Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
Cell ; 185(5): 777-793.e20, 2022 03 03.
Article in English | MEDLINE | ID: mdl-35196500

ABSTRACT

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.


Subject(s)
Blastocyst , Embryo, Mammalian , Endoderm , Animals , Blastocyst/metabolism , Cell Differentiation/physiology , Cell Lineage/physiology , Cell Membrane/metabolism , Embryo, Mammalian/metabolism , Embryonic Development , Endoderm/metabolism , Mammals , Mice , Protein Transport
2.
Cell ; 184(7): 1914-1928.e19, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33730596

ABSTRACT

Embryo morphogenesis is impacted by dynamic changes in tissue material properties, which have been proposed to occur via processes akin to phase transitions (PTs). Here, we show that rigidity percolation provides a simple and robust theoretical framework to predict material/structural PTs of embryonic tissues from local cell connectivity. By using percolation theory, combined with directly monitoring dynamic changes in tissue rheology and cell contact mechanics, we demonstrate that the zebrafish blastoderm undergoes a genuine rigidity PT, brought about by a small reduction in adhesion-dependent cell connectivity below a critical value. We quantitatively predict and experimentally verify hallmarks of PTs, including power-law exponents and associated discontinuities of macroscopic observables. Finally, we show that this uniform PT depends on blastoderm cells undergoing meta-synchronous divisions causing random and, consequently, uniform changes in cell connectivity. Collectively, our theoretical and experimental findings reveal the structural basis of material PTs in an organismal context.


Subject(s)
Embryo, Nonmammalian/physiology , Embryonic Development , Animals , Blastoderm/cytology , Blastoderm/physiology , Cadherins/antagonists & inhibitors , Cadherins/genetics , Cadherins/metabolism , Cell Adhesion , Embryo, Nonmammalian/cytology , Morpholinos/metabolism , Rheology , Viscosity , Zebrafish/growth & development
3.
Cell ; 184(26): 6313-6325.e18, 2021 12 22.
Article in English | MEDLINE | ID: mdl-34942099

ABSTRACT

How tissues acquire complex shapes is a fundamental question in biology and regenerative medicine. Zebrafish semicircular canals form from invaginations in the otic epithelium (buds) that extend and fuse to form the hubs of each canal. We find that conventional actomyosin-driven behaviors are not required. Instead, local secretion of hyaluronan, made by the enzymes uridine 5'-diphosphate dehydrogenase (ugdh) and hyaluronan synthase 3 (has3), drives canal morphogenesis. Charged hyaluronate polymers osmotically swell with water and generate isotropic extracellular pressure to deform the overlying epithelium into buds. The mechanical anisotropy needed to shape buds into tubes is conferred by a polarized distribution of actomyosin and E-cadherin-rich membrane tethers, which we term cytocinches. Most work on tissue morphogenesis ascribes actomyosin contractility as the driving force, while the extracellular matrix shapes tissues through differential stiffness. Our work inverts this expectation. Hyaluronate pressure shaped by anisotropic tissue stiffness may be a widespread mechanism for powering morphological change in organogenesis and tissue engineering.


Subject(s)
Extracellular Space/chemistry , Hyaluronic Acid/pharmacology , Morphogenesis , Organ Specificity , Pressure , Semicircular Canals/cytology , Semicircular Canals/embryology , Actomyosin/metabolism , Animals , Anisotropy , Behavior, Animal , Extracellular Matrix/metabolism , Hyaluronic Acid/biosynthesis , Models, Biological , Morphogenesis/drug effects , Organ Specificity/drug effects , Osmotic Pressure , Semicircular Canals/diagnostic imaging , Stereotyped Behavior , Zebrafish/embryology , Zebrafish Proteins/metabolism
4.
Cell ; 181(3): 604-620.e22, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32259486

ABSTRACT

During embryonic and postnatal development, organs and tissues grow steadily to achieve their final size at the end of puberty. However, little is known about the cellular dynamics that mediate postnatal growth. By combining in vivo clonal lineage tracing, proliferation kinetics, single-cell transcriptomics, and in vitro micro-pattern experiments, we resolved the cellular dynamics taking place during postnatal skin epidermis expansion. Our data revealed that harmonious growth is engineered by a single population of developmental progenitors presenting a fixed fate imbalance of self-renewing divisions with an ever-decreasing proliferation rate. Single-cell RNA sequencing revealed that epidermal developmental progenitors form a more uniform population compared with adult stem and progenitor cells. Finally, we found that the spatial pattern of cell division orientation is dictated locally by the underlying collagen fiber orientation. Our results uncover a simple design principle of organ growth where progenitors and differentiated cells expand in harmony with their surrounding tissues.


Subject(s)
Epidermal Cells/metabolism , Epidermis/growth & development , Skin/growth & development , Animals , Animals, Outbred Strains , Cell Differentiation/physiology , Cell Division/physiology , Cell Lineage/genetics , Cell Proliferation/physiology , Cells, Cultured , Epidermal Cells/pathology , Epidermis/metabolism , Female , Male , Mice , Mice, Transgenic , Stem Cells/cytology
5.
Nat Immunol ; 23(8): 1246-1255, 2022 08.
Article in English | MEDLINE | ID: mdl-35817845

ABSTRACT

Lymph nodes (LNs) comprise two main structural elements: fibroblastic reticular cells that form dedicated niches for immune cell interaction and capsular fibroblasts that build a shell around the organ. Immunological challenge causes LNs to increase more than tenfold in size within a few days. Here, we characterized the biomechanics of LN swelling on the cellular and organ scale. We identified lymphocyte trapping by influx and proliferation as drivers of an outward pressure force, causing fibroblastic reticular cells of the T-zone (TRCs) and their associated conduits to stretch. After an initial phase of relaxation, TRCs sensed the resulting strain through cell matrix adhesions, which coordinated local growth and remodeling of the stromal network. While the expanded TRC network readopted its typical configuration, a massive fibrotic reaction of the organ capsule set in and countered further organ expansion. Thus, different fibroblast populations mechanically control LN swelling in a multitier fashion.


Subject(s)
Lymph Nodes , Stromal Cells , Animals , Fibroblasts , Lymphocytes , Mice , Mice, Inbred C57BL
6.
Cell ; 178(1): 12-25, 2019 06 27.
Article in English | MEDLINE | ID: mdl-31251912

ABSTRACT

There is increasing evidence that both mechanical and biochemical signals play important roles in development and disease. The development of complex organisms, in particular, has been proposed to rely on the feedback between mechanical and biochemical patterning events. This feedback occurs at the molecular level via mechanosensation but can also arise as an emergent property of the system at the cellular and tissue level. In recent years, dynamic changes in tissue geometry, flow, rheology, and cell fate specification have emerged as key platforms of mechanochemical feedback loops in multiple processes. Here, we review recent experimental and theoretical advances in understanding how these feedbacks function in development and disease.


Subject(s)
Biomechanical Phenomena/physiology , Cell Communication/physiology , Cell Differentiation/physiology , Cell Proliferation/physiology , Feedback, Physiological , Animals , Cell Size , Cytoskeleton/physiology , Extracellular Matrix/physiology , Humans , Protein Conformation , Rheology
7.
Cell ; 177(6): 1463-1479.e18, 2019 05 30.
Article in English | MEDLINE | ID: mdl-31080065

ABSTRACT

Segregation of maternal determinants within the oocyte constitutes the first step in embryo patterning. In zebrafish oocytes, extensive ooplasmic streaming leads to the segregation of ooplasm from yolk granules along the animal-vegetal axis of the oocyte. Here, we show that this process does not rely on cortical actin reorganization, as previously thought, but instead on a cell-cycle-dependent bulk actin polymerization wave traveling from the animal to the vegetal pole of the oocyte. This wave functions in segregation by both pulling ooplasm animally and pushing yolk granules vegetally. Using biophysical experimentation and theory, we show that ooplasm pulling is mediated by bulk actin network flows exerting friction forces on the ooplasm, while yolk granule pushing is achieved by a mechanism closely resembling actin comet formation on yolk granules. Our study defines a novel role of cell-cycle-controlled bulk actin polymerization waves in oocyte polarization via ooplasmic segregation.


Subject(s)
Actins/metabolism , Cell Cycle/physiology , Oocytes/metabolism , Actins/physiology , Animals , Cell Polarity/physiology , Cytoplasm/metabolism , Egg Yolk/physiology , Polymerization , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins/metabolism , Zygote
8.
Cell ; 171(1): 242-255.e27, 2017 Sep 21.
Article in English | MEDLINE | ID: mdl-28938116

ABSTRACT

The morphogenesis of branched organs remains a subject of abiding interest. Although much is known about the underlying signaling pathways, it remains unclear how macroscopic features of branched organs, including their size, network topology, and spatial patterning, are encoded. Here, we show that, in mouse mammary gland, kidney, and human prostate, these features can be explained quantitatively within a single unifying framework of branching and annihilating random walks. Based on quantitative analyses of large-scale organ reconstructions and proliferation kinetics measurements, we propose that morphogenesis follows from the proliferative activity of equipotent tips that stochastically branch and randomly explore their environment but compete neutrally for space, becoming proliferatively inactive when in proximity with neighboring ducts. These results show that complex branched epithelial structures develop as a self-organized process, reliant upon a strikingly simple but generic rule, without recourse to a rigid and deterministic sequence of genetically programmed events.


Subject(s)
Kidney/growth & development , Mammary Glands, Human/growth & development , Models, Biological , Morphogenesis , Prostate/growth & development , Animals , Female , Humans , Kidney/embryology , Male , Mammary Glands, Human/embryology , Mice , Prostate/embryology
10.
Nature ; 607(7919): 548-554, 2022 07.
Article in English | MEDLINE | ID: mdl-35831497

ABSTRACT

The morphology and functionality of the epithelial lining differ along the intestinal tract, but tissue renewal at all sites is driven by stem cells at the base of crypts1-3. Whether stem cell numbers and behaviour vary at different sites is unknown. Here we show using intravital microscopy that, despite similarities in the number and distribution of proliferative cells with an Lgr5 signature in mice, small intestinal crypts contain twice as many effective stem cells as large intestinal crypts. We find that, although passively displaced by a conveyor-belt-like upward movement, small intestinal cells positioned away from the crypt base can function as long-term effective stem cells owing to Wnt-dependent retrograde cellular movement. By contrast, the near absence of retrograde movement in the large intestine restricts cell repositioning, leading to a reduction in effective stem cell number. Moreover, after suppression of the retrograde movement in the small intestine, the number of effective stem cells is reduced, and the rate of monoclonal conversion of crypts is accelerated. Together, these results show that the number of effective stem cells is determined by active retrograde movement, revealing a new channel of stem cell regulation that can be experimentally and pharmacologically manipulated.


Subject(s)
Cell Count , Cell Movement , Intestines , Stem Cells , Animals , Intestinal Mucosa/cytology , Intestine, Small/cytology , Intestines/cytology , Mice , Receptors, G-Protein-Coupled , Stem Cells/cytology , Wnt Proteins
11.
PLoS Biol ; 21(10): e3002315, 2023 10.
Article in English | MEDLINE | ID: mdl-37792696

ABSTRACT

To meet the physiological demands of the body, organs need to establish a functional tissue architecture and adequate size as the embryo develops to adulthood. In the liver, uni- and bipotent progenitor differentiation into hepatocytes and biliary epithelial cells (BECs), and their relative proportions, comprise the functional architecture. Yet, the contribution of individual liver progenitors at the organ level to both fates, and their specific proportion, is unresolved. Combining mathematical modelling with organ-wide, multispectral FRaeppli-NLS lineage tracing in zebrafish, we demonstrate that a precise BEC-to-hepatocyte ratio is established (i) fast, (ii) solely by heterogeneous lineage decisions from uni- and bipotent progenitors, and (iii) independent of subsequent cell type-specific proliferation. Extending lineage tracing to adulthood determined that embryonic cells undergo spatially heterogeneous three-dimensional growth associated with distinct environments. Strikingly, giant clusters comprising almost half a ventral lobe suggest lobe-specific dominant-like growth behaviours. We show substantial hepatocyte polyploidy in juveniles representing another hallmark of postembryonic liver growth. Our findings uncover heterogeneous progenitor contributions to tissue architecture-defining cell type proportions and postembryonic organ growth as key mechanisms forming the adult liver.


Subject(s)
Liver , Zebrafish , Animals , Cell Lineage , Liver/metabolism , Hepatocytes/metabolism , Epithelial Cells , Cell Differentiation , Cell Proliferation
12.
Semin Cell Dev Biol ; 150-151: 58-65, 2023 12.
Article in English | MEDLINE | ID: mdl-36470715

ABSTRACT

Homeostatic balance in the intestinal epithelium relies on a fast cellular turnover, which is coordinated by an intricate interplay between biochemical signalling, mechanical forces and organ geometry. We review recent modelling approaches that have been developed to understand different facets of this remarkable homeostatic equilibrium. Existing models offer different, albeit complementary, perspectives on the problem. First, biomechanical models aim to explain the local and global mechanical stresses driving cell renewal as well as tissue shape maintenance. Second, compartmental models provide insights into the conditions necessary to keep a constant flow of cells with well-defined ratios of cell types, and how perturbations can lead to an unbalance of relative compartment sizes. A third family of models address, at the cellular level, the nature and regulation of stem fate choices that are necessary to fuel cellular turnover. We also review how these different approaches are starting to be integrated together across scales, to provide quantitative predictions and new conceptual frameworks to think about the dynamics of cell renewal in complex tissues.


Subject(s)
Signal Transduction , Stem Cells , Animals , Stem Cells/metabolism , Intestinal Mucosa , Homeostasis , Mammals
13.
Nature ; 570(7759): 107-111, 2019 06.
Article in English | MEDLINE | ID: mdl-31092921

ABSTRACT

Adult intestinal stem cells are located at the bottom of crypts of Lieberkühn, where they express markers such as LGR51,2 and fuel the constant replenishment of the intestinal epithelium1. Although fetal LGR5-expressing cells can give rise to adult intestinal stem cells3,4, it remains unclear whether this population in the patterned epithelium represents unique intestinal stem-cell precursors. Here we show, using unbiased quantitative lineage-tracing approaches, biophysical modelling and intestinal transplantation, that all cells of the mouse intestinal epithelium-irrespective of their location and pattern of LGR5 expression in the fetal gut tube-contribute actively to the adult intestinal stem cell pool. Using 3D imaging, we find that during fetal development the villus undergoes gross remodelling and fission. This brings epithelial cells from the non-proliferative villus into the proliferative intervillus region, which enables them to contribute to the adult stem-cell niche. Our results demonstrate that large-scale remodelling of the intestinal wall and cell-fate specification are closely linked. Moreover, these findings provide a direct link between the observed plasticity and cellular reprogramming of differentiating cells in adult tissues following damage5-9, revealing that stem-cell identity is an induced rather than a hardwired property.


Subject(s)
Cell Lineage , Intestines/cytology , Stem Cells/cytology , Animals , Cell Differentiation , Cellular Reprogramming , Female , Fetus/cytology , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Intestines/growth & development , Male , Mice , Receptors, G-Protein-Coupled/metabolism , Regeneration , Stem Cell Niche
14.
J Cell Sci ; 134(14)2021 07 15.
Article in English | MEDLINE | ID: mdl-34323278

ABSTRACT

Proper control of division orientation and symmetry, largely determined by spindle positioning, is essential to development and homeostasis. Spindle positioning has been extensively studied in cells dividing in two-dimensional (2D) environments and in epithelial tissues, where proteins such as NuMA (also known as NUMA1) orient division along the interphase long axis of the cell. However, little is known about how cells control spindle positioning in three-dimensional (3D) environments, such as early mammalian embryos and a variety of adult tissues. Here, we use mouse embryonic stem cells (ESCs), which grow in 3D colonies, as a model to investigate division in 3D. We observe that, at the periphery of 3D colonies, ESCs display high spindle mobility and divide asymmetrically. Our data suggest that enhanced spindle movements are due to unequal distribution of the cell-cell junction protein E-cadherin between future daughter cells. Interestingly, when cells progress towards differentiation, division becomes more symmetric, with more elongated shapes in metaphase and enhanced cortical NuMA recruitment in anaphase. Altogether, this study suggests that in 3D contexts, the geometry of the cell and its contacts with neighbors control division orientation and symmetry. This article has an associated First Person interview with the first author of the paper.


Subject(s)
Anaphase , Spindle Apparatus , Animals , Intercellular Junctions , Metaphase , Mice , Mitosis , Stem Cells
15.
Nature ; 545(7652): 103-107, 2017 05 04.
Article in English | MEDLINE | ID: mdl-28296858

ABSTRACT

During epithelial cytokinesis, the remodelling of adhesive cell-cell contacts between the dividing cell and its neighbours has profound implications for the integrity, arrangement and morphogenesis of proliferative tissues. In both vertebrates and invertebrates, this remodelling requires the activity of non-muscle myosin II (MyoII) in the interphasic cells neighbouring the dividing cell. However, the mechanisms that coordinate cytokinesis and MyoII activity in the neighbours are unknown. Here we show that in the Drosophila notum epithelium, each cell division is associated with a mechanosensing and transmission event that controls MyoII dynamics in neighbouring cells. We find that the ring pulling forces promote local junction elongation, which results in local E-cadherin dilution at the ingressing adherens junction. In turn, the reduction in E-cadherin concentration and the contractility of the neighbouring cells promote self-organized actomyosin flows, ultimately leading to accumulation of MyoII at the base of the ingressing junction. Although force transduction has been extensively studied in the context of adherens junction reinforcement to stabilize adhesive cell-cell contacts, we propose an alternative mechanosensing mechanism that coordinates actomyosin dynamics between epithelial cells and sustains the remodelling of the adherens junction in response to mechanical forces.


Subject(s)
Actomyosin/metabolism , Adherens Junctions/metabolism , Cadherins/metabolism , Cytokinesis , Drosophila melanogaster/cytology , Myosin Type II/metabolism , Animals , Cell Adhesion , Cell Division , Epithelial Cells/cytology , Epithelial Cells/metabolism
16.
Nature ; 542(7641): 313-317, 2017 02 16.
Article in English | MEDLINE | ID: mdl-28135720

ABSTRACT

During puberty, the mouse mammary gland develops into a highly branched epithelial network. Owing to the absence of exclusive stem cell markers, the location, multiplicity, dynamics and fate of mammary stem cells (MaSCs), which drive branching morphogenesis, are unknown. Here we show that morphogenesis is driven by proliferative terminal end buds that terminate or bifurcate with near equal probability, in a stochastic and time-invariant manner, leading to a heterogeneous epithelial network. We show that the majority of terminal end bud cells function as highly proliferative, lineage-committed MaSCs that are heterogeneous in their expression profile and short-term contribution to ductal extension. Yet, through cell rearrangements during terminal end bud bifurcation, each MaSC is able to contribute actively to long-term growth. Our study shows that the behaviour of MaSCs is not directly linked to a single expression profile. Instead, morphogenesis relies upon lineage-restricted heterogeneous MaSC populations that function as single equipotent pools in the long term.


Subject(s)
Cell Lineage , Mammary Glands, Animal/cytology , Morphogenesis , Stem Cells/cytology , Stem Cells/metabolism , Animals , Cell Proliferation , Female , Gene Expression Profiling , Mice , Models, Molecular , Sexual Maturation , Single-Cell Analysis , Stochastic Processes
17.
Proc Natl Acad Sci U S A ; 117(29): 16969-16975, 2020 07 21.
Article in English | MEDLINE | ID: mdl-32611816

ABSTRACT

Understanding to what extent stem cell potential is a cell-intrinsic property or an emergent behavior coming from global tissue dynamics and geometry is a key outstanding question of systems and stem cell biology. Here, we propose a theory of stem cell dynamics as a stochastic competition for access to a spatially localized niche, giving rise to a stochastic conveyor-belt model. Cell divisions produce a steady cellular stream which advects cells away from the niche, while random rearrangements enable cells away from the niche to be favorably repositioned. Importantly, even when assuming that all cells in a tissue are molecularly equivalent, we predict a common ("universal") functional dependence of the long-term clonal survival probability on distance from the niche, as well as the emergence of a well-defined number of functional stem cells, dependent only on the rate of random movements vs. mitosis-driven advection. We test the predictions of this theory on datasets of pubertal mammary gland tips and embryonic kidney tips, as well as homeostatic intestinal crypts. Importantly, we find good agreement for the predicted functional dependency of the competition as a function of position, and thus functional stem cell number in each organ. This argues for a key role of positional fluctuations in dictating stem cell number and dynamics, and we discuss the applicability of this theory to other settings.


Subject(s)
Cell Lineage , Cell Self Renewal , Stem Cell Niche , Animals , Cell Survival , Female , Homeostasis , Intestines/cytology , Intestines/growth & development , Kidney/cytology , Kidney/growth & development , Mammary Glands, Animal/cytology , Mammary Glands, Animal/growth & development , Mice , Models, Theoretical , Signal-To-Noise Ratio , Stem Cells/cytology , Stem Cells/physiology
18.
Nature ; 536(7616): 298-303, 2016 08 18.
Article in English | MEDLINE | ID: mdl-27459053

ABSTRACT

The changes in cell dynamics after oncogenic mutation that lead to the development of tumours are currently unknown. Here, using skin epidermis as a model, we assessed the effect of oncogenic hedgehog signalling in distinct cell populations and their capacity to induce basal cell carcinoma, the most frequent cancer in humans. We found that only stem cells, and not progenitors, initiated tumour formation upon oncogenic hedgehog signalling. This difference was due to the hierarchical organization of tumour growth in oncogene-targeted stem cells, characterized by an increase in symmetric self-renewing divisions and a higher p53-dependent resistance to apoptosis, leading to rapid clonal expansion and progression into invasive tumours. Our work reveals that the capacity of oncogene-targeted cells to induce tumour formation is dependent not only on their long-term survival and expansion, but also on the specific clonal dynamics of the cancer cell of origin.


Subject(s)
Carcinoma, Basal Cell/pathology , Clone Cells/pathology , Neoplastic Stem Cells/pathology , Skin Neoplasms/pathology , Animals , Apoptosis , Carcinoma, Basal Cell/genetics , Cell Self Renewal , Cell Survival , Disease Progression , Epidermis/pathology , Female , Hedgehog Proteins/metabolism , Homeostasis , Male , Mice , Mutation/genetics , Oncogenes/genetics , Signal Transduction , Skin Neoplasms/genetics , Tail/pathology , Tumor Suppressor Protein p53/metabolism
19.
Proc Natl Acad Sci U S A ; 116(12): 5344-5349, 2019 03 19.
Article in English | MEDLINE | ID: mdl-30819884

ABSTRACT

The formation of self-organized patterns is key to the morphogenesis of multicellular organisms, although a comprehensive theory of biological pattern formation is still lacking. Here, we propose a minimal model combining tissue mechanics with morphogen turnover and transport to explore routes to patterning. Our active description couples morphogen reaction and diffusion, which impact cell differentiation and tissue mechanics, to a two-phase poroelastic rheology, where one tissue phase consists of a poroelastic cell network and the other one of a permeating extracellular fluid, which provides a feedback by actively transporting morphogens. While this model encompasses previous theories approximating tissues to inert monophasic media, such as Turing's reaction-diffusion model, it overcomes some of their key limitations permitting pattern formation via any two-species biochemical kinetics due to mechanically induced cross-diffusion flows. Moreover, we describe a qualitatively different advection-driven Keller-Segel instability which allows for the formation of patterns with a single morphogen and whose fundamental mode pattern robustly scales with tissue size. We discuss the potential relevance of these findings for tissue morphogenesis.


Subject(s)
Body Patterning/physiology , Morphogenesis/physiology , Protein Transport/physiology , Animals , Cell Differentiation/physiology , Diffusion , Kinetics , Models, Biological
20.
Phys Biol ; 18(4)2021 04 14.
Article in English | MEDLINE | ID: mdl-33276350

ABSTRACT

The way in which interactions between mechanics and biochemistry lead to the emergence of complex cell and tissue organization is an old question that has recently attracted renewed interest from biologists, physicists, mathematicians and computer scientists. Rapid advances in optical physics, microscopy and computational image analysis have greatly enhanced our ability to observe and quantify spatiotemporal patterns of signalling, force generation, deformation, and flow in living cells and tissues. Powerful new tools for genetic, biophysical and optogenetic manipulation are allowing us to perturb the underlying machinery that generates these patterns in increasingly sophisticated ways. Rapid advances in theory and computing have made it possible to construct predictive models that describe how cell and tissue organization and dynamics emerge from the local coupling of biochemistry and mechanics. Together, these advances have opened up a wealth of new opportunities to explore how mechanochemical patterning shapes organismal development. In this roadmap, we present a series of forward-looking case studies on mechanochemical patterning in development, written by scientists working at the interface between the physical and biological sciences, and covering a wide range of spatial and temporal scales, organisms, and modes of development. Together, these contributions highlight the many ways in which the dynamic coupling of mechanics and biochemistry shapes biological dynamics: from mechanoenzymes that sense force to tune their activity and motor output, to collectives of cells in tissues that flow and redistribute biochemical signals during development.


Subject(s)
Biomechanical Phenomena , Morphogenesis , Signal Transduction , Models, Biological
SELECTION OF CITATIONS
SEARCH DETAIL