Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Development ; 151(1)2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38031972

ABSTRACT

Cilia are microtubule (MT)-based organelles present on the surface of nearly all vertebrate cells. MTs are polymers of α- and ß-tubulins that are each encoded by multiple, individual isotype genes. Tubulin isotype composition is thought to influence MT behaviors. Ciliary MTs differ from other MTs in the cell in terms of organization, stability and post-translational modifications. However, little is known about the tubulin isotypes that build ciliary MTs and the functional requirements for tubulin isotypes in cilia have not been examined in vertebrates. Here, we have tested the role of the ß-tubulin isotype genes in the mouse that harbor a conserved amino acid motif associated with ciliated organisms. We found that Tubb4b localizes to cilia in multi-ciliated cells (MCCs) specifically. In respiratory and oviduct MCCs, Tubb4b is asymmetrically localized within multi-cilia, indicating that the tubulin isotype composition changes along the length of the ciliary axonemal MTs. Deletion of Tubb4b resulted in striking structural defects within the axonemes of multi-cilia, without affecting primary cilia. These studies show that Tubb4b is essential for the formation of a specific MT-based subcellular organelle and sheds light on the requirements of tubulin isotypes in cilia.


Subject(s)
Cilia , Tubulin , Animals , Mice , Axoneme/metabolism , Cilia/metabolism , Microtubules/metabolism , Protein Processing, Post-Translational , Tubulin/genetics , Tubulin/metabolism
2.
Cell ; 151(4): 847-858, 2012 Nov 09.
Article in English | MEDLINE | ID: mdl-23141541

ABSTRACT

The primary cilium has critical roles in human development and disease, but the mechanisms that regulate ciliogenesis are not understood. Here, we show that Tau tubulin kinase 2 (TTBK2) is a dedicated regulator of the initiation of ciliogenesis in vivo. We identified a null allele of mouse Ttbk2 based on loss of Sonic hedgehog activity, a signaling pathway that requires the primary cilium. Despite a normal basal body template, Ttbk2 mutants lack cilia. TTBK2 acts at the distal end of the basal body, where it promotes the removal of CP110, which caps the mother centriole, and promotes recruitment of IFT proteins, which build the ciliary axoneme. Dominant truncating mutations in human TTBK2 cause spinocerebellar ataxia type 11 (SCA11); these mutant proteins do not promote ciliogenesis and inhibit ciliogenesis in wild-type cells. We propose that cell-cycle regulators target TTBK2 to the basal body, where it modifies specific targets to initiate ciliogenesis.


Subject(s)
Cilia/metabolism , Mutation , Protein Serine-Threonine Kinases/metabolism , Animals , Cell Cycle Proteins/metabolism , Hedgehog Proteins/metabolism , Humans , Mice , Microtubule-Associated Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Spinocerebellar Ataxias/metabolism
3.
J Am Soc Nephrol ; 30(11): 2103-2111, 2019 11.
Article in English | MEDLINE | ID: mdl-31451534

ABSTRACT

BACKGROUND: PKD1 or PKD2, the two main causal genes for autosomal dominant polycystic kidney disease (ADPKD), encode the multipass transmembrane proteins polycystin-1 (PC1) and polycystin-2 (PC2), respectively. Polycystins localize to the primary cilium, an organelle essential for cell signaling, including signal transduction of the Hedgehog pathway. Mutations in ciliary genes that build and maintain the cilium also cause renal cystic disease through unknown pathways. Although recent studies have found alterations in Hedgehog signaling in ADPKD-related models and tissues, the relationship between Hedgehog and polycystic kidney disease is not known. METHODS: To examine the potential role of cell-autonomous Hedgehog signaling in regulating kidney cyst formation in vivo in both early- and adult-onset mouse models of ADPKD, we used conditional inactivation of Pkd1 combined with conditional modulation of Hedgehog signaling components in renal epithelial cells, where mutations in Pkd1 initiate cyst formation. After increasing or decreasing levels of Hedgehog signaling in cells that underwent inactivation of Pkd1, we evaluated the effects of these genetic manipulations on quantitative parameters of polycystic kidney disease severity. RESULTS: We found that in Pkd1 conditional mutant mouse kidneys, neither downregulation nor activation of the Hedgehog pathway in epithelial cells along the nephron significantly influenced the severity of the polycystic kidney phenotype in mouse models of developmental or adult-onset of ADPKD. CONCLUSIONS: These data suggest that loss of Pkd1 function results in kidney cysts through pathways that are not affected by the activity of the Hedgehog pathway.


Subject(s)
Hedgehog Proteins/physiology , Polycystic Kidney, Autosomal Dominant/etiology , Animals , Disease Models, Animal , Mice , Signal Transduction/physiology , TRPP Cation Channels/genetics , TRPP Cation Channels/physiology , Zinc Finger Protein GLI1/physiology
4.
Development ; 140(6): 1262-71, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23406901

ABSTRACT

The planar cell polarity (PCP; non-canonical Wnt) pathway is required to orient the cells within the plane of an epithelium. Here, we show that cofilin 1 (Cfl1), an actin-severing protein, and Vangl2, a core PCP protein, cooperate to control PCP in the early mouse embryo. Two aspects of planar polarity can be analyzed quantitatively at cellular resolution in the mouse embryo: convergent extension of the axial midline; and posterior positioning of cilia on cells of the node. Analysis of the spatial distribution of brachyury(+) midline cells shows that the Cfl1 mutant midline is normal, whereas Vangl2 mutants have a slightly wider midline. By contrast, midline convergent extension fails completely in Vangl2 Cfl1 double mutants. Planar polarity is required for the posterior positioning of cilia on cells in the mouse node, which is essential for the initiation of left-right asymmetry. Node cilia are correctly positioned in Cfl1 and Vangl2 single mutants, but cilia remain in the center of the cell in Vangl2 Cfl1 double mutants, leading to randomization of left-right asymmetry. In both the midline and node, the defect in planar polarity in the double mutants arises because PCP protein complexes fail to traffic to the apical cell membrane, although other aspects of apical-basal polarity are unaffected. Genetic and pharmacological experiments demonstrate that F-actin remodeling is essential for the initiation, but not maintenance, of PCP. We propose that Vangl2 and cofilin cooperate to target Rab11(+) vesicles containing PCP proteins to the apical membrane during the initiation of planar cell polarity.


Subject(s)
Body Patterning/genetics , Cell Polarity/genetics , Cofilin 1/physiology , Embryonic Development/genetics , Nerve Tissue Proteins/physiology , Animals , Cells, Cultured , Cilia/genetics , Cilia/metabolism , Cilia/physiology , Cofilin 1/genetics , Cofilin 1/metabolism , Embryo Culture Techniques , Embryo, Mammalian , Embryonic Development/physiology , Epistasis, Genetic , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Phenotype
5.
Development ; 140(23): 4672-82, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24173802

ABSTRACT

Mammalian neuroepithelial stem cells divide using a polarized form of cytokinesis, which is not well understood. The cytokinetic furrow cleaves the cell by ingressing from basal to apical, forming the midbody at the apical membrane. The midbody mediates abscission by recruiting many factors, including the Kinesin-6 family member Kif20b. In developing embryos, Kif20b mRNA is most highly expressed in neural stem/progenitor cells. A loss-of-function mutant in Kif20b, magoo, was found in a forward genetic screen. magoo has a small cerebral cortex, with reduced production of progenitors and neurons, but preserved layering. In contrast to other microcephalic mouse mutants, mitosis and cleavage furrows of cortical stem cells appear normal in magoo. However, apical midbodies show changes in number, shape and positioning relative to the apical membrane. Interestingly, the disruption of abscission does not appear to result in binucleate cells, but in apoptosis. Thus, Kif20b is required for proper midbody organization and abscission in polarized cortical stem cells and has a crucial role in the regulation of cerebral cortex growth.


Subject(s)
Cerebral Cortex/metabolism , Cytokinesis/physiology , Kinesins/metabolism , Neural Stem Cells/metabolism , Animals , Cell Polarity/genetics , Gene Expression , Kinesins/genetics , Mice , Mice, Inbred C57BL , Microtubules/metabolism , RNA, Messenger/biosynthesis
6.
Dev Biol ; 385(1): 67-82, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24161523

ABSTRACT

The long bones of the vertebrate body are built by the initial formation of a cartilage template that is later replaced by mineralized bone. The proliferation and maturation of the skeletal precursor cells (chondrocytes) within the cartilage template and their replacement by bone is a highly coordinated process which, if misregulated, can lead to a number of defects including dwarfism and other skeletal deformities. This is exemplified by the fact that abnormal bone development is one of the most common types of human birth defects. Yet, many of the factors that initiate and regulate chondrocyte maturation are not known. We identified a recessive dwarf mouse mutant (pug) from an N-ethyl-N-nitrosourea (ENU) mutagenesis screen. pug mutant skeletal elements are patterned normally during development, but display a ~20% length reduction compared to wild-type embryos. We show that the pug mutation does not lead to changes in chondrocyte proliferation but instead promotes premature maturation and early ossification, which ultimately leads to disproportionate dwarfism. Using sequence capture and high-throughput sequencing, we identified a missense mutation in the Xylosyltransferase 1 (Xylt1) gene in pug mutants. Xylosyltransferases catalyze the initial step in glycosaminoglycan (GAG) chain addition to proteoglycan core proteins, and these modifications are essential for normal proteoglycan function. We show that the pug mutation disrupts Xylt1 activity and subcellular localization, leading to a reduction in GAG chains in pug mutants. The pug mutant serves as a novel model for mammalian dwarfism and identifies a key role for proteoglycan modification in the initiation of chondrocyte maturation.


Subject(s)
Bone and Bones/embryology , Chondrocytes/metabolism , Osteogenesis/genetics , Pentosyltransferases/physiology , Animals , Base Sequence , Bone and Bones/metabolism , Cell Differentiation/genetics , Cell Proliferation , Dwarfism/genetics , Fibroblast Growth Factors/metabolism , Hedgehog Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation, Missense , Parathyroid Hormone-Related Protein/metabolism , Pentosyltransferases/genetics , Sequence Analysis, DNA , Signal Transduction/genetics , UDP Xylose-Protein Xylosyltransferase
7.
Hum Mol Genet ; 21(8): 1808-23, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22228095

ABSTRACT

Mutations in components of the intraflagellar transport (IFT) machinery required for assembly and function of the primary cilium cause a subset of human ciliopathies characterized primarily by skeletal dysplasia. Recently, mutations in the IFT-A gene IFT144 have been described in patients with Sensenbrenner and Jeune syndromes, which are associated with short ribs and limbs, polydactyly and craniofacial defects. Here, we describe an N-ethyl-N-nitrosourea-derived mouse mutant with a hypomorphic missense mutation in the Ift144 gene. The mutant twinkle-toes (Ift144(twt)) phenocopies a number of the skeletal and craniofacial anomalies seen in patients with human skeletal ciliopathies. Like other IFT-A mouse mutants, Ift144 mutant embryos display a generalized ligand-independent expansion of hedgehog (Hh) signalling, in spite of defective ciliogenesis and an attenuation of the ability of mutant cells to respond to upstream stimulation of the pathway. This enhanced Hh signalling is consistent with cleft palate and polydactyly phenotypes in the Ift144(twt) mutant, although extensive rib branching, fusion and truncation phenotypes correlate with defects in early somite patterning and may reflect contributions from multiple signalling pathways. Analysis of embryos harbouring a second allele of Ift144 which represents a functional null, revealed a dose-dependent effect on limb outgrowth consistent with the short-limb phenotypes characteristic of these ciliopathies. This allelic series of mouse mutants provides a unique opportunity to uncover the underlying mechanistic basis of this intriguing subset of ciliopathies.


Subject(s)
Abnormalities, Multiple/genetics , Cilia , Craniofacial Abnormalities/genetics , Proteins/genetics , Abnormalities, Multiple/embryology , Abnormalities, Multiple/metabolism , Animals , Chromosome Mapping , Cilia/physiology , Cilia/ultrastructure , Craniofacial Abnormalities/embryology , Craniofacial Abnormalities/metabolism , Cytoskeletal Proteins , Embryo, Mammalian , Fibroblast Growth Factors/metabolism , Forelimb/abnormalities , Forelimb/metabolism , Hedgehog Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins , Mice , Mutagenesis , Mutation, Missense , Phenotype , Polydactyly/embryology , Polydactyly/genetics , Polydactyly/metabolism , Proteins/chemistry , Ribs/abnormalities , Signal Transduction
8.
Development ; 138(24): 5333-43, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22110054

ABSTRACT

TRIM28 is a transcriptional regulator that is essential for embryonic development and is implicated in a variety of human diseases. The roles of TRIM28 in distinct biological processes are thought to depend on its interaction with factors that determine its DNA target specificity. However, functional evidence linking TRIM28 to specific co-factors is scarce. chatwo, a hypomorphic allele of Trim28, causes embryonic lethality and defects in convergent extension and morphogenesis of extra-embryonic tissues. These phenotypes are remarkably similar to those of mutants in the Krüppel-associated box (KRAB) zinc finger protein ZFP568, providing strong genetic evidence that ZFP568 and TRIM28 control morphogenesis through a common molecular mechanism. We determined that chatwo mutations decrease TRIM28 protein stability and repressive activity, disrupting both ZFP568-dependent and ZFP568-independent roles of TRIM28. These results, together with the analysis of embryos bearing a conditional inactivation of Trim28 in embryonic-derived tissues, revealed that TRIM28 is differentially required by ZFP568 and other factors during the early stages of mouse embryogenesis. In addition to uncovering novel roles of TRIM28 in convergent extension and morphogenesis of extra-embryonic tissues, our characterization of chatwo mutants demonstrates that KRAB domain proteins are essential to determine some of the biological functions of TRIM28.


Subject(s)
Carrier Proteins/metabolism , Morphogenesis , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Amino Acid Sequence , Animals , Cell Line , Female , Gastrula/metabolism , Gastrulation , Gene Expression Regulation, Developmental , Male , Mice , Molecular Sequence Data , Mutation , Protein Stability , Tripartite Motif-Containing Protein 28
9.
Mol Biol Cell ; 34(3): ar18, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36652335

ABSTRACT

The primary cilium is a nexus for cell signaling and relies on specific protein trafficking for function. The tubby family protein TULP3 transports integral membrane proteins into cilia through interactions with the intraflagellar transport complex-A (IFT-A) and phosphoinositides. It was previously shown that short motifs called ciliary localization sequences (CLSs) are necessary and sufficient for TULP3-dependent ciliary trafficking of transmembrane cargoes. However, the mechanisms by which TULP3 regulates ciliary compartmentalization of nonintegral, membrane-associated proteins and whether such trafficking requires TULP3-dependent CLSs is unknown. Here we show that TULP3 is required for ciliary transport of the Joubert syndrome-linked palmitoylated GTPase ARL13B through a CLS. An N-terminal amphipathic helix, preceding the GTPase domain of ARL13B, couples with the TULP3 tubby domain for ciliary trafficking, irrespective of palmitoylation. ARL13B transport requires TULP3 binding to IFT-A but not to phosphoinositides, indicating strong membrane-proximate interactions, unlike transmembrane cargo transport requiring both properties of TULP3. TULP3-mediated trafficking of ARL13B also regulates ciliary enrichment of farnesylated and myristoylated downstream effectors of ARL13B. The lipidated cargoes show distinctive depletion kinetics from kidney epithelial cilia with relation to Tulp3 deletion-induced renal cystogenesis. Overall, these findings indicate an expanded role of the tubby domain in capturing analogous helical secondary structural motifs from diverse cargoes.


Subject(s)
Cilia , Membrane Proteins , Cilia/metabolism , Protein Transport , Membrane Proteins/metabolism , GTP Phosphohydrolases/metabolism , Phosphatidylinositols/metabolism
10.
Proc Natl Acad Sci U S A ; 106(32): 13377-82, 2009 Aug 11.
Article in English | MEDLINE | ID: mdl-19666503

ABSTRACT

Mammalian Sonic hedgehog (Shh) signaling is essential for embryonic development and stem cell maintenance and has critical roles in tumorigenesis. Although core components of the Shh pathway are conserved in evolution, important aspects of mammalian Shh signaling are not shared with the Drosophila pathway. Perhaps the most dramatic difference between the Drosophila and mammalian pathways is that Shh signaling in the mouse requires a microtubule-based organelle, the primary cilium. Proteins that are required for the response to Shh are enriched in the cilium, but it is not clear why the cilium provides an appropriate venue for signal transduction. Here, we demonstrate that Kif7, a mammalian homologue of Drosophila Costal2 (Cos2), is a cilia-associated protein that regulates signaling from the membrane protein Smoothened (Smo) to Gli transcription factors. By using a Kif7 mutant allele identified in a reporter-based genetic screen, we show that, similar to Drosophila and zebrafish Cos2, mouse Kif7 acts downstream of Smo and upstream of Gli2 and has both negative and positive roles in Shh signal transduction. Mouse Kif7 activity depends on the presence of cilia and Kif7-eGFP localizes to base of the primary cilium in the absence of Shh. Activation of the Shh pathway promotes trafficking of Kif7-eGFP from the base to the tip of the cilium, and localization to the tip of the cilium is disrupted in a motor domain mutant. We conclude that Kif7 is a core regulator of Shh signaling that may also act as a ciliary motor.


Subject(s)
Cilia/metabolism , Hedgehog Proteins/metabolism , Kinesins/metabolism , Signal Transduction , Animals , Cell Lineage , Flagella/metabolism , Kinesins/genetics , Mice , Mutation/genetics , Neural Tube/cytology , Neural Tube/metabolism , Phenotype , Protein Transport
11.
Sci Adv ; 8(7): eabf7262, 2022 02 18.
Article in English | MEDLINE | ID: mdl-35171680

ABSTRACT

Mutations in tubulins cause distinct neurodevelopmental and degenerative diseases termed "tubulinopathies"; however, little is known about the functional requirements of tubulins or how mutations cause cell-specific pathologies. Here, we identify a mutation in the gene Tubb4a that causes degeneration of cerebellar granule neurons and myelination defects. We show that the neural phenotypes result from a cell type-specific enrichment of a dominant mutant form of Tubb4a relative to the expression other ß-tubulin isotypes. Loss of Tubb4a function does not underlie cellular pathology but is compensated by the transcriptional up-regulation of related tubulin genes in a cell type-specific manner. This work establishes that the expression of a primary tubulin mutation in mature neurons is sufficient to promote cell-autonomous cell death, consistent with a causative association of microtubule dysfunction with neurodegenerative diseases. These studies provide evidence that mutations in tubulins cause specific phenotypes based on expression ratios of tubulin isotype genes.


Subject(s)
Models, Genetic , Tubulin , Cerebellum/metabolism , Microtubules/metabolism , Mutation , Tubulin/genetics , Tubulin/metabolism
12.
Front Genet ; 13: 1021037, 2022.
Article in English | MEDLINE | ID: mdl-36276950

ABSTRACT

Patient variants in Tubby Like Protein-3 (TULP3) have recently been associated with progressive fibrocystic disease in tissues and organs. TULP3 is a ciliary trafficking protein that links membrane-associated proteins to the intraflagellar transport complex A. In mice, mutations in Tulp3 drive phenotypes consistent with ciliary dysfunction which include renal cystic disease, as part of a ciliopathic spectrum. Here we report two sisters from consanguineous parents with fibrocystic renal and hepatic disease harboring a homozygous missense mutation in TULP3 (NM_003324.5: c.1144C>T, p.Arg382Trp). The R382W patient mutation resides within the C-terminal Tubby domain, a conserved domain required for TULP3 to associate with phosphoinositides. We show that inner medullary collecting duct-3 cells expressing the TULP3 R382W patient variant have a severely reduced ability to localize the membrane-associated proteins ARL13b, INPP5E, and GPR161 to the cilium, consistent with a loss of TULP3 function. These studies establish Arginine 382 as a critical residue in the Tubby domain, which is essential for TULP3-mediated protein trafficking within the cilium, and expand the phenotypic spectrum known to result from recessive deleterious mutations in TULP3.

13.
Neuroscience ; 450: 3-14, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32682825

ABSTRACT

Cell specification in the ventral spinal cord is a well-studied model system to understand how tissue pattern develops in response to a morphogen gradient. Ventral cell types including motor neurons (MNs) are induced in the neural tube in response to graded Sonic Hedgehog (Shh) signaling. We performed a forward genetic screen in the mouse that incorporated a GFP-expressing transgene to visualize MNs to identify genes regulating ventral patterning. Here we contrast the neural patterning phenotypes of two mouse lines carrying induced mutations in ciliary trafficking genes. We show that a hypomorphic mutation in the gene Tubby-like protein 3 (Tulp3) resulted in a dorsal expansion of MNs consistent with an up-regulation of Shh signaling. Interestingly, patterning defects in Tulp3 mutants were restricted to posterior regions of the spinal cord as patterning was similar to WT in the anterior spinal cord. In contrast, a mutation in the ciliary trafficking gene cytoplasmic dynein 2 heavy chain 1 (Dync2h1), led to a complete loss of MNs in anterior regions of the spinal cord, indicating a strong down-regulation of Shh signaling. However, this severe phenotype was restricted to the cervical region as MNs developed posteriorly. Mutations in cilia trafficking genes affect Shh-dependent signaling in the neural tube differentially along the anterior-posterior (A-P) axis in a process that is not understood.


Subject(s)
Hedgehog Proteins , Neural Tube , Animals , Body Patterning/genetics , Gene Expression Regulation, Developmental , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Intracellular Signaling Peptides and Proteins , Mice , Mutation , Neural Tube/metabolism
14.
Curr Biol ; 29(5): 803-812.e5, 2019 03 04.
Article in English | MEDLINE | ID: mdl-30799240

ABSTRACT

The primary cilium is an organelle essential for cell signaling pathways. One of the most common human genetic diseases is autosomal dominant polycystic kidney disease (ADPKD), which is caused by mutations in the PKD1 or PKD2 genes that encode Polycystin 1 and 2 (PC1/2), transmembrane proteins that translocate to the cilium. Mutations in genes that disrupt ciliogenesis also cause kidney cysts as part of a "ciliopathic" disease spectrum. The molecular mechanisms that link cilia function with renal cystic diseases are not well understood, and the mechanistic relationship between ADPKD and ciliopathic PKD is not known. Here we identify the gene Tubby-like protein-3 (Tulp3) as a key regulator of renal cystic disease from a forward genetic screen in the mouse. Mice homozygous for a hypomorphic missense mutation within the conserved Tubby domain of Tulp3 develop cysts at late embryonic stages, leading to severe postnatal loss of kidney function. In contrast to other ciliopathic disease models, Tulp3 mutations do not affect ciliogenesis. Instead, we demonstrate that Tulp3 is essential for the trafficking of the Joubert syndrome-associated small GTPase Arl13b into kidney cilia. We show that reduction of Pkd1 dosage promotes cystogenesis in the Tulp3 conditional ciliopathic PKD model. However, in an adult model of ADPKD utilizing inducible conditional Pkd1 deletion, concomitant removal of Tulp3 surprisingly ameliorates cystic disease. Therefore, Tulp3 controls distinct ciliary pathways that positively or negatively regulate cystogenesis depending on the cellular context.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Polycystic Kidney Diseases/genetics , Protein Transport , Animals , Female , Male , Mice , Mice, Knockout , Polycystic Kidney Diseases/metabolism
15.
J Morphol ; 280(12): 1865-1870, 2019 12.
Article in English | MEDLINE | ID: mdl-31638290

ABSTRACT

The king-of-the-salmon, Trachipterus altivelis (Lampriformes), has an unusual set of oral jaws which allow it the ability to protrude the entire upper jaw, containing the premaxilla and the maxilla bones, to extreme distances. Here, we provide a short description of the cranial anatomy and mechanism of jaw protrusion in T. altivelis using hand-drawn illustrations (by KF), supplemented by CT-scans. We then place the protrusion abilities of T. altivelis into context by comparing anatomical jaw protrusion with protrusion from other members of the Lampriformes, other unrelated species with highly protrusile jaws, and unrelated species with more stereotypical amounts of jaw protrusion. Through these comparisons we demonstrate that T. altivelis is indeed, capable of some of the most extreme premaxillary protrusion as of yet discovered, even when taking into account the extreme morphological modifications that facilitate said protrusion. That is to say, T. altivelis can protrude the premaxilla farther than one would predict from the length of the ascending process alone.


Subject(s)
Feeding Behavior , Maxilla/anatomy & histology , Salmonidae/anatomy & histology , Animals , Biomechanical Phenomena , Jaw/anatomy & histology , Maxilla/physiology , Salmonidae/physiology
16.
Cell Rep ; 29(11): 3356-3366.e3, 2019 12 10.
Article in English | MEDLINE | ID: mdl-31825820

ABSTRACT

The small GTPase Arl13b is enriched in primary cilia and regulates Sonic hedgehog (Shh) signaling. During neural development, Shh controls patterning and proliferation through a canonical, transcription-dependent pathway that requires the primary cilium. Additionally, Shh controls axon guidance through a non-canonical, transcription-independent pathway whose connection to the primary cilium is unknown. Here we show that inactivation of Arl13b results in defective commissural axon guidance in vivo. In vitro, we demonstrate that Arl13b functions autonomously in neurons for their Shh-dependent guidance response. We detect Arl13b protein in axons and growth cones, far from its well-established ciliary enrichment. To test whether Arl13b plays a non-ciliary function, we used an engineered, cilia-localization-deficient Arl13b variant and found that it was sufficient to mediate Shh axon guidance in vitro and in vivo. Together, these results indicate that, in addition to its ciliary role in canonical Shh signaling, Arl13b plays a cilia-independent role in Shh-mediated axon guidance.


Subject(s)
ADP-Ribosylation Factors/metabolism , Axon Guidance , Cilia/metabolism , Hedgehog Proteins/metabolism , Animals , Cells, Cultured , Growth Cones/metabolism , Mice , Signal Transduction
17.
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
18.
Nat Cell Biol ; 16(7): 663-72, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24952464

ABSTRACT

Mammalian Hedgehog (Hh) signal transduction requires a primary cilium, a microtubule-based organelle, and the Gli-Sufu complexes that mediate Hh signalling, which are enriched at cilia tips. Kif7, a kinesin-4 family protein, is a conserved regulator of the Hh signalling pathway and a human ciliopathy protein. Here we show that Kif7 localizes to the cilium tip, the site of microtubule plus ends, where it limits cilium length and controls cilium structure. Purified recombinant Kif7 binds the plus ends of growing microtubules in vitro, where it reduces the rate of microtubule growth and increases the frequency of microtubule catastrophe. Kif7 is not required for normal intraflagellar transport or for trafficking of Hh pathway proteins into cilia. Instead, a central function of Kif7 in the mammalian Hh pathway is to control cilium architecture and to create a single cilium tip compartment, where Gli-Sufu activity can be correctly regulated.


Subject(s)
Cilia/metabolism , Hedgehog Proteins/metabolism , Kinesins/metabolism , Signal Transduction , Animals , Axoneme/genetics , Axoneme/metabolism , Cell Line , Cells, Cultured , Cilia/chemistry , Fibroblasts/metabolism , HEK293 Cells , Humans , Kinesins/genetics , Mice , Microtubules/metabolism , Mutation , NIH 3T3 Cells , Protein Binding , Protein Transport , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
19.
J Cell Biol ; 197(6): 789-800, 2012 Jun 11.
Article in English | MEDLINE | ID: mdl-22689656

ABSTRACT

Two intraflagellar transport (IFT) complexes, IFT-A and IFT-B, build and maintain primary cilia and are required for activity of the Sonic hedgehog (Shh) pathway. A weak allele of the IFT-A gene, Ift144, caused subtle defects in cilia structure and ectopic activation of the Shh pathway. In contrast, strong loss of IFT-A, caused by either absence of Ift144 or mutations in two IFT-A genes, blocked normal ciliogenesis and decreased Shh signaling. In strong IFT-A mutants, the Shh pathway proteins Gli2, Sufu, and Kif7 localized correctly to cilia tips, suggesting that these pathway components were trafficked by IFT-B. In contrast, the membrane proteins Arl13b, ACIII, and Smo failed to localize to primary cilia in the absence of IFT-A. We propose that the increased Shh activity seen in partial loss-of-function IFT-A mutants may be a result of decreased ciliary ACIII and that the loss of Shh activity in the absence of IFT-A is a result of severe disruptions of cilia structure and membrane protein trafficking.


Subject(s)
Cilia/ultrastructure , Hedgehog Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Proteins/metabolism , Signal Transduction , Animals , Cilia/metabolism , Cytoskeletal Proteins , Flagella/metabolism , Hedgehog Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Kinesins/genetics , Kinesins/metabolism , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Electron , Proteins/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Zinc Finger Protein Gli2
20.
Mech Dev ; 127(7-8): 321-8, 2010.
Article in English | MEDLINE | ID: mdl-20420902

ABSTRACT

Lens regeneration in adult salamanders occurs at the pupillary margin of the mid-dorsal iris where pigmented epithelial cells (PEC) re-enter the cell cycle and transdifferentiate into lens. It is not understood how the injury caused by removal of the lens (lentectomy) in one location is linked to initiating the response in a different spatial location (dorsal iris) and to this particular sector. We propose that the blood provides a link between the localised coagulation and signal transduction pathways that lead to regeneration. A transmembrane protein (tissue factor) is expressed in a striking patch-like domain in the dorsal iris of the newt that localises coagulation specifically to this location, but is not expressed in the axolotl, a related species that does not show thrombin activation after lentectomy and cannot regenerate its lens. Our hypothesis is that tissue factor expression localises the initiation of regeneration through the activation of thrombin and the recruitment of blood cells, leading to local growth factor release. This is the first example of gene expression in a patch of cells that prefigures the location of a regenerative response, and links the immune system with the initiation of a regenerative program.


Subject(s)
Iris/enzymology , Lens, Crystalline/physiology , Regeneration/physiology , Salamandridae/metabolism , Thrombin/metabolism , Thromboplastin/metabolism , Ambystoma mexicanum/metabolism , Amino Acid Sequence , Animals , Cell Transdifferentiation , Enzyme Activation , Fibrin , Lens, Crystalline/cytology , Lens, Crystalline/metabolism , Lens, Crystalline/surgery , Models, Biological , Molecular Sequence Data , Salamandridae/physiology , Thromboplastin/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL