The community site for and by
developmental and stem cell biologists

New Intern for the Node

Posted by , on 16 September 2024

Hi everyone! 

My name is Ryan Harrison, and I will be supporting the community sites at The Company of Biologists as part of a three-month internship. 

I am currently pursuing a PhD between the labs of Timothy Saunders and James Briscoe as part of the MRC DTP in Interdisciplinary Biomedical Research, where I am researching organoids that mimic embryonic development of the lower back and spine.  

Fluorescence images of some organoids I have grown in the lab. In the organoid on the left, we can see how different cell fates emerge (red and yellow), and on the right we can see the some of the cellular cytoarchitecture (green) in the organoid. 

At university, I am also part of the Ambassadors for a Better Research Culture (ABRC), where we aim to improve the research environment on the medical school campus for postgraduate students and staff. Here, I am part of the ‘LGBTQIA+ Inclusion’ subgroup where we run monthly events to foster a community of LGBTQIA+ researchers at the medical school campus. We have also established a larger series called ‘Pride in STEM’, where we invite external speakers to discuss their experiences of being queer in different STEM career environments. I would like to carry this into my internship and compile some LGBTQIA+ voices for a post in the Honest Conversations blog series here on the Node. If anyone is interested in sharing their experiences of being queer in academia, please feel free to get in touch at ryan.harrison@biologists.com or thenode@biologists.com

Thumbs up (1 votes)
Loading...

Tags: ,
Categories: News, Uncategorized

Navigate the archive

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

Human developmental biology: the past, present and future

Posted by , on 16 September 2024

This year, 2024, marks the 10th anniversary of the first Development ‘From Stem Cells to Human Development’ meeting, and today is the beginning of the sixth meeting at Wotton House in the UK. The influence the meeting has had on the field is discussed in a recent article by science historian Nick Hopwood (Hopwood, 2024a), who suggests that human developmental biology has experienced peaks of attention and periods of neglect, fuelled by the productivity of technical innovations. In the current issue of Development, we have published a complementary Perspective article by Nick highlighting key aspects of the history of the field for an audience of stem cell and developmental biologists (Hopwood, 2024b). In addition, Development invited researchers from eight countries around the world to respond to these ideas and comment on how human development is perceived in their country of work, discussing how they believe their local legal, political, regulatory, societal and technological frameworks are influencing the field’s trajectory (Clark et al., 2024). The authors and some highlights from the Perspective are shown in the image below, and you can click the image to read the whole article.

Recognising that this article only manages to capture a small sample of the breadth of human development and stem cell research worldwide, we encourage you, readers of the Node, to share your opinion on human developmental biology in your country of work. Do you believe that interest in human developmental biology is cyclical, as suggested by Hopwood? If so, what lies ahead? Are we experiencing a boom or bust in support of human development research? How long might this trajectory lead before turning on its head? What societal undercurrents might contribute to maintaining or changing the field’s course? The floor is yours…

References

Clark, A.T., Goolam, M., Hanna, J.H., Long, K., Nicol, D., Petropoulos, S., Saitou, M., Tam, P.L., Wang, H. Human developmental biology – a global perspective. Development. 151(17). https://doi.org/10.1242/dev.203092

Hopwood, N. (2024a). Species Choice and Model Use: Reviving Research on Human Development. In Journal of the History of Biology. Springer Science and Business Media LLC. https://doi.org/10.1007/s10739-024-09775-7

Hopwood, N. (2024b). Past and future of human developmental biology. Development. 151(17). https://doi.org/10.1242/dev.203085

Thumbs up (No Ratings Yet)
Loading...

Tags: , , ,
Categories: Discussion, Research

Faculty Position in Developmental, Cell or Stem Cell Biology, King’s College London

Posted by , on 12 September 2024

Closing Date: 6 October 2024

Are you ready to be a group leader? Are you a multi-disciplinary biologist with interests in developmental cell biology? Join a robust, collaborative and supportive department with world-class research.

We seek to appoint a Lecturer (Assistant Prof Equivalent) in developmental, cell or stem cell biology, genetics or genomics with a focus on human disease modelling and/or craniofacial biology.

Who are you? You are a promising postdoc with an excellent publication profile, high-quality collaborative connections and ambitious plans for your independent research programme. You should be an early career scientists with an outstanding research track record and excellent potential to develop an internationally competitive research programme and to collaborate within the Centre and across King’s.

Who are we? The Centre for Craniofacial & Regenerative Biology at King’s College London is one of the leading centres for Craniofacial and Stem Cell Biology worldwide. Our Centre comprises 19 collaborative groups with interests in craniofacial and stem cell biology, innovative bioengineering strategies to regeneration and repair, and big data approaches to understand the complexity of development and disease. Our research spans basic, clinical and translational sciences. The Centre offers a vibrant, collaborative, and interactive research and teaching environment in the heart of London.

https://www.kcl.ac.uk/dentistry/research/centre-for-craniofacial-regenerative-biology

Successful candidates are expected to establish their independent research group in the Centre, to contribute to our educational programmes and to training the next generation of interdisciplinary scientists, and to support the strategic vision of the Centre and King’s. They will have access to a variety of PhD programmes, as well as mentorship and career development opportunities. They will work with outstanding scientists across King’s https://www.kcl.ac.uk/research and access our world-class Research Facilities: https://www.kcl.ac.uk/research/facilities

CLOSING DATE 6 October. Application link below. Informal inquiries are welcome to Head of Department Professor Andrea Streit or to relevant Faculty members.

https://rb.gy/ksftkv

Thumbs up (No Ratings Yet)
Loading...

Tags: , ,
Categories: Careers, Jobs, Uncategorized

A short rant on the present and future of developmental biology

Posted by , on 11 September 2024

Observing a cluster of migrating cells or a developing embryo through the lens of a microscope can be a visceral experience; one is struck by the ephemeral beauty, layered complexity, and alien intelligence displayed by such specimens. For those who seek a scientific understanding of these striking phenomena, it is also a humbling experience. There are so many moving parts here, so many subsystems within subsystems, so much noise, so much nonlinearity, so much contingency… how could we possibly hope to capture this in the simple yet powerful models that make scientific explanations so satisfying and useful?

I’ve been grappling with this question ever since my undergrad, and as anyone who does so, I have found plenty of reasons to be pessimistic about it.

High-resolution confocal microscopy image of zebrafish cranial neural crest cells migrating through the embryo.
Neural crest cells swarming through a zebrafish embryo are just one of countless phenomena in cell and developmental biology that are truly spectacular to behold. [Main panel: live-imaged AiryScan volume; by author. Bottom-right inset: fixed lightsheet volume for spatial reference; by Zimeng Wu, UCL.]

Though we have extensive knowledge of the molecular machines that form the building blocks of biological systems, putting this giant puzzle together from the bottom up seems an impossibly complicated task. Instead, the field’s still-dominant approach is to link particular perturbations to particular outcomes, usually by lifting out a handful of mechanisms or genes from the broader system, drawing arrows between them, and calling the result a pathway. But whilst the models this produces are of an appealing simplicity, they lack power; they often fail to explain or predict anything outside the narrow set of conditions and observations that were considered in the original study. At least we are indisputably making progress in developing new tools to collect more and better data, ever more quickly, ever more precisely… but alas, this progress is closely shadowed by the realization that it can only take us so far; more data does not yield more understanding if we don’t know how to ask the right questions.

With these problems permeating the field, it comes as no surprise that there is a measure of discontent in the community. Some argue that we have an attitude problem [1]; perhaps young researchers spend too much time on twitter and not enough time in the library? Others contend that we have an image problem [2]; perhaps we should be spending more time on twitter, reassuring each other and the wider public that our field remains essential – or even that it has recently entered, as some would have it [3], a “new golden age”? Like so many developmental biology papers, these viewpoints may not be entirely wrong, but they’re also not particularly compelling.

I’d like for us to entertain the possibility that we are in fact facing a science problem. That our progress is not bottlenecked by modern attitudes or public misperceptions, but by the profound intellectual challenge of finding new and better ways of thinking about the spectacles that play out under our microscopes. I’d like to take seriously the above reasons for pessimism and treat them as real scientific challenges for us to tackle and overcome. If the molecular details are intractable, we should search for new and better systems-level abstractions to subsume them. If the current standard of mechanistic explanation is inadequate, we should look to build new and better conceptual frameworks that set a higher standard. If it is hard to distill meaning from the deluge of high-throughput data, we should aim to develop new and better models that yield strong inductive priors for big-data analysis.

This is much easier said than done, of course, but in grappling with these issues I have also come across a few reasons for optimism!

Looking back in history, the challenge faced by Darwin and his contemporaries in seeking to unify the diversity of living organisms must have seemed no less daunting than our current predicament, yet they persevered and emerged with an entirely new understanding of life. Returning to modern times, a new theory of cell types established about a decade ago shows brilliantly that deep conceptual progress is possible even today [4]. And not only that; it also shows that such progress really does have the impact we would hope to see! For one, it has inspired new ways of analyzing and interpreting transcriptomics data (see e.g. [5]). For another, I have personally witnessed how much more productive the discourse on cell types can be within a group of researchers who know this theory (even if they don’t all fully endorse it) compared to a group who do not. These and other inspirational observations are always in the back of my mind as I explore my own ideas for tackling the field’s fundamental problems.

One such idea is the Core & Periphery (C&P) hypothesis, which was published last week [6] and serves as the occasion for this post.

The C&P idea originated from discussions between first author Elisa Gallo and me on the prospects of discovering principles that generalize well across different biological systems and phenomena. It is often implicitly assumed that the diversity of cellular and multi-cellular behaviors results from the contingent combination of various modular parts or subprocesses, much like sequence diversity on the molecular level. This would leave us with limited avenues to pursue explanations that generalize over many such contingent assemblies.

Mulling over this in search for alternative perspectives eventually led us to an almost metaphysical argument: if there do exist principles that can explain a diverse set of biological phenomena in a unified manner, then they must be generative principles, that is to say they must comprise a mechanism by which the explained diversity is generated. But are there any such mechanisms in cellular and developmental systems, or does contingency reign supreme?

As we started looking with fresh eyes, it turned out that many of the biological phenomena we are interested in (including cytoskeletal dynamics, reaction-diffusion patterning, different aspects of multicellular morphogenesis, and even embryo-scale processes like gastrulation) can indeed be decomposed into what we have come to call a versatile core and a function-specifying periphery. A versatile core is a mechanism that implements a generative principle and hence is capable of producing a wide range of different behaviors or outputs. The periphery, then, is what configures such a core to produce one particular function out of the many in its large behavior inventory.

Illustrated examples of Core & Periphery systems, including actomyosin, Rho GTPases, gastrulation, the DITH, and the BSW Turing system.
A medley of examples of C&P architectures compiled from our recent paper. The same cores (orange disks) are reused with different peripheries (blue leaves) to generate different functional behaviors or outputs. [The exquisite illustrations in the paper were created by first author Elisa Gallo, UZH.]

Intriguingly, we expect systems with a C&P architecture to be highly evolvable because the core’s large behavioral space is readily accessible through modifications in the periphery. As a consequence, cores will tend to spread widely and become deeply conserved in evolution, even as their peripheries diversify to exploit the full range of the core’s versatility. If follows that a generative principle that describes how a core works will generalize across the many different systems and phenomena wherein that core is reused. In other words, the C&P decomposition helps us separate the generalizable (the core) from the contingent (the periphery).

A more systematic introduction and comprehensive discussion of what the C&P hypothesis proposes is of course found in the paper. For my ramblings here, what matters most is that working on this project has greatly increased my optimism, to the point where I now believe that it really is possible to discover human-interpretable yet powerful theories that capture the essence of complex living systems. It’s just that the structure of such theories may need to differ considerably from that of the classical mechanistic accounts we are accustomed to, which is what makes it so hard (and so exciting) to pursue them.

This pursuit requires conceptual work, which means reading widely, thinking deeply, and engaging in intense and interdisciplinary discussion. As it turns out, this is surprisingly difficult and time-consuming; it is real scientific work. Unfortunately, our current research ecosystem does very little to incentivize and support such efforts. Young researchers in particular feel the pressure to pipette and/or code as fast as we can, just to stay in place in an ever-accelerating academic rat race. Taking time to think outside established lines seems wasteful, let alone taking time to pursue an explicitly conceptual project. In my case, it was only through a combination of luck, privilege, and the generosity of a few individuals that I was able to take a sabbatical year and invest the time necessary to arrive at the C&P hypothesis as it now stands. If we want the pace of conceptual innovation to pick up, this will need to change. Fortunately, there are positive signals here, too, as some leading institutes are now building up new theory-focused research programs.

In conclusion, I see many serious obstacles that we must face on our quest to better understand the complexity, intelligence, and beauty of cells and embryos. But if we take these obstacles seriously, I dare hope that we can overcome them, and that the dawn of a new golden age is indeed on the horizon.

Many thanks to Elisa Gallo and Matyas Bubna-Litic for their feedback on a draft version of this post.

[1] C.D. Stern, Reflections on the past, present and future of developmental biology, Developmental Biology 488 (2022) 30–34. https://doi.org/10.1016/j.ydbio.2022.05.001.
[2] J.B. Wallingford, We Are All Developmental Biologists, Developmental Cell 50 (2019) 132–137. https://doi.org/10.1016/j.devcel.2019.07.006.
[3] P. Liberali, A.F. Schier, The evolution of developmental biology through conceptual and technological revolutions, Cell 187 (2024) 3461–3495. https://doi.org/10.1016/j.cell.2024.05.053.
[4] D. Arendt, J.M. Musser, C.V.H. Baker, A. Bergman, C. Cepko, D.H. Erwin, M. Pavlicev, G. Schlosser, S. Widder, M.D. Laubichler, G.P. Wagner, The origin and evolution of cell types, Nat Rev Genet 17 (2016) 744–757. https://doi.org/10.1038/nrg.2016.127.
[5] A.J. Tarashansky, J.M. Musser, M. Khariton, P. Li, D. Arendt, S.R. Quake, B. Wang, Mapping single-cell atlases throughout Metazoa unravels cell type evolution, eLife 10 (2021) e66747. https://doi.org/10.7554/eLife.66747.
[6] E. Gallo, S. De Renzis, J. Sharpe, R. Mayor, J. Hartmann, Versatile system cores as a conceptual basis for generality in cell and developmental biology, Cell Systems (2024). https://doi.org/10.1016/j.cels.2024.08.001.

Thumbs up (No Ratings Yet)
Loading...

Tags: , , ,
Categories: Research

The bumpy journey to the signal that kicks off endosperm development

Posted by , on 9 September 2024

Here, Sara Simonini and Ueli Grossniklaus from the Institute of Plant and Microbial Biology and Zurich-Basel Plant Science Center, University of Zurich, tell the story behind the paper “A paternal signal induces endosperm proliferation upon fertilization in Arabidopsis”.

Fertilization is one of the most fascinating events during the development of an organism. In sexually reproducing multicellular organisms like animals and plants, fertilization involves the fusion of two gametes – a female egg cell and a male sperm. Gametes are highly specialized cells that, upon reaching maturity, await fertilization in a quiescent state. One way to achieve this is by inhibition of cell cycle progression, thus allowing gametes to arrest at a precise, stable stage. This aspect is crucial because uncontrolled gamete proliferation could have dramatic consequences, such as abortion of the progeny or a waste of resources.

Fertilization in plants is unique

When the egg cell and sperm fuse, their quiescent state is lifted and the cell cycle reactivated, so that the product of fertilization, the zygote, can initiate cell division. The molecular mechanisms that control the establishment of the quiescent state and its exit are still poorly understood.

In flowering plants, the fertilization event is rather unique as they produce two types of female gametes, called egg cell and central cell. During the process of double fertilization, the two female gametes are fertilized by one sperm cell each, giving rise to the embryo and endosperm, respectively, the latter being a placental-like, nourishing tissue that sustains embryonic growth.

Typically, the egg cell and central cell derive from consecutive mitotic events of the same haploid megaspore, making them genetically identical. However, despite their genetic similarity, the egg cell and central cell have distinct identities, unique transcriptomes, different DNA contents (the central cell is homodiploid at maturity), and behave very differently once fertilized.

The fertilized egg undergoes morphological changes soon after fertilization. It progressively elongates, and its nucleus strongly polarizes towards the apical domain of the cell. The first cell division occurs approximately 20-24 hours after fertilization, resulting in an apical cell (forming the embryo proper) and a basal cell (forming the suspensor). In contrast, the fertilized central cell takes a different rhythm, committing its first division to initiate endosperm production already about 6-8 hours after fertilization.

Figure 1. Ovule and developing seeds imaged with confocal microscopy. The central cell and the endosperm nuclei express a yellow fluorescent protein. The cell wall is labeled by propidium iodide. The first division of the central cell to produce the primary endosperm nuclei occurs just 4-6 hours after fertilization.

Cell cycle stage at which Arabidopsis gametes arrest

Over two decades ago, a hypothesis emerged suggesting the presence of a mechanism in the central cell that regulates the cell cycle, distinct from the one operating in the egg cell. The proposed idea was that a molecular brake prevents central cell division, and that fertilization acts as a trigger to release this brake, allowing division. This hypothesis stemmed from the observation of the rapid proliferation of the central cell after fertilization, as well as from the phenotypes exhibited by certain mutants where the central cell either divides in the absence of fertilization or is unable to divide once fertilized.

To understand fertilization’s impact on central cell quiescence, we initially determined the cell cycle stage at which the mature female gametes arrest. Quantifying DNA content in the female gametes is quite challenging as they cannot be collected in sufficient quantity for conventional ploidy analysis, such as flow cytometry. Our approaches involved propidium iodide staining to quantify DNA content, for which a reliable protocol was already established, and the imaging of histones tagged with fluorescent protein to infer the chromatin content in different nuclei of the ovule. These two approaches worked well and were reasonably straightforward. However, when it came to assessing DNA synthesis through nucleotide-analogue incorporation (EdU), well, we hit our head against a wall for about six months. It took a multitude of adjustments, trials, and a certain level of DIY attitude before we were able to establish a reliable, efficient protocol. But we made it!

It took a multitude of adjustments, trials, and a certain level of DIY attitude before we were able to establish a reliable, efficient protocol. But we made it!

The results of our ploidy analysis were both surprising and exciting. While we could confirm that the egg cell arrests in G2 as previously suggested, the central cell presented a completely different story. Its ploidy and behaviour suggested that its DNA synthesis (S phase) had initiated but not finished, and we could observe that fertilization was necessary for the S phase to be completed.

Figure 2. Ovules embedded in wax and sliced into 7μm thick sections. In this section, the central cell (cyan), the egg cell (pink) and the two synergids cells (orange) are clearly distinguishable. These sections are used for Laser Assisted Microdissection (LAM) microscopy, where single cell types can be cut with a laser and isolated. Here, we have used this technique for a transcriptome analysis of central cells at different time points around the moment of fertilization.

Finding the brake

Now that we knew the central cell is arrested in S phase, we wanted to identify the factor causing this arrest in DNA synthesis. Almost immediately, we considered RBR1, because it is a conserved cell cycle inhibitor known for regulating entry and progression through S-phase, and its absence causes central cells to proliferate in the absence of fertilization. The first confirmation that indeed RBR1 was our candidate came during a day at the microscope, observing the dynamics of an RBR1-YFP fusion protein during fertilization. For this type of experiments, we emasculated almost ready-to-bloom flowers by removing the stamens, so that self-pollination was avoided. The next day, we pollinated the pistils, marking the “0” time point. Then, after 4, 6, 8, 10, or 12 hours after pollination, we dissected the pistils and imaged the ovules using a multiphoton microscope. Normally, we pollinated between 8 and 9 in the morning, meaning that we had to spend quite some evenings at the microscope.

During these observations, we noticed that some central cells showed a RBR1-YFP signal, while others did not. After confirming the homozygosity of the RBR1-YFP line, it became evident that RBR1-YFP disappeared from the central cell only in fertilized ovules. This led us to the conclusion that something was degrading RBR1 at fertilization. Therefore, RBR1 acted as the brake, and fertilization somehow triggered RBR1 degradation, allowing the cell cycle to proceed.

Searching the signal that releases the brake

Just shortly after observing the turnover of RBR1 during fertilization, we received the sequencing results of transcriptomes from central cells at different time points before and shortly after fertilization that we had isolated by Laser-Assisted Microdissection (LAM). In practical terms, this technique allows us to isolate single cells from fixed, paraffin-embedded, and sliced tissues of interest. Completing this experiment took almost a year and a half for various reasons. The first significant obstacle was the global pandemic. We had just started to collect material when the institute went into a complete lockdown for about eight weeks, which meant that we lost at least two plant generations. The re-start was problematic too, because we had to do shifts to prevent overcrowding the labs, and experiments proceeded rather slowly. The second challenge was the time required make semi-thin sections of the material used for LAM. It takes approximately five days to gather enough material for a single replicate; our analysis covered four developmental time points, each performed in triplicate.

However, the results justified the long waiting time. Among the cell cycle-related genes potentially involved in RBR1 degradation, one D-type cyclin, CYCD7;1, caught our attention. Its expression peaked just around the moment when RBR1 is degraded in the central cell. Moreover, the literature indicated that CYCD7;1 is expressed only in stomata and pollen, and its ectopic expression in the female gametophyte was previously shown to induce proliferation of the unfertilized central cell. This led us to hypothesize that CYCD7;1 is paternally produced and stored in the sperm cells, and only upon fertilization, would CYCD7;1 be present in the same place and at the same time as RBR1, triggering its degradation. Observing CYCD7;1 messenger RNA location and delivery, as well as CYCD7;1 protein dynamics, confirmed our hypothesis. We also found that ectopic expression of CYCD7;1 in the central cell was sufficient to trigger RBR1 degradation and central cell division.

The only missing element was a visible phenotype. Mutant lines for CYCD7;1 (T-DNA and CRISPR-Cas9) were growing, and I (Sara) was confident in predicting the cycd7;1 mutant phenotype: paternal-effect seed abortion. This means that seeds would fail to develop when cycd7;1 mutant pollen was used as a male in a cross with a wild-type plant. Because RBR1 wouldn’t be degraded, the central cell wouldn’t divide, and no endosperm could be produced. However, upon inspecting the first cycd7;1 siliques under the microscope to evaluate the level of seed abortion, the result was hard to accept. All four cycd7;1 mutants I analysed exhibited a perfectly fine seed set – no seed abortion. We accepted the disappointing result that absence of paternal CYCD7;1 did not impact seed development. We went back to the LAM transcriptome, searching for alternative candidates, and stopped working on CYCD7;1. Sometime later, Ueli and I were having a meeting to discuss new hypotheses and strategies to further develop the project. As we revisited the CYCD7;1-related data, Ueli asked me which seed developmental stages I had been looking at for the phenotypical analysis, and he added “Do it again, look closer to the moment of fertilization”.

As we revisited the CYCD7;1-related data, Ueli asked me which seed developmental stages I had been looking at for the phenotypical analysis, and he added “Do it again, look closer to the moment of fertilization”.

That very afternoon, I sowed all the plant lines, and six weeks later, I made reciprocal crosses between wild-type and cycd7;1 plants again. This time, instead of looking at fully grown siliques, I sampled seeds at 12 hours after pollination, and the phenotype was evident: seeds generated by cycd7;1 pollen had fewer – or even no – endosperm nuclei compared to those derived from wild-type pollen. This meant that paternal delivery of CYCD7;1 is required for normal central cell division after fertilization. Central cells that receive a sperm cell lacking CYCD7;1 are blind to the fertilization event and do not divide immediately as they should. However, cycd7;1 mutant had no seed abortion, meaning that seed development can proceed normally even in absence of CYCD7;1 and, indeed, at 24 hours after pollination, cycd7;1-derived seeds showed endosperm proliferation. How can this happen? We hypothesized that other D-type cyclins, expressed from the maternal and/or paternal genome soon after fertilization, might compensate for CYCD7;1’s absence. This hypothesis turned out to be correct as we were able to delay endosperm proliferation even further when using pollen from plants mutated for four D-type cyclins.

Our results have not only addressed the fundamental question of how a cell determines the appropriate timing for division, but have also uncovered new and intriguing research directions. These include the understanding of how the central cell can arrest in S-phase, elucidating the mechanisms by which the CYCD7;1 messenger RNA is selectively stored in the sperm nucleus without degradation, and exploring the broader question of which other paternal or maternal signals regulate cell cycle arrest and progression in gametes. It also taught us the important lesson of formulating the right biological questions and designing the right strategies to address them. This is especially important when looking at developmental transitions, growth progression, and developmental processes in general: we cannot look at development if we do not take into consideration the time factor. 

Thumbs up (2 votes)
Loading...

Tags: , ,
Categories: Research

Catch up on Development presents… September webinar on cardiac development and regeneration

Posted by , on 6 September 2024

The 4 September 2024 Development presents… webinar was chaired by Development Senior Editor Alex Eve and featured two talks on the topic of cardiac development and regeneration. Catch up on the talks below.

Catch up on previous webinars and register for new ones.

Sign up to the Development presents… mailing list to learn about the upcoming webinars as they are announced.

Lydia Djenoune (MGH, Harvard Medical School)

Talk and Q&A

Keaton Schuster (New York University)

Talk and Q&A
Thumbs up (2 votes)
Loading...

Tags: , , , , ,
Categories: Development presents..., Video

Development presents… October webinar on environment, evolution and development

Posted by , on 4 September 2024


On the topic of environment, evolution and development, chaired by Development’s Guest Editor, Karen Sears (UCLA).

Wednesday 2 October – 15:00 BST

Girish Kale (University of Hohenheim)
‘Elevated temperature fatally disrupts nuclear divisions in the early Drosophila embryo’

Natasha Shylo (Stowers Institute for Medical Research)
‘Gastrulation and Left-Right patterning in veiled chameleons’

Sergio Menchero (The Francis Crick Institute)
‘Understanding temporal diversity in mammalian developmental programmes using marsupial single-cell transcriptomics’

At the speakers’ discretion, the webinar will be recorded for viewing on demand. To see the other webinars scheduled in our series, and to catch up on previous talks, please visit: thenode.biologists.com/devpres

Thumbs up (2 votes)
Loading...

Tags: , ,
Categories: Development presents...

August in preprints

Posted by , on 4 September 2024

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

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

Developmental biology

Cell Biology

Modelling

Tools & Resources

Developmental biology

| Patterning & signalling

Multiple Notch ligands in the synchronization of the segmentation clock

Marcos Wappner, Koichiro Uriu, Andrew C. Oates, Luis G. Morelli

Spatiotemporal Characterization of Cyclooxygenase Pathway Enzymes During Vertebrate Embryonic Development

Tess A. Leathers, Raneesh Ramarapu, Crystal D. Rogers

The multi-level effect of chlorpyrifos during clownfish metamorphosis

Mathieu Reynaud, Stefano Vianello, Shu-Hua Lee, Pauline Salis, Mélanie Dusseune, Kai Wu, Bruno Frederich, David Lecchini, Laurence Besseau, Natacha Roux, Vincent Laudet

Retinoic acid breakdown is required for proximodistal positional identity during amphibian limb regeneration

 Timothy J. Duerr, Melissa Miller, Sage Kumar, Dareen Bakr, Jackson R. Griffiths, Aditya K. Gautham, Danielle Douglas, S. Randal Voss, James R. Monaghan

Post-eclosion growth in the Drosophila Ejaculatory Duct is driven by Juvenile Hormone signaling and is essential for male fertility

Navyashree A Ramesh, Allison M. Box, Laura Buttitta

Eomes directs the formation of spatially and functionally diverse extra-embryonic hematovascular tissues

Bart Theeuwes, Luke TG Harland, Alexandra Bisia, Ita Costello, Mai-Linh Ton, Tim Lohoff, Stephen J Clark, Ricard Argelaguet, Nicola K Wilson, Wolf Reik, Elizabeth Bikoff, Elizabeth J Robertson, Berthold Gottgens

ETS TRANSCRIPTION FACTOR POINTED CONTROLS GERMLINE SURVIVAL IN DROSOPHILA

Alicia E. Rosales-Nieves, Miriam Marín-Menguiano, Lourdes López-Onieva, Juan Garrido-Maraver, Acaimo González-Reyes

Blastocoel expansion and AMOT degradation cooperatively promote YAP nuclear localization during epiblast formation

Hinako Maeda, Hiroshi Sasaki

Mycn regulates vascular development through PI3K signaling pathway in zebrafish

Guo-Qin Zhao, Tao Cheng, Peng-Yun Wang, Jing Mo, Feng Yu, Yang Dong, Yun-Fei Li, Yu Feng, Peng-Fei Xu, Li-Ping Shu

From Zhao et al. This image is made available under a CC-BY 4.0 International license.

NOTCH-driven oscillations control cell fate decisions during intestinal homeostasis

 View ORCID ProfileSonja D. C. Weterings, Hiromune Eto, Jan-Daniël de Leede, Amir Giladi, Mirjam E. Hoekstra, Wouter F. Beijk, Esther J. M. Liefting, Karen B. van den Anker, Jacco van Rheenen, Alexander van Oudenaarden, Katharina F. Sonnen

Canonical Wnt signalling from the Area Opaca induces and maintains the Marginal Zone in pre-primitive-streak stage chick embryos

 View ORCID ProfileYara Fadaili, Hui-Chun Lu, Hyung Chul Lee, Amra Ryazapova, Claudio D. Stern

Effects Of Aryl Hydrocarbon Receptor Ligand TCDD On Human Trophoblast Cell Development

 View ORCID ProfileVinay Shukla, Khursheed Iqbal, Hiroaki Okae, Takahiro Arima, Michael J. Soares

Comprehensive profiling of migratory primordial germ cells reveals niche-specific differences in non-canonical Wnt and Nodal-Lefty signaling in anterior vs posterior migrants

 View ORCID ProfileRebecca Garrett Jaszczak, Jay W Zussman, Daniel E Wagner, Diana J Laird

| Morphogenesis & mechanics

Material Properties Of The Embryonic Small Intestine During Buckling Morphogenesis

Jenny Gao, Lucia Martin, Elise A. Loffet, John F. Durel, Panagiotis Oikonomou, Nandan L. Nerurkar

BMP signalling attenuates intercellular adhesion to drive mesenchyme migration during fin fold morphogenesis

Bitan Saha, Harsha Mahabaleshwar, Charmaine Ho Min, Leslie Boon Haw Leong, Levene Wenqian Chua, Samuel Kwok, Tom J Carney

Morphogenic, molecular, and cellular adaptations for unidirectional airflow in the chicken lung

Kamryn N Gerner-Mauro, Lisandra Vila Ellis, Guolun Wang, Richa Nayak, Peter Y Lwigale, Ross A Poché, Jichao Chen

Mll4 regulates tooth enamel development

Jung-Mi Lee, Hunmin Jung, Qinghuang Tang, Woojung An, Soo-Kyung Lee, Jae W. Lee, Yungki Park, Hyuk-Jae Edward Kwon

Developmental control of E-cadherin junctions by mechanical contractility in Drosophila embryos

Na Zhang, Wangfang Liu, Huiying Lu, Matthias Haering, Fred Wolf, Joerg Grosshans, Zhiyi Lv, Deqing Kong

Mast cells are not essential for pubertal mammary gland branching.

Simran Kapoor, Jimmy Marsden, Clara M Munz, Cyril Carvalho, Marlene Magalhaes Pinto, Bert Malengier-Devlies, Solvig Becker, Guillaume Seuzaret, Katelyn Patatsos, Ramazan Akyol, Marc DALOD, Amy B Pedersen, Gillian J Wilson, Rebecca Gentek

| Genes & genomes

PROSER1 Modulates DNA Demethylation through Dual Mechanisms to Prevent Syndromic Developmental Malformations

Anna Fleming, Elena V. Knatko, Xiang Li, Ansgar Zoch, Zoe Heckhausen, Stephanie Stransky, Alejandro J. Brenes, Simone Sidoli, Petra Hajkova, Dónal O’Carroll, Kasper D. Rasmussen

Members of an array of zinc finger proteins specify distinct Hox chromatin boundaries

Havva Ortabozkoyun, Pin-Yao Huang, Edgar Gonzalez-Buendia, Hyein Cho, Sang Y. Kim, Aristotelis Tsirigos, Esteban O. Mazzoni, Danny Reinberg

Structural perturbation of chromatin domains with multiple developmental regulators can severely impact gene regulation and development

Shreeta Chakraborty, Nina Wenzlitschke, Matthew J. Anderson, Ariel Eraso, Manon Baudic, Joyce J. Thompson, Alicia A. Evans, Lilly M. Shatford‑Adams, Raj Chari, Parirokh Awasthi, Ryan K. Dale, Mark Lewandoski, Timothy J. Petros, Pedro P. Rocha

Temporally restricted activities of En1 regulatory elements underlie distinct limb malformations

Alessa R. Ringel, Andreas Magg, Natalia Benetti, Robert Schöpflin, Mira Kühnlein, Asita Carola Stiege, Ute Fischer, Lars Wittler, Stephan Lorenz, Stefan Mundlos, Lila Allou

Mechanistic basis of lineage restriction

Bohou Wu, Jae Hyun Lee, Kara M. Foshay, Li Zhang, Croydon J. Fernandes, Boyang Gao, Xiaoyang Dou, Chris Z. Zhang, Guoping Fan, Becky X. Xiao, Bruce T. Lahn

A sperm-enriched 5’fragment of tRNA-Valine regulates preimplantation embryonic transcriptome and development

Simeiyun Liu, Andrew D. Holmes, Sol Katzman, Upasna Sharma

Chromatin remodeler CHD4 establishes chromatin states required for ovarian reserve formation, maintenance, and germ cell survival

Yasuhisa Munakata, Mengwen Hu, Yuka Kitamura, Adam L Bynder, Amelia S Fritz, Richard M Schultz, Satoshi H Namekawa

Diverse somatic Transformer and sex chromosome karyotype pathways regulate gene expression in Drosophila gonad development

Sharvani Mahadevaraju, Soumitra Pal, Pradeep Bhaskar, Brennan D. McDonald, Leif Benner, Luca Denti, Davide Cozzi, Paola Bonizzoni, Teresa M. Przytycka, Brian Oliver

A newly evolved gene is essential for efficient sperm entry into eggs in Drosophila melanogaster

Sara Y. Guay, Prajal H. Patel, Jonathon M. Thomalla, Kerry L. McDermott, Jillian M. O’Toole, Sarah E. Arnold, Sarah J. Obrycki, Mariana F. Wolfner, Geoffrey D. Findlay

Mechanistic origin and preimplantation development of uniparental and polyploid blastomeres

Yan Zhao, Andrea Fernández-Montoro, Greet Peeters, Tatjana Jatsenko, Tine De Coster, Daniel Angel-Velez, Thomas Lefevre, Thierry Voet, Olga Tšuiko, Ants Kurg, Katrien Smits, Ann Van Soom, Joris Robert Vermeesch

Dynamic Wt1 expression in the gastrulation-stage mouse embryo specifies vascular and visceral smooth muscle cell fate independently from mesothelial fate.

Suad Hassan Alsukari, Huei Teng Ng, Lilly Lang, Sharna Lunn, Shanthi Beglinger, Lauren Carr, Michael Boyes, David Andrew Turner, Bettina Wilm

The E3 ubiquitin ligase RNF220 maintains hindbrain Hox expression patterns through regulation of WDR5 stability

Huishan Wang, Xingyan Liu, Yamin Liu, Chencheng Yang, Yaxin Ye, Xiaomei Yu, Nengyin Sheng, Shihua Zhang, Bingyu Mao, Pengcheng Ma

Mesenchymal Meis2 controls whisker development independently from trigeminal sensory innervation

Mehmet Mahsum Kaplan, Erika Hudacova, Miroslav Matejcek, Haneen Tuaima, Jan Krivanek, Ondrej Machon

From Kaplan et al. This image is made available under a CC-BY 4.0 International license.

Neuronal progenitors suffer genotoxic stress in the Drosophila clock mutant per0

Nunzia Colonna Romano, Marcella Marchetti, Anna Marangoni, Laura Leo, Diletta Retrosi, Ezio Rosato, Laura Fanti

Mutations in ErbB2 accumulating in the male germline measured by error-corrected sequencing

Atena Yasari, Monika Heinzl, Theresa Mair, Tina Karimian, Shehab Moukbel Ali Aldawla, Ingrid Hartl, Andrea J. Betancourt, Peter Lanzerstorfer, Irene Tiemann-Boege

Traffic Jam activates the Flamenco piRNA cluster locus and the Piwi pathway to ensure transposon silencing and Drosophila fertility

Austin Rivera, Jou-Hsuan Roxie Lee, Shruti Gupta, Linda Yang, Raghuveera Kumar Goel, Joseph Zaia, Nelson C. Lau

The Transcription Factor TCF21 is necessary for adoption of cell fates by Foxd1+ stromal progenitors during kidney development

Gal Finer, Mohammad D. Khan, Yalu Zhou, Gaurav Gadhvi, George S. Yacu, Joo-Seop Park, R. Ariel Gomez, Maria Luisa Sequeira-Lopez, Susan E. Quaggin, Deborah R. Winter

Fate specification triggers a positive feedback loop of TEAD–YAP and NANOG to promote epiblast formation in preimplantation embryos

Naoki Hirono, Masakazu Hashimoto, Hinako Maeda, Hiromi Shimojo, Hiroshi Sasaki

Distinct checkpoint and homolog biorientation pathways regulate meiosis I in Drosophila oocytes

Joanatta G. Shapiro, Neha Changela, Janet K. Jang, Jay N. Joshi, Kim S. McKim

A whole-organism landscape of X-inactivation in humans

Björn Gylemo, Maike Bensberg, Colm E. Nestor

Epigenetic and transcriptional regulation of ovarian development altered in ErβKO ovaries

Ryan Mohamadi, Kevin Vo, Yashica Sharma, Amelia Mohamadi, Elizabeth S. Bahadursingh, Patrick E. Fields, M. A. Karim Rumi

Castor is a temporal transcription factor that specifies early born central complex neuron identity

Noah R. Dillon, Chris Q. Doe

From Dillon et al. This image is made available under a CC-BY 4.0 International license.

Dynamic convergence of autism disorder risk genes across neurodevelopment

Meilin Fernandez Garcia, Kayla Retallick-Townsley, April Pruitt, Elizabeth Davidson, Yi Dai, Sarah E. Fitzpatrick, Annabel Sen, Sophie Cohen, Olivia Livoti, Suha Khan, Grace Dossou, Jen Cheung, P.J. Michael Deans, Zuoheng Wang, Laura Huckins, Ellen Hoffman, Kristen Brennand

H3K4me2 distinguishes a distinct class of enhancers during the maternal-to-zygotic transition

Matthew D. Hurton, Jennifer M. Miller, Miler T. Lee

Early Hox Gene Expression in Echinoderms

 Olga V. Ezhova, Natalya V. Ageenko, Konstantin V. Kiselev, Anastasiya I. Lukinykh, Vladimir V. Malakhov

A sperm-enriched 5’fragment of tRNA-Valine regulates preimplantation embryonic transcriptome and development

Simeiyun Liu, Andrew D. Holmes, Sol Katzman, Upasna Sharma

A master regulatory loop that activates genes in a temporally coordinated manner in muscle cells of ascidian embryos

Izumi Oda, Yutaka Satou

The Transcription Factor TCF21 is necessary for adoption of cell fates by Foxd1+ stromal progenitors during kidney development

Gal Finer, Mohammad D. Khan, Yalu Zhou, Gaurav Gadhvi, George S. Yacu, Joo-Seop Park, R. Ariel Gomez, Maria Luisa Sequeira-Lopez, Susan E. Quaggin, Deborah R. Winter

Differential regulation of eye specification in Drosophila by Polycomb Group (PcG) epigenetic repressors

Haley E. Brown, Brandon P. Weasner, Justin P. Kumar

A dual ribosomal system in the zebrafish soma and germline

Arish N Shah, Friederike Leesch, Laura Lorenzo-Orts, Lorenz Grundmann, Maria Novatchkova, David Haselbach, Eliezer Calo, Andrea Pauli

| Stem cells, regeneration & disease modelling

The transcription factor Chronophage/BCL11A/B promotes intestinal stem cell proliferation and endocrine differentiation in the Drosophila intestine

Emer Aisling King, Eleanor Jacobsen, Nicholas Woolner, Joaquín de Navascués, Owen J Marshall, Jerome Korzelius

PDK-1/S6K and mTORC1 bypass systemic growth restrictions to promote regeneration

Ananthakrishnan Vijayakumar Maya, Liyne Nogay, Lara Heckmann, Isabelle Grass, Katrin Kierdorf, Jörg Büscher, Anne-Kathrin Classen

Canonical and non-canonical PRC1 differentially contribute to the regulation of neural stem cell fate

Janine Hoffmann, Theresa M. Schütze, Annika Kolodziejczyk, Annekathrin Kränkel, Susanne Reinhardt, Razvan P. Derihaci, Cahit Birdir, Pauline Wimberger, Haruhiko Koseki, Mareike Albert

Serotonin neuromodulation directs optic nerve regeneration

 Kristian Saied-Santiago, Melissa Baxter, Jaffna Mathiaparanam, Michael Granato

Compound screening in primary human airway basal cells identifies Wnt pathway activators as potential pro-regenerative therapies

Yuki Ishii, Jessica C. Orr, Marie-Belle El Mdawar, Denise R. Bairros de Pilger, David R. Pearce, Kyren A. Lazarus, Rebecca E. Graham, Marko Z. Nikolic, Robin Ketteler, Neil O. Carragher, Sam M. Janes, Robert E. Hynds

A Sox2 Enhancer Cluster Regulates Region-Specific Neural Fates from Mouse Embryonic Stem Cells

Ian C Tobias, Sakthi D Moorthy, Virlana M Shchuka, Lida Langroudi, Mariia Cherednychenko, Zoe E Gillespie, Andrew G Duncan, Ruxiao Tian, Natalia A Gajewska, Raphaël B Di Roberto, Jennifer A Mitchell

The level of HAND1 controls the specification of multipotent cardiac and extraembryonic progenitors

Adam T Lynch, Naomi Phillips, Megan Douglas, Marta Dorgnach, I-Hsuan Lin, Antony D Adamson, Zoulfia Darieva, Jessica Whittle, Neil A Hanley, Nicoletta Bobola, Matthew J Birket

The microbiota affects stem cell decision making in Hydra

Jinru He, Alexander Klimovich, Sabine Kock, Linus Dahmke, Sören Franzenburg, Thomas C.G. Bosch

Proliferative events ameliorate DNA damage accumulation without affecting function in hematopoietic stem cells

Shubham Haribhau Mehatre, Harsh Agrawal, Irene Mariam Roy, Sarah Schouteden, Satish Khurana

Appendage regeneration requires IMPDH2 and creates a sensitized environment for enzyme filament formation

Morgan E. McCartney, Gavin M. Wheeler, Audrey G. O’Neill, Jeet H. Patel, Zoey R. Litt, S. John Calise, Justin M. Kollman, Andrea E. Wills

Chromatin activity of IκBα mediates the exit from naïve pluripotency

Luis G. Palma, Daniel Álvarez-Villanueva, María Maqueda, Mercedes Barrero, Arnau Iglesias, Joan Bertran, Damiana Álvarez-Errico, Carlos A. García-Prieto, Cecilia Ballaré, Virginia Rodriguez-Cortez, Clara Bueno, August Vidal, Alberto Villanueva, Pablo Menéndez, Gregoire Stik, Luciano Di Croce, Bernhard Payer, Manel Esteller, Lluís Espinosa, Anna Bigas

Voltage-gated calcium channels generate blastema Ca2+ fluxes restraining zebrafish fin regenerative outgrowth

Heather K. Le Bleu, Rea G. Kioussi, Astra L. Henner, Victor M. Lewis, Scott Stewart, Kryn Stankunas

FGF9 treatment reduces off-target chondrocytes from iPSC-derived kidney organoids

Virginie Joris, Anika Schumacher, Paula Marks, Maria Eischen-Loges, Martijn van Griensven, Vanessa L.S. LaPointe

From Joris et al. This image is made available under a CC-BY 4.0 International license.

Transcriptomic analysis and high throughput functional characterization of human induced pluripotent stem cell derived sensory neurons

Vincent Truong, Jackson Brougher, Tim Strassmaier, Irene Lu, Dale George, Theodore J. Price, Alison Obergrussberger, Aaron Randolph, Rodolfo J. Haedo, Niels Fertig, Patrick Walsh

Mitochondrial activity-driven hematopoietic stem cell fate and lympho-myeloid lineage choice is first established in the aorta-gonad-mesonephros

Aishwarya Prakash, Maneesha S. Inamdar

Readaptation of mesenchymal stem cells to high stiffness and oxygen environments modulate the extracellular matrix

Inês Caramelo, Catarina Domingues, Vera M. Mendes, Sandra I. Anjo, Margarida Geraldo, Carla M. P. Cardoso, Mário Grãos, Bruno Manadas

Metadichol induces CD14 Glycoprotein Expression in Human Embryonic Stem Cells and Fibroblasts

P. R. Raghavan

Nuclear actin is a critical regulator of Drosophila female germline stem cell maintenance

Nicole M. Green, Danielle Talbot, Tina L. Tootle

Survival of hepatocytes from executioner caspase activation promotes liver regeneration by enhancing JAK/STAT3 activity

Zhiyuan Cao, Lining Qin, Kaixuan Liu, Chen Yao, Enhong Li, Xiaoyu Hao, Molin Wang, Baichun Jiang, Yongxin Zou, Huili Hu, Qiao Liu, Changshun Shao, Yaoqin Gong, Gongping Sun

Posterior specification of multi-lineage axial assembloids from human pluripotent stem cells.

Nigel Kee, Mélanie Leboeuf, Silvia Gómez, Charles Petipré, Irene Mei, Salim Benlefki, Daniel W Hagey, José Dias, François Lallemend, Samir EL Andaloussi, Johan Ericson, Eva Hedlund

| Plant development

snRNA-seq analysis of the moss Physcomitrium patens reveals a conserved cytokinin-ESR module promoting pluripotent stem cell identity

Yuki Hata, Nicola Hetherington, Kai Battenberg, Atsuko Hirota, Aki Minoda, Makoto Hayashi, Junko Kyozuka

Knockout of the tomato HAIRY MERISTEM 4 alters phloem-characteristics and impairs development

Jackson Khedia, Abhay Pratap Vishwakarma, Ortal Galsurker, Shira Corem, Suresh Kumar Gupta, Tzahi Arazi

The contrasting flowering-time among coffee genotypes is associated with ectopic and differential expressions of genes related to environment, floral development, and hormonal regulation

Marlon Enrique López, Raphael Ricon de Oliveira, Lillian Magalhães Azevedo, Iasminy Silva Santos, Thales Henrique Cherubino Ribeiro, Dapeng Zhang, Antonio Chalfun-Junior

Physiological and Molecular Responses of Projected Future Temperatures on Potato Tuberization

Abigail M. Guillemette, Guillian Hernández Casanova, John P. Hamilton, Eva Pokorná, Petre I. Dobrev, Václav Motyka, Aaron M. Rashotte, Courtney P. Leisner

Genome sequence of the ornamental plant Digitalis purpurea reveals the molecular basis of flower color and morphology variation

Jakob Maximilian Horz, Katharina Wolff, Ronja Friedhoff, Boas Pucker

From Horz et al. This image is made available under a CC-BY 4.0 International license.

Single-nucleus transcriptomics resolves differentiation dynamics between shoot stem cells and primary stem

Sebastián R. Moreno, Martin O. Lenz, Elliot M Meyerowitz, James CW Locke, Henrik Jönsson

A cell fractionation and quantitative proteomics pipeline to enable functional analyses of cotton fiber development

Youngwoo Lee, Heena Rani, Eileen L. Mallery, Daniel B Szymanski

Viral delivery of an RNA-guided genome editor for transgene-free germline editing in Arabidopsis

Trevor Weiss, Maris Kamalu, Honglue Shi, Zheng Li, Jasmine Amerasekera, Zhenhui Zhong, Benjamin A Adler, Michelle Song, Kamakshi Vohra, Gabriel Wirnowski, Sidharth Chitkara, Charlie Ambrose, Noah Steinmetz, Ananya Sridharan, Diego Sahagun, Jill Banfield, Jennifer Doudna, Steven E. Jacobsen

Natural variation suggests new mechanisms for bract development in Arabidopsis, desynchronising bract suppression from the floral transition

Sana Dieudonne Dr., Ruth Kristianingsih Ms, Stephanie Laine Ms, Beline JESSON Ms, Veronique VIDAL Ms, Rachel Wells Dr., Richard Morris Pr., Fabrice Besnard Dr.

MAC3A and MAC3B modulate FLM splicing to repress photoperiod-dependent floral transition

Yu-Wen Huang, Chih-Yen Tseng, Yi-Tsung Tu, Hsin-Yu Hsieh, Yu-Sen Wang, Yun-Tung Ly, Yu-Zhen Chen, Shih-Long Tu, Chin-Mei Lee

Conserved role of the SERK–BIR module in development and immunity across land plants

Yijia Yan, Jaqueline Mellüh, Martin A. Mecchia, Hyung-Woo Jeon, Katharina Melkonian, Clemens Holzberger, Anne Harzen, Sara Christina Stolze, Rainer Franzen, Yuki Hirakawa, Ana I. Caño Delgado, Hirofumi Nakagami

A leaf-expressed TERMINAL FLOWER1 ortholog from coffee with alternate splice forms alters flowering time and inflorescence branching in Arabidopsis

 Carlos Henrique Cardon, Victoria Lesy, Catherine Fust, Thales Henrique Cherubino Ribeiro, Owen Hebb, Raphael Ricon de Oliveira, Mark Minow, Antonio Chalfun Junior, Joseph Colasanti

Two distinct oscillatory auxin signals define the plasticity of lateral rooting in Arabidopsis thaliana

Chengzhi Ren, Jule Bodendorf, Jurgen Kleine-Vehn

Natural alleles of LEAFY and WAPO1 interact to regulate spikelet number per spike in wheat

Junli Zhang, German F Burguener, Francine Paraiso, Jorge Dubcovsky

Developmental variability in cotton fiber cell wall properties linked to important agronomic traits

Michael C Wilson, Alexander H Howell, Anika Sood, Youngwoo Lee, Pengcheng Yang, Heena Rani, Elena Yu, Eileen L. Mallery, Sivakumar Swaminathan, Corrinne E. Grover, Jonathan F. Wendel, Olga A. Zabotina, Jun Xie, Chelsea S. Davis, Daniel Szymanski

Flower bud cooling protects pollen development and improves fertility during heatwaves

Martijn J. Jansen, Stuart Y. Jansma, Klaske M. Kuipers, Wim H. Vriezen, Frank F. Millenaar, Teresa Montoro, Carolien G.F. de Kovel, Fred A. van Eeuwijk, Eric J.W. Visser, Ivo Rieu

Assessing male reproductive investment in Papaveraceae using flow cytometry reveals lineage-specific trajectories to pollen-to-ovule ratio reduction

Jurriaan M. de Vos, Yannick Woudstra, Ilia J. Leitch, Oriane Hidalgo

The three-dimensional anatomy and dorsoventral asymmetry of the mature Marchantia polymorpha meristem develops from a symmetrical gemma meristem

Victoria Spencer, Eva-Sophie Wallner, Katharina Jandrasits, Natalie Edelbacher, Magdalena Mosiolek, Liam Dolan

OsWHY1/OsTRXz/OsMORFs complex is essential for RNA modification and early chloroplast development in rice

Xiangzi Zheng, Qingzheng Lu, Yuling Luo, Jiaxuan Xu, Weiqi Wang, Min Tan, Dongmei Liao, Wuqiang Hong, Sirong Chen, Chuheng Lin, Xiaoli Wang, Chunlan Fan, Habiba, Xiaowei Wang, Yanyun Li, Yu Zhang, Wenfang Lin, Ying Miao

Transcriptomic dynamics of petal development in the one-day flower species, Japanese morning glory (Ipomoea nil)

Soya Nakagawa, Atsushi Hoshino, Kazuyo Ito, Hiroyo Nishide, Katsuhiro Shiratake, Atsushi J Nagano, Yasubumi Sakakibara

Guard-cell phytosterol homeostasis is critical for proper stomatal development

Chih-Chung Yen, Ya-Wen Hsu, Kuan-Chieh Leu, Sheau-Shyang Chen, Tzu-Yun Chen, Chien-Ta Juan, Chi Kuan, Jei-Fu Shaw, Chin-Min Kimmy Ho, Guang-Yuh Jauh

The receptor-like kinase ALE2 promotes giant cell formation in the sepal epidermis

Frances K Clark, Jessica McGory, Nicholas Russell, Pau Formosa-Jordan, Adrienne H. K. Roeder

Multi-scale dynamics influence the division potential of stomatal lineage ground cells in Arabidopsis

Hannah F. Fung, Gabriel O. Amador, Renee Dale, Yan Gong, Macy Vollbrecht, Joel M. Erberich, Andrea Mair, Dominique C. Bergmann

From Fung et al. This image is made available under a CC-BY 4.0 International license.

| Evo-devo

Reverse development in the ctenophore Mnemiopsis leidyi

Joan J. Soto-Angel, Pawel Burkhardt

Protein degradation shapes developmental tempo in mouse and human neural progenitors

Shota Nakanoh, Despina Stamataki, Lorena Garcia-Perez, Chiara Azzi, Hayley L Carr, Alexandra Pokhilko, Lu Yu, Steven Howell, Mark Skehel, David Oxley, Simon Andrews, James Briscoe, Teresa Rayon

A punctuated burst of massive genomic rearrangements and the origin of non-marine annelids

Carlos Vargas-Chávez, Lisandra Benítez-Álvarez, Gemma I. Martínez-Redondo, Lucía Álvarez-González, Judit Salces-Ortiz, Klara Eleftheriadi, Nuria Escudero, Nadège Guiglielmoni, Jean-François Flot, Marta Novo, Aurora Ruiz-Herrera, Aoife McLysaght, Rosa Fernández

Developmental plasticity and genetic selection shaped cereal evolution in the Early Holocene southern Levant

Jade Whitlam, Pascal Flohr, Amy Bogaard, Mike Charles, Bill Finlayson, Cheryl A. Makarewicz

A c-opsin functions in a ciliary-marginal zone-like stem cell region of an invertebrate camera-type eye

Nadja Milivojev, Camila L. Velastegui Gamboa, Gabriele Andreatta, Florian Raible, Kristin Tessmar-Raible

Mammalian retinal specializations for high acuity vision evolve in response to both foraging strategies and morphological constraints

Emily E. K. Kopania, Nathan L. Clark

Microglia cannibalism and efferocytosis leads to shorter lifespans of developmental microglia

Hannah Gordon, Zachary Schafer, Cody J. Smith

Functional divergence of conserved developmental plasticity genes between two distantly related nematodes

Sara Wighard, Hanh Witte, Ralf J. Sommer

Less, but more: new insights from appendicularians on chordate Fgf evolution and the divergence of tunicate lifestyles

Gaspar Sánchez-Serna, Jordi Badia-Ramentol, Paula Bujosa, Alfonso Ferrández-Roldán, Nuria P. Torres-Águila, Marc Fabregà-Torrus, Johannes N. Wibisana, Michael J. Mansfield, Charles Plessy, Nicholas M. Luscombe, Ricard Albalat, Cristian Cañestro

From Sánchez-Serna et al. This image is made available under a CC-BY-NC 4.0 International license.

Cell Biology

Polo-like kinase 1 prevents excess microtubule polymerization in C. elegans oocytes to ensure faithful meiosis

Juhi G. Narula, Sarah M. Wignall

Sirt5 regulates chondrocyte metabolism and osteoarthritis development through protein lysine malonylation

Huanhuan Liu, Anupama Binoy, Siqi Ren, Thomas C. Martino, Anna E. Miller, Craig R. G. Willis, Shivakumar R. Veerabhadraiah, Abhijit Sukul, Joanna Bons, Jacob P. Rose, Birgit Schilling, Michael J. Jurynec, Shouan Zhu

Actin dynamics switches two distinct modes of endosomal fusion in yolk sac visceral endoderm cells

Seiichi Koike, Masashi Tachikawa, Motosuke Tsutsumi, Takuya Okada, Tomomi Nemoto, Kazuko Keino-Masu, Masayuki Masu

A maternally inherited Chromosomal Passenger Complex regulates germ plasm ribonucleoparticle aggregation in Zebrafish

Cara Moravec, Francisco Pelegri

AalpiRNA-18529 regulates vitellogenesis of Aedes albopictus via the Gadd45a-mediated JNK-dependent nurse cell apoptosis pathway

Lu Yang, Yonghui Gao, Yulan Chen, Shuyi Ren, Yifan Guo, Peiwen Liu, Khadija Batool, Jianxia Tang, Jinbao Gu

From Yang et al. This image is made available under a CC-BY 4.0 International license.

Versatile gamma-tubulin complexes contribute to the dynamic organization of MTOCs during Drosophila spermatogenesis

Elham Alzyoud, Dóra Németh, Viktor Vedelek, Titanilla Szögi, Viktória Petra Tóth, Mónika Krecsmarik, Edit Ábrahám, Zoltán Lipinszki, Rita Sinka

Proteome asymmetry in mouse and human embryos before fate specification

Lisa K. Iwamoto-Stohl, Aleksandra A. Petelski, Maciej Meglicki, Audrey Fu, Saad Khan, Harrison Specht, Gray Huffman, Jason Derks, Victoria Jorgensen, Bailey A.T. Weatherbee, Antonia Weberling, Carlos W. Gantner, Rachel S. Mandelbaum, Richard J. Paulson, Lisa Lam, Ali Ahmady, Estefania Sanchez Vasquez, Nikolai Slavov, Magdalena Zernicka-Goetz

The PIDDosome controls cardiomyocyte polyploidization during postnatal heart development

M Leone, N Kinz, F Eichin, D Obwegs, VC Sladky, D Rizzotto, C Manzl, K Moos, ED Jacotot, C Savko, MA Sussman, M Boerries, A Villunger

Erythropoietin-dependent Acquisition of CD71hiCD105hi Phenotype within CD235a- Early Erythroid Progenitors

Natascha Schippel, Jing Wei, Xiaokuang Ma, Mrinalini Kala, Shenfeng Qiu, Peter Stoilov, Shalini Sharma

Sustained fertility from first-wave follicle oocytes that pause their growth

Bikem Soygur, Eliza A. Gaylord, Mariko H. Foecke, Steven A. Cincotta, Tegan S. Horan, Anna Wood, Paula E. Cohen, Diana J. Laird

Modelling

Physical modeling of embryonic transcriptomes identifies collective modes of gene expression

 Dominic J. Skinner, Patrick Lemaire, Madhav Mani

Computing hematopoietic stem and progenitor cell plasticity in response to genetic mutations and environmental stimulations

Yuchen Wen, Hang He, Yunxi Ma, Lorie Chen Cai, Huaquan Wang, Yanmei Li, Baobing Zhao, Zhigang Cai

Spatial model of cell-fate choice uncovers strong links between tissue morphology and tissue regeneration

Somya Mani, Tsvi Tlusty

Role of Data-driven Regional Growth Model in Shaping Brain Folding Patterns

Jixin Hou, Zhengwang Wu, Xianyan Chen, Dajiang Zhu, Tianming Liu, Gang Li, Xianqiao Wang

A Computational Framework for Modeling Emergence of Color Vision in the Human Brain

Atsunobu Kotani, Ren Ng

Streamline tractography of the fetal brain in utero with machine learning

Weide Liu, Camilo Calixto, Simon K. Warfield, Davood Karimi

Multiple Notch ligands in the synchronization of the segmentation clock

Marcos Wappner, Koichiro Uriu, Andrew C. Oates, Luis G. Morelli

Tools & Resources

Silk-Ovarioids: Establishment and characterization of human ovarian primary cells 3D-model system

Valentina Di Nisio, Tianyi Li, Zhijie Xiao, Kiriaki Papaikonomou, Anastasios Damdimopoulos, Ákos Végvári, Filipa Lebre, Ernesto Alfaro-Moreno, Mikael Pedersen, Terje Svingen, Roman Zubarev, Ganesh Acharya, Pauliina Damdimopoulou, Andres Salumets

Phylogeny, morphology, and behavior of the new ciliate species Stentor stipatus

D. Rajan, B. Lee, A. Albright, E. Tang, A. Maravillas, C. Vargas, W. F. Marshall, D. Cortes

CRISPR-Cas13d as a molecular tool to achieve targeted gene expression knockdown in chick embryos

Minyoung Kim, Erica J. Hutchins

Compatibility of time-lapse dry incubator on in vitro production of bovine embryos

Haruhisa Tsuji, Hiroki Nagai, Sayaka Kobinata, Hinata Koyama, Atchalalt Khurchabilig, Noritaka Fukunaga, Yoshimasa Asada, Satoshi Sugimura

A transgene-free, human peri-gastrulation embryo model with trilaminar embryonic disc-, amnion- and yolk sac-like structures

Shiyu Sun, Yi Zheng, Yung Su Kim, Zheng Zhong, Norio Kobayashi, Xufeng Xue, Yue Liu, Zhuowei Zhou, Yanhong Xu, Jinglei Zhai, Hongmei Wang, Jianping Fu

Morphological criteria for staging near-hatching embryos of the domesticated Mallard (Anas platyrhynchos) and Swan Goose (Anser cygnoides)

 Bassel Arnaout, Kaylen Brzezinski, Benjamin Steventon, Daniel J. Field

From Arnaout et al. This image is made available under a CC-BY 4.0 International license.

Three-dimensional culture in a bioengineered matrix and somatic cell complementation to improve growth and survival of bovine preantral follicles

 Juliana I. Candelaria, Ramon C. Botigelli, Carly Guiltinan, Ariella Shikanov, Anna C. Denicol

Centimeter-scale, physiologically relevant intestinal organoids generated entirely from pluripotent stem cells

Zhen Qi, Zhanguang Zuo, Yi Dong, Jingyu Shao, Chong Wang, Rosanna Zhang

A human induced pluripotent stem (hiPS) cell model for the holistic study of epithelial to mesenchymal transitions (EMTs)

Caroline Hookway, Antoine Borensztejn, Leigh K. Harris, Sara Carlson, Gokhan Dalgin, Suraj Mishra, Nivedita Nivedita, Ellen M. Adams, Tiffany Barszczewski, Julie C. Dixon, Jacqueline H. Edmonds, Erik A. Ehlers, Alexandra J. Ferrante, Margaret A. Fuqua, Philip Garrison, Janani Gopalan, Benjamin W. Gregor, Maxwell J. Hedayati, Kyle N. Klein, Chantelle L. Leveille, Sean L. Meharry, Haley S. Morris, Gouthamrajan Nadarajan, Sandra A. Oluoch, Serge E. Parent, Amber Phan, Brock Roberts, Emmanuel E. Sanchez, M. Filip Sluzewski, Lev S. Snyder, Derek J. Thirstrup, John Paul Thottam, Julia R. Torvi, Gaea Turman, Matheus P. Viana, Lyndsay Wilhelm, Chamari S. Wijesooriya, Jie Yao, Julie A. Theriot, Susanne M. Rafelski, Ruwanthi N. Gunawardane

A simple inland culture system provides insights into ascidian post-embryonic developmental physiology

Birthe Thuesen Mathiesen, Mayu Ohta, Boris Pinto De Magalhaes, Chiara Castelletti, Vincenzo Perria, Lionel Christiaen, Naoyuki Ohta

Three-dimension transcriptomics maps of whole mouse embryo during organogenesis

Mengnan Cheng, Huiwen Zheng, Qi Fang, Yinqi Bai, Chao Liu, Hailin Pan, Zhewei Zhang, Qin Lu, Chang Shi, Tianyi Xia, Zehua Jing, Huanlin Liu, Ning Feng, Guojun Fu, Yumei Li, Jing Feng, Zepeng Li, Jingjing Wang, Yuanyuan Chen, Lianying Wang, Zhonghan Deng, Mei Li, Longqi Liu, Ao Chen, Xun Xu

A Human Biomimetic Intestinal Mucosa Model to Study Gastrointestinal Development and Disease

Alessandro Dei, Carlemi Calitz, Joep Korsten, Nina Johannesson, Eline Freeze, Allen Eaves, John Stingl, Ryan K Condor, Wing Chang, Dasja Pajkrt, Katja C. Wolthers, Adithya Sridhar, Salvatore Simmini

Moult cycle and setal development of the Atlantic ditch shrimp Palaemon varians Leach, 1814

Kenneth Kim, Jonathan Antcliffe, Allison C. Daley, Marc Robinson-Rechavi

Whole-embryo Spatial Transcriptomics at Subcellular Resolution from Gastrulation to Organogenesis

Yinan Wan, Jakob El Kholtei, Ignatius Jenie, Mariona Colomer-Rosell, Jialin Liu, Joaquin Navajas Acedo, Lucia Y. Du, Mireia Codina-Tobias, Mengfan Wang, Ahilya Sawh, Edward Lin, Tzy-Harn Chuang, Susan E. Mango, Guoqiang Yu, Bogdan Bintu, Alexander F. Schier

Dissecting the regulatory logic of specification and differentiation during vertebrate embryogenesis

Jialin Liu, Sebastian M. Castillo-Hair, Lucia Y. Du, Yiqun Wang, Adam N. Carte, Mariona Colomer-Rosell, Christopher Yin, Georg Seelig, Alexander F. Schier

A single-cell atlas of spatial and temporal gene expression in the mouse cranial neural plate

Eric R. Brooks, Andrew R. Moorman, Bhaswati Bhattacharya, Ian Prudhomme, Max Land, Heather L. Alcorn, Roshan Sharma, Dana Pe’er, Jennifer A. Zallen

From Brooks et al. This image is made available under a CC-BY 4.0 International license.

Thumbs up (No Ratings Yet)
Loading...

Tags: ,
Categories: Highlights, Research

How we learned to build a gliding mammal

Posted by , on 2 September 2024

A new hope to study convergent evolution

Convergent evolution — the independent emergence of analogous structures among species whose last common ancestors lacked the trait— has long fascinated me. This phenomenon represents an exciting opportunity to study the genomic constraints that shape organisms during development to produce specific forms and functions. In the fall of 2018, as I was searching for a PhD program, I heard a talk by Dr. Ricardo Mallarino. He was interested in studying one such example of convergence, the evolution of the patagium, a skin membrane that, like a wingsuit, allows animals to glide through the air as a means of locomotion. Of particular interest was the patagium of a small marsupial possum: sugar gliders. The patagium has evolved many times across the tree of life including rodents, primates, marsupials, lizards, and frogs. Marsupials however, offered a uniquely tractable group to study how similar novel morphological traits evolved independently, as three closely related possums had acquired this adaptation. Here I will cover the findings of our recent publication.

Adult sugar glider extending its patagium (red arrow).

Biology strikes back: identifying candidate regulatory elements

Previous RNAseq-based exploration of the developing patagium indicated that a major difference between the early patagium and neighboring skin was the differential regulation of an early gene regulatory network1. Given this, I set out to identify cis-regulatory elements (CREs) which had similar patterns of evolution in gliding species, which may harbor the causative changes driving differential gene regulation, evidenced by previous work2,3. We produced 15 new marsupial genomes which included gliding (sugar glider, greater glider, and feathertail glider) and non-gliding species. To identify cis-regulatory elements in the developing patagium tissue, we used ChIP- and ATAC-seq, which when used in combination provided a candidate list of active and poised CREs. Then using both our biological data and our genomes, we measured the rate of nucleotide change across these identified cis-regulatory elements, giving us an indication of which CREs are experiencing selection as these species have evolved their patagium. Through this analysis, we identified thousands of candidate glider accelerated regions (GARs)— elements which showed a substantial increase in nucleotide substitution. By focusing only on the GARs that showed shared patterns across the three gliding species we analyzed, we hoped to find a smaller pool of CREs that could be involved in the evolution of the patagium. We were surprised to find that not a single CRE matched that description. Then in our darkest times, a moment of brilliance: what if the cause was not a single CRE but instead different CREs across species all regulating the same gene. We had previously conducted Micro-C on the developing patagium and thought to use this data and identify topologically associating domains (TADs) to inform an analysis of GAR distribution and abundance. Using these TADs, we assigned CREs to the genes that were in the same TADs and asked if any gene had an overabundance of GARs.

Return of a key gene

Previous work from our group uncovered that the sugar glider patagium develops through the deployment of a conserved network of genes, and that Wnt5a is heavily involved in the early development of the gliding membrane1. One of the other genes identified was the transcription factor Emx2. It just so happened, that our analysis for GAR enrichment identified Emx2 as our strongest candidate, having GARs from each of the three gliding species in its vicinity. We produced an shRNA lentivirus for Emx2 and began testing the effects of downregulating Emx2 in the developing patagium. Taking advantage of marsupial biology, that is they give birth to their young, or joeys, quite early in their development, we could then probe the early patagium. We conducted injections into the patagium primordium of sugar glider joeys and found that indeed downregulation of Emx2 caused a decrease in the area of the developing patagium. This was one of my favorite experiments and served to remind me how fascinating working in science can be. For some time, I was one of the only people in the world who knew that lack of Emx2 led to incorrect patagium growth. We then explored how Emx2 may be regulated. We had several candidates to test but initially focused on just two, one was positioned in what we presumed to be the promoter of Emx2 while the second seemed to be a distal enhancer located 1mb away from the Emx2 promoter but with a strong contact loop in the Micro-C dataset. The former was accelerated in the sugar glider and the latter accelerated in the feathertail glider. We wanted to test if the acceleration observed had an effect on the element’s ability to regulate expression and so we decided to use luciferase assays in an immortalized sugar glider cell line. This experiment works by placing your CRE of interest upstream of the luciferase gene (which originates from fireflies) to measure the amount of fluorescence produced by the cells to see if your CRE has regulatory function. In our case, our results indicated that the distal enhancer had accumulated changes that made it a stronger enhancer in the feathertail glider compared to its non-gliding sister and the sugar glider. We later found that the other gliding species, the greater glider, also had an enhancer that showed the same pattern.

Emx2 shRNA injection results in a decrease in patagium size

Duo of fate: Our two favorite genes are important for patagium development

Now we knew that Emx2 had a phenotypic effect on the patagium and had some clues as to how it could end up being highly expressed in gliding species, but I became interested in what was happening at the molecular level in the patagium when we disrupted Emx2 expression. We did another round of shRNA injections, and this time collected the tissue for RNA sequencing. We found that many of the genes that were normally upregulated in the patagium when compared to surrounding skin were now downregulated. Among the ~400 genes downregulated, Wnt5a was one of the more strongly affected genes. This prompted us to investigate if Emx2 regulates all these genes directly and specifically how it may be regulating Wnt5a. We did an Emx2-ChIP-seq experiment and found that many genes did indeed have Emx2 binding sites, and we were able to identify multiple binding sites in the Wnt5a promoter. An example of great peer reviewing led to another of my favorite experiments as we set out to test the Emx2 binding sites found in the Wnt5a promoter. We again used luciferase this time testing two versions of the same promoter, one was unaltered while in the second we mutated the Emx2 binding sites. We found that the loss of these sites led to a complete loss of regulatory ability. Then to see if Emx2 was responsible for activating this promoter we co-transfected an Emx2-producing plasmid with our luciferase reporter plasmids. This experiment showed that the wild-type promoter increased its production of luciferase nearly 4-fold when Emx2 was present.

Emx2 awakens a conserved pathway

Our final goal for the paper was to establish if the spatial expression and function of Emx2 was novel to sugar gliders or if it was conserved in non-gliding mammals. We decided to test this hypothesis in mice, a much more amenable system for testing overexpression of genes. We found that Emx2, as reported previously4, was expressed in mice in a similar spatial pattern as that of the patagium, however this expression was only present for ~2 days whereas in the developing sugar gliders it was present for at least 14 days. To test whether overexpression of emx2 was sufficient to produce early patagium phenotypes, like we had previously observed with Wnt5, we extended the duration of Emx2 expression in mice while maintaining its endogenous spatial pattern. This however resulted in mice whose forebrain grew uncontrollably and resulted in non-viability; previous work had implicated Emx2 in brain development5. Therefore, we restricted the overexpression of Emx2 to only the skin. We found that indeed this overexpression was capable of recapitulating phenotypes observed in the early patagium such as increased cell proliferation, density, and the thickening of the epidermis1. These experiments showed that Emx2 has a conserved role in driving proliferation, potentially via regulation of the Wnt pathway, further indicating that evolution has re-used existing cellular programs to evolve a new adaptation.

The last remarks

In my opinion, the key message of this paper is that the evolution of convergent traits can occur independently via similar pathways/mechanisms, but the path to get there can be different. Our work showed that the redeployment of a shared developmental pathway can be an effective mechanism by which adaptations evolve.

I am very happy to see this work published; it took many years to get to this point. There were many bumps along the way, and it was the culmination of the hard work of many people involved. I want to thank the reviewers for their candid and helpful words, truly they made the paper better than when we first submitted. As someone who finds great joy in working on emerging model systems and has received countless advice in the past to just work on “model” organisms, I am incredibly pleased with how this paper is received. I continue to work on non-model systems with fascinating biology now as a postdoc and advise anyone who is interested in working on new models to go for it! It is rewarding to work on questions that can only be asked in a new system and uncover and share new findings.

References

1          Feigin, C. Y. et al. Convergent deployment of ancestral functions during the evolution of mammalian flight membranes. Science Advances 9, eade7511 (2023). https://doi.org/doi:10.1126/sciadv.ade7511

2          Booker, B. M. et al. Bat Accelerated Regions Identify a Bat Forelimb Specific Enhancer in the HoxD Locus. PLOS Genetics 12, e1005738 (2016). https://doi.org/10.1371/journal.pgen.1005738

3          Capra, J. A., Erwin, G. D., McKinsey, G., Rubenstein, J. L. R. & Pollard, K. S. Many human accelerated regions are developmental enhancers. Philosophical Transactions of the Royal Society B: Biological Sciences 368, 20130025 (2013). https://doi.org/doi:10.1098/rstb.2013.0025

4          Pellegrini, M., Pantano, S., Fumi, M. P., Lucchini, F. & Forabosco, A. Agenesis of the Scapula in Emx2 Homozygous Mutants. Developmental Biology 232, 149-156 (2001). https://doi.org/https://doi.org/10.1006/dbio.2001.0159

5          Yoshida, M. et al. Emx1 and Emx2 functions in development of dorsal telencephalon. Development 124, 101-111 (1997). https://doi.org/10.1242/dev.124.1.101

Thumbs up (2 votes)
Loading...

Tags: , , ,
Categories: Research

Help optimise the SAFE Labs Handbook by filling in our survey

Posted by , on 30 August 2024

We need your help to optimise the SAFE Labs Handbook before it is disseminated throughout the academic community. Please complete our this survey to help optimize this tool for the academic community.

This handbook is an outcome of the 2024 SAFE Labs workshop, where new bioscience group leaders from across Europe discussed Starting Aware, Fair, and Equitable Labs. The primary goal of this workshop (funded through the UCL Global Engagement award) was to better-understand the common, and divergent, problems faced by new researchers trying to build successful, equitable, fair, and environmentally sustainable labs with a positive research culture. There were no scientific talks as part of the program (there are plenty of meetings for that!).

Through this workshop, we realised that a major obstacle to improving lab culture was the lack of any resource with actionable “commitments” for group leaders that could be implemented in the absence of institutional support. Existing resources raised important issues, but lacked specific actions for group leaders to take.

The thirteen attendees unanimously agreed to collaborate on creating a SAFE Labs Handbook, covering commitments that span different topics: Policies, Teams, and Careers. Of course, there are many effective strategies to run a team, reflecting differences across groups, institutions, and countries. Thus, the Handbook is not prescriptive; rather it aims to increase transparency and minimise expectation mismatch between the group leader and lab members to create positive and equitable lab culture. Commitments may require a policy to be documented, but the content of that policy is decided by the group leader.

No commitment relies on institutional support to implement, and all commitments are actionable, and can be verifiably implemented. This is because:

  1. Implementable commitments are the most helpful to group leaders and lab members.
  2. Verifiable commitments allow for accountability and specific feedback. 

For example, “I commit to supporting a healthy work-life balance for my lab members” will not be in this handbook because the commitment cannot be verified.

The handbook does not represent an optimal or exhaustive template: it is the product of thirteen group leaders who are all passionate about improving lab culture, and are all fallible. Although this handbook will not solve every problem, we hope it takes a significant step toward addressing key challenges faced in daily lab life. The handbook will improve through community feedback and further dedicated workshops.

However, to ensure that this ensure that the SAFE Labs Handbook reflects the needs of our entire community, we need a diversity of feedback on the initial document: from all career levels and countries. The results will directly influence the content of the handbook and ensure that the commitments are something that are universally valued.

Please take the time to complete this survey, which can take less than 15 minutes (or 30 minutes if reading the commitments in more detail).

We hope the SAFE Labs Handbook becomes a valuable tool to community. Our intention is that labs implementing all commitments would feature the SAFE Labs logo on their website. This demonstrates to potential and existing lab members that the group leader is committed to creating a SAFE Lab.

We have secured funding to hold a 2025 workshop to further improve, expand, and refine this initiative. If you are interested in participating, please add your email address at the end of the survey.

Thank you for helping us to make the SAFE Labs Handbook as useful as possible!

The SAFE Labs organisers,

Pip Coen, Letizia Mariotti, Stéphane Bugeon, and Federico Rossi

2024 SAFE Labs Workshop Attendees
Thumbs up (1 votes)
Loading...

Tags: ,
Categories: Resources