Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Development ; 151(15)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39092607

ABSTRACT

Branching morphogenesis is a characteristic feature of many essential organs, such as the lung and kidney, and most glands, and is the net result of two tissue behaviors: branch point initiation and elongation. Each branched organ has a distinct architecture customized to its physiological function, but how patterning occurs in these ramified tubular structures is a fundamental problem of development. Here, we use quantitative 3D morphometrics, time-lapse imaging, manipulation of ex vivo cultured mouse embryonic organs and mice deficient in the planar cell polarity component Vangl2 to address this question in the developing mammary gland. Our results show that the embryonic epithelial trees are highly complex in topology owing to the flexible use of two distinct modes of branch point initiation: lateral branching and tip bifurcation. This non-stereotypy was contrasted by the remarkably constant average branch frequency, indicating a ductal growth invariant, yet stochastic, propensity to branch. The probability of branching was malleable and could be tuned by manipulating the Fgf10 and Tgfß1 pathways. Finally, our in vivo data and ex vivo time-lapse imaging suggest the involvement of tissue rearrangements in mammary branch elongation.


Subject(s)
Mammary Glands, Animal , Morphogenesis , Animals , Mammary Glands, Animal/embryology , Mammary Glands, Animal/growth & development , Mice , Female , Fibroblast Growth Factor 10/metabolism , Fibroblast Growth Factor 10/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Transforming Growth Factor beta1/metabolism , Time-Lapse Imaging , Cell Polarity , Embryo, Mammalian/metabolism , Signal Transduction
2.
Mol Cell Proteomics ; 18(2): 277-293, 2019 02.
Article in English | MEDLINE | ID: mdl-30404858

ABSTRACT

Integrin-mediated laminin adhesions mediate epithelial cell anchorage to basement membranes and are critical regulators of epithelial cell polarity. Integrins assemble large multiprotein complexes that link to the cytoskeleton and convey signals into the cells. Comprehensive proteomic analyses of actin network-linked focal adhesions (FA) have been performed, but the molecular composition of intermediate filament-linked hemidesmosomes (HD) remains incompletely characterized. Here we have used proximity-dependent biotin identification (BioID) technology to label and characterize the interactome of epithelia-specific ß4-integrin that, as α6ß4-heterodimer, forms the core of HDs. The analysis identified ∼150 proteins that were specifically labeled by BirA-tagged integrin-ß4. In addition to known HDs proteins, the interactome revealed proteins that may indirectly link integrin-ß4 to actin-connected protein complexes, such as FAs and dystrophin/dystroglycan complexes. The specificity of the screening approach was validated by confirming the HD localization of two candidate ß4-interacting proteins, utrophin (UTRN) and ELKS/Rab6-interacting/CAST family member 1 (ERC1). Interestingly, although establishment of functional HDs depends on the formation of α6ß4-heterodimers, the assembly of ß4-interactome was not strictly dependent on α6-integrin expression. Our survey to the HD interactome sets a precedent for future studies and provides novel insight into the mechanisms of HD assembly and function of the ß4-integrin.


Subject(s)
Integrin beta4/chemistry , Integrin beta4/metabolism , Proteomics/methods , Animals , Biotinylation , Chromatography, Liquid , Dogs , Madin Darby Canine Kidney Cells , Protein Interaction Maps , Protein Multimerization , Tandem Mass Spectrometry
3.
Nat Cell Biol ; 26(2): 207-218, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38302719

ABSTRACT

Morphogenesis and cell state transitions must be coordinated in time and space to produce a functional tissue. An excellent paradigm to understand the coupling of these processes is mammalian hair follicle development, which is initiated by the formation of an epithelial invagination-termed placode-that coincides with the emergence of a designated hair follicle stem cell population. The mechanisms directing the deformation of the epithelium, cell state transitions and physical compartmentalization of the placode are unknown. Here we identify a key role for coordinated mechanical forces stemming from contractile, proliferative and proteolytic activities across the epithelial and mesenchymal compartments in generating the placode structure. A ring of fibroblast cells gradually wraps around the placode cells to generate centripetal contractile forces, which, in collaboration with polarized epithelial myosin activity, promote elongation and local tissue thickening. These mechanical stresses further enhance compartmentalization of Sox9 expression to promote stem cell positioning. Subsequently, proteolytic remodelling locally softens the basement membrane to facilitate a release of pressure on the placode, enabling localized cell divisions, tissue fluidification and epithelial invagination into the underlying mesenchyme. Together, our experiments and modelling identify dynamic cell shape transformations and tissue-scale mechanical cooperation as key factors for orchestrating organ formation.


Subject(s)
Hair Follicle , Mammals , Animals , Cell Shape , Epithelium , Morphogenesis , Cell Division , Hair Follicle/metabolism
4.
iScience ; 26(4): 106490, 2023 Apr 21.
Article in English | MEDLINE | ID: mdl-37096043

ABSTRACT

Maintaining apicobasal polarity (ABP) is crucial for epithelial integrity and homeostasis during tissue development. Although intracellular mechanisms underlying ABP establishment have been well studied, it remains to be addressed how the ABP coordinates tissue growth and homeostasis. By studying Scribble, a key ABP determinant, we address molecular mechanisms underlying ABP-mediated growth control in the Drosophila wing imaginal disc. Our data reveal that genetic and physical interactions between Scribble, Septate junction complex and α-Catenin appear to be key for sustaining ABP-mediated growth control. Cells with conditional scribble knockdown instigate the loss of α-Catenin, ultimately leading to the formation of neoplasia accompanying with activation of Yorkie. In contrast, cells expressing wild type scribble progressively restore ABP in scribble hypomorphic mutant cells in a non-autonomous manner. Our findings provide unique insights into cellular communication among optimal and sub-optimal cells to regulate epithelial homeostasis and growth.

5.
J Cell Biol ; 222(9)2023 09 04.
Article in English | MEDLINE | ID: mdl-37367826

ABSTRACT

Branching morphogenesis is an evolutionary solution to maximize epithelial function in a compact organ. It involves successive rounds of branch elongation and branch point formation to generate a tubular network. In all organs, branch points can form by tip splitting, but it is unclear how tip cells coordinate elongation and branching. Here, we addressed these questions in the embryonic mammary gland. Live imaging revealed that tips advance by directional cell migration and elongation relies upon differential cell motility that feeds a retrograde flow of lagging cells into the trailing duct, supported by tip proliferation. Tip bifurcation involved localized repression of cell cycle and cell motility at the branch point. Cells in the nascent daughter tips remained proliferative but changed their direction to elongate new branches. We also report the fundamental importance of epithelial cell contractility for mammary branching morphogenesis. The co-localization of cell motility, non-muscle myosin II, and ERK activities at the tip front suggests coordination/cooperation between these functions.


Subject(s)
Epithelial Cells , Mammary Glands, Animal , Morphogenesis , Cell Division , Cell Movement , Mammary Glands, Animal/embryology , Morphogenesis/physiology , Mammals , Myosin Type II/physiology
6.
Front Cell Dev Biol ; 10: 912001, 2022.
Article in English | MEDLINE | ID: mdl-36211469

ABSTRACT

Epithelial homeostasis is an emergent property of both physical and biochemical signals emanating from neighboring cells and across tissue. A recent study reveals that Scribble, an apico-basal polarity determinant, cooperates with α-Catenin, an adherens junction component, to regulate tissue homeostasis in the Drosophila wing imaginal disc. However, it remains to be addressed whether similar mechanisms are utilized in vertebrates. In this study, we first address how α-Catenin cooperates with Scribble to regulate epithelial homeostasis and growth in mammalian cells. Our data show that α-Catenin and Scribble interact physically in mammalian cells. We then found that both α-Catenin and Scribble are required for regulating nuclear translocation of YAP, an effector of the Hippo signaling pathway. Furthermore, ectopic Scribble suffices to suppress YAP in an α-Catenin-dependent manner. Then, to test our hypothesis that Scribble amounts impact epithelial growth, we use the Drosophila wing imaginal disc. We show that Scribble expression is complementary to Yorkie signal, the Drosophila ortholog of YAP. Ectopic expression of full-length Scribble or Scribble Leucine Rich Region (LRR):α-Catenin chimera sufficiently down-regulates Yorkie signal, leading to smaller wing size. Moreover, Scribble LRR:α-Catenin chimera rescues scribble mutant clones in the wing imaginal disc to maintain tissue homeostasis. Taken together, our studies suggest that the association of cell polarity component Scribble with α-Catenin plays a conserved role in epithelial homeostasis and growth.

7.
Methods Mol Biol ; 2471: 1-18, 2022.
Article in English | MEDLINE | ID: mdl-35175589

ABSTRACT

Mammary gland development starts during embryogenesis, and the process continues after birth. During development, the mammary gland undergoes massive morphological and physiological alterations including growth, invasion, and branching morphogenesis providing an ideal model for stem cell and cancer biology studies. Great efforts have been made in understanding mammary gland development during puberty and adulthood; however, the process during embryogenesis is still elusive. One reason is that the tools to study tissue dynamics during development are limited, which is partially due to the lack of an ex vivo culture method. Here we describe an updated organ culture protocol of the murine embryonic mammary gland. This powerful tool allows monitoring of growth and branching morphogenesis of mammary gland ex vivo by live imaging. In addition, we introduce a novel method for culturing intact, stroma-free mammary rudiments from late gestation mouse embryos in 3D in Matrigel. This approach can be used to identify the direct stromal cues for branching morphogenesis.


Subject(s)
Epithelial Cells , Mammary Glands, Animal , Animals , Female , Mice , Morphogenesis , Organ Culture Techniques , Pregnancy
8.
Methods Mol Biol ; 2217: 57-69, 2021.
Article in English | MEDLINE | ID: mdl-33215377

ABSTRACT

Integrins are heterodimeric adhesion receptors that maintain cell-extracellular matrix (ECM) interactions in diverse tissue microenvironments. They mediate cell adhesion and signaling through the assembly of large cytoplasmic multiprotein complexes that focally connect with the cytoskeleton. Integrin adhesion complexes (IAC) are specialized by the type of integrin-ECM contact and are sensitive to mechanical forces. Thus, they encrypt context-dependent information about the microenvironment in their composition. Signals mediated through IACs modulate many aspects of cell behavior, which allows cells to adapt to their surroundings. To gain insights into their function, IACs have been isolated from cultured cells and explored by proteomics. IACs are insoluble by nature and held together by transient/weak interactions, which makes it challenging to isolate intact IACs. Usually all IACs coupled to a specified ECM, which may employ different integrins, are isolated. Here we describe an alternative method based on proximity-dependent biotin identification (BioID), where specific integrin interaction partners are labeled in live cells and isolated without the need to isolate intact IACs.


Subject(s)
Biological Assay , Carbon-Nitrogen Ligases/metabolism , Escherichia coli Proteins/metabolism , Integrin alpha Chains/metabolism , Integrin beta Chains/metabolism , Protein Interaction Mapping/methods , Recombinant Fusion Proteins/metabolism , Repressor Proteins/metabolism , Amino Acid Sequence , Animals , Biotin/chemistry , Biotin/metabolism , Biotinylation , Carbon-Nitrogen Ligases/genetics , Cell Adhesion , Cell Membrane/chemistry , Cell Membrane/metabolism , Dogs , Escherichia coli Proteins/genetics , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Integrin alpha Chains/classification , Integrin alpha Chains/genetics , Integrin beta Chains/classification , Integrin beta Chains/genetics , Madin Darby Canine Kidney Cells , Plasmids/chemistry , Plasmids/metabolism , Protein Binding , Protein Multimerization , Recombinant Fusion Proteins/genetics , Repressor Proteins/genetics , Staining and Labeling/methods , Transfection
9.
J Cell Biol ; 220(8)2021 08 02.
Article in English | MEDLINE | ID: mdl-34042944

ABSTRACT

The mammary gland develops from the surface ectoderm during embryogenesis and proceeds through morphological phases defined as placode, hillock, bud, and bulb stages followed by branching morphogenesis. During this early morphogenesis, the mammary bud undergoes an invagination process where the thickened bud initially protrudes above the surface epithelium and then transforms to a bulb and sinks into the underlying mesenchyme. The signaling pathways regulating the early morphogenetic steps have been identified to some extent, but the underlying cellular mechanisms remain ill defined. Here, we use 3D and 4D confocal microscopy to show that the early growth of the mammary rudiment is accomplished by migration-driven cell influx, with minor contributions of cell hypertrophy and proliferation. We delineate a hitherto undescribed invagination mechanism driven by thin, elongated keratinocytes-ring cells-that form a contractile rim around the mammary bud and likely exert force via the actomyosin network. Furthermore, we show that conditional deletion of nonmuscle myosin IIA (NMIIA) impairs invagination, resulting in abnormal mammary bud shape.


Subject(s)
Actomyosin/metabolism , Cell Movement , Epithelial Cells/metabolism , Mammary Glands, Animal/metabolism , Mechanotransduction, Cellular , Animals , Cell Proliferation , Epithelial Cells/ultrastructure , Female , Gene Expression Regulation, Developmental , Gestational Age , Hypertrophy , Keratinocytes/metabolism , Keratinocytes/ultrastructure , Mammary Glands, Animal/embryology , Mammary Glands, Animal/ultrastructure , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Microscopy, Electron, Scanning , Microscopy, Fluorescence , Morphogenesis
10.
Curr Opin Cell Biol ; 61: 72-78, 2019 12.
Article in English | MEDLINE | ID: mdl-31387017

ABSTRACT

Branching morphogenesis is a fundamental developmental program that generates large epithelial surfaces in a limited three-dimensional space. It is regulated by inductive tissue interactions whose effects are mediated by soluble signaling molecules, and cell-cell and cell-extracellular matrix interactions. Here, we will review recent studies on inductive signaling interactions governing branching morphogenesis in light of phenotypes of mouse mutants and ex vivo organ culture studies with emphasis on developing mammary and salivary glands. We will highlight advances in understanding how cell fate decisions are intimately linked with branching morphogenesis. We will also discuss novel insights into the molecular control of cellular mechanisms driving the formation of these arborized ductal structures and reflect upon how distinct spatial patterns are generated.


Subject(s)
Mammary Glands, Animal/embryology , Mammary Glands, Animal/metabolism , Morphogenesis/physiology , Salivary Glands/embryology , Salivary Glands/metabolism , Animals , Breast/embryology , Cell Differentiation , Epithelial Cells/cytology , Extracellular Matrix , Female , Mice , Organ Culture Techniques , Signal Transduction
11.
Elife ; 72018 07 31.
Article in English | MEDLINE | ID: mdl-30063206

ABSTRACT

Mesenchymal condensation is a critical step in organogenesis, yet the underlying molecular and cellular mechanisms remain poorly understood. The hair follicle dermal condensate is the precursor to the permanent mesenchymal unit of the hair follicle, the dermal papilla, which regulates hair cycling throughout life and bears hair inductive potential. Dermal condensate morphogenesis depends on epithelial Fibroblast Growth Factor 20 (Fgf20). Here, we combine mouse models with 3D and 4D microscopy to demonstrate that dermal condensates form de novo and via directional migration. We identify cell cycle exit and cell shape changes as early hallmarks of dermal condensate morphogenesis and find that Fgf20 primes these cellular behaviors and enhances cell motility and condensation. RNAseq profiling of immediate Fgf20 targets revealed induction of a subset of dermal condensate marker genes. Collectively, these data indicate that dermal condensation occurs via directed cell movement and that Fgf20 orchestrates the early cellular and molecular events.


Subject(s)
Cell Cycle , Cell Movement , Dermis/cytology , Fibroblast Growth Factors/metabolism , Hair Follicle/cytology , Actins/metabolism , Animals , Cell Aggregation , Cell Lineage , Cell Shape , Dermis/ultrastructure , Fibroblast Growth Factor 9/pharmacology , Fibroblasts/cytology , Fibroblasts/metabolism , Mice, Inbred C57BL , Morphogenesis , Receptors, Fibroblast Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , SOXB1 Transcription Factors/metabolism , Signal Transduction , Transcription, Genetic
12.
Article in English | MEDLINE | ID: mdl-28159878

ABSTRACT

Laminin, a basement membrane protein discovered in 1979, was shortly thereafter implicated in the polarization of epithelial cells in both mammals and a variety of lower organisms. To transduce a spatial cue to the intrinsic polarization machinery, laminin must polymerize into a dense network that forms the foundation of the basement membrane. Evidence suggests that activation of the small GTPase Rac1 by ß1-integrins mobilizes laminin-binding integrins and dystroglycan to consolidate formation of the laminin network and initiate rearrangements of both the actin and microtubule cytoskeleton to help establish the apicobasal axis. A key coordinator of spatial signals from laminin is the serine-threonine kinase Par-1, which is known to affect dystroglycan availability, microtubule and actin organization, and lumen formation. The signaling protein integrin-linked kinase (ILK) may also play a role. Despite significant advances, knowledge of the mechanism by which assembled laminin produces a spatial signal remains fragmentary, and much more research into the complex functions of laminin in polarization and other cellular processes is needed.


Subject(s)
Cell Polarity , Epithelial Cells/physiology , Laminin/physiology , Animals
13.
PLoS One ; 6(5): e19453, 2011 May 06.
Article in English | MEDLINE | ID: mdl-21573123

ABSTRACT

BACKGROUND: Formation of apical compartments underlies the morphogenesis of most epithelial organs during development. The extracellular matrix (ECM), particularly the basement membrane (BM), plays an important role in orienting the apico-basal polarity and thereby the positioning of apical lumens. Integrins have been recognized as essential mediators of matrix-derived polarity signals. The importance of ß1-integrins in epithelial polarization is well established but the significance of the accompanying α-subunits have not been analyzed in detail. PRINCIPAL FINDINGS: Here we demonstrate that two distinct integrin-dependent pathways regulate formation of apical lumens to ensure robust apical membrane biogenesis under different microenvironmental conditions; 1) α2ß1- and α6ß4-integrins were required to establish a basal cue that depends on Rac1-activity and guides apico-basal cell polarization. 2) α3ß1-integrins were implicated in positioning of mitotic spindles in cysts, a process that is essential for Cdc42-driven epithelial hollowing. SIGNIFICANCE: Identification of the separate processes driven by particular integrin receptors clarifies the functional hierarchies between the different integrins co-expressed in epithelial cells and provides valuable insight into the complexity of cell-ECM interactions thereby guiding future studies addressing the molecular basis of epithelial morphogenesis during development and disease.


Subject(s)
Epithelial Cells/cytology , Epithelial Cells/metabolism , Integrin alpha2beta1/metabolism , Integrin alpha6beta4/metabolism , Animals , Cell Adhesion/genetics , Cell Adhesion/physiology , Cell Line , Cell Polarity/genetics , Cell Polarity/physiology , Dogs , Fluorescent Antibody Technique , Immunoblotting , Integrin alpha2beta1/genetics , Integrin alpha3beta1/genetics , Integrin alpha3beta1/metabolism , Integrin alpha6beta4/genetics , Microscopy, Confocal , Microscopy, Fluorescence , RNA, Small Interfering
SELECTION OF CITATIONS
SEARCH DETAIL