue at the embryo surface formed during early development (only four hours post fertilization in zebrafish embryos), could function as a scavenger of dead cells at these stages. This was a challenging idea, as specific clearance functions of epithelial cells have not been previously considered to occur in the early embryo. Phagocytosis of dead cells by epithelia was known to be important in certain adult tissues2-3, suggesting that epithelial cells could perform similar functions in the early embryo. We reasoned that this function could have an essential protective role in development, as an accumulation of dead bodies might lead to multiple defects, from mechanical interference with gastrulation movements to the uncontrolled release of intracellular contents by secondary necrosis, including signaling molecules and toxic factors.   

 

Figure 1. Epithelial clearance of apoptotic cells in the early zebrafish embryo. (A) An internal progenitor cell dies and is ingested by the superficial epithelial layer (EVL). (B,C) Images (B) and quantification (C) showing epithelial clearance of abundant progenitor cell death induced by mosaic expression of the proapoptotic protein Bax (red cells). (D) Morphodynamic analysis of a single phagocytic event. The apoptotic cell is shown in red. Adapted from Hoijman et al, Nature 2021, 590:618.

Motivated by the possible relevance of these observations, Esteban combined his expertise in live embryo imaging with experience in cell apoptosis that he gained during his PhD. We developed a single cell in vivo morphodynamic analysis of phagocytosis that allowed us to study epithelial phagocytic events inside a living animal (Figure 1B). Our study revealed that the phagocytic cup formed by epithelial cells exerts compressive forces during uptake of an apoptotic cell, a feature recently discovered to be shared by macrophages ingesting synthetic phagocytic targets in culture4

Next, we decided to evaluate if this process was conserved in mammals, as this could have relevant implications for the progression of human embryonic development. Filming mouse blastocysts, we observed that the surface epithelium (in this case the trophoblast) was also able to clear apoptotic cells by recognizing surface molecules of dying cells, specifically phosphatidylserine. This observation indicated that the early epithelial phagocytic process we first observed in zebrafish embryos was evolutionary conserved and based on similar molecular mechanisms to the ones used by professional phagocytic cells of the immune system such as macrophages. These results were exciting, as they suggested that the first tissue formed at the surface of a vertebrate embryo is used for its protection. This is particularly relevant considering that the main cause of early miscarriages in human embryos are errors in mitosis leading to cell death5.

We observed that phagocytic clearance was sensitive enough to detect single cells dying, while being efficient enough to be able to clear hundreds of cells dying simultaneously (Figure 1). We were thus wondering how epithelial cells, which are sessile cells in the epithelium, can achieve such high efficiency for clearance, as compared to immune phagocytes which are motile to perform their (clearance) task. The answer was hidden in our movies of live zebrafish embryos. We detected two surprising features: first, dead cells moved much more than what we expected before the uptake, significantly faster than the live neighboring cells undergoing gastrulation movements. Although it was proposed that apoptotic cells can acquire specific motility inside tissues, this was not directly proven to be cell-autonomous6-8. Our dynamic analysis of actomyosin network organization, which showed a static localization and non-polarized organization in apoptotic cells, was however supporting that they do not have autonomous motility. Second, by visualizing F-actin dynamics inside the whole embryo, we observed the formation of actin accumulations in contact with the external rear surface of motile apoptotic cells. We therefore questioned, could these two observations be linked? Surprised by the observation of these actin structures, we performed both high-speed imaging to analyze actin localization dynamics and single cell staining to identify the cells in which they are formed. This allowed us to determine that these actin structures formed in epithelial cells and constitute a new elongated protrusion, with actin enriched at the protrusion tip in contact with apoptotic cells and moving coordinately with them, a protrusion type that we called “epithelial arms” (Figure 2A,B).

Figure 2. Epithelial arms propel apoptotic cells improving tissue clearance efficiency. (A) An epithelial arm forms upon contacting an apoptotic cell (red). (B) Epithelial cells form two types of protrusions when recognize an apoptotic cell: phagocytic cups to ingest and epithelial arms to push. (C) Consecutive pushing by epithelial arms increases the number of epithelial cells encountered by the each apoptotic cell (red). (D) Modeling indicates that the time to complete clearance of an apoptotic mass is reduced when velocity of the apoptotic targets increases. Nmax: maximum number of targets ingested by each epithelial cell. Adapted from Hoijman et al, Nature 2021, 590:618.

We came up with different hypotheses that might explain this observation: Could these protrusions mechanically push the apoptotic cells? Or are protrusions just “chasing” apoptotic cells? In the latter case, we should probably observe some apoptotic cells moving without contacting epithelial arms. However, we observed a stringent correlation between arms and apoptotic movement with both occurring hand in hand. To evaluate our first hypothesis, we designed an experiment in which we transplanted synthetic apoptotic targets made of lipid aggregates into the embryo as a source of objects with no intrinsic motility. Interestingly, these targets were efficiently phagocytosed and, furthermore, they acquired similar motility as apoptotic cells in terms of velocity fluctuations, directionality, and association with epithelial arms. The movement of the surrogate targets indicated that epithelial arms can propel apoptotic targets using actin-dependent forces. Looking at the whole tissue, we further found that apoptotic cells moved along long-range trajectories, caused via consecutive pushing by multiple epithelial arms formed by different epithelial cells (Figure 2C), with the path for each dead cell target being described by a random-walk.

We therefore asked, how does this apoptotic target movement influence the overall efficiency of tissue clearance? In other words, from the point of view of a single epithelial cell, why would it be meaningful to push a target instead of ingesting it directly? Analyzing population movement of apoptotic targets, we observed that the random-walk behavior led to the overall spreading of the apoptotic mass. A larger area of the epithelial tissue was therefore “visited” by apoptotic targets over time. Comparing this behavior to professional phagocytes, which usually move towards an apoptotic cell mass to perform efficient clearance, we interpreted the observed spreading of the apoptotic mass as an inversion of the clearance process, with motile targets moving around sessile epithelial phagocytes.

The immediate consequence of apoptotic motility was an increased number of epithelial cells encountered by each target. We hypothesized that several possibilities could lead to an advantage for cells clearing the tissue when increasing the phagocyte-target encounter rate. We therefore set out to mathematically model the clearance process and its efficiency. For this, Verena teamed up with Stefan Wieser (ICFO Barcelona) and Andrew Callan-Jones (CNRS/Université de Paris) to perform simulations and derived a theoretical formula that predicted that increasing the speed of target spreading (mediated by arm pushing) decreases the time required to clear the whole apoptotic mass, thus making the process more efficient (Figure 2D). Importantly, this was specifically relevant when a limited uptake capacity for each epithelial cell exists, as we determined experimentally.

The successful combination of this theoretical and experimental analysis indicated that the stochastic spreading allows the recruitment of new epithelial phagocytes near the vicinity of those cells that already reached their maximal uptake capacity, thereby accelerating the clearance process at the tissue level.

In summary, the mechanical cooperation between epithelial cells mediated by the active dispersal of apoptotic targets by epithelial arms improves the protection of the embryo at the earliest stages of development. We are now exploring the impact of this phagocytic system at the CRG Barcelona (Verena) and the newly established group at the University of Barcelona (Esteban).

As many other researchers experienced in 2020-2021, performing a revision in the peak of the pandemic was a challenging situation that led us to deal with complex situations and demanded creative solutions and support between labs to solve unpredictable problems (i.e. we needed a reagent during the revision when companies paused their supply; we were lucky to receive it as a gift from the lab next door). We believe that our work is a good example of how advanced imaging technologies that can nowadays capture single cell and molecular dynamics inside live animals open new exciting opportunities for the discovery of relevant in vivo cell and tissue functions and their underlying mechanisms.

  1. Hoijman et al, Nature 2021, 590:618, doi: 10.1038/s41586-021-03200-3
  2. Akhtar et al, Dev Cell 2016, 38:522, doi: 10.1016/j.devcel.2016.08.005
  3. Juncadella et al, Nature 2013, 493:547, doi: 10.1038/nature11714
  4. Vorselen et al, bioarxiv 2021, doi.org/10.1101/2021.03.14.435346
  5. Bolton et al, Nat Commun 2016, 7:11165, doi: 10.1038/ncomms11165
  6. Yamaguchi et al, J Cell Biol 2011, 195:1047, doi: 10.1083/jcb.201104057
  7. van Ham, Curr Biol 2012, 22:830, doi: 10.1016/j.cub.2012.03.027
  8. Dzhagalov et al, PLoS Biol 2013, 11:e1001566, doi: 10.1371/journal.pbio.1001566
Thumbs up (1 votes)
Loading...

Tags: , , , , , ,
Categories: Research

Genetics Unzipped podcast: Genes for all – Making sure everyone benefits from genetics research

Posted by , on 15 July 2021

Masai Tribe Kenya – Willy Horsch, CC BY-SA 3.0 via Wikimedia Commons

In the latest episode of the Genetics Unzipped podcast we’re meeting some of the researchers who are working to make sure that everyone gets the benefits of genetic research – from underserved communities to entire continents.

Kat Arney chats with Charles Rotimi, director of the Centre for Genomics and Global Health within the NIH National Human Genome Research Institute at Bethesda, Maryland in the US, and a distinguished NIH investigator. He’s also the founder of the African Society of Human Genetics and the driving force behind a major genomics project called Human Heredity and Health in Africa, or H3Africa, which he helped to establish ten years ago.

She also talks with Laura Koehly – a senior investigator at the National Human Genome Research Institute, with a special interest in helping people unlock the information hidden in their family health histories, particularly focusing on underserved and less privileged communities.

Finally, Kat hears from bioethicist Sara Hull, who has worked at the National Institutes of Health for more than 20 years helping researchers make sure their work is done ethically and doesn’t cause harm to the people involved.

Genetics Unzipped is the podcast from The Genetics Society. Full transcript, links and references available online at GeneticsUnzipped.com.

Subscribe from Apple podcasts, Spotify, or wherever you get your podcasts.

Head over to GeneticsUnzipped.com to catch up on our extensive back catalogue.

If you enjoy the show, please do rate and review on Apple podcasts and help to spread the word on social media. And you can always send feedback and suggestions for future episodes and guests to podcast@geneticsunzipped.com Follow us on Twitter – @geneticsunzip

Thumbs up (No Ratings Yet)
Loading...

Tags: , , , , ,
Categories: Outreach, Podcast, Resources, Societies

The people behind the papers – Clément Dubois, Shivam Gupta, Andrew Mugler and Marie-Anne Félix

Posted by , on 12 July 2021

This interview, the 95th in our series, was published in Development earlier this year

Cell migration needs to be precisely regulated during development so that cells stop in the right position. A new paper in Development investigates the robustness of neuroblast migration in the C. elegans larva in the face of both genetic and environmental variation. To hear more about the story, we met the paper’s four authors: Clément Dubois and Shivam Gupta, and their respective supervisors Andrew Mugler (currently Assistant Professor at the Department of Physics and Astronomy at the University of Pittsburgh, where his lab recently moved from Purdue University) and Marie-Anne Félix (Principal Investigator at Institut de Biologie de l’Ecole Normale Supérieure in Paris and Research Director at CNRS).

Clément (top left), Shivam Gupta (top right), Marie-Anne (bottom left) and Andrew (bottom right).

Andrew and Marie-Anne, can you give us your scientific biographies and the questions your labs are trying to answer?

AM: After a PhD and postdoctoral research on theoretical biological physics, I began my own position investigating noise in biological systems. Cells contend with unavoidable noise from environmental fluctuations, small molecule numbers and many other sources. My group uses tools from theoretical physics to uncover strategies that cells use to precisely sense their environment, undergo controlled developmental or phenotypic changes, and execute collective behaviours.

M-AF: After a PhD in cell biology, I wanted to combine evolutionary biology with cell/developmental biology. In Paul Sternberg’s lab at Caltech and in my lab’s early years, we compared vulva development in different nematode species, giving flesh to the concept of developmental systems drift. Over the past 20 years, we moved to a microevolutionary scale: studying the sensitivities of developmental systems to various types of perturbation and comparing these with their evolutionary variation. The overall thread is to assess how much development biases phenotypic evolution, with some phenotypes being more easily reached than others upon random genetic variation or quantitative tuning of parameters.

Studying natural variation led us to seek C. elegans in nature, which was a lot of fun. With many colleagues, we have since been probing its population genetics structure, and the zoo of its associated microbes. Bringing real worms back to the lab also led us to discover natural variation in the duration of their multigenerational memory.

Clément and Shivam – how did you come to work in your respective labs and what drives your research today?

CD: I did an MSc in ecology and evolution, and was specifically interested in host-pathogen interactions. After a first internship in the Pasteur Institute working on mosquito-virus interactions, my former supervisor suggested I apply to Marie-Anne’s lab. I started working on the intraspecific evolution of C. elegans resistance to microsporidia, and then did a year as a technician working on the evolution of a peculiar cell in C. elegans called P3.p. This year was really formative. I gained a lot of scientific maturity, and realised how rich and friendly the C. elegans community is. Then Marie-Anne proposed I do a PhD on the evolution of QR neuroblast migration. Considering the different topics I worked on, I would say that my research is mostly driven by curiosity and the question of how much genotypes can shape phenotypes.

SG: I came to Purdue University as a grad student in 2015. I joined Dr Mugler’s lab after doing a lab rotation to get a feel for the research. The research work in the Mugler lab was very challenging, and it was intriguing to see how fundamental physics can explain the behaviours of microscopic organisms.

How has your research been affected by the COVID-19 pandemic?

M-AF: The lab closed for 3 months in the spring of 2020; since then it has been somewhat restricted, but we can do experiments. Of course, C. elegans is a great organism to work on because we do not lose strains: they can be kept frozen or can live for 3 months in diapause (a pre-adaptation to SARS-CoV2!). But some projects were affected for more than 3 months. To enhance social interactions, since the fall we decided to meet after lunch every day in a hybrid format (real life and video). This compensates for the usual lunch-time exchanges, be they scientific or not. On the positive side, the pandemic also gave us the impetus to meet more readily with colleagues worldwide, attending each other’s lab meetings or organising informal meetings.

AM: As a theory group, we could continue most of our work from home without interruption. However, developing ideas in any area benefits from true collective brainstorming, which has been slowed without the ability to physically meet at the whiteboard. Additionally, the pandemic has increased my childcare responsibilities, which has left less time to dedicate to students. On the positive side, I moved institutions in the middle of the pandemic, and I now hold hybrid group meetings with group members from both places, which is perfectly natural to do virtually in these times.

How did your labs come to collaborate on this project?

AM & M-AF: This was all started by our colleague Rik Korswagen, who has been working on C. elegans QR migration as a model. He contacted us both to write an application for a collaborative grant from the Human Frontier Science Program. We got the grant, and this has been a great experience. The modelling of body size variation was not in the grant, but the collaboration took place naturally once we were networking.

Can you give us the key results of the paper in a paragraph?

CD, SG, AM & M-AF: The goal was to measure the precision and natural variation in the final position of a cell that migrates during development. Other studies in various systems have examined the directionality of cell migration, but to our knowledge not the precision in the cell’s final position. Specifically, we studied two descendants of the QR neuroblast lineage that migrate a long way during C. elegans development. The Korswagen lab had shown that the end of their migration is specified by a temporal rather than a spatial mechanism. Given this temporal regulation, we hypothesised that a change in body size would affect the final position of the neurons relative to body landmarks. Indeed, we observed that a smaller body size resulted in the cell migrating further. Cell position even changed as a function of maternal age, a factor previously known to affect body size. We developed a mathematical model of the expectation, taking into account larval growth during the migration. The data did not fit quantitatively the simplest form of the model. Then, a model with partial compensation of body size, grounded on measurements of cell speed, fitted the experimental results without any free parameters. Finally, we revealed natural variation among wild isolates of C. elegans in the neurons’ final position, large enough that they result in a change in their neighbouring cells.

Late L1 larvae showing examples of the relative position of QR.pax in different strains: posterior (strain XZ1516, top) and anterior (strain EG4725, bottom).

Do you have any idea why cell migration speed is dependent on body size?

CD & M-AF: The first that comes to mind is that a larger body size means larger cells (at least for a species such as C. elegans, which has a quasi-invariant number of cells). But interestingly, we could not find much published on whether a larger cell would go faster or slower than a smaller one. Both appear possible, depending on specific motility mechanisms. Another possibility is that the external environment of the cell is altered as a function of body size; for example, a change in the Wnt gradient that influences QR migration, either its initial concentration or its decay length.

On the other hand, in the wild isolates, the eventual position of QR.pax did not correlate with body size: why do you think this is?

CD & M-AF: We did not detect a significant correlation, and we cannot rule there would be one with a larger set of experiments. In any case, this means that much of the variation is due to other factors that can go in another direction than the effect of body size. Variation in many genes and processes can affect a quantitative trait such as cell position.

Your paper was published in Development through the Review Commons route – how did you find the experience?

CD, SG, AM & M-AF: Great! This was our first time. With Review Commons, the manuscript is sent to reviewers without any submission to a specific journal. The reviewers do not have any specific journal in mind. In our case, the reviews were very constructive, especially by suggesting that we assessed the effect of maternal age. Once the reviews arrived, the authors can contact one of the participating journals, specifying what is planned for responding to reviews. Thanks to Development, this went very well.

When doing the research, did you have any particular result or eureka moment that has stuck with you?

CD: The first eureka moment was to see the preliminary results on the natural variation of QR.pax final position. To be able to detect differences between wild isolates for the first time was really exciting and promising. From then on, I was excited at the end of each experiment after making plots from the data (and I spent a lot of time on them!).

SG: The first few months of my PhD work was mainly building mathematical models for temporal precision in gene expression. At first, I was a bit pessimistic that the rules of physics would explain the complex behaviour of organisms. After analysing experimental data from C. elegans, it was very satisfying to see that the behaviour of microscopic cells can be explained by our models.

And what about the flipside: any moments of frustration or despair?

CD: I did not have a particularly memorable moment of frustration or despair – even negative results were informative. Nonetheless, the most challenging part was probably the measurements of embryo and larva lengths at different time points, in parallel on four genotypes. It required a lot of organisation and efficacy, but it was worth it.

SG: To solve complex biological problems, we start with a simple model and slowly build up the model. After a point, our model became very complex and it took days to run the computer program. It was frustrating to wait days to see results, but in the end, when results were promising, it was very joyful.

What next for you two after this paper?

CD: In the short term I’m doing two things: writing my PhD manuscript and working on the intraspecific evolution of the final position of QR.pax. Using Recombinant Inbred Lines between CB4932 and JU1242 (two genotypes with an opposite phenotype), I found a QTL on chromosome IV. The goal now is to find the polymorphism associated with the difference in QR.pax final position.

SG: I have successfully defended my thesis and secured a job in the pharmaceutical industry. My ambition is to advance drug discovery and safety of drugs through developing pharmacokinetics/pharmacodynamics models and analysing clinical trial data.

Simple mathematical models can prove very useful for sharpening basic understanding of biological systems

Where will this story take the Mugler and Félix labs?

AM: This has been one of the most positive demonstrations that simple mathematical models can prove very useful for sharpening basic understanding of biological systems; I hope to continue in this vein with this and other collaborations.

M-AF: I hope we can find the polymorphism behind the divergent phenotypes of two wild C. elegans isolates. Beyond, I will keep vulva development as our main developmental model, but, as both are regulated by the same Wnt gradient, we will likely come back again to QR migration.

Finally, let’s move outside the lab – what do you like to do in your spare time in Paris and West Lafayette?

CD: Paris is full of small and hidden restaurants that are really good. I like to explore them with my partner. I also enjoy spending time with my friends playing board games, climbing in the gym or in Fontainebleau, a forest close to Paris known for its large boulders.

SG: I enjoy exploring new places around West Lafayette for hiking and biking.

AM: Biking, exploring new restaurants and travelling.

M-AF: Hunting. For nematodes.

Thumbs up (No Ratings Yet)
Loading...

Tags:
Categories: Interview

A bad day for the angel on my shoulder: Practical advice for grant writing from the #devbiolwriteclub and #devbiolgrantclub

Posted by , on 11 July 2021

Hello!  Welcome back to the #devbiolwriteclub!  Over the last year or so, I’ve harangued you on Twitter and on The Node about practicing the craft of writing.  I’ve ignored any practical advice on what you should actually put on a page and instead I have focused on how to build the habits of mind that allow you to grow as a writer.  Yesterday, I launched a new Twitter project called #devbiolgrantclub, where I’ll be offering random bits of grantsmithing advice.  Today, I’ll present a “crossover episode” of the two projects. 

As academic scientists, we all know that we must master two types of writing: papers and grants.  But I don’t think enough people really grasp the fact that the two represent entirely distinct disciplines within scientific writing.  In fact, the best way to write a grant is totally, totally different from the best way to write a paper. 

Ask anyone in my lab, and they’ll tell you that I love papers like druids love trees.  Writing papers is one my life’s greatest joys. It’s also the ultimate goal of science, which is why papers are the hard currency of our field.  In fact, call me sentimental, but I see something noble in our endeavor to elegantly reveal to the world the new knowledge that we have discovered.

Just the same, doing this noble work while simultaneously contemplating my next grant proposal brings to mind a great Melvern Taylor song, from which I borrowed this post’s title.  That angel is perched there on your shoulder, egging you on as you do the good work of science.  But let me tell you, successful grant writing absolutely requires that you ignore that angel and listen very carefully to the devil on your other shoulder.

The best grant writing advice I ever heard came from a clinician from UT, San Antonio at a grant writing workshop I attended when I was in Berkeley as a postdoc.  Between solid practical advice and a riotously funny story about tequila and a big stack of grants to review, he said this:

“Listen.  You don’t have to be proud of what you wrote.  You have to get the money.”

Think about that.  Let it sink in.  It’s harsh, for sure.  It may even be antithetical to how we see ourselves as academic writers.  But it’s the ultimate truth of grant writing.   You don’t have to be proud of what you wrote.  You just have to get the money. 

Now let me be clear:  I am not talking about being sleezy or self-aggrandizing, and I’m definitely not talking about making stuff up.  Rather, I am simply proposing that you be intentional about grant writing by ignoring what you want, learning exactly what your audience needs, and providing exactly that.

To understand what I mean, consider this:  Every once in a while, I find myself with a totally free afternoon.  So, what do I do?  I go get a cup of coffee, sit somewhere pleasant, and read papers.  Maybe I grab a few from the stack on my desk, maybe I hit Pubmed to look for something new, or maybe I click a link on Twitter.  It’s a joy.

On the other hand, no one in the history of science ever said to themselves: “Gee, I’d really like to read and carefully review ten randomly assigned grants in my free time today.”  This gives us the First Principle of grant writing:

1.  Assume your readers do NOT want to read your grant. 

This is possibly the biggest concept that grant writers fail to grasp.  When you write a paper, you can reasonably assume that whoever reads it wants to read it.  They very likely share your interest in the subject.  You might even say they want to know how the story ends.  Thus, they are actually quite likely to overlook a confusing paragraph, or power through a difficult passage.  At the very least, they’ll give it a read and likely learn something. 

But your reviewer did not choose to read your grant.  Your reviewer was assigned your grant by a grant officer.  The cynical (and effective) grant writer therefore assumes that the reviewer does not care how the story ends, but does know exactly when it will end:  When the review is written and submitted to that grant officer.  

Presented with this hard fact, you might feel compelled to try to write the grant in such way as to MAKE the reviewer want to read the grant.  Don’t.  It’s too risky.  There are just too many variables.  (E.g., Sorry, you cannot ever make me want to read a grant about… well, lots of things, but I won’t name them.)   

On the other hand, there is one thing that will make all reviewers happy, and that is making the grant EASY for the reviewer to read and absorb.  More thoughts on how to do this later, but for now, let me drive home why this strategy works by presenting the Second Principle of grant writing:

2.  Accept that your readers MUST compare your grant to other grants, mentally ranking them in real time. 

This is another critical distinction between grants and papers that people usually don’t consider when writing.  If I am doing my work as a scientist, I am judging the quality of every single paper.  Am I convinced by the data?  Do I care about the conclusions?  Just the same, I am not sitting there thinking hard about whether your paper is better or worse than the last one I read.  And I am absolutely not trying to mentally rank the last ten papers I read.  On the other hand, if I am reading your grant, this is exactly what I am doing. 

This simple fact creates a very different workflow for grant readers, as compared to paper readers.  The grant reader won’t just start reading ten grants in the order they were assigned.  Instead, most reviewers will at least glance through the grants they are assigned, but then will pretty quickly decide which ones to read first – or last.  So, in very short order, a mental ranking of some sort is already starting to emerge.  This bears directly on our work as grant writers. 

Here’s another thought exercise.  You are a reviewer with a stack of nine grants.  Two are in your specific sub-field and are asking questions that you find interesting.  One grant has the minimum-allowed 0.5-inch margins, long paragraphs taking up nearly half a page, no white space breaking up the text, tables filled with huge amounts of data in small fonts, and no color anywhere.  The other grant has larger margins and bullet points and colorful diagrams to summarize key points; these are well separated by white space from the main text, and that text is presented in short, easy-to-digest paragraphs.  This second grant stops several inches short of the bottom of that last page.  Tell me, which of those two grants -that you have to read today- will you choose to read first?

This brings us to the Third Principle.

3.  Help your readers; they have to WRITE a careful review of your grant, AND ALSO several other grants.

This is the last major distinction between paper and grant readers. When I am reading a paper, I may or may not need to write about it, but if I do, I will usually end up doing so in a pretty indirect way.  I’m very unlikely to do that writing the same day I read the paper.  But consider the beleaguered grant reviewer.  Just a few weeks to read an entire stack of grants, choose which ones are better and worse, and write reviews for all of them, justifying their decisions. 

So, as a writer, this is where I am least proud of my grant writing:  In my quest to make my grants easy for Reviewers, I consider that there are sections of NIH grant review forms titled Significance and Innovation.  So, my grants include prominent, underlined sentences that read: “This grant is significant because…” or “This grant is innovative because…”.  It’s clunky, unsubtle language, and I hate it.  I hate bullet points, too.  I also despise witless diagrams that I know are oversimplified.  But if I provide these things, and do so with care and intent, it will help me get the money.

So those are three Principles to keep in mind when writing grants.  Next, let’s see how they relate to the Rules of #devbiolwriteclub.

Rules #1 and #2 tell us to do the work.  Make grant writing a specific craft you practice with intent throughout your career, not just this thing you need to do when you’re out of money.   

Rule #3 tells us to revise and edit, again and again.  When you are doing this, though, keep the Principles foremost in your mind.  Revise and edit to make the grant EASIER to read, EASIER to rank, and EASIER to review.

Rule #4 tells us to read with intent.  So, read grants with intent.  Ask you PI and your peers for grants they wrote.  Read the ones that got funded, but also read the ones that DID NOT get funded.  Read the reviews!  Here’s an idea I just came up with:  Instead of journal club, have grant reviews club.  Read a grant and the reviews together as a group.  Try to figure it out.

Rule #5 says you can’t do it alone.  So, when you beg your friends to read your grant, don’t ask them if they found typos.  Ask them if it was easy to read.  Also, recall that grants are assigned to reviewers, frequently outside their core area.  So, ask people WHO ARE NOT IN YOUR lab to read your grants. 

Finally, leave waxing lyrical and fighting the good fight for your papers. 

Write grants with a singularity of purpose: Get the Money.

Thumbs up (17 votes)
Loading...

Tags: ,
Categories: Discussion

June in preprints

Posted by , on 6 July 2021

Welcome to our monthly trawl for developmental biology (and related) preprints.

The preprints this month are hosted on bioRxiv, arXiv and preprints.org – use these links to get to the section you want.

Developmental biology

Cell Biology

Modelling

Tools & Resources

Research practice & education

Developmental biology

| Patterning & signalling

Fly embryos from Poovathumkadavil, et al.

Ssdp influences Wg expression and embryonic somatic muscle identity in Drosophila melanogaster
Preethi Poovathumkadavil, Jean-Philippe Da Ponte, Krzysztof Jagla

Inhibition of aryl hydrocarbon receptor signaling promotes the terminal differentiation of human erythrocytes
Yijin Chen, Yong Dong, Xulin Lu, Wanjing Li, Yimeng Zhang, Bin Mao, Xu Pan, Xiaohong Li, Ya Zhou, Quanming An, Fangxin Xie, Shihui Wang, Yuan Xue, Xinping Cai, Mowen Lai, Qiongxiu Zhou, Yan Yan, Ruohan Fu, Hong Wang, Tatsutoshi Nakahata, Xiuli An, Lihong Shi, Yonggang Zhang, Feng Ma

Insulin is expressed by enteroendocrine cells during human fetal development
Adi Egozi, Dhana Llivichuzhca-Loja, Blake McCourt, Lydia Farack, Xiaojing An, Fujing Wang, Kong Chen, Liza Konnikova, Shalev Itzkovitz

Fish patterns from Owen, et al.

Differential growth is a critical determinant of zebrafish pigment pattern formation
Jennifer P. Owen, Christian A. Yates, Robert N. Kelsh

Quail embryos from Barzilai-Tutsch, et al.

TopFlash transgenic quail reveals dynamic TCF/β-catenin signaling during avian embryonic development
Hila Barzilai-Tutsch, Valerie Morin, Gauthier Toulouse, Stephen Firth, Christophe Marcelle, Olivier Serralbo

In vitro endoderm emergence and self-organisation in the absence of extraembryonic tissues and embryonic architecture
Stefano Vianello, Matthias P. Lutolf

Serotonergic neuron ribosomes regulate the neuroendocrine control of Drosophila development
Lisa P. Deliu, Deeshpaul Jadir, Abhishek Ghosh, Savraj S. Grewal

Differential repression of Otx2 underlies the capacity of NANOG and ESRRB to induce germline entry
Matúš Vojtek, Jingchao Zhang, Juanjuan Sun, Man Zhang, Ian Chambers

The ciliary gene INPP5E confers dorsal telencephalic identity to human cortical organoids by negatively regulating Sonic Hedgehog signalling
Leah Schembs, Ariane Willems, Kerstin Hasenpusch-Theil, James D. Cooper, Katie Whiting, Karen Burr, Sunniva M.K. Bøstrand, Bhuvaneish T. Selvaraj, Siddharthan Chandran, Thomas Theil

Naa12 compensates for Naa10 in mice in the amino-terminal acetylation pathway
Hyae Yon Kweon, Mi-Ni Lee, Max Dörfel, Seungwoon Seo, Leah Gottlieb, Thomas Papazyan, Nina McTiernan, Rasmus Ree, David Bolton, Andrew Garcia, Michael Flory, Jonathan Crain, Alison Sebold, Scott Lyons, Ahmed Ismail, Elaine Marchi, Seong-keun Sonn, Se-Jin Jeong, Sejin Jeon, Shinyeong Ju, Simon J. Conway, TaeSoo Kim, Hyun-Seok Kim, Cheolju Lee, Tae-Young Roh, Thomas Arnesen, Ronen Marmorstein, Goo Taeg Oh, Gholson J. Lyon

Mouse neurons from Wen, et al.

Developmental Regulation of Homeostatic Plasticity in Mouse Primary Visual Cortex
Wei Wen, Gina G. Turrigiano

Patterning on the move: the effects of Hh morphogen source movement on signaling dynamics
D. G. Míguez, A. Iannini, D. García-Morales, F. Casares

Preterm birth alters the development of cortical microstructure and morphology at term-equivalent age
Ralica Dimitrova, Maximilian Pietsch, Judit Ciarrusta, Sean P. Fitzgibbon, Logan Z. J. Williams, Daan Christiaens, Lucilio Cordero-Grande, Dafnis Batalle, Antonios Makropoulos, Andreas Schuh, Anthony N. Price, Jana Hutter, Rui PAG Teixeira, Emer Hughes, Andrew Chew, Shona Falconer, Olivia Carney, Alexia Egloff, J-Donald Tournier, Grainne McAlonan, Mary A. Rutherford, Serena J. Counsell, Emma C. Robinson, Joseph V. Hajnal, Daniel Rueckert, A. David Edwards, Jonathan O’Muircheartaigh

Connectomes across development reveal principles of brain maturation
Daniel Witvliet, Ben Mulcahy, James K. Mitchell, Yaron Meirovitch, Daniel R. Berger, Yuelong Wu, Yufang Liu, Wan Xian Koh, Rajeev Parvathala, Douglas Holmyard, Richard L. Schalek, Nir Shavit, Andrew D. Chisholm, Jeff W. Lichtman, Aravinthan D.T. Samuel, Mei Zhen

Fly eyes from Mishra, et al.

Homothorax Controls a Binary Rhodopsin Switch in Drosophila Ocelli
Abhishek Kumar Mishra, Cornelia Fritsch, Roumen Voutev, Richard S. Mann, Simon G. Sprecher

Zebrafish in Hasan, et al.

Genetic and chemical inhibition of autophagy in zebrafish induced myeloproliferation
Kazi Md Mahmudul Hasan, Xiang-Ke Chen, Zhen-Ni Yi, Jack Jark-Yin Lau, Alvin Chun-hang Ma

The asymmetric Pitx2 regulates intestinal muscular-lacteal development and protects against fatty liver disease
Shing Hu, Aparna Mahadevan, Isaac F. Elysee, Joseph Choi, Nathan R. Souchet, Gloria H. Bae, Alessandra K. Taboada, Gerald E. Duhamel, Carolyn S. Sevier, Ge Tao, Natasza A. Kurpios

The atypical RNA-binding protein TAF15 regulates dorsoanterior neural development through diverse mechanisms in Xenopus tropicalis
Caitlin S. DeJong, Darwin S. Dichmann, Cameron R. T. Exner, Yuxiao Xu, Richard M. Harland

Estrogen regulates early embryonic development of the olfactory sensory system via estrogen-responsive glia
Aya Takesono, Paula Schirrmacher, Aaron Scott, Jon M. Green, Okhyun Lee, Matthew J. Winter, Tetsuhiro Kudoh, Charles R. Tyler

Midbrain organoids from Kim, et al.

Chronic opioid treatment arrests neurodevelopment and alters synaptic activity in human midbrain organoids
Hye Sung Kim, Yang Xiao, Xuejing Chen, Siyu He, Jongwon Im, Moshe J. Willner, Michael O. Finlayson, Cong Xu, Huixiang Zhu, Se Joon Choi, Eugene V. Mosharov, Hae-Won Kim, Bin Xu, Kam W. Leong

Polypeptides IGF-1C and P24 synergistically promote osteogenic differentiation of bone marrow mesenchymal stem cells in vitro through the p38 and JNK signaling pathways
Gaoying Ran, Wei Fang, Lifang Zhang, Yuting Peng, Jiatong Li, Xianglong Ding, Shuguang Zeng, Yan He

Amyloid precursor protein localises to ependymal cilia in vertebrates and is required for ciliogenesis and brain development in zebrafish
Jasmine Chebli, Maryam Rahmati, Tammaryn Lashley, Birgitta Edeman, Anders Oldfors, Henrik Zetterberg, Alexandra Abramsson

Notch pathway is required for protection against heat-stress in spermatogonial stem cells
Omar D. Moreno Acosta, Agustín F. Boan, Ricardo S. Hattori, Juan I. Fernandino

In vivo imaging of mammary epithelial cell dynamics in response to lineage-biased Wnt/β-catenin activation
Bethan Lloyd-Lewis, Francesca Gobbo, Meghan Perkins, Guillaume Jacquemin, Marisa M Faraldo, Silvia Fre

Apoptotic Find-me Signals are an Essential Driver of Stem Cell Conversion To The Cardiac Lineage
Loic Fort, Vivian Gama, Ian G. Macara

Notch-dependent Abl signaling regulates cell motility during ommatidial rotation in Drosophila
Yildiz Koca, Linh T. Vuong, Jaskirat Singh, Edward Giniger, Marek Mlodzik

EXC-4/CLIC, Gα, and Rho/Rac signaling regulate tubulogenesis in C. elegans
Anthony F. Arena, Daniel D. Shaye

Release of Notch activity coordinated by IL-1β signalling confers differentiation plasticity of airway progenitors via Fosl2 during alveolar regeneration
Jinwook Choi, Yu Jin Jang, Catherine Dabrowska, Elhadi Iich, Kelly V. Evans, Helen Hall, Sam M. Janes, Benjamin D. Simons, Bon-Kyoung Koo, Jonghwan Kim, Joo-Hyeon Lee

Reciprocal EGFR signaling in the Anchor Cell ensures precise inter-organ connection during C. elegans vulval morphogenesis
Silvan Spiri, Simon Berger, Louisa Mereu, Andrew DeMello, Alex Hajnal

Synergistic TOR and ERK inhibition mitigates the hereditary haemorrhagic telangiectasia-like phenotype and excess kugel formation in endoglin mutant zebrafish
Ryan O. Snodgrass, Helen M. Arthur, Timothy J.A. Chico

Gene-teratogen interactions influence the penetrance of birth defects by altering Hedgehog signaling strength
Jennifer H. Kong, Cullen B. Young, Ganesh V. Pusapati, F Hernán Espinoza, Chandni B. Patel, Francis Beckert, Sebastian Ho, Bhaven B. Patel, George C. Gabriel, L. Aravind, J Fernando Bazan, Teresa M. Gunn, Cecilia W. Lo, Rajat Rohatgi

GLI transcriptional repression is inert prior to Hedgehog pathway activation
Rachel K. Lex, Weiqiang Zhou, Zhicheng Ji, Kristin N. Falkenstein, Kaleigh E. Schuler, Kathryn E. Windsor, Joseph D. Kim, Hongkai Ji, Steven A Vokes

FGF/ERK autocrine signaling is enhanced by NANOG in a subpopulation of pluripotent stem cells to execute autoregulation and induce heterogeneity
Hanuman T Kale, Rajendra Singh Rajpurohit, Debabrata Jana, Vishnu V Vijay, Mansi Srivastava, Preeti R Mourya, Gunda Srinivas, P Chandra Shekar

Neuronal KGB-1 JNK MAPK signaling regulates the dauer developmental decision in response to environmental stress in C. elegans
Deepshikha Dogra, Warakorn Kulalert, Frank C. Schroeder, Dennis H. Kim

Self-Organogenesis from 2D Micropatterns to 3D Biomimetic Biliary Trees

Emilie Gontran, Lorena Loarca, Cyrille El Khassis, Latifa Bouzhir, Dmitry Ayollo, Elsa Mazari-Arrighi, Alexandra Fuchs, Pascale Dupuis-Williams

| Morphogenesis & mechanics

Differential adhesion regulates neurite placement via a retrograde zippering mechanism
Titas Sengupta, Noelle L. Koonce, Mark W. Moyle, Leighton H. Duncan, Nabor Vázquez-Martínez, Sarah E. Emerson, Xiaofei Han, Lin Shao, Yicong Wu, Anthony Santella, Li Fan, Zhirong Bao, William A. Mohler, Hari Shroff, Daniel A. Colón-Ramos

Acetylated microtubules are required for maintenance of the barrier between two adjacent tissues
Matthew Antel, Taylor Simao, Muhammed Burak Bener, Mayu Inaba

A midbody component homolog, too much information/prc1-like, is required for microtubule reorganization during both cytokinesis and axis induction in the early zebrafish embryo
S Nair, E.L. Welch, C.E. Moravec, R.L. Trevena, F. Pelegri

Pak1 and PP2A antagonize aPKC function to support cortical tension induced by the Crumbs-Yurt complex
Cornélia Biehler, Katheryn E. Rothenberg, Alexandra Jetté, Hélori-Mael Gaudé, Rodrigo Fernandez-Gonzalez, Patrick Laprise

Tracking cells in Xenopus, in Christodoulou and Skourides

Distinct spatiotemporal contribution of morphogenetic events and mechanical tissue coupling during Xenopus neural tube closure
Neophytos Christodoulou, Paris A. Skourides

The worm brush border in Bidaud-Meynard, et al.

High resolution dynamic mapping of the C. elegans intestinal brush border
Aurélien Bidaud-Meynard, Flora Demouchy, Ophélie Nicolle, Anne Pacquelet, Grégoire Michaux

Met-signaling Controls Dendritic Cell Migration by Regulating Podosome Formation and Function
Ahmed E.I. Hamouda, Carmen Schalla, Antonio Sechi, Martin Zenke, Thomas Hieronymus

Cell-matrix adhesion contributes to permeability control in human colon organoids
James Varani, Shannon D. McClintock, Muhammad N. Aslam

Cell adhesions link subcellular actomyosin dynamics to tissue scale force production during vertebrate convergent extension
Robert J. Huebner, Shinuo Weng, Chanjae Lee, Sena Sarıkaya, Ophelia Papoulas, Rachael M. Cox, Edward M. Marcotte, John B. Wallingford

Convergent extension requires adhesion-dependent biomechanical integration of cell crawling and junction contraction
Shinuo Weng, Robert J. Huebner, John B. Wallingford

A new approach to measure forces at junction vertices in an epithelium
Clémentine Villeneuve, Samuel Mathieu, Emilie Lagoutte, Bruno Goud, Philippe Chavrier, Jean-Baptiste Manneville, Carine Rossé

Pressure and curvature control of contact inhibition in epithelia growing under spherical confinement
Ilaria Di Meglio, Anastasiya Trushko, Pau Guillamat, Carles Blanch-Mercader, Aurélien Roux

Mechanical Stimulation via Muscle Activity is Necessary for the Maturation of Tendon Multiscale Mechanics during Embryonic Development
Benjamin E Peterson, Rebecca A. Rolfe, Allen Kunselman, Paula Murphy, Spencer E. Szczesny

Laminin-binding Integrins Regulate Angiogenesis by Distinct and Overlapping Mechanisms in Organotypic Cell Culture Models
Hao Xu, Susan E LaFlamme

Bmper is required for morphogenesis of the anterior and posterior semicircular canal ducts in the developing zebrafish inner ear
Sarah Baxendale, Esther C. Maier, Nikolaus D. Obholzer, Sarah Burbridge, Joseph Zinski, Francesca B. Tuazon, Nicholas J. van Hateren, M. Montserrat Garcia Romero, Mar Marzo, Kazutomo Yokoya, Robert D. Knight, Sean G. Megason, Mary C. Mullins, Tanya T. Whitfield

Characterisation of the transcriptional dynamics underpinning the function, fate, and migration of the mouse Anterior Visceral Endoderm
Shifaan Thowfeequ, Jonathan Fiorentino, Di Hu, Maria Solovey, Sharon Ruane, Maria Whitehead, Bart Vanhaesebroeck, Antonio Scialdone, Shankar Srinivas

IFT20 is critical for early chondrogenesis during endochondral ossification
Hiroyuki Yamaguchi, Megumi Kitami, Karin H. Uchima Koecklin, Li He, Jianbo Wang, Daniel S. Perrien, William R. Lagor, Yoshihiro Komatsu

| Genes & genomes

Caenorhabditis elegans ETR-1/CELF has broad effects on the muscle cell transcriptome, including genes that regulate translation and neuroblast migration
Matthew E. Ochs, Rebecca McWhirter, Rob Unckless, David Miller, Erik A Lundquist

A comprehensive series of temporal transcription factors in the fly visual system
Nikolaos Konstantinides, Anthony M. Rossi, Aristides Escobar, Liébaut Dudragne, Yen-Chung Chen, Thinh Tran, Azalia Martinez Jaimes, Mehmet Neset Özel, Félix Simon, Zhiping Shao, Nadejda M. Tsankova, John F. Fullard, Uwe Walldorf, Panos Roussos, Claude Desplan

Expansion of RNA sequence diversity and RNA editing rates throughout human cortical development
Ryn Cuddleston, Laura Sloofman, Lindsay Liang, Enrico Mossotto, Xuanjia Fan, Minghui Wang, Bin Zhang, Jiebiao Wang, Nenad Sestan, Bernie Devlin, Kathryn Roeder, Joseph D. Buxbaum, Stephan J. Sanders, Michael S. Breen

Mouse heads from Van Otterloo, et al.

AP-2α and AP-2β cooperatively function in the craniofacial surface ectoderm to regulate chromatin and gene expression dynamics during facial development
Eric Van Otterloo, Isaac Milanda, Hamish Pike, Hong Li, Kenneth L Jones, Trevor Williams

Fly brains from Zhu, et al.

A comprehensive temporal patterning gene network in Drosophila medulla neuroblasts revealed by single-cell RNA sequencing
Hailun Zhu, Sihai Dave Zhao, Alokananda Ray, Yu Zhang, Xin Li

Heterogeneity and molecular programming of progenitors for motor neurons and oligodendrocytes
Lingyan Xing, Rui Chai, Jiaqi Wang, Jiaqi Lin, Hanyang Li, Yueqi Wang, Biqin Lai, Junjie Sun, Gang Chen

A DNA Replication-Independent Function of the pre-Replication Complex during Cell Invasion in C. elegans
Evelyn Lattmann, Ting Deng, Michael Walser, Patrizia Widmer, Charlotte Rexha-Lambert, Vibhu Prasad, Ossia Eichhoff, Michael Daube, Reinhard Dummer, Mitchell P. Levesque, Alex Hajnal

CDK12 is Necessary to Promote Epidermal Differentiation through Transcription Elongation
Jingting Li, Manisha Tiwari, Yifang Chen, George L. Sen

Systematic reconstruction of the cellular trajectories of mammalian embryogenesis
Chengxiang Qiu, Junyue Cao, Tony Li, Sanjay Srivatsan, Xingfan Huang, Diego Calderon, William Stafford Noble, Christine M. Disteche, Malte Spielmann, Cecilia B. Moens, Cole Trapnell, Jay Shendure

Identification of enamel knot gene signature within the developing mouse molar
Emma Wentworth Winchester, Justin Cotney

Oligodendrocyte maps from Hilscher, et al.

Spatial cell type mapping of the oligodendrocyte lineage in the mouse juvenile and adult CNS with in situ sequencing
Markus M. Hilscher, Christoffer Mattsson Langseth, Petra Kukanja, Chika Yokota, Mats Nilsson, Gonçalo Castelo-Branco

Application of ATAC-Seq for genome-wide analysis of the chromatin state at single myofiber resolution
Korin Sahinyan, Darren M Blackburn, Marie-Michelle Simon, Felicia Lazure, Tony Kwan, Guillaume Bourque, Vahab D Soleimani

Fly spermatids from Rivard, et al.

A putative de novo evolved gene required for spermatid chromatin condensation in Drosophila melanogaster
Emily L. Rivard, Andrew G. Ludwig, Prajal H. Patel, Anna Grandchamp, Sarah E. Arnold, Alina Berger, Emilie M. Scott, Brendan J. Kelly, Grace C. Mascha, Erich Bornberg-Bauer, Geoffrey D. Findlay

Klf5 establishes bi-potential cell fate by dual regulation of ICM and TE specification genes
Martin Kinisu, Yong Jin Choi, Claudia Cattoglio, Ke Liu, Hector Roux de Bezieux, Raeline Valbuena, Nicole Pum, Sandrine Dudoit, Haiyan Huang, Zhenyu Xuan, Sang Yong Kim, Lin He

C. elegans TFIIH subunit GTF-2H5/TTDA is a non-essential transcription factor indispensable for DNA repair
Karen L. Thijssen, Melanie van der Woude, Carlota Davó-Martínez, Mariangela Sabatella, Wim Vermeulen, Hannes Lans

Adult fibroblasts retain organ-specific transcriptomic identity
Elvira Forte, Mirana Ramialison, Hieu T. Nim, Madison Mara, Rachel Cohn, Sandra L. Daigle, Sarah Boyd, J. Travis Hinson, Mauro W. Costa, Nadia A. Rosenthal, Milena B. Furtado

Differential Bcd activation of two hunchback promoters emerges from unified kinetics of enhancer-promoter interaction
Jingyao Wang, Shihe Zhang, Hongfang Lu, Heng Xu

Hypothalamic neurons from Cabera Zapata, et al.

X-Linked Histone H3K27 Demethylase Kdm6a Regulates Sexually Dimorphic Differentiation of Hypothalamic Neurons
Lucas E. Cabrera Zapata, Carla D. Cisternas, Camila Sosa, Maria Angeles Arevalo, Luis Miguel Garcia-Segura, María Julia Cambiasso

Environment-driven reprogramming of gamete DNA methylation occurs during maturation and is transmitted intergenerationally in salmon
Kyle Wellband, David Roth, Tommi Linnansaari, R. Allen Curry, Louis Bernatchez

ETV2 primes hematoendothelial gene enhancers prior to hematoendothelial fate commitment
Jeffrey D. Steimle, Chul Kim, Rangarajan D. Nadadur, Zhezhen Wang, Andrew D. Hoffmann, Erika Hanson, Junghun Kweon, Tanvi Sinha, Kyunghee Choi, Brian L. Black, John M. Cunningham, Kohta Ikegami, Ivan P. Moskowitz

Multivariate genome-wide association studies on the tissue compartments of human brain identify novel loci underpinning brain development and neuropsychiatric outcomes
Chun Chieh Fan, Robert Loughnan, Carolina Makowski, Diliana Pechva, Chi-Hua Chen, Donald Hagler, Wesley K. Thompson, Dennis van der Meer, Oleksandr Frei, Ole Andreassen, Anders M. Dale

Chromatin dynamics during hematopoiesis reveal discrete regulatory modules instructing differentiation
Grigorios Georgolopoulos, Nikoletta Psatha, Mineo Iwata, Andrew Nishida, Tannishtha Som, Minas Yiangou, John A. Stamatoyannopoulos, Jeff Vierstra

H3K9 tri-methylation at Nanog times differentiation commitment and enables the acquisition of primitive endoderm fate
A. Dubois, L. Vincenti, A. Chervova, S. Vandormael-Pournin, M. Cohen-Tannoudji, P. Navarro

Deconvolution of the epigenetic age discloses distinct inter-personal variability in epigenetic aging patterns
Tamar Shahal, Elad Segev, Thomas Konstantinovsky, Yonit Marcus, Gabi Shefer, Metsada Pasmanik-Chor, Assaf Buch, Yuval Ebenstein, Paul Zimmet, Naftali Stern

The Caenorhabditis elegans TDRD5/7-like protein, LOTR-1, interacts with the helicase ZNFX-1 to balance epigenetic signals in the germline
Elisabeth A. Marnik, Miguel V. Almeida, P. Giselle Cipriani, George Chung, Edoardo Caspani, Emil Karaulanov, Falk Butter, Catherine S. Sharp, John Zinno, Hin Hark Gan, Fabio Piano, René F Ketting, Kristin C. Gunsalus, Dustin L. Updike

Blm Helicase Facilitates Rapid Replication of Repetitive DNA Sequences in early Drosophila Development
Jolee M. Ruchert, Morgan M Brady, Susan McMahan, Karly J. Lacey, Leigh C. Latta, Jeff Sekelsky, Eric P. Stoffregen

Translesion DNA synthesis-driven mutagenesis in very early embryogenesis of fast cleaving embryos
Elena Lo Furno, Isabelle Busseau, Claudio Lorenzi, Cima Saghira, Matt C Danzi, Stephan Zuchner, Domenico Maiorano

Identification of PAX6 and NFAT4 as the transcriptional regulators of lncRNA Mrhl in neuronal progenitors
Debosree Pal, Sangeeta Dutta, Dhanur P Iyer, Utsa Bhaduri, M.R.S Rao

Conserved Transcription Factors Control Chromatin Accessibility and Gene Expression to Maintain Cell Fate Stability and Restrict Reprogramming of Differentiated Cells
Maria A. Missinato, Sean A. Murphy, Michaela Lynott, Anaïs Kervadec, Michael S. Yu, Yu-Ling Chang, Suraj Kannan, Mafalda Loreti, Christopher Lee, Prashila Amatya, Hiroshi Tanaka, Chun-Teng Huang, Pier Lorenzo Puri, Chulan Kwon, Peter D. Adams, Li Qian, Alessandra Sacco, Peter Andersen, Alexandre R. Colas

ZFP462 targets heterochromatin to transposon-derived enhancers restricting transcription factor binding and expression of lineage-specifying genes
Ramesh Yelagandula, Karin Stecher, Maria Novatchkova, Luca Michetti, Georg Michlits, Jingkui Wang, Pablo Hofbauer, Carina Pribitzer, Gintautas Vainorius, Luke Isbel, Sasha Mendjan, Dirk Schübeler, Ulrich Elling, Julius Brennecke, Oliver Bell

MicroRNA-202 prevents precocious spermatogonial differentiation and meiotic initiation during mouse spermatogenesis
Jian Chen, Chenxu Gao, Xiwen Lin, Yan Ning, Wei He, Chunwei Zheng, Daoqin Zhang, Lin Yan, Binjie Jiang, Yuting Zhao, Md Alim Hossen, Chunsheng Han

RNAseq analysis reveals dynamic metaboloepigenetic profiles of human, mouse and bovine pre-implantation embryos
Marcella Pecora Milazzotto, Michael James Noonan, Marcia de Almeida Monteiro Melo Ferraz

| Stem cells, regeneration & disease modelling

The NF-κB pathway regulates heterochromatin at intronic young LINE-1 elements and hematopoietic stem cell gene expression during irradiation stress
Yanis Pelinski, Donia Hidaoui, François Hermetet, Anne Stolz, M’boyba Khadija Diop, Amir M. Chioukh, Françoise Porteu, Emilie Elvira-Matelot

Planarian in situs from Wong, et al.

Intestine-enriched apolipoprotein b orthologs are required for stem cell differentiation and regeneration in planarians
Lily L. Wong, Christina G. Bruxvoort, Nicholas I. Cejda, Jannette Rodriguez Otero, David J. Forsthoefel

Proliferation maintains the undifferentiated status of stem cells: the role of the planarian cell cycle regulator Cdh1
Yuki Sato, Yoshihiko Umesono, Yoshihito Kuroki, Kiyokazu Agata, Chikara Hashimoto

Investigation of Thyroid Hormone Associated Gene-Regulatory Networks during Hepatogenesis using an Induced Pluripotent Stem Cell based Model
Audrey Ncube, Nina Graffmann, Jan Greulich, Bo Scherer, Wasco Wruck, James Adjaye

Human enteroids from Zeve, et al.

Robust differentiation of human enteroendocrine cells from intestinal stem cells
Daniel Zeve, Eric Stas, Joshua de Sousa Casal, Prabhath Mannam, Wanshu Qi, Xiaolei Yin, Sarah Dubois, Manasvi S. Shah, Erin P. Syverson, Sophie Hafner, Jeffrey M. Karp, Diana L. Carlone, Jose Ordovas-Montanes, David T. Breault

ES cells from Vojtek & Chambers

Loss of Resf1 reduces the efficiency of embryonic stem cell self-renewal and germline entry
Matúš Vojtek, Ian Chambers

Hypertrophic Chondrocytes Serve as a Reservoir for Unique Marrow Associated Skeletal Stem and Progenitor Cells, Osteoblasts, and Adipocytes During Skeletal Development
Jason T. Long, Abigail Leinroth, Yihan Liao, Yinshi Ren, Anthony J. Mirando, Tuyet Nguyen, Wendi Guo, Deepika Sharma, Colleen Wu, Kathryn Song Eng Cheah, Courtney M. Karner, Matthew J. Hilton

Identification of SUMO targets required to maintain human stem cells in the pluripotent state
Barbara Mojsa, Michael H. Tatham, Lindsay Davidson, Magda Liczmanska, Emma Branigan, Ronald T. Hay

Stem cell therapy for skin regeneration using mesenchymal stem cells derived from the progeroid Werner syndrome-specific iPS cells
Shinichiro Funayama, Hisaya Kato, Hiyori Kaneko, Kentaro Kosaka, Daisuke Sawada, Aki Takada-Watanabe, Takuya Minamizuka, Yusuke Baba, Masaya Koshizaka, Akira Shimamoto, Yasuo Ouchi, Atsushi Iwama, Yusuke Endo, Naoya Takayama, Koji Eto, Yoshiro Maezawa, Koutaro Yokote

Gene expression heatmaps from Sinigaglia, et al.

Distinct gene expression dynamics in developing and regenerating limbs
Chiara Sinigaglia, Alba Almazan, Marie Semon, Benjamin Gillet, Sandrine Hughes, Eric Edsinger, Michalis Averof, Mathilde Paris

Single cell chronoatlas of regenerating mouse livers reveals early Kupffer cell proliferation
Daniel Sánchez-Taltavull, Tess Brodie, Joel Zindel, Noëlle Dommann, Bas G.J. Surewaard, Adrian Keogh, Nicolas Mélin, Isabel Büchi, Riccardo Tombolini, Paul Kubes, Daniel Candinas, Guido Beldi, Deborah Stroka

Vestibular and auditory hair cell regeneration following targeted ablation of hair cells with diphtheria toxin in zebrafish
Erin Jimenez, Claire C. Slevin, Luis Colón-Cruz, Shawn M. Burgess

Notch signaling via Hey1 and Id2b regulates Müller glia’s regenerative response to retinal injury
Aresh Sahu, Sulochana Devi, Jonathan Jui, Daniel Goldman

Anal skin-like epithelium mediates colonic wound healing
Cambrian Y. Liu, Nandini Girish, Marie L. Gomez, Philip E. Dubé, M. Kay Washington, Benjamin D. Simons, D. Brent Polk

Zebrafish retinas from Lu, et al.

mTOR activity is essential for retinal pigment epithelium regeneration in zebrafish
Fangfang Lu, Lyndsay L. Leach, Jeffrey M. Gross

Potential therapy for progressive vision loss due to PCDH15-associated Usher Syndrome developed in an orthologous Usher mouse
Saumil Sethna, Wadih M. Zein, Sehar Riaz, Arnaud P. J. Giese, Julie M. Schultz, Todd Duncan, Robert B. Hufnagel, Carmen C. Brewer, Andrew J. Griffith, T. Michael Redmond, Saima Riazuddin, Thomas B. Friedman, Zubair M. Ahmed

A functional network signature in the developing cerebellum: evidence from a preclinical model of autism
María Berenice Soria-Ortiz, Atáulfo Martínez Torres, Daniel Reyes-Haro

Pharmacological inhibition of the VCP/proteasome axis rescues photoreceptor degeneration in RHOP23H rat retinal explants
Merve Sen, Oksana Kutsyr, Bowen Cao, Sylvia Bolz, Blanca Arango-Gonzalez, Marius Ueffing

Dissecting the molecular basis of human interneuron migration in forebrain assembloids from Timothy syndrome
Fikri Birey, Min-Yin Li, Aaron Gordon, Mayuri Thete, Alfredo M Valencia, Omer Revah, Anca M Pasca, Daniel H Geschwind, Sergiu P Pasca

Chick embryos from Wilerding, et al.

Sustained experimental activation of FGF8/ERK in the developing chicken spinal cord reproducibly models early events in ERK-mediated tumorigenesis
Axelle Wilmerding, Lauranne Bouteille, Nathalie Caruso, Ghislain Bidaut, Heather Etchevers, Yacine Graba, Marie-Claire Delfini

Mutations in SIX1 associated with Branchio-oto-renal Syndrome (BOR) differentially affect otic expression of putative target genes
Tanya Mehdizadeh, Himani Datta Majumdar, Sarah Ahsan, Andre Tavares, Sally A. Moody

Comparative therapeutic strategies for preventing aortic rupture in a mouse model of vascular Ehlers Danlos syndrome
Anne Legrand, Charline Guery, Julie Faugeroux, Erika Fontaine, Carole Beugnon, Amélie Gianfermi, Irmine Loisel-Ferreira, Marie-Christine Verpont, Salma Adham, Tristan Mirault, Juliette Hadchouel, Xavier Jeunemaitre

Mouse embryos from Manivannan, et al.

Maternal hyperglycemia impedes second heart field-derived cardiomyocyte differentiation to elevate the risk of congenital heart defects
Sathiyanarayanan Manivannan, Corrin Mansfield, Xinmin Zhang, Karthik M. Kodigepalli, Uddalak Majumdar, Vidu Garg, Madhumita Basu

Analysis of CHD-7 defective dauer nematodes implicates collagen misregulation in CHARGE syndrome features
Diego M. Jofré, Dane K. Hoffman, Ailen S. Cervino, McKenzie Grundy, Sijung Yun, Francis RG. Amrit, Donna B. Stolz, Esteban Salvatore, Fabiana A. Rossi, Arjumand Ghazi, M. Cecilia Cirio, Judith L. Yanowitz, Daniel Hochbaum

Identifying developing interneurons as a potential target for multiple genetic autism risk factors in human and rodent forebrain
Yifei Yang, Sam A. Booker, James M. Clegg, Idoia Quintana Urzainqui, Anna Sumera, Zrinko Kozic, Owen Dando, Sandra Martin Lorenzo, Yann Herault, Peter C. Kind, David J. Price, Thomas Pratt

BRN2 and PTN unveil multiple neurodevelopmental mechanisms in Schizophrenia patient-derived cerebral organoids
Michael Notaras, Aiman Lodhi, Friederike Dundar, Paul Collier, Nicole Sayles, Hagen Tilgner, David Greening, Dilek Colak

Inter- and intrapopulational heterogeneity of characteristic markers in adult human neural crest-derived stem cells
Beatrice A. Windmöller, Anna L. Höving, Johannes F.W. Greiner

PpRPK2 modulates auxin homeostasis and transport to specify stem cell identity and plant shape in the moss Physcomitrella
Zoe Nemec Venza, Connor Madden, Amy Stewart, Wei Liu, Ondřej Novák, Aleš Pěnčík, Andrew C. Cuming, Yasuko Kamisugi, C. Jill Harrison

Fetal-like reversion in the regenerating intestine is regulated by mesenchymal Asporin
Sharif Iqbal, Simon Andersson, Ernesta Nestaite, Nalle Pentinmikko, Ashish Kumar, Daniel Borshagovski, Anna Webb, Tuure Saarinen, Anne Juuti, Alessandro Ori, Markku Varjosalo, Kirsi H. Pietiläinen, Kim B. Jensen, Menno Oudhoff, Pekka Katajisto

A single-cell atlas of de novo β-cell regeneration reveals the contribution of hybrid β/δ cells to diabetes recovery in zebrafish
Sumeet Pal Singh, Prateek Chawla, Alisa Hnatiuk, Margrit Kamel, Luis Delgadillo Silva, Bastiaan Spanjard, Sema Elif Eski, Sharan Janjuha, Pedro Olivares, Oezge Kayisoglu, Fabian Rost, Juliane Bläsche, Annekathrin Kränkel, Andreas Petzold, Thomas Kurth, Susanne Reinhardt, Jan Philipp Junker, Nikolay Ninov

Modular, Cascade-like Transcriptional Program of Regeneration in Stentor
Pranidhi Sood, Athena Lin, Rebecca McGillivary, Wallace F. Marshall

A multimodal iPSC platform for cystic fibrosis drug testing
Andrew Berical, Rhianna E. Lee, Junjie Lu, Mary Lou Beermann, Jake A. LeSeur, Aditya Mithal, Dylan Thomas, Nicole Ranallo, Megan Peasley, Alex Stuffer, Jan Harrington, Kevin Coote, Killian Hurley, Paul McNally, Gustavo Mostovslavsky, John Mahoney, Scott H. Randell, Finn J. Hawkins

Dystonia-specific mutations in THAP1 alter transcription of genes associated with neurodevelopment and myelin
Aloysius Domingo, Rachita Yadav, Shivangi Shah, William T. Hendriks, Serkan Erdin, Dadi Gao, Kathryn O’Keefe, Benjamin Currall, James F. Gusella, Nutan Sharma, Laurie J. Ozelius, Michelle E. Ehrlich, Michael E. Talkowski, D. Cristopher Bragg

Inhibition of N-myristoyltransferase Promotes Naive Pluripotency in Mouse and Human Pluripotent Stem Cells
Junko Yoshida, Hitomi Watanabe, Kaori Yamauchi, Takumi Nishikubo, Ayako Isotani, Satoshi Ohtsuka, Hitoshi Niwa, Hidenori Akutsu, Akihiro Umezawa, Hirofumi Suemori, Yasuhiro Takashima, Gen Kondoh, Junji Takeda, Kyoji Horie

The extracellular matrix controls stem cell specification and crypt morphology in the developing and adult gut
R. Ramadan, SM. van Neerven, VM. Wouters, T. Martins Garcia, V. Muncan, OD. Franklin, M. Battle, KS. Carlson, J. Leach, OJ. Sansom, L. Vermeulen, JP. Medema, DJ. Huels

Stem cell-free therapy for glaucoma to preserve vision
Ajay Kumar, Xiong Siqi, Minwen Zhou, Wen Chen, Enzhi Yang, Andrew Price, Liang Le, Ying Zhang, Laurence Florens, Michael Washburn, Akshay Kumar, Yunshu Li, Yi Xu, Kira Lathrop, Katherine Davoli, Yuanyuan Chen, Joel S. Schuman, Ting Xie, Yiqin Du

Tfap2b specifies an embryonic melanocyte stem cell population that retains adult multi-fate potential
Alessandro Brombin, Daniel J. Simpson, Jana Travnickova, Hannah R. Brunsdon, Zhiqiang Zeng, Yuting Lu, Tamir Chandra, E. Elizabeth Patton

Single-cell RNA sequencing-based characterization of resident lung mesenchymal stromal cells in bronchopulmonary dysplasia
I. Mižíková, F. Lesage, C. Cyr-Depauw, D. P. Cook, M. Hurskainen, S.M. Hänninen, A. Vadivel, P. Bardin, S. Zhong, O. Carpen, B. C. Vanderhyden, B. Thébaud

Human iPSC-derived cerebral organoids model features of Leigh Syndrome and reveal abnormal corticogenesis
Alejandra I. Romero-Morales, Gabriella L. Robertson, Anuj Rastogi, Megan L. Rasmussen, Hoor Temuri, Gregory Scott McElroy, Ram Prosad Chakrabarty, Lawrence Hsu, Paula M. Almonacid, Bryan A. Millis, Navdeep S. Chandel, Jean-Philippe Cartailler, Vivian Gama

Zebrafish pigment cells develop directly from persistent highly multipotent progenitors
Masataka Nikaido, Tatiana Subkhankulova, Leonid A. Uroshlev, Artem J. Kasianov, Karen Camargo Sosa, Gemma Bavister, Xueyan Yang, Frederico S. L. M. Rodrigues, Thomas J. Carney, Hartmut Schwetlick, Jonathan H.P. Dawes, Andrea Rocco, Vsevelod Makeev, Robert N. Kelsh

Basal neural stem cells drive postnatal neurogenesis whereas apical stem cells act as proliferation gatekeepers by regulating notch activation in the postnatal ventricular-subventricular zone
Katja Baur, Yomn Abdullah, Claudia Mandl, Gabriele Hoelzl-Wenig, Yan Shi, Udo Schmidt-Edelkraut, Priti Khatri, Francesca Ciccolini

The role of Kabuki Syndrome genes KMT2D and KDM6A in development: Analysis in Human sequencing data and compared to mice and zebrafish
Rwik Sen, Ezra Lencer, Elizabeth A. Geiger, Kenneth Jones, Tamim H. Shaikh, Kristin Bruk Artinger

Single-cell transcriptome analysis of embryonic and adult endothelial cells allows to rank the hemogenic potential of post-natal endothelium
Artem Adamov, Yasmin Natalia Serina Secanechia, Christophe Lancrin

Control of Arabidopsis shoot stem cell homeostasis by two antagonistic CLE peptide signalling pathways
Jenia Schlegel, Grégoire Denay, Karine Gustavo Pinto, Yvonne Stahl, Julia Schmid, Patrick Blümke, Rüdiger Simon

Skeletal dysplasia-causing TRPV4 mutations suppress the hypertrophic differentiation of human iPSC-derived chondrocytes
Amanda R. Dicks, Grigory I. Maksaev, Zainab Harissa, Alireza Savadipour, Ruhang Tang, Nancy Steward, Wolfgang Liedtke, Colin G. Nichols, Chia-Lung Wu, Farshid Guilak

ZFP541 is indispensable for pachytene progression by interacting with KCTD19 and activates meiotic gene expression in mouse spermatogenesis
Yushan Li, Ranran Meng, Shanze Li, Bowen Gu, Xiaotong Xu, Haihang Zhang, Tianyu Shao, Jiawen Wang, Yinghua Zhuang, Fengchao Wang

Generation of liver organoids from human induced pluripotent stem cells as liver fibrosis and steatosis models
Hoi Ying Tsang, Paulisally Hau Yi Lo, Kenneth Ka Ho Lee

Oncofetal protein CRIPTO regulates wound healing and fibrogenesis in regenerating liver and is associated with the initial stages of cardiac fibrosis
Sofia Karkampouna, Danny van der Helm, Bart van Hoek, Hein W Verspaget, Marie Jose TH Goumans, Minneke Coenraad, Boudewijn TH Kruithof, Marianna Kruithof-deJulio

Unbiased in vivo exploration of nuclear bodies-enhanced sumoylation reveals that PML orchestrates embryonic stem cell fate
Sarah Tessier, Omar Ferhi, Marie-Claude Geoffroy, Roman Gonzalez-Prieto, Antoine Canat, Samuel Quentin, Marika Pla, Michiko Niwa-Kawakita, Pierre Bercier, Domitille Rerolle, Pierre Therizols, Emmanuelle Fabre, Alfred C.O. Vertegaal, Hugues de The, Valerie Lallemand-Breitenbach

Distinct epicardial gene regulatory programmes drive development and regeneration of the zebrafish heart
Michael Weinberger, Filipa C. Simoes, Tatjana Sauka-Spengler, Paul R. Riley

Kidney organoids: A system to study human basement membrane assembly in health and disease
Mychel RPT Morais, Pinyuan Tian, Craig Lawless, Syed Murtuza-Baker, Louise Hopkinson, Steven Woods, Aleksandr Mironov, David A Long, Daniel Gale, Telma MT Zorn, Roy Zent, Rachel Lennon

Snail maintains the stem/progenitor state of skin epithelial cells and carcinomas through the autocrine effect of the matricellular protein Mindin
Krithika Badarinath, Binita Dam, Sunny Kataria, Ravindra K. Zirmire, Rakesh Dey, Randhir Singh, Tafheem A. Masudi, Janani Sambath, Prashanth Kumar, Akash Gulyani, You-Wen He, Sudhir Krishna, Colin Jamora

Glypican-6 deficiency causes dose-dependent conotruncal congenital heart malformations through abnormal remodelling of the endocardial cushions
Gennadiy Tenin, Alexander Crozier, Kathryn E. Hentges, Bernard Keavney

TLR4 regulation in human fetal membranes as an explicative mechanism of a pathological preterm case
Corinne Belville, Flora Ponelle-Chachuat, Marion Rouzaire, Christelle Gross, Bruno Pereira, Denis Gallot, Vincent Sapin, Loïc Blanchon

Cell-autonomous differentiation of human primed embryonic stem cells into trophoblastic syncytia through the nascent amnion-like cell state
Masatoshi Ohgushi, Mototsugu Eiraku

Intrinsic and extrinsic regulation of human fetal bone marrow haematopoiesis and perturbations in Down syndrome
Laura Jardine, Simone Webb, Issac Goh, Mariana Quiroga Londoño, Gary Reynolds, Michael Mather, Bayanne Olabi, Emily Stephenson, Rachel A. Botting, Dave Horsfall, Justin Engelbert, Daniel Maunder, Nicole Mende, Caitlin Murnane, Emma Dann, Jim McGrath, Hamish King, Iwo Kucinski, Rachel Queen, Christopher D Carey, Caroline Shrubsole, Elizabeth Poyner, Meghan Acres, Claire Jones, Thomas Ness, Rowan Coulthard, Natalina Elliott, Sorcha O’Byrne, Myriam L. R. Haltalli, John E Lawrence, Steven Lisgo, Petra Balogh, Kerstin B Meyer, Elena Prigmore, Kirsty Ambridge, Mika Sarkin Jain, Mirjana Efremova, Keir Pickard, Thomas Creasey, Jaume Bacardit, Deborah Henderson, Jonathan Coxhead, Andrew Filby, Rafiqul Hussain, David Dixon, David McDonald, Dorin-Mirel Popescu, Monika S. Kowalczyk, Bo Li, Orr Ashenberg, Marcin Tabaka, Danielle Dionne, Timothy L. Tickle, Michal Slyper, Orit Rozenblatt-Rosen, Aviv Regev, Sam Behjati, Elisa Laurenti, Nicola K. Wilson, Anindita Roy, Berthold Göttgens, Irene Roberts, Sarah A. Teichmann, Muzlifah Haniffa

Inflammatory blockade prevents injury to the developing pulmonary gas exchange surface in preterm primates
Andrea Toth, Shelby Steinmeyer, Paranthaman Kannan, Jerilyn Gray, Courtney M. Jackson, Shibabrata Mukherjee, Martin Demmert, Joshua R. Sheak, Daniel Benson, Joe Kitzmiller, Joseph A. Wayman, Pietro Presicce, Christopher Cates, Rhea Rubin, Kashish Chetal, Yina Du, Yifei Miao, Mingxia Gu, Minzhe Guo, Vladimir V. Kalinichenko, Suhas G. Kallapur, Emily R. Miraldi, Yan Xu, Daniel Swarr, Ian Lewkowich, Nathan Salomonis, Lisa Miller, Jennifer S. Sucre, Jeffrey A. Whitsett, Claire A. Chougnet, Alan H. Jobe, Hitesh Deshmukh, William J. Zacharias

Post-embryonic development and aging of the appendicular skeleton in Ambystoma mexicanum
Camilo Riquelme-Guzmán, Maritta Schuez, Alexander Böhm, Dunja Knapp, Sandra Edwards-Jorquera, Alberto S. Ceccarelli, Osvaldo Chara, Martina Rauner, Tatiana Sandoval-Guzmán

| Plant development

Populus ERF85 balances xylem cell expansion and secondary cell wall formation in hybrid aspen
Carolin Seyfferth, Bernard A Wessels, Jorma Vahala, Jaakko Kangasjarvi, Nicolas Delhomme, Torgeir R Hvidsten, Hannele Tuominen, Judith Felten

B1L regulates lateral root development by exocytic vesicular trafficking-mediated polar auxin transport in Arabidopsis
Gang Yang, Bi-xia Chen, Tao Chen, Jia-hui Chen, Rui Sun, Cong-cong Liu, Jiao Jia, Xiu-le Yue, Li-zhe An, Hua Zhang

Moss mitosis from Kozgunova, et al.

Spindle position dictates division site during asymmetric cell division in moss
Elena Kozgunova, Mari W. Yoshida, Ralf Reski, Gohta Goshima

Maize Brittle Stalk2-Like3, encoding a COBRA protein, functions in cell wall formation and carbohydrate partitioning
Benjamin T. Julius, Tyler J. McCubbin, Rachel A. Mertz, Nick Baert, Jan Knoblauch, DeAna G. Grant, Kyle Conner, Saadia Bihmidine, Paul Chomet, Ruth Wagner, Jeff Woessner, Karen Grote, Jeanette Peevers, Thomas L. Slewinski, Maureen C. McCann, Nicholas C. Carpita, Michael Knoblauch, David M. Braun

Protein turnover in the developing Triticum aestivum grain
Hui Cao, Owen Duncan, A. Harvey Millar

Arabidopss roots from Shukla, et al.

Suberin plasticity to developmental and exogenous cues is regulated by a set of MYB transcription factors
Vinay Shukla, Jian-Pu Han, Fabienne Cléard, Linnka Lefebvre- Legendre, Kay Gully, Paulina Flis, Alice Berhin, Tonni Grube Andersen, David E Salt, Christiane Nawrath, Marie Barberon

Endogenous RNA editing of a nuclear gene BOSS triggers flowering in tomato
Wenqian Wang, Jie Ye, Chuying Yu, Qingmin Xie, Xin Wang, Huiyang Yu, Jianwen Song, Changxing Li, Long Cui, Heyou Han, Changxian Yang, Hanxia Li, Yongen Lu, Taotao Wang, Yuyang Zhang, Junhong Zhang, Bo Ouyang, Zhibiao Ye

Transcriptomic analysis of temporal shifts in berry development between two grapevine cultivars of the Pinot family reveals potential genes controlling ripening time
Jens Theine, Daniela Holtgräwe, Katja Herzog, Florian Schwander, Anna Kicherer, Ludger Hausmann, Prisca Viehöver, Reinhard Töpfer, Bernd Weisshaar

Chromatin enrichment for Proteomics in Plants (ChEP-P) implicates the histone reader ALFIN-LIKE 6 in jasmonate signalling
Isabel Cristina Vélez-Bermúdez, Wolfgang Schmidt

Fern divisions from Wu, et al.

Timing of meristem initiation and maintenance determines the morphology of fern gametophytes
Xiao Wu, An Yan, Scott McAdam, Jo Ann Banks, Shaoling Zhang, Yun Zhou

The Genetic Architecture of Strawberry Yield and Fruit Quality Traits
Helen M. Cockerton, Amanda Karlström, Abigail W. Johnson, Bo Li, Eleftheria Stavridou, Katie J. Hopson, Adam B. Whitehouse, Richard J. Harrison

The REF6-dependent H3K27 demethylation establishes transcriptional competence to promote germination in Arabidopsis
Jie Pan, Huairen Zhang, Zhenping Zhan, Ting Zhao, Danhua Jiang

Arabidopsis seedlings from Cook, et al.

Regulation of suberin biosynthesis and Casparian strip development in the root endodermis by two plant auxins
Sam David Cook, Seisuke Kimura, Qi Wu, Rochus Benni Franke, Takehiro Kamiya, Hiroyuki Kasahara

Estimation of cell cycle kinetics in higher plant root meristem links organ position with cellular fate and chromatin structure
Taras Pasternak, Stefan Kircher, Klaus Palme

The VIL gene CRAWLING ELEPHANT controls maturation and differentiation in tomato via polycomb silencing
Ido Shwartz, Chen Yahav, Neta Kovetz, Alon Israeli, Maya Bar, Matan Levy, Katherine L. Duval, José M. Jiménez-Gómez, Roger B. Deal, Naomi Ori

A quantitative gibberellin signalling biosensor reveals a role for gibberellins in internode specification at the shoot apical meristem
Bihai Shi, Amelia Felipo-Benavent, Guillaume Cerutti, Carlos Galvan-Ampudia, Lucas Jilli, Geraldine Brunoud, Jérome Mutterer, Lali Sakvarelidze-Achard, Jean-Michel Davière, Alejandro Navarro-Galiano, Ankit Walia, Shani Lazary, Jonathan Legrand, Roy Weinstein, Alexander M. Jones, Salomé Prat, Patrick Achard, Teva Vernoux

Adaptive reprogramming during early seed germination requires temporarily enhanced fermentation – a critical role for alternative oxidase (AOX) regulation that concerns also microbiota effectiveness
Bharadwaj Revuru, Carlos Noceda, Mohanapriya Gunasekaran, Sarma Rajeev Kumar, Karine Leitão Lima Thiers, José Hélio Costa, Elisete Santos Macedo, Aprajita Kumari, Kapuganti Jagadis Gupta, Shivani Srivastava, Alok Adholeya, Manuela Oliveira, Isabel Velada, Debabrata Sircar, Ramalingam Sathishkumar, Birgit Arnholdt-Schmitt

Ovule siRNAs methylate and silence protein-coding genes in trans
Diane Burgess, Hiu Tung Chow, Jeffrey W. Grover, Michael Freeling, Rebecca A. Mosher

SlKIX8 and SlKIX9 are negative regulators of leaf and fruit growth in tomato
Gwen Swinnen, Jean-Philippe Mauxion, Alexandra Baekelandt, Rebecca De Clercq, Jan Van Doorsselaere, Dirk Inzé, Nathalie Gonzalez, Alain Goossens, Laurens Pauwels

Arabidopsis roots from Rosado, et al.

Shade-induced WRKY transcription factors restrict root growth during the shade avoidance response
Daniele Rosado, Amanda Ackermann, Olya Spassibojko, Magdalena Rossi, Ullas V. Pedmale

The U1 snRNP component RBP45d regulates temperature-responsive flowering in Arabidopsis thaliana
Ping Chang, Hsin-Yu Hsieh, Shih-Long Tu

INDEHISCENT regulates explosive seed dispersal
Anahit Galstyan, Penny Sarchet, Rafael Campos-Martin, Milad Adibi, Lachezar A. Nikolov, Miguel Pérez Antón, Léa Rambaud-Lavigne, Xiangchao Gan, Angela Hay

Seed morphological traits as a tool to quantify variation maintained in ex situ collections: a case study in Pinus torreyana (Parry)
Lionel N Di Santo, Monica Polgar, Storm Nies, Paul Hodgkiss, Courtney A Canning, Jessica W Wright, Jill A Hamilton

DNA METHYLTRANSFERASE 3 (MET3) is regulated by Polycomb Group complex during Arabidopsis endosperm development
Louis Tirot, Pauline E. Jullien

Developmental Effects on Relative Use of PEPCK and NADP-ME Pathways of C4 Photosynthesis in Maize
Jennifer J. Arp, Shrikaar Kambhampati, Kevin L. Chu, Somnath Koley, Lauren M. Jenkins, Todd C. Mockler, Doug K. Allen

GTL1 is required for a robust root hair growth response to avoid nutrient overloading
Michitaro Shibata, David S. Favero, Ryu Takebayashi, Ayako Kawamura, Bart Rymen, Yoichiroh Hosokawa, Keiko Sugimoto

Transcriptional activation of auxin biosynthesis drives developmental reprogramming of differentiated cells
Yuki Sakamoto, Ayako Kawamura, Takamasa Suzuki, Shoji Segami, Masayoshi Maeshima, Stefanie Polyn, Lieven De Veylder, Keiko Sugimoto

Genetic basis and dual adaptive role of floral pigmentation in sunflowers
Marco Todesco, Natalia Bercovich, Amy Kim, Ivana Imerovski, Gregory L. Owens, Óscar Dorado Ruiz, Srinidhi V. Holalu, Lufiani L. Madilao, Mojtaba Jahani, Jean-Sébastien Légaré, Benjamin K. Blackman, Loren H. Rieseberg

Arabidopsis stomatal polarity protein BASL mediates distinct processes before and after cell division to coordinate cell size and fate asymmetries
Yan Gong, Julien Alassimone, Andrew Muroyama, Gabriel Amador, Rachel Varnau, Ao Liu, Dominique C. Bergmann

Arabidopsis ABIG1 Functions in Laminar Growth and Polarity Formation through Regulation by REVOLUTA and KANADI
Jesus Preciado, Kevin Begcy, Tie Liu

3D reconstruction identifies loci linked to variation in angle of individual sorghum leaves
Michael C. Tross, Mathieu Gaillard, Mackenzie Zweiner, Chenyong Miao, Bosheng Li, Bedrich Benes, James C. Schnable

Members of the ELMOD protein family specify formation of distinct aperture domains on the Arabidopsis pollen surface
Yuan Zhou, Prativa Amom, Sarah H. Reeder, Byung Ha Lee, Adam Helton, Anna A. Dobritsa

Ureides are similarly accumulated in response to UV-C irradiation and wound but differently remobilized during recovery in Arabidopsis leaves.
Aigerim Soltabayeva, Aizat Bekturova, Assylay Kurmanbayeva, Dinara Oshanova, Zhadyrassyn Nurbekova, Sudhakar Srivastava, Dominic Standing, Moshe Sagi

| Evo-devo

Evolution of the nitric oxide synthase family in vertebrates and novel insights in gill development
Giovanni Annona, Iori Sato, Juan Pascual-Anaya, Ingo Braasch, Randal Voss, Jan Stundl, Vladimir Soukup, Sihigeru Kuratani, John Postlethwait, Salvatore D’Aniello

Evolutionary dynamics of sex-biased genes expressed in cricket brains and gonads
Carrie A. Whittle, Arpita Kulkarni, Cassandra G. Extavour

Single-nucleus transcriptomes reveal functional and evolutionary properties of cell types in the Drosophila accessory gland
Alex C. Majane, Julie M. Cridland, David J. Begun

Evolutionary transition of doublesex regulation in termites and cockroaches: from sex-specific splicing to male-specific transcription
Satoshi Miyazaki, Kokuto Fujiwara, Keima Kai, Yudai Masuoka, Hiroki Gotoh, Teruyuki Niimi, Yoshinobu Hayashi, Shuji Shigenobu, Kiyoto Maekawa

Complete metamorphosis and microbiota turnover in insects
Christin Manthey, Paul R Johsnton, Jens Rolff

Facultative release from developmental constraints through polyphenism promotes adaptively flexible maturation
Flor T. Rhebergen, Isabel M. Smallegange

A large disordered region confers a wide spanning volume to vertebrate Suppressor of Fused as shown in a trans-species solution study
Staëlle Makamte, Amira Jabrani, Annick Paquelin, Anne Plessis, Mathieu Sanial, Aurélien Thureau, Olga Rudenko, Francesco Oteri, Marc Baaden, Valérie Biou

Developmental plasticity in male courtship in Bicyclus anynana butterflies is driven by hormone regulation of the yellow gene
Heidi Connahs, Eunice Jingmei Tan, Yi Ting Ter, Emilie Dion, Yuji Matsuoka, Ashley Bear, Antónia Monteiro

Convergent adaptation and ecological speciation result from unique genomic mechanisms in sympatric extremophile fishes
Ryan Greenway, Anthony P. Brown, Henry Camarillo, Cassandra Delich, Kerry L. McGowan, Joel Nelson, Lenin Arias-Rodriguez, Joanna L. Kelley, Michael Tobler

Behavioural adaptations in egg laying ancestors facilitate evolutionary transitions to live birth
Amanda K. Pettersen, Nathalie Feiner, Daniel W.A. Noble, Geoffrey M. While, Charlie K. Cornwallis, Tobias Uller

Functional divergence of the bag of marbles gene in the Drosophila melanogaster species group
Jaclyn E. Bubnell, Cynthia K.S. Ulbing, Paula Fernandez-Begne, Charles F. Aquadro

Adaptive shifts underlie the divergence in wing morphology in bombycoid moths
Brett R. Aiello, Milton Tan, Usama Bin Sikandar, Alexis J. Alvey, Burhanuddin Bhinderwala, Katalina C. Kimball, Jesse R. Barber, Chris A. Hamilton, Akito Y. Kawahara, Simon Sponberg

An evolutionarily conserved odontode gene regulatory network underlies head armor formation in suckermouth armored catfish
Shunsuke Mori, Tetsuya Nakamura

Evolution of lbx spinal cord expression and function
José Luis Juárez-Morales, Frida Weierud, Samantha J. England, Celia Demby, Nicole Santos, Ginny Grieb, Sylvie Mazan, Katharine E. Lewis

Evolution of Drosophila buzzatii wings: Modular genetic organization, sex-biased integrative selection and intralocus sexual conflict
PP Iglesias, FA Machado, S Llanes, E Hasson, EM Soto

Sex-Specific Plasticity Explains Genetic Variation in Sexual Size Dimorphism in Drosophila
Isabelle M Vea, Austin Wilcox, W. Anthony Frankino, Alexander W Shingleton

Cell Biology

A mechano-osmotic feedback couples cell volume to the rate of cell deformation
Larisa Venkova, Amit Singh Vishen, Sergio Lembo, Nishit Srivastava, Baptiste Duchamp, Artur Ruppel, Stéphane Vassilopoulos, Alexandre Deslys, Juan Manuel Garcia Arcos, Alba Diz-Muñoz, Martial Balland, Jean-François Joanny, Damien Cuvelier, Pierre Sens, Matthieu Piel

Volume growth in animal cells is cell cycle dependent and shows additive fluctuations
Clotilde Cadart, Matthieu Piel, Marco Cosentino Lagomarsino

C. elegans embryos from Chang, et al.

A particle size threshold governs diffusion and segregation of PAR-3 during cell polarization
Yiran Chang, Daniel J. Dickinson

Mouse hair placodes from Moreci and Lechler

KIF18B is a cell-type specific regulator of spindle orientation in the epidermis
Rebecca S. Moreci, Terry Lechler

A ciliopathy complex builds distal appendages to initiate ciliogenesis
Dhivya Kumar, Addison Rains, Vicente Herranz-Pérez, Quanlong Lu, Xiaoyu Shi, Danielle L. Swaney, Erica Stevenson, Nevan J. Krogan, Bo Huang, Christopher Westlake, Jose Manuel Garcia-Verdugo, Bradley Yoder, Jeremy F. Reiter

Mouse oocytes do not contain a Balbiani body
Laasya Dhandapani, Marion C. Salzer, Juan M. Duran, Gabriele Zaffagnini, Cristian De Guirior, Maria Angeles Martínez-Zamora, Elvan Böke

SLC1A5 provides glutamine and asparagine necessary for bone development in mice
Deepika Sharma, Yilin Yu, Leyao Shen, Guo-Fang Zhang, Courtney Karner

The Transcriptional Co-Activator Taz Contributes to the Differentiation of a Salivary Gland Epithelial Cell Line Towards a Myoepithelial Phenotype
Renee F. Thiemann, Scott Varney, Nicholas Moskwa, John Lamar, Melinda Larsen, Susan E. LaFlamme

Anoikis resistance in mammary epithelial cells is mediated by semaphorin 7a
Taylor R. Rutherford, Alan M Elder, Traci R. Lyons

A steroid hormone regulates growth in response to oxygen availability
George P. Kapali, Viviane Callier, Hailey Broeker, Parth Tank, Samuel J.L. Gascoigne, Jon F Harrison, Alexander W. Shingleton

Modelling

Cell types and ontologies of the Human Cell Atlas

David Osumi-Sutherland, Chuan Xu, Maria Keays, Peter V. Kharchenko, Aviv Regev, Ed Lein, Sarah A. Teichmann

Computational modelling of cell motility modes emerging from cell-matrix adhesion dynamics
Leonie van Steijn, Clément Sire, Loïc Dupré, Guy Theraulaz, Roeland M.H. Merks

A landscape model for cell fate decisions during mesoendoderm differentiation in C. elegans based on Wnt dynamics
Shyr-Shea Chang, Zhirong Bao, Eric D. Siggia

Modelling the worm embryo from Serin-Lee & Kimura

The extra-embryonic space is a geometric constraint regulating cell arrangement in nematodes
Sungrim Seirin-Lee, Akatsuki Kimura

From heterogeneous datasets to predictive models of embryonic development
Sayantan Dutta, Aleena L. Patel, Shannon E. Keenan, Stanislav Y. Shvartsman

Graph-based machine learning reveals rules of spatiotemporal cell interactions in tissues
Takaki Yamamoto, Katie Cockburn, Valentina Greco, Kyogo Kawaguchi

Blastocoel morphogenesis: a biophysics perspective

Mathieu Le-Verge-Serandour, Hervé Turlier

Disorder in cellular packing can alter proliferation dynamics to regulate growth

Chandrashekar Kuyyamudi, Shakti N. Menon, Fernando Casares, Sitabhra Sinha

Tools & Resources

hei-tag: a highly efficient tag to boost targeted genome editing
Thomas Thumberger, Tinatini Tavhelidse, Jose Arturo Gutierrez-Triana, Rebekka Medert, Alex Cornean, Bettina Welz, Marc Freichel, Joachim Wittbrodt

NanoDam identifies novel temporal transcription factors conserved between the Drosophila central brain and visual system
Jocelyn L.Y. Tang, Anna E. Hakes, Robert Krautz, Takumi Suzuki, Esteban G. Contreras, Paul M. Fox, Andrea H. Brand

Worm mapping in Skuhersky, et al.

Toward a More Accurate 3D Atlas of C. elegans Neurons
Michael Skuhersky, Tailin Wu, Eviatar Yemini, Edward Boyden, Max Tegmark

Genome editing in animals with minimal PAM CRISPR-Cas9 enzymes
Jeremy Vicencio, Carlos Sánchez-Bolaños, Ismael Moreno-Sánchez, David Brena, Dmytro Kukhtar, Miguel Ruiz-López, Mariona Cots-Ponjoan, Charles E. Vejnar, Alejandro Rubio, Natalia Rodrigo Melero, Carlo Carolis, Antonio J. Pérez-Pulido, Antonio J. Giráldez, Benjamin P. Kleinstiver, Julián Cerón, Miguel A. Moreno-Mateos

A CRISPR toolbox for generating intersectional genetic mice for functional, molecular, and anatomical circuit mapping
Savannah J. Lusk, Andrew McKinney, Patrick J. Hunt, Paul G. Fahey, Jay Patel, Jenny J. Sun, Vena K. Martinez, Ping Jun Zhu, Jeremy R. Egbert, Xiaolong Jiang, Benjamin R. Arenkiel, Andreas S. Tolias, Mauro Costa-Mattioli, Russell S. Ray

Extremely bright, near-IR emitting spontaneously blinking fluorophores enable ratiometric multicolor nanoscopy in live cells
Jonathan Tyson, Kevin Hu, Shuai Zheng, Phylicia Kidd, Neville Dadina, Ling Chu, Derek Toomre, Joerg Bewersdorf, Alanna Schepartz

CeLINC worms from Kroll, et al.

CeLINC, a fluorescence-based protein-protein interaction assay in C. elegans
Jason R Kroll, Sanne Remmelzwaal, Mike Boxem

Anatomical Structures, Cell Types, and Biomarkers Tables Plus 3D Reference Organs in Support of a Human Reference Atlas
Katy Börner, Sarah A. Teichmann, Ellen M. Quardokus, James Gee, Kristen Browne, David Osumi-Sutherland, Bruce W. Herr II, Andreas Bueckle, Hrishikesh Paul, Muzlifah A. Haniffa, Laura Jardine, Amy Bernard, Song-Lin Ding, Jeremy A. Miller, Shin Lin, Marc Halushka, Avinash Boppana, Teri A. Longacre, John Hickey, Yiing Lin, M. Todd Valerius, Yongqun He, Gloria Pryhuber, Xin Sun, Marda Jorgensen, Andrea J. Radtke, Clive Wasserfall, Fiona Ginty, Jonhan Ho, Joel Sunshine, Rebecca T. Beuschel, Maigan Brusko, Sujin Lee, Rajeev Malhotra, Sanjay Jain, Griffin Weber

Fast 3D Clear: A Fast, Aqueous-Based, Reversible Three-Day Tissue Clearing Method for Adult and Embryonic Mouse Brain and Whole Body
Stylianos Kosmidis, Adrian Negrean, Alex Dranovsky, Attila Losonczy, Eric R. Kandel

The developmental transcriptome of Parhyale hawaiensis: microRNAs and mRNAs show different expression dynamics during the maternal-zygotic transition
Llilians Calvo, Maria Birgaoanu, Tom Pettini, Matthew Ronshaugen, Sam Griffiths-Jones

Tissue-specific modification of cellular bioelectrical activities using the chemogenetic tool, DREADD, in zebrafish
Martin R. Silic, GuangJun Zhang

Single nucleus pituitary transcriptomic and epigenetic landscape reveals human stem cell heterogeneity with diverse regulatory mechanisms
Zidong Zhang, Michel Zamojski, Gregory R. Smith, Thea L. Willis, Val Yianni, Natalia Mendelev, Hanna Pincas, Nitish Seenarine, Mary Anne S. Amper, Mital Vasoya, Venugopalan D. Nair, Judith L. Turgeon, Daniel J. Bernard, Olga G. Troyanskaya, Cynthia L. Andoniadou, Stuart C. Sealfon, Frederique Ruf-Zamojski

Zebrafish Cre/lox regulated UFlip alleles generated by CRISPR/Cas targeted integration provide cell-type specific conditional gene inactivation
Maira P. Almeida, Sekhar Kambakam, Fang Liu, Zhitao Ming, Jordan M. Welker, Wesley A. Wierson, Laura E. Schultz-Rogers, Stephen C. Ekker, Karl J. Clark, Jeffrey J. Essner, Maura McGrail

A highly efficient reporter system for identifying and characterizing in vitro expanded hematopoietic stem cells
James L.C. Che, Daniel Bode, Iwo Kucinski, Alyssa H. Cull, Fiona Bain, Melania Barile, Grace Boyd, Miriam Belmonte, Maria Jassinskaja, Juan Rubio-Lara, Mairi S. Shepherd, Anna Clay, Adam C. Wilkinson, Hiromitsu Nakauchi, Satoshi Yamazaki, Berthold Göttgens, David G. Kent

Forward genetics combined with unsupervised classifications identified zebrafish mutants affecting biliary system formation
Divya Jyoti Singh, Kathryn M. Tuscano, Karen L. Ortega, Manali Dimri, Kevin Tae, William Lee, Muslim A. Muslim, Jay L. Liu, Lain X. Pierce, Allyson McClendon, Gregory Naegele, Isabel Gibson, Jodi Livesay, Takuya F. Sakaguchi

Research practice & education

Building Back More Equitable STEM Education: Teach Science by Engaging Students in Doing Science
Sarah C R Elgin, Shan Hays, Vida Mingo, Christopher D Shaffer, Jason Williams

TeamTree analysis: a new approach to evaluate scientific production
Frank W. Pfrieger

I, We, and They: A Linguistic and Narrative Exploration of the Authorship Process
Abigail Konopasky, Bridget C O’Brien, Anthony R Artino Jr., Erik W Driessen, Christopher J Watling, Lauren A Maggio

Thumbs up (No Ratings Yet)
Loading...

Tags:
Categories: Highlights

The people behind the papers – Brandon Carpenter and David Katz

Posted by , on 5 July 2021

This interview, the 94th in our series, was published in Development earlier this year

A dynamic pattern of histone methylation and demethylation controls gene expression during development, with some processes such as formation of the zygote involving large-scale reprogramming of methylation states. A new paper in Development investigates how inherited histone methylation regulates developmental timing and the germline/soma distinction in Caenorhabditis elegans. To hear more about the story we caught up with first author and postdoctoral researcher Brandon Carpenter, and his supervisor David Katz, Associate Professor in the Department of Cell Biology at Emory University School of Medicine in Atlanta, Georgia.

Brandon (L) and David (R)

David, can you give us your scientific biography and the questions your lab is trying to answer?

DK: As a graduate student, I worked with Dr Shirley Tilghman at Princeton on the regulation of genomic imprinting. We provided in vivo evidence for the first chromatin boundary formed by CTCF at the H19 locus in mouse. As a postdoc, I worked for Dr Bill Kelly at Emory University on the regulation of histone methylation in the germline of C. elegans. We provided the first evidence of a transgenerational phenotype (sterility) caused by the build-up of histone methylation, when the H3K4me1/2 demethylase Lsd1 is mutated. In my own lab at the Emory University School of Medicine, we have worked on both C. elegans and mouse model systems to study the mechanisms that regulate histone methylation and how inappropriately inherited histone methylation gives rise to phenotypes. We have also implicated LSD1 as a crucial molecule that may contribute to Alzheimer’s disease: our data suggest it is being inhibited by pathological aggregates in dementia patients. We are currently trying to develop a therapeutic intervention based on what we have learned about the function of LSD1 in the Alzheimer’s disease pathway.

Brandon – how did you come to work in David’s lab and what drives your research today?

BC: After obtaining my doctoral degree, I knew I wanted to continue studying development, and that I wanted to focus on a model system that would allow undergraduates to develop projects related to my research. As a graduate student, I fell in love with mentoring students and wanted to find opportunities to inspire students in the classroom as well as at the bench. Thus, my passion for mentoring and studying developmental biology led me to the Katz lab, where I could work with the beautiful model system, C. elegans, to study how epigenetic inheritance affects developmental cell fates.

At Emory University, I joined the Katz lab as a Fellowship In Research and Science Teaching (FIRST) Fellow (part of the National Institutes of Health-funded IRACDA programme) where, in parallel with my research, I was able to develop my teaching and mentorship skills. The most exciting part of joining the Katz lab was being able to bring my research into the classroom at Oglethorpe University, a nearby small liberal arts college. The Katz lab has a longstanding collaboration with Dr Karen Schmeichel from the biology department at Oglethorpe, integrating C. elegans experiments into the entire curriculum. As part of this, I was able to teach a semester-long Course-based Undergraduate Research Experience (CURE) based on the research I was conducting in the Katz lab. As part of this semester-long CURE, Oglethorpe students became inspired by the science we are doing in the Katz Lab and generated data for this manuscript. Jovan Brockett, an undergraduate student, is an author on this manuscript for research he did in the classroom! My passion for understanding how an organism develops from a single cell drives my research, and the feeling I get when I see my mentees succeed while studying these mechanisms provides the fuel that keeps me going.

How has your research been affected by the COVID-19 pandemic?

BC: For me, COVID-19 hit right as I was finishing the experiments for two of my postdoctoral projects. During the ∼2-3 month lab shut down I was able to stay productive by submitting and revising manuscripts, but being away from the lab made it hard to advance interesting new ideas. The thing I miss most about not being able to go into lab is my ability to bounce crazy ideas off my talented Katz lab colleagues.

Before your work, what was known about the role of inherited histone methylation in the germline/soma distinction?

BC, DK: This paper is really about two major lines of research coming together to create a new story. We had been working on how two histone modifying enzymes, the H3K4me1/2 demethylase LSD1 (SPR-5 in C. elegans) and the H3K9 methyltransferase MET-2 cooperate to reprogramme histone methylation at fertilization to prevent the inappropriate chromatin environment from being passed on from one generation to the next. We had found that a failure to reprogramme histone methylation in spr-5; met-2 double mutants causes a maternal effect developmental delay and sterility phenotype. We were interested in how the inappropriate inheritance of histone methylation causes the developmental delay. Dr Susan Strome, with some help from Dr Bill Kelly, had performed some beautiful work showing how maternal deposition of the H3K6 methyltransferase is required transgenerationally to help specify the germline in progeny. Brandon had noticed some similarities between the developmental delay that we were observing and some high temperature phenotypes that Susan Strome had shown and were continuing to be worked on by Dr Lisa Patrella in her own lab. As detailed below, Brandon was able to show that the MES-4 system and the SPR-5; MET-2 reprogramming mechanism antagonize one another. It is also important to note that several labs have identified somatic repression mechanisms that antagonize the MES-4 system. We are interested in seeing how these systems interface with SPR-5; MET-2 reprogramming, so stay tuned!

Can you give us the key results of the paper in a paragraph?

BC, DK: We had previously shown that SPR-5 and MET-2 act together to repress germline genes at fertilization. In this paper, we found that H3K36 methylation antagonizes this repression to prevent these germline genes from being completely shut down. Without inherited H3K36 methylation, the germline is not properly specified. In contrast, without SPR-5; MET-2 repression, H3K36 is inappropriately propagated to the soma, resulting in germline genes being inappropriately expressed there. The inappropriate expression of germline genes in the soma results in a developmental delay. Thus, neither SPR-5; MET-2 reprogramming nor the MES-4 germline inheritance system can properly function without each other. Instead SPR-5, MET-2 and MES-4 coordinately balance three difference histone modifications (H3K4, H3K9 and H3K36 methylation) to ensure that germline versus soma is properly specified.

Single molecule fluorescence in situ hybridization image of a C. elegans L1 larvae ectopically expressing a germline-specific gene, htp-1, in somatic tissues (grey, htp-1mRNA; blue; DAPI).

Why do you think inappropriate somatic expression of germline genes causes developmental delay?

BC, DK: As discussed in the paper, we think that there are two possible mechanisms for how the ectopic expression of germline genes causes developmental delay in spr-5; met-2 mutants. One possibility is that transcription of the germline programme itself causes the developmental delay. For example, if germline transcription factors are competing with somatic transcription factors to turn on genes, it is possible that the mix of proteins generated is simply too confusing for the cell to commit to its proper cell fate. The alternative is that a part of the germline function interferes with somatic development; for example, the germline precursors undergo a cell cycle arrest. It is possible that the proteins involved in this germline cell cycle checkpoint slow the progression of somatic cells via cell cycle regulation. Consistent with this latter possibility, we show that spr-5; met-2 mutants can silence an extrachromosomal array in the soma. This function, which is normally confined to the germline, suggests that somatic tissues in spr-5; met-2 mutants make proteins that can perform some germline functions. Thus, it is possible that a germline function acting in the soma prevents somatic cells from quickly adopting their proper cell fate.

What relevance do your data have for human patients harbouring mutations in histone-modifying enzymes?

BC, DK: Recent genome sequencing has revealed that several neurodevelopmental disorders are caused by mutations in histone-modifying enzymes. These include mutations in: (1) the H3K36 methyltransferase Setd2, the H3K27 demethylase Kdm6a and the H3K4 methyltransferase Kmt2d, which cause Kabuki Syndrome; (2) the human orthologue of spr-5LSD1, which causes a Kabuki-like Syndrome; and (3) the H3K36 methyltransferase Nsd1, which causes Sotos Syndrome. Similar to what we observed in spr-5; met-2 mutant progeny, many of the human patients with mutations in these histone-modifying enzymes suffer from global developmental delay. Based on our model, it is possible that the developmental delay in these patients may be caused by the failure to properly regulate histone methylation during essential developmental transitions. Consistent with this, we have recapitulated some phenotypes in a maternal hypomorphic mutant of Lsd1 in mice that are reminiscent of Kabuki Syndrome. We hope that by continuing to study how mutations in histone-modifying enzymes in C. elegans and mice give rise to developmental defects, we will shed light on the human diseases caused by defects in histone-modifying enzymes.

When doing the research, did you have any particular result or eureka moment that has stuck with you?

BC: As detailed above, I had made the observation that the developmental delay we were observing had some similarities to some high-temperature phenotypes that Susan Strome had shown and were continuing to be worked on by Lisa Patrella in her own lab. This raised the possibility of a connection between SPR-5; MET-2 reprogramming and the MES-4 inheritance system. The crucial test of this potential connection was to knock down mes-4 via RNAi and see if it rescued the developmental delay, and we were very excited to find that it did rescue it, and even more excited when our RNA-seq subsequently showed that MES-4 germline genes are expressed in the somatic tissues of spr-5; met-2 mutants. After this independent confirmation of the connection between the two systems, we were confident that we had figured it out.

And what about the flipside: any moments of frustration or despair?

BC: The most frustrating part of this project was trying to gather enough L1 larvae to perform the initial genomic experiments. When we first started the project, there were no strains available that could balance the spr-5 mutant allele. I had to genotype every single hermaphrodite parent! At one point, I thought I would never get enough larvae to perform the genomic experiments. But David saw on science Twitter that the Caenorhabditis Genetics Center (CGC), which houses C. elegans strains, was developing new balancer strains. I contacted them and was able to get the FX30208 tmC27 [unc-75(tmls1239)](I) balancer even before they made it available to the broader C. elegans community. By reporting back that it worked well, I was also able to give back to the C. elegans community.

What next for you after this paper?

BC: I am officially on the academic job market searching for a tenure-track position and developing exciting new projects of my own that stem from this paper. We have mounting evidence that mutations in highly conserved histone-modifying enzymes may give rise to developmental phenotypes in vertebrates that are similar to what we observe in C. elegans. I want to take advantage of C. elegans mutants that fail to properly inherit histone methylation to further investigate how inherited chromatin states affect complex developmental processes like cell-to-cell communication and cell migration. I am also interested in potentially introducing the human version of Lsd1 into C. elegans to humanize the worm so that I can generate mutations that have been found in the human LSD1 patients. This type of approach is on-going through the NIH-funded Undiagnosed Diseases Network (UDN).

Where will this story take the Katz lab?

DK: We believe that spr-5; met-2 double mutants provide an excellent model for understanding how cells respond to inappropriately inherited histone methylation. We are taking advantage of the invariant embryonic cell lineage in C. elegans by performing automated lineage tracing experiments in spr-5; met-2 mutants. This will enable us to understand cell by cell how inappropriately inherited histone methylation affects processes such as cell division timing, cell migration, programmed cell death, etc. We hope to combine this with single cell RNA-seq to ask how each cell responds transcriptionally to this inappropriately inherited histone methylation. So stay tuned.

Remarkably, trying to understand the regulation of histone methylation in the germline has also taken us into the Alzheimer’s disease field. While trying to understand whether SPR-5; MET-2 reprogramming is conserved in mice, we serendipitously discovered that LSD1 is continually required for the survival of hippocampus and cortex neurons. We are interested in the possibility that terminally differentiated cells continually employ histone-modifying enzymes, such as LSD1, to maintain their cell fate. In the meantime, we have gone on to provide evidence that LSD1 is inhibited by pathological aggregates of tau in mice and human Alzheimer’s disease patients. We believe that this inhibition is a crucial part of how pathological tau induces neurodegeneration. So, you never know where developmental biology will lead!

You never know where developmental biology will lead!

Finally, let’s move outside the lab – what do you like to do in your spare time in Atlanta?

BC: I like to go hiking with my 5-year-old twin daughters, play golf, and find cool breweries who push the edge on brewing delicious stouts and IPAs!

DK: I enjoy soccer with my 13-year-old twins and co-host a popular Atlanta United podcast. I also co-founded a very small vineyard just outside Atlanta at a friend’s house, and we have just produced our first successful vintage of a Norton/Cabernet Sauvignon blend. But I am also happy to drink a local beer with my outstanding postdoc Brandon.

Thumbs up (No Ratings Yet)
Loading...

Tags:
Categories: Interview