Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.439
Filtrar
1.
J Agric Food Chem ; 72(17): 9746-9754, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38602331

RESUMEN

The latex of Euphorbia peplus and its major component 20-deoxyingenol-3-angelate (DI3A) displayed significant nematicidal activity against Caenorhabditis elegans and Panagrellus redivivus. DI3A treatment inhibited the growth and development of nematodes and caused significantly negative effects on locomotion behavior, reproduction, and accumulation of reactive oxygen species. Transcriptome analysis indicated that differential expression genes in DI3A-treated C. elegans were mainly associated with the metabolism, growth, and development process, which were further confirmed by RT-qPCR experiments. The expression level of TPA-1 gene encoding a protein kinase C isotype was obviously upregulated by DI3A treatment, and knockdown of TPA-1 by RNAi technology in the nematode could relieve the growth-inhibitory effect of DI3A. Metabolic analysis indicated that DI3A was hardly metabolized by C. elegans, but a glycosylated indole derivative was specifically accumulated likely due to the activation of detoxification. Overall, our findings suggested that DI3A from E. peplus latex exerted a potent nematicidal effect through the gene TPA-1, which provides a potential target for the control of nematodes and also suggests the potential application value of E. peplus latex and DI3A as botanical nematicides.


Asunto(s)
Antinematodos , Caenorhabditis elegans , Euphorbia , Látex , Proteína Quinasa C , Animales , Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/genética , Caenorhabditis elegans/enzimología , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/crecimiento & desarrollo , Látex/química , Látex/metabolismo , Antinematodos/farmacología , Antinematodos/química , Antinematodos/metabolismo , Euphorbia/química , Proteína Quinasa C/metabolismo , Proteína Quinasa C/genética , Extractos Vegetales/farmacología , Extractos Vegetales/química
2.
Cell Chem Biol ; 31(5): 1011-1022.e6, 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38183989

RESUMEN

Methyl ketone (MK)-ascarosides represent essential components of several pheromones in Caenorhabditis elegans, including the dauer pheromone, which triggers the stress-resistant dauer larval stage, and the male-attracting sex pheromone. Here, we identify an acyl-CoA thioesterase, ACOT-15, that is required for the biosynthesis of MK-ascarosides. We propose a model in which ACOT-15 hydrolyzes the ß-keto acyl-CoA side chain of an ascaroside intermediate during ß-oxidation, leading to decarboxylation and formation of the MK. Using comparative metabolomics, we identify additional ACOT-15-dependent metabolites, including an unusual piperidyl-modified ascaroside, reminiscent of the alkaloid pelletierine. The ß-keto acid generated by ACOT-15 likely couples to 1-piperideine to produce the piperidyl ascaroside, which is much less dauer-inducing than the dauer pheromone, asc-C6-MK (ascr#2, 1). The bacterial food provided influences production of the piperidyl ascaroside by the worm. Our work shows how the biosynthesis of MK- and piperidyl ascarosides intersect and how bacterial food may impact chemical signaling in the worm.


Asunto(s)
Caenorhabditis elegans , Feromonas , Animales , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/enzimología , Feromonas/metabolismo , Feromonas/biosíntesis , Feromonas/química , Proteínas de Caenorhabditis elegans/metabolismo , Tioléster Hidrolasas/metabolismo
3.
Nature ; 622(7982): 402-409, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37758951

RESUMEN

Transposable elements are genomic parasites that expand within and spread between genomes1. PIWI proteins control transposon activity, notably in the germline2,3. These proteins recognize their targets through small RNA co-factors named PIWI-interacting RNAs (piRNAs), making piRNA biogenesis a key specificity-determining step in this crucial genome immunity system. Although the processing of piRNA precursors is an essential step in this process, many of the molecular details remain unclear. Here, we identify an endoribonuclease, precursor of 21U RNA 5'-end cleavage holoenzyme (PUCH), that initiates piRNA processing in the nematode Caenorhabditis elegans. Genetic and biochemical studies show that PUCH, a trimer of Schlafen-like-domain proteins (SLFL proteins), executes 5'-end piRNA precursor cleavage. PUCH-mediated processing strictly requires a 7-methyl-G cap (m7G-cap) and a uracil at position three. We also demonstrate how PUCH interacts with PETISCO, a complex that binds to piRNA precursors4, and that this interaction enhances piRNA production in vivo. The identification of PUCH concludes the search for the 5'-end piRNA biogenesis factor in C. elegans and uncovers a type of RNA endonuclease formed by three SLFL proteins. Mammalian Schlafen (SLFN) genes have been associated with immunity5, exposing a molecular link between immune responses in mammals and deeply conserved RNA-based mechanisms that control transposable elements.


Asunto(s)
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Endorribonucleasas , ARN de Interacción con Piwi , Animales , Proteínas Argonautas/metabolismo , Caenorhabditis elegans/enzimología , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/química , Proteínas de Caenorhabditis elegans/metabolismo , Elementos Transponibles de ADN/genética , Endorribonucleasas/química , Endorribonucleasas/metabolismo , Holoenzimas/química , Holoenzimas/metabolismo , ARN de Interacción con Piwi/química , ARN de Interacción con Piwi/genética , ARN de Interacción con Piwi/metabolismo , Análogos de Caperuza de ARN/química , Análogos de Caperuza de ARN/metabolismo
4.
J Biol Chem ; 299(9): 105149, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37567477

RESUMEN

Alanyl-tRNA synthetase retains a conserved prototype structure throughout its biology. Nevertheless, its C-terminal domain (C-Ala) is highly diverged and has been shown to play a role in either tRNA or DNA binding. Interestingly, we discovered that Caenorhabditis elegans cytoplasmic C-Ala (Ce-C-Alac) robustly binds both ligands. How Ce-C-Alac targets its cognate tRNA and whether a similar feature is conserved in its mitochondrial counterpart remain elusive. We show that the N- and C-terminal subdomains of Ce-C-Alac are responsible for DNA and tRNA binding, respectively. Ce-C-Alac specifically recognized the conserved invariant base G18 in the D-loop of tRNAAla through a highly conserved lysine residue, K934. Despite bearing little resemblance to other C-Ala domains, C. elegans mitochondrial C-Ala robustly bound both tRNAAla and DNA and maintained targeting specificity for the D-loop of its cognate tRNA. This study uncovers the underlying mechanism of how C. elegans C-Ala specifically targets the D-loop of tRNAAla.


Asunto(s)
Alanina-ARNt Ligasa , Caenorhabditis elegans , Motivos de Nucleótidos , ARN de Transferencia de Alanina , Animales , Alanina-ARNt Ligasa/química , Alanina-ARNt Ligasa/metabolismo , Caenorhabditis elegans/enzimología , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Secuencia Conservada , Citoplasma/enzimología , ADN/química , ADN/metabolismo , Ligandos , Lisina/metabolismo , Mitocondrias/enzimología , Dominios Proteicos , ARN de Transferencia de Alanina/química , ARN de Transferencia de Alanina/metabolismo , Especificidad por Sustrato , Conformación de Ácido Nucleico
5.
Nucleic Acids Res ; 51(5): 2434-2446, 2023 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-36794723

RESUMEN

In Caenorhabditis elegans, the N6-methyladenosine (m6A) modification by METT10, at the 3'-splice sites in S-adenosyl-l-methionine (SAM) synthetase (sams) precursor mRNA (pre-mRNA), inhibits sams pre-mRNA splicing, promotes alternative splicing coupled with nonsense-mediated decay of the pre-mRNAs, and thereby maintains the cellular SAM level. Here, we present structural and functional analyses of C. elegans METT10. The structure of the N-terminal methyltransferase domain of METT10 is homologous to that of human METTL16, which installs the m6A modification in the 3'-UTR hairpins of methionine adenosyltransferase (MAT2A) pre-mRNA and regulates the MAT2A pre-mRNA splicing/stability and SAM homeostasis. Our biochemical analysis suggested that C. elegans METT10 recognizes the specific structural features of RNA surrounding the 3'-splice sites of sams pre-mRNAs, and shares a similar substrate RNA recognition mechanism with human METTL16. C. elegans METT10 also possesses a previously unrecognized functional C-terminal RNA-binding domain, kinase associated 1 (KA-1), which corresponds to the vertebrate-conserved region (VCR) of human METTL16. As in human METTL16, the KA-1 domain of C. elegans METT10 facilitates the m6A modification of the 3'-splice sites of sams pre-mRNAs. These results suggest the well-conserved mechanisms for the m6A modification of substrate RNAs between Homo sapiens and C. elegans, despite their different regulation mechanisms for SAM homeostasis.


Asunto(s)
Caenorhabditis elegans , Metiltransferasas , Animales , Caenorhabditis elegans/enzimología , Caenorhabditis elegans/metabolismo , Homeostasis/genética , Metionina Adenosiltransferasa/genética , Metionina Adenosiltransferasa/metabolismo , Metilación , Metiltransferasas/química , Precursores del ARN
6.
J Cell Biol ; 221(10)2022 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-36121422

RESUMEN

How cells spatially organize their plasma membrane, cytoskeleton, and cytoplasm remains a central question for cell biologists. In this issue of JCB, Calvi et al. (2022. J. Cell Biol.https://doi.org/10.1083/jcb.202201048) identify PP1 phosphatases as key regulators of C. elegans anterior-posterior polarity, by counterbalancing aPKC-mediated phosphorylation of PAR-2.


Asunto(s)
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Polaridad Celular , Fosfoproteínas Fosfatasas , Proteína Quinasa C , Animales , Caenorhabditis elegans/citología , Caenorhabditis elegans/enzimología , Proteínas de Caenorhabditis elegans/metabolismo , Membrana Celular , Citoplasma , Microtúbulos/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Fosforilación , Proteína Quinasa C/metabolismo
7.
J Biol Chem ; 298(9): 102343, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35933017

RESUMEN

Proximity-dependent protein labeling provides a powerful in vivo strategy to characterize the interactomes of specific proteins. We previously optimized a proximity labeling protocol for Caenorhabditis elegans using the highly active biotin ligase TurboID. A significant constraint on the sensitivity of TurboID is the presence of abundant endogenously biotinylated proteins that take up bandwidth in the mass spectrometer, notably carboxylases that use biotin as a cofactor. In C. elegans, these comprise POD-2/acetyl-CoA carboxylase alpha, PCCA-1/propionyl-CoA carboxylase alpha, PYC-1/pyruvate carboxylase, and MCCC-1/methylcrotonyl-CoA carboxylase alpha. Here, we developed ways to remove these carboxylases prior to streptavidin purification and mass spectrometry by engineering their corresponding genes to add a C-terminal His10 tag. This allows us to deplete them from C. elegans lysates using immobilized metal affinity chromatography. To demonstrate the method's efficacy, we use it to expand the interactome map of the presynaptic active zone protein ELKS-1. We identify many known active zone proteins, including UNC-10/RIM, SYD-2/liprin-alpha, SAD-1/BRSK1, CLA-1/CLArinet, C16E9.2/Sentryn, as well as previously uncharacterized potentially synaptic proteins such as the ortholog of human angiomotin, F59C12.3 and the uncharacterized protein R148.3. Our approach provides a quick and inexpensive solution to a common contaminant problem in biotin-dependent proximity labeling. The approach may be applicable to other model organisms and will enable deeper and more complete analysis of interactors for proteins of interest.


Asunto(s)
Biotinilación , Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Carboxiliasas , Acetil-CoA Carboxilasa/metabolismo , Animales , Biotinilación/métodos , Caenorhabditis elegans/enzimología , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Carboxiliasas/genética , Carboxiliasas/metabolismo , Proteínas Portadoras/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Metilmalonil-CoA Descarboxilasa/metabolismo , Piruvato Carboxilasa/metabolismo , Estreptavidina
8.
J Biol Chem ; 298(10): 102415, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36007615

RESUMEN

Virtually all age-related neurodegenerative diseases (NDs) can be characterized by the accumulation of proteins inside and outside the cell that are thought to significantly contribute to disease pathogenesis. One of the cell's primary systems for the degradation of misfolded/damaged proteins is the ubiquitin proteasome system (UPS), and its impairment is implicated in essentially all NDs. Thus, upregulating this system to combat NDs has garnered a great deal of interest in recent years. Various animal models have focused on stimulating 26S activity and increasing 20S proteasome levels, but thus far, none have targeted intrinsic activation of the 20S proteasome itself. Therefore, we constructed an animal model that endogenously expresses a hyperactive, open gate proteasome in Caenorhabditis elegans. The gate-destabilizing mutation that we introduced into the nematode germline yielded a viable nematode population with enhanced proteasomal activity, including peptide, unstructured protein, and ubiquitin-dependent degradation activities. We determined these nematodes showed a significantly increased lifespan and substantial resistance to oxidative and proteotoxic stress but a significant decrease in fecundity. Our results show that introducing a constitutively active proteasome into a multicellular organism is feasible and suggests targeting the proteasome gating mechanism as a valid approach for future age-related disease research efforts in mammals.


Asunto(s)
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Longevidad , Estrés Oxidativo , Complejo de la Endopetidasa Proteasomal , Proteostasis , Animales , Caenorhabditis elegans/enzimología , Caenorhabditis elegans/genética , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo , Activación Enzimática , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo
9.
Nature ; 607(7919): 571-577, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35794472

RESUMEN

Individuals can exhibit differences in metabolism that are caused by the interplay of genetic background, nutritional input, microbiota and other environmental factors1-4. It is difficult to connect differences in metabolism to genomic variation and derive underlying molecular mechanisms in humans, owing to differences in diet and lifestyle, among others. Here we use the nematode Caenorhabditis elegans as a model to study inter-individual variation in metabolism. By comparing three wild strains and the commonly used N2 laboratory strain, we find differences in the abundances of both known metabolites and those that have not to our knowledge been previously described. The latter metabolites include conjugates between 3-hydroxypropionate (3HP) and several amino acids (3HP-AAs), which are much higher in abundance in one of the wild strains. 3HP is an intermediate in the propionate shunt pathway, which is activated when flux through the canonical, vitamin-B12-dependent propionate breakdown pathway is perturbed5. We show that increased accumulation of 3HP-AAs is caused by genetic variation in HPHD-1, for which 3HP is a substrate. Our results suggest that the production of 3HP-AAs represents a 'shunt-within-a-shunt' pathway to accommodate a reduction-of-function allele in hphd-1. This study provides a step towards the development of metabolic network models that capture individual-specific differences of metabolism and more closely represent the diversity that is found in entire species.


Asunto(s)
Caenorhabditis elegans , Redes y Vías Metabólicas , Animales , Humanos , Oxidorreductasas de Alcohol/genética , Oxidorreductasas de Alcohol/metabolismo , Aminoácidos/metabolismo , Caenorhabditis elegans/clasificación , Caenorhabditis elegans/enzimología , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Ácido Láctico/análogos & derivados , Ácido Láctico/metabolismo , Redes y Vías Metabólicas/genética , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Modelos Animales , Propionatos/metabolismo , Vitamina B 12/metabolismo
10.
Proc Natl Acad Sci U S A ; 119(3)2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-35022236

RESUMEN

The fidelity of a signaling pathway depends on its tight regulation in space and time. Extracellular signal-regulated kinase (ERK) controls wide-ranging cellular processes to promote organismal development and tissue homeostasis. ERK activation depends on a reversible dual phosphorylation on the TEY motif in its active site by ERK kinase (MEK) and dephosphorylation by DUSPs (dual specificity phosphatases). LIP-1, a DUSP6/7 homolog, was proposed to function as an ERK (MPK-1) DUSP in the Caenorhabditis elegans germline primarily because of its phenotype, which morphologically mimics that of a RAS/let-60 gain-of-function mutant (i.e., small oocyte phenotype). Our investigations, however, reveal that loss of lip-1 does not lead to an increase in MPK-1 activity in vivo. Instead, we show that loss of lip-1 leads to 1) a decrease in MPK-1 phosphorylation, 2) lower MPK-1 substrate phosphorylation, 3) phenocopy of mpk-1 reduction-of-function (rather than gain-of-function) allele, and 4) a failure to rescue mpk-1-dependent germline or fertility defects. Moreover, using diverse genetic mutants, we show that the small oocyte phenotype does not correlate with increased ectopic MPK-1 activity and that ectopic increase in MPK-1 phosphorylation does not necessarily result in a small oocyte phenotype. Together, these data demonstrate that LIP-1 does not function as an MPK-1 DUSP in the C. elegans germline. Our results caution against overinterpretation of the mechanistic underpinnings of orthologous phenotypes, since they may be a result of independent mechanisms, and provide a framework for characterizing the distinct molecular targets through which LIP-1 may mediate its several germline functions.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/enzimología , Proteínas de Ciclo Celular/metabolismo , Células Germinativas/enzimología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Animales , Proteínas de Caenorhabditis elegans/genética , Proteínas de Ciclo Celular/genética , Proliferación Celular , Activación Enzimática , Mutación/genética , Oocitos/citología , Oocitos/metabolismo , Fase Paquiteno , Fenotipo , Fosforilación , Proteínas Tirosina Fosfatasas/genética , Especificidad por Sustrato , Complejo Sinaptonémico/metabolismo , Temperatura
11.
Int J Mol Sci ; 24(1)2022 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-36614054

RESUMEN

Altered metabolism is a hallmark of aging. The tricarboxylic acid cycle (TCA cycle) is an essential metabolic pathway and plays an important role in lifespan regulation. Supplementation of α-ketoglutarate, a metabolite converted by isocitrate dehydrogenase alpha-1 (idha-1) in the TCA cycle, increases lifespan in C. elegans. However, whether idha-1 can regulate lifespan in C. elegans remains unknown. Here, we reported that the expression of idha-1 modulates lifespan and oxidative stress tolerance in C. elegans. Transgenic overexpression of idha-1 extends lifespan, increases the levels of NADPH/NADP+ ratio, and elevates the tolerance to oxidative stress. Conversely, RNAi knockdown of idha-1 exhibits the opposite effects. In addition, the longevity of eat-2 (ad1116) mutant via dietary restriction (DR) was reduced by idha-1 knockdown, indicating that idha-1 may play a role in DR-mediated longevity. Furthermore, idha-1 mediated lifespan may depend on the target of rapamycin (TOR) signaling. Moreover, the phosphorylation levels of S6 kinase (p-S6K) inversely correlate with idha-1 expression, supporting that the idha-1-mediated lifespan regulation may involve the TOR signaling pathway. Together, our data provide new insights into the understanding of idha-1 new function in lifespan regulation probably via DR and TOR signaling and in oxidative stress tolerance in C. elegans.


Asunto(s)
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Isocitrato Deshidrogenasa , Longevidad , Estrés Oxidativo , Animales , Caenorhabditis elegans/enzimología , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Isocitrato Deshidrogenasa/genética , Isocitrato Deshidrogenasa/metabolismo , Longevidad/genética
12.
Cell Rep ; 37(13): 110166, 2021 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-34965433

RESUMEN

Animals encounter microorganisms in their habitats, adapting physiology and behavior accordingly. The nematode Caenorhabditis elegans is found in microbe-rich environments; however, its responses to fungi are not extensively studied. Here, we describe interactions of C. elegans and Penicillium brevicompactum, an ecologically relevant mold. Transcriptome studies reveal that co-culture upregulates stress response genes, including xenobiotic-metabolizing enzymes (XMEs), in C. elegans intestine and AMsh glial cells. The nuclear hormone receptors (NHRs) NHR-45 and NHR-156 are induction regulators, and mutants that cannot induce XMEs in the intestine when exposed to P. brevicompactum experience mitochondrial stress and exhibit developmental defects. Different C. elegans wild isolates harbor sequence polymorphisms in nhr-156, resulting in phenotypic diversity in AMsh glia responses to microbe exposure. We propose that P. brevicompactum mitochondria-targeting mycotoxins are deactivated by intestinal detoxification, allowing tolerance to moldy environments. Our studies support the idea that C. elegans NHRs may be regulated by environmental cues.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/efectos de los fármacos , Tracto Gastrointestinal/enzimología , Mitocondrias/enzimología , Neuroglía/enzimología , Penicillium/fisiología , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Caenorhabditis elegans/enzimología , Caenorhabditis elegans/microbiología , Proteínas de Caenorhabditis elegans/genética , Inducción Enzimática , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/microbiología , Regulación del Desarrollo de la Expresión Génica , Mitocondrias/efectos de los fármacos , Mitocondrias/microbiología , Neuroglía/efectos de los fármacos , Neuroglía/microbiología
13.
Cell Rep ; 37(4): 109901, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34706230

RESUMEN

The Wnt family contains conserved secretory proteins required for developmental patterning and tissue homeostasis. However, how Wnt is targeted to the endoplasmic reticulum (ER) for processing and secretion remains poorly understood. Here, we report that CATP-8/P5A ATPase directs neuronal migration non-cell autonomously in Caenorhabditis elegans by regulating EGL-20/Wnt biogenesis. CATP-8 likely functions as a translocase to translocate nascent EGL-20/Wnt polypeptide into the ER by interacting with the highly hydrophobic core region of EGL-20 signal sequence. Such regulation of Wnt biogenesis by P5A ATPase is common in C. elegans and conserved in human cells. These findings describe the physiological roles of P5A ATPase in neural development and identify Wnt proteins as direct substrates of P5A ATPase for ER translocation.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/enzimología , Movimiento Celular , Neuronas/enzimología , Vía de Señalización Wnt , Adenosina Trifosfatasas/genética , Animales , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Línea Celular Tumoral , Retículo Endoplásmico/enzimología , Retículo Endoplásmico/genética , Células HEK293 , Humanos
14.
Biochemistry ; 60(42): 3137-3151, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34648259

RESUMEN

Eukaryotic cells partition enzymes and other cellular components into distinct subcellular compartments to generate specialized biochemical niches. A subclass of these compartments form in the absence of lipid membranes, via liquid-liquid phase separation of proteins to form biomolecular condensates or "membraneless organelles" such as nucleoli, stress granules, and P-bodies. Because of their propensity to form compartments from simple starting materials, membraneless organelles are an attractive target for engineering new functionalities in both living cells and protocells. In this work, we demonstrate incorporation of a novel enzymatic activity in protein coacervates with the light-generating enzyme, NanoLuc, to produce bioluminescence. Using condensates comprised of the disordered RGG domain of Caenorhabditis elegans LAF-1, we functionalized condensates with enzymatic activity in vitro and show that enzyme localization to coacervates enhances assembly and activity of split enzymes. To build condensates that function as light-emitting reactors, we designed a NanoLuc enzyme flanked by RGG domains. The resulting condensates concentrated NanoLuc by 10-fold over bulk solution and displayed significantly increased reaction rates. We further show that condensate viscosity impacts light emission due to diffusion-limited behavior. Because our model condensates have low viscosities, we predict NanoLuc diffusion-limited behavior in most other condensates and thus propose the condensate-Nanoluc system as a potential strategy for high-throughput screening of condensate targeting drugs. By splitting the NanoLuc enzyme into its constituent components, we demonstrate that NanoLuc activity can be reconstituted via co-condensation. In addition, we demonstrate control of the spatial localization of the enzyme within condensates by targettng NanoLuc to the surface of in vitro condensates. Collectively, this work demonstrates that membraneless organelles can be endowed with localized enzymatic activity and that this activity can be spatially and temporally controlled via biochemical reconstitution and design of protein surfactants.


Asunto(s)
Proteínas de Caenorhabditis elegans/química , Luciferasas/química , Sustancias Macromoleculares/química , ARN Helicasas/química , Animales , Caenorhabditis elegans/enzimología , Luminiscencia , Dominios Proteicos , Ingeniería de Proteínas
15.
J Biol Chem ; 297(5): 101263, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34600886

RESUMEN

Autophagy is a major cellular quality control system responsible for the degradation of proteins and organelles in response to stress and damage to maintain homeostasis. Ubiquitination of autophagy-related proteins or regulatory components is important for the precise control of autophagy pathways. Here, we show that the deubiquitinase ubiquitin-specific protease 11 (USP11) restricts autophagy and that KO of USP11 in mammalian cells results in elevated autophagic flux. We also demonstrate that depletion of the USP11 homolog H34C03.2 in Caenorhabditis elegans triggers hyperactivation of autophagy and protects the animals against human amyloid-ß peptide 42 aggregation-induced paralysis. USP11 coprecipitated with autophagy-specific class III phosphatidylinositol 3-kinase complex I and limited its interaction with nuclear receptor-binding factor 2, thus decreasing lipid kinase activity of class III phosphatidylinositol 3-kinase complex I and subsequent recruitment of effectors such as WD-repeat domain phosphoinositide-interacting proteins to the autophagosomal membrane. Accordingly, more WD-repeat domain phosphoinositide-interacting protein 2 puncta accumulated in USP11 KO cells. In addition, USP11 interacts with and stabilizes the serine/threonine kinase mechanistic target of rapamycin, thereby further contributing to the regulation of autophagy induction. Taken together, our data suggested that USP11 impinges on the autophagy pathway at multiple sites and that inhibiting USP11 alleviates symptoms of proteotoxicity, which is a major hallmark of neurodegenerative diseases.


Asunto(s)
Autofagia , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/enzimología , Tioléster Hidrolasas/metabolismo , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Animales , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Línea Celular , Técnicas de Inactivación de Genes , Humanos , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Tioléster Hidrolasas/genética
16.
Science ; 373(6558): 984-991, 2021 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-34446600

RESUMEN

Protein kinase activity must be precisely regulated, but how a cell governs hyperactive kinases remains unclear. In this study, we generated a constitutively active mitogen-activated protein kinase DYF-5 (DYF-5CA) in Caenorhabditis elegans that disrupted sensory cilia. Genetic suppressor screens identified that mutations of ADR-2, an RNA adenosine deaminase, rescued ciliary phenotypes of dyf-5CA We found that dyf-5CA animals abnormally transcribed antisense RNAs that pair with dyf-5CA messenger RNA (mRNA) to form double-stranded RNA, recruiting ADR-2 to edit the region ectopically. RNA editing impaired dyf-5CA mRNA splicing, and the resultant intron retentions blocked DYF-5CA protein translation and activated nonsense-mediated dyf-5CA mRNA decay. The kinase RNA editing requires kinase hyperactivity. The similar RNA editing-dependent feedback regulation restricted the other ciliary kinases NEKL-4/NEK10 and DYF-18/CCRK, which suggests a widespread mechanism that underlies kinase regulation.


Asunto(s)
Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/enzimología , Cilios/metabolismo , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Edición de ARN , Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo , Animales , Caenorhabditis elegans/genética , Núcleo Celular/metabolismo , Cilios/enzimología , Activación Enzimática , Fenotipo , Biosíntesis de Proteínas , Proteínas Serina-Treonina Quinasas/metabolismo , Precursores del ARN/genética , Precursores del ARN/metabolismo , Empalme del ARN , Estabilidad del ARN , ARN sin Sentido/genética , ARN sin Sentido/metabolismo , ARN Bicatenario/genética , ARN Bicatenario/metabolismo , ARN de Helminto/genética , ARN de Helminto/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Transcripción Genética
17.
Biosci Rep ; 41(9)2021 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-34397087

RESUMEN

For decades, Parkinson's disease (PD) cases have been genetically categorised into familial, when caused by mutations in single genes with a clear inheritance pattern in affected families, or idiopathic, in the absence of an evident monogenic determinant. Recently, genome-wide association studies (GWAS) have revealed how common genetic variability can explain up to 36% of PD heritability and that PD manifestation is often determined by multiple variants at different genetic loci. Thus, one of the current challenges in PD research stands in modelling the complex genetic architecture of this condition and translating this into functional studies. Caenorhabditis elegans provide a profound advantage as a reductionist, economical model for PD research, with a short lifecycle, straightforward genome engineering and high conservation of PD relevant neural, cellular and molecular pathways. Functional models of PD genes utilising C. elegans show many phenotypes recapitulating pathologies observed in PD. When contrasted with mammalian in vivo and in vitro models, these are frequently validated, suggesting relevance of C. elegans in the development of novel PD functional models. This review will discuss how the nematode C. elegans PD models have contributed to the uncovering of molecular and cellular mechanisms of disease, with a focus on the genes most commonly found as causative in familial PD and risk factors in idiopathic PD. Specifically, we will examine the current knowledge on a central player in both familial and idiopathic PD, Leucine-rich repeat kinase 2 (LRRK2) and how it connects to multiple PD associated GWAS candidates and Mendelian disease-causing genes.


Asunto(s)
Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/genética , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Mutación , Neuronas/enzimología , Enfermedad de Parkinson/genética , Animales , Animales Modificados Genéticamente , Caenorhabditis elegans/enzimología , Proteínas de Caenorhabditis elegans/metabolismo , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Herencia , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Degeneración Nerviosa , Neuronas/patología , Enfermedad de Parkinson/enzimología , Enfermedad de Parkinson/patología , Fenotipo , Factores de Riesgo
18.
PLoS Biol ; 19(7): e3001302, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34252079

RESUMEN

Defects in mitochondrial function activate compensatory responses in the cell. Mitochondrial stress that is caused by unfolded proteins inside the organelle induces a transcriptional response (termed the "mitochondrial unfolded protein response" [UPRmt]) that is mediated by activating transcription factor associated with stress 1 (ATFS-1). The UPRmt increases mitochondrial protein quality control. Mitochondrial dysfunction frequently causes defects in the import of proteins, resulting in the accumulation of mitochondrial proteins outside the organelle. In yeast, cells respond to mistargeted mitochondrial proteins by increasing activity of the proteasome in the cytosol (termed the "unfolded protein response activated by mistargeting of proteins" [UPRam]). The presence and relevance of this response in higher eukaryotes is unclear. Here, we demonstrate that defects in mitochondrial protein import in Caenorhabditis elegans lead to proteasome activation and life span extension. Both proteasome activation and life span prolongation partially depend on ATFS-1, despite its lack of influence on proteasomal gene transcription. Importantly, life span prolongation depends on the fully assembled proteasome. Our data provide a link between mitochondrial dysfunction and proteasomal activity and demonstrate its direct relevance to mechanisms that promote longevity.


Asunto(s)
Caenorhabditis elegans/fisiología , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Estrés Fisiológico , Animales , Caenorhabditis elegans/enzimología , Proteínas de Caenorhabditis elegans/genética , Activación Enzimática , Técnicas de Silenciamiento del Gen , Respuesta de Proteína Desplegada
19.
Cell Cycle ; 20(16): 1519-1526, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34266376

RESUMEN

A central player in meiotic chromosome dynamics is the conserved Polo-like kinase (PLK) family. PLKs are dynamically localized to distinct structures during meiotic prophase and phosphorylate a diverse group of substrates to control homolog pairing, synapsis, and meiotic recombination. In a recent study, we uncovered the mechanisms that control the targeting of a meiosis-specific PLK-2 in C. elegans. In early meiotic prophase, PLK-2 localizes to special chromosome regions known as pairing centers and drives homolog pairing and synapsis. PLK-2 then relocates to the synaptonemal complex (SC) after crossover designation and mediates chromosome remodeling required for homolog separation. What controls this intricate targeting of PLK-2 in space and time? We discuss recent findings and remaining questions for the future.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/enzimología , Emparejamiento Cromosómico , Cromosomas , Meiosis , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Sitios de Unión , Caenorhabditis elegans/genética , Caenorhabditis elegans/crecimiento & desarrollo , Proteínas de Caenorhabditis elegans/genética , Regulación de la Expresión Génica , Profase Meiótica I , Fosforilación , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal
20.
Food Funct ; 12(15): 6841-6850, 2021 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-34124721

RESUMEN

Consumption of 4-desmethylsterols has been claimed to have many beneficial effects, but the benefits of 4,4-dimethylsterols are less appreciated. We utilized a nematode model, Caenorhabditis elegans (C. elegans), to explore the anti-obesity effects of different classes of 4,4-dimethylsterols purified from rice bran oil (RST) and shea nut butter (SST). Both SST and RST significantly reduced fat deposition in C. elegans with smaller sizes and numbers of lipid droplets. But the food intake was not significantly affected. Metabolomics analysis indicated a significantly altered pathway after treatment with 4,4-dimethylsterols. Finally, it was found that 4,4-dimethylsterols targeted stearoyl-CoA desaturases (SCD) and nuclear hormone receptor-49 (NHR-49), resulting in a reduced desaturation index as proved by a lower ratio of oleic acid (C18:1n-9) to stearic acid (C18:0). Overall, 4,4-dimethylsterols can inhibit fat deposition via regulating the NHR-49/SCD pathway in C. elegans.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Grasas/metabolismo , Fitosteroles , Aceites de Plantas/química , Receptores Citoplasmáticos y Nucleares/metabolismo , Estearoil-CoA Desaturasa/metabolismo , Animales , Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/enzimología , Caenorhabditis elegans/metabolismo , Metaboloma/efectos de los fármacos , Fitosteroles/química , Fitosteroles/farmacología , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA