Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Nat Commun ; 12(1): 4469, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34294704

ABSTRACT

The basal body of the bacterial flagellum is a rotary motor that consists of several rings (C, MS and LP) and a rod. The LP ring acts as a bushing supporting the distal rod for its rapid and stable rotation without much friction. Here, we use electron cryomicroscopy to describe the LP ring structure around the rod, at 3.5 Å resolution, from Salmonella Typhimurium. The structure shows 26-fold rotational symmetry and intricate intersubunit interactions of each subunit with up to six partners, which explains the structural stability. The inner surface is charged both positively and negatively. Positive charges on the P ring (the part of the LP ring that is embedded within the peptidoglycan layer) presumably play important roles in its initial assembly around the rod with a negatively charged surface.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/ultrastructure , Flagella/chemistry , Flagella/ultrastructure , Molecular Motor Proteins/chemistry , Molecular Motor Proteins/ultrastructure , Bacterial Proteins/physiology , Basal Bodies/chemistry , Basal Bodies/physiology , Basal Bodies/ultrastructure , Cryoelectron Microscopy , Flagella/physiology , Models, Molecular , Molecular Motor Proteins/physiology , Movement/physiology , Protein Interaction Domains and Motifs , Protein Structure, Quaternary , Protein Subunits , Salmonella typhimurium/chemistry , Salmonella typhimurium/physiology , Salmonella typhimurium/ultrastructure , Static Electricity
2.
Mol Biol Cell ; 32(20): ar12, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34319756

ABSTRACT

Motile cilia of multiciliated epithelial cells undergo synchronized beating to produce fluid flow along the luminal surface of various organs. Each motile cilium consists of an axoneme and a basal body (BB), which are linked by a "transition zone" (TZ). The axoneme exhibits a characteristic 9+2 microtubule arrangement important for ciliary motion, but how this microtubule system is generated is not yet fully understood. Here we show that calmodulin-regulated spectrin-associated protein 3 (CAMSAP3), a protein that can stabilize the minus-end of a microtubule, concentrates at multiple sites of the cilium-BB complex, including the upper region of the TZ or the axonemal basal plate (BP) where the central pair of microtubules (CP) initiates. CAMSAP3 dysfunction resulted in loss of the CP and partial distortion of the BP, as well as the failure of multicilia to undergo synchronized beating. These findings suggest that CAMSAP3 plays pivotal roles in the formation or stabilization of the CP by localizing at the basal region of the axoneme and thereby supports the coordinated motion of multicilia in airway epithelial cells.


Subject(s)
Cilia/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Actin Cytoskeleton/metabolism , Animals , Axoneme/physiology , Basal Bodies/physiology , Epithelial Cells/metabolism , Female , Male , Mice , Mice, Inbred ICR , Mice, Transgenic , Movement/physiology , Trachea/physiology
4.
J Mol Biol ; 433(13): 167004, 2021 06 25.
Article in English | MEDLINE | ID: mdl-33891903

ABSTRACT

The bacterial flagellum consists of a long extracellular filament that is rotated by a motor embedded in the cell envelope. While flagellar assembly has been extensively studied,1 the disassembly process remains less well understood. In addition to the programmed flagellar ejection that occurs during the life cycle of Caulobacter crescentus, we and others have recently shown that many bacterial species lose their flagella under starvation conditions, leaving relic structures in the outer membrane.2-7 However, it remains unknown whether the programmed flagellar ejection of C. crescentus leaves similar relics or not. Here, we imaged the various stages of the C. crescentus life cycle using electron cryo-tomography (cryo-ET) and found that flagellar relic subcomplexes, akin to those produced in the starvation-induced process, remain as a result of flagellar ejection during cell development. This similarity suggests that the programmed flagellar ejection of C. crescentus might share a common evolutionary path with the more general, and likely more ancient,3 starvation-related flagellar loss.


Subject(s)
Caulobacter crescentus/physiology , Cell Wall/metabolism , Flagella/physiology , Lipopolysaccharides/metabolism , Peptidoglycan/metabolism , Basal Bodies/physiology , Basal Bodies/ultrastructure , Caulobacter crescentus/metabolism , Caulobacter crescentus/ultrastructure , Cell Wall/ultrastructure , Electron Microscope Tomography/methods , Fimbriae, Bacterial/physiology , Fimbriae, Bacterial/ultrastructure , Flagella/ultrastructure
5.
PLoS One ; 16(4): e0249908, 2021.
Article in English | MEDLINE | ID: mdl-33861760

ABSTRACT

The single mitochondrial nucleoid (kinetoplast) of Trypanosoma brucei is found proximal to a basal body (mature (mBB)/probasal body (pBB) pair). Kinetoplast inheritance requires synthesis of, and scission of kinetoplast DNA (kDNA) generating two kinetoplasts that segregate with basal bodies into daughter cells. Molecular details of kinetoplast scission and the extent to which basal body separation influences the process are unavailable. To address this topic, we followed basal body movements in bloodstream trypanosomes following depletion of protein kinase TbCK1.2 which promotes kinetoplast division. In control cells we found that pBBs are positioned 0.4 um from mBBs in G1, and they mature after separating from mBBs by at least 0.8 um: mBB separation reaches ~2.2 um. These data indicate that current models of basal body biogenesis in which pBBs mature in close proximity to mBBs may need to be revisited. Knockdown of TbCK1.2 produced trypanosomes containing one kinetoplast and two nuclei (1K2N), increased the percentage of cells with uncleaved kDNA 400%, decreased mBB spacing by 15%, and inhibited cytokinesis 300%. We conclude that (a) separation of mBBs beyond a threshold of 1.8 um correlates with division of kDNA, and (b) TbCK1.2 regulates kDNA scission. We propose a Kinetoplast Division Factor hypothesis that integrates these data into a pathway for biogenesis of two daughter mitochondrial nucleoids.


Subject(s)
Basal Bodies/physiology , Casein Kinase I/metabolism , DNA, Kinetoplast/metabolism , Protozoan Proteins/metabolism , Trypanosoma brucei brucei/metabolism , Casein Kinase I/antagonists & inhibitors , Casein Kinase I/genetics , Cytokinesis/physiology , Cytoplasm/metabolism , DNA Cleavage , DNA Replication , Mitochondria/metabolism , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction
6.
PLoS Comput Biol ; 16(2): e1007649, 2020 02.
Article in English | MEDLINE | ID: mdl-32084125

ABSTRACT

In multi-ciliated cells, directed and synchronous ciliary beating in the apical membrane occurs through appropriate configuration of basal bodies (BBs, roots of cilia). Although it has been experimentally shown that the position and orientation of BBs are coordinated by apical cytoskeletons (CSKs), such as microtubules (MTs), and planar cell polarity (PCP), the underlying mechanism for achieving the patterning of BBs is not yet understood. In this study, we propose that polarity in bundles of apical MTs play a crucial role in the patterning of BBs. First, the necessity of the polarity was discussed by theoretical consideration on the symmetry of the system. The existence of the polarity was investigated by measuring relative angles between the MTs and BBs using published experimental data. Next, a mathematical model for BB patterning was derived by combining the polarity and self-organizational ability of CSKs. In the model, BBs were treated as finite-size particles in the medium of CSKs and excluded volume effects between BBs and CSKs were taken into account. The model reproduces the various experimental observations, including normal and drug-treated phenotypes. Our model with polarity provides a coherent and testable mechanism for apical BB pattern formation. We have also discussed the implication of our study on cell chirality.


Subject(s)
Basal Bodies/physiology , Cilia/physiology , Cytoskeleton/physiology , Animals , Cell Membrane , Cell Polarity , Computer Simulation , Elasticity , Epithelial Cells/cytology , Mice , Microtubules/physiology , Models, Theoretical , Nocodazole/pharmacology , Phenotype , Trachea/physiology
7.
Mol Microbiol ; 113(6): 1122-1139, 2020 06.
Article in English | MEDLINE | ID: mdl-32039533

ABSTRACT

The Lyme disease bacterium Borrelia burgdorferi has 7-11 periplasmic flagella (PF) that arise from the cell poles and extend toward the midcell as a flat-ribbon, which is distinct from other bacteria. FlhF, a signal recognition particle (SRP)-like GTPase, has been found to regulate the flagellar number and polarity; however, its role in B. burgdorferi remains unknown. B. burgdorferi has an FlhF homolog (BB0270). Structural and biochemical analyses show that BB0270 has a similar structure and enzymatic activity as its counterparts from other bacteria. Genetics and cryo-electron tomography studies reveal that deletion of BB0270 leads to mutant cells that have less PF (4 ± 2 PF per cell tip) and fail to form a flat-ribbon, indicative of a role of BB0270 in the control of PF number and configuration. Mechanistically, we demonstrate that BB0270 localizes at the cell poles and controls the number and position of PF via regulating the flagellar protein stability and the polar localization of the MS-ring protein FliF. Our study not only provides the detailed characterizations of BB0270 and its profound impacts on flagellar assembly, morphology and motility in B. burgdorferi, but also unveils mechanistic insights into how spirochetes control their unique flagellar patterns.


Subject(s)
Bacterial Proteins/metabolism , Borrelia burgdorferi/metabolism , Flagella/metabolism , Flagella/physiology , Monomeric GTP-Binding Proteins/metabolism , Bacterial Proteins/genetics , Basal Bodies/physiology , Borrelia burgdorferi/genetics , Electron Microscope Tomography , Flagella/genetics , Gene Deletion , Locomotion/genetics , Monomeric GTP-Binding Proteins/genetics
8.
J Cell Biol ; 219(1)2020 01 06.
Article in English | MEDLINE | ID: mdl-31740506

ABSTRACT

Multi-ciliary arrays promote fluid flow and cellular motility using the polarized and coordinated beating of hundreds of motile cilia. Tetrahymena basal bodies (BBs) nucleate and position cilia, whereby BB-associated striated fibers (SFs) promote BB anchorage and orientation into ciliary rows. Mutants that shorten SFs cause disoriented BBs. In contrast to the cytotaxis model, we show that disoriented BBs with short SFs can regain normal orientation if SF length is restored. In addition, SFs adopt unique lengths by their shrinkage and growth to establish and maintain BB connections and cortical interactions in a ciliary force-dependent mechanism. Tetrahymena SFs comprise at least eight uniquely localizing proteins belonging to the SF-assemblin family. Loss of different proteins that localize to the SF base disrupts either SF steady-state length or ciliary force-induced SF elongation. Thus, the dynamic regulation of SFs promotes BB connections and cortical interactions to organize ciliary arrays.


Subject(s)
Basal Bodies/physiology , Cilia/physiology , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Protozoan Proteins/metabolism , Tetrahymena thermophila/growth & development , Tetrahymena thermophila/metabolism , Mechanical Phenomena , Microtubule-Associated Proteins/genetics , Protozoan Proteins/genetics , Tetrahymena thermophila/genetics
9.
Development ; 146(6)2019 03 15.
Article in English | MEDLINE | ID: mdl-30877126

ABSTRACT

Motile cilia on multiciliated cells (MCCs) function in fluid clearance over epithelia. Studies with Xenopus embryos and individuals with the congenital respiratory disorder reduced generation of multiple motile cilia (RGMC), have implicated the nuclear protein MCIDAS (MCI), in the transcriptional regulation of MCC specification and differentiation. Recently, a paralogous protein, geminin coiled-coil domain containing (GMNC), was also shown to be required for MCC formation. Surprisingly, in contrast to the presently held view, we find that Mci mutant mice can specify MCC precursors. However, these precursors cannot produce multiple basal bodies, and mature into single ciliated cells. We identify an essential role for MCI in inducing deuterosome pathway components for the production of multiple basal bodies. Moreover, GMNC and MCI associate differentially with the cell-cycle regulators E2F4 and E2F5, which enables them to activate distinct sets of target genes (ciliary transcription factor genes versus basal body amplification genes). Our data establish a previously unrecognized two-step model for MCC development: GMNC functions in the initial step for MCC precursor specification. GMNC induces Mci expression that drives the second step of basal body production for multiciliation.


Subject(s)
Cell Cycle Proteins/physiology , Cilia/physiology , Mice, Mutant Strains , Nuclear Proteins/physiology , Animals , Basal Bodies/physiology , Carrier Proteins/physiology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Differentiation , Cell Nucleus/physiology , Ciliopathies , Gene Expression Regulation, Developmental , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/genetics , Protein Domains , Zebrafish
10.
Prog Retin Eye Res ; 71: 26-56, 2019 07.
Article in English | MEDLINE | ID: mdl-30590118

ABSTRACT

Photoreceptors are polarized neurons, with very specific subcellular compartmentalization and unique requirements for protein expression and trafficking. Each photoreceptor contains an outer segment, the site of photon capture that initiates vision, an inner segment that houses the biosynthetic machinery and a synaptic terminal for signal transmission to downstream neurons. Outer segments and inner segments are connected by a connecting cilium (CC), the equivalent of a transition zone (TZ) of primary cilia. The connecting cilium is part of the basal body/axoneme backbone that stabilizes the outer segment. This report will update the reader on late developments in photoreceptor ciliogenesis and transition zone formation, specifically in mouse photoreceptors, focusing on early events in photoreceptor ciliogenesis. The connecting cilium, an elongated and narrow structure through which all outer segment proteins and membrane components must traffic, functions as a gate that controls access to the outer segment. Here we will review genes and their protein products essential for basal body maturation and for CC/TZ genesis, sorted by phenotype. Emphasis is given to naturally occurring mouse mutants and gene knockouts that interfere with CC/TZ formation and ciliogenesis.


Subject(s)
Cilia/physiology , Photoreceptor Cells/physiology , Animals , Basal Bodies/physiology , Membrane Proteins/metabolism , Models, Animal , Protein Transport/physiology , Signal Transduction/physiology
11.
Dev Cell ; 46(5): 595-610.e3, 2018 09 10.
Article in English | MEDLINE | ID: mdl-30205038

ABSTRACT

The actin cytoskeleton is critical to shape cells and pattern intracellular organelles, which collectively drives tissue morphogenesis. In multiciliated cells (MCCs), apical actin drives expansion of the cell surface necessary to host hundreds of cilia. The apical actin also forms a lattice to uniformly distribute basal bodies. This apical actin network is dynamically remodeled, but the molecules that regulate its architecture remain poorly understood. We identify the chromatin modifier, WDR5, as a regulator of apical F-actin in MCCs. Unexpectedly in MCCs, WDR5 has a function independent of chromatin modification. We discover a scaffolding role for WDR5 between the basal body and F-actin. Specifically, WDR5 binds to basal bodies and migrates apically, where F-actin organizes around WDR5. Using a monomer trap for G-actin, we show that WDR5 stabilizes F-actin to maintain lattice architecture. In summary, we identify a non-chromatin role for WDR5 in stabilizing F-actin in MCCs.


Subject(s)
Actin Cytoskeleton/physiology , Basal Bodies/physiology , Cell Membrane/metabolism , Cilia/physiology , Embryo, Nonmammalian/physiology , Histone-Lysine N-Methyltransferase/metabolism , Animals , Embryo, Nonmammalian/cytology , Female , Histone-Lysine N-Methyltransferase/genetics , Humans , Intracellular Signaling Peptides and Proteins , Male , Mice , Morphogenesis , Xenopus
12.
PLoS Genet ; 14(2): e1007198, 2018 02.
Article in English | MEDLINE | ID: mdl-29425198

ABSTRACT

Pericentrin is a conserved centrosomal protein whose dysfunction has been linked to several human diseases. It has been implicated in many aspects of centrosome and cilia function, but its precise role is unclear. Here, we examine Drosophila Pericentrin-like-protein (PLP) function in vivo in tissues that form both centrosomes and cilia. Plp mutant centrioles exhibit four major defects: (1) They are short and have subtle structural abnormalities; (2) They disengage prematurely, and so overduplicate; (3) They organise fewer cytoplasmic MTs during interphase; (4) When forming cilia, they fail to establish and/or maintain a proper connection to the plasma membrane-although, surprisingly, they can still form an axoneme-like structure that can recruit transition zone (TZ) proteins. We show that PLP helps assemble "pericentriolar clouds" of electron-dense material that emanate from the central cartwheel spokes and spread outward to surround the mother centriole. We propose that the partial loss of these structures may largely explain the complex centriole, centrosome and cilium defects we observe in Plp mutant cells.


Subject(s)
Centrioles/metabolism , Centrosome/metabolism , Drosophila Proteins/physiology , Microtubules/metabolism , Animals , Animals, Genetically Modified , Basal Bodies/metabolism , Basal Bodies/physiology , Calmodulin-Binding Proteins , Centrioles/genetics , Cilia/genetics , Cilia/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Interphase/physiology , Multiprotein Complexes/metabolism , Mutation/physiology , Protein Multimerization/physiology , Protein Stability , Sensory Receptor Cells/chemistry , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/ultrastructure
13.
J Cell Biol ; 216(6): 1659-1671, 2017 06 05.
Article in English | MEDLINE | ID: mdl-28411189

ABSTRACT

Cilia are cellular projections that assemble on centriole-derived basal bodies. While cilia assembly is absolutely dependent on centrioles, it is not known to what extent they contribute to downstream events. The nematode C. elegans provides a unique opportunity to address this question, as centrioles do not persist at the base of mature cilia. Using fluorescence microscopy and electron tomography, we find that centrioles degenerate early during ciliogenesis. The transition zone and axoneme are not completely formed at this time, indicating that cilia maturation does not depend on intact centrioles. The hydrolethalus syndrome protein HYLS-1 is the only centriolar protein known to remain at the base of mature cilia and is required for intraflagellar transport trafficking. Surprisingly, targeted degradation of HYLS-1 after initiation of ciliogenesis does not affect ciliary structures. Taken together, our results indicate that while centrioles are essential to initiate cilia formation, they are dispensable for cilia maturation and maintenance.


Subject(s)
Basal Bodies/physiology , Caenorhabditis elegans/physiology , Centrioles/physiology , Neurogenesis , Sensory Receptor Cells/physiology , Animals , Animals, Genetically Modified , Axoneme/physiology , Basal Bodies/metabolism , Basal Bodies/ultrastructure , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/ultrastructure , Caenorhabditis elegans Proteins/metabolism , Centrioles/metabolism , Centrioles/ultrastructure , Cilia/physiology , Electron Microscope Tomography , Microscopy, Fluorescence , Microscopy, Video , Proteolysis , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/ultrastructure , Time Factors , Time-Lapse Imaging
14.
Development ; 143(24): 4654-4664, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27864379

ABSTRACT

Multiciliated cell (MCC) differentiation involves extensive organelle biogenesis required to extend hundreds of motile cilia. Key transcriptional regulators known to drive the gene expression required for this organelle biogenesis are activated by the related coiled-coil proteins Multicilin and Gemc1. Here we identify foxn4 as a new downstream target of Multicilin required for MCC differentiation in Xenopus skin. When Foxn4 activity is inhibited in Xenopus embryos, MCCs show transient ciliogenesis defects similar to those seen in mutants of Foxj1, a known key regulator of genes required for motile ciliation. RNAseq analysis indicates that Foxn4 co-activates some Foxj1 target genes strongly and many Foxj1 targets weakly. ChIPseq suggests that whereas Foxn4 and Foxj1 frequently bind to different targets at distal enhancers, they largely bind together at MCC gene promoters. Consistent with this co-regulation, cilia extension by MCCs is more severely compromised in foxn4 and foxj1 double mutants than in single mutants. In contrast to Foxj1, Foxn4 is not required to extend a single motile cilium by cells involved in left-right patterning. These results indicate that Foxn4 complements Foxj1 transcriptionally during MCC differentiation, thereby shaping the levels of gene expression required for the timely and complete biogenesis of multiple motile cilia.


Subject(s)
Cilia/metabolism , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Developmental , Skin/embryology , Xenopus Proteins/genetics , Xenopus laevis/embryology , Animals , Basal Bodies/physiology , CRISPR-Cas Systems/genetics , Carrier Proteins/genetics , Cell Cycle Proteins , Cell Differentiation/genetics , Cell Differentiation/physiology , DNA-Binding Proteins/genetics , Forkhead Transcription Factors/antagonists & inhibitors , Forkhead Transcription Factors/metabolism , Morpholinos/genetics , Nerve Tissue Proteins/genetics , Xenopus Proteins/antagonists & inhibitors , Xenopus Proteins/metabolism
15.
J Cell Biol ; 213(4): 435-50, 2016 05 23.
Article in English | MEDLINE | ID: mdl-27185836

ABSTRACT

Centrioles are the foundation of two organelles, centrosomes and cilia. Centriole numbers and functions are tightly controlled, and mutations in centriole proteins are linked to a variety of diseases, including microcephaly. Loss of the centriole protein Asterless (Asl), the Drosophila melanogaster orthologue of Cep152, prevents centriole duplication, which has limited the study of its nonduplication functions. Here, we identify populations of cells with Asl-free centrioles in developing Drosophila tissues, allowing us to assess its duplication-independent function. We show a role for Asl in controlling centriole length in germline and somatic tissue, functioning via the centriole protein Cep97. We also find that Asl is not essential for pericentriolar material recruitment or centrosome function in organizing mitotic spindles. Lastly, we show that Asl is required for proper basal body function and spermatid axoneme formation. Insights into the role of Asl/Cep152 beyond centriole duplication could help shed light on how Cep152 mutations lead to the development of microcephaly.


Subject(s)
Centrioles/metabolism , Centrioles/physiology , Drosophila Proteins/metabolism , Spermatozoa/growth & development , Spermatozoa/metabolism , Animals , Axoneme/metabolism , Axoneme/physiology , Basal Bodies/metabolism , Basal Bodies/physiology , Cell Cycle Proteins/metabolism , Drosophila melanogaster/metabolism , Drosophila melanogaster/physiology , Male , Mitosis/physiology , Spermatozoa/physiology
16.
Nat Commun ; 6: 8964, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26667778

ABSTRACT

Eukaryotic cilia/flagella exhibit two characteristic ultrastructures reflecting two main functions; a 9+2 axoneme for motility and a 9+0 axoneme for sensation and signalling. Whether, and if so how, they interconvert is unclear. Here we analyse flagellum length, structure and molecular composition changes in the unicellular eukaryotic parasite Leishmania during the transformation of a life cycle stage with a 9+2 axoneme (the promastigote) to one with a 9+0 axoneme (the amastigote). We show 9+0 axonemes can be generated by two pathways: by de novo formation and by restructuring of existing 9+2 axonemes associated with decreased intraflagellar transport. Furthermore, pro-basal bodies formed under conditions conducive for 9+2 axoneme formation can form a 9+0 axoneme de novo. We conclude that pro-centrioles/pro-basal bodies are multipotent and not committed to form either a 9+2 or 9+0 axoneme. In an alternative pathway structures can also be removed from existing 9+2 axonemes to convert them to 9+0.


Subject(s)
Axoneme/metabolism , Basal Bodies/physiology , Flagella/physiology , Leishmania mexicana/physiology , Animals , Axoneme/ultrastructure , Cell Division , Cells, Cultured , Chlamydomonas reinhardtii , Gene Expression Regulation , Green Fluorescent Proteins , Leishmania mexicana/ultrastructure , Macrophages/physiology , Mice , Mice, Inbred C57BL , Organisms, Genetically Modified , Recombinant Proteins
17.
Cytoskeleton (Hoboken) ; 72(10): 503-16, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26446290

ABSTRACT

In wild type (WT) tracheal epithelial cells, ciliary basal bodies are oriented such that all cilia on the cell surface beat in the same upward direction. This precise alignment of basal bodies and, as a result, the ciliary axoneme, is termed rotational planar cell polarity (PCP). Rotational PCP in the multi-ciliated epithelial cells of the trachea is perturbed in rats lacking myosin Id (Myo1d). Myo1d is localized in the F-actin and basal body rich subapical cortex of the ciliated tracheal epithelial cell. Scanning and transmission electron microscopy of Myo1d knock out (KO) trachea revealed that the unidirectional bending pattern is disrupted. Instead, cilia splay out in a disordered, often radial pattern. Measurement of the alignment axis of the central pair axonemal microtubules was much more variable in the KO, another indicator that rotational PCP is perturbed. The asymmetric localization of the PCP core protein Vangl1 is lost. Both the velocity and linearity of cilia-driven movement of beads above the tracheal mucosal surface was impaired in the Myo1d KO. Multi-ciliated brain ependymal epithelial cells exhibit a second form of PCP termed translational PCP in which basal bodies and attached cilia are clustered at the anterior side of the cell. The precise asymmetric clustering of cilia is disrupted in the ependymal cells of the Myo1d KO rat. While basal body clustering is maintained, left-right positioning of the clusters is lost.


Subject(s)
Ependyma/physiology , Epithelial Cells/cytology , Myosin Type I/physiology , Myosins/physiology , Actins/physiology , Animals , Animals, Genetically Modified , Axoneme/physiology , Basal Bodies/physiology , Brain/physiology , Cell Polarity , Cilia/physiology , In Situ Nick-End Labeling , Intestines/physiology , Male , Mice, Knockout , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Myosin Type I/genetics , Myosins/genetics , Neuroglia/physiology , Phenotype , Rats , Rats, Inbred F344 , Rotation , Trachea/physiology
19.
Methods Cell Biol ; 127: 131-59, 2015.
Article in English | MEDLINE | ID: mdl-25837389

ABSTRACT

Cilia are key organelles in development and homeostasis. The ever-expanding complement of cilia associated proteins necessitates rapid and tractable models for in vivo functional investigation. Xenopus laevis provides an attractive model for such studies, having multiple ciliated populations, including primary and multiciliated tissues. The rapid external development of Xenopus and the large cells make it an especially excellent platform for imaging studies. Here we present embryological and cell biological methods for the investigation of cilia structure and function in X. laevis, with a focus on quantitative live and fixed imaging.


Subject(s)
Cilia/physiology , Embryo, Nonmammalian/cytology , Animals , Axoneme/physiology , Basal Bodies/physiology , Fluorescent Antibody Technique/methods , Image Processing, Computer-Assisted , Microscopy, Confocal , Morpholinos/genetics , Oligonucleotides, Antisense/genetics , Staining and Labeling/methods , Tissue Fixation/methods , Xenopus laevis
20.
Methods Cell Biol ; 127: 457-85, 2015.
Article in English | MEDLINE | ID: mdl-25837404

ABSTRACT

Paramecium is a single cell able to divide in its morphologically differentiated stage that has many cilia anchored at its cell surface. Many thousands of cilia are thus assembled in a short period of time during division to duplicate the cell pattern while the cell continues swimming. Most, but not all, of these sensory cilia are motile and involved in two main functions: prey capture and cell locomotion. These cilia display heterogeneity, both in their length and their biochemical properties. Thanks to these properties, as well as to the availability of many postgenomic tools and the possibility to follow the regrowth of cilia after deciliation, Paramecium offers a nice opportunity to study the assembly of the cilia, as well as the genesis of their diversity within a single cell. In this paper, after a brief survey of Paramecium morphology and cilia properties, we describe the tools and the protocols currently used for immunofluorescence, transmission electron microscopy, and ultrastructural immunocytochemistry to analyze cilia, with special recommendations to overcome the problem raised by cilium diversity.


Subject(s)
Cell Movement/physiology , Cilia/physiology , Paramecium/physiology , Antibodies/immunology , Basal Bodies/physiology , Basal Bodies/ultrastructure , Cell Membrane/metabolism , Cilia/ultrastructure , Fluorescent Antibody Technique/methods , Immunohistochemistry/methods , Microscopy, Electron, Transmission/methods , Paramecium/genetics , Staining and Labeling/methods , Tubulin/immunology , Tubulin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...