Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Biol Open ; 10(2)2021 02 03.
Article in English | MEDLINE | ID: mdl-33504488

ABSTRACT

Regulation of cell architecture is critical in the formation of tissues during animal development. The mechanisms that control cell shape must be both dynamic and stable in order to establish and maintain the correct cellular organization. Previous work has identified Shroom family proteins as essential regulators of cell morphology during vertebrate development. Shroom proteins regulate cell architecture by directing the subcellular distribution and activation of Rho-kinase, which results in the localized activation of non-muscle myosin II. Because the Shroom-Rock-myosin II module is conserved in most animal model systems, we have utilized Drosophila melanogaster to further investigate the pathways and components that are required for Shroom to define cell shape and tissue architecture. Using a phenotype-based heterozygous F1 genetic screen for modifiers of Shroom activity, we identified several cytoskeletal and signaling protein that may cooperate with Shroom. We show that two of these proteins, Enabled and Short stop, are required for ShroomA-induced changes in tissue morphology and are apically enriched in response to Shroom expression. While the recruitment of Ena is necessary, it is not sufficient to redefine cell morphology. Additionally, this requirement for Ena appears to be context dependent, as a variant of Shroom that is apically localized, binds to Rock, but lacks the Ena binding site, is still capable of inducing changes in tissue architecture. These data point to important cellular pathways that may regulate contractility or facilitate Shroom-mediated changes in cell and tissue morphology.


Subject(s)
Biomarkers , Cytoskeletal Proteins/metabolism , Cytoskeleton/metabolism , Drosophila Proteins/metabolism , Morphogenesis , Animals , Cell Shape/genetics , Cytoskeletal Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/physiology , Gene Expression Regulation, Developmental , Morphogenesis/genetics , Organ Specificity/genetics , Organogenesis , Phenotype , Signal Transduction
2.
Nat Commun ; 8: 14536, 2017 03 06.
Article in English | MEDLINE | ID: mdl-28262662

ABSTRACT

Shrm4, a protein expressed only in polarized tissues, is encoded by the KIAA1202 gene, whose mutations have been linked to epilepsy and intellectual disability. However, a physiological role for Shrm4 in the brain is yet to be established. Here, we report that Shrm4 is localized to synapses where it regulates dendritic spine morphology and interacts with the C terminus of GABAB receptors (GABABRs) to control their cell surface expression and intracellular trafficking via a dynein-dependent mechanism. Knockdown of Shrm4 in rat severely impairs GABABR activity causing increased anxiety-like behaviour and susceptibility to seizures. Moreover, Shrm4 influences hippocampal excitability by modulating tonic inhibition in dentate gyrus granule cells, in a process involving crosstalk between GABABRs and extrasynaptic δ-subunit-containing GABAARs. Our data highlights a role for Shrm4 in synaptogenesis and in maintaining GABABR-mediated inhibition, perturbation of which may be responsible for the involvement of Shrm4 in cognitive disorders and epilepsy.


Subject(s)
Hippocampus/metabolism , Microfilament Proteins/genetics , Nerve Tissue Proteins/genetics , Neurons/metabolism , Receptors, GABA-A/genetics , Receptors, GABA-B/genetics , Synaptic Transmission/genetics , Animals , Dentate Gyrus/metabolism , Dentate Gyrus/pathology , Dentate Gyrus/ultrastructure , Embryo, Mammalian , Epilepsy/genetics , Epilepsy/metabolism , Epilepsy/pathology , Gene Expression Regulation , HEK293 Cells , Hippocampus/pathology , Hippocampus/ultrastructure , Humans , Injections, Intraventricular , Intellectual Disability/genetics , Intellectual Disability/metabolism , Intellectual Disability/pathology , Microfilament Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neural Inhibition , Neurogenesis/genetics , Neurons/pathology , Neurons/ultrastructure , Primary Cell Culture , Rats , Rats, Wistar , Receptor Cross-Talk , Receptors, GABA-A/metabolism , Receptors, GABA-B/metabolism , Synapses/metabolism , Synapses/pathology , Synapses/ultrastructure
3.
J Vis Exp ; (119)2017 01 06.
Article in English | MEDLINE | ID: mdl-28117766

ABSTRACT

Obtaining crystals for structure determination can be a difficult and time consuming proposition for any protein. Coiled-coil proteins and domains are found throughout nature, however, because of their physical properties and tendency to aggregate, they are traditionally viewed as being especially difficult to crystallize. Here, we utilize a variety of quick and simple techniques designed to identify a series of possible domain boundaries for a given coiled-coil protein, and then quickly characterize the behavior of these proteins in solution. With the addition of a strongly fluorescent tag (mRuby2), protein characterization is simple and straightforward. The target protein can be readily visualized under normal lighting and can be quantified with the use of an appropriate imager. The goal is to quickly identify candidates that can be removed from the crystallization pipeline because they are unlikely to succeed, affording more time for the best candidates and fewer funds expended on proteins that do not produce crystals. This process can be iterated to incorporate information gained from initial screening efforts, can be adapted for high-throughput expression and purification procedures, and is augmented by robotic screening for crystallization.


Subject(s)
Proteins/chemistry , Amino Acid Sequence , Computational Biology , Crystallization , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Plasmids/genetics , Plasmids/metabolism , Point Mutation , Protein Domains , Proteins/genetics , Proteins/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Sequence Alignment , User-Computer Interface
4.
J Cell Sci ; 130(1): 243-259, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27802160

ABSTRACT

Epithelia within tubular organs form and expand lumens. Failure of these processes can result in serious developmental anomalies. Although tight junction assembly is crucial to epithelial polarization, the contribution of specific tight junction proteins to lumenogenesis is undefined. Here, we show that ZO-1 (also known as TJP1) is necessary for the formation of single lumens. Epithelia lacking this tight junction scaffolding protein form cysts with multiple lumens and are defective in the earliest phases of polarization, both in two and three dimensions. Expression of ZO-1 domain-deletion mutants demonstrated that the actin-binding region and U5-GuK domain are crucial to single lumen development. For actin-binding region, but not U5-GuK domain, mutants, this could be overcome by strong polarization cues from the extracellular matrix. Analysis of the U5-GuK binding partners shroom2, α-catenin and occludin showed that only occludin deletion led to multi-lumen cysts. Like ZO-1-deficiency, occludin deletion led to mitotic spindle orientation defects. Single lumen formation required the occludin OCEL domain, which binds to ZO-1. We conclude that ZO-1-occludin interactions regulate multiple phases of epithelial polarization by providing cell-intrinsic signals that are required for single lumen formation.


Subject(s)
Actins/metabolism , Cell Culture Techniques/methods , Cell Polarity , Epithelial Cells/cytology , Epithelial Cells/metabolism , Occludin/metabolism , Zonula Occludens-1 Protein/metabolism , Cell Line , Cell Proliferation , Gene Knockdown Techniques , Humans , Mitosis , Morphogenesis , Phenotype , Protein Binding , Protein Transport , Tight Junctions/metabolism , Zonula Occludens-1 Protein/chemistry , alpha Catenin/metabolism
5.
J Biol Chem ; 291(49): 25364-25374, 2016 Dec 02.
Article in English | MEDLINE | ID: mdl-27758857

ABSTRACT

Shroom-mediated remodeling of the actomyosin cytoskeleton is a critical driver of cellular shape and tissue morphology that underlies the development of many tissues including the neural tube, eye, intestines, and vasculature. Shroom uses a conserved SD2 domain to direct the subcellular localization of Rho-associated kinase (Rock), which in turn drives changes in the cytoskeleton and cellular morphology through its ability to phosphorylate and activate non-muscle myosin II. Here, we present the structure of the human Shroom-Rock binding module, revealing an unexpected stoichiometry for Shroom in which two Shroom SD2 domains bind independent surfaces on Rock. Mutation of interfacial residues impaired Shroom-Rock binding in vitro and resulted in altered remodeling of the cytoskeleton and loss of Shroom-mediated changes in cellular morphology. Additionally, we provide the first direct evidence that Shroom can function as a Rock activator. These data provide molecular insight into the Shroom-Rock interface and demonstrate that Shroom directly participates in regulating cytoskeletal dynamics, adding to its known role in Rock localization.


Subject(s)
G-Protein-Coupled Receptor Kinase 1/chemistry , Membrane Proteins/chemistry , Microfilament Proteins/chemistry , Multiprotein Complexes/chemistry , G-Protein-Coupled Receptor Kinase 1/genetics , G-Protein-Coupled Receptor Kinase 1/metabolism , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Myosin Type II/chemistry , Myosin Type II/genetics , Myosin Type II/metabolism , Protein Domains , Protein Structure, Quaternary , Structure-Activity Relationship
6.
Development ; 142(7): 1305-14, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25742799

ABSTRACT

The genetic control of mammalian epithelial polarity and dynamics can be studied in vivo at cellular resolution during morphogenesis of the mouse neural tube. The mouse neural plate is a simple epithelium that is transformed into a columnar pseudostratified tube over the course of ∼ 24 h. Apical F-actin is known to be important for neural tube closure, but the precise roles of actin dynamics in the neural epithelium are not known. To determine how the organization of the neural epithelium and neural tube closure are affected when actin dynamics are blocked, we examined the cellular basis of the neural tube closure defect in mouse mutants that lack the actin-severing protein cofilin 1 (CFL1). Although apical localization of the adherens junctions, the Par complex, the Crumbs complex and SHROOM3 is normal in the mutants, CFL1 has at least two distinct functions in the apical and basal domains of the neural plate. Apically, in the absence of CFL1 myosin light chain does not become phosphorylated, indicating that CFL1 is required for the activation of apical actomyosin required for neural tube closure. On the basal side of the neural plate, loss of CFL1 has the opposite effect on myosin: excess F-actin and myosin accumulate and the ectopic myosin light chain is phosphorylated. The basal accumulation of F-actin is associated with the assembly of ectopic basal tight junctions and focal disruptions of the basement membrane, which eventually lead to a breakdown of epithelial organization.


Subject(s)
Cell Polarity , Cofilin 1/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Morphogenesis , Neural Plate/embryology , Neural Plate/metabolism , Actins/metabolism , Actomyosin/metabolism , Animals , Basement Membrane/metabolism , Biomarkers/metabolism , Cell Count , Cell Shape , Cytoplasm/metabolism , Cytoskeletal Proteins/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Embryo, Mammalian/ultrastructure , Epithelium/embryology , Epithelium/metabolism , Female , Male , Membrane Proteins/metabolism , Mice , Microfilament Proteins/metabolism , Mutation/genetics , Neural Plate/cytology , Neural Plate/ultrastructure , Neural Tube/cytology , Neural Tube/embryology , Neural Tube/metabolism , Phosphorylation , Tight Junction Proteins/metabolism
7.
Biol Open ; 4(2): 186-96, 2015 Jan 16.
Article in English | MEDLINE | ID: mdl-25596276

ABSTRACT

Neural tube closure is a critical developmental event that relies on actomyosin contractility to facilitate specific processes such as apical constriction, tissue bending, and directional cell rearrangements. These complicated processes require the coordinated activities of Rho-Kinase (Rock), to regulate cytoskeletal dynamics and actomyosin contractility, and the Planar Cell Polarity (PCP) pathway, to direct the polarized cellular behaviors that drive convergent extension (CE) movements. Here we investigate the role of Shroom3 as a direct linker between PCP and actomyosin contractility during mouse neural tube morphogenesis. In embryos, simultaneous depletion of Shroom3 and the PCP components Vangl2 or Wnt5a results in an increased liability to NTDs and CE failure. We further show that these pathways intersect at Dishevelled, as Shroom3 and Dishevelled 2 co-distribute and form a physical complex in cells. We observed that multiple components of the Shroom3 pathway are planar polarized along mediolateral cell junctions in the neural plate of E8.5 embryos in a Shroom3 and PCP-dependent manner. Finally, we demonstrate that Shroom3 mutant embryos exhibit defects in planar cell arrangement during neural tube closure, suggesting a role for Shroom3 activity in CE. These findings support a model in which the Shroom3 and PCP pathways interact to control CE and polarized bending of the neural plate and provide a clear illustration of the complex genetic basis of NTDs.

8.
Biol Open ; 3(9): 850-60, 2014 Aug 29.
Article in English | MEDLINE | ID: mdl-25171888

ABSTRACT

Shroom3 is an actin-associated regulator of cell morphology that is required for neural tube closure, formation of the lens placode, and gut morphogenesis in mice and has been linked to chronic kidney disease and directional heart looping in humans. Numerous studies have shown that Shroom3 likely regulates these developmental processes by directly binding to Rho-kinase and facilitating the assembly of apically positioned contractile actomyosin networks. We have characterized the molecular basis for the neural tube defects caused by an ENU-induced mutation that results in an arginine-to-cysteine amino acid substitution at position 1838 of mouse Shroom3. We show that this substitution has no effect on Shroom3 expression or localization but ablates Rock binding and renders Shroom3 non-functional for the ability to regulate cell morphology. Our results indicate that Rock is the major downstream effector of Shroom3 in the process of neural tube morphogenesis. Based on sequence conservation and biochemical analysis, we predict that the Shroom-Rock interaction is highly conserved across animal evolution and represents a signaling module that is utilized in a variety of biological processes.

9.
Development ; 141(16): 3177-87, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25038041

ABSTRACT

Apical constriction (AC) is a widely utilized mechanism of cell shape change whereby epithelial cells transform from a cylindrical to conical shape, which can facilitate morphogenetic movements during embryonic development. Invertebrate epithelial cells undergoing AC depend on the contraction of apical cortex-spanning actomyosin filaments that generate force on the apical junctions and pull them toward the middle of the cell, effectively reducing the apical circumference. A current challenge is to determine whether these mechanisms are conserved in vertebrates and to identify the molecules responsible for linking apical junctions with the AC machinery. Utilizing the developing mouse eye as a model, we have uncovered evidence that lens placode AC may be partially dependent on apically positioned myosin-containing filaments associated with the zonula adherens. In addition we found that, among several junctional components, p120-catenin genetically interacts with Shroom3, a protein required for AC during embryonic morphogenesis. Further analysis revealed that, similar to Shroom3, p120-catenin is required for AC of lens cells. Finally, we determined that p120-catenin functions by recruiting Shroom3 to adherens junctions. Together, these data identify a novel role for p120-catenin during AC and further define the mechanisms required for vertebrate AC.


Subject(s)
Catenins/physiology , Gene Expression Regulation, Developmental , Lens, Crystalline/embryology , Microfilament Proteins/physiology , Actomyosin/metabolism , Adherens Junctions/metabolism , Animals , Cytoskeleton/metabolism , Gene Deletion , Genotype , Mice , Mice, Transgenic , Microfilament Proteins/genetics , Morphogenesis , Nonmuscle Myosin Type IIB/metabolism , Time Factors , Delta Catenin
10.
PLoS One ; 8(12): e81075, 2013.
Article in English | MEDLINE | ID: mdl-24349032

ABSTRACT

Rho-associated coiled coil containing protein kinase (Rho-kinase or Rock) is a well-defined determinant of actin organization and dynamics in most animal cells characterized to date. One of the primary effectors of Rock is non-muscle myosin II. Activation of Rock results in increased contractility of myosin II and subsequent changes in actin architecture and cell morphology. The regulation of Rock is thought to occur via autoinhibition of the kinase domain via intramolecular interactions between the N-terminus and the C-terminus of the kinase. This autoinhibited state can be relieved via proteolytic cleavage, binding of lipids to a Pleckstrin Homology domain near the C-terminus, or binding of GTP-bound RhoA to the central coiled-coil region of Rock. Recent work has identified the Shroom family of proteins as an additional regulator of Rock either at the level of cellular distribution or catalytic activity or both. The Shroom-Rock complex is conserved in most animals and is essential for the formation of the neural tube, eye, and gut in vertebrates. To address the mechanism by which Shroom and Rock interact, we have solved the structure of the coiled-coil region of Rock that binds to Shroom proteins. Consistent with other observations, the Shroom binding domain is a parallel coiled-coil dimer. Using biochemical approaches, we have identified a large patch of residues that contribute to Shrm binding. Their orientation suggests that there may be two independent Shrm binding sites on opposing faces of the coiled-coil region of Rock. Finally, we show that the binding surface is essential for Rock colocalization with Shroom and for Shroom-mediated changes in cell morphology.


Subject(s)
Microfilament Proteins/metabolism , rho-Associated Kinases/metabolism , Fluorescence Polarization , Fluorescent Antibody Technique , Humans , Microfilament Proteins/genetics , Myosin Type II/genetics , Myosin Type II/metabolism , Protein Binding , rho-Associated Kinases/chemistry , rho-Associated Kinases/genetics
11.
Mol Biol Cell ; 23(11): 2131-42, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22493320

ABSTRACT

Shroom (Shrm) proteins are essential regulators of cell shape and tissue morpho-logy during animal development that function by interacting directly with the coiled-coil region of Rho kinase (Rock). The Shrm-Rock interaction is sufficient to direct Rock subcellular localization and the subsequent assembly of contractile actomyosin networks in defined subcellular locales. However, it is unclear how the Shrm-Rock interaction is regulated at the molecular level. To begin investigating this issue, we present the structure of Shrm domain 2 (SD2), which mediates the interaction with Rock and is required for Shrm function. SD2 is a unique three-segmented dimer with internal symmetry, and we identify conserved residues on the surface and within the dimerization interface that are required for the Rock-Shrm interaction and Shrm activity in vivo. We further show that these residues are critical in both vertebrate and invertebrate Shroom proteins, indicating that the Shrm-Rock signaling module has been functionally and molecularly conserved. The structure and biochemical analysis of Shrm SD2 indicate that it is distinct from other Rock activators such as RhoA and establishes a new paradigm for the Rock-mediated assembly of contractile actomyosin networks.


Subject(s)
Cell Polarity , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/metabolism , Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Microfilament Proteins/chemistry , Microfilament Proteins/metabolism , Protein Multimerization , rho-Associated Kinases/metabolism , Animals , Conserved Sequence , Crystallography, X-Ray , Dogs , Drosophila melanogaster , Humans , Mice , Mutation/genetics , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Structure-Activity Relationship
12.
Development ; 138(23): 5177-88, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22031541

ABSTRACT

Epithelial invagination is a common feature of embryogenesis. An example of invagination morphogenesis occurs during development of the early eye when the lens placode forms the lens pit. This morphogenesis is accompanied by a columnar-to-conical cell shape change (apical constriction or AC) and is known to be dependent on the cytoskeletal protein Shroom3. Because Shroom3-induced AC can be Rock1/2 dependent, we hypothesized that during lens invagination, RhoA, Rock and a RhoA guanine nucleotide exchange factor (RhoA-GEF) would also be required. In this study, we show that Rock activity is required for lens pit invagination and that RhoA activity is required for Shroom3-induced AC. We demonstrate that RhoA, when activated and targeted apically, is sufficient to induce AC and that RhoA plays a key role in Shroom3 apical localization. Furthermore, we identify Trio as a RhoA-GEF required for Shroom3-dependent AC in MDCK cells and in the lens pit. Collectively, these data indicate that a Trio-RhoA-Shroom3 pathway is required for AC during lens pit invagination.


Subject(s)
Cell Shape/physiology , Epithelial Cells/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Lens, Crystalline/embryology , Microfilament Proteins/metabolism , Morphogenesis/physiology , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , rho GTP-Binding Proteins/metabolism , Animals , Cell Line , Chick Embryo , Cryoultramicrotomy , Dogs , Electroporation , Fluorescent Antibody Technique , Mice , Regression Analysis , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein
13.
Development ; 138(20): 4423-32, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21880782

ABSTRACT

The cellular mechanisms that drive growth and remodeling of the early intestinal epithelium are poorly understood. Current dogma suggests that the murine fetal intestinal epithelium is stratified, that villi are formed by an epithelial remodeling process involving the de novo formation of apical surface at secondary lumina, and that radial intercalation of the stratified cells constitutes a major intestinal lengthening mechanism. Here, we investigate cell polarity, cell cycle dynamics and cell shape in the fetal murine intestine between E12.5 and E14.5. We show that, contrary to previous assumptions, this epithelium is pseudostratified. Furthermore, epithelial nuclei exhibit interkinetic nuclear migration, a process wherein nuclei move in concert with the cell cycle, from the basal side (where DNA is synthesized) to the apical surface (where mitosis takes place); such nuclear movements were previously misinterpreted as the radial intercalation of cells. We further demonstrate that growth of epithelial girth between E12.5 and E14.5 is driven by microtubule- and actinomyosin-dependent apicobasal elongation, rather than by progressive epithelial stratification as was previously thought. Finally, we show that the actin-binding protein Shroom3 is crucial for the maintenance of the single-layered pseudostratified epithelium. In mice lacking Shroom3, the epithelium is disorganized and temporarily stratified during villus emergence. These results favor an alternative model of intestinal morphogenesis in which the epithelium remains single layered and apicobasally polarized throughout early intestinal development.


Subject(s)
Intestinal Mucosa/embryology , Animals , Cell Cycle , Cell Polarity , Cell Shape , Female , Gene Expression Regulation, Developmental , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Morphogenesis , Pregnancy
14.
Mol Biol Cell ; 22(6): 795-805, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21248203

ABSTRACT

The intrinsic contractile, migratory, and adhesive properties of endothelial cells are central determinants in the formation of vascular networks seen in vertebrate organisms. Because Shroom2 (Shrm2) is expressed within the endothelium, is localized to cortical actin and cell-cell adhesions, and contains a conserved Rho kinase (Rock) binding domain, we hypothesized that Shrm2 may participate in the regulation of endothelial cell behavior during vascular morphogenesis. Consistent with this hypothesis, depletion of Shrm2 results in elevated branching and sprouting angiogenic behavior of endothelial cells. This is recapitulated in human umbilical vein endothelial cells and in a vasculogenesis assay in which differentiated embryonic stem cells depleted for Shrm2 form a more highly branched endothelial network. Further analyses indicate that the altered behavior observed following Shrm2 depletion is due to aberrant cell contractility, as evidenced by decreased stress fiber organization and collagen contraction with an increase in cellular migration. Because Shrm2 directly interacts with Rock, and Shrm2 knockdown results in the loss of Rock and activated myosin II from sites of cell-cell adhesion, we conclude that Shrm2 facilitates the formation of a contractile network within endothelial cells, the loss of which leads to an increase in endothelial sprouting, migration, and angiogenesis.


Subject(s)
Endothelium, Vascular/physiology , Membrane Proteins/metabolism , Morphogenesis/physiology , Muscle Contraction/physiology , Animals , Cell Line , Cell Movement/physiology , Endothelial Cells/cytology , Endothelial Cells/physiology , Endothelium, Vascular/cytology , Humans , Membrane Proteins/genetics , Mice , Microfilament Proteins , Neovascularization, Physiologic , RNA Interference
15.
Am J Physiol Renal Physiol ; 299(5): F1178-84, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20702601

ABSTRACT

Here, we compared the effects of nucleofection and lipid-based approaches to introduce siRNA duplexes on the subsequent development of membrane polarity in kidney cells. Nucleofection of Madin-Darby canine kidney (MDCK) cells, even with control siRNA duplexes, disrupted the initial surface polarity as well as the steady-state distribution of membrane proteins. Transfection using lipofectamine yielded slightly less efficient knockdown but did not disrupt membrane polarity. Polarized secretion was unaffected by nucleofection, suggesting a selective defect in the development of membrane polarity. Cilia frequency and length were not altered by nucleofection. However, the basolateral appearance of a fluorescent lipid tracer added to the apical surface of nucleofected cells was dramatically enhanced relative to untransfected controls or lipofectamine-treated cells. In contrast, [(3)H]inulin diffusion and transepithelial electrical resistance were not altered in nucleofected cells compared with untransfected ones. We conclude that nucleofection selectively hinders development of the tight junction fence function in MDCK cells.


Subject(s)
Cell Polarity/physiology , Epithelial Cells/physiology , Kidney/physiology , Tight Junctions/physiology , Adenoviridae/genetics , Animals , Biotinylation , Cell Line , Cell Membrane/physiology , Cilia/ultrastructure , Dogs , Fluorescent Dyes , Gene Transfer Techniques , Genetic Vectors , Inulin , Kidney/cytology , Lipids , Membrane Potentials/physiology , Microscopy, Fluorescence , RNA, Small Interfering/genetics , Transfection
16.
Dev Dyn ; 239(7): 2078-93, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20549743

ABSTRACT

Vertebrate Shroom proteins define cytoskeletal organization and cellular architecture by binding directly to F-actin and Rho-kinase and spatially regulating the activity of nonmuscle myosin II (myosin II). Here, we report characterization and gain-of-function analysis of Drosophila Shroom. The dShrm locus expresses at least two protein isoforms, dShrmA and dShrmB, which localize to adherens junctions and the apical membrane, respectively. dShrmA and dShrmB exhibit differing abilities to induce apical constriction that are based on their subcellular distribution and the subsequent assembly of spatially and organizationally distinct actomyosin networks that are dependent on the ability to engage Rho-kinase and the activity of myosin II. These data show that the differential subcellular distribution of naturally occurring isoforms of Shroom proteins can define both the position and organization of actomyosin networks in vivo. We further hypothesize that differentially positioned contractile arrays have distinct effects on cellular morphologies and behaviors.


Subject(s)
Cytoskeletal Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila/metabolism , Protein Isoforms/metabolism , Actomyosin/genetics , Actomyosin/metabolism , Adherens Junctions/metabolism , Adherens Junctions/ultrastructure , Animals , Blotting, Western , COS Cells , Cell Line , Chlorocebus aethiops , Cytoskeletal Proteins/genetics , Cytoskeleton/metabolism , Dogs , Drosophila/embryology , Drosophila/ultrastructure , Drosophila Proteins/genetics , Female , Fluorescent Antibody Technique , Male , Microscopy, Electron, Scanning , Myosin Type II/genetics , Myosin Type II/metabolism , Protein Isoforms/genetics , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
17.
Development ; 137(3): 405-15, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20081189

ABSTRACT

Embryonic development requires a complex series of relative cellular movements and shape changes that are generally referred to as morphogenesis. Although some of the mechanisms underlying morphogenesis have been identified, the process is still poorly understood. Here, we address mechanisms of epithelial morphogenesis using the vertebrate lens as a model system. We show that the apical constriction of lens epithelial cells that accompanies invagination of the lens placode is dependent on Shroom3, a molecule previously associated with apical constriction during morphogenesis of the neural plate. We show that Shroom3 is required for the apical localization of F-actin and myosin II, both crucial components of the contractile complexes required for apical constriction, and for the apical localization of Vasp, a Mena family protein with F-actin anti-capping function that is also required for morphogenesis. Finally, we show that the expression of Shroom3 is dependent on the crucial lens-induction transcription factor Pax6. This provides a previously missing link between lens-induction pathways and the morphogenesis machinery and partly explains the absence of lens morphogenesis in Pax6-deficient mutants.


Subject(s)
Eye Proteins/physiology , Homeodomain Proteins/physiology , Lens, Crystalline/embryology , Microfilament Proteins/genetics , Morphogenesis , Paired Box Transcription Factors/physiology , Repressor Proteins/physiology , Actins/physiology , Animals , Cell Adhesion Molecules/physiology , Cell Line , Embryonic Development , Epithelial Cells/physiology , Lens, Crystalline/growth & development , Mice , Mice, Mutant Strains , Microfilament Proteins/physiology , Myosin Type II/physiology , PAX6 Transcription Factor , Phosphoproteins/physiology
18.
Cell Motil Cytoskeleton ; 64(1): 49-63, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17009331

ABSTRACT

All animal cells utilize a specialized set of cytoskeletal proteins to determine their overall shape and the organization of their intracellular compartments and organelles. During embryonic development, the dynamic nature of the actin cytoskeleton is critical for virtually all morphogenic events requiring changes in cell shape, migration, adhesion, and division. The behavior of the actin cytoskeleton is modulated by a myriad of accessory proteins. Shroom3 is an actin binding protein that regulates neural tube morphogenesis by eliciting changes in cell shape through a myosin II-dependent pathway. The Shroom-related gene SHROOM4 (formerly called KIAA1202) has also been implicated in neural development, as mutations in this gene are associated with human X-linked mental retardation. To better understand the function of Shrm4 in embryonic development, we have cloned mouse Shroom4 and characterized its protein product in vivo and in vitro. Shroom4 is expressed in a wide range of cell types during mouse development, including vascular endothelium and the polarized epithelium of the neural tube and kidney. In endothelial cells and embryo fibroblasts, endogenous Shroom4 co-distributes with myosin II to a distinct cytoplasmic population of F-actin and ectopic expression of Shroom4 in multiple cell types enhances or induces the formation of this actin-based structure. This localization is mediated, at least in part, by the direct interaction of Shroom4 and F-actin. Our results suggest that Shroom4 is a regulator of cytoskeletal architecture that may play an important role in vertebrate development.


Subject(s)
Actins/metabolism , Cytoskeleton/metabolism , Microfilament Proteins/metabolism , Actins/ultrastructure , Animals , Brain , Cloning, Molecular , Cytoplasm/metabolism , Cytoskeleton/ultrastructure , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Endothelium, Vascular/metabolism , Epithelium/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Library , Kidney/metabolism , Mice , Microfilament Proteins/genetics , Myosin Type II/metabolism , Rabbits
19.
Development ; 133(20): 4109-18, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16987870

ABSTRACT

Shroom family proteins have been implicated in the control of the actin cytoskeleton, but so far only a single family member has been studied in the context of developing embryos. Here, we show that the Shroom-family protein, Shroom2 (previously known as APXL) is both necessary and sufficient to govern the localization of pigment granules at the apical surface of epithelial cells. In Xenopus embryos that lack Shroom2 function, we observed defects in pigmentation of the eye that stem from failure of melanosomes to mature and to associate with the apical cell surface. Ectopic expression of Shroom2 in naïve epithelial cells facilitates apical pigment accumulation, and this activity specifically requires the Rab27a GTPase. Most interestingly, we find that Shroom2, like Shroom3 (previously called Shroom), is sufficient to induce a dramatic apical accumulation of the microtubule-nucleating protein gamma-tubulin at the apical surfaces of naïve epithelial cells. Together, our data identify Shroom2 as a central regulator of RPE pigmentation, and suggest that, despite their diverse biological roles, Shroom family proteins share a common activity. Finally, because the locus encoding human SHROOM2 lies within the critical region for two distinct forms of ocular albinism, it is possible that SHROOM2 mutations may be a contributing factor in these human visual system disorders.


Subject(s)
Eye/embryology , Melanosomes/genetics , Organogenesis/genetics , Pigment Epithelium of Eye/embryology , Pigmentation/genetics , Xenopus Proteins/physiology , Animals , Epithelial Cells/chemistry , Epithelial Cells/metabolism , Eye/ultrastructure , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Melanosomes/chemistry , Melanosomes/metabolism , Pigment Epithelium of Eye/chemistry , Pigment Epithelium of Eye/ultrastructure , Tubulin/analysis , Tubulin/metabolism , Xenopus/embryology , Xenopus/genetics , Xenopus/metabolism , Xenopus Proteins/analysis , Xenopus Proteins/genetics
20.
J Biol Chem ; 281(29): 20542-54, 2006 Jul 21.
Article in English | MEDLINE | ID: mdl-16684770

ABSTRACT

Shroom is an actin-associated determinant of cell morphology that is required for neural tube closure in both mice and frogs. Shroom regulates this process by causing apical constriction of epithelial cells via a pathway involving myosin II. Here we report on characterization of the Shroom-related proteins Apxl and KIAA1202 and their role in cell architecture. Shroom, Apxl, and KIAA1202 exhibit differing abilities to interact with the actin cytoskeleton. In fibroblasts, Shroom readily associates with actin stress fibers and induces bundling, Apxl is found on cortical actin, and KIAA1202 is localized to a cytoplasmic population of F-actin. In epithelial cells, Apxl and KIAA1202 do not induce apical constriction as Shroom does, but have the capacity to do so if targeted to the apical junctional complex. To determine whether the activity of Shroom-like proteins is conserved in invertebrates, we have tested the ability of the lone Shroomrelated protein in Drosophila, CG8603, to activate the constriction pathway. A chimeric protein consisting of the Shroom targeting domain and the Drosophila protein elicits constriction. Finally, we show that Apxl is involved in regulating the cytoskeletal organization and architecture of endothelial cells. We predict that the ability of Shroom-like proteins to regulate cellular morphology is conserved in evolution and is regulated in part by subcellular localization.


Subject(s)
Evolution, Molecular , Microfilament Proteins/genetics , Actins/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cell Line , Cloning, Molecular , Conserved Sequence , Dogs , Endothelium, Vascular/physiology , Gene Expression Regulation , Invertebrates , Mice , Microfilament Proteins/metabolism , Molecular Sequence Data , Muscle, Skeletal/physiology , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Vertebrates
SELECTION OF CITATIONS
SEARCH DETAIL
...