TY - JOUR AB - In development, lineage segregation is coordinated in time and space. An important example is the mammalian inner cell mass, in which the primitive endoderm (PrE, founder of the yolk sac) physically segregates from the epiblast (EPI, founder of the fetus). While the molecular requirements have been well studied, the physical mechanisms determining spatial segregation between EPI and PrE remain elusive. Here, we investigate the mechanical basis of EPI and PrE sorting. We find that rather than the differences in static cell surface mechanical parameters as in classical sorting models, it is the differences in surface fluctuations that robustly ensure physical lineage sorting. These differential surface fluctuations systematically correlate with differential cellular fluidity, which we propose together constitute a non-equilibrium sorting mechanism for EPI and PrE lineages. By combining experiments and modeling, we identify cell surface dynamics as a key factor orchestrating the correct spatial segregation of the founder embryonic lineages. AU - Yanagida, Ayaka AU - Corujo-Simon, Elena AU - Revell, Christopher K. AU - Sahu, Preeti AU - Stirparo, Giuliano G. AU - Aspalter, Irene M. AU - Winkel, Alex K. AU - Peters, Ruby AU - De Belly, Henry AU - Cassani, Davide A.D. AU - Achouri, Sarra AU - Blumenfeld, Raphael AU - Franze, Kristian AU - Hannezo, Edouard B AU - Paluch, Ewa K. AU - Nichols, Jennifer AU - Chalut, Kevin J. ID - 10825 IS - 5 JF - Cell SN - 00928674 TI - Cell surface fluctuations regulate early embryonic lineage sorting VL - 185 ER - TY - JOUR AB - Embryo development requires biochemical signalling to generate patterns of cell fates and active mechanical forces to drive tissue shape changes. However, how these processes are coordinated, and how tissue patterning is preserved despite the cellular flows occurring during morphogenesis, remains poorly understood. Gastrulation is a crucial embryonic stage that involves both patterning and internalization of the mesendoderm germ layer tissue. Here we show that, in zebrafish embryos, a gradient in Nodal signalling orchestrates pattern-preserving internalization movements by triggering a motility-driven unjamming transition. In addition to its role as a morphogen determining embryo patterning, graded Nodal signalling mechanically subdivides the mesendoderm into a small fraction of highly protrusive leader cells, able to autonomously internalize via local unjamming, and less protrusive followers, which need to be pulled inwards by the leaders. The Nodal gradient further enforces a code of preferential adhesion coupling leaders to their immediate followers, resulting in a collective and ordered mode of internalization that preserves mesendoderm patterning. Integrating this dual mechanical role of Nodal signalling into minimal active particle simulations quantitatively predicts both physiological and experimentally perturbed internalization movements. This provides a quantitative framework for how a morphogen-encoded unjamming transition can bidirectionally couple tissue mechanics with patterning during complex three-dimensional morphogenesis. AU - Nunes Pinheiro, Diana C AU - Kardos, Roland AU - Hannezo, Edouard B AU - Heisenberg, Carl-Philipp J ID - 12209 IS - 12 JF - Nature Physics KW - General Physics and Astronomy SN - 1745-2473 TI - Morphogen gradient orchestrates pattern-preserving tissue morphogenesis via motility-driven unjamming VL - 18 ER - TY - JOUR AB - The development dynamics and self-organization of glandular branched epithelia is of utmost importance for our understanding of diverse processes ranging from normal tissue growth to the growth of cancerous tissues. Using single primary murine pancreatic ductal adenocarcinoma (PDAC) cells embedded in a collagen matrix and adapted media supplementation, we generate organoids that self-organize into highly branched structures displaying a seamless lumen connecting terminal end buds, replicating in vivo PDAC architecture. We identify distinct morphogenesis phases, each characterized by a unique pattern of cell invasion, matrix deformation, protein expression, and respective molecular dependencies. We propose a minimal theoretical model of a branching and proliferating tissue, capturing the dynamics of the first phases. Observing the interaction of morphogenesis, mechanical environment and gene expression in vitro sets a benchmark for the understanding of self-organization processes governing complex organoid structure formation processes and branching morphogenesis. AU - Randriamanantsoa, S. AU - Papargyriou, A. AU - Maurer, H. C. AU - Peschke, K. AU - Schuster, M. AU - Zecchin, G. AU - Steiger, K. AU - Öllinger, R. AU - Saur, D. AU - Scheel, C. AU - Rad, R. AU - Hannezo, Edouard B AU - Reichert, M. AU - Bausch, A. R. ID - 12217 JF - Nature Communications KW - General Physics and Astronomy KW - General Biochemistry KW - Genetics and Molecular Biology KW - General Chemistry KW - Multidisciplinary SN - 2041-1723 TI - Spatiotemporal dynamics of self-organized branching in pancreas-derived organoids VL - 13 ER - TY - JOUR AB - The sculpting of germ layers during gastrulation relies on the coordinated migration of progenitor cells, yet the cues controlling these long-range directed movements remain largely unknown. While directional migration often relies on a chemokine gradient generated from a localized source, we find that zebrafish ventrolateral mesoderm is guided by a self-generated gradient of the initially uniformly expressed and secreted protein Toddler/ELABELA/Apela. We show that the Apelin receptor, which is specifically expressed in mesodermal cells, has a dual role during gastrulation, acting as a scavenger receptor to generate a Toddler gradient, and as a chemokine receptor to sense this guidance cue. Thus, we uncover a single receptor–based self-generated gradient as the enigmatic guidance cue that can robustly steer the directional migration of mesoderm through the complex and continuously changing environment of the gastrulating embryo. AU - Stock, Jessica AU - Kazmar, Tomas AU - Schlumm, Friederike AU - Hannezo, Edouard B AU - Pauli, Andrea ID - 12253 IS - 37 JF - Science Advances SN - 2375-2548 TI - A self-generated Toddler gradient guides mesodermal cell migration VL - 8 ER - TY - JOUR AB - Cell migration in confining physiological environments relies on the concerted dynamics of several cellular components, including protrusions, adhesions with the environment, and the cell nucleus. However, it remains poorly understood how the dynamic interplay of these components and the cell polarity determine the emergent migration behavior at the cellular scale. Here, we combine data-driven inference with a mechanistic bottom-up approach to develop a model for protrusion and polarity dynamics in confined cell migration, revealing how the cellular dynamics adapt to confining geometries. Specifically, we use experimental data of joint protrusion-nucleus migration trajectories of cells on confining micropatterns to systematically determine a mechanistic model linking the stochastic dynamics of cell polarity, protrusions, and nucleus. This model indicates that the cellular dynamics adapt to confining constrictions through a switch in the polarity dynamics from a negative to a positive self-reinforcing feedback loop. Our model further reveals how this feedback loop leads to stereotypical cycles of protrusion-nucleus dynamics that drive the migration of the cell through constrictions. These cycles are disrupted upon perturbation of cytoskeletal components, indicating that the positive feedback is controlled by cellular migration mechanisms. Our data-driven theoretical approach therefore identifies polarity feedback adaptation as a key mechanism in confined cell migration. AU - Brückner, David AU - Schmitt, Matthew AU - Fink, Alexandra AU - Ladurner, Georg AU - Flommersfeld, Johannes AU - Arlt, Nicolas AU - Hannezo, Edouard B AU - Rädler, Joachim O. AU - Broedersz, Chase P. ID - 12277 IS - 3 JF - Physical Review X KW - General Physics and Astronomy SN - 2160-3308 TI - Geometry adaptation of protrusion and polarity dynamics in confined cell migration VL - 12 ER -