Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
PLoS One ; 13(11): e0207470, 2018.
Article in English | MEDLINE | ID: mdl-30440051

ABSTRACT

Numerous observations have suggested a connection between the maintenance of cell polarity and control of cell proliferation; however, the mechanisms underlying these connections remain poorly understood. Here we found that ectopic expression of CRB3, which was previously shown to restore tight junctions and membrane polarity in MCF-10A cells, induced a hyperproliferative phenotype, with significantly enlarged acini in basement membrane culture, similar to structures induced by expression of proliferative oncogenes such as cyclinD1. We found that CRB3-induced proliferation is epidermal growth factor (EGF)-independent and occurs through a mechanism that involves secretion of the EGF-family ligand, amphiregulin (AREG). The increase in AREG secretion is associated with an increase in the number and size of both early and late endosomes. Both the proliferative and endocytic phenotypes associated with CRB3 expression require the FERM-binding domain (FBD) but not the PDZ-binding domain of CRB3, arguing that this proliferative phenotype is independent of the PDZ-dependent polarity signaling by CRB3. We identified the FBD-containing protein, EPB41L4B, as an essential mediator of CRB3-driven proliferation and observed that the CRB3-dependent changes in endocytic trafficking were also dependent on EPB41L4B. Taken together, these data reveal a previously uncharacterized role for CRB3 in regulating proliferation in mammalian cells that is associated with changes in the endocytic trafficking machinery.


Subject(s)
Amphiregulin/genetics , Cell Polarity/genetics , Cytoskeletal Proteins/genetics , Membrane Glycoproteins/genetics , Amphiregulin/biosynthesis , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin D1/genetics , Epithelial Cells/metabolism , FERM Domains/genetics , Gene Expression Regulation, Neoplastic , Humans , Mammary Glands, Human/drug effects , Mammary Glands, Human/metabolism , PDZ Domains/genetics , Phenotype , Protein Binding , RNA, Small Interfering/genetics
2.
Article in English | MEDLINE | ID: mdl-28289060

ABSTRACT

The Crumbs proteins are evolutionarily conserved apical transmembrane proteins. Drosophila Crumbs was discovered via its crucial role in epithelial polarity during fly embryogenesis. Crumbs proteins have variable extracellular domains but a highly conserved intracellular domain that can bind FERM and PDZ domain proteins. Mammals have three Crumbs genes and this review focuses on Crumbs3, the major Crumbs isoform expressed in mammalian epithelial cells. Although initial work has highlighted the role of Crumbs3 in polarity, more recent studies have found it has an important role in tissue morphogenesis functioning as a linker between the apical membrane and the actin cytoskeleton. In addition, recent publications have linked Crumbs3 to growth control via regulation of the Hippo/Yap pathway.


Subject(s)
Cell Polarity , Membrane Glycoproteins/physiology , Animals , Cell Proliferation , Cytoskeleton/physiology , Drosophila , Humans , Membrane Glycoproteins/chemistry , Morphogenesis
3.
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
4.
Exp Cell Res ; 328(2): 239, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25193077
5.
EMBO Rep ; 15(4): 428-37, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24591568

ABSTRACT

Apical lumen formation is a key step during epithelial morphogenesis. The establishment of the apical lumen is a complex process that involves coordinated changes in plasma membrane composition, endocytic transport, and cytoskeleton organization. These changes are accomplished, at least in part, by the targeting and fusion of Rab11/FIP5-containing apical endosomes with the apical membrane initiation site (AMIS). Although AMIS formation and polarized transport of Rab11/FIP5-containing endosomes are crucial for the formation of a single apical lumen, the spatiotemporal regulation of this process remains poorly understood. Here, we demonstrate that the formation of the midbody during cytokinesis is a symmetry-breaking event that establishes the location of the AMIS. The interaction of FIP5 with SNX18, which is required for the formation of apical endocytic carriers, is inhibited by GSK-3 phosphorylation at FIP5-T276. Importantly, we show that FIP5-T276 phosphorylation occurs specifically during metaphase and anaphase, to ensure the fidelity and timing of FIP5-endosome targeting to the AMIS during apical lumen formation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Mitosis , Protein Processing, Post-Translational , Amino Acid Sequence , Animals , Cell Membrane/metabolism , Cell Polarity , Cytokinesis , Dogs , HeLa Cells , Humans , Madin Darby Canine Kidney Cells , Phosphorylation , Protein Binding , Protein Transport
6.
Mol Cell Biol ; 34(1): 43-56, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24164893

ABSTRACT

First identified in Drosophila, the Crumbs (Crb) proteins are important in epithelial polarity, apical membrane formation, and tight junction (TJ) assembly. The conserved Crb intracellular region includes a FERM (band 4.1/ezrin/radixin/moesin) binding domain (FBD) whose mammalian binding partners are not well understood and a PDZ binding motif that interacts with mammalian Pals1 (protein associated with lin seven) (also known as MPP5). Pals1 binds Patj (Pals1-associated tight-junction protein), a multi-PDZ-domain protein that associates with many tight junction proteins. The Crb complex also binds the conserved Par3/Par6/atypical protein kinase C (aPKC) polarity cassette that restricts migration of basolateral proteins through phosphorylation. Here, we describe a Crb3 knockout mouse that demonstrates extensive defects in epithelial morphogenesis. The mice die shortly after birth, with cystic kidneys and proteinaceous debris throughout the lungs. The intestines display villus fusion, apical membrane blebs, and disrupted microvilli. These intestinal defects phenocopy those of Ezrin knockout mice, and we demonstrate an interaction between Crumbs3 and ezrin. Taken together, our data indicate that Crumbs3 is crucial for epithelial morphogenesis and plays a role in linking the apical membrane to the underlying ezrin-containing cytoskeleton.


Subject(s)
Epithelium/metabolism , Kidney/metabolism , Lung/metabolism , Membrane Proteins/metabolism , Animals , Blotting, Western , Cell Line , Cell Polarity , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Cytoskeleton/metabolism , Epithelial Cells/metabolism , Epithelial Cells/ultrastructure , Epithelium/embryology , Epithelium/ultrastructure , Female , Kidney/embryology , Lung/embryology , Male , Membrane Glycoproteins , Membrane Proteins/genetics , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Survival Analysis , Tight Junctions/metabolism
7.
PLoS One ; 7(11): e48967, 2012.
Article in English | MEDLINE | ID: mdl-23145041

ABSTRACT

From a genetic screen for Drosophila melanogaster mutants with altered ethanol tolerance, we identified intolerant (intol), a novel allele of discs large 1 (dlg1). Dlg1 encodes Discs Large 1, a MAGUK (Membrane Associated Guanylate Kinase) family member that is the highly conserved homolog of mammalian PSD-95 and SAP97. The intol mutation disrupted specifically the expression of DlgS97, a SAP97 homolog, and one of two major protein isoforms encoded by dlg1 via alternative splicing. Expression of the major isoform, DlgA, a PSD-95 homolog, appeared unaffected. Ethanol tolerance in the intol mutant could be partially restored by transgenic expression of DlgS97, but not DlgA, in specific neurons of the fly's brain. Based on co-immunoprecipitation, DlgS97 forms a complex with N-methyl-D-aspartate (NMDA) receptors, a known target of ethanol. Consistent with these observations, flies expressing reduced levels of the essential NMDA receptor subunit dNR1 also showed reduced ethanol tolerance, as did mutants in the gene calcium/calmodulin-dependent protein kinase (caki), encoding the fly homolog of mammalian CASK, a known binding partner of DlgS97. Lastly, mice in which SAP97, the mammalian homolog of DlgS97, was conditionally deleted in adults failed to develop rapid tolerance to ethanol's sedative/hypnotic effects. We propose that DlgS97/SAP97 plays an important and conserved role in the development of tolerance to ethanol via NMDA receptor-mediated synaptic plasticity.


Subject(s)
Ethanol/toxicity , Guanylate Kinases/genetics , Membrane Proteins/genetics , Neurons/metabolism , Alleles , Alternative Splicing , Animals , Discs Large Homolog 1 Protein , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster , Female , Guanylate Kinases/metabolism , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mutation/genetics , Protein Isoforms , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
9.
Traffic ; 13(8): 1170-85, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22554228

ABSTRACT

During epithelial to mesenchymal transition (EMT), cells modulate expression of proteins resulting in loss of apical-basal polarity. Effectors of this EMT switch target the polarity protein Crumbs3a, a small transmembrane protein that is essential for generation of the apical membrane and tight junctions of mammalian epithelial cells. We previously showed that the Crumbs3 gene is a direct target of transcriptional regulation by Snail, a potent inducer of EMT. However, Snail has also been shown to have multiple non-transcriptional roles, including regulation of cell adhesion, proliferation and survival. Using SNAP-tag labeling, we determined that cell surface Crumbs3a has a half-life of approximately 3 h and that this cell surface half-life is significantly reduced when EMT is induced by Snail. We further observe that Snail induces differential glycosylation of Crumbs3a, including sialylation, suggesting a mechanism by which Crumbs3a may be destabilized. These results indicate that Crumbs3a is a post-translational target of Snail, in addition to being a transcriptional target. We conclude that Snail's ability to post-translationally modify and destabilize Crumbs3a augments the depolarizing process of EMT.


Subject(s)
Membrane Glycoproteins/metabolism , Transcription Factors/metabolism , Animals , Cell Line , Cell Membrane/metabolism , Dogs , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Glycosylation , Humans , Protein Processing, Post-Translational , Protein Stability , Snail Family Transcription Factors , Tight Junctions/metabolism
10.
Nat Cell Biol ; 14(4): 431-7, 2012 Mar 04.
Article in English | MEDLINE | ID: mdl-22388888

ABSTRACT

The cilium is a microtubule-based organelle that contains a unique complement of proteins for cell motility and signalling functions. Entry into the ciliary compartment is proposed to be regulated at the base of the cilium. Recent work demonstrated that components of the nuclear import machinery, including the Ran GTPase and importins, regulate ciliary entry. We hypothesized that the ciliary base contains a ciliary pore complex whose molecular nature and selective mechanism are similar to those of the nuclear pore complex. By microinjecting fluorescently labelled dextrans and recombinant proteins of various sizes, we characterize a size-dependent diffusion barrier for the entry of cytoplasmic molecules into primary cilia in mammalian cells. We demonstrate that nucleoporins localize to the base of primary and motile cilia and that microinjection of nucleoporin-function-blocking reagents blocks the ciliary entry of kinesin-2 KIF17 motors. Together, this work demonstrates that the physical and molecular nature of the ciliary pore complex is similar to that of the nuclear pore complex, and further extends functional parallels between nuclear and ciliary import.


Subject(s)
Biological Transport , Cilia/metabolism , Nuclear Pore Complex Proteins/metabolism , Cells, Cultured , Cytoplasm/metabolism , Dextrans , Humans , Molecular Weight , Permeability , Protein Transport
11.
Exp Cell Res ; 318(9): 1033-9, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22421511

ABSTRACT

The apicobasal polarization of epithelia is critical for many aspects of kidney function. Over the last decade there have been major advances in our understanding of the mechanisms that underlie this polarity. Critical to this understanding has been the identification of protein complexes on the apical and basolateral sides of epithelial cells that act in a mutually antagonistic manner to define these domains. Concomitant with the creation of apical and basolateral domains is the formation of highly specialized cell-cell junctions including adherens junctions and tight junctions. Recent research points to variability in the polarity and junctional complexes amongst different species and between different cell types of the kidney. Defects in apicobasal polarity are prominent in several disorders including acute renal failure and polycystic kidney disease.


Subject(s)
Cell Polarity , Epithelial Cells/metabolism , Kidney/metabolism , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Adherens Junctions/metabolism , Animals , Epithelial Cells/cytology , Humans , Intercellular Junctions/metabolism , Kidney/cytology , Polycystic Kidney Diseases/metabolism , Polycystic Kidney Diseases/pathology
12.
Mol Biol Cell ; 22(23): 4539-48, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21998203

ABSTRACT

The small GTPase Ran and the importin proteins regulate nucleocytoplasmic transport. New evidence suggests that Ran GTP and the importins are also involved in conveying proteins into cilia. In this study, we find that Ran GTP accumulation at the basal bodies is coordinated with the initiation of ciliogenesis. The Ran-binding protein 1 (RanBP1), which indirectly accelerates Ran GTP → Ran GDP hydrolysis and promotes the dissociation of the Ran/importin complex, also localizes to basal bodies and cilia. To confirm the crucial link between Ran GTP and ciliogenesis, we manipulated the levels of RanBP1 and determined the effects on Ran GTP and primary cilia formation. We discovered that RanBP1 knockdown results in an increased concentration of Ran GTP at basal bodies, leading to ciliogenesis. In contrast, overexpression of RanBP1 antagonizes primary cilia formation. Furthermore, we demonstrate that RanBP1 knockdown disrupts the proper localization of KIF17, a kinesin-2 motor, at the distal tips of primary cilia in Madin-Darby canine kidney cells. Our studies illuminate a new function for Ran GTP in stimulating cilia formation and reinforce the notion that Ran GTP and the importins play key roles in ciliogenesis and ciliary protein transport.


Subject(s)
Cilia/metabolism , Epithelial Cells/metabolism , Nuclear Proteins/metabolism , ran GTP-Binding Protein/metabolism , Animals , Cell Differentiation , Cell Line , Centrosome/metabolism , Dogs , Down-Regulation , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Kinesins/genetics , Kinesins/metabolism , Mice , Nuclear Proteins/genetics , ran GTP-Binding Protein/genetics
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.
Organogenesis ; 7(3): 147-53, 2011.
Article in English | MEDLINE | ID: mdl-21791971

ABSTRACT

Cilia are microtubule-based organelles that arise from the centrosome and project from the surface of many cells. Defects in cilia-localized proteins are felt to lead to polycystic kidney disease as well as ciliopathies with multiple organ involvement. Movement of proteins along mammalian cilia is a specialized process that is highly related to the intraflagellar movement of proteins in lower organisms. Entry of proteins into the cilia appears to be a tightly regulated process. Several cilia-targeting sequences have been identified that appear to mediate the movement of proteins into cilia, although the molecular basis through which these sequences operate is still being elucidated. Entry of proteins into cilia appears to be regulated at the base of the cilia at a region known as the transition zone. It has been proposed that a ciliary pore exists in this zone that controls entry of proteins into the cilia, similar to the nuclear pore that controls entry of proteins into the nucleus. Our group at the University of Michigan has found that proteins important in nuclear import appear to function similarly in cilia entry. In particular, we have identified roles for the small GTPase, Ran and its binding partners, the importins, in regulating cilia entry of specific proteins.


Subject(s)
Cilia/metabolism , Karyopherins/metabolism , ran GTP-Binding Protein/metabolism , Animals , Cell Movement/physiology , Cell Nucleus/metabolism , Chlamydomonas reinhardtii/metabolism , Humans , Monomeric GTP-Binding Proteins/metabolism , Protein Transport
15.
J Cell Sci ; 124(Pt 5): 718-26, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21285245

ABSTRACT

Ciliopathies represent a newly emerging group of human diseases that share a common etiology resulting from dysfunction of the cilium or centrosome. The gene encoding the retinitis pigmentosa 2 protein (RP2) is mutated in X-linked retinitis pigmentosa. RP2 localizes to the ciliary base and this requires the dual acylation of the N-terminus, but the precise mechanism by which RP2 is trafficked to the cilia is unknown. Here we have characterized an interaction between RP2 and Importin ß2 (transportin-1), a member of the Importin-ß family that regulates nuclear-cytoplasmic shuttling. We demonstrate that Importin ß2 is necessary for localization of RP2 to the primary cilium because ablation of Importin ß2 by shRNA blocks entry both of endogenous and exogenous RP2 to the cilium. Furthermore, we identify two distinct binding sites of RP2, which interact independently with Importin ß2. One binding site is a nuclear localization signal (NLS)-like sequence that is located at the N-terminus of RP2 and the other is an M9-like sequence within the tubulin folding cofactor C (TBCC) domain. Mutation of the NLS-like consensus sequence did not abolish localization of RP2 to cilia, suggesting that the sequence is not essential for RP2 ciliary targeting. Interestingly, we found that several missense mutations that cause human disease fall within the M9-like sequence of RP2 and these mutations block entry of RP2 into the cilium, as well as its interaction with Importin ß2. Together, this work further highlights a role of Importin ß2 in regulation of the entry of RP2 and other proteins into the ciliary compartment.


Subject(s)
Cilia/metabolism , Eye Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , beta Karyopherins/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cell Line , Centrosome/metabolism , Cilia/ultrastructure , Epithelial Cells/metabolism , Epithelial Cells/ultrastructure , Eye Proteins/genetics , GTP-Binding Proteins , Gene Knockdown Techniques , Humans , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Molecular Sequence Data , Multiprotein Complexes/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , beta Karyopherins/genetics
16.
Am J Physiol Renal Physiol ; 300(3): F589-601, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21228104

ABSTRACT

Establishment of epithelial apicobasal polarity is crucial for proper kidney development and function. In recent years, there have been important advances in our understanding of the factors that mediate the initiation of apicobasal polarization. Key among these are the polarity complexes that are evolutionarily conserved from simple organisms to humans. Three of these complexes are discussed in this review: the Crumbs complex, the Par complex, and the Scribble complex. The apical Crumbs complex consists of three proteins, Crumbs, PALS1, and PATJ, whereas the apical Par complex consists of Par-3, Par-6, and atypical protein kinase C. The lateral Scribble complex consists of Scribble, discs large, and lethal giant larvae. These complexes modulate kinase and small G protein activity such that the apical and basolateral complexes signal antagonistically, leading to the segregation of the apical and basolateral membranes. The polarity complexes also serve as scaffolds to direct and retain proteins at the apical membrane, the basolateral membrane, or the intervening tight junction. There is plasticity in apicobasal polarity, and this is best seen in the processes of epithelial-to-mesenchymal transition and the converse mesenchymal-to-epithelial transition. These transitions are important in kidney disease as well as kidney development, and modulation of the polarity complexes are critical for these transitions.


Subject(s)
Cell Polarity/physiology , Epithelial Cells/physiology , Kidney/cytology , Multiprotein Complexes/physiology , Cell Membrane/physiology , Eye Proteins/physiology , Humans , Kidney/physiology , Membrane Proteins/physiology , Neoplasm Proteins/physiology , Nerve Tissue Proteins/physiology , Tumor Suppressor Proteins/physiology
17.
Hum Mol Genet ; 19(22): 4330-44, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20729296

ABSTRACT

Ciliopathies represent a growing group of human genetic diseases whose etiology lies in defects in ciliogenesis or ciliary function. Given the established entity of renal-retinal ciliopathies, we have been examining the role of cilia-localized proteins mutated in retinitis pigmentosa (RP) in regulating renal ciliogenesis or cilia-dependent signaling cascades. Specifically, this study examines the role of the RP2 gene product with an emphasis on renal and vertebrate development. We demonstrate that in renal epithelia, RP2 localizes to the primary cilium through dual acylation of the amino-terminus. We also show that RP2 forms a calcium-sensitive complex with the autosomal dominant polycystic kidney disease protein polycystin 2. Ablation of RP2 by shRNA promotes swelling of the cilia tip that may be a result of aberrant trafficking of polycystin 2 and other ciliary proteins. Morpholino-mediated repression of RP2 expression in zebrafish results in multiple developmental defects that have been previously associated with ciliary dysfunction, such as hydrocephalus, kidney cysts and situs inversus. Finally, we demonstrate that, in addition to our observed physical interaction between RP2 and polycystin 2, dual morpholino-mediated knockdown of polycystin 2 and RP2 results in enhanced situs inversus, indicating that these two genes also regulate a common developmental process. This work suggests that RP2 may be an important regulator of ciliary function through its association with polycystin 2 and provides evidence of a further link between retinal and renal cilia function.


Subject(s)
Cilia/physiology , Eye Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , TRPP Cation Channels/metabolism , Acylation , Animals , Cilia/genetics , Cilia/metabolism , GTP-Binding Proteins , Gene Knockdown Techniques , Humans , Kidney/metabolism , Kidney/physiopathology , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/metabolism , Mutation , Vertebrates/genetics , Vertebrates/metabolism , Zebrafish/genetics , Zebrafish/metabolism
18.
Nat Cell Biol ; 12(7): 703-10, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20526328

ABSTRACT

The biogenesis, maintenance and function of primary cilia are controlled through intraflagellar transport (IFT) driven by two kinesin-2 family members, the heterotrimeric KIF3A/KIF3B/KAP complex and the homodimeric KIF17 motor. How these motors and their cargoes gain access to the ciliary compartment is poorly understood. Here, we identify a ciliary localization signal (CLS) in the KIF17 tail domain that is necessary and sufficient for ciliary targeting. Similarities between the CLS and classic nuclear localization signals (NLSs) suggest that similar mechanisms regulate nuclear and ciliary import. We hypothesize that ciliary targeting of KIF17 is regulated by a ciliary-cytoplasmic gradient of the small GTPase Ran, with high levels of GTP-bound Ran (RanGTP) in the cilium. Consistent with this, cytoplasmic expression of GTP-locked Ran(G19V) disrupts the gradient and abolishes ciliary entry of KIF17. Furthermore, KIF17 interacts with the nuclear import protein importin-beta2 in a manner dependent on the CLS and inhibited by RanGTP. We propose that Ran has a global role in regulating cellular compartmentalization by controlling the shuttling of cytoplasmic proteins into nuclear and ciliary compartments.


Subject(s)
Cilia/metabolism , Kinesins/metabolism , beta Karyopherins/metabolism , ran GTP-Binding Protein/metabolism , Animals , Dogs , Humans , Kinesins/genetics , Mice , NIH 3T3 Cells , Protein Binding , beta Karyopherins/genetics , ran GTP-Binding Protein/genetics
20.
Mol Biol Cell ; 20(22): 4652-63, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19776356

ABSTRACT

Although lumen generation has been extensively studied through so-called cyst-formation assays in Madin-Darby canine kidney (MDCK) cells, an underlying mechanism that leads to the initial appearance of a solitary lumen remains elusive. Lumen formation is thought to take place at early stages in aggregates containing only a few cells. Evolutionarily conserved polarity protein complexes, namely the Crumbs, Par, and Scribble complexes, establish apicobasal polarity in epithelial cells, and interference with their function impairs the regulated formation of solitary epithelial lumina. Here, we demonstrate that MDCK cells form solitary lumina during their first cell division. Before mitosis, Crumbs3a becomes internalized and concentrated in Rab11-positive recycling endosomes. These compartments become partitioned in both daughter cells and are delivered to the site of cytokinesis, thus forming the first apical membrane, which will eventually form a lumen. Endosome trafficking in this context appears to depend on the mitotic spindle apparatus and midzone microtubules. Furthermore, we show that this early lumen formation is regulated by the apical polarity complexes because Crumbs3 assists in the recruitment of aPKC to the forming apical membrane and interference with their function can lead to the formation of a no-lumen or multiple-lumen phenotype at the two-cell stage.


Subject(s)
Cell Polarity , Cytokinesis/physiology , Membrane Proteins/metabolism , Morphogenesis , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Cytoskeleton/metabolism , Dogs , Endocytosis/physiology , Endosomes/metabolism , Membrane Proteins/genetics , Microtubules/metabolism , Multiprotein Complexes/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Kinase C/metabolism , Protein Transport/physiology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...