Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Front Plant Sci ; 13: 928949, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35845691

RESUMO

Wheat streak mosaic virus (WSMV) is an economically important viral pathogen that threatens global wheat production, particularly in the Great Plains of the United States. The Wsm2 locus confers resistance to WSMV and has been widely deployed in common wheat varieties adapted to this region. Characterizing the underlying causative genetic variant would contribute to our understanding of viral resistance mechanisms in wheat and aid the development of perfect markers for breeding. In this study, linkage mapping in a doubled-haploid (DH) mapping population confirmed Wsm2 as a major locus conferring WSMV resistance in wheat. The Wsm2 flanking markers were mapped to a 4.0 Mbp region at the distal end of chromosome 3BS containing 142 candidate genes. Eight haplotypes were identified from seventeen wheat genotypes collected from different agroecological zones, indicating that Wsm2 lies in a dynamic region of the genome with extensive structural variation and that it is likely a rare allele in most available genome assemblies of common wheat varieties. Exome sequencing of the variety "Snowmass", which carries Wsm2, revealed several loss-of-function mutations and copy number variants in the 142 candidate genes within the Wsm2 interval. Six of these genes are differentially expressed in "Snowmass" compared to "Antero," a variety lacking Wsm2, including a gene that encodes a nucleotide-binding site leucine-rich repeat (NBS-LRR) type protein with homology to RPM1. A de novo assembly of unmapped RNA-seq reads identified nine transcripts expressed only in "Snowmass," three of which are also induced in response to WSMV inoculation. This study sheds light on the variation underlying Wsm2 and provides a list of candidate genes for subsequent validation.

2.
Biochem Biophys Res Commun ; 527(1): 124-130, 2020 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-32446355

RESUMO

Resistance to Pseudomonas syringae pv. Maculicola 1 (RPM1) is a crucial immune receptor conferring plant enhanced resistance to pathogenic bacteria. RPM1-interacting protein 13 (RIN13) enhances RPM1-mediated disease resistance through interacting with the central domain of RPM1 in Arabidopsis, while the underlying mechanism remains elusive. Here, we report the subcellular localization and function of RIN13 using the Nicotiana benthamiana (N. benthamiana) transient expression system. Our results showed that RIN13 is exclusively localized in the nucleus, and RIN13 (231-300) fragment is responsible for its nuclear localization. Transient expression of RIN13 in N. benthamiana leaves can accelerate leaf senescence and cell death, and affect the activities of ROS-scavenging enzymes, and the C-terminus of RIN13 is crucial for its function. Furthermore, we identified a RIN13-interacting protein, Auxin Response Factor 1 (ARF1), and found that similar to RIN13, ARF1 can also promote leaf senescence and cell death. In addition, expression of RIN13 in N. benthamiana leaves can facilitate the translocation of ARF1 into the nucleus. Collectively, our study revealed a possible mechanism of RIN13 in accelerating leaf senescence and cell death by changing the subcellular localization of ARF1.


Assuntos
Proteínas de Arabidopsis/metabolismo , Arabidopsis/metabolismo , Núcleo Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Fatores de Transcrição/metabolismo , Arabidopsis/citologia , Arabidopsis/genética , Proteínas de Arabidopsis/análise , Proteínas de Arabidopsis/genética , Morte Celular , Núcleo Celular/genética , Proteínas de Ligação a DNA/análise , Proteínas de Ligação a DNA/genética , Resistência à Doença , Expressão Gênica , Doenças das Plantas/microbiologia , Pseudomonas syringae/fisiologia , Nicotiana/citologia , Nicotiana/genética , Nicotiana/metabolismo , Fatores de Transcrição/análise , Fatores de Transcrição/genética
3.
Elife ; 82019 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-30652969

RESUMO

Synapse formation is comprised of target cell recognition, synapse assembly, and synapse maintenance. Maintaining established synaptic connections is essential for generating functional circuitry and synapse instability is a hallmark of neurodegenerative disease. While many molecules impact synapse formation generally, we know little about molecules that affect synapse maintenance in vivo. Using genetics and developmental time course analysis in C.elegans, we show that the α-tubulin acetyltransferase ATAT-2 and the signaling hub RPM-1 are required presynaptically to maintain stable synapses. Importantly, the enzymatic acetyltransferase activity of ATAT-2 is required for synapse maintenance. Our analysis revealed that RPM-1 is a hub in a genetic network composed of ATAT-2, PTRN-1 and DLK-1. In this network, ATAT-2 functions independent of the DLK-1 MAPK and likely acts downstream of RPM-1. Thus, our study reveals an important role for tubulin acetyltransferase activity in presynaptic maintenance, which occurs via the RPM-1/ATAT-2 pathway.


Assuntos
Acetiltransferases/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Terminações Pré-Sinápticas/fisiologia , Transdução de Sinais , Tubulina (Proteína)/metabolismo , Animais , Aprendizagem , Sistema de Sinalização das MAP Quinases , Microtúbulos/efeitos dos fármacos , Microtúbulos/metabolismo
4.
Development ; 145(18)2018 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-30093552

RESUMO

Axons must correctly reach their targets for proper nervous system function, although we do not fully understand the underlying mechanism, particularly for the first 'pioneer' axons. In C. elegans, AVG is the first neuron to extend an axon along the ventral midline, and this pioneer axon facilitates the proper extension and guidance of follower axons that comprise the ventral nerve cord. Here, we show that the ubiquitin ligase RPM-1 prevents the overgrowth of the AVG axon by repressing the activity of the DLK-1/p38 MAPK pathway. Unlike in damaged neurons, where this pathway activates CEBP-1, we find that RPM-1 and the DLK-1 pathway instead regulate the response to extracellular Wnt cues in developing AVG axons. The Wnt LIN-44 promotes the posterior growth of the AVG axon. In the absence of RPM-1 activity, AVG becomes responsive to a different Wnt, EGL-20, through a mechanism that appears to be independent of canonical Fz-type receptors. Our results suggest that RPM-1 and the DLK-1 pathway regulate axon guidance and growth by preventing Wnt signaling crosstalk.


Assuntos
Axônios/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/embriologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Animais , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Proteínas de Caenorhabditis elegans/genética , Diferenciação Celular/genética , Proliferação de Células/genética , Glicoproteínas/metabolismo , Fatores de Troca do Nucleotídeo Guanina/genética , MAP Quinase Quinase Quinases/genética , Neurônios/metabolismo , Proteínas Wnt/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
5.
Plant Signal Behav ; 13(5): e1473666, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29969363

RESUMO

Considering the crude methods used to control phytoplasma diseases, a deeper knowledge on the defence mechanisms recruited by the plant to face phytoplasma invasion is required. Recently, we demonstrated that Arabidopsis mutants lacking AtSEOR1 gene showed a low phytoplasma titre. In wild type plants AtSEOR1 and AtSEOR2 are tied in filamentous proteins. Knockout of the AtSEOR1 gene may pave the way for an involvement of free AtSEOR2 proteins in defence mechanisms. Among the proteins conferring resistance against pathogenic bacteria, AtRPM1-interacting protein has been found to interact with AtSEOR2 in a high-quality, matrix-based yeast-two hybrid assay. For this reason, we investigated the expression levels of Arabidopsis AtRIN4, and the associated AtRPM1 and AtRPS2 genes in healthy and Chrysanthemum yellows-infected wild-type and Atseor1ko lines.


Assuntos
Proteínas de Arabidopsis/metabolismo , Arabidopsis/metabolismo , Phytoplasma/metabolismo , Proteínas de Plantas/metabolismo , Proteínas de Arabidopsis/genética , Doenças das Plantas/genética , Transdução de Sinais
6.
J Biol Chem ; 293(36): 13897-13909, 2018 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-29997255

RESUMO

PHR (PAM/Highwire/RPM-1) proteins are conserved RING E3 ubiquitin ligases that function in developmental processes, such as axon termination and synapse formation, as well as axon degeneration. At present, our understanding of how PHR proteins form ubiquitin ligase complexes remains incomplete. Although genetic studies indicate NMNAT2 is an important mediator of PHR protein function in axon degeneration, it remains unknown how PHR proteins inhibit NMNAT2. Here, we decipher the biochemical basis for how the human PHR protein PAM, also called MYCBP2, forms a noncanonical Skp/Cullin/F-box (SCF) complex that contains the F-box protein FBXO45 and SKP1 but lacks CUL1. We show FBXO45 does not simply function in substrate recognition but is important for assembly of the PAM/FBXO45/SKP1 complex. Interestingly, we demonstrate a novel role for SKP1 as an auxiliary component of the target recognition module that enhances binding of FBXO45 to NMNAT2. Finally, we provide biochemical evidence that PAM polyubiquitinates NMNAT2 and regulates NMNAT2 protein stability and degradation by the proteasome.


Assuntos
Amidina-Liases/química , Oxigenases de Função Mista/química , Nicotinamida-Nucleotídeo Adenililtransferase/química , Proteínas Ligases SKP Culina F-Box/química , Ubiquitinação , Proteínas Adaptadoras de Transdução de Sinal , Animais , Caenorhabditis elegans , Proteínas F-Box/metabolismo , Humanos , Complexos Multiproteicos/química , Complexos Multiproteicos/fisiologia , Nicotinamida-Nucleotídeo Adenililtransferase/metabolismo , Ligação Proteica , Proteínas Quinases Associadas a Fase S , Proteínas Ligases SKP Culina F-Box/fisiologia , Ubiquitina-Proteína Ligases
7.
Front Plant Sci ; 9: 1991, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30713545

RESUMO

RPM1 is a plant immune receptor that specially recognizes pathogen-released effectors to activate effector-triggered immunity (ETI) in Arabidopsis thaliana. RPM1 triggers ETI and hypersensitive response (HR) for disease resistance. Previous reports indicated that Phospholipase D (PLD) positively regulated RPM1-mediated HR. However, single, double, and triple pld knock-out mutants of 12 members of the PLD family in A. thaliana did not show suppressed RPM1-mediated HR, indicating the functional redundancy among PLD members. In this study, we revealed that PLD could negatively regulate the function of RPM1. We found that RPM1 interacted with PLDδ, but did not interact with PLDß1, PLDß2, and PLDγ3. Overexpression of PLDδ conducted to a reduction of protein level and corresponding activity of RPM1. We found that abscisic acid (ABA) reduced the protein level of RPM1, and the ABA-induced RPM1 reduction required PLD activity and PLD-derived phosphatidic acid (PA). Our study shows that PLD plays both negative and positive roles regulating the protein level and activity of RPM1 during stress responses in plants. PLD proteins are regulating points to integrate the abiotic and biotic responses of plants.

8.
Development ; 144(24): 4658-4672, 2017 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-29084805

RESUMO

Axon termination is essential for efficient and accurate nervous system construction. At present, relatively little is known about how growth cone collapse occurs prior to axon termination in vivo Using the mechanosensory neurons of C. elegans, we found collapse prior to axon termination is protracted, with the growth cone transitioning from a dynamic to a static state. Growth cone collapse prior to termination is facilitated by the signaling hub RPM-1. Given the prominence of the cytoskeleton in growth cone collapse, we assessed the relationship between RPM-1 and regulators of actin dynamics and microtubule stability. Our results reveal several important findings about how axon termination is orchestrated: (1) RPM-1 functions in parallel to RHO-1 and CRMP/UNC-33, but is suppressed by the Rac isoform MIG-2; (2) RPM-1 opposes the function of microtubule stabilizers, including tubulin acetyltransferases; and (3) genetic epistasis suggests the microtubule-stabilizing protein Tau/PTL-1 potentially inhibits RPM-1. These findings provide insight into how growth cone collapse is regulated during axon termination in vivo, and suggest that RPM-1 signaling destabilizes microtubules to facilitate growth cone collapse and axon termination.


Assuntos
Axônios/fisiologia , Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/embriologia , Cones de Crescimento/fisiologia , Fatores de Troca do Nucleotídeo Guanina/genética , Microtúbulos/fisiologia , Acetiltransferases/metabolismo , Citoesqueleto de Actina/metabolismo , Animais , Animais Geneticamente Modificados , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Fatores de Crescimento Neural/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Transdução de Sinais , Proteínas rac de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo
9.
Dev Biol ; 429(1): 240-248, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28673818

RESUMO

The 3' untranslated regions (3' UTRs) of mRNAs mediate post-transcriptional regulation of genes in many biological processes. Cis elements in 3' UTRs can interact with RNA-binding factors in sequence-specific or structure-dependent manners, enabling regulation of mRNA stability, translation, and localization. Caenorhabditis elegans CEBP-1 is a conserved transcription factor of the C/EBP family, and functions in diverse contexts, from neuronal development and axon regeneration to organismal growth. Previous studies revealed that the levels of cebp-1 mRNA in neurons depend on its 3' UTR and are also negatively regulated by the E3 ubiquitin ligase RPM-1. Here, by systematically dissecting cebp-1's 3' UTR, we test the roles of specific cis elements in cebp-1 expression and function in neurons. We present evidence for a putative stem-loop in the cebp-1 3' UTR that contributes to basal expression levels of mRNA and to negative regulation by rpm-1. Mutant animals lacking the endogenous cebp-1 3' UTR showed a noticeable increased expression of cebp-1 mRNA and enhanced the neuronal developmental phenotypes of rpm-1 mutants. Our data reveal multiple cis elements within cebp-1's 3' UTR that help to optimize CEBP-1 expression levels in neuronal development.


Assuntos
Regiões 3' não Traduzidas/genética , Proteínas Estimuladoras de Ligação a CCAAT/genética , Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/genética , Regulação da Expressão Gênica no Desenvolvimento , Neurônios/metabolismo , Sequências Reguladoras de Ácido Nucleico/genética , Animais , Axônios/metabolismo , Sequência de Bases , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Sistemas CRISPR-Cas/genética , Proteínas de Caenorhabditis elegans/metabolismo , Conformação de Ácido Nucleico , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Regeneração , Deleção de Sequência/genética , Transgenes
10.
Cell Rep ; 19(4): 822-835, 2017 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-28445732

RESUMO

Genetic changes in the HECT ubiquitin ligase HUWE1 are associated with intellectual disability, but it remains unknown whether HUWE1 functions in post-mitotic neurons to affect circuit function. Using genetics, pharmacology, and electrophysiology, we show that EEL-1, the HUWE1 ortholog in C. elegans, preferentially regulates GABAergic presynaptic transmission. Decreasing or increasing EEL-1 function alters GABAergic transmission and the excitatory/inhibitory (E/I) balance in the worm motor circuit, which leads to impaired locomotion and increased sensitivity to electroshock. Furthermore, multiple mutations associated with intellectual disability impair EEL-1 function. Although synaptic transmission defects did not result from abnormal synapse formation, sensitizing genetic backgrounds revealed that EEL-1 functions in the same pathway as the RING family ubiquitin ligase RPM-1 to regulate synapse formation and axon termination. These findings from a simple model circuit provide insight into the molecular mechanisms required to obtain E/I balance and could have implications for the link between HUWE1 and intellectual disability.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Neurônios GABAérgicos/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Aldicarb/toxicidade , Animais , Animais Geneticamente Modificados/metabolismo , Axônios/metabolismo , Proteínas de Caenorhabditis elegans/antagonistas & inibidores , Proteínas de Caenorhabditis elegans/genética , Eletrochoque , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Hipersensibilidade/etiologia , Locomoção/efeitos dos fármacos , Mutagênese Sítio-Dirigida , Terminações Pré-Sinápticas/metabolismo , Interferência de RNA , Transdução de Sinais , Sinapses/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Ubiquitina-Proteína Ligases/genética
11.
J Biol Chem ; 292(6): 2519-2530, 2017 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-27979965

RESUMO

The intracellular signaling protein regulator of presynaptic morphology 1 (RPM-1) is a conserved regulator of synapse formation and axon termination in Caenorhabditis elegans RPM-1 functions in a ubiquitin ligase complex with the F-box protein FSN-1 and functions through the microtubule binding protein RAE-1. Using a structure-function approach and positive selection for transgenic C. elegans, we explored the biochemical relationship between RPM-1, FSN-1, and RAE-1. This led to the identification of two new domains in RPM-1 that are sufficient for binding to FSN-1, called FSN-1 binding domain 2 (FBD2) and FBD3. Furthermore, we map the RAE-1 binding domain to a much smaller region of RPM-1. Point mutations in RPM-1 that reduce binding to RAE-1 did not affect FSN-1 binding, indicating that RPM-1 utilizes different biochemical mechanisms to bind these molecules. Analysis of RPM-1 protein complexes in the neurons of C. elegans elucidated two further discoveries: FSN-1 binds to RAE-1, and this interaction is not mediated by RPM-1, and RPM-1 binding to FSN-1 and RAE-1 reduces FSN-1·RAE-1 complex formation. These results indicate that RPM-1 uses different mechanisms to recruit FSN-1 and RAE-1 into independent signaling complexes in neurons.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Neurônios/metabolismo , Transdução de Sinais , Sinapses/metabolismo , Animais , Animais Geneticamente Modificados , Proteínas de Caenorhabditis elegans/genética , Ligação Proteica , Relação Estrutura-Atividade
12.
Neural Dev ; 11: 8, 2016 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-27008623

RESUMO

During development, a coordinated and integrated series of events must be accomplished in order to generate functional neural circuits. Axons must navigate toward target cells, build synaptic connections, and terminate outgrowth. The PHR proteins (consisting of mammalian Phr1/MYCBP2, Drosophila Highwire and C. elegans RPM-1) function in each of these events in development. Here, we review PHR function across species, as well as the myriad of signaling pathways PHR proteins regulate. These findings collectively suggest that the PHR proteins are intracellular signaling hubs, a concept we explore in depth. Consistent with prominent developmental functions, genetic links have begun to emerge between PHR signaling networks and neurodevelopmental disorders, such as autism, schizophrenia and intellectual disability. Finally, we discuss the recent and important finding that PHR proteins regulate axon degeneration, which has further heightened interest in this fascinating group of molecules.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Axônios/metabolismo , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Drosophila/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Transdução de Sinais , Ubiquitina-Proteína Ligases/metabolismo , Animais , Axônios/fisiologia , Caenorhabditis elegans , Drosophila melanogaster , Humanos , Transtornos do Neurodesenvolvimento/genética , Transtornos do Neurodesenvolvimento/metabolismo , Especificidade da Espécie , Sinapses/metabolismo
13.
Sci China Life Sci ; 58(11): 1084-8, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26563175

RESUMO

Since Caenorhabditis elegans was chosen as a model organism by Sydney Brenner in 1960's, genetic studies in this organism have been instrumental in discovering the function of genes and in deciphering molecular signaling network. The small size of the organism and the simple nervous system enable the complete reconstruction of the first connectome. The stereotypic developmental program and the anatomical reproducibility of synaptic connections provide a blueprint to dissect the mechanisms underlying synapse formation. Recent technological innovation using laser surgery of single axons and in vivo imaging has also made C. elegans a new model for axon regeneration. Importantly, genes regulating synaptogenesis and axon regeneration are highly conserved in function across animal phyla. This mini-review will summarize the main approaches and the key findings in understanding the mechanisms underlying the development and maintenance of the nervous system. The impact of such findings underscores the awesome power of C. elegans genetics.


Assuntos
Axônios/fisiologia , Caenorhabditis elegans/fisiologia , Regeneração Nervosa/fisiologia , Sinapses/fisiologia , Animais , Axônios/metabolismo , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Modelos Neurológicos , Mutação , Regeneração Nervosa/genética , Transdução de Sinais/genética , Sinapses/metabolismo
14.
G3 (Bethesda) ; 5(12): 2745-57, 2015 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-26464359

RESUMO

The PAM/Highwire/RPM-1 (PHR) proteins are signaling hubs that function as important regulators of neural development. Loss of function in Caenorhabditis elegans rpm-1 and Drosophila Highwire results in failed axon termination, inappropriate axon targeting, and abnormal synapse formation. Despite broad expression in the nervous system and relatively dramatic defects in synapse formation and axon development, very mild abnormalities in behavior have been found in animals lacking PHR protein function. Therefore, we hypothesized that large defects in behavior might only be detected in scenarios in which evoked, prolonged circuit function is required, or in which behavioral plasticity occurs. Using quantitative approaches in C. elegans, we found that rpm-1 loss-of-function mutants have relatively mild abnormalities in exploratory locomotion, but have large defects in evoked responses to harsh touch and learning associated with tap habituation. We explored the nature of the severe habituation defects in rpm-1 mutants further. To address what part of the habituation circuit was impaired in rpm-1 mutants, we performed rescue analysis with promoters for different neurons. Our findings indicate that RPM-1 function in the mechanosensory neurons affects habituation. Transgenic expression of RPM-1 in adult animals failed to rescue habituation defects, consistent with developmental defects in rpm-1 mutants resulting in impaired habituation. Genetic analysis showed that other regulators of neuronal development that function in the rpm-1 pathway (including glo-4, fsn-1, and dlk-1) also affected habituation. Overall, our findings suggest that developmental defects in rpm-1 mutants manifest most prominently in behaviors that require protracted or plastic circuit function, such as learning.


Assuntos
Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/fisiologia , Fatores de Troca do Nucleotídeo Guanina/genética , Animais , Animais Geneticamente Modificados , Comportamento Animal , Proteínas de Caenorhabditis elegans/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Habituação Psicofisiológica/genética , Locomoção/genética , Mecanotransdução Celular , Mutação , Neurônios/fisiologia
15.
16.
Genetics ; 198(3): 1101-15, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25194163

RESUMO

The PHR (Pam/Highwire/RPM-1) family of ubiquitin E3 ligases plays conserved roles in axon patterning and synaptic development. Genetic modifier analysis has greatly aided the discovery of the signal transduction cascades regulated by these proteins. In Caenorhabditis elegans, loss of function in rpm-1 causes axon overgrowth and aberrant presynaptic morphology, yet the mutant animals exhibit little behavioral deficits. Strikingly, rpm-1 mutations strongly synergize with loss of function in the presynaptic active zone assembly factors, syd-1 and syd-2, resulting in severe locomotor deficits. Here, we provide ultrastructural evidence that double mutants, between rpm-1 and syd-1 or syd-2, dramatically impair synapse formation. Taking advantage of the synthetic locomotor defects to select for genetic suppressors, previous studies have identified the DLK-1 MAP kinase cascade negatively regulated by RPM-1. We now report a comprehensive analysis of a large number of suppressor mutations of this screen. Our results highlight the functional specificity of the DLK-1 cascade in synaptogenesis. We also identified two previously uncharacterized genes. One encodes a novel protein, SUPR-1, that acts cell autonomously to antagonize RPM-1. The other affects a conserved protein ESS-2, the homolog of human ES2 or DGCR14. Loss of function in ess-2 suppresses rpm-1 only in the presence of a dlk-1 splice acceptor mutation. We show that ESS-2 acts to promote accurate mRNA splicing when the splice site is compromised. The human DGCR14/ES2 resides in a deleted chromosomal region implicated in DiGeorge syndrome, and its mutation has shown high probability as a risk factor for schizophrenia. Our findings provide the first functional evidence that this family of proteins regulate mRNA splicing in a context-specific manner.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/genética , Genes Supressores , Fatores de Troca do Nucleotídeo Guanina/genética , Splicing de RNA/genética , Animais , Proteínas de Caenorhabditis elegans/genética , Genes de Helmintos , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Mutação/genética , Fenótipo , Terminações Pré-Sinápticas/metabolismo , Terminações Pré-Sinápticas/ultraestrutura , Sítios de Splice de RNA/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Vesículas Sinápticas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA