Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS Genet ; 18(5): e1010209, 2022 05.
Article in English | MEDLINE | ID: mdl-35604893

ABSTRACT

In the nervous system, the specific identity of a neuron is established and maintained by terminal selector transcription factors that directly activate large batteries of terminal differentiation genes and positively regulate their own expression via feedback loops. However, how this is achieved in a reliable manner despite noise in gene expression, genetic variability or environmental perturbations remains poorly understood. We addressed this question using the AIY cholinergic interneurons of C. elegans, whose specification and differentiation network is well characterized. Via a genetic screen, we found that a loss of function of PRC1 chromatin factors induces a stochastic loss of AIY differentiated state in a small proportion of the population. PRC1 factors act directly in the AIY neuron and independently of PRC2 factors. By quantifying mRNA and protein levels of terminal selector transcription factors in single neurons, using smFISH and CRISPR tagging, we observed that, in PRC1 mutants, terminal selector expression is still initiated during embryonic development but the level is reduced, and expression is subsequently lost in a stochastic manner during maintenance phase in part of the population. We also observed variability in the level of expression of terminal selectors in wild type animals and, using correlation analysis, established that this noise comes from both intrinsic and extrinsic sources. Finally, we found that PRC1 factors increase the resistance of AIY neuron fate to environmental stress, and also secure the terminal differentiation of other neuron types. We propose that PRC1 factors contribute to the consistency of neuronal cell fate specification and maintenance by protecting neurons against noise and perturbations in their differentiation program.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cell Differentiation/genetics , Chromatin/genetics , Chromatin/metabolism , Gene Expression Regulation, Developmental , Neurons/metabolism , Transcription Factors/metabolism
2.
Nucleic Acids Res ; 48(16): 9019-9036, 2020 09 18.
Article in English | MEDLINE | ID: mdl-32725242

ABSTRACT

Cilia assembly is under strict transcriptional control during animal development. In vertebrates, a hierarchy of transcription factors (TFs) are involved in controlling the specification, differentiation and function of multiciliated epithelia. RFX TFs play key functions in the control of ciliogenesis in animals. Whereas only one RFX factor regulates ciliogenesis in C. elegans, several distinct RFX factors have been implicated in this process in vertebrates. However, a clear understanding of the specific and redundant functions of different RFX factors in ciliated cells remains lacking. Using RNA-seq and ChIP-seq approaches we identified genes regulated directly and indirectly by RFX1, RFX2 and RFX3 in mouse ependymal cells. We show that these three TFs have both redundant and specific functions in ependymal cells. Whereas RFX1, RFX2 and RFX3 occupy many shared genomic loci, only RFX2 and RFX3 play a prominent and redundant function in the control of motile ciliogenesis in mice. Our results provide a valuable list of candidate ciliary genes. They also reveal stunning differences between compensatory processes operating in vivo and ex vivo.


Subject(s)
Cilia/physiology , Ependyma/cytology , Gene Expression Regulation, Developmental , Regulatory Factor X Transcription Factors/physiology , Regulatory Factor X1/physiology , Animals , Cilia/genetics , Mice , Mice, Inbred C57BL
3.
Nat Commun ; 9(1): 1741, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29717108

ABSTRACT

Many membranes must merge during cellular trafficking, but fusion and fission events initiating at exoplasmic (non-cytosolic) membrane surfaces are not well understood. Here we show that the C. elegans cell-cell fusogen anchor-cell fusion failure 1 (AFF-1) is required for membrane trafficking events during development of a seamless unicellular tube. EGF-Ras-ERK signaling upregulates AFF-1 expression in the excretory duct tube to promote tube auto-fusion and subsequent lumen elongation. AFF-1 is required for scission of basal endocytic compartments and for apically directed exocytosis to extend the apical membrane. Lumen elongation also requires the transcytosis factor Rab11, but occurs independently of dynamin and clathrin. These results support a transcytosis model of seamless tube lumen growth and show that cell-cell fusogens also can play roles in intracellular membrane trafficking events.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Cell Fusion , Endocytosis/physiology , Animals , Animals, Genetically Modified , Caenorhabditis elegans/genetics , Epidermal Growth Factor/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Protein Transport , Signal Transduction , ras Proteins/metabolism
4.
Semin Cell Dev Biol ; 60: 136-145, 2016 12.
Article in English | MEDLINE | ID: mdl-27436685

ABSTRACT

Cells adopt specific shapes that are necessary for specific functions. For example, some neurons extend elaborate arborized dendrites that can contact multiple targets. Epithelial and endothelial cells can form tiny seamless unicellular tubes with an intracellular lumen. Recent advances showed that cells can auto-fuse to acquire those specific shapes. During auto-fusion, a cell merges two parts of its own plasma membrane. In contrast to cell-cell fusion or macropinocytic fission, which result in the merging or formation of two separate membrane bound compartments, auto-fusion preserves one compartment, but changes its shape. The discovery of auto-fusion in C. elegans was enabled by identification of specific protein fusogens, EFF-1 and AFF-1, that mediate cell-cell fusion. Phenotypic characterization of eff-1 and aff-1 mutants revealed that fusogen-mediated fusion of two parts of the same cell can be used to sculpt dendritic arbors, reconnect two parts of an axon after injury, or form a hollow unicellular tube. Similar auto-fusion events recently were detected in vertebrate cells, suggesting that auto-fusion could be a widely used mechanism for shaping neurons and tubes.


Subject(s)
Endothelial Cells/physiology , Epithelial Cells/physiology , Neurons/cytology , Animals , Caenorhabditis elegans/cytology , Cell Fusion , Morphogenesis , Nerve Regeneration
5.
Mol Biol Cell ; 25(8): 1276-86, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24554765

ABSTRACT

Cilia play major functions in physiology and development, and ciliary dysfunctions are responsible for several diseases in humans called ciliopathies. Cilia motility is required for cell and fluid propulsion in organisms. In humans, cilia motility deficiencies lead to primary ciliary dyskinesia, with upper-airways recurrent infections, left-right asymmetry perturbations, and fertility defects. In Drosophila, we identified hemingway (hmw) as a novel component required for motile cilia function. hmw encodes a 604-amino acid protein characterized by a highly conserved coiled-coil domain also found in the human orthologue, KIAA1430. We show that HMW is conserved in species with motile cilia and that, in Drosophila, hmw is expressed in ciliated sensory neurons and spermatozoa. We created hmw-knockout flies and found that they are hearing impaired and male sterile. hmw is implicated in the motility of ciliated auditory sensory neurons and, in the testis, is required for elongation and maintenance of sperm flagella. Because HMW is absent from mature flagella, we propose that HMW is not a structural component of the motile axoneme but is required for proper acquisition of motile properties. This identifies HMW as a novel, evolutionarily conserved component necessary for motile cilium function and flagella assembly.


Subject(s)
Cilia/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Sperm Tail/metabolism , Amino Acid Sequence , Animals , Animals, Genetically Modified , Binding Sites , Ciliary Motility Disorders , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila Proteins/biosynthesis , Drosophila Proteins/genetics , Gene Knockout Techniques , Hearing Loss/genetics , Infertility, Male , Male , Molecular Sequence Data , Promoter Regions, Genetic , Protein Structure, Tertiary , Regulatory Factor X Transcription Factors , Sequence Alignment , Spermatogenesis/physiology , Transcription Factors/genetics , Transcription Factors/metabolism
6.
Hum Mol Genet ; 23(3): 563-77, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24067530

ABSTRACT

Cilia are evolutionarily conserved organelles endowed with essential physiological and developmental functions. In humans, disruption of cilia motility or signaling leads to complex pleiotropic genetic disorders called ciliopathies. Cilia motility requires the assembly of multi-subunit motile components such as dynein arms, but mechanisms underlying their assembly pathway and transport into the axoneme are still largely unknown. We identified a previously uncharacterized coiled-coil domain containing protein CCDC151, which is evolutionarily conserved in motile ciliated species and shares ancient features with the outer dynein arm-docking complex 2 of Chlamydomonas. In Drosophila, we show that CG14127/CCDC151 is associated with motile intraflagellar transport (IFT)-dependent cilia and required for geotaxis behavior of adult flies. In zebrafish, Ccdc151 is expressed in tissues with motile cilia, and morpholino-induced depletion of Ccdc151 leads to left-right asymmetry defects and kidney cysts. We demonstrate that Ccdc151 is required for proper motile function of cilia in the Kupffer's vesicle and in the pronephros by controlling dynein arm assembly, showing that Ccdc151 is a novel player in the control of IFT-dependent dynein arm assembly in animals. However, we observed that CCDC151 is also implicated in other cellular functions in vertebrates. In zebrafish, ccdc151 is involved in proper orientation of cell divisions in the pronephros and genetically interacts with prickle1 in this process. Furthermore, knockdown experiments in mammalian cells demonstrate that CCDC151 is implicated in the regulation of primary cilium length. Hence, CCDC151 is required for motile cilia function in animals but has acquired additional non-motile functions in vertebrates.


Subject(s)
Cilia/metabolism , Drosophila Proteins/metabolism , Zebrafish Proteins/metabolism , Amino Acid Sequence , Animals , Animals, Genetically Modified , Axoneme/metabolism , Biological Transport , Cell Polarity , Cilia/genetics , Conserved Sequence , Drosophila/embryology , Drosophila/genetics , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Embryo, Nonmammalian/cytology , Ependyma/cytology , Flagella/metabolism , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Kidney Diseases/genetics , Kidney Diseases/pathology , Mice , Phylogeny , Protein Structure, Tertiary , Proteins/chemistry , Proteins/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics
7.
J Cell Biol ; 197(2): 313-25, 2012 Apr 16.
Article in English | MEDLINE | ID: mdl-22508513

ABSTRACT

Centriole-to-basal body conversion, a complex process essential for ciliogenesis, involves the progressive addition of specific proteins to centrioles. CHIBBY (CBY) is a coiled-coil domain protein first described as interacting with ß-catenin and involved in Wg-Int (WNT) signaling. We found that, in Drosophila melanogaster, CBY was exclusively expressed in cells that require functional basal bodies, i.e., sensory neurons and male germ cells. CBY was associated with the basal body transition zone (TZ) in these two cell types. Inactivation of cby led to defects in sensory transduction and in spermatogenesis. Loss of CBY resulted in altered ciliary trafficking into neuronal cilia, irregular deposition of proteins on spermatocyte basal bodies, and, consequently, distorted axonemal assembly. Importantly, cby(1/1) flies did not show Wingless signaling defects. Hence, CBY is essential for normal basal body structure and function in Drosophila, potentially through effects on the TZ. The function of CBY in WNT signaling in vertebrates has either been acquired during vertebrate evolution or lost in Drosophila.


Subject(s)
Carrier Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Nuclear Proteins/metabolism , Sensory Receptor Cells/metabolism , Spermatozoa/metabolism , Wnt1 Protein/metabolism , Amino Acid Sequence , Animals , Carrier Proteins/biosynthesis , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cells, Cultured , Centrioles/metabolism , Cilia/metabolism , DNA-Binding Proteins/metabolism , Drosophila Proteins/biosynthesis , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Infertility, Male , Male , Mice , Molecular Sequence Data , Nuclear Proteins/biosynthesis , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Protein Transport , Regulatory Factor X Transcription Factors , Transcription Factors/metabolism , Wnt Signaling Pathway
8.
Biol Cell ; 102(9): 499-513, 2010 Jul 09.
Article in English | MEDLINE | ID: mdl-20690903

ABSTRACT

Cilia and flagella have essential functions in a wide range of organisms. Cilia assembly is dynamic during development and different types of cilia are found in multicellular organisms. How this dynamic and specific assembly is regulated remains an important question in cilia biology. In metazoans, the regulation of the overall expression level of key components necessary for cilia assembly or function is an important way to achieve ciliogenesis control. The FOXJ1 (forkhead box J1) and RFX (regulatory factor X) family of transcription factors have been shown to be important players in controlling ciliary gene expression. They fulfill a complementary and synergistic function by regulating specific and common target genes. FOXJ1 is essential to allow for the assembly of motile cilia in vertebrates through the regulation of genes specific to motile cilia or necessary for basal body apical transport, whereas RFX proteins are necessary to assemble both primary and motile cilia in metazoans, in particular, by regulating genes involved in intraflagellar transport. Recently, different transcription factors playing specific roles in cilia biogenesis and physiology have also been discovered. All these factors are subject to complex regulation to allow for the dynamic and specific regulation of ciliogenesis in metazoans.


Subject(s)
Cilia/genetics , Cilia/physiology , Gene Expression Regulation, Developmental , Morphogenesis/genetics , Transcription Factors/physiology , Transcription, Genetic , Animals , Cell Differentiation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/physiology , Humans , Regulatory Factor X Transcription Factors , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...