Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.873
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 187(3): 659-675.e18, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38215760

RESUMEN

The electron transport chain (ETC) of mitochondria, bacteria, and archaea couples electron flow to proton pumping and is adapted to diverse oxygen environments. Remarkably, in mice, neurological disease due to ETC complex I dysfunction is rescued by hypoxia through unknown mechanisms. Here, we show that hypoxia rescue and hyperoxia sensitivity of complex I deficiency are evolutionarily conserved to C. elegans and are specific to mutants that compromise the electron-conducting matrix arm. We show that hypoxia rescue does not involve the hypoxia-inducible factor pathway or attenuation of reactive oxygen species. To discover the mechanism, we use C. elegans genetic screens to identify suppressor mutations in the complex I accessory subunit NDUFA6/nuo-3 that phenocopy hypoxia rescue. We show that NDUFA6/nuo-3(G60D) or hypoxia directly restores complex I forward activity, with downstream rescue of ETC flux and, in some cases, complex I levels. Additional screens identify residues within the ubiquinone binding pocket as being required for the rescue by NDUFA6/nuo-3(G60D) or hypoxia. This reveals oxygen-sensitive coupling between an accessory subunit and the quinone binding pocket of complex I that can restore forward activity in the same manner as hypoxia.


Asunto(s)
Caenorhabditis elegans , Complejo I de Transporte de Electrón , Hipoxia , Animales , Ratones , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Complejo I de Transporte de Electrón/metabolismo , Hipoxia/genética , Hipoxia/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Oxígeno/metabolismo
2.
Cell ; 187(17): 4605-4620.e17, 2024 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-38959891

RESUMEN

The ability of mitochondria to coordinate stress responses across tissues is critical for health. In C. elegans, neurons experiencing mitochondrial stress elicit an inter-tissue signaling pathway through the release of mitokine signals, such as serotonin or the Wnt ligand EGL-20, which activate the mitochondrial unfolded protein response (UPRMT) in the periphery to promote organismal health and lifespan. We find that germline mitochondria play a surprising role in neuron-to-periphery UPRMT signaling. Specifically, we find that germline mitochondria signal downstream of neuronal mitokines, Wnt and serotonin, and upstream of lipid metabolic pathways in the periphery to regulate UPRMT activation. We also find that the germline tissue itself is essential for UPRMT signaling. We propose that the germline has a central signaling role in coordinating mitochondrial stress responses across tissues, and germline mitochondria play a defining role in this coordination because of their inherent roles in germline integrity and inter-tissue signaling.


Asunto(s)
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Células Germinativas , Mitocondrias , Transducción de Señal , Respuesta de Proteína Desplegada , Animales , Caenorhabditis elegans/metabolismo , Mitocondrias/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Células Germinativas/metabolismo , Neuronas/metabolismo , Serotonina/metabolismo , Proteínas Wnt/metabolismo
3.
Cell ; 186(12): 2574-2592.e20, 2023 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-37192620

RESUMEN

Serotonin influences many aspects of animal behavior. But how serotonin acts on its diverse receptors across the brain to modulate global activity and behavior is unknown. Here, we examine how serotonin release in C. elegans alters brain-wide activity to induce foraging behaviors, like slow locomotion and increased feeding. Comprehensive genetic analyses identify three core serotonin receptors (MOD-1, SER-4, and LGC-50) that induce slow locomotion upon serotonin release and others (SER-1, SER-5, and SER-7) that interact with them to modulate this behavior. SER-4 induces behavioral responses to sudden increases in serotonin release, whereas MOD-1 induces responses to persistent release. Whole-brain imaging reveals widespread serotonin-associated brain dynamics, spanning many behavioral networks. We map all sites of serotonin receptor expression in the connectome, which, together with synaptic connectivity, helps predict which neurons show serotonin-associated activity. These results reveal how serotonin acts at defined sites across a connectome to modulate brain-wide activity and behavior.


Asunto(s)
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Animales , Caenorhabditis elegans/metabolismo , Serotonina/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Receptores de Serotonina/genética , Receptores de Serotonina/metabolismo , Conducta Animal/fisiología , Encéfalo/metabolismo
4.
Cell ; 186(19): 4134-4151.e31, 2023 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-37607537

RESUMEN

Changes in an animal's behavior and internal state are accompanied by widespread changes in activity across its brain. However, how neurons across the brain encode behavior and how this is impacted by state is poorly understood. We recorded brain-wide activity and the diverse motor programs of freely moving C. elegans and built probabilistic models that explain how each neuron encodes quantitative behavioral features. By determining the identities of the recorded neurons, we created an atlas of how the defined neuron classes in the C. elegans connectome encode behavior. Many neuron classes have conjunctive representations of multiple behaviors. Moreover, although many neurons encode current motor actions, others integrate recent actions. Changes in behavioral state are accompanied by widespread changes in how neurons encode behavior, and we identify these flexible nodes in the connectome. Our results provide a global map of how the cell types across an animal's brain encode its behavior.


Asunto(s)
Caenorhabditis elegans , Conectoma , Animales , Encéfalo/citología , Encéfalo/metabolismo , Modelos Estadísticos , Neuronas/metabolismo
5.
Cell ; 186(13): 2911-2928.e20, 2023 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-37269832

RESUMEN

Animals with complex nervous systems demand sleep for memory consolidation and synaptic remodeling. Here, we show that, although the Caenorhabditis elegans nervous system has a limited number of neurons, sleep is necessary for both processes. In addition, it is unclear if, in any system, sleep collaborates with experience to alter synapses between specific neurons and whether this ultimately affects behavior. C. elegans neurons have defined connections and well-described contributions to behavior. We show that spaced odor-training and post-training sleep induce long-term memory. Memory consolidation, but not acquisition, requires a pair of interneurons, the AIYs, which play a role in odor-seeking behavior. In worms that consolidate memory, both sleep and odor conditioning are required to diminish inhibitory synaptic connections between the AWC chemosensory neurons and the AIYs. Thus, we demonstrate in a living organism that sleep is required for events immediately after training that drive memory consolidation and alter synaptic structures.


Asunto(s)
Caenorhabditis elegans , Odorantes , Animales , Caenorhabditis elegans/fisiología , Olfato , Sueño/fisiología , Sinapsis/fisiología
6.
Cell ; 184(20): 5122-5137.e17, 2021 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-34534446

RESUMEN

Natural goal-directed behaviors often involve complex sequences of many stimulus-triggered components. Understanding how brain circuits organize such behaviors requires mapping the interactions between an animal, its environment, and its nervous system. Here, we use brain-wide neuronal imaging to study the full performance of mating by the C. elegans male. We show that as mating unfolds in a sequence of component behaviors, the brain operates similarly between instances of each component but distinctly between different components. When the full sensory and behavioral context is taken into account, unique roles emerge for each neuron. Functional correlations between neurons are not fixed but change with behavioral dynamics. From individual neurons to circuits, our study shows how diverse brain-wide dynamics emerge from the integration of sensory perception and motor actions in their natural context.


Asunto(s)
Encéfalo/fisiología , Caenorhabditis elegans/fisiología , Sensación/fisiología , Conducta Sexual Animal/fisiología , Animales , Mapeo Encefálico , Copulación/fisiología , Cortejo , Bases de Datos como Asunto , Retroalimentación , Femenino , Masculino , Modelos Biológicos , Movimiento , Neuronas/fisiología , Descanso , Procesamiento de Señales Asistido por Computador , Sinapsis/fisiología , Vulva/fisiología
7.
Cell ; 184(18): 4697-4712.e18, 2021 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-34363756

RESUMEN

Animals face both external and internal dangers: pathogens threaten from the environment, and unstable genomic elements threaten from within. C. elegans protects itself from pathogens by "reading" bacterial small RNAs, using this information to both induce avoidance and transmit memories for four generations. Here, we found that memories can be transferred from either lysed animals or from conditioned media to naive animals via Cer1 retrotransposon-encoded virus-like particles. Moreover, Cer1 functions internally at the step of transmission of information from the germline to neurons and is required for learned avoidance. The presence of the Cer1 retrotransposon in wild C. elegans strains correlates with the ability to learn and inherit small-RNA-induced pathogen avoidance. Together, these results suggest that C. elegans has co-opted a potentially dangerous retrotransposon to instead protect itself and its progeny from a common pathogen through its inter-tissue signaling ability, hijacking this genomic element for its own adaptive immunity benefit.


Asunto(s)
Elementos Transponibles de ADN/genética , Transferencia de Gen Horizontal/genética , Patrón de Herencia/genética , Memoria/fisiología , Animales , Reacción de Prevención , Conducta Animal , Caenorhabditis elegans/genética , Caenorhabditis elegans/fisiología , Vesículas Extracelulares/metabolismo , Regulación de la Expresión Génica , Genoma , Células Germinativas/metabolismo , ARN/metabolismo , Interferencia de ARN , Virión/metabolismo
8.
Cell ; 184(16): 4329-4347.e23, 2021 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-34237253

RESUMEN

We have produced gene expression profiles of all 302 neurons of the C. elegans nervous system that match the single-cell resolution of its anatomy and wiring diagram. Our results suggest that individual neuron classes can be solely identified by combinatorial expression of specific gene families. For example, each neuron class expresses distinct codes of ∼23 neuropeptide genes and ∼36 neuropeptide receptors, delineating a complex and expansive "wireless" signaling network. To demonstrate the utility of this comprehensive gene expression catalog, we used computational approaches to (1) identify cis-regulatory elements for neuron-specific gene expression and (2) reveal adhesion proteins with potential roles in process placement and synaptic specificity. Our expression data are available at https://cengen.org and can be interrogated at the web application CengenApp. We expect that this neuron-specific directory of gene expression will spur investigations of underlying mechanisms that define anatomy, connectivity, and function throughout the C. elegans nervous system.


Asunto(s)
Caenorhabditis elegans/metabolismo , Sistema Nervioso/metabolismo , Animales , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Colorantes Fluorescentes/metabolismo , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Larva/metabolismo , Neuronas/metabolismo , Neuropéptidos/genética , Neuropéptidos/metabolismo , Motivos de Nucleótidos/genética , RNA-Seq , Secuencias Reguladoras de Ácidos Nucleicos/genética , Transducción de Señal/genética , Factores de Transcripción/metabolismo , Transcripción Genética
9.
Cell ; 176(5): 1014-1025.e12, 2019 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-30794773

RESUMEN

Bioactive molecules can pass between microbiota and host to influence host cellular functions. However, general principles of interspecies communication have not been discovered. We show here in C. elegans that nitric oxide derived from resident bacteria promotes widespread S-nitrosylation of the host proteome. We further show that microbiota-dependent S-nitrosylation of C. elegans Argonaute protein (ALG-1)-at a site conserved and S-nitrosylated in mammalian Argonaute 2 (AGO2)-alters its function in controlling gene expression via microRNAs. By selectively eliminating nitric oxide generation by the microbiota or S-nitrosylation in ALG-1, we reveal unforeseen effects on host development. Thus, the microbiota can shape the post-translational landscape of the host proteome to regulate microRNA activity, gene expression, and host development. Our findings suggest a general mechanism by which the microbiota may control host cellular functions, as well as a new role for gasotransmitters.


Asunto(s)
Interacciones Microbiota-Huesped/genética , MicroARNs/metabolismo , Óxido Nítrico/metabolismo , Animales , Proteínas Argonautas/genética , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Células HEK293 , Células HeLa , Humanos , MicroARNs/fisiología , Microbiota/genética , Óxido Nítrico/fisiología , Procesamiento Proteico-Postraduccional/genética , Proteoma/genética , Proteoma/metabolismo , Proteómica/métodos , Proteínas de Unión al ARN/genética
10.
Cell ; 177(7): 1814-1826.e15, 2019 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-31178120

RESUMEN

It is unknown whether the activity of the nervous system can be inherited. In Caenorhabditis elegans nematodes, parental responses can transmit heritable small RNAs that regulate gene expression transgenerationally. In this study, we show that a neuronal process can impact the next generations. Neurons-specific synthesis of RDE-4-dependent small RNAs regulates germline amplified endogenous small interfering RNAs (siRNAs) and germline gene expression for multiple generations. Further, the production of small RNAs in neurons controls the chemotaxis behavior of the progeny for at least three generations via the germline Argonaute HRDE-1. Among the targets of these small RNAs, we identified the conserved gene saeg-2, which is transgenerationally downregulated in the germline. Silencing of saeg-2 following neuronal small RNA biogenesis is required for chemotaxis under stress. Thus, we propose a small-RNA-based mechanism for communication of neuronal processes transgenerationally.


Asunto(s)
Conducta Animal , Caenorhabditis elegans , Neuronas/metabolismo , ARN de Helminto , ARN Pequeño no Traducido , Animales , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Caenorhabditis elegans/citología , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Neuronas/citología , ARN de Helminto/biosíntesis , ARN de Helminto/genética , ARN Pequeño no Traducido/biosíntesis , ARN Pequeño no Traducido/genética
11.
Cell ; 178(6): 1299-1312.e29, 2019 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-31474368

RESUMEN

Metformin is the first-line therapy for treating type 2 diabetes and a promising anti-aging drug. We set out to address the fundamental question of how gut microbes and nutrition, key regulators of host physiology, affect the effects of metformin. Combining two tractable genetic models, the bacterium E. coli and the nematode C. elegans, we developed a high-throughput four-way screen to define the underlying host-microbe-drug-nutrient interactions. We show that microbes integrate cues from metformin and the diet through the phosphotransferase signaling pathway that converges on the transcriptional regulator Crp. A detailed experimental characterization of metformin effects downstream of Crp in combination with metabolic modeling of the microbiota in metformin-treated type 2 diabetic patients predicts the production of microbial agmatine, a regulator of metformin effects on host lipid metabolism and lifespan. Our high-throughput screening platform paves the way for identifying exploitable drug-nutrient-microbiome interactions to improve host health and longevity through targeted microbiome therapies. VIDEO ABSTRACT.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Microbioma Gastrointestinal/efectos de los fármacos , Interacciones Microbiota-Huesped/efectos de los fármacos , Hipoglucemiantes/uso terapéutico , Metformina/uso terapéutico , Agmatina/metabolismo , Animales , Caenorhabditis elegans/microbiología , Proteína Receptora de AMP Cíclico , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Humanos , Hipoglucemiantes/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Longevidad/efectos de los fármacos , Metformina/farmacología , Nutrientes/metabolismo
12.
Cell ; 176(1-2): 85-97.e14, 2019 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-30580965

RESUMEN

Animals must respond to the ingestion of food by generating adaptive behaviors, but the role of gut-brain signaling in behavioral regulation is poorly understood. Here, we identify conserved ion channels in an enteric serotonergic neuron that mediate its responses to food ingestion and decipher how these responses drive changes in foraging behavior. We show that the C. elegans serotonergic neuron NSM acts as an enteric sensory neuron that acutely detects food ingestion. We identify the novel and conserved acid-sensing ion channels (ASICs) DEL-7 and DEL-3 as NSM-enriched channels required for feeding-dependent NSM activity, which in turn drives slow locomotion while animals feed. Point mutations that alter the DEL-7 channel change NSM dynamics and associated behavioral dynamics of the organism. This study provides causal links between food ingestion, molecular and physiological properties of an enteric serotonergic neuron, and adaptive feeding behaviors, yielding a new view of how enteric neurons control behavior.


Asunto(s)
Canales Iónicos Sensibles al Ácido/metabolismo , Sistema Nervioso Entérico/metabolismo , Conducta Alimentaria/fisiología , Canales Iónicos Sensibles al Ácido/fisiología , Animales , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/fisiología , Proteínas de Caenorhabditis elegans/metabolismo , Sistema Nervioso Entérico/fisiología , Alimentos , Canales Iónicos/metabolismo , Canales Iónicos/fisiología , Locomoción , Neuronas/metabolismo , Células Receptoras Sensoriales/metabolismo , Neuronas Serotoninérgicas/metabolismo , Neuronas Serotoninérgicas/fisiología , Serotonina , Transducción de Señal
13.
Cell ; 175(1): 57-70.e17, 2018 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-30220455

RESUMEN

Neurons in Caenorhabditis elegans and other nematodes have been thought to lack classical action potentials. Unexpectedly, we observe membrane potential spikes with defining characteristics of action potentials in C. elegans AWA olfactory neurons recorded under current-clamp conditions. Ion substitution experiments, mutant analysis, pharmacology, and modeling indicate that AWA fires calcium spikes, which are initiated by EGL-19 voltage-gated CaV1 calcium channels and terminated by SHK-1 Shaker-type potassium channels. AWA action potentials result in characteristic signals in calcium imaging experiments. These calcium signals are also observed when intact animals are exposed to odors, suggesting that natural odor stimuli induce AWA spiking. The stimuli that elicit action potentials match AWA's specialized function in climbing odor gradients. Our results provide evidence that C. elegans neurons can encode information through regenerative all-or-none action potentials, expand the computational repertoire of its nervous system, and inform future modeling of its neural coding and network dynamics.


Asunto(s)
Potenciales de Acción/fisiología , Nervio Olfatorio/fisiología , Olfato/fisiología , Animales , Caenorhabditis elegans/fisiología , Proteínas de Caenorhabditis elegans/metabolismo , Calcio/metabolismo , Canales de Calcio/fisiología , Quimiotaxis/fisiología , Potenciales de la Membrana/fisiología , Odorantes , Neuronas Receptoras Olfatorias/metabolismo
14.
Cell ; 169(3): 470-482.e13, 2017 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-28431247

RESUMEN

Aging is attended by a progressive decline in protein homeostasis (proteostasis), aggravating the risk for protein aggregation diseases. To understand the coordination between proteome imbalance and longevity, we addressed the mechanistic role of the quality-control ubiquitin ligase CHIP, which is a key regulator of proteostasis. We observed that CHIP deficiency leads to increased levels of the insulin receptor (INSR) and reduced lifespan of worms and flies. The membrane-bound INSR regulates the insulin and IGF1 signaling (IIS) pathway and thereby defines metabolism and aging. INSR is a direct target of CHIP, which triggers receptor monoubiquitylation and endocytic-lysosomal turnover to promote longevity. However, upon proteotoxic stress conditions and during aging, CHIP is recruited toward disposal of misfolded proteins, reducing its capacity to degrade the INSR. Our study indicates a competitive relationship between proteostasis and longevity regulation through CHIP-assisted proteolysis, providing a mechanistic concept for understanding the impact of proteome imbalance on aging.


Asunto(s)
Envejecimiento , Antígenos CD/metabolismo , Receptor de Insulina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Caenorhabditis elegans , Drosophila melanogaster , Endocitosis , Humanos , Longevidad , Lisosomas/metabolismo , Proteolisis , Proteoma , Transducción de Señal , Somatomedinas , Ubiquitinación
15.
Cell ; 169(3): 431-441.e8, 2017 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-28431244

RESUMEN

The human microbiota greatly affects physiology and disease; however, the contribution of bacteria to the response to chemotherapeutic drugs remains poorly understood. Caenorhabditis elegans and its bacterial diet provide a powerful system to study host-bacteria interactions. Here, we use this system to study how bacteria affect the C. elegans response to chemotherapeutics. We find that different bacterial species can increase the response to one drug yet decrease the effect of another. We perform genetic screens in two bacterial species using three chemotherapeutic drugs: 5-fluorouracil (5-FU), 5-fluoro-2'-deoxyuridine (FUDR), and camptothecin (CPT). We find numerous bacterial nucleotide metabolism genes that affect drug efficacy in C. elegans. Surprisingly, we find that 5-FU and FUDR act through bacterial ribonucleotide metabolism to elicit their cytotoxic effects in C. elegans rather than by thymineless death or DNA damage. Our study provides a blueprint for characterizing the role of bacteria in the host response to chemotherapeutics.


Asunto(s)
Antineoplásicos/metabolismo , Caenorhabditis elegans/microbiología , Comamonas/metabolismo , Escherichia coli/metabolismo , Microbioma Gastrointestinal , Animales , Antineoplásicos/farmacología , Camptotecina/metabolismo , Camptotecina/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Comamonas/genética , Desoxiuridina/análogos & derivados , Desoxiuridina/metabolismo , Desoxiuridina/farmacología , Dieta , Escherichia coli/genética , Fluorouracilo/metabolismo , Fluorouracilo/farmacología , Humanos , Modelos Animales , Nucleósidos de Pirimidina/metabolismo
16.
Cell ; 169(3): 442-456.e18, 2017 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-28431245

RESUMEN

Fluoropyrimidines are the first-line treatment for colorectal cancer, but their efficacy is highly variable between patients. We queried whether gut microbes, a known source of inter-individual variability, impacted drug efficacy. Combining two tractable genetic models, the bacterium E. coli and the nematode C. elegans, we performed three-way high-throughput screens that unraveled the complexity underlying host-microbe-drug interactions. We report that microbes can bolster or suppress the effects of fluoropyrimidines through metabolic drug interconversion involving bacterial vitamin B6, B9, and ribonucleotide metabolism. Also, disturbances in bacterial deoxynucleotide pools amplify 5-FU-induced autophagy and cell death in host cells, an effect regulated by the nucleoside diphosphate kinase ndk-1. Our data suggest a two-way bacterial mediation of fluoropyrimidine effects on host metabolism, which contributes to drug efficacy. These findings highlight the potential therapeutic power of manipulating intestinal microbiota to ensure host metabolic health and treat disease.


Asunto(s)
Antineoplásicos/metabolismo , Escherichia coli/metabolismo , Fluorouracilo/metabolismo , Microbioma Gastrointestinal , Animales , Autofagia , Caenorhabditis elegans , Muerte Celular , Neoplasias Colorrectales/tratamiento farmacológico , Dieta , Escherichia coli/enzimología , Escherichia coli/genética , Humanos , Modelos Animales , Pentosiltransferasa/genética
17.
Cell ; 165(2): 396-409, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-27020753

RESUMEN

Multiple division cycles without growth are a characteristic feature of early embryogenesis. The female germline loads proteins and RNAs into oocytes to support these divisions, which lack many quality control mechanisms operating in somatic cells undergoing growth. Here, we describe a small RNA-Argonaute pathway that ensures early embryonic divisions in C. elegans by employing catalytic slicing activity to broadly tune, instead of silence, germline gene expression. Misregulation of one target, a kinesin-13 microtubule depolymerase, underlies a major phenotype associated with pathway loss. Tuning of target transcript levels is guided by the density of homologous small RNAs, whose generation must ultimately be related to target sequence. Thus, the tuning action of a small RNA-catalytic Argonaute pathway generates oocytes capable of supporting embryogenesis. We speculate that the specialized nature of germline chromatin led to the emergence of small RNA-catalytic Argonaute pathways in the female germline as a post-transcriptional control layer to optimize oocyte composition.


Asunto(s)
Caenorhabditis elegans/embriología , Caenorhabditis elegans/metabolismo , Embrión no Mamífero/metabolismo , Redes y Vías Metabólicas , Oocitos/metabolismo , Animales , Proteínas Argonautas/metabolismo , Secuencia de Bases , Caenorhabditis elegans/citología , Proteínas de Caenorhabditis elegans/metabolismo , División Celular , Embrión no Mamífero/citología , Desarrollo Embrionario , Femenino , Cinesinas/metabolismo , Microtúbulos/metabolismo , Datos de Secuencia Molecular , Procesamiento Postranscripcional del ARN
18.
Cell ; 167(7): 1705-1718.e13, 2016 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-27984722

RESUMEN

Metformin has utility in cancer prevention and treatment, though the mechanisms for these effects remain elusive. Through genetic screening in C. elegans, we uncover two metformin response elements: the nuclear pore complex (NPC) and acyl-CoA dehydrogenase family member-10 (ACAD10). We demonstrate that biguanides inhibit growth by inhibiting mitochondrial respiratory capacity, which restrains transit of the RagA-RagC GTPase heterodimer through the NPC. Nuclear exclusion renders RagC incapable of gaining the GDP-bound state necessary to stimulate mTORC1. Biguanide-induced inactivation of mTORC1 subsequently inhibits growth through transcriptional induction of ACAD10. This ancient metformin response pathway is conserved from worms to humans. Both restricted nuclear pore transit and upregulation of ACAD10 are required for biguanides to reduce viability in melanoma and pancreatic cancer cells, and to extend C. elegans lifespan. This pathway provides a unified mechanism by which metformin kills cancer cells and extends lifespan, and illuminates potential cancer targets. PAPERCLIP.


Asunto(s)
Metformina/farmacología , Acil-CoA Deshidrogenasa/genética , Envejecimiento , Animales , Tamaño Corporal , Caenorhabditis elegans/crecimiento & desarrollo , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Línea Celular Tumoral , Proteínas de Unión al ADN/metabolismo , Humanos , Longevidad , Diana Mecanicista del Complejo 1 de la Rapamicina , Mitocondrias/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Complejos Multiproteicos/metabolismo , Neoplasias/tratamiento farmacológico , Poro Nuclear/metabolismo , Fosforilación Oxidativa , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Factores de Transcripción/metabolismo
19.
Mol Cell ; 83(11): 1810-1826.e8, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37267903

RESUMEN

Microprocessor (MP), DROSHA-DGCR8, processes primary miRNA transcripts (pri-miRNAs) to initiate miRNA biogenesis. The canonical cleavage mechanism of MP has been extensively investigated and comprehensively validated for two decades. However, this canonical mechanism cannot account for the processing of certain pri-miRNAs in animals. In this study, by conducting high-throughput pri-miRNA cleavage assays for approximately 260,000 pri-miRNA sequences, we discovered and comprehensively characterized a noncanonical cleavage mechanism of MP. This noncanonical mechanism does not need several RNA and protein elements essential for the canonical mechanism; instead, it utilizes previously unrecognized DROSHA dsRNA recognition sites (DRESs). Interestingly, the noncanonical mechanism is conserved across animals and plays a particularly significant role in C. elegans. Our established noncanonical mechanism elucidates MP cleavage in numerous RNA substrates unaccounted for by the canonical mechanism in animals. This study suggests a broader substrate repertoire of animal MPs and an expanded regulatory landscape for miRNA biogenesis.


Asunto(s)
MicroARNs , Animales , MicroARNs/metabolismo , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Unión al ARN/metabolismo , Ribonucleasa III/metabolismo , ARN Bicatenario , Procesamiento Postranscripcional del ARN
20.
Mol Cell ; 82(17): 3239-3254.e11, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-36027913

RESUMEN

The high substrate selectivity of the ubiquitin/proteasome system is mediated by a large group of E3 ubiquitin ligases. The ubiquitin ligase CHIP regulates the degradation of chaperone-controlled and chaperone-independent proteins. To understand how CHIP mediates substrate selection and processing, we performed a structure-function analysis of CHIP and addressed its physiological role in Caenorhabditis elegans and human cells. The conserved function of CHIP in chaperone-assisted degradation requires dimer formation to mediate proteotoxic stress resistance and to prevent protein aggregation. The CHIP monomer, however, promotes the turnover of the membrane-bound insulin receptor and longevity. The dimer-monomer transition is regulated by CHIP autoubiquitylation and chaperone binding, which provides a feedback loop that controls CHIP activity in response to cellular stress. Because CHIP also binds other E3 ligases, such as Parkin, the molecular switch mechanism described here could be a general concept for the regulation of substrate selectivity and ubiquitylation by combining different E3s.


Asunto(s)
Proteínas de Caenorhabditis elegans , Ubiquitina-Proteína Ligasas , Ubiquitina , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Humanos , Chaperonas Moleculares/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA