Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 304
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(35): e2404969121, 2024 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-39172783

RESUMO

The abundance of CaV2 voltage-gated calcium channels is linked to presynaptic homeostatic plasticity (PHP), a process that recalibrates synaptic strength to maintain the stability of neural circuits. However, the molecular and cellular mechanisms governing PHP and CaV2 channels are not completely understood. Here, we uncover a previously not described form of PHP in Caenorhabditis elegans, revealing an inverse regulatory relationship between the efficiency of neurotransmitter release and the abundance of UNC-2/CaV2 channels. Gain-of-function unc-2SL(S240L) mutants, which carry a mutation analogous to the one causing familial hemiplegic migraine type 1 in humans, showed markedly reduced channel abundance despite increased channel functionality. Reducing synaptic release in these unc-2SL(S240L) mutants restored channel levels to those observed in wild-type animals. Conversely, loss-of-function unc-2DA(D726A) mutants, which harbor the D726A mutation in the channel pore, exhibited a marked increase in channel abundance. Enhancing synaptic release in unc-2DA mutants reversed this increase in channel levels. Importantly, this homeostatic regulation of UNC-2 channel levels is accompanied by the structural remodeling of the active zone (AZ); specifically, unc-2DA mutants, which exhibit increased channel abundance, showed parallel increases in select AZ proteins. Finally, our forward genetic screen revealed that WWP-1, a HECT family E3 ubiquitin ligase, is a key homeostatic mediator that removes UNC-2 from synapses. These findings highlight a self-tuning PHP regulating UNC-2/CaV2 channel abundance along with AZ reorganization, ensuring synaptic strength and stability.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Neurotransmissores , Animais , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Neurotransmissores/metabolismo , Terminações Pré-Sinápticas/metabolismo , Canais de Cálcio/metabolismo , Canais de Cálcio/genética , Transmissão Sináptica/fisiologia , Plasticidade Neuronal , Mutação , Canais de Cálcio Tipo N/metabolismo , Canais de Cálcio Tipo N/genética , Neurônios/metabolismo , Proteínas de Membrana
2.
Nat Commun ; 15(1): 7144, 2024 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-39164296

RESUMO

FOXO transcription factors modulate aging-related pathways and influence longevity in multiple species, but the transcriptional targets that mediate these effects remain largely unknown. Here, we identify an evolutionarily conserved FOXO target gene, Oxidative stress-responsive serine-rich protein 1 (OSER1), whose overexpression extends lifespan in silkworms, nematodes, and flies, while its depletion correspondingly shortens lifespan. In flies, overexpression of OSER1 increases resistance to oxidative stress, starvation, and heat shock, while OSER1-depleted flies are more vulnerable to these stressors. In silkworms, hydrogen peroxide both induces and is scavenged by OSER1 in vitro and in vivo. Knockdown of OSER1 in Caenorhabditis elegans leads to increased ROS production and shorter lifespan, mitochondrial fragmentation, decreased ATP production, and altered transcription of mitochondrial genes. Human proteomic analysis suggests that OSER1 plays roles in oxidative stress response, cellular senescence, and reproduction, which is consistent with the data and suggests that OSER1 could play a role in fertility in silkworms and nematodes. Human studies demonstrate that polymorphic variants in OSER1 are associated with human longevity. In summary, OSER1 is an evolutionarily conserved FOXO-regulated protein that improves resistance to oxidative stress, maintains mitochondrial functional integrity, and increases lifespan in multiple species. Additional studies will clarify the role of OSER1 as a critical effector of healthy aging.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Proteínas de Drosophila , Fatores de Transcrição Forkhead , Longevidade , Estresse Oxidativo , Animais , Longevidade/genética , Caenorhabditis elegans/genética , Caenorhabditis elegans/fisiologia , Caenorhabditis elegans/metabolismo , Humanos , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Fatores de Transcrição Forkhead/metabolismo , Fatores de Transcrição Forkhead/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Bombyx/genética , Bombyx/metabolismo , Bombyx/fisiologia , Drosophila melanogaster/genética , Mitocôndrias/metabolismo , Mitocôndrias/genética , Espécies Reativas de Oxigênio/metabolismo , Regulação da Expressão Gênica
3.
Elife ; 132024 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-39161220

RESUMO

Since double-stranded RNA (dsRNA) is effective for silencing a wide variety of genes, all genes are typically considered equivalent targets for such RNA interference (RNAi). Yet, loss of some regulators of RNAi in the nematode Caenorhabditis elegans can selectively impair the silencing of some genes. Here, we show that such selective requirements can be explained by an intersecting network of regulators acting on genes with differences in their RNA metabolism. In this network, the Maelstrom domain-containing protein RDE-10, the intrinsically disordered protein MUT-16, and the Argonaute protein NRDE-3 work together so that any two are required for silencing one somatic gene, but each is singly required for silencing another somatic gene, where only the requirement for NRDE-3 can be overcome by enhanced dsRNA processing. Quantitative models and their exploratory simulations led us to find that (1) changing cis-regulatory elements of the target gene can reduce the dependence on NRDE-3, (2) animals can recover from silencing in non-dividing cells, and (3) cleavage and tailing of mRNAs with UG dinucleotides, which makes them templates for amplifying small RNAs, are enriched within 'pUG zones' matching the dsRNA. Similar crosstalk between pathways and restricted amplification could result in apparently selective silencing by endogenous RNAs.


A variety of diseases are treated with therapeutics that switch off genes via a mechanism called RNA interference (or RNAi for short). Each gene has the instructions cells need to build a particular protein. To achieve this, the DNA sequence must first be copied into a single-stranded mRNA molecule than can be translated into protein. RNAi interferes with this process by generating a double-stranded RNA molecule which contains the same DNA sequence as the gene being turned off. A set of proteins (known as regulators) then progressively trigger a series of events that allow the double-stranded RNA to be processed into short pieces that interact with the mRNA and target it for degradation. While cells use RNAi to regulate the expression of their own genes, researchers can also artificially switch off genes by synthesizing double-stranded RNA molecules in the laboratory. However, some genes are trickier to turn off than others, and why this happens is poorly understood. To investigate, Knudsen-Palmer et al. studied how two genes (bli-1 and unc-22) are switched off by RNAi in the roundworm Caenorhabditis elegans. A previous study discovered that bli-1 and unc-22 require different regulators for their expression to be disrupted. Knudsen-Palmer et al. found that this was because the RNAi process involves an intersecting network of multiple regulators, rather than a linear pathway of regulators working one after the other. For example, bli-1 requires three regulators (MUT-16, RDE-10 and NRDE-3), whereas unc-22 only needs any two of these regulators to be switched off. Further experiments revealed that which regulators are required depends on how the gene being silenced is naturally regulated in the cell. Analysis through a computational model showed that the regulators needed for RNAi could be altered in many ways, including by changing the regions that regulators bind to on the mRNA of the target gene. These findings provide new insights into why some genes respond differently to double-stranded RNA molecules. They also suggest that testing how natural regulation of a target gene influences its response to the RNAi process could potentially lead to better therapeutics.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Interferência de RNA , RNA de Cadeia Dupla , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Animais , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , RNA de Cadeia Dupla/metabolismo , RNA de Cadeia Dupla/genética , Proteínas de Ligação a RNA
4.
Nat Commun ; 15(1): 6879, 2024 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-39128917

RESUMO

Mechanical stress during muscle contraction is a constant threat to proteome integrity. However, there is a lack of experimental systems to identify critical proteostasis regulators under mechanical stress conditions. Here, we present the transgenic Caenorhabditis elegans model OptIMMuS (Optogenetic Induction of Mechanical Muscle Stress) to study changes in the proteostasis network associated with mechanical forces. Repeated blue light exposure of a muscle-expressed Chlamydomonas rheinhardii channelrhodopsin-2 variant results in sustained muscle contraction and mechanical stress. Using OptIMMuS, combined with proximity labeling and mass spectrometry, we identify regulators that cooperate with the myosin-directed chaperone UNC-45 in muscle proteostasis. One of these is the TRIM E3 ligase NHL-1, which interacts with UNC-45 and muscle myosin in genetic epistasis and co-immunoprecipitation experiments. We provide evidence that the ubiquitylation activity of NHL-1 regulates myosin levels and functionality under mechanical stress. In the future, OptIMMuS will help to identify muscle-specific proteostasis regulators of therapeutic relevance.


Assuntos
Animais Geneticamente Modificados , Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Optogenética , Proteostase , Estresse Mecânico , Ubiquitina-Proteína Ligases , Ubiquitinação , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/genética , Animais , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/genética , Miosinas/metabolismo , Miosinas/genética , Contração Muscular/fisiologia , Músculos/metabolismo , Chaperonas Moleculares
5.
J Cell Biol ; 223(11)2024 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-39105756

RESUMO

Mitosis in early embryos often proceeds at a rapid pace, but how this pace is achieved is not understood. Here, we show that cyclin B3 is the dominant driver of rapid embryonic mitoses in the C. elegans embryo. Cyclins B1 and B2 support slow mitosis (NEBD to anaphase ∼600 s), but the presence of cyclin B3 dominantly drives the approximately threefold faster mitosis observed in wildtype. Multiple mitotic events are slowed down in cyclin B1 and B2-driven mitosis, and cyclin B3-associated Cdk1 H1 kinase activity is ∼25-fold more active than cyclin B1-associated Cdk1. Addition of cyclin B1 to fast cyclin B3-only mitosis introduces an ∼60-s delay between completion of chromosome alignment and anaphase onset; this delay, which is important for segregation fidelity, is dependent on inhibitory phosphorylation of the anaphase activator Cdc20. Thus, cyclin B3 dominance, coupled to a cyclin B1-dependent delay that acts via Cdc20 phosphorylation, sets the rapid pace and ensures mitotic fidelity in the early C. elegans embryo.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Ciclina B1 , Embrião não Mamífero , Mitose , Animais , Caenorhabditis elegans/embriologia , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Ciclina B1/metabolismo , Ciclina B1/genética , Embrião não Mamífero/metabolismo , Fosforilação , Proteína Quinase CDC2/metabolismo , Proteína Quinase CDC2/genética , Ciclina B/metabolismo , Ciclina B/genética , Proteínas Cdc20/metabolismo , Proteínas Cdc20/genética , Ciclina B2/metabolismo , Ciclina B2/genética
6.
Biol Lett ; 20(8): 20240278, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39137892

RESUMO

The heat-shock response plays a key role in the immune defence against viruses across various organisms. Studies on model organisms have shown that inducing this response prior to viral exposure enhances host resistance to infections, while deficient responses make individuals more susceptible. Moreover, viruses rely on components of the heat-shock response for their own stability and viral infections improve thermal tolerance in plants, giving infected individuals an advantage in extreme conditions, which aids the virus in replication and transmission. Here, we examine the interaction between the nematode Caenorhabditis elegans and its natural pathogen the Orsay virus (OrV) under heat stress. We found that OrV infection leads to differential expression of heat-stress-related genes, and infected populations show increased resistance to heat-shock. This resistance correlates with increased expression of argonautes alg-1 and alg-2, which are crucial for survival after heat-shock and for OrV replication. Overall, our study suggests an environmental-dependent mutualistic relationship between the nematode and OrV, potentially expanding the animal's ecological niche and providing the virus with extra opportunities for replication and adaptation to extreme conditions.


Assuntos
Caenorhabditis elegans , Resposta ao Choque Térmico , Animais , Caenorhabditis elegans/virologia , Caenorhabditis elegans/fisiologia , Caenorhabditis elegans/genética , Caenorhabditis elegans/imunologia , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Interações Hospedeiro-Patógeno
7.
Nat Commun ; 15(1): 6594, 2024 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-39097618

RESUMO

Neurons coordinate inter-tissue protein homeostasis to systemically manage cytotoxic stress. In response to neuronal mitochondrial stress, specific neuronal signals coordinate the systemic mitochondrial unfolded protein response (UPRmt) to promote organismal survival. Yet, whether chemical neurotransmitters are sufficient to control the UPRmt in physiological conditions is not well understood. Here, we show that gamma-aminobutyric acid (GABA) inhibits, and acetylcholine (ACh) promotes the UPRmt in the Caenorhabditis elegans intestine. GABA controls the UPRmt by regulating extra-synaptic ACh release through metabotropic GABAB receptors GBB-1/2. We find that elevated ACh levels in animals that are GABA-deficient or lack ACh-degradative enzymes induce the UPRmt through ACR-11, an intestinal nicotinic α7 receptor. This neuro-intestinal circuit is critical for non-autonomously regulating organismal survival of oxidative stress. These findings establish chemical neurotransmission as a crucial regulatory layer for nervous system control of systemic protein homeostasis and stress responses.


Assuntos
Acetilcolina , Caenorhabditis elegans , Mitocôndrias , Transdução de Sinais , Animais , Acetilcolina/metabolismo , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Receptor Nicotínico de Acetilcolina alfa7/genética , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/genética , Caenorhabditis elegans/fisiologia , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Ácido gama-Aminobutírico/metabolismo , Intestinos/fisiologia , Mitocôndrias/metabolismo , Neurônios/metabolismo , Estresse Oxidativo , Receptores de GABA-B/metabolismo , Receptores de GABA-B/genética , Estresse Fisiológico , Transmissão Sináptica/fisiologia , Resposta a Proteínas não Dobradas
8.
PLoS Genet ; 20(8): e1011362, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39110773

RESUMO

A recently reported Schizophrenia-associated genetic variant in the 3'UTR of the human furin gene, a homolog of C. elegans kpc-1, highlights an important role of the furin 3'UTR in neuronal development. We isolate three kpc-1 mutants that display abnormal dendrite arborization in PVD neurons and defective male mating behaviors. We show that the kpc-1 3'UTR participates in dendrite branching and self-avoidance. The kpc-1 3'UTR facilitates mRNA localization to branching points and contact points between sibling dendrites and promotes translation efficiency. A predicted secondary structural motif in the kpc-1 3'UTR is required for dendrite self-avoidance. Animals with over-expression of DMA-1, a PVD dendrite receptor, exhibit similar dendrite branching and self-avoidance defects that are suppressed with kpc-1 over-expression. Our results support a model in which KPC-1 proteins are synthesized at branching points and contact points to locally down-regulate DMA-1 receptors to promote dendrite branching and self-avoidance of a mechanosensory neuron important for male courtship.


Assuntos
Regiões 3' não Traduzidas , Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Corte , Dendritos , RNA Mensageiro , Animais , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Masculino , Dendritos/metabolismo , Dendritos/genética , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Regiões 3' não Traduzidas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Biossíntese de Proteínas , Comportamento Sexual Animal/fisiologia , Humanos , Mutação , Proteínas de Membrana
9.
Methods Mol Biol ; 2845: 67-77, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39115658

RESUMO

The autophagy-lysosomal pathway enables the controlled degradation of cellular contents. Nucleophagy is the selective autophagic recycling of nuclear components upon delivery to the lysosome. Although methods to monitor and quantify autophagy as well as selective types of autophagy have been developed and implemented in cells and in vivo, methods monitoring nucleophagy remain scarce. Here, we describe a procedure to monitor the autophagic engagement of an endogenous nuclear envelope component, i.e., ANC-1, the nematode homologue of the mammalian Nesprins in vivo, utilizing super-resolution microscopy.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Animais , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Autofagia/fisiologia , Lisossomos/metabolismo , Membrana Nuclear/metabolismo , Núcleo Celular/metabolismo , Macroautofagia
10.
Methods Mol Biol ; 2845: 55-66, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39115657

RESUMO

Preserving mitochondrial homeostasis is vital, particularly for the energetically demanding and metabolically active nerve cells. Mitophagy, the selective autophagic removal of mitochondria, stands out as a prominent mechanism for efficient mitochondrial turnover, which is crucial for proper neuronal development and function. Dysfunctional mitochondria and disrupted mitophagy pathways have been linked to a diverse array of neurological disorders. The nematode Caenorhabditis elegans, with its well-defined nervous system, serves as an excellent model to unravel the intricate involvement of mitophagy in developing neurons. This chapter describes the use of Rosella biosensor in C. elegans to monitor neuronal mitophagy, providing a user-friendly platform for screening genes and drugs affecting mitophagic pathways under physiological conditions or in the context of neurodevelopmental pathologies.


Assuntos
Caenorhabditis elegans , Mitocôndrias , Mitofagia , Neurônios , Animais , Caenorhabditis elegans/metabolismo , Neurônios/metabolismo , Neurônios/citologia , Mitocôndrias/metabolismo , Técnicas Biossensoriais/métodos , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética
11.
Nat Commun ; 15(1): 6869, 2024 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-39127676

RESUMO

In C. elegans mechanisms by which peripheral organs relay internal state information to the nervous system remain unknown, although strong evidence suggests that such signals do exist. Here we report the discovery of a peptide of the ancestral insulin superfamily called INS-7 that functions as an enteroendocrine peptide and is secreted from specialized cells of the intestine. INS-7 secretion is stimulated by food withdrawal, increases during fasting and acts as a bona fide gut-to-brain peptide that attenuates the release of a neuropeptide that drives fat loss in the periphery. Thus, INS-7 functions as a homeostatic signal from the intestine that gates the neuronal drive to stimulate fat loss during food shortage. Mechanistically, INS-7 functions as an antagonist at the canonical DAF-2 receptor and functions via FOXO and AMPK signaling in ASI neurons. Phylogenetic analysis suggests that INS-7 bears greater resemblance to members of the broad insulin/relaxin superfamily than to conventional mammalian insulin and IGF peptides. The discovery of an endogenous insulin antagonist secreted by specialized intestinal cells with enteroendocrine functions suggests unexpected and important properties of the intestine and its role in directing neuronal functions.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Homeostase , Insulina , Neurônios , Animais , Neurônios/metabolismo , Neurônios/efeitos dos fármacos , Insulina/metabolismo , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Receptor de Insulina/metabolismo , Receptor de Insulina/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/efeitos dos fármacos , Neuropeptídeos/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Fatores de Transcrição Forkhead/genética , Intestinos , Filogenia , Jejum , Mucosa Intestinal/metabolismo
12.
Nat Commun ; 15(1): 6884, 2024 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-39128930

RESUMO

Fertility requires the faithful proliferation of germ cells and their differentiation into gametes. Controlling these cellular states demands precise timing and expression of gene networks. Nucleic acid binding proteins (NBPs) play critical roles in gene expression networks that influence germ cell development. There has, however, been no functional analysis of the entire NBP repertoire in controlling in vivo germ cell development. Here, we analyzed germ cell states and germline architecture to systematically investigate the function of 364 germline-expressed NBPs in the Caenorhabditis elegans germ line. Using germline-specific knockdown, automated germ cell counting, and high-content analysis of germ cell nuclei and plasma membrane organization, we identify 156 NBPs with discrete autonomous germline functions. By identifying NBPs that control the germ cell cycle, proliferation, differentiation, germline structure and fertility, we have created an atlas for mechanistic dissection of germ cell behavior and gamete production.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Células Germinativas , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Células Germinativas/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Regulação da Expressão Gênica no Desenvolvimento , Diferenciação Celular/genética , Proteínas de Ligação a RNA/metabolismo , Proteínas de Ligação a RNA/genética , Fertilidade/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , Proliferação de Células/genética , Ciclo Celular/genética , Masculino , Redes Reguladoras de Genes
13.
Methods Mol Biol ; 2818: 195-212, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39126476

RESUMO

The Caenorhabditis elegans germline is arranged spatiotemporally and is therefore a powerful model system for the interrogation of meiotic molecular dynamics. Coupling this property with the temporal control that the auxin-inducible degron (AID) system allows can unveil new/unappreciated roles for critical meiotic factors in specific germline regions. Here we describe a widely used approach for the introduction of degron tags to specific targets and provide a procedure for applying the AID system to C. elegans meiotic DSB repair dynamics in the germline.


Assuntos
Caenorhabditis elegans , Quebras de DNA de Cadeia Dupla , Meiose , Caenorhabditis elegans/genética , Animais , Reparo do DNA , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Células Germinativas/metabolismo
14.
Development ; 151(13)2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38984540

RESUMO

Germ granules have been hypothesized to deliver mRNAs of germ cell fate determinants to primordial germ cells. Now, a new study in Development finds that many mRNAs enriched in germ granules are not involved in germline development in Caenorhabditis elegans. To find out more about the story behind the paper, we caught up with first author Alyshia Scholl, second author Yihong Liu and corresponding author Geraldine Seydoux, Professor at Johns Hopkins University School of Medicine.


Assuntos
Caenorhabditis elegans , Animais , Caenorhabditis elegans/genética , Células Germinativas/metabolismo , Biologia do Desenvolvimento/história , História do Século XXI , História do Século XX , Humanos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética
15.
Dis Model Mech ; 17(8)2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-38946472

RESUMO

Ras genes are important oncogenes that are frequently mutated in cancer. Human oncogenic variants exhibit functional distinctions in terms of their representation in different cancer types, impact on cellular targets and sensitivity to pharmacological treatments. However, how these distinct variants influence and respond to the cellular networks in which they are embedded is poorly understood. To identify novel participants in the complex interplay between Ras genotype and cell interaction networks in vivo, we have developed and tested an experimental framework using a simple vulva-development assay in the nematode C. elegans. Using this system, we evaluated a set of Ras oncogenic substitution changes at G12, G13 and Q61. We found that these variants fall into distinct groups based on phenotypic differences, sensitivity to gene dosage and inhibition of the downstream kinase MEK and their response to genetic modulators that influence Ras activity in a non-autonomous manner. Together, our results demonstrated that oncogenic C. elegans Ras variants exhibit clear distinctions in how they interface with the vulva-development network and showed that extracellular modulators yield variant-restricted effects in vivo.


Assuntos
Caenorhabditis elegans , Vulva , Proteínas ras , Caenorhabditis elegans/genética , Animais , Vulva/patologia , Vulva/metabolismo , Proteínas ras/metabolismo , Proteínas ras/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Feminino , Fenótipo , Mutação/genética , Oncogenes/genética , Humanos
16.
Nat Genet ; 56(8): 1737-1749, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39039278

RESUMO

The structural maintenance of chromosome (SMC) complexes-cohesin and condensins-are crucial for chromosome separation and compaction during cell division. During the interphase, mammalian cohesins additionally fold the genome into loops and domains. Here we show that, in Caenorhabditis elegans, a species with holocentric chromosomes, condensin I is the primary, long-range loop extruder. The loss of condensin I and its X-specific variant, condensin IDC, leads to genome-wide decompaction, chromosome mixing and disappearance of X-specific topologically associating domains, while reinforcing fine-scale epigenomic compartments. In addition, condensin I/IDC inactivation led to the upregulation of X-linked genes and unveiled nuclear bodies grouping together binding sites for the X-targeting loading complex of condensin IDC. C. elegans condensin I/IDC thus uniquely organizes holocentric interphase chromosomes, akin to cohesin in mammals, as well as regulates X-chromosome gene expression.


Assuntos
Adenosina Trifosfatases , Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Proteínas de Ligação a DNA , Complexos Multiproteicos , Cromossomo X , Animais , Caenorhabditis elegans/genética , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Cromossomo X/genética , Proteínas Cromossômicas não Histona/metabolismo , Proteínas Cromossômicas não Histona/genética , Coesinas , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Interfase/genética , Genoma Helmíntico , Genes Ligados ao Cromossomo X , Cromossomos/genética
17.
Nutrients ; 16(14)2024 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-39064700

RESUMO

Velvet antler, a traditional tonic widely used in East Asia for its health benefits, is explored in this study for its protective effects against hypoxia-induced damage using Caenorhabditis elegans (C. elegans) as a model. Hypoxia, characterized by low oxygen availability, induces significant physiological stress and potential tissue damage. Our research demonstrates that methanol extracts from velvet antler (MEs) enhance the survival of C. elegans under hypoxic conditions. This enhancement is achieved through the stabilization of hypoxia-inducible factor-1 (HIF-1) and the promotion of lipid accumulation, both of which are crucial for mitigating cellular damage. Specifically, MEs improve mitochondrial function, increase ATP production, and aid in the recovery of physical activity in C. elegans post-hypoxia or following hypoxia-reoxygenation (HR). The pivotal role of HIF-1 is underscored by the loss of these protective effects when HIF-1 function is inhibited. Additionally, our findings reveal that the gene related to lipid metabolism, ech-8, significantly contributes to the lipid accumulation that enhances resilience to hypoxia in C. elegans treated with MEs. These results not only highlight the therapeutic potential of velvet antler in modern medical applications, particularly for conditions involving hypoxic stress, but also provide insights into the molecular mechanisms by which MEs confer protection against hypoxic damage.


Assuntos
Chifres de Veado , Caenorhabditis elegans , Hipóxia , Metabolismo dos Lipídeos , Metanol , Animais , Caenorhabditis elegans/efeitos dos fármacos , Chifres de Veado/química , Metabolismo dos Lipídeos/efeitos dos fármacos , Hipóxia/metabolismo , Metanol/química , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Fator 1 Induzível por Hipóxia/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Substâncias Protetoras/farmacologia
18.
Commun Biol ; 7(1): 903, 2024 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-39060347

RESUMO

Pathological tau disrupts protein homeostasis (proteostasis) within neurons in Alzheimer's disease (AD) and related disorders. We previously showed constitutive activation of the endoplasmic reticulum unfolded protein response (UPRER) transcription factor XBP-1s rescues tauopathy-related proteostatic disruption in a tau transgenic Caenorhabditis elegans (C. elegans) model of human tauopathy. XBP-1s promotes clearance of pathological tau, and loss of function of the ATF-6 branch of the UPRER prevents XBP-1s rescue of tauopathy in C. elegans. We conducted transcriptomic analysis of tau transgenic and xbp-1s transgenic C. elegans and found 116 putative target genes significantly upregulated by constitutively active XBP-1s. Among these were five candidate XBP-1s target genes with human orthologs and a previously known association with ATF6 (csp-1, dnj-28, hsp-4, ckb-2, and lipl-3). We examined the functional involvement of these targets in XBP-1s-mediated tauopathy suppression and found loss of function in any one of these genes completely disrupts XBP-1s suppression of tauopathy. Further, we demonstrate upregulation of HSP-4, C. elegans BiP, partially rescues tauopathy independent of other changes in the transcriptional network. Understanding how the UPRER modulates pathological tau accumulation will inform neurodegenerative disease mechanisms and direct further study in mammalian systems with the long-term goal of identifying therapeutic targets in human tauopathies.


Assuntos
Animais Geneticamente Modificados , Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Tauopatias , Resposta a Proteínas não Dobradas , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Animais , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Tauopatias/metabolismo , Tauopatias/genética , Humanos , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/genética , Proteínas tau/metabolismo , Proteínas tau/genética , Proteína 1 de Ligação a X-Box/metabolismo , Proteína 1 de Ligação a X-Box/genética , Modelos Animais de Doenças , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Regulação da Expressão Gênica , Proteínas de Transporte
19.
Life Sci Alliance ; 7(9)2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38960623

RESUMO

In many animal species, the oocyte meiotic spindle, which is required for chromosome segregation, forms without centrosomes. In some systems, Ran-GEF on chromatin initiates spindle assembly. We found that in Caenorhabditis elegans oocytes, endogenously-tagged Ran-GEF dissociates from chromatin during spindle assembly but re-associates during meiotic anaphase. Meiotic spindle assembly occurred after auxin-induced degradation of Ran-GEF, but anaphase I was faster than controls and extrusion of the first polar body frequently failed. In search of a possible alternative pathway for spindle assembly, we found that soluble tubulin concentrates in the nuclear volume during germinal vesicle breakdown. We found that the concentration of soluble tubulin in the metaphase spindle region is enclosed by ER sheets which exclude cytoplasmic organelles including mitochondria and yolk granules. Measurement of the volume occupied by yolk granules and mitochondria indicated that volume exclusion would be sufficient to explain the concentration of tubulin in the spindle volume. We suggest that this concentration of soluble tubulin may be a redundant mechanism promoting spindle assembly near chromosomes.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Meiose , Oócitos , Tubulina (Proteína) , Proteína ran de Ligação ao GTP , Animais , Anáfase , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Cromatina/metabolismo , Segregação de Cromossomos , Guanosina Trifosfato/metabolismo , Meiose/fisiologia , Oócitos/metabolismo , Prometáfase , Proteína ran de Ligação ao GTP/metabolismo , Fuso Acromático/metabolismo , Tubulina (Proteína)/metabolismo
20.
Elife ; 122024 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-38963411

RESUMO

Precise developmental timing control is essential for organism formation and function, but its mechanisms are unclear. In C. elegans, the microRNA lin-4 critically regulates developmental timing by post-transcriptionally downregulating the larval-stage-fate controller LIN-14. However, the mechanisms triggering the activation of lin-4 expression toward the end of the first larval stage remain unknown. We demonstrate that the transmembrane transcription factor MYRF-1 is necessary for lin-4 activation. MYRF-1 is initially localized on the cell membrane, and its increased cleavage and nuclear accumulation coincide with lin-4 expression timing. MYRF-1 regulates lin-4 expression cell-autonomously and hyperactive MYRF-1 can prematurely drive lin-4 expression in embryos and young first-stage larvae. The tandem lin-4 promoter DNA recruits MYRF-1GFP to form visible loci in the nucleus, suggesting that MYRF-1 directly binds to the lin-4 promoter. Our findings identify a crucial link in understanding developmental timing regulation and establish MYRF-1 as a key regulator of lin-4 expression.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Regulação da Expressão Gênica no Desenvolvimento , MicroRNAs , Fatores de Transcrição , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/crescimento & desenvolvimento , Animais , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , MicroRNAs/metabolismo , MicroRNAs/genética , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Regiões Promotoras Genéticas , Transcrição Gênica , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Núcleo Celular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA