Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Cell Sci ; 136(1)2023 01 01.
Article in English | MEDLINE | ID: mdl-36602106

ABSTRACT

Branched epithelial networks are generated through an iterative process of elongation and bifurcation. We sought to understand bifurcation of the mammary epithelium. To visualize this process, we utilized three-dimensional (3D) organotypic culture and time-lapse confocal microscopy. We tracked cell migration during bifurcation and observed local reductions in cell speed at the nascent bifurcation cleft. This effect was proximity dependent, as individual cells approaching the cleft reduced speed, whereas cells exiting the cleft increased speed. As the cells slow down, they orient both migration and protrusions towards the nascent cleft, while cells in the adjacent branches orient towards the elongating tips. We next tested the hypothesis that TGF-ß signaling controls mammary branching by regulating cell migration. We first validated that addition of TGF-ß1 (TGFB1) protein increased cleft number, whereas inhibition of TGF-ß signaling reduced cleft number. Then, consistent with our hypothesis, we observed that pharmacological inhibition of TGF-ß1 signaling acutely decreased epithelial migration speed. Our data suggest a model for mammary epithelial bifurcation in which TGF-ß signaling regulates cell migration to determine the local sites of bifurcation and the global pattern of the tubular network.


Subject(s)
Mammary Glands, Animal , Transforming Growth Factor beta1 , Animals , Transforming Growth Factor beta1/pharmacology , Transforming Growth Factor beta1/metabolism , Morphogenesis , Epithelium/metabolism , Cell Movement , Epithelial Cells/metabolism
2.
Mol Biol Cell ; 33(14): br26, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36222834

ABSTRACT

Animals are shaped through the movement of large cellular collectives. Such morphogenetic processes require cadherin-based cell adhesion to maintain tissue cohesion and planar cell polarity to coordinate movement. Despite a vast literature surrounding cadherin-based adhesion and planar cell polarity, it is unclear how these molecular networks interface. Here we investigate the relationship between cadherins and planar cell polarity during gastrulation cell movements in Xenopus laevis. We first assessed bulk cadherin localization and found that cadherins were enriched at a specific subset of morphogenetically active cell-cell junctions. We then found that cadherin and actin had coupled temporal dynamics and that disruption of planar cell polarity uncoupled these dynamics. Next, using superresolution time-lapse microscopy and quantitative image analysis, we were able to measure the lifespan and size of individual cadherin clusters. Finally, we show that planar cell polarity not only controls the size of cadherin clusters but, more interestingly, regulates cluster stability. These results reveal an intriguing link between two essential cellular properties, adhesion and planar polarity, and provide insight into the molecular control of morphogenetic cell movements.


Subject(s)
Cadherins , Gastrulation , Animals , Cadherins/metabolism , Morphogenesis , Gastrulation/physiology , Xenopus laevis/metabolism , Cell Adhesion/physiology
3.
Dev Cell ; 57(9): 1119-1131.e5, 2022 05 09.
Article in English | MEDLINE | ID: mdl-35476939

ABSTRACT

The design of an animal's body plan is encoded in the genome, and the execution of this program is a mechanical progression involving coordinated movement of proteins, cells, and whole tissues. Thus, a challenge to understanding morphogenesis is connecting events that occur across various length scales. Here, we describe how a poorly characterized adhesion effector, Arvcf catenin, controls Xenopus head-to-tail axis extension. We find that Arvcf is required for axis extension within the intact organism but not within isolated tissues. We show that the organism-scale phenotype results from a defect in tissue-scale force production. Finally, we determine that the force defect results from the dampening of the pulsatile recruitment of cell adhesion and cytoskeletal proteins to membranes. These results provide a comprehensive understanding of Arvcf function during axis extension and produce an insight into how a cellular-scale defect in adhesion results in an organism-scale failure of development.


Subject(s)
Armadillo Domain Proteins , Catenins , Animals , Armadillo Domain Proteins/genetics , Armadillo Domain Proteins/metabolism , Cadherins/metabolism , Cell Adhesion Molecules/metabolism , Morphogenesis , Phosphoproteins/metabolism , Xenopus laevis/metabolism
4.
Cell Rep ; 39(4): 110666, 2022 04 26.
Article in English | MEDLINE | ID: mdl-35476988

ABSTRACT

Convergent extension (CE) is an evolutionarily conserved collective cell movement that elongates several organ systems during development. Studies have revealed two distinct cellular mechanisms, one based on cell crawling and the other on junction contraction. Whether these two behaviors collaborate is unclear. Here, using live-cell imaging, we show that crawling and contraction act both independently and jointly but that CE is more effective when they are integrated via mechano-reciprocity. We thus developed a computational model considering both crawling and contraction. This model recapitulates the biomechanical efficacy of integrating the two modes and further clarifies how the two modes and their integration are influenced by cell adhesion. Finally, we use these insights to understand the function of an understudied catenin, Arvcf, during CE. These data are significant for providing interesting biomechanical and cell biological insights into a fundamental morphogenetic process that is implicated in human neural tube defects and skeletal dysplasias.


Subject(s)
Cell Adhesion Molecules , Neural Tube Defects , Cell Adhesion , Cell Adhesion Molecules/metabolism , Cell Movement , Humans , Morphogenesis
5.
Elife ; 102021 05 25.
Article in English | MEDLINE | ID: mdl-34032216

ABSTRACT

Morphogenesis is governed by the interplay of molecular signals and mechanical forces across multiple length scales. The last decade has seen tremendous advances in our understanding of the dynamics of protein localization and turnover at subcellular length scales, and at the other end of the spectrum, of mechanics at tissue-level length scales. Integrating the two remains a challenge, however, because we lack a detailed understanding of the subcellular patterns of mechanical properties of cells within tissues. Here, in the context of the elongating body axis of Xenopus embryos, we combine tools from cell biology and physics to demonstrate that individual cell-cell junctions display finely-patterned local mechanical heterogeneity along their length. We show that such local mechanical patterning is essential for the cell movements of convergent extension and is imparted by locally patterned clustering of a classical cadherin. Finally, the patterning of cadherins and thus local mechanics along cell-cell junctions are controlled by Planar Cell Polarity signaling, a key genetic module for CE that is mutated in diverse human birth defects.


Subject(s)
Cadherins/metabolism , Intercellular Junctions/metabolism , Single-Cell Analysis , Xenopus/embryology , Animals , Body Patterning , Cell Polarity , Morphogenesis
6.
Dev Dyn ; 250(5): 717-731, 2021 05.
Article in English | MEDLINE | ID: mdl-33368695

ABSTRACT

BACKGROUND: Explanted tissues from vertebrate embryos reliably develop in culture and have provided essential paradigms for understanding embryogenesis, from early embryological investigations of induction, to the extensive study of Xenopus animal caps, to the current studies of mammalian gastruloids. Cultured explants of the Xenopus dorsal marginal zone ("Keller" explants) serve as a central paradigm for studies of convergent extension cell movements, yet we know little about the global patterns of gene expression in these explants. RESULTS: In an effort to more thoroughly develop this important model system, we provide here a time-resolved bulk transcriptome for developing Keller explants. CONCLUSIONS: The dataset reported here provides a useful resource for those using Keller explants for studies of morphogenesis and provide genome-scale insights into the temporal patterns of gene expression in an important tissue when explanted and grown in culture.


Subject(s)
Embryo Culture Techniques , Gastrula/metabolism , Transcriptome , Xenopus laevis/metabolism , Animals , Xenopus laevis/genetics
7.
8.
Dev Cell ; 46(4): 389-396, 2018 08 20.
Article in English | MEDLINE | ID: mdl-30130529

ABSTRACT

Cell motility is a widespread biological property that is best understood in the context of individual cell migration. Indeed, studies of migration in culture have provided tremendous insight into the signals and mechanics involved and have laid the foundation for our understanding of similar migrations by larger cellular collectives. By contrast, our understanding of another flavor of movement, cell intercalation during convergent extension, is only now emerging. Here, we integrate divergent findings related to intercalation in different settings into a unifying model, paying attention to how this model does and does not resemble current models for directed cell migration.


Subject(s)
Body Patterning/physiology , Cell Movement/physiology , Models, Biological , Morphogenesis/physiology , Animals , Cell Culture Techniques , Consensus , Humans
9.
Dev Cell ; 45(1): 67-82.e6, 2018 04 09.
Article in English | MEDLINE | ID: mdl-29634937

ABSTRACT

We sought to understand how cells collectively elongate epithelial tubes. We first used 3D culture and biosensor imaging to demonstrate that epithelial cells enrich Ras activity, phosphatidylinositol (3,4,5)-trisphosphate (PIP3), and F-actin to their leading edges during migration within tissues. PIP3 enrichment coincided with, and could enrich despite inhibition of, F-actin dynamics, revealing a conserved migratory logic compared with single cells. We discovered that migratory cells can intercalate into the basal tissue surface and contribute to tube elongation. We then connected molecular activities to subcellular mechanics using force inference analysis. Migration and transient intercalation required specific and similar anterior-posterior ratios of interfacial tension. Permanent intercalations were distinguished by their capture at the boundary through time-varying tension dynamics. Finally, we integrated our experimental and computational data to generate a finite element model of tube elongation. Our model revealed that intercalation, interfacial tension dynamics, and high basal stress are together sufficient for mammary morphogenesis.


Subject(s)
Actins/metabolism , Cell Movement/physiology , Epithelial Cells/cytology , Mammary Glands, Animal/cytology , Morphogenesis/physiology , ras Proteins/metabolism , Animals , Cell Proliferation , Cells, Cultured , Epithelial Cells/metabolism , Female , Mammary Glands, Animal/metabolism , Mice , Mice, Transgenic , Signal Transduction , Surface Tension
10.
Development ; 143(6): 983-93, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26839364

ABSTRACT

Mammary branching morphogenesis is regulated by receptor tyrosine kinases (RTKs). We sought to determine how these RTK signals alter proliferation and migration to accomplish tube elongation in mouse. Both behaviors occur but it has been difficult to determine their relative contribution to elongation in vivo, as mammary adipocytes scatter light and limit the depth of optical imaging. Accordingly, we utilized 3D culture to study elongation in an experimentally accessible setting. We first used antibodies to localize RTK signals and discovered that phosphorylated ERK1/2 (pERK) was spatially enriched in cells near the front of elongating ducts, whereas phosphorylated AKT was ubiquitous. We next observed a gradient of cell migration speeds from rear to front of elongating ducts, with the front characterized by both high pERK and the fastest cells. Furthermore, cells within elongating ducts oriented both their protrusions and their migration in the direction of tube elongation. By contrast, cells within the organoid body were isotropically protrusive. We next tested the requirement for proliferation and migration. Early inhibition of proliferation blocked the creation of migratory cells, whereas late inhibition of proliferation did not block continued duct elongation. By contrast, pharmacological inhibition of either MEK or Rac1 signaling acutely blocked both cell migration and duct elongation. Finally, conditional induction of MEK activity was sufficient to induce collective cell migration and ductal elongation. Our data suggest a model for ductal elongation in which RTK-dependent proliferation creates motile cells with high pERK, the collective migration of which acutely requires both MEK and Rac1 signaling.


Subject(s)
Cell Movement , Epithelial Cells/cytology , Extracellular Signal-Regulated MAP Kinases/metabolism , Mammary Glands, Animal/cytology , Animals , Anisotropy , Cell Proliferation , Female , Humans , Mice, Transgenic , Mitogen-Activated Protein Kinase Kinases/metabolism , Morphogenesis , Organoids/metabolism , Phosphorylation , Signal Transduction , rac1 GTP-Binding Protein/metabolism
11.
Proc Natl Acad Sci U S A ; 113(6): E679-88, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26792522

ABSTRACT

Collective cell responses to exogenous cues depend on cell-cell interactions. In principle, these can result in enhanced sensitivity to weak and noisy stimuli. However, this has not yet been shown experimentally, and little is known about how multicellular signal processing modulates single-cell sensitivity to extracellular signaling inputs, including those guiding complex changes in the tissue form and function. Here we explored whether cell-cell communication can enhance the ability of cell ensembles to sense and respond to weak gradients of chemotactic cues. Using a combination of experiments with mammary epithelial cells and mathematical modeling, we find that multicellular sensing enables detection of and response to shallow epidermal growth factor (EGF) gradients that are undetectable by single cells. However, the advantage of this type of gradient sensing is limited by the noisiness of the signaling relay, necessary to integrate spatially distributed ligand concentration information. We calculate the fundamental sensory limits imposed by this communication noise and combine them with the experimental data to estimate the effective size of multicellular sensory groups involved in gradient sensing. Functional experiments strongly implicated intercellular communication through gap junctions and calcium release from intracellular stores as mediators of collective gradient sensing. The resulting integrative analysis provides a framework for understanding the advantages and limitations of sensory information processing by relays of chemically coupled cells.


Subject(s)
Cell Communication , Morphogenesis , Animals , Cadherins/metabolism , Calcium/metabolism , Calcium Signaling/drug effects , Cell Communication/drug effects , Cell Movement/drug effects , Computer Simulation , Epidermal Growth Factor/pharmacology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Female , Gap Junctions/drug effects , Gap Junctions/metabolism , Ions , Ligands , Mammary Glands, Animal/cytology , Models, Biological , Morphogenesis/drug effects , Organoids/cytology , Organoids/drug effects , Rats , Time Factors
12.
Methods Mol Biol ; 1189: 135-62, 2015.
Article in English | MEDLINE | ID: mdl-25245692

ABSTRACT

Epithelia are fundamental tissues that line cavities, glands, and outer body surfaces. We use three-dimensional (3D) embedded culture of primary murine mammary epithelial ducts, called "organoids," to recapitulate in days in culture epithelial programs that occur over weeks deep within the body. Modulating the composition of the extracellular matrix (ECM) allows us to model cell- and tissue-level behaviors observed in normal development, such as branching morphogenesis, and in cancer, such as invasion and dissemination. Here, we describe a collection of protocols for 3D culture of mammary organoids in different ECMs and for immunofluorescence staining of 3D culture samples and mammary gland tissue sections. We illustrate expected phenotypic outcomes of each assay and provide troubleshooting tips for commonly encountered technical problems.


Subject(s)
Cell Culture Techniques/methods , Epithelial Cells/cytology , Mammary Glands, Animal/growth & development , Morphogenesis , Animals , Biological Assay , Cell Separation , Cell Shape/drug effects , Collagen/pharmacology , Collagen Type I/pharmacology , Drug Combinations , Epithelial Cells/drug effects , Female , Fluorescent Antibody Technique , Laminin/pharmacology , Mammary Glands, Animal/cytology , Mammary Glands, Animal/drug effects , Mice , Morphogenesis/drug effects , Organoids/cytology , Organoids/drug effects , Phenotype , Proteoglycans/pharmacology , Rats , Serum Albumin, Bovine/metabolism , Staining and Labeling
13.
Semin Cell Dev Biol ; 31: 124-31, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24747369

ABSTRACT

The mammary gland is composed of a highly branched network of epithelial tubes, embedded within a complex stroma. The mammary epithelium originates during embryonic development from an epidermal placode. However, the majority of ductal elongation and bifurcation occurs postnatally, in response to steroid hormone and growth factor receptor signaling. The process of pubertal branching morphogenesis involves both elongation of the primary ducts across the length of the fat pad and a wave of secondary branching that elaborates the ductal network. Recent studies have revealed that mammary epithelial morphogenesis is accomplished by transitions between simple and stratified organization. During active morphogenesis, the epithelium is stratified, highly proliferative, has few intercellular junctions, and exhibits incomplete apico-basal polarity. In this review, we discuss recent advances in our understanding of the relationship between epithelial architecture, epithelial polarity, and ductal elongation.


Subject(s)
Mammary Glands, Animal/cytology , Mammary Glands, Animal/growth & development , Mammary Glands, Human/cytology , Mammary Glands, Human/growth & development , Morphogenesis , Animals , Epithelium/growth & development , Female , Humans
14.
Development ; 141(5): 1085-94, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24550116

ABSTRACT

Mammary ducts are elongated during development by stratified epithelial structures, known as terminal end buds (TEBs). TEBs exhibit reduced apicobasal polarity and extensive proliferation. A major unanswered question concerns the mechanism by which the simple ductal epithelium stratifies during TEB formation. We sought to elucidate this mechanism using real-time imaging of growth factor-induced stratification in 3D cultures of mouse primary epithelial organoids. We hypothesized that stratification could result from vertical divisions in either the apically positioned luminal epithelial cells or the basally positioned myoepithelial cells. Stratification initiated exclusively from vertical apical cell divisions, both in 3D culture and in vivo. During vertical apical divisions, only the mother cell retained tight junctions and segregated apical membranes. Vertical daughter cells initiated an unpolarized cell population located between the luminal and myoepithelial cells, similar to the unpolarized body cells in the TEB. As stratification and loss of apicobasal polarity are early hallmarks of cancer, we next determined the cellular mechanism of oncogenic stratification. Expression of activated ERBB2 induced neoplastic stratification through analogous vertical divisions of apically positioned luminal epithelial cells. However, ERBB2-induced stratification was accompanied by tissue overgrowth and acute loss of both tight junctions and apical polarity. Expression of phosphomimetic MEK (MEK1DD), a major ERBB2 effector, also induced stratification through vertical apical cell divisions. However, MEK1DD-expressing organoids exhibited normal levels of growth and retained apicobasal polarity. We conclude that both normal and neoplastic stratification are accomplished through receptor tyrosine kinase signaling dependent vertical cell divisions within the luminal epithelial cell layer.


Subject(s)
Epithelial Cells/metabolism , Mammary Glands, Animal/cytology , Animals , Cells, Cultured , Epithelial Cells/cytology , Mice , Mice, Transgenic , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Signal Transduction/physiology , Tight Junctions/genetics , Tight Junctions/metabolism , Zonula Occludens-1 Protein/genetics , Zonula Occludens-1 Protein/metabolism
15.
J Cell Sci ; 125(Pt 11): 2638-54, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22344263

ABSTRACT

Normal mammary morphogenesis involves transitions between simple and multilayered epithelial organizations. We used electron microscopy and molecular markers to determine whether intercellular junctions and apico-basal polarity were maintained in the multilayered epithelium. We found that multilayered elongating ducts had polarized apical and basal tissue surfaces both in three-dimensional culture and in vivo. However, individual cells were only polarized on surfaces in contact with the lumen or extracellular matrix. The basolateral marker scribble and the apical marker atypical protein kinase C zeta localized to all interior cell membranes, whereas PAR3 displayed a cytoplasmic localization, suggesting that the apico-basal polarity was incomplete. Despite membrane localization of E-cadherin and ß-catenin, we did not observe a defined zonula adherens connecting interior cells. Instead, interior cells were connected through desmosomes and exhibited complex interdigitating membrane protrusions. Single-cell labeling revealed that individual cells were both protrusive and migratory within the epithelial multilayer. Inhibition of Rho kinase (ROCK) further reduced intercellular adhesion on apical and lateral surfaces but did not disrupt basal tissue organization. Following morphogenesis, segregated membrane domains were re-established and junctional complexes re-formed. We observed similar epithelial organization during mammary morphogenesis in organotypic culture and in vivo. We conclude that mammary epithelial morphogenesis involves a reversible, spatially limited, reduction in polarity and intercellular junctions and active individualistic cell migration. Our data suggest that reductions in polarity and adhesion during breast cancer progression might reflect partial recapitulation of a normal developmental program.


Subject(s)
Cell Movement , Epithelial Cells/cytology , Epithelium/metabolism , Mammary Glands, Animal/cytology , Amides/pharmacology , Animals , Cell Communication/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cell Movement/drug effects , Cell Polarity/drug effects , Cell Shape/drug effects , Cell Surface Extensions/metabolism , Cell Surface Extensions/ultrastructure , Collagen , Desmosomes/drug effects , Desmosomes/metabolism , Desmosomes/ultrastructure , Drug Combinations , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Epithelial Cells/ultrastructure , Epithelium/drug effects , Epithelium/ultrastructure , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Female , Laminin , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/ultrastructure , Mice , Models, Biological , Morphogenesis/drug effects , Myosin-Light-Chain Kinase/metabolism , Organoids/cytology , Organoids/drug effects , Organoids/ultrastructure , Proteoglycans , Pyridines/pharmacology , Tight Junctions/metabolism , Tight Junctions/ultrastructure , Tissue Culture Techniques , rac GTP-Binding Proteins/metabolism , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...