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.
Cells ; 10(12)2021 12 08.
Article in English | MEDLINE | ID: mdl-34943966

ABSTRACT

Werner syndrome (WS) is a rare recessive genetic disease characterized by premature aging. Individuals with this disorder develop normally during childhood, but their physiological conditions exacerbate the aging process in late adolescence. WS is caused by mutation of the human WS gene (WRN), which encodes two main domains, a 3'-5' exonuclease and a 3'-5' helicase. Caenorhabditis elegans expresses human WRN orthologs as two different proteins: MUT-7, which has a 3'-5' exonuclease domain, and C. elegans WRN-1 (CeWRN-1), which has only helicase domains. These unique proteins dynamically regulate olfactory memory in C. elegans, providing insight into the molecular roles of WRN domains in humans. In this review, we specifically focus on characterizing the function of MUT-7 in small interfering RNA (siRNA) synthesis in the cytoplasm and the roles of siRNA in directing nuclear CeWRN-1 loading onto a heterochromatin complex to induce negative feedback regulation. Further studies on the different contributions of the 3'-5' exonuclease and helicase domains in the molecular mechanism will provide clues to the accelerated aging processes in WS.


Subject(s)
Aging, Premature/genetics , Caenorhabditis elegans Proteins/genetics , DNA Helicases/genetics , Exoribonucleases/genetics , Werner Syndrome/genetics , Animals , Caenorhabditis elegans/genetics , Heterochromatin/genetics , Humans , Mutation/genetics , RNA, Small Interfering/genetics , Werner Syndrome/pathology
2.
Elife ; 102021 03 01.
Article in English | MEDLINE | ID: mdl-33646120

ABSTRACT

Caenorhabditis elegans expresses human Werner syndrome protein (WRN) orthologs as two distinct proteins: MUT-7, with a 3'-5' exonuclease domain, and CeWRN-1, with helicase domains. How these domains cooperate remains unclear. Here, we demonstrate the different contributions of MUT-7 and CeWRN-1 to 22G small interfering RNA (siRNA) synthesis and the plasticity of neuronal signaling. MUT-7 acts specifically in the cytoplasm to promote siRNA biogenesis and in the nucleus to associate with CeWRN-1. The import of siRNA by the nuclear Argonaute NRDE-3 promotes the loading of the heterochromatin-binding protein HP1 homolog HPL-2 onto specific loci. This heterochromatin complex represses the gene expression of the guanylyl cyclase ODR-1 to direct olfactory plasticity in C. elegans. Our findings suggest that the exonuclease and helicase domains of human WRN may act in concert to promote RNA-dependent loading into a heterochromatin complex, and the failure of this entire process reduces plasticity in postmitotic neurons.


Subject(s)
DNA Helicases/metabolism , Werner Syndrome Helicase/metabolism , Werner Syndrome/metabolism , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/metabolism , DNA Helicases/genetics , Exonucleases , Heterochromatin , Neuronal Plasticity , RNA, Small Interfering/metabolism , Werner Syndrome/genetics , Werner Syndrome Helicase/genetics
3.
PLoS One ; 11(4): e0154723, 2016.
Article in English | MEDLINE | ID: mdl-27124114

ABSTRACT

Spinal muscular atrophy (SMA) is an autosomal recessive motor neuron disease caused by deficiency of the survival of motor neuron (SMN) protein, which leads to synaptic defects and spinal motor neuron death. Neuromuscular junction (NMJ) abnormalities have been found to be involved in SMA pathogenesis in the SMNΔ7 SMA mouse model. However, whether similar NMJ pathological findings present in another commonly used mouse model, the Taiwanese SMA mouse, has not been fully investigated. To examine the NMJs of the Taiwanese severe SMA mouse model (Smn-/-; SMN2tg/0), which is characterized by severe phenotype and death before postnatal day (P) 9, we investigated 25 axial and appendicular muscles from P1 to P9. We labelled the muscles with anti-neurofilament and anti-synaptophysin antibodies for nerve terminals and α-bungarotoxin for acetylcholine receptors (AChRs). We found that severe NMJ denervation (<50% fully innervated endplates) selectively occurred in the flexor digitorum brevis 2 and 3 (FDB-2/3) muscles from P5, and an increased percentage of fully denervated endplates correlated with SMA progression. Furthermore, synaptophysin signals were absent at the endplate compared to control littermate mice, suggesting that vesicle transport might only be affected at the end stage. Subsequently, we treated the Taiwanese severe SMA mice with morpholino (MO) antisense oligonucleotides (80 µg/g) via subcutaneous injection at P0. We found that MO significantly reversed the NMJ denervation in FDB-2/3 muscles and extended the survival of Taiwanese severe SMA mice. We conclude that early NMJ denervation in the FDB-2/3 muscles of Taiwanese severe SMA mice can be reversed by MO treatment. The FDB-2/3 muscles of Taiwanese severe SMA mice provide a very sensitive platform for assessing the effectiveness of drug treatments in SMA preclinical studies.


Subject(s)
Morpholinos/therapeutic use , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/therapy , Nerve Degeneration/therapy , Neuromuscular Junction/pathology , Oligonucleotides, Antisense/therapeutic use , Animals , Disease Models, Animal , Mice , Mice, Knockout , Motor Neurons/pathology , Muscle, Skeletal/innervation , Muscular Atrophy, Spinal/pathology , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Neuromuscular Junction/genetics , Survival of Motor Neuron 1 Protein/genetics
4.
Hum Mol Genet ; 23(18): 4945-59, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-24821701

ABSTRACT

Fragile X-associated tremor/ataxia syndrome (FXTAS) is a severe neurodegenerative disorder that affects carriers of premutation CGG-repeat expansion alleles of the fragile X mental retardation 1 (FMR1) gene; current evidence supports a causal role of the expanded CGG repeat within the FMR1 mRNA in the pathogenesis of FXTAS. Though the mRNA has been observed to induce cellular toxicity in FXTAS, the mechanisms are unclear. One common neurophysiological characteristic of FXTAS patients is their inability to properly attenuate their response to an auditory stimulus upon receipt of a small pre-stimulus. Therefore, to gain genetic and cell biological insight into FXTAS, we examined the effect of expanded CGG repeats on the plasticity of the olfactory response of the genetically tractable nematode, Caenorhabditis elegans (C. elegans). While C. elegans is innately attracted to odors, this response can be downregulated if the odor is paired with starvation. We found that expressing expanded CGG repeats in olfactory neurons interfered with this plasticity without affecting either the innate odor-seeking response or the olfactory neuronal morphology. Interrogation of three RNA regulatory pathways indicated that the expanded CGG repeats act via the C. elegans microRNA (miRNA)-specific Argonaute ALG-2 to diminish olfactory plasticity. This observation suggests that the miRNA-Argonaute pathway may play a pathogenic role in subverting neuronal function in FXTAS.


Subject(s)
Butanones/pharmacology , Caenorhabditis elegans/physiology , Fragile X Mental Retardation Protein/genetics , Olfactory Receptor Neurons/metabolism , Sensory Receptor Cells/metabolism , Animals , Animals, Genetically Modified , Argonaute Proteins/genetics , Ataxia/genetics , Ataxia/pathology , Caenorhabditis elegans/genetics , Disease Models, Animal , Fragile X Syndrome/genetics , Fragile X Syndrome/pathology , Humans , Neuronal Plasticity , Smell , Tremor/genetics , Tremor/pathology , Trinucleotide Repeat Expansion
5.
Cell ; 154(5): 1010-1022, 2013 Aug 29.
Article in English | MEDLINE | ID: mdl-23993094

ABSTRACT

Most eukaryotic cells express small regulatory RNAs. The purpose of one class, the somatic endogenous siRNAs (endo-siRNAs), remains unclear. Here, we show that the endo-siRNA pathway promotes odor adaptation in C. elegans AWC olfactory neurons. In adaptation, the nuclear Argonaute NRDE-3, which acts in AWC, is loaded with siRNAs targeting odr-1, a gene whose downregulation is required for adaptation. Concomitant with increased odr-1 siRNA in AWC, we observe increased binding of the HP1 homolog HPL-2 at the odr-1 locus in AWC and reduced odr-1 mRNA in adapted animals. Phosphorylation of HPL-2, an in vitro substrate of the EGL-4 kinase that promotes adaption, is necessary and sufficient for behavioral adaptation. Thus, environmental stimulation amplifies an endo-siRNA negative feedback loop to dynamically repress cognate gene expression and shape behavior. This class of siRNA may act broadly as a rheostat allowing prolonged stimulation to dampen gene expression and promote cellular memory formation. PAPERFLICK:


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/physiology , Down-Regulation , Guanylate Cyclase/genetics , RNA Interference , Sensory Receptor Cells/metabolism , Adaptation, Physiological , Animals , Butanones/chemistry , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Odorants , Phosphorylation , RNA, Helminth/metabolism , RNA, Small Interfering/metabolism , RNA-Binding Proteins/metabolism
6.
PLoS Genet ; 9(7): e1003619, 2013.
Article in English | MEDLINE | ID: mdl-23874221

ABSTRACT

Signaling levels within sensory neurons must be tightly regulated to allow cells to integrate information from multiple signaling inputs and to respond to new stimuli. Herein we report a new role for the cGMP-dependent protein kinase EGL-4 in the negative regulation of G protein-coupled nociceptive chemosensory signaling. C. elegans lacking EGL-4 function are hypersensitive in their behavioral response to low concentrations of the bitter tastant quinine and exhibit an elevated calcium flux in the ASH sensory neurons in response to quinine. We provide the first direct evidence for cGMP/PKG function in ASH and propose that ODR-1, GCY-27, GCY-33 and GCY-34 act in a non-cell-autonomous manner to provide cGMP for EGL-4 function in ASH. Our data suggest that activated EGL-4 dampens quinine sensitivity via phosphorylation and activation of the regulator of G protein signaling (RGS) proteins RGS-2 and RGS-3, which in turn downregulate Gα signaling and behavioral sensitivity.


Subject(s)
Behavior, Animal/physiology , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , Cyclic GMP-Dependent Protein Kinases/genetics , Cyclic GMP/metabolism , Animals , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Phosphorylation , RGS Proteins/genetics , RGS Proteins/metabolism , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/physiology , Signal Transduction/genetics
7.
Proc Natl Acad Sci U S A ; 107(13): 6016-21, 2010 Mar 30.
Article in English | MEDLINE | ID: mdl-20220099

ABSTRACT

To navigate a complex and changing environment, an animal's sensory neurons must continually adapt to persistent cues while remaining responsive to novel stimuli. Long-term exposure to an inherently attractive odor causes Caenorhabditis elegans to ignore that odor, a process termed odor adaptation. Odor adaptation is likely to begin within the sensory neuron, because it requires factors that act within these cells at the time of odor exposure. The process by which an olfactory sensory neuron makes a decisive shift over time from a receptive state to a lasting unresponsive one remains obscure. In C. elegans, adaptation to odors sensed by the AWC pair of olfactory neurons requires the cGMP-dependent protein kinase EGL-4. Using a fully functional, GFP-tagged EGL-4, we show here that prolonged odor exposure sends EGL-4 into the nucleus of the stimulated AWC neuron. This odor-induced nuclear translocation correlates temporally with the stable dampening of chemotaxis that is indicative of long-term adaptation. Long-term adaptation requires cGMP binding residues as well as an active EGL-4 kinase. We show here that EGL-4 nuclear accumulation is both necessary and sufficient to induce long-lasting odor adaptation. After it is in the AWC nucleus, EGL-4 decreases the animal's responsiveness to AWC-sensed odors by acting downstream of the primary sensory transduction. Thus, the EGL-4 protein kinase acts as a sensor that integrates odor signaling over time, and its nuclear translocation is an instructive switch that allows the animal to ignore persistent odors.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/physiology , Cyclic GMP-Dependent Protein Kinases/physiology , Smell/physiology , Active Transport, Cell Nucleus , Adaptation, Physiological , Animals , Animals, Genetically Modified , Behavior, Animal/physiology , Binding Sites , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinases/chemistry , Cyclic GMP-Dependent Protein Kinases/genetics , Gene Expression , Genes, Helminth , Models, Neurological , Odorants , Olfactory Receptor Neurons/physiology , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction
8.
Biochemistry ; 44(30): 10396-405, 2005 Aug 02.
Article in English | MEDLINE | ID: mdl-16042417

ABSTRACT

The human cellular factor (HCF) is a multidomain protein that is implicated in processes of cell cycle progression, and it is recruited into a multicomponent assembly that triggers the expression of the herpes simplex virus genome. The amino-terminal domain of HCF has been proposed to form a "kelch" type beta-propeller fold, and the carboxy-terminal domain contains a repeat of a fibronectin-like motif. We describe the expression, purification, and characterization of the domains from the human HCF and of the full-length HCF from Caenorhabditis elegans. The purified recombinant C. elegans HCF can substitute for the human HCF in efficiently forming a multiprotein complex on a herpes simplex virus promoter element. As noted in earlier studies, a segment of human HCF encompassing the human kelch domain forms a stable complex on a viral promoter element. The purified fibronectin domain can also be recruited into this complex, but not into the stable complex formed with the minimal kelch domain. These results suggest that the fibronectin domain can interact with HCF in the transcriptional activating complex and that the association requires a region outside the putative beta-propeller.


Subject(s)
Caenorhabditis elegans Proteins/isolation & purification , Transcription Factors/isolation & purification , Animals , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cell Line , DNA-Binding Proteins/genetics , Drosophila , Fibronectins/biosynthesis , Fibronectins/genetics , Fibronectins/isolation & purification , Herpes Simplex Virus Protein Vmw65/physiology , Host Cell Factor C1 , Humans , Microfilament Proteins/genetics , Microfilament Proteins/physiology , Octamer Transcription Factor-1 , Peptide Fragments/biosynthesis , Peptide Fragments/genetics , Peptide Fragments/isolation & purification , Protein Folding , Protein Structure, Tertiary/genetics , Protein Transport/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Solubility , Subcellular Fractions/chemistry , Subcellular Fractions/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...