Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Curr Biol ; 34(3): 615-622.e4, 2024 02 05.
Article in English | MEDLINE | ID: mdl-38199065

ABSTRACT

Convergent extension (CE) requires the coordinated action of the planar cell polarity (PCP) proteins1,2 and the actin cytoskeleton,3,4,5,6 but this relationship remains incompletely understood. For example, PCP signaling orients actomyosin contractions, yet actomyosin is also required for the polarized localization of PCP proteins.7,8 Moreover, the actin-regulating Septins play key roles in actin organization9 and are implicated in PCP and CE in frogs, mice, and fish5,6,10,11,12 but execute only a subset of PCP-dependent cell behaviors. Septin loss recapitulates the severe tissue-level CE defects seen after core PCP disruption yet leaves overt cell polarity intact.5 Together, these results highlight the general fact that cell movement requires coordinated action by distinct but integrated actin populations, such as lamella and lamellipodia in migrating cells13 or medial and junctional actin populations in cells engaged in apical constriction.14,15 In the context of Xenopus mesoderm CE, three such actin populations are important, a superficial meshwork known as the "node-and-cable" system,4,16,17,18 a contractile network at deep cell-cell junctions,6,19 and mediolaterally oriented actin-rich protrusions, which are present both superficially and deeply.4,19,20,21 Here, we exploited the amenability of the uniquely "two-dimensional" node and cable system to probe the relationship between PCP proteins, Septins, and the polarization of this actin network. We find that the PCP proteins Vangl2 and Prickle2 and Septins co-localize at nodes, and that the node and cable system displays a cryptic, PCP- and Septin-dependent anteroposterior (AP) polarity in its organization and dynamics.


Subject(s)
Actins , Septins , Mice , Animals , Septins/metabolism , Actins/metabolism , Actomyosin/metabolism , Actin Cytoskeleton/metabolism , Cell Movement/physiology , Cell Polarity/physiology , Membrane Proteins/metabolism , LIM Domain Proteins/metabolism
2.
bioRxiv ; 2023 Nov 08.
Article in English | MEDLINE | ID: mdl-37986924

ABSTRACT

Understanding biomechanics of biological systems is crucial for unraveling complex processes like tissue morphogenesis. However, current methods for studying cellular mechanics in vivo are limited by the need for specialized equipment and often provide limited spatiotemporal resolution. Here we introduce two new techniques, Tension by Transverse Fluctuation (TFlux) and in vivo microrheology, that overcome these limitations. They both offer time-resolved, subcellular biomechanical analysis using only fluorescent reporters and widely available microscopes. Employing these two techniques, we have revealed a planar cell polarity (PCP)-dependent mechanical gradient both in the cell cortex and the cytoplasm of individual cells engaged in convergent extension. Importantly, the non-invasive nature of these methods holds great promise for its application for uncovering subcellular mechanical variations across a wide array of biological contexts. Summary: Non-invasive imaging-based techniques providing time-resolved biomechanical analysis at subcellular scales in developing vertebrate embryos.

3.
J Cell Sci ; 134(14)2021 07 15.
Article in English | MEDLINE | ID: mdl-34060614

ABSTRACT

The dynamic control of the actin cytoskeleton is a key aspect of essentially all animal cell movements. Experiments in single migrating cells and in vitro systems have provided an exceptionally deep understanding of actin dynamics. However, we still know relatively little of how these systems are tuned in cell-type-specific ways, for example in the context of collective cell movements that sculpt the early embryo. Here, we provide an analysis of the actin-severing and depolymerization machinery during vertebrate gastrulation, with a focus on Twinfilin1 (Twf1) in Xenopus. We find that Twf1 is essential for convergent extension, and loss of Twf1 results in a disruption of lamellipodial dynamics and polarity. Moreover, Twf1 loss results in a failure to assemble polarized cytoplasmic actin cables, which are essential for convergent extension. These data provide an in vivo complement to our more-extensive understanding of Twf1 action in vitro and provide new links between the core machinery of actin regulation and the specialized cell behaviors of embryonic morphogenesis.


Subject(s)
Actins , Gastrulation , Actin Cytoskeleton , Actins/genetics , Animals , Pseudopodia , Xenopus laevis
4.
Elife ; 92020 12 02.
Article in English | MEDLINE | ID: mdl-33263282

ABSTRACT

Ciliary motility is driven by axonemal dyneins that are assembled in the cytoplasm before deployment to cilia. Motile ciliopathy can result from defects in the dyneins themselves or from defects in factors required for their cytoplasmic pre-assembly. Recent work demonstrates that axonemal dyneins, their specific assembly factors, and broadly-acting chaperones are concentrated in liquid-like organelles in the cytoplasm called DynAPs (Dynein Axonemal Particles). Here, we use in vivo imaging in Xenopus to show that inner dynein arm (IDA) and outer dynein arm (ODA) subunits are partitioned into non-overlapping sub-regions within DynAPs. Using affinity- purification mass-spectrometry of in vivo interaction partners, we also identify novel partners for inner and outer dynein arms. Among these, we identify C16orf71/Daap1 as a novel axonemal dynein regulator. Daap1 interacts with ODA subunits, localizes specifically to the cytoplasm, is enriched in DynAPs, and is required for the deployment of ODAs to axonemes. Our work reveals a new complexity in the structure and function of a cell-type specific liquid-like organelle that is directly relevant to human genetic disease.


Subject(s)
Axonemal Dyneins/metabolism , Organelles/metabolism , Animals , Cilia/metabolism , Cytoplasm/metabolism , Immunoprecipitation , Mass Spectrometry , Tandem Affinity Purification , Xenopus laevis/embryology
5.
Dev Biol ; 467(1-2): 108-117, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32898505

ABSTRACT

Cell-type specific RNA-associated proteins are essential for development and homeostasis in animals. Despite a massive recent effort to systematically identify RNA-associated proteins, we currently have few comprehensive rosters of cell-type specific RNA-associated proteins in vertebrate tissues. Here, we demonstrate the feasibility of determining the RNA-associated proteome of a defined vertebrate embryonic tissue using DIF-FRAC, a systematic and universal (i.e., label-free) method. Application of DIF-FRAC to cultured tissue explants of Xenopus mucociliary epithelium identified dozens of known RNA-associated proteins as expected, but also several novel RNA-associated proteins, including proteins related to assembly of the mitotic spindle and regulation of ciliary beating. In particular, we show that the inner dynein arm tether Cfap44 is an RNA-associated protein that localizes not only to axonemes, but also to liquid-like organelles in the cytoplasm called DynAPs. This result led us to discover that DynAPs are generally enriched for RNA. Together, these data provide a useful resource for a deeper understanding of mucociliary epithelia and demonstrate that DIF-FRAC will be broadly applicable for systematic identification of RNA-associated proteins from embryonic tissues.


Subject(s)
Cilia/metabolism , Embryo, Nonmammalian/metabolism , RNA-Binding Proteins/metabolism , Xenopus Proteins/metabolism , Animals , Epithelium/embryology , Tissue Culture Techniques , Xenopus
6.
Mol Biol Rep ; 46(6): 6243-6252, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31583563

ABSTRACT

CDKN2A is an evolutionarily conserved gene encoding proteins implicated in tumor suppression, ocular development, aging, and metabolic diseases. Like the human form, mouse Cdkn2a encodes two distinct proteins-p16Ink4a, which blocks cyclin-dependent kinase activity, and p19Arf, which is best known as a positive regulator of the p53 tumor suppressor-and their functions have been well-studied in genetically engineered mouse models. Relatively little is known about how expression of the two transcripts is controlled in normal development and in certain disease states. To better understand their coordinate and transcript-specific expression in situ, we used a transposase-aided approach to generate a new BAC transgenic mouse model in which the first exons encoding Arf and Ink4a are replaced by fluorescent reporters. We show that mouse embryo fibroblasts generated from the transgenic lines faithfully display induction of each transgenic reporter in cell culture models, and we demonstrate the expected expression of the Arf reporter in the normal testis, one of the few places where that promoter is normally expressed. Interestingly, the TGFß-2-dependent induction of the Arf reporter in the eye-a process essential for normal eye development-does not occur. Our findings illustrate the value of BAC transgenesis in mapping key regulatory elements in the mouse by revealing the genomic DNA required for Cdkn2a induction in cultured cells and the developing testis, and the apparent lack of elements driving expression in the developing eye.


Subject(s)
ADP-Ribosylation Factors/genetics , Gene Expression Regulation , Promoter Regions, Genetic , Testis/metabolism , Transposases/metabolism , ADP-Ribosylation Factors/metabolism , Animals , Cell Line , Chromosomes, Artificial, Bacterial , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Disease Models, Animal , Humans , Male , Mice , Mice, Transgenic
7.
Exp Eye Res ; 145: 224-229, 2016 04.
Article in English | MEDLINE | ID: mdl-26778750

ABSTRACT

Arf encodes an important tumor suppressor, p19(Arf), which also plays a critical role to control hyperplasia in the primary vitreous during mouse eye development. In the absence of Arf, mice are born blind and display a phenotype closely mimicking severe forms of the human eye disease, persistent hyperplastic primary vitreous (PHPV). In this report, we characterize p19(Arf) expression in perivascular cells that normally populate the primary vitreous and express the Arf promoter. Using a new ex vivo model, we show that these cells respond to exogenous Tgfß, despite being isolated at a time when Tgfß has already turned on the Arf promoter. Treatment of the cells with PDGF-B ligand doubles the population of cells in S-phase and ectopic expression of Arf blunts that effect. We show this effect is mediated through Pdgfrß as expression of Arf represses expression of Pdgfrß mRNA and protein to approximately 60%. p53 is not required for Arf-dependent blockade of PDGF-B driven proliferation and repression of Pdgfrß protein as ectopic expression of Arf is still able to inhibit the 2-fold increase in the S-phase fraction of cells upon treatment with PDGF-B. Finally, induction of mature miR-34a, a microRNA previously identified to be regulated by p19(Arf) does not depend on p53 while the expression of the primary transcript does require p53. These data corroborate that, as in vivo, p19(Arf) functions to inhibit PDGF-B driven proliferation ex vivo.


Subject(s)
Cell Proliferation/physiology , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Proto-Oncogene Proteins c-sis/physiology , Retinal Diseases/physiopathology , Vitreous Body/cytology , Animals , Blotting, Western , Cell Cycle/physiology , Cells, Cultured , Mice , Receptor, Platelet-Derived Growth Factor beta/pharmacology , Tumor Suppressor Protein p53 , Vitreous Body/drug effects
8.
Biotechniques ; 56(5): 239-49, 2014 May.
Article in English | MEDLINE | ID: mdl-24806224

ABSTRACT

Although many researchers have successfully uncovered novel functions of the tumor suppressor p19(Arf) utilizing various types of cultured cancer cells and immortalized fibroblasts, these systems do not accurately reflect the endogenous environment in which Arf is developmentally expressed. We addressed this by isolating perivascular cells (PVCs) from the primary vitreous of the mouse eye. This rare cell type normally expresses the p19(Arf) tumor suppressor in a non-pathological, developmental context. We utilized fluorescence activated cell sorting (FACS) to purify the cells by virtue of a GFP reporter driven by the native Arf promoter and then characterized their morphology and gene expression pattern. We further examined the effects of reintroduction of Arf expression in the Arf(GFP/GFP) PVCs to verify expected downstream effectors of p19(Arf) as well as uncover novel functions of Arf as a regulator of vasculogenesis. This methodology and cell culture model should serve as a useful tool to examine p19(Arf) biology.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/genetics , Eye/growth & development , Flow Cytometry/methods , Gene Expression Regulation, Developmental , Vitreous Body/cytology , Animals , Cell Culture Techniques , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Green Fluorescent Proteins/genetics , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic , Vitreous Body/growth & development
9.
PLoS One ; 8(8): e70371, 2013.
Article in English | MEDLINE | ID: mdl-23940569

ABSTRACT

Recent studies show that Arf, a bona fide tumor suppressor, also plays an essential role during mouse eye development. Tgfß is required for Arf promoter activation in developing mouse eyes, and its capacity to induce Arf depends on Smads 2/3 as well as p38 Mapk. Substantial delay between activation of these pathways and increased Arf transcription imply that changes in the binding of additional transcription factors help orchestrate changes in Arf expression. Focusing on proteins with putative DNA binding elements near the mouse Arf transcription start, we now show that Tgfß induction of this gene correlated with decreased expression and DNA binding of C/ebpß to the proximal Arf promoter. Ectopic expression of C/ebpß in mouse embryo fibroblasts (MEFs) blocked Arf induction by Tgfß. Although basal levels of Arf mRNA were increased by C/ebpß loss in MEFs and in the developing eye, Tgfß was still able to increase Arf, indicating that derepression was not the sole factor. Chromatin immunoprecipitation (ChIP) assay showed increased Sp1 binding to the Arf promotor at 24 and 48 hours after Tgfß treatment, at which time points Arf expression was significantly induced by Tgfß. Chemical inhibition of Sp1 and its knockdown by RNA interference blocked Arf induction by Tgfß in MEFs. In summary, our results indicate that C/ebpß and Sp1 are negative and positive Arf regulators that are influenced by Tgfß.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Sp1 Transcription Factor/metabolism , ADP-Ribosylation Factors/genetics , ADP-Ribosylation Factors/metabolism , Animals , Blotting, Western , CCAAT-Enhancer-Binding Protein-beta/genetics , Chromatin Immunoprecipitation , Laser Capture Microdissection , Mice , Mice, Mutant Strains , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sp1 Transcription Factor/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
10.
Dev Biol ; 380(1): 49-57, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23665474

ABSTRACT

The Arf tumor suppressor represents one of several genes encoded at the Cdkn2a and Cdkn2b loci in the mouse. Beyond its role blunting the growth of incipient cancer cells, the Arf gene also plays an essential role in development: its gene product, p19(Arf), is induced by Tgfß2 in the developing eye to dampen proliferative signals from Pdgfrß, which effect ultimately fosters the vascular remodeling required for normal vision in the mouse. Mechanisms underlying Arf induction by Tgfß2 are not fully understood. Using the chr4(Δ70 kb/Δ70 kb) mouse, we now show that deletion of the coronary artery disease (CAD) risk interval lying upstream of the Cdkn2a/b locus represses developmentally-timed induction of Arf resulting in eye disease mimicking the persistent hyperplastic primary vitreous (PHPV) found in Arf-null mice and in children. Using mouse embryo fibroblasts, we demonstrate that Arf induction by Tgfß is blocked in cis to the 70 kb deletion, but Arf induction by activated RAS and cell culture "shock" is not. Finally, we show that Arf induction by Tgfß is derailed by preventing RNA polymerase II recruitment following Smad 2/3 binding to the promoter. These findings provide the first evidence that the CAD risk interval, located at a distance from Arf, acts as a cis enhancer of Tgfß2-driven induction of Arf during development.


Subject(s)
ADP-Ribosylation Factor 1/metabolism , Enhancer Elements, Genetic , Eye/embryology , Gene Expression Regulation, Developmental , Transforming Growth Factor beta/metabolism , Animals , Coronary Artery Disease/genetics , DNA, Intergenic/metabolism , Disease Models, Animal , Eye Diseases/genetics , Fibroblasts/metabolism , Gene Deletion , Mice , Mice, Transgenic , Persistent Hyperplastic Primary Vitreous/genetics , Phenotype , Receptor, Platelet-Derived Growth Factor beta/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...