Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Development ; 150(18)2023 09 15.
Article in English | MEDLINE | ID: mdl-37681301

ABSTRACT

Drosophila blood cells called hemocytes form an efficient barrier against infections and tissue damage. During metamorphosis, hemocytes undergo tremendous changes in their shape and behavior, preparing them for tissue clearance. Yet, the diversity and functional plasticity of pupal blood cells have not been explored. Here, we combine single-cell transcriptomics and high-resolution microscopy to dissect the heterogeneity and plasticity of pupal hemocytes. We identified undifferentiated and specified hemocytes with different molecular signatures associated with distinct functions such as antimicrobial, antifungal immune defense, cell adhesion or secretion. Strikingly, we identified a highly migratory and immune-responsive pupal cell population expressing typical markers of the posterior signaling center (PSC), which is known to be an important niche in the larval lymph gland. PSC-like cells become restricted to the abdominal segments and are morphologically very distinct from typical Hemolectin (Hml)-positive plasmatocytes. G-TRACE lineage experiments further suggest that PSC-like cells can transdifferentiate to lamellocytes triggered by parasitoid wasp infestation. In summary, we present the first molecular description of pupal Drosophila blood cells, providing insights into blood cell functional diversification and plasticity during pupal metamorphosis.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Drosophila/metabolism , Drosophila melanogaster/metabolism , Transcriptome/genetics , Cell Differentiation , Blood Cells/metabolism , Drosophila Proteins/metabolism , Hemocytes , Larva/metabolism
2.
Front Cell Dev Biol ; 11: 1171930, 2023.
Article in English | MEDLINE | ID: mdl-37025173

ABSTRACT

The actin cytoskeleton represents a highly dynamic filament system providing cell structure and mechanical forces to drive a variety of cellular processes. The dynamics of the actin cytoskeleton are controlled by a number of conserved proteins that maintain the pool of actin monomers, promote actin nucleation, restrict the length of actin filaments and cross-link filaments into networks or bundles. Previous work has been established that cytoplasmic calcium is an important signal to rapidly relay information to the actin cytoskeleton, but the underlying mechanisms remain poorly understood. Here, we summarize new recent perspectives on how calcium fluxes are transduced to the actin cytoskeleton in a physiological context. In this mini-review we will focus on three calcium-binding EF-hand-containing actin cross-linking proteins, α-actinin, plastin and EFHD2/Swiprosin-1, and how these conserved proteins affect the cell's actin reorganization in the context of cell migration and wound closure in response to calcium.

3.
J Cell Sci ; 136(6)2023 03 15.
Article in English | MEDLINE | ID: mdl-36727484

ABSTRACT

Exocytosis is a fundamental cellular process by which cells secrete cargos from their apical membrane into the extracellular lumen. Cargo release proceeds in sequential steps that depend on coordinated assembly and organization of an actin cytoskeletal network. Here, we identified the conserved actin-crosslinking protein Swip-1 as a novel regulator controlling exocytosis of glue granules in the Drosophila salivary gland. Real-time imaging revealed that Swip-1 is simultaneously recruited with F-actin onto secreting granules in proximity to the apical membrane. We observed that Swip-1 is rapidly cleared at the point of secretory vesicle fusion and colocalizes with actomyosin network around the fused vesicles. Loss of Swip-1 function impairs secretory cargo expulsion, resulting in strongly delayed secretion. Thus, our results uncover a novel role of Swip-1 in secretory vesicle compression and expulsion of cargo during regulated exocytosis. Remarkably, this function neither requires Ca2+ binding nor dimerization of Swip-1. Our data rather suggest that Swip-1 regulates actomyosin activity upstream of Rho-GTPase signaling to drive proper vesicle membrane crumpling and expulsion of cargo.


Subject(s)
Actins , Drosophila , Animals , Drosophila/metabolism , Actins/metabolism , Actomyosin/metabolism , Exocytosis/physiology , Secretory Vesicles/metabolism , Salivary Glands/metabolism
4.
Methods Mol Biol ; 2608: 117-129, 2023.
Article in English | MEDLINE | ID: mdl-36653705

ABSTRACT

Collective cell migration has a key role in tissue morphogenesis, wound healing, tissue regeneration, and cancer invasion. In recent years, different animal models have been established to analyze how chemical and mechanical stimuli shape the behavior of single cells into tissues and organs. At present, there are still only a few model systems that allow to genetically dissect underlying molecular mechanisms driving cell motility during tissue morphogenesis at high resolution in real time. Here, we provide a detailed protocol and toolbox for ex vivo culturing of Drosophila testes for 4D live imaging of myotube collective migration, which allows to genetically address a wide range of developmental and cell biological questions regarding modes of filopodia-based protrusion/locomotion, cell-cell adhesion, cytoskeletal modes of collective decision-making, and collective closure processes. Additionally, this protocol has been successfully used in combination with laser-induced single-cell ablation and pharmacological treatments, but it can also be used with confocal microscopy after tissue fixation.


Subject(s)
Cell Movement , Drosophila , Animals , Male , Morphogenesis , Muscle Fibers, Skeletal , Testis
5.
Front Mol Biosci ; 9: 963635, 2022.
Article in English | MEDLINE | ID: mdl-36060241

ABSTRACT

Actin, as an ancient and fundamental protein, participates in various cytoplasmic as well as nuclear functions in eukaryotic cells. Based on its manifold tasks in the nucleus, it is a reasonable assumption that the nuclear presence of actin is essential for the cell, and consequently, its nuclear localization is ensured by a robust system. However, today only a single nuclear import and a single nuclear export pathway is known which maintain the dynamic balance between cytoplasmic and nuclear actin pools. In our work, we tested the robustness of the nuclear import of actin, and investigated whether the perturbations of nuclear localization affect the viability of the whole organism. For this aim, we generated a genetic system in Drosophila, in which we rescued the lethal phenotype of the null mutation of the Actin5C gene with transgenes that express different derivatives of actin, including a Nuclear Export Signal (NES)-tagged isoform which ensures forced nuclear export of the protein. We also disrupted the SUMOylation site of actin, suggested earlier to be responsible for nuclear retention, and eliminated the activity of the single nuclear import factor dedicated to actin. We found that, individually, none of the above mentioned manipulations led to a notable reduction in nuclear actin levels and thus, fully rescued lethality. However, the NES tagging of actin, together with the knock out of its importin, significantly reduced the amount of nuclear actin and induced lethality, confirming that the presence of actin in the nucleus is essential, and thereby, over-secured. Supporting this, we identified novel nuclear importins specific to actin, which sheds light on the mechanism behind the robustness of nuclear localization of actin, and supports the idea of essentiality of its nuclear functions.

6.
Nat Commun ; 13(1): 2492, 2022 05 06.
Article in English | MEDLINE | ID: mdl-35524157

ABSTRACT

Changes in cell morphology require the dynamic remodeling of the actin cytoskeleton. Calcium fluxes have been suggested as an important signal to rapidly relay information to the actin cytoskeleton, but the underlying mechanisms remain poorly understood. Here, we identify the EF-hand domain containing protein EFhD2/Swip-1 as a conserved lamellipodial protein strongly upregulated in Drosophila macrophages at the onset of metamorphosis when macrophage behavior shifts from quiescent to migratory state. Loss- and gain-of-function analysis confirm a critical function of EFhD2/Swip-1 in lamellipodial cell migration in fly and mouse melanoma cells. Contrary to previous assumptions, TIRF-analyses unambiguously demonstrate that EFhD2/Swip-1 proteins efficiently cross-link actin filaments in a calcium-dependent manner. Using a single-cell wounding model, we show that EFhD2/Swip-1 promotes wound closure in a calcium-dependent manner. Mechanistically, our data suggest that transient calcium bursts reduce EFhD2/Swip-1 cross-linking activity and thereby promote rapid reorganization of existing actin networks to drive epithelial wound closure.


Subject(s)
Actins , Calcium-Binding Proteins , Calcium , Wound Healing , Actins/metabolism , Animals , Calcium/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Movement , Drosophila , Drosophila Proteins , Mice , Signal Transduction
7.
J Cell Sci ; 134(23)2021 12 01.
Article in English | MEDLINE | ID: mdl-34730182

ABSTRACT

The WAVE regulatory complex (WRC) is the main activator of the Arp2/3 complex, promoting lamellipodial protrusions in migrating cells. The WRC is basally inactive but can be activated by Rac1 and phospholipids, and through phosphorylation. However, the in vivo relevance of the phosphorylation of WAVE proteins remains largely unknown. Here, we identified casein kinase I alpha (CK1α) as a regulator of WAVE, thereby controlling cell shape and cell motility in Drosophila macrophages. CK1α binds and phosphorylates WAVE in vitro. Phosphorylation of WAVE by CK1α appears not to be required for activation but, rather, regulates its stability. Pharmacologic inhibition of CK1α promotes ubiquitin-dependent degradation of WAVE. Consistently, loss of Ck1α but not ck2 function phenocopies the depletion of WAVE. Phosphorylation-deficient mutations in the CK1α consensus sequences within the VCA domain of WAVE can neither rescue mutant lethality nor lamellipodium defects. By contrast, phosphomimetic mutations rescue all cellular and developmental defects. Finally, RNAi-mediated suppression of 26S proteasome or E3 ligase complexes substantially rescues lamellipodia defects in CK1α-depleted macrophages. Therefore, we conclude that basal phosphorylation of WAVE by CK1α protects it from premature ubiquitin-dependent degradation, thus promoting WAVE function in vivo. This article has an associated First Person interview with the first author of the paper.


Subject(s)
Casein Kinase Ialpha , Casein Kinase Ialpha/genetics , Casein Kinase Ialpha/metabolism , Cell Shape , Humans , Immunity , Phosphorylation , Wiskott-Aldrich Syndrome Protein Family/metabolism
8.
Bioessays ; 43(11): e2100124, 2021 11.
Article in English | MEDLINE | ID: mdl-34480489

ABSTRACT

Collective migration is a key process that is critical during development, as well as in physiological and pathophysiological processes including tissue repair, wound healing and cancer. Studies in genetic model organisms have made important contributions to our current understanding of the mechanisms that shape cells into different tissues during morphogenesis. Recent advances in high-resolution and live-cell-imaging techniques provided new insights into the social behavior of cells based on careful visual observations within the context of a living tissue. In this review, we will compare Drosophila testis nascent myotube migration with established in vivo model systems, elucidate similarities, new features and principles in collective cell migration.


Subject(s)
Muscle Fibers, Skeletal , Pseudopodia , Cell Movement , Morphogenesis , Social Behavior
9.
Nat Commun ; 12(1): 791, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33542237

ABSTRACT

Cells migrate collectively to form tissues and organs during morphogenesis. Contact inhibition of locomotion (CIL) drives collective migration by inhibiting lamellipodial protrusions at cell-cell contacts and promoting polarization at the leading edge. Here, we report a CIL-related collective cell behavior of myotubes that lack lamellipodial protrusions, but instead use filopodia to move as a cohesive cluster in a formin-dependent manner. We perform genetic, pharmacological and mechanical perturbation analyses to reveal the essential roles of Rac2, Cdc42 and Rho1 in myotube migration. These factors differentially control protrusion dynamics and cell-matrix adhesion formation. We also show that active Rho1 GTPase localizes at retracting free edge filopodia and that Rok-dependent actomyosin contractility does not mediate a contraction of protrusions at cell-cell contacts, but likely plays an important role in the constriction of supracellular actin cables. Based on these findings, we propose that contact-dependent asymmetry of cell-matrix adhesion drives directional movement, whereas contractile actin cables contribute to the integrity of the migrating cell cluster.


Subject(s)
Cell Movement/physiology , Morphogenesis/physiology , Muscle Fibers, Skeletal/physiology , Pseudopodia/metabolism , Actin Cytoskeleton/metabolism , Actomyosin/metabolism , Animals , Cadherins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster , GTP-Binding Proteins/metabolism , Intravital Microscopy , rac GTP-Binding Proteins/metabolism , rho GTP-Binding Proteins/metabolism , RAC2 GTP-Binding Protein
10.
Development ; 146(7)2019 04 04.
Article in English | MEDLINE | ID: mdl-30910826

ABSTRACT

The formation of neuronal dendrite branches is fundamental for the wiring and function of the nervous system. Indeed, dendrite branching enhances the coverage of the neuron's receptive field and modulates the initial processing of incoming stimuli. Complex dendrite patterns are achieved in vivo through a dynamic process of de novo branch formation, branch extension and retraction. The first step towards branch formation is the generation of a dynamic filopodium-like branchlet. The mechanisms underlying the initiation of dendrite branchlets are therefore crucial to the shaping of dendrites. Through in vivo time-lapse imaging of the subcellular localization of actin during the process of branching of Drosophila larva sensory neurons, combined with genetic analysis and electron tomography, we have identified the Actin-related protein (Arp) 2/3 complex as the major actin nucleator involved in the initiation of dendrite branchlet formation, under the control of the activator WAVE and of the small GTPase Rac1. Transient recruitment of an Arp2/3 component marks the site of branchlet initiation in vivo These data position the activation of Arp2/3 as an early hub for the initiation of branchlet formation.


Subject(s)
Actin-Related Protein 2-3 Complex/metabolism , Dendrites/metabolism , Actin Cytoskeleton/metabolism , Actin-Related Protein 2-3 Complex/genetics , Actins/metabolism , Animals , Drosophila , Drosophila melanogaster , Sensory Receptor Cells/metabolism
11.
J Biol Chem ; 294(18): 7202-7218, 2019 05 03.
Article in English | MEDLINE | ID: mdl-30737279

ABSTRACT

Whereas myosin 18B (Myo18B) is known to be a critical sarcomeric protein, the function of myosin 18A (Myo18A) is unclear, although it has been implicated in cell motility and Golgi shape. Here, we show that homozygous deletion (homozygous tm1a, tm1b, or tm1d alleles) of Myo18a in mouse is embryonic lethal. Reminiscent of Myo18b, Myo18a was highly expressed in the embryo heart, and cardiac-restricted Myo18a deletion in mice was embryonic lethal. Surprisingly, using Western blot analysis, we were unable to detect the known isoforms of Myo18A, Myo18Aα and Myo18Aß, in mouse heart using a custom C-terminal antibody. However, alternative anti-Myo18A antibodies detected a larger than expected protein, and RNA-Seq analysis indicated that a novel Myo18A transcript is expressed in mouse ventricular myocytes (and human heart). Cloning and sequencing revealed that this cardiac isoform, denoted Myo18Aγ, lacks the PDZ-containing N terminus of Myo18Aα but includes an alternative N-terminal extension and a long serine-rich C terminus. EGFP-tagged Myo18Aγ expressed in ventricular myocytes localized to the level of A-bands in sarcomeres, and Myo18a knockout embryos at day 10.5 exhibited disorganized sarcomeres with wavy thick filaments. We additionally generated myeloid-restricted Myo18a knockout mice to investigate the role of Myo18A in nonmuscle cells, exemplified by macrophages, which express more Myo18Aß than Myo18Aα, but no defects in cell shape, motility, or Golgi shape were detected. In summary, we have identified a previously unrecognized sarcomere component, a large novel isoform (denoted Myo18Aγ) of Myo18A. Thus, both members of class XVIII myosins are critical components of cardiac sarcomeres.


Subject(s)
Myocardium/metabolism , Myosins/metabolism , Sarcomeres/metabolism , Animals , Gene Deletion , Genes, Lethal , Green Fluorescent Proteins/metabolism , Humans , Mice , Mice, Knockout , Myosins/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism
12.
Methods Mol Biol ; 1749: 227-238, 2018.
Article in English | MEDLINE | ID: mdl-29526001

ABSTRACT

The most abundant immune cells in Drosophila are macrophage-like plasmatocytes that fulfill central roles in morphogenesis, immune and tissue damage response. The various genetic tools available in Drosophila together with high-resolution and live-imaging microscopy techniques make Drosophila macrophages an excellent model system that combines many advantages of cultured cells with in vivo genetics. Here, we describe the isolation and staining of macrophages from larvae for ex vivo structured illumination microscopy (SIM), the preparation of white prepupae for in vivo 2D random cell migration analysis, and the preparation of pupae (18 h after puparium formation, APF) for in vivo 3D directed cell migration analysis upon wounding using spinning disk microscopy.


Subject(s)
Cell Movement/physiology , Cell Shape/physiology , Macrophages/cytology , Microscopy/methods , Animals , Cells, Cultured , Drosophila , Larva/cytology , Microscopy, Fluorescence , Pupa/cytology
13.
J Cell Sci ; 130(20): 3427-3435, 2017 Oct 15.
Article in English | MEDLINE | ID: mdl-29032357

ABSTRACT

The actin cytoskeleton and associated motor proteins provide the driving forces for establishing the astonishing morphological diversity and dynamics of mammalian cells. Aside from functions in protruding and contracting cell membranes for motility, differentiation or cell division, the actin cytoskeleton provides forces to shape and move intracellular membranes of organelles and vesicles. To establish the many different actin assembly functions required in time and space, actin nucleators are targeted to specific subcellular compartments, thereby restricting the generation of specific actin filament structures to those sites. Recent research has revealed that targeting and activation of actin filament nucleators, elongators and myosin motors are tightly coordinated by conserved protein complexes to orchestrate force generation. In this Cell Science at a Glance article and the accompanying poster, we summarize and discuss the current knowledge on the corresponding protein complexes and their modes of action in actin nucleation, elongation and force generation.


Subject(s)
Actin Cytoskeleton/physiology , Pseudopodia/physiology , Actin Cytoskeleton/ultrastructure , Actins/physiology , Actins/ultrastructure , Animals , Cell Physiological Phenomena , Cells, Cultured , Humans , Protein Multimerization , Pseudopodia/ultrastructure
14.
Article in English | MEDLINE | ID: mdl-28096264

ABSTRACT

Cadherin-based adherens junctions are conserved structures that mediate epithelial cell-cell adhesion in invertebrates and vertebrates. Despite their pivotal function in epithelial integrity, adherens junctions show a remarkable plasticity that is a prerequisite for tissue architecture and morphogenesis. Epithelial cadherin (E-cadherin) is continuously turned over and undergoes cycles of endocytosis, sorting and recycling back to the plasma membrane. Mammalian cell culture and genetically tractable model systems such as Drosophila have revealed conserved, but also distinct, mechanisms in the regulation of E-cadherin membrane trafficking. Here, we discuss our current knowledge about molecules and mechanisms controlling endocytosis, sorting and recycling of E-cadherin during junctional remodeling.


Subject(s)
Adherens Junctions/physiology , Cadherins/metabolism , Animals , Biological Transport , Catenins/physiology , Endocytosis , Endosomes/metabolism , Protein Processing, Post-Translational
15.
J Cell Sci ; 130(2): 344-359, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27884932

ABSTRACT

The Wiskott-Aldrich syndrome protein and SCAR homolog (WASH; also known as Washout in flies) is a conserved actin-nucleation-promoting factor controlling Arp2/3 complex activity in endosomal sorting and recycling. Previous studies have identified WASH as an essential regulator in Drosophila development. Here, we show that homozygous wash mutant flies are viable and fertile. We demonstrate that Drosophila WASH has conserved functions in integrin receptor recycling and lysosome neutralization. WASH generates actin patches on endosomes and lysosomes, thereby mediating both aforementioned functions. Consistently, loss of WASH function results in cell spreading and cell migration defects of macrophages, and an increased lysosomal acidification that affects efficient phagocytic and autophagic clearance. WASH physically interacts with the vacuolar (V)-ATPase subunit Vha55 that is crucial to establish and maintain lysosome acidification. As a consequence, starved flies that lack WASH function show a dramatic increase in acidic autolysosomes, causing a reduced lifespan. Thus, our data highlight a conserved role for WASH in the endocytic sorting and recycling of membrane proteins, such as integrins and the V-ATPase, that increase the likelihood of survival under nutrient deprivation.


Subject(s)
Cell Movement , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Integrin alpha Chains/metabolism , Lysosomes/metabolism , Vesicular Transport Proteins/metabolism , Acids/metabolism , Actins/metabolism , Animals , Autophagy , Cell Adhesion , Endosomes/metabolism , Fertility , Homozygote , Macrophages/cytology , Macrophages/metabolism , Mutation/genetics , Oogenesis , Phagocytosis , Phagosomes/metabolism , Protein Binding , Protein Subunits/metabolism , Pupa/cytology , Vacuolar Proton-Translocating ATPases/metabolism
16.
Handb Exp Pharmacol ; 235: 285-310, 2017.
Article in English | MEDLINE | ID: mdl-27757759

ABSTRACT

The actin cytoskeleton provides mechanical support for cells and generates forces to drive cell shape changes and cell migration in morphogenesis. Molecular understanding of actin dynamics requires a genetically traceable model system that allows interdisciplinary experimental approaches to elucidate the regulatory network of cytoskeletal proteins in vivo. Here, we will discuss some examples of how advances in Drosophila genetics and high-resolution imaging techniques contribute to the discovery of new actin functions, signaling pathways, and mechanisms of actin regulation in vivo.


Subject(s)
Actin Cytoskeleton/physiology , Drosophila/physiology , Animals , Cell Movement , Humans , Janus Kinases/physiology , Macrophages/physiology , STAT Transcription Factors/physiology , Signal Transduction , Wiskott-Aldrich Syndrome Protein Family/physiology
18.
J Cell Biol ; 212(5): 591-603, 2016 Feb 29.
Article in English | MEDLINE | ID: mdl-26903538

ABSTRACT

Directional cell movements during morphogenesis require the coordinated interplay between membrane receptors and the actin cytoskeleton. The WAVE regulatory complex (WRC) is a conserved actin regulator. Here, we found that the atypical cadherin Fat2 recruits the WRC to basal membranes of tricellular contacts where a new type of planar-polarized whip-like actin protrusion is formed. Loss of either Fat2 function or its interaction with the WRC disrupts tricellular protrusions and results in the formation of nonpolarized filopodia. We provide further evidence for a molecular network in which the receptor tyrosine phosphatase Dlar interacts with the WRC to couple the extracellular matrix, the membrane, and the actin cytoskeleton during egg elongation. Our data uncover a mechanism by which polarity information can be transduced from a membrane receptor to a key actin regulator to control collective follicle cell migration during egg elongation. 4D-live imaging of rotating MCF10A mammary acini further suggests an evolutionary conserved mechanism driving rotational motions in epithelial morphogenesis.


Subject(s)
Cadherins/metabolism , Cell Movement , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Multiprotein Complexes/metabolism , Actins/metabolism , Animals , Drosophila melanogaster/embryology , Extracellular Matrix/metabolism , Rotation
19.
J Cell Sci ; 129(3): 604-20, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26675239

ABSTRACT

Wiskott-Aldrich syndrome proteins (WASPs) are nucleation-promoting factors (NPF) that differentially control the Arp2/3 complex. In Drosophila, three different family members, SCAR (also known as WAVE), WASP and WASH (also known as CG13176), have been analyzed so far. Here, we characterized WHAMY, the fourth Drosophila WASP family member. whamy originated from a wasp gene duplication and underwent a sub-neofunctionalization. Unlike WASP, we found that WHAMY specifically interacted with activated Rac1 through its two CRIB domains, which were sufficient for targeting WHAMY to lamellipodial and filopodial tips. Biochemical analyses showed that WHAMY promoted exceptionally fast actin filament elongation, although it did not activate the Arp2/3 complex. Loss- and gain-of-function studies revealed an important function of WHAMY in membrane protrusions and cell migration in macrophages. Genetic data further implied synergistic functions between WHAMY and WASP during morphogenesis. Double mutants were late-embryonic lethal and showed severe defects in myoblast fusion. Trans-heterozygous mutant animals showed strongly increased defects in sensory cell fate specification. Thus, WHAMY is a novel actin polymerase with an initial partitioning of ancestral WASP functions in development and subsequent acquisition of a new function in cell motility during evolution.


Subject(s)
Actins/metabolism , Cell Movement/physiology , Drosophila Proteins/metabolism , Drosophila/metabolism , Macrophages/metabolism , Microfilament Proteins/metabolism , Myoblasts/metabolism , Organogenesis/physiology , Actin Cytoskeleton/metabolism , Animals , Drosophila/physiology , Morphogenesis/physiology , Muscle Development/physiology , Wiskott-Aldrich Syndrome Protein/metabolism
20.
Histochem Cell Biol ; 144(6): 517-32, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26275669

ABSTRACT

Endothelial junctions are dynamic structures organized by multi-protein complexes that control monolayer integrity, homeostasis, inflammation, cell migration and angiogenesis. Newly developed methods for both the genetic manipulation of endothelium and microscopy permit time-lapse recordings of fluorescent proteins over long periods of time. Quantitative data analyses require automated methods. We developed a software package, the CellBorderTracker, allowing quantitative analysis of fluorescent-tagged cell junction protein dynamics in time-lapse sequences. The CellBorderTracker consists of the CellBorderExtractor that segments cells and identifies cell boundaries and mapping tools for data extraction. The tool is illustrated by analyzing fluorescent-tagged VE-cadherin the backbone of adherence junctions in endothelium. VE-cadherin displays high dynamics that is forced by junction-associated intermittent lamellipodia (JAIL) that are actin driven and WASP/ARP2/3 complex controlled. The manual segmentation and the automatic one agree to 90 %, a value that indicates high reliability. Based on segmentations, different maps were generated allowing more detailed data extraction. This includes the quantification of protein distribution pattern, the generation of regions of interest, junction displacements, cell shape changes, migration velocities and the visualization of junction dynamics over many hours. Furthermore, we demonstrate an advanced kymograph, the J-kymograph that steadily follows irregular cell junction dynamics in time-lapse sequences for individual junctions at the subcellular level. By using the CellBorderTracker, we demonstrate that VE-cadherin dynamics is quickly arrested upon thrombin stimulation, a phenomenon that was largely due to transient inhibition of JAIL and display a very heterogeneous subcellular and divers VE-cadherin dynamics during intercellular gap formation and resealing.


Subject(s)
Cadherins/analysis , Endothelium, Vascular/cytology , Intercellular Junctions/metabolism , Software , Animals , Cadherins/metabolism , Cells, Cultured , Drosophila , Endothelium, Vascular/metabolism , Fluorescence , Fluorescent Antibody Technique , Humans , Intercellular Junctions/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...